bims-mistre Biomed News
on Mito stress
Issue of 2025–11–30
thirty-two papers selected by
Ellen Siobhan Mitchell, MitoQ



  1. Life (Basel). 2025 Nov 13. pii: 1744. [Epub ahead of print]15(11):
      Mitochondrial dysfunction contributes to female reproductive endocrine disorders and is frequently associated with multisystem symptoms. Insulin resistance (IR) is a common metabolic disorder strongly linked to polycystic ovary syndrome (PCOS), while premature ovarian insufficiency (POI) also impairs fertility. Mitochondrial DNA (mtDNA) deletions and the stress-responsive cytokine growth differentiation factor 15 (GDF-15) have recently emerged as complementary biomarkers of mitochondrial impairment. In this retrospective observational study, we examined reproductive hormones, plasma GDF-15, mtDNA deletions, and clinical symptoms in insulin-resistant women, including those with PCOS or POI. Eighty-one patients were divided into three subgroups: IR-only (n = 49), IR-PCOS (n = 19), and IR-POI (n = 13). IR was defined based on elevated insulin levels during oral glucose tolerance testing (>10 mU/L at 0 min, >50 mU/L at 60 min, >30 mU/L at 120 min) according to national gynecological endocrinology guidelines, acknowledging that IR is not universally accepted as a distinct clinical entity. POI was defined as reduced ovarian reserve before age 40 with anti-Müllerian hormone (AMH) <1.0 ng/mL. Clinical symptoms were assessed using a questionnaire, medical record, and physical examination. MtDNA deletions were detected by long-range PCR, and GDF-15 was measured by ELISA. Free thyroxine (T4) emerged as an independent predictor of GDF-15, suggesting that thyroid function modulates mitochondrial stress signaling in insulin-resistant women. MtDNA deletions and/or elevated GDF-15 correlated with endocrine, gastrointestinal, and neuropsychiatric symptoms, and reduced AMH/FSH ratios indicated impaired ovarian function. Cross-sectional analysis further revealed lower AMH and AMH/FSH ratios in older women with mtDNA deletions, consistent with a trend toward accelerated reproductive aging. Overall, these findings support the role of GDF-15 and mtDNA deletions as complementary biomarkers of mitonuclear stress, with potential relevance for both systemic and reproductive health.
    Keywords:  GDF-15; PCOS; POI; insulin resistance; mitochondrial dysfunction; mtDNA deletion
    DOI:  https://doi.org/10.3390/life15111744
  2. Int J Mol Sci. 2025 Nov 11. pii: 10923. [Epub ahead of print]26(22):
      Nicotinamide adenine dinucleotide (NAD+) boosting can sustain energy metabolism and neurovascular stability in the retinal tissue. Depletion of NAD+ is linked to the development of pathological retinal conditions, such as age-related macular degeneration (AMD) and diabetic retinopathy (DR). Mitochondrial dysfunction, oxidative stress, and inflammation occur in these diseases. This review summarizes substantial evidence of therapeutic NAD+ boosters, including nicotinamide, nicotinamide mononucleotide, or nicotinamide riboside. They help improve mitochondrial function and lessen neurovascular injury. We also emphasize the importance of natural products and sirtuins in facilitating cytoprotective effects through the regulation of mitochondrial balance and inflammation. Developments in drug delivery methods, such as nanoparticle encapsulation and targeted eye treatments, are promising for enhancing the bioavailability and effectiveness of NAD+ boosters. The novelty of this work is its combination of mechanistic insights regarding NAD+ metabolism with efficacy data from preclinical studies. Furthermore, natural products may work together to boost their therapeutic effects against retinal damage. Together, our review article highlights NAD+ metabolism as a potential therapeutic target for addressing retinal degeneration and maintaining vision in aging, neurologic disorders, and various metabolic diseases, including diabetes.
    Keywords:  B vitamins; inflammation; mitochondrial function; neuroprotection; nicotinamide; nicotinamide mononucleotide; oxidative stress; retinopathy; sirtuins
    DOI:  https://doi.org/10.3390/ijms262210923
  3. Int J Mol Sci. 2025 Nov 16. pii: 11077. [Epub ahead of print]26(22):
      Impaired glucose metabolism elevates the risk of neurodegenerative diseases by activating the receptor for advanced glycation end products (RAGE), thereby promoting oxidative and endoplasmic reticulum (ER) stress that leads to neuronal apoptosis. C-phycocyanin (C-PC), a natural pigment-protein complex derived from algae, possesses potent antioxidant and antiglycation properties; however, its capacity to modulate RAGE-mediated neurotoxicity remains to be fully elucidated. In this study, we established a RAGE-driven neuronal injury model by exposing differentiated SH-SY5Y cells to advanced glycation end products (AGEs; 300 μg/mL). Pretreatment with C-PC (15-50 μmol/L) improved cell viability, preserved neuronal morphology, and attenuated AGEs-induced oxidative stress, as indicated by reduced intracellular reactive oxygen species and mitochondrial superoxide levels. Furthermore, C-PC inhibited activation of the PERK-CHOP pathway, and upregulated Bcl-2 while downregulating Bax, thereby preventing cytochrome c release and reducing caspase-9/3 activation as well as apoptotic DNA fragmentation. These neuroprotective effects of C-PC were comparable to those observed with the selective RAGE antagonist FPS-ZM1. In conclusion, our findings demonstrate that C-PC confers robust protection against AGEs-induced neuronal injury by suppressing oxidative and ER stress pathways downstream of RAGE activation, highlighting its potential as a natural modulator of the AGE-RAGE axis for the prevention or treatment of diabetes-associated neurodegeneration.
    Keywords:  C-phycocyanin; SH-SY5Y cells; advanced glycation end products; endoplasmic reticulum stress; receptor for advanced glycation end products
    DOI:  https://doi.org/10.3390/ijms262211077
  4. Pharmaceuticals (Basel). 2025 Nov 08. pii: 1698. [Epub ahead of print]18(11):
      Mammalians are constantly exposed to exogenous and endogenous sources of free radicals that have both favorable and harmful effects on the cellular systems. Oxidative stress (OS) is an imbalance of reactive oxygen species (ROS) and antioxidants in the body that can lead to serious cell damage. It is associated with many diseases such as cancer, Alzheimer's disease and heart disease. Background/Objectives: The Nuclear factor-2 erythroid-related factor-2 (Nrf2) is a transcription factor that controls the cellular oxidation state using antioxidant systems in the body and affects mitochondrial activities. Increased Nrf2 levels serve to protect cells from mitochondrial toxins; however, Nrf2 activity is inhibited in mitochondria-related diseases. In addition, Nrf2 is involved in mitochondrial activities for OS control. Methods: As mitochondrial wellbeing and activity is the chief controller for cellular metabolism, Nrf2 is a critical regulator for metabolic pathways. Thus, Nrf2 is the chief organizer of protection against OS in the cells. Nrf2 activator molecules support mitochondrial activity by stimulating mitophagy and helping to battle OS-related permeability transition. Conclusions: This review describes the influence of Nrf2 on OS and the way Nrf2 modulates mitochondrial function. Furthermore, we highlight recent studies of Nrf2 regarding its possible role in cell systems as well as pharmacological implications. Furthermore, this review emphasizes the importance of the mitochondria in the development of life-threatening diseases; pharmacological activation of Nrf2 is an important strategy to counter mitochondrial dysfunction.
    Keywords:  Nrf2; ROS; antioxidant; mitochondrial function; oxidative stress
    DOI:  https://doi.org/10.3390/ph18111698
  5. Ageing Res Rev. 2025 Nov 25. pii: S1568-1637(25)00303-4. [Epub ahead of print] 102957
      This review delves into the intricate relationship between serotonin signaling, mitophagy and mitochondrial dysfunction in Alzheimer's disease (AD), with a focus on the mechanistic pathways that link these processes and their potential therapeutic implications. A neurodegenerative condition called Alzheimer's disease is marked by cognitive deterioration. It is increasingly recognized as being influenced by impaired mitochondrial function and mitophagy, the selective degradation of damaged mitochondria. Serotonin, a neurotransmitter traditionally known for its role in mood regulation, has emerged as a critical modulator of mitochondrial dynamics and quality control through its interaction with key pathways such as the PINK1-Parkin and cAMP/PKA signaling pathways. In AD, alterations in serotonin levels and receptor function are associated with disruptions in mitophagy, leading to the accumulation of dysfunctional mitochondria, increased oxidative stress, and subsequent neuronal damage. This review synthesizes current evidence that links serotonin dysregulation to mitochondrial pathology in AD, exploring how impaired serotonin signaling exacerbates mitochondrial dysfunction and contributes to amyloid-beta (Aβ), phosphorylation of Tau (p-Tau) accumulations, and increased neuroinflammation. Additionally, we assessed the therapeutic potential of serotonin-targeting agents, particularly selective serotonin reuptake inhibitors (SSRIs), in restoring mitophagy, enhancing mitochondrial integrity, and attenuating neurodegeneration. By highlighting existing knowledge gaps and key controversies, this review underscores the promise of serotonin-mitochondria pathways as novel therapeutic targets and advocates for focused investigation into receptor-specific, mitophagy-centered interventions for AD.
    Keywords:  Alzheimer's disease; PINK1-Parkin pathway; mitochondrial dysfunction; mitophagy; selective serotonin reuptake inhibitors (SSRIs); serotonin
    DOI:  https://doi.org/10.1016/j.arr.2025.102957
  6. Free Radic Biol Med. 2025 Nov 21. pii: S0891-5849(25)01385-1. [Epub ahead of print]243 245-259
      APOE4, the strongest genetic risk factor for sporadic Alzheimer's disease (AD), is closely associated with mitochondrial dysfunction, yet the mechanisms remain poorly defined. We identify a previously unrecognized failure of the Nrf2-PINK1/Parkin axis in APOE4 neurons that compromises mitochondrial quality control. Unlike APOE3, APOE4 neurons fail to activate PINK1/Parkin-dependent mitophagy under stress, a defect compounded by impaired Nrf2 signaling and weakened antioxidant defenses. In vivo, APOE4 mice show age-dependent collapse of this pathway, correlating with progressive mitochondrial dysfunction and disrupted mito-nuclear communication. Pharmacological activation of Nrf2 or PINK1 restores mitochondrial clearance, highlighting the axis as a druggable node. These findings provide a mechanistic link between APOE4 and mitochondrial failure, establishing the Nrf2-PINK1/Parkin pathway as a critical driver of neurodegeneration and a promising target for therapeutic intervention in AD.
    Keywords:  APOE4; Alzheimer's disease (AD); Mito-nuclear communication; Mitochondrial stress; Mitophagy; Nrf2-PINK/Parkin
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2025.11.040
  7. Mol Hum Reprod. 2025 Nov 26. pii: gaaf058. [Epub ahead of print]
      Ovarian fibrosis is increasingly recognized as a pivotal factor contributing to ovarian aging, dysfunction and female infertility. It results from chronic or repetitive ovarian injury, such as that caused by repeated ovulation, which induces inflammation and excessive extracellular matrix (ECM) deposition, predominantly by activated fibroblasts and myofibroblasts. The key molecular pathways driving ovarian fibrosis include transforming growth factor-beta (TGF-β) /Smad signaling, Wnt/β-catenin and PI3K/Akt pathways, which orchestrate fibroblast activation, ECM remodeling and tissue stiffening. Elevated collagen types I and III, fibronectin and hyaluronan characterize the fibrotic ovarian stroma, disrupting normal folliculogenesis and steroidogenesis. Ovarian fibrosis is also implicated in reproductive pathologies such as polycystic ovary syndrome (PCOS), premature ovarian insufficiency (POI) and endometriosis, and may contribute to an increased risk of ovarian cancer, although definitive causal links require further elucidation. Current therapeutic strategies remain largely experimental, focusing on antifibrotic agents such as pirfenidone, TGF-β inhibitors and modulation of oxidative stress, alongside emerging interventions such as stem cell therapies, which are offer potential avenues for intervention in the ovary. This review synthesizes current insights into the cellular and molecular mechanisms driving ovarian fibrosis, its association with reproductive disorders, and emerging therapeutic strategies. It underscores key knowledge gaps and emphasizes the need for future research focused on fibroblast activation, inflammatory signaling and immune-ECM interactions to facilitate the development of targeted, long-term interventions aimed at preventing or reversing ovarian fibrosis and preserving female fertility.
    Keywords:  female fertility; ovarian aging; ovarian dysfunction; ovarian fibrosis; therapeutic targets
    DOI:  https://doi.org/10.1093/molehr/gaaf058
  8. Cell Mol Biol Lett. 2025 Nov 24. 30(1): 140
       BACKGROUND: Ovarian aging-induced decline in oocyte quality has been a main issue in women of advanced maternal age. However, the potential mechanism remains elusive, and there are no effective strategies to ameliorate aged oocyte quality. The lipid metabolism of oocytes has drawn great attention, but the intrinsic regulation of oocyte quality by metabolites, metabolic enzymes, and intracellular mediators is less well-characterized.
    METHODS: Targeted lipidomics was employed to detect the neutral lipids in oocytes during maturation. We used 4,4-difluoro-1,3,5,7,8-pentamethyl-4-bora-3a,4a-diaza-s-indacene (BODIPY 493/503) and Filipin to stain cholesteryl ester and free cholesterol, respectively. The Cholesterol/Cholesteryl Ester Quantification Assay kit was used further to quantify cholesterol-related metabolites. Western blotting was performed to evaluate acyl-coenzyme A: cholesterol acyltransferase 1/2 (ACAT1/2) expression. Immunofluorescence and quantitative real-time polymerase chain reaction (qRT-PCR) were conducted to validate the knockdown efficiency of ACAT1. Avasimibe treatment and ACAT1 small interfering RNA (siRNA) microinjection were performed to investigate the effect of impaired cholesterol-cholesteryl ester metabolism on oocyte quality. Single-oocyte RNA sequencing was conducted to explore the mechanism. Mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) production, reactive oxygen species (ROS), and mitochondrial autophagosomes were detected to evaluate mitochondrial function and mitophagy.
    RESULTS: There is a profound increase in the conversion of cholesterol to cholesteryl ester in oocytes during maturation, which depends on ACAT1. Conversely, disturbing the homeostasis of cholesterol-cholesteryl ester metabolism by manipulating ACAT1 impairs oocyte quality, primarily manifested as decreased polar body extrusion (PBE), increased meiotic defects, and abnormal early embryonic development. Mechanistically, the impaired conversion of cholesterol to cholesteryl ester reduces oocyte mitophagy, leading to mitochondrial dysfunction, including reduced MMP and ATP production, and excessive accumulation of ROS. Notably, we also reveal that this metabolic homeostasis is impaired in aged oocytes, accompanied by decreased ACAT1 levels. Moreover, cholesteryl ester supplementation via cholesterol conjugated to methyl-β-cyclodextrin (CCM) can effectively ameliorate aged oocyte quality by enhancing mitophagy.
    CONCLUSIONS: This study reveals the mechanism by which cholesterol-cholesteryl ester metabolism regulates oocyte quality and thus participates in the process of oocyte aging by influencing mitophagy and mitochondrial function.
    Keywords:  ACAT1; Cholesterol–cholesteryl ester metabolism; Mitochondrial function; Mitophagy; Oocyte aging; Oocyte maturation
    DOI:  https://doi.org/10.1186/s11658-025-00811-w
  9. bioRxiv. 2025 Oct 14. pii: 2025.10.10.681622. [Epub ahead of print]
      Menopause affects the aging process in women through significant ovarian hormone production decline in midlife. Women who experience early menopause face an accelerated physiological aging rate, along with impaired memory and increased risks of neurodegenerative diseases. However, it remains elusive how the timing of menopause affects brain activity, which could be crucial for understanding menopause-related acceleration of aging and increased risk of dementia. Recent studies have revealed a highly structured infra-slow (< 0.1 Hz) global brain activity across species and linked it to arousal and memory functions, as well as waste clearance in Alzheimer's diseases (AD). In this study, we examined how this global brain activity relates to age of menopause using resting-state fMRI data from the Human Connectome Project-Aging dataset. We found that women who experienced earlier menopause (mean menopausal age 45±3.5 yr) exhibited weaker global brain activity ( p = 5.0 × 10 -4 ) with reduced coupling to cerebrospinal fluid (CSF) flow ( p = 0.017) compared to age-matched later-menopausal women (mean menopausal age 54±1.2 yr). Differences appeared mainly in higher-order brain regions, where activation levels correlated with memory performance in earlier but not in intermediate or later menopausal women. These findings highlight brain activity changes linked to early menopause, suggesting a potential mechanism underlying memory decline and the increased risk of AD and dementias in early-onset menopausal women.
    DOI:  https://doi.org/10.1101/2025.10.10.681622
  10. Front Psychiatry. 2025 ;16 1667639
      Creatine, as a naturally occurring organic compound, has gained attention for its potential role in psychiatric health. The creatine kinase-phosphocreatine energy buffer system plays a crucial role in maintaining energy supply in the brain. Brain bioenergetic deficits, particularly those related to mitochondrial dysfunction, plays a critical role in the pathophysiology of psychiatric illnesses. A growing body of literature has focused on the potential therapeutic role of creatine supplementation in psychiatric illnesses. This review summarizes findings from preclinical, epidemiological, clinical and neuroimaging studies to examine creatine's role as both a biomarker and therapeutic agent in psychiatric disorders, including Major Depressive Disorder, Anxiety Disorders, Posttraumatic Stress Disorder, and Substance Use Disorder.
    Keywords:  anxiety disorder; creatine; major depressive disorder; posttraumatic stress disorder; substance use disorder
    DOI:  https://doi.org/10.3389/fpsyt.2025.1667639
  11. Nutrients. 2025 Nov 15. pii: 3571. [Epub ahead of print]17(22):
      Background: Skeletal muscle aging is characterized by impaired myogenic differentiation, disrupted circadian rhythms, elevated oxidative stress, and mitochondrial dysfunction. Rutin, a natural flavonoid with antioxidant properties, has been suggested to mitigate aging processes; however, its effects on circadian regulation and muscle homeostasis remain unclear. Methods: In vitro, differentiated C2C12 myotubes were treated with D-galactose (D-gal, 20 g/L) with or without rutin (20 μM). In vivo, C57BL/6 mice were supplemented with rutin (100 mg/kg b.w.) via oral gavage in a D-gal-induced aging mouse model (150 mg/kg b.w., i.p.). Results: D-gal induced cellular senescence, impaired myogenic differentiation, disrupted circadian oscillations, increased oxidative stress, and compromised mitochondrial function. Rutin treatment restored myotube formation, enhanced circadian rhythmicity of differentiation-related genes, and corrected the antiphase patterns of Per2 and Rorc. It also reduced reactive oxygen species and malondialdehyde levels; increased superoxide dismutase, catalase, and glutathione peroxidase activity; improved ATP production and membrane potential; and decreased mitochondrial oxidative aging, as confirmed by pMitoTimer imaging. Furthermore, rutin reinstated the rhythmic expression of oxidative phosphorylation proteins and Pgc1α. In vivo, rutin supplementation enhanced muscle performance (prolonged hanging time) and oxidative capacity, particularly at night (ZT14-ZT16), without altering muscle fiber-type distribution, and normalized circadian rhythmicity of core clock genes. Conclusions: Rutin attenuates D-gal-induced cellular senescence by modulating circadian rhythms, reducing oxidative stress, and improving mitochondrial function. Importantly, its in vivo effects on muscle performance and circadian regulation suggest that rutin is a promising therapeutic strategy to counteract skeletal muscle aging and sarcopenia.
    Keywords:  circadian rhythm; mitochondrial function; muscle senescence; oxidative stress; rutin
    DOI:  https://doi.org/10.3390/nu17223571
  12. Ageing Res Rev. 2025 Nov 21. pii: S1568-1637(25)00301-0. [Epub ahead of print] 102955
      Type 2 diabetes mellitus (T2DM) is increasingly recognized as a shared pathological substrate for both sarcopenia and cognitive decline, particularly Alzheimer's disease (AD). This review synthesizes current evidence on the converging molecular pathways linking insulin resistance, hyperglycaemia, mitochondrial dysfunction, oxidative stress, and chronic inflammation to muscle wasting and neurodegeneration. Central to this interplay is the muscle-brain axis, a bidirectional communication network mediated by myokines, exercise-induced cytokines that influence metabolic and neural homeostasis. Key myokines such as IGF-1, irisin, BDNF, FGF21, and SPARC promote myogenesis, synaptic plasticity, and neuroprotection, while others including myostatin, IL-8, and GDF-15 exert detrimental effects. Context-dependent molecules such as IL-6, IL-15, lactate, and cathepsin-B show dual roles modulated by aging, inflammation, and metabolic state. Emerging data support that improved glycaemic control, enhanced insulin sensitivity, and sustained physical activity can attenuate both sarcopenia and cognitive decline. This review aims to summarize current evidence describing how insulin resistance, chronic hyperglycaemia, mitochondrial dysfunction, oxidative stress, and inflammation interact to promote both muscle wasting and neurodegeneration. Keywords.
    Keywords:  Brain; diabetes; muscle; myokines; sarcopenia
    DOI:  https://doi.org/10.1016/j.arr.2025.102955
  13. bioRxiv. 2025 Nov 06. pii: 2025.11.04.686626. [Epub ahead of print]
      Asymptomatic Alzheimer's disease (AsymAD) refers to individuals who, despite exhibiting amyloid-β plaques and tau pathology comparable to Alzheimer's disease (AD), maintain cognitive performance similar to cognitively normal individuals. The resilience mechanism in these AsymAD individual remains understudied. We performed a systematic analysis comparing AsymAD and AD across multiple cohorts (ROSMAP, Banner and Mount Sinai), brain regions (BA6, BA9, BA36 and BA37) and neuronal and glial cell types using proteomics and transcriptomics data. AsymAD brains exhibited preserved mitochondrial bioenergetics, characterized by enhanced oxidative phosphorylation (OXPHOS), electron transport chain (ETC) activity, fatty acid and lipid metabolism, and branched-chain amino acid (BCAA) utilization. Pathways regulating mitochondrial complex biogenesis and calcium homeostasis were also upregulated. Key mitochondrial proteins such as MRPL47, CPT2, BCAT2, and IDH2, were consistently upregulated in AsymAD, whereas MACROD1 was downregulated. At the cellular level, excitatory neurons, including superficial, mid-layer, and deep-layer subtypes, exhibited the most preserved mitochondrial function, whereas vulnerable inhibitory subtypes, including PVALB and SST neurons, showed increased cellular abundance and bioenergetic activity. In contrast, microglia and oligodendrocytes proportions were reduced in AsymAD relative to AD. Our findings identify preserved mitochondrial bioenergetics as a defining feature of resilience in AD and suggest that enhancing NADH metabolism via NAD+ precursor-based interventions may potentially help in maintaining cognitive function despite amyloid and tau pathology.
    DOI:  https://doi.org/10.1101/2025.11.04.686626
  14. Appl Psychophysiol Biofeedback. 2025 Nov 27.
      Heart rate variability biofeedback (HRV-B) is a technique that has been shown to have benefits for both physical and mental health conditions. The aim of the present systematic review and meta-analysis is to describe the methodological approach and examine the efficacy of remote HRV-B interventions for mental health symptoms, including depression, stress, anxiety, and sleep as well as heart rate variability (HRV), a measure of autonomic function. A systematic literature search was conducted from five databases and eighteen studies with a total of 1352 subjects from different populations were included in this systematic review and meta-analysis based on the inclusion criteria. Meta-analysis showed a medium effect size of HRV-B for improving both depression (g = - 0.41 [- 0.049, - 0.772], p = 0.026, I2 = 72.623%, k = 10) and HRV (g = 0.443, [0.718, 0.167], p = 0.002, I2 = 56.81%, k = 10) compared to control conditions. These results remained significant after sensitivity analyses excluding high-risk, non-randomized, and outlier studies. Effect for stress was not significant (p = 0.152, k = 8). Meta-regressions identified study and protocol characteristics as significant moderators of the effect on HRV, stress, and anxiety. Specifically, maximizing resonance, screen on the device, less lab visits, less than twenty minutes of practice time per day, and female gender were beneficial for the intervention effectiveness. Overall, this study indicates that HRV-B is an effective intervention for decreasing depressive symptoms and increasing HRV across populations, and some intervention variables may influence the intervention outcomes.
    Keywords:   Biofeedback; Heart rate variability; Mental health
    DOI:  https://doi.org/10.1007/s10484-025-09750-w
  15. Int J Mol Sci. 2025 Nov 18. pii: 11155. [Epub ahead of print]26(22):
      Reactive oxygen species (ROS), inevitable by-products of aerobic metabolism, act both as regulators of signaling pathways and as mediators of oxidative stress and aging-related damage. Protein oxidative post-translational modifications (Ox-PTMs) are recognized hallmarks of aging and metabolic decline, yet the persistence of protein oxidation under different physiological conditions, such as age and diet, remains unclear. Here, we applied proteomics to mitochondrial and membrane-enriched fractions of male Fischer 344 rat cerebrum and heart, comparing Ox-PTMs across young and aged animals subjected to ad libitum nutrition (AL) or calorie restriction (CR). We identified 139 mitochondrial and membrane-associated proteins consistently exhibiting high levels of oxidation, including tricarboxylic acid (TCA) cycle enzymes, respiratory chain subunits, ATP synthase components, cytoskeletal proteins, and synaptic vesicle regulators. Functional enrichment and network analyses revealed that oxidized proteins clustered in modules related to mitochondrial energy metabolism, membrane transport, and excitation-contraction coupling. Notably, many proteins remained persistently oxidized, predominantly as mono-oxidation, without significant changes during aging or CR. Moreover, the enzymatic activity of mitochondrial complexes was not only preserved but significantly enhanced in specific contexts, and the structural integrity of the respiratory chain was maintained. These findings indicate a dual strategy for coping with oxidative stress: CR reduces ROS production to limit oxidative burden, while protein and network robustness enable functional adaptation to persistent oxidation, collectively shaping mitochondrial function and cellular homeostasis under differing physiological conditions.
    Keywords:  aging; calorie restriction; mitochondria; oxidative post-translational modifications; rat cerebrum; rat heart; reactive oxygen species
    DOI:  https://doi.org/10.3390/ijms262211155
  16. Antioxidants (Basel). 2025 Oct 24. pii: 1278. [Epub ahead of print]14(11):
      Dysregulated redox signaling, mitochondrial dysfunction and impaired autophagy form an interconnected network that drives inflammatory and immune responses in cardiovascular disease. Among these, disturbances in redox balance, largely mediated by reactive oxygen species (ROS), serve as key drivers linking inflammatory signaling to adverse cardiovascular outcomes. Mitochondria are essential for energy production and cellular homeostasis, but their dysfunction leads to the accumulation of excessive ROS, which triggers inflammation. This pro-oxidative milieu disrupts immune regulation by activating inflammasomes, promoting cytokine secretion, triggering immune cell infiltration and ultimately contributing to cardiovascular injury. Conversely, intracellular degradation processes such as mitophagy alleviate these effects by selectively eliminating dysfunctional mitochondria, thereby decreasing ROS levels and maintaining immune homoeostasis. These interconnected processes influence myeloid cell function, including mitochondrial reprogramming, macrophage polarization and autophagic activity. The modulation of these immune responses is crucial for determining the severity and resolution of cardiac and vascular inflammation, and consequently the extent of cellular injury. This review examines the latest developments and understanding of the intricate relationships between redox signaling, mitochondrial dysfunction, autophagy and oxidative stress in modulating inflammation and immune responses in cardiovascular diseases. Understanding these interrelationships will inform future studies and therapeutic solutions for the prevention and treatment of cardiovascular diseases.
    Keywords:  autophagy; cardiovascular disease; inflammation; mitochondrial dysfunction; oxidative stress; redox signaling
    DOI:  https://doi.org/10.3390/antiox14111278
  17. Antioxidants (Basel). 2025 Oct 24. pii: 1276. [Epub ahead of print]14(11):
      Rotenone, a classical inhibitor of mitochondrial complex I, disrupts electron transport and promotes the generation of reactive oxygen species (ROS), contributing to inflammation and cell death. However, the precise molecular mechanisms linking mitochondrial dysfunction to inflammatory signaling remain incompletely understood. In this study, we investigated the role of the cGAS-STING pathway in rotenone-induced NLRP3 inflammasome activation in PMA-differentiated THP-1 macrophages. Rotenone treatment activated the cGAS-STING axis, as evidenced by increased cGAS expression and the phosphorylation of STING and TBK1. This activation led to the nuclear translocation of NF-κB and the upregulation of NLRP3, promoting inflammasome priming and IL-1β secretion. Inhibition of STING using H-151 markedly suppressed NLRP3 expression, NF-κB activation, and IL-1β release. Similarly, cyclosporin A, an inhibitor of mitochondrial permeability transition pore opening, reduced mitochondrial ROS, cytosolic oxidized mitochondrial DNA, and downstream activation of the cGAS-STING pathway, thereby attenuating inflammasome activation. These findings demonstrate that rotenone activates the NLRP3 inflammasome via mitochondrial ROS-mediated release of mtDNA and subsequent activation of the cGAS-STING-NF-κB signaling axis in THP-1-derived macrophages.
    Keywords:  NF-κB; NLRP3 inflammasome; cGAS–STING pathway; mitochondrial DNA; rotenone
    DOI:  https://doi.org/10.3390/antiox14111276
  18. bioRxiv. 2025 Nov 11. pii: 2025.11.10.687744. [Epub ahead of print]
      Aging leads to quantitative and qualitative changes in platelet (Plt) production, with increased risk for thrombosis and other adverse cardiovascular events. Recent reports showed that aging promotes the emergence of non-canonical (nc) megakaryocyte progenitors (MkPs) directly from hematopoietic stem cells (HSCs), leading to the production of hyperactive Plts. The higher engraftment potential of ncMkPs compared to both young and old canonical (c)MkPs, contrasts with the functional decline of old HSCs. Emerging reports suggest that mitochondrial function critically regulates lineage commitment and cellular functionality, but how mitochondrial activity affects aging megakaryopoiesis is unknown. Here, we demonstrate that aged MkPs sustain unique mitochondrial activity, characterized by higher mitochondrial membrane potential, higher ATP content, and lower ROS levels compared to their younger counterparts. This contrasts with the dysfunctional mitochondrial state observed in old HSCs, suggesting lineage-specific organelle adaptations upon aging. Notably, we observed that the elevated mitochondrial capacity in aged MkPs is driven selectively by the age-specific ncMkPs. Paradoxically, in vivo pharmacological enhancement of mitochondrial activity in old mice reduced in situ Plt production, but increased Plt reconstitution by transplanted HSCs. These discoveries link uniquely regulated mitochondrial capacity to the intrinsic properties of age-specific MkPs, raising the possibility of therapeutic targeting to prevent aging-induced megakaryopoiesis.
    HIGHLIGHTS: Aging-specific MkPs have elevated mitochondrial capacity, the inverse of aged HSCsMitochondrial enhancement differentially alters platelet counts in young and old miceEnhancement of mitochondrial capacity increases platelet repopulation by both young and old HSCs.
    DOI:  https://doi.org/10.1101/2025.11.10.687744
  19. Geroscience. 2025 Nov 25.
      Growth differentiation factor 15 (GDF-15) has been proposed as a potential biomarker for the geriatric syndromes of frailty and sarcopenia. This study investigated plasma GDF-15 as a biomarker associated with sarcopenia and frailty in a cohort of mobility-limited older adults. Patients (≥ 65 years) were recruited from a geriatric outpatient clinic for fall assessment. Frailty was identified using the Clinical Frailty Scale with a cut-off ≥ 5. Sarcopenia was defined according to the European Working Group on Sarcopenia in Older People 2. Plasma GDF-15 was analyzed using electrochemiluminescence assays (Meso Scale Discovery, Rockville, MD, USA). Receiver operating characteristic (ROC) analysis was performed to assess the diagnostic accuracy of GDF-15 for identifying frailty and sarcopenia. The optimum GDF-15 cut-off for frailty was determined using Youden's Index. A total of 429 patients (age 79.6 ± 6.2 years, 64% female) were included. Of these, 16% had sarcopenia and 25% had frailty. Median GDF-15 levels in patients with/without sarcopenia and with/without frailty were 1916 pg/mL vs. 1569 pg/mL (p = 0.035), and 2252 pg/mL vs. 1438 pg/mL (p < 0.001), respectively. The area under the curve (AUC) for frailty was 0.681 (95% CI: 0.623-0.739) with an optimal cut-off of 2047 pg/mL. For sarcopenia, AUC was 0.577 (95% CI: 0.501-0.653). GDF-15 plasma levels appear to be elevated in mobility-limited older adults with frailty and sarcopenia. The diagnostic accuracy of GDF-15 for frailty appears of moderate magnitude, suggesting a potential clinical role of this biomarker in identifying frailty in mobility-limited older adults. In contrast, the diagnostic accuracy of GDF-15 for sarcopenia was poor. ClinicalTrials.gov Identifier: NCT05795556.
    Keywords:  Biomarker; Frailty; GDF-15; Mobility-Limited Patients; Old Age; Sarcopenia
    DOI:  https://doi.org/10.1007/s11357-025-01946-6
  20. Cells. 2025 Nov 14. pii: 1786. [Epub ahead of print]14(22):
      Habituation is a fundamental form of non-associative learning that allows organisms to filter out repetitive, non-salient stimuli but declines with age. While the kynurenine pathway (KP) of tryptophan metabolism is implicated in psychiatric and neurodegenerative diseases, its role in age-related habituation deficits has been overlooked. This review proposes a systems-level framework suggesting that age-related, chronic inflammation KP dysregulation is a key driver of habituation deficits. We present evidence showing that neurotoxic metabolites from the kynurenine-3-monooxygenase (KMO)-dependent branch drive a self-reinforcing cycle of oxidative stress, excitotoxicity, and glial reactivity that destabilizes the neural circuits required for habituation. This framework redefines KP modulation as context dependent: metabolites such as kynurenic acid (KYNA), which can be disruptive when elevated in youth, may become compensatory under the oxidative load of aging. Our findings that genetic KMO deletion preserves habituation in aged and old mice provide the first direct in vivo evidence supporting this model. We propose that inhibiting the KMO branch preserves habituation not by simply altering metabolite levels but by restoring homeostatic balance across neuroimmune, redox, and plasticity networks. KMO thus emerges as a critical node for maintaining cognitive resilience, offering a therapeutic target for preserving brain function during aging.
    Keywords:  KMO; aging; habituation; kynurenine pathway; non-associative learning and memory
    DOI:  https://doi.org/10.3390/cells14221786
  21. Res Sq. 2025 Oct 13. pii: rs.3.rs-7634140. [Epub ahead of print]
      Mitochondrial dysfunction and pro-inflammatory signaling are each key drivers of aging. However, a clear understanding of the connections between mitochondrial homeostasis, inflammation and lifespan determination remains elusive. Upon mitochondrial stress or damage, mtDNA can be released into the cytosol thus encountering cytosolic DNA sensors and activating pro-inflammatory responses. Here, we report a striking age-related increase in cytosolic mtDNA, which can be counteracted by mitophagy, in Drosophila brain and muscle tissue. We find that upregulation of DNase II, an acid DNase which digests DNA in the autophagy-lysosome system, reduces cytosolic mtDNA levels in aged flies and prolongs healthspan. Reducing the abundance of cytosolic DNA in aged flies also dampens Rel/NF-κB pro-inflammatory signaling. Furthermore, we show that inhibition of EYA, a Rel/NF-κB-binding protein involved in immune sensing of DNA, in aging neurons counteracts brain aging and prolongs healthspan. Our findings identify DNase II and EYA as therapeutic targets to prolong healthspan.
    DOI:  https://doi.org/10.21203/rs.3.rs-7634140/v1
  22. Aging Dis. 2025 Nov 16.
      Endothelial senescence is a critical contributor of arterial dysfunction and age-related cardiovascular diseases. This study demonstrates that long-term senolytic treatment with dasatinib plus quercetin (D+Q; 5 mg/kg + 50 mg/kg biweekly for 8 months) in mice significantly attenuates vascular endothelial senescence. D+Q lowered senescence markers (p21 protein and SA-β-gal positivity) in aged mesenteric arteries and human umbilical vein endothelial cells (HUVECs), while maintaining endothelial integrity. Transcriptomic analysis indicated activation of the relaxin signaling pathway and upregulation of nitric oxide synthase isoforms. Mechanistically, D+Q reversed age-related eNOS uncoupling by promoting dimerization, increased nitric oxide bioavailability, and reduced mitochondrial dysfunction, evidenced by restored mitochondrial ultrastructure, decreased mitochondrial mass, and lowered reactive oxygen species (ROS) production. Consequently, D+Q restored endothelium-dependent vasodilation and enhanced blood flow in aged mesenteric arteries following acetylcholine stimulation. These findings demonstrate that clearance of senescent endothelial cells via senolytic therapy mitigates arterial aging by restoring mitochondrial homeostasis and eNOS function, highlighting its therapeutic potential for age-related vascular dysfunction.
    DOI:  https://doi.org/10.14336/AD.2025.0976
  23. Aging Dis. 2025 Nov 09.
      Cardiovascular diseases (CVDs) have long been the leading cause of mortality among the elderly worldwide. Despite substantial progress, a complete understanding of the initiation and progression of CVDs remains elusive. Emerging evidence suggests that the risk of developing CVDs increases with aging and prolonged insulin resistance (IR). Insulin is a key metabolic hormone crucial for regulating glucose and lipid metabolism in various tissues. An impaired tissue response to insulin stimulation results in IR. With aging, pathological changes such as visceral obesity, chronic inflammation, and oxidative stress collectively exacerbate IR, leading to dyslipidemia, hyperglycemia, and hypertension. These conditions highlight IR as a critical factor linking aging to various CVDs, including atherosclerosis, heart failure, and hypertension. Thus, preventing IR is essential for preserving cardiac function. In this review, the pathological mechanisms of IR in elderly individuals are summarized, emphasizing their association with aging-related CVDs. Additionally, potential therapeutic targets associated with IR for treating CVDs are discussed, along with current limitations and future directions for cardiac recovery strategies.
    DOI:  https://doi.org/10.14336/AD.2025.1108
  24. Redox Biol. 2025 Nov 14. pii: S2213-2317(25)00440-9. [Epub ahead of print]88 103927
      Ageing is associated with attenuated exercise responses in skeletal muscle, which may be related to a failure of muscle redox signalling. The attenuation of redox responses to exercise in aged muscle has been linked to perturbations in redox homeostasis induced by age-related increases in mitochondrial oxidative stress. Accordingly, we investigated the effects of supplementation with the mitochondria-targeted antioxidant MitoQ on mitochondrial bioenergetics and H2O2 emission as well as acute exercise-induced redox responses in skeletal muscle of older individuals. In a randomised, double-blind, placebo-controlled, parallel design, 10 males and 12 females aged 65-80 years were randomised to receive either MitoQ (20 mg/day) or a placebo for 12 weeks before completing a single bout of exercise. Vastus lateralis muscle biopsies were collected before supplementation and before, immediately post- and 4 h post-exercise. MitoQ supplementation reduced mitochondrial H2O2 emission capacity in skeletal muscle but did not impact mitochondrial respiration, H2O2 emission in the presence of ADP, or the sensitivity for ADP to stimulate respiration (apparent Km) and attenuate H2O2 emission (apparent IC50). Acute exercise-induced peroxiredoxin oxidation in skeletal muscle was not altered by MitoQ supplementation. Similarly, MitoQ had no effect on the phosphorylation of several redox-sensitive protein kinases (AMPK, p38 MAPK, and ERK1/2) or the upregulation of mitochondrial and antioxidant genes following exercise. Collectively, these findings indicate that MitoQ supplementation did not influence the basal myocellular redox state or redox responses to exercise in skeletal muscle of older individuals.
    Keywords:  Ageing; Antioxidant; Mitochondria; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.redox.2025.103927
  25. Sci Rep. 2025 Nov 28. 15(1): 42766
      Alzheimer's disease (AD) is a progressive neurodegenerative condition that has become a global health challenge due to an aging world population and no available effective treatment. Mitochondrial dysfunction plays a crucial role in the development of AD due to its critical role in neuronal survival and function. However, the specific mitochondrial genes and pathways involved in AD pathogenesis remain poorly defined. In this study, we incorporated seven AD human postmortem and three AD iPSC-derived neurons (iNs) gene expression datasets to identify mitochondria-related Differentially Expressed Genes (mitoDEGs) between AD and control. The Gene Ontology (GO) analysis is conducted to investigate the AD biological mechanisms, and a random forest model is developed to assess how well the key mitoDEGs differentiate AD and control groups. Through our analysis, we identified fourteen key mitochondria related genes that show significant dysregulation in both postmortem brain tissues and iNs derived from AD patients. These genes have strong connections to oxidative stress, indicating mitochondrial dysfunction plays a crucial role in Alzheimer's disease pathology. Our study identified the key genes and pathways as promising targets for future research and therapeutic interventions, highlighting the importance of mitigating oxidative stress and restoring mitochondrial function in AD.
    Keywords:  Alzheimer disease; Autopsy; Induced pluripotent stem cells; Mitochondria; Reactive oxygen species
    DOI:  https://doi.org/10.1038/s41598-025-27019-4
  26. bioRxiv. 2025 Nov 07. pii: 2025.11.06.687021. [Epub ahead of print]
       Study Objectives: Women experience more sleep disruptions than men, particularly during hormonal transitions such as puberty, pregnancy, and menopause. This study investigated the role of estradiol (E2) in regulating sleep-wake behavior in female rats and identified the brain regions involved.
    Methods: Using an exogenous E2 replacement model in ovariectomized rats, we assessed changes in sleep-wake patterns via EEG/EMG telemetry. The effects of E2 and progesterone, selective estrogen receptor agonists, and direct brain infusions of E2 and receptor antagonists were evaluated.
    Results: E2 administration increased wakefulness, reduced non-rapid eye movement (NREM) and rapid eye movement (REM) sleep, and decreased NREM slow-wave activity (SWA), predominantly during the dark phase. These effects required both estrogen receptor alpha (ERα) and beta (ERβ) activation and were mediated by estrogen receptor signaling within the median preoptic nucleus (MnPO). Direct infusion of E2 into the MnPO was sufficient to replicate systemic effects, while local infusion of the pure estrogen receptor antagonist ICI 182,780 (Fulvestrant) attenuated them. Progesterone did not augment E2's actions, and males showed no sleep-wake changes in response to E2, highlighting sex-specific mechanisms.
    Conclusions: The MnPO is a critical site where E2 regulates sleep-wake behavior. These findings provide a neurobiological framework for understanding how ovarian hormones contribute to sleep disruptions in women, offering potential therapeutic targets for sleep disorders related to hormonal changes.
    Statement of significance: Sleep disturbances disproportionately affect women, particularly during hormonal transitions such as puberty, pregnancy, and menopause. Despite this, the underlying mechanisms by which estradiol (E2) influences sleep-wake behavior have remained unclear. This study identifies the median preoptic nucleus (MnPO) as a critical site where E2 signaling regulates sleep-wake patterns in female rats. Our findings demonstrate that E2 increases wakefulness and reduces both non-rapid eye movement (NREM) and rapid eye movement (REM) sleep, with effects primarily observed during the dark phase. By showing that local E2 signaling in the MnPO is both necessary and sufficient for these changes, this work offers a novel neurobiological framework for understanding how fluctuations in ovarian hormones contribute to sleep disruptions in women. These insights lay the groundwork for developing targeted therapies to improve sleep health in women, particularly those experiencing hormonal changes.
    DOI:  https://doi.org/10.1101/2025.11.06.687021
  27. Diabetes Obes Metab. 2025 Nov 24.
       AIMS: Polycystic ovary syndrome (PCOS) is a common endocrine disorder in reproductive-age women, associated with insulin resistance, hyperandrogenism, and menstrual irregularities. Nutraceutical interventions-bioactive compounds derived from foods or supplements that provide health benefits beyond basic nutrition-may support conventional therapy. This study aimed to systematically evaluate the effects of nutraceutical interventions on reproductive and metabolic outcomes in PCOS.
    MATERIALS AND METHODS: This systematic review and meta-analysis was registered in PROSPERO (CRD42024521879) and followed PRISMA 2020. Searches were performed in PubMed, Embase, Cochrane Library, Web of Science, and regional databases (January 2013-December 2024) and included 78 eligible studies. Eligible randomized and quasi-experimental studies assessed nutraceuticals such as vitamin D, inositol, and quercetin. Risk of bias was evaluated with standardized tools; certainty of evidence was graded by GRADE.
    RESULTS: Seventy-eight studies met inclusion criteria. Nutraceutical interventions significantly reduced fasting insulin (MD = -2.14 μIU/mL; 95% CI -3.12 to -1.16) and luteinizing hormone (MD = -1.34 mIU/mL; 95% CI -2.10 to -0.58) and increased sex-hormone-binding globulin (MD = +3.72 nmol/L; 95% CI 1.35 to 6.09). Vitamin D supplementation showed the strongest metabolic and hormonal improvements. Results for ovarian follicle count and menstrual bleeding were inconsistent.
    CONCLUSIONS: Nutraceutical interventions targeting metabolic and hormonal regulation may complement lifestyle therapy in women with PCOS. Interpretation is limited by study heterogeneity and variable methodological quality. No external funding was received.
    Keywords:  PCOS; dietary supplement; nutraceuticals; phytonutrient‐rich foods; reproductive health
    DOI:  https://doi.org/10.1111/dom.70307
  28. J Ovarian Res. 2025 Nov 28. 18(1): 287
       OBJECTIVES: Ovarian aging is considered the "pacemaker" and "biological clock" of systemic female aging, with early manifestations that are often insidious. In this study, we analyzed the shared and distinct molecular signatures between physiological and pathological ovarian aging models using proteomic approaches, with the aim of identifying early predictive markers and therapeutic targets for ovarian aging, evaluating model fidelity, and elucidating underlying molecular mechanisms.
    METHODS: Ovarian tissue samples were collected from female C57/BL6 mice representing chemotherapeutic ovarian aging (8-week-old, Cyclophosphamide-Busulfan model) and natural ovarian aging (18-month-old). Initial validation of the models was conducted through histological assessment and serum hormone measurements. Quantitative proteomics was employed to profile protein expression. Differentially expressed proteins were subjected to GO, KEGG, and WikiPathways functional enrichment and clustering analyses, followed by validation of selected candidate genes using quantitative real-time polymerase chain reaction (RT-qPCR).
    RESULTS: The cyclophosphamide-busulfan induced pathological ovarian aging model exhibited partial consistency with natural ovarian aging in terms of histological morphology, hormone levels, and proteomic profiles. protein-protein interaction (PPI) network construction and pathway enrichment analyses provided further evidence supporting the strong association between alterations in the subcortical maternal complex (SCMC) and ovarian dysfunction. Notably, Cyp17a1 and Lhcgr were identified as potential early biomarkers for ovarian aging. Additionally, 7 key molecular targets (pbk, sdhd, Gsta3, Gstm6, Nlrp5, Nlrp4f, and Nlrp14) closely related to chronic inflammation and oxidative stress were identified, some of which may reveal the close relationship between aging and tumors. Comparative analysis further revealed that pathological ovarian aging is predominantly characterized by DNA damage and cell cycle dysregulation, whereas physiological aging predominantly involved immune dysfunction, abnormal lipid metabolism, and chronic low-grade inflammation, underscoring the molecular heterogeneity between aging subtypes.
    CONCLUSIONS: This study confirmed the validity of the cyclophosphamide-busulfan induced mouse model of ovarian senescence, highlighted both the shared and distinct molecular mechanisms of physiological and pathological ovarian aging, and identified promising early biomarkers and therapeutic intervention targets.
    Keywords:  Indicator prediction; Model evaluation; Ovarian aging; Proteomics; Therapeutic targets
    DOI:  https://doi.org/10.1186/s13048-025-01874-1
  29. Nat Commun. 2025 Nov 22.
      Sex-biased differences in Alzheimer's disease (AD) are well documented, but the mechanisms underlying increased vulnerability in postmenopausal women remain unclear. This study aimed to model the effects of perimenopausal hormonal fluctuations on AD pathophysiology. Using a VCD-induced accelerated ovarian failure model in young female C57BL/6 J and 3xTg mice, we simulated a perimenopausal state with hormonal changes characterised by elevated oestradiol levels and reduced progesterone levels. Supporting human brain transcriptomic and metabolomic data from the ROSMAP study revealed that impaired oestrogen-related receptor alpha (ERRα) function was a key driver of female sex-biased vulnerability. In female mice, progesterone-guided oestrogen receptor signalling maintained ERRα activity by regulating neuronal cholesterol homoeostasis and the TCA cycle. Hormonal imbalances disrupted this mechanism, triggering an aspartate-driven "minicycle," which increased glutamate release, neuronal excitability, ATP depletion, and energy crisis susceptibility. This study demonstrates how perimenopausal hormonal imbalances exacerbate AD risk via ERRα dysfunction, linking neuronal cholesterol and energy homeostasis to disease vulnerability.
    DOI:  https://doi.org/10.1038/s41467-025-66726-4
  30. Curr Issues Mol Biol. 2025 Nov 17. pii: 955. [Epub ahead of print]47(11):
      Cardiovascular diseases (CVDs) remain a leading cause of mortality worldwide. According to the WHO, every year, there is an increase in the rate of death globally due to CVDs, stroke, and myocardial infarction. Several risk factors contribute to the development of CVDs, one of which is hypoxia, defined as a reduction in oxygen levels. This major stressor affects aerobic species and plays a crucial role in the development of cardiovascular disease. Research has uncovered the "hypoxia-inducible factors (HIFs) switch" and investigated the onset, progression, acute and chronic effects, and adaptations of hypoxia, particularly at high altitudes. The hypoxia signalling pathways are closely linked to natural rhythms such as the circadian rhythm and hibernation. In addition to genetic and evolutionary factors, epigenetics also plays an important role in postnatal cardiovascular responses to hypoxia. Oxidized LDL-C initiates atherosclerosis amidst oxidative stress, inflammation, endothelial dysfunction, and vascular remodelling in CVD pathogenesis. Anti-inflammatory and antioxidant biomarkers are needed to identify individuals at risk of cardiovascular events and enhance risk prediction. Among these, C-reactive protein (CRP) is a recognized marker of vascular inflammation in coronary arteries. Elevated pro-atherogenic oxidized LDL (oxLDL) expression serves as an antioxidant marker, predicting coronary heart disease in apparently healthy men. Natural antioxidants and anti-inflammatory molecules protect the heart by reducing oxidative stress, enhancing vasodilation, and improving endothelial function. For instance, the flavonoid quercetin exerts antioxidant and anti-inflammatory effects primarily by activating the Nrf2/HO-1 signaling pathway, thereby enhancing cellular antioxidant defense and reducing reactive oxygen species. Carotenoids, such as astaxanthin, exhibit potent antioxidant activity by scavenging free radicals and preserving mitochondrial integrity. The alkaloid berberine mediates cardiovascular benefits through activation of AMO-activated protein kinase (AMPK) and inhibition of nuclear factor kappa B [NF-kB] signalling, improving lipid metabolism and suppressing inflammatory cytokines. Emerging evidence highlights microRNAs (miRNAs) as potential regulators of oxidative stress via endothelial nitric oxide synthase (eNOS) and silent mating-type information regulation 2 homolog (SIRT1). While the exact mechanisms remain unclear, their benefits are likely to include antioxidant and anti-inflammatory effects, notably reducing the susceptibility of low-density lipoproteins to oxidation. Additionally, the interactions between organs under hypoxia signalling underscore the need for a comprehensive regulatory framework that can support the identification of therapeutic targets, advance clinical research, and enhance treatments, including FDA-approved drugs and those in clinical trials. Promising natural products, including polysaccharides, alkaloids, saponins, flavonoids, and peptides, as well as traditional Indian medicines, have demonstrated anti-hypoxic properties. Their mechanisms of action include increasing haemoglobin, glycogen, and ATP levels, reducing oxidative stress and lipid peroxidation, preserving mitochondrial function, and regulating genes related to apoptosis. These findings emphasise the importance of anti-hypoxia research for the development of effective therapies to combat this critical health problem. A recent approach to controlling CVDs involves the use of antioxidant and anti-inflammatory therapeutics through low-dose dietary supplementation. Despite their effectiveness at low doses, further research on ROS, antioxidants, and nutrition, supported by large multicentre trials, is needed to optimize this strategy.
    Keywords:  anti-inflammatory; antioxidant; cardiovascular diseases
    DOI:  https://doi.org/10.3390/cimb47110955
  31. Biochimie. 2025 Nov 20. pii: S0300-9084(25)00283-4. [Epub ahead of print]
      The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that is thought to play important roles in aging, oxidative stress, and cellular senescence. We have previously shown that the AhR agonist kynurenine (Kyn), a tryptophan metabolite that increases with age, can induce muscle atrophy in young mice. AhR overexpression can also lead to muscle atrophy and neuromuscular junction degradation. Here we utilized existing GEO data sets from skeletal muscles of aged mice to examine the impact of two longevity-related interventions, calorie restriction (CR) or treatment with the drug rapamycin (RM), on the expression of genes in the Kyn-AhR pathway. Data were examined in four skeletal muscles: soleus, gastrocnemius, tibialis anterior and triceps brachii. Results show that AhR expression increased with age in the triceps but was decreased with CR in the soleus and gastrocnemius. RM treatment did not significantly alter AhR expression in any of the four muscles of aged mice. Three enzymes that convert kynurenine to kynurenic acid in skeletal muscle, Kyat1, Kyat3 and Got2/Kyat4, are known to increase with endurance exercise and all three increased significantly with CR in aged skeletal muscle. In contrast, RM treatment did not increase Kyat1 expression in aged muscle and RM significantly decreased Kyat3 expression levels in muscles from aged mice. Together these data point to kynurenine aminotransferases as mediating some of the positive effects of CR on skeletal muscle with aging, and support prior research suggesting that CR and RM modulate different patterns of muscle-specific gene expression.
    Keywords:  Got2; Kyat1; Kyat3; Sarcopenia; Tryptophan
    DOI:  https://doi.org/10.1016/j.biochi.2025.11.010
  32. Invest Educ Enferm. 2025 Sep;43(3):
       Objective: To evaluate the impact of multidimensional lifestyle interventions on aging outcomes, including cognitive function, physical health, emotional well-being, and longevity.
    Methods: This systematic review included 35 randomized controlled trials from 2014 through 2024 using MeSH terms such as "healthy aging," "nutrition," "physical activity," "mental health," "social connections," and "preventive healthcare." Inclusion was made based on whether studies have explored at least one lifestyle intervention among populations of 50 years and above with a reported outcome related to health and aging.
    Results: 35 RCTs (n ≈ 25 000 participants) were included. Key findings were: Nutrition: The Mediterranean diet, antioxidant-rich foods, and protein intake were associated with cardiovascular benefits (RR = 0.78), reduced cognitive decline (OR = 0.72), and improved muscle mass (SMD = 0.45). Physical Exercise: Aerobic and resistance exercises enhanced cardiovascular fitness (MD in VO2 max = 3.6 mL/kg/min) as well as risk of frailty (RR = 0.67); Mental Health: Cognitive stimulation and mindfulness interventions reduced the risk of dementia (OR = 0.75) as well as stress levels (Standard Mean Difference -SMD = -0.65); Network Social: Friendship support interventions with community involvement attenuated the susceptibility to depression, by 30% (RR = 0.70), with improved life overall satisfaction (SMD = 0.55); Safe Harm Avoidance: Smoking treatment reduced cardiovascular outcome risks (OR = 0.68), though moderate alcoholism was associated with better liver overall function (SMD = -0.38); Sleep: Insomnia was related to a 25% reduced risk of cognitive decline when kept at 7-8 hours (RR = 0.75), and Cognitive Behavioral Therapy for Insomnia was highly effective in enhancing quality of sleep (SMD = 0.74); Preventive Healthcare: Routine checkup reduced the un-diagnosed chronic conditions by 40% (RR = 0.60) and the vaccination programs reduced the influenza-related hospital admissions (OR = 0.58).
    Conclusion: Lifestyle interventions are significant in promoting life expectancy, cognitive performance, and overall well-being. The most comprehensive benefits of delay of age-related decline will be offered by the integration of multiple lifestyle factors, including balanced diets, regular exercise, cognitive engagement, strong social ties, and preventive healthcare.
    Keywords:  cognitive dysfunction; depression; diet, healthy; exercise; healthy aging; life style; mindfulness; quality of life; risk factors; sleep initiation and maintenance disorders
    DOI:  https://doi.org/10.17533/udea.iee.v43n3e06