bims-mistre Biomed News
on Mito stress
Issue of 2025–12–14
24 papers selected by
Ellen Siobhan Mitchell, MitoQ



  1. Metabolism. 2025 Dec 04. pii: S0026-0495(25)00329-4. [Epub ahead of print] 156460
      Heart failure with preserved ejection fraction (HFpEF) is increasingly recognized as an age-predominant syndrome characterized by diastolic dysfunction despite preserved systolic performance. In the aged myocardium, fatty acid oxidation capacity declines, while glycolytic flux increases; however, impaired pyruvate oxidation limits mitochondrial glucose oxidation, resulting in suboptimal ATP yield per oxygen molecule and worsening energetic inefficiency. Mitochondrial deficits, marked by reduced biogenesis, NAD+ depletion related to reduced sirtuin activity and consequent hyperacetylation of oxidative enzymes, and impaired electron-transport capacity, further diminish bioenergetic reserve and elevate reactive oxygen species generation. Concurrently, inflammaging and proteostatic collapse promote chronic low-grade inflammation, misfolded protein accumulation, and myocardial fibrosis, collectively contributing to increased ventricular stiffness and progressive HFpEF development. Therapeutic strategies targeting these interconnected pathways show considerable promise. Preclinical studies suggest that interventions such as NAD+ precursor supplementation, mTORC1 inhibition, and β-hydroxybutyrate administration can ameliorate HFpEF-like phenotypes by improving mitochondrial efficiency and reducing inflammation. SGLT2 inhibitors and GLP-1 receptor agonists confer clinically proven benefits in HFpEF, likely via systemic metabolic reprogramming toward more oxygen-efficient substrates and attenuation of inflammation. This review underscores the critical role of aging-associated metabolic and mitochondrial derangements in HFpEF pathogenesis and highlights mechanistically tailored interventions as the next frontier in managing this challenging, age-related syndrome.
    Keywords:  Cardiac aging; Heart failure with preserved ejection fraction; Metabolic remodeling; Mitochondrial dysfunction; Therapeutic targets
    DOI:  https://doi.org/10.1016/j.metabol.2025.156460
  2. Molecules. 2025 Dec 01. pii: 4621. [Epub ahead of print]30(23):
      In vitro and in vivo evidence demonstrates that EGT exerts neuroprotective effects through multiple mechanisms: scavenging reactive oxygen species, suppressing neuroinflammatory cytokines (TNF-α, IL-1β, IL-6), activating Nrf2 antioxidant pathways, and preserving mitochondrial integrity. Low blood EGT levels correlate with cognitive decline and dementia, supporting its role as a conditionally essential micronutrient for healthy aging. Mushroom by-products retain EGT concentrations comparable to commercial fruiting bodies, making them viable sources for dietary supplements and functional foods. Mushroom processing generates substantial residual biomass-including stems, culls, and spent substrate-that represents an underexploited dietary source of ergothioneine (EGT), a naturally occurring antioxidant with exceptional neuroprotective and anti-inflammatory properties. Since humans cannot synthesize EGT endogenously, dietary intake is essential for maintaining neuroprotection against neurodegenerative diseases. This review examines sustainable extraction strategies-including hot-water, ultrasound-assisted, and high-hydrostatic-pressure methods-enabling integration into circular biorefinery systems. Applications in nutraceuticals and pharmaceuticals targeting oxidative stress-related neurodegeneration are highlighted. Despite challenges in standardization and regulatory approval, valorizing mushroom residuals offers a sustainable pathway to increase dietary availability of this neuroprotective antioxidant, supporting both environmental sustainability and therapeutic innovation for neurodegenerative disease prevention.
    Keywords:  anti-inflammatory; circular bioeconomy; ergothioneine; mushroom by-products; neuroprotection; oxidative stress; sustainable extraction
    DOI:  https://doi.org/10.3390/molecules30234621
  3. Brain Behav. 2025 Dec;15(12): e71130
       PURPOSE: Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline, posing a significant challenge to global public health. As a core signaling pathway in the mammalian innate immune system, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a pivotal role in maintaining intracellular homeostasis. This review aims to systematically elucidate the role and therapeutic potential of the cGAS-STING signaling pathway in AD, focusing on its involvement in key pathological processes and its relevance to AD risk factors.
    METHOD: Through literature search, we summarized the molecular mechanisms of the cGAS-STING pathway and its dysregulation in AD, emphasizing the integrated evidence linking cGAS-STING to neuroinflammation, autophagy impairment, and neuronal death, as well as its interactions with aging, obesity, cardiovascular disease, and diabetes.
    FINDINGS: The cGAS-STING pathway is critically involved in AD pathogenesis, contributing to neuroinflammation, defective autophagy, and neuronal loss. Its activation is associated with multiple AD risk factors, suggesting a broad influence on disease progression. Pharmacological inhibition of cGAS-STING shows promise in attenuating these pathological features in preclinical models.
    CONCLUSION: The cGAS-STING signaling pathway plays a central regulatory role in the central nervous system, and its dysregulation promotes neuroinflammation and is closely associated with AD. This pathway forms a vicious cycle by integrating multiple pathological signals, including mitochondrial dysfunction and endoplasmic reticulum stress. Small-molecule inhibitors and natural products targeting this pathway have demonstrated significant efficacy in preclinical studies, providing a basis for developing disease-modifying therapies for AD. Future efforts should focus on multi-target combination strategies (e.g., STING inhibitors co-administered with Aβ/tau drugs) and dynamically deciphering pathway alterations across AD stages to advance personalized treatment approaches.
    Keywords:  STING; alzheimer's disease; cGAS; neuroinflammation
    DOI:  https://doi.org/10.1002/brb3.71130
  4. Nutrients. 2025 Nov 30. pii: 3771. [Epub ahead of print]17(23):
      Background/Objectives: Extracellular vesicles derived from edible plants have emerged as bioactive nanostructures with potential therapeutic and nutraceutical properties and are currently being investigated as natural carriers for the treatment of oxidative stress-induced damage and oxidative stress-related diseases, including neurodegenerative disorders such as Alzheimer's disease (AD). Recent studies suggest that PDEVs exhibit high stability within the gastrointestinal tract and selective tissue-targeting abilities, facilitating the efficient delivery of bioactive molecules. Methods: This study investigates the antioxidant effects of Rheum rhabarbarum-derived EVs by assessing the antioxidant activity through different in vitro assays and their effects on oxidative stress and energy metabolism in the cellular model of Alzheimer's disease. Results: Rhubarb-derived EVs showed measurable antioxidant capacity in chemical assays and were non-toxic under the tested conditions. Treatment reduced intracellular ROS levels and modulated oxidative stress-related proteins, suggesting a potential protective effect against oxidative damage. Moreover, metabolic analysis revealed a decrease in glycolytic activity, indicating a potential restoration of cellular bioenergetic homeostasis. Conclusions: These results provide preliminary evidence supporting the nutraceutical interest of rhubarb-derived EVs in counteracting oxidative stress, while further studies will be needed to confirm their biological relevance and therapeutic potential.
    Keywords:  Alzheimer’s disease (AD); antioxidant activity; drug delivery; oxidative stress; plant-derived extracellular vesicles (PDEVs); reactive oxygen species (ROS)
    DOI:  https://doi.org/10.3390/nu17233771
  5. Cells. 2025 Nov 26. pii: 1861. [Epub ahead of print]14(23):
      Mitochondria are essential for β-cell function, coupling glucose metabolism to ATP production and insulin secretion. In diabetes, β-cell mitochondrial dysfunction arises from oxidative stress, impaired quality control and disrupted dynamics, leading to reduced oxidative phosphorylation, defective insulin release and progressive cell loss. Key transcriptional regulators link genetic susceptibility to mitochondrial dysfunction in both type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). These disruptions impair mitophagy, mitochondrial translation and redox homeostasis. Therapeutic strategies that restore mitochondrial function, including mitophagy enhancers, mitochondrial antioxidants, and transcriptional regulators, have shown potential in preserving β-cell integrity. As mitochondrial failure precedes β-cell loss, targeting mitochondrial pathways may represent a critical approach to modifying diabetes progression.
    Keywords:  diabetes; mitochondria; mitochondrial dysfunction; mitophagy; β-cell
    DOI:  https://doi.org/10.3390/cells14231861
  6. Synapse. 2026 Jan;80(1): e70036
      In recent years, global life expectancy has risen significantly, leading to a marked increase in the elderly population. Aging disproportionately affects the brain, resulting in cognitive decline and increased susceptibility to neurodegenerative diseases. This vulnerability is largely due to the high metabolic activity of neural tissue and its sensitivity to reactive oxygen species (ROS). Consequently, oxidative and inflammatory processes are key contributors to cellular damage and age-related cognitive deficits. These oxidative processes may act as primary drivers of neuronal damage during aging. Recent evidence suggests that phenolic compounds may play a crucial role in neuroprotective strategies. Among these, gallic acid (GA) has garnered attention due to its potent antioxidant and anti-inflammatory properties, which enhance neuronal resilience in animal models. However, the effects of GA on hippocampal neurodegeneration during aging remain unclear. In this study, we evaluated the impact of GA on learning, memory, redox balance, neuroinflammation, apoptosis, and synaptic plasticity in the hippocampus (Hp) of aged rats. Male rats aged 18 months received a daily dose of GA (20 mg/kg) for 60 days. GA treatment significantly improved short- and long-term recognition memory. In the Hp, ROS, tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and lipid peroxidation were reduced, while superoxide dismutase (SOD) and catalase (CAT) activities increased. GA administration also decreased caspase-3 expression. Collectively, these findings indicate that GA enhances antioxidant and anti-inflammatory defenses, thereby mitigating hippocampal damage and supporting its potential as a promising therapeutic strategy to prevent or reduce neurodegenerative disorders associated with aging.
    Keywords:  brain aging; neuroinflammation; neuroplasticity; oxidative stress; polyphenols; recognition memory
    DOI:  https://doi.org/10.1002/syn.70036
  7. Nat Metab. 2025 Dec 10.
      Metabolic flexibility allows cells to adapt to different fuel sources, which is particularly important for cells with high metabolic demands1-3. In contrast, neurons, which are major energy consumers, are considered to rely essentially on glucose and its derivatives to support their metabolism. Here, using Drosophila melanogaster, we show that memory formed after intensive massed training is dependent on mitochondrial fatty acid (FA) β-oxidation to produce ATP in neurons of the mushroom body (MB), a major integrative centre in insect brains. We identify cortex glia as the provider of lipids to sustain the usage of FAs for this type of memory. Furthermore, we demonstrate that massed training is associated with mitochondria network remodelling in the soma of MB neurons, resulting in increased mitochondrial size. Artificially increasing mitochondria size in adult MB neurons increases ATP production in their soma and, at the behavioural level, strikingly results in improved memory performance after massed training. These findings challenge the prevailing view that neurons are unable to use FAs for energy production, revealing, on the contrary, that in vivo neuronal FA oxidation has an essential role in cognitive function, including memory formation.
    DOI:  https://doi.org/10.1038/s42255-025-01416-5
  8. Eur J Prev Cardiol. 2025 Dec 11. pii: zwaf726. [Epub ahead of print]
       AIMS: This study aims to investigate the role of biological aging pace and quantify its contributions in explaining sex disparities in cardiovascular disease (CVD) risk.
    METHODS AND RESULTS: A population-based study was conducted, including 371 032 participants in the UK Biobank. Two measures of biological aging pace were assessed, including phenotypic age acceleration (PhenoAgeAccel) and telomere length. The chronological age was similar between females [56.1 (8.0) years] and males [56.1 (8.2) years]. Male participants consistently had higher CVD risks than female participants, including any CVD [hazard ratio (HR), 1.66; 95% confidence interval (CI), 1.54-1.80], atrial fibrillation (HR, 1.78; 95% CI, 1.73-1.83), coronary heart disease (HR, 2.11; 95% CI, 2.05-2.18), heart failure (HR, 1.72; 95% CI, 1.65-1.80), and stroke (HR, 1.44; 95% CI, 1.38-1.51). Females had 0.71-2.90 years longer time to CVD than males. Males had a faster biological aging pace of 1.02 (95% CI, 0.99-1.06) years in PhenoAgeAccel than females, with 17.1% explained by unhealthy lifestyles, followed by prevalent chronic diseases (8.6%) and metabolic factors (6.5%). The sex disparities in PhenoAgeAccel significantly explained 65.0% of male-associated any CVD, 64.7% of atrial fibrillation, 68.2% of coronary heart disease, 64.4% of heart failure, and 60.4% of stroke, respectively. Similar findings were observed when using the telomere length for evaluating biological aging pace or controlling for sex hormones.
    CONCLUSION: Our findings reveal that the biological aging pace might partially explain sex disparities in CVD risk, highlighting the importance of monitoring the biological aging pace to address the huge sex gap and promote sex-specific CVD primary prevention.
    Keywords:  Biological aging; Cardiovascular disease; Phenotypic age; Sex disparities; Telomere length
    DOI:  https://doi.org/10.1093/eurjpc/zwaf726
  9. Sci Rep. 2025 Dec 10. 15(1): 43527
      This study aimed to evaluate the anti-aging effects of L-theanine in a D-galactose-induced aging model in rats and to elucidate its underlying molecular mechanisms. Aging was induced via subcutaneous administration of D-galactose, while L-theanine was delivered orally for eight weeks. Skin aging was induced in rats by daily subcutaneous injection of D-galactose. L-theanine was administered orally for eight weeks. Skin condition was assessed through by macroscopic observation and histological examination. Enzyme-linked immunosorbent assays were conducted to measure oxidative stress parameters, inflammatory cytokines, and AGEs/RAGE expression levels. L-theanine administration significantly improved skin integrity, maintained epidermal thickness and collagen architecture, administration significantly preserved skin integrity, maintaining epidermal thickness and collagen architecture. It reduced AGEs and receptor for advanced glycation end products (RAGE) expression, enhanced antioxidant enzyme activities (SOD, CAT, GSH-Px, T-AOC), and downregulated reduced the accumulation of AGEs and the expression of RAGE, increased activities of antioxidant enzymes (SOD, CAT, GSH-Px, T-AOC), and decreased levels of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6) in a dose-dependent manner. L-theanine effectively delays delayed skin aging in rats by inhibiting the AGEs-RAGE signaling cascade signaling pathway, thereby modulating attenuating oxidative stress and inflammatory responses.
    Keywords:  Advanced glycation end products; Antioxidant defense; Inflammation; L-theanine; NF-κB; Skin aging
    DOI:  https://doi.org/10.1038/s41598-025-27566-w
  10. Int J Gynaecol Obstet. 2025 Dec 09.
      Menopause represents a significant physiologic transition in a woman's life, often accompanied by psychological challenges, including depression, anxiety, and cognitive decline. The postmenopausal decline in estrogen levels has been implicated in increased risk of neuropsychiatric disorders and cognitive impairment, with studies highlighting estrogen's neuroprotective and regenerative effects. While hormone replacement therapy (HRT) has been widely used for the relief of vasomotor and physical symptoms of menopause, a definitive consensus on its efficacy in the mitigation and management of mental health symptoms is lacking. This narrative review examines the role of HRT in managing menopausal mental health, evaluating the effects of estrogen-only therapy, combined estrogen-progesterone therapy, and alternative hormonal treatments on mood disorders, anxiety, and cognitive function. Findings suggest that estrogen therapy, in particular, transdermal 17β-estradiol, may have antidepressant effects in perimenopausal women, although its efficacy in postmenopausal depression is less clear. Combined estrogen-progesterone hormone therapy, while possibly beneficial for mood regulation, may attenuate some cognitive benefits associated with estrogen alone. Additionally, the effects of progesterone remain ambiguous, with synthetic progestins potentially counteracting estrogen's neuroprotective effects. Emerging evidence suggests a possible role for testosterone and selective estrogen receptor modulators in cognitive and mood stabilization, though further research is warranted. Cognitive outcomes associated with HRT appear to be time-dependent, aligning with the "timing hypothesis", which posits that early initiation of HRT post-menopause may confer neuroprotective benefits, whereas late-life initiation could contribute to an increased risk of dementia. The heterogeneity of findings emphasizes the importance of individualized, carefully considered treatment approaches, underscoring the need for further large-scale, robust trials to further inform the use of HRT, as appropriate, in menopausal mental health management.
    Keywords:  anxiety; cognitive function; depression; estrogen therapy; hormone replacement therapy; menopause; mental health; neuroprotection
    DOI:  https://doi.org/10.1002/ijgo.70728
  11. Commun Biol. 2025 Dec 11. 8(1): 1759
      Hypermetabolism, a futile cycle of energy production and consumption, has been proposed as an adaptative response to deficiencies in mitochondrial oxidative phosphorylation. However, the cellular costs of hypermetabolism remain largely unknown. Here we studied the consequences of hypermetabolism in human motor neurons harboring a heteroplasmic mutation in MT-ATP6, which impairs ATP synthase assembly. Respirometry, metabolomics, and proteomics analyses of the motor neurons showed that elevated ATP production rates were accompanied with increased demand for acetyl-Coenzyme A (acetyl-CoA) and depleted pantothenate (vitamin B5), and the proteome was remodeled to support the metabolic adaptation. Mitochondrial membrane potential and coupling efficiency remained stable, and the therapeutic agent avanafil did not affect metabolite levels. However, a redistribution of acetyl-CoA usage resulted in metabolic trade-offs, including reduced histone acetylation and altered maintenance of the neurotransmitter acetylcholine, revealing potential vulnerabilities in motor neurons. These findings advance the understanding of cellular metabolic consequences imposed by hypermetabolic conditions.
    DOI:  https://doi.org/10.1038/s42003-025-09149-7
  12. Nutr Res Pract. 2025 Dec;19(6): 867-879
       BACKGROUND/OBJECTIVES: Aging is often accompanied by chronic oxidative stress, contributing to the progression of metabolic disorders. A previous study reported that supplementation with collagen tripeptide (CTP) inhibits fatty liver and lipid hypertrophy, but the mechanism is unclear.
    MATERIALS/METHODS: This study examined the effects of 12-week CTP supplementation on oxidative stress, insulin resistance, and adenosine monophosphate-activated protein kinase (AMPK) α activation, key factors in metabolic disorders in aged mice. Thirty-eight male C57BL/6J mice (58 weeks old) were assigned to 4 groups: aging control, 0.03% Gly-Pro-Hyp, 0.9% CTP, and 0.9% granulated collagen tripeptide (gCTP) supplementation. A group of young mice (YC, n = 8) served as the positive control.
    RESULTS: The results showed that CTP and gCTP enhance antioxidant capacity by reducing thiobarbituric acid reactive substances and H2O2 and improving the antioxidant enzyme activities, including superoxide dismutase and glutathione peroxidase. CTP also improved insulin sensitivity by increasing the expression of insulin receptors and glucose transporters in the liver and white adipose tissue. Furthermore, CTP and gCTP improved energy metabolism and mitochondrial function via AMPKα activation.
    CONCLUSION: CTP supplementation is a promising intervention to mitigate comorbidities in older adults, such as obesity and fatty liver disease. CTP and gCTP supplementation may serve as functional ingredients to improve energy metabolism and mitochondrial function. Their potential to reduce oxidative stress, improve insulin resistance, and enhance AMPKα expression supports their use in managing obesity and fatty liver disease, particularly in older adults.
    Keywords:  Collagen; aging; insulin resistance; mitochondria; oxidative stress
    DOI:  https://doi.org/10.4162/nrp.2025.19.6.867
  13. Mol Neurobiol. 2025 Dec 08. 63(1): 270
      Aging has emerged as a prominent area of academic inquiry. Brain aging is a complex physiological process characterized by features such as enhanced apoptosis, oxidative stress, neuroinflammation, mitochondrial dysfunction, and impaired autophagy. Currently, effective preventative or therapeutic approaches for age-related neurodegenerative disorders remain elusive. Ectoine, a naturally occurring compatible solute, possesses diverse applications in biological engineering, cosmetics, medicine, and the food industry. Ectoine is reported to exhibit anti-inflammatory, antioxidant, and anti-apoptotic properties, making it a potential anti-aging agent. Consequently, the present study investigated the potential neuroprotective effects of Ectoine against D-galactose (D-gal)-induced brain aging. Accelerated aging was induced by subcutaneous injection of D-gal. Rats were subsequently divided into a control group, an aged group, and Ectoine-supplemented groups, receiving daily doses of 10, 20, and 40 mg/kg, respectively. Our findings revealed that Ectoine effectively and dose-dependently protected against D-gal-induced brain aging by inhibiting oxidative stress, enhancing the antioxidant system, decreasing neuroinflammation, restoring autophagy and mitochondrial homeostasis, and inhibiting apoptosis. Furthermore, Ectoine significantly restored the expression of miR-124 and its target genes; however, this effect is correlative and warrants further mechanistic validation. Additionally, while Ectoine's neuroprotective effects were observed at the tissue level, its cell-type specificity remains to be determined. These findings suggest that Ectoine may exert multi-pathway neuroprotective effects in brain aging. However, the current data are exploratory and warrant further validation to define causality and translational applicability.
    Keywords:  Aging; Apoptosis; Autophagy; D-galactose; Ectoine; MiR-124; Mitochondrial homeostasis; Neuroinflammation; Neuroprotection; Oxidative stress
    DOI:  https://doi.org/10.1007/s12035-025-05451-x
  14. Am J Psychiatry. 2025 Dec 10. appiajp20241115
       OBJECTIVE: Studies evaluating the effect of anti-inflammatory treatment on depressive symptom severity and anhedonia in depressed individuals report mixed results. In this preregistered systematic review and meta-analysis, the authors evaluated whether anti-inflammatory treatments, compared to placebo, reduce anhedonia and depressive symptom severity in depressed individuals with an inflammatory phenotype.
    METHODS: The authors included randomized controlled trials of pharmacological anti-inflammatory treatments that assessed anhedonia or depressive symptom severity and recruited depressed individuals with an inflammatory phenotype or measured baseline inflammatory biomarkers that permitted post hoc analysis. A search was conducted in February 2025 of MEDLINE, Embase, Web of Science Core Collection, Cochrane Central Register of Controlled Trials, ClinicalTrials.gov, and PsycINFO. Multiple reviewers independently applied criteria, and discrepancies were resolved via consensus. Two reviewers independently extracted data and cross-checked for errors.
    RESULTS: In randomized controlled trials (k=11) using an established cutoff for elevated inflammation (C-reactive protein ≥2 mg/L), both anhedonia (Hedges' g=0.40, 95% CI=0.08, 0.71) and depressive symptoms (Hedges' g=0.35, 95% CI=0.05, 0.64) were reduced, but no differences in treatment response (relative risk=1.28, 95% CI=0.997, 1.64) or remission rates (relative risk=1.18, 95% CI=0.71, 1.95) were observed. Results did not vary by clinical, interventional, or demographic characteristics.
    CONCLUSIONS: Anti-inflammatory treatments may be safe and effective at reducing depressive symptoms and anhedonia in depressed individuals with heightened inflammation. Not accounting for inflammatory status may help explain prior mixed findings.
    Keywords:  Depressive Disorders; Inflammation
    DOI:  https://doi.org/10.1176/appi.ajp.20241115
  15. Front Aging. 2025 ;6 1706117
       Introduction: Menopause triggers declining estradiol, causing vasomotor symptoms, bone loss, and urogenital changes. Despite hormone therapy's effectiveness, safety concerns drive 40%-50% of Western women toward natural alternatives.
    Aim: To evaluate evidence supporting non-pharmacological interventions that modulate endogenous estrogen activity in postmenopausal women.
    Methods: 48 high-quality publications (2015-2025) examining dietary interventions, micronutrient supplementation, gut microbiome modulation, lifestyle modifications, and botanical remedies for menopausal symptoms were analyzed and included in this review. Selection criteria included randomized controlled trials, systematic reviews, meta-analyses, and cohort studies specifically addressing natural interventions in perimenopausal and postmenopausal women.
    Results: Multiple natural approaches demonstrated clinically meaningful effects. Dietary phytoestrogens (50-80 mg/day isoflavones) reduced severe hot flashes by up to 92%, improved metabolic parameters, and were confirmed safe for reproductive tissues. Flaxseed lignans reduced perimenopausal symptoms. Combined vitamin E and omega-3 lowered hot flush intensity, while vitamin E alone showed estrogenic receptor activation. Curcumin (500 mg/day) reduced hot flashes after 4 weeks and improved metabolic profiles. Probiotics containing L. brevis KABP052 increased circulating estrogens by up to 26% over 12 weeks. Stress reduction interventions improved quality of life, and cognitive behavioral therapy reduced insomnia severity. Botanicals including black cohosh, red clover, and rhapontic rhubarb reduced vasomotor symptoms, while resveratrol (75 mg twice daily) significantly improved bone mineral density over 12 months. Research gaps remain regarding dosing and genetic variability.
    Conclusion: A multi-domain approach incorporating phytoestrogen-rich foods, targeted micronutrients, gut microbiome optimization, and regular exercise provides evidence-based options for managing estrogen decline. While these approaches cannot fully replace hormone therapy, they provide valuable alternatives for women with contraindications or preferences against pharmaceutical intervention. Future research should focus on personalized approaches incorporating genetic profiling.
    Keywords:  dietary supplements; estrogens; gastrointestinal microbiome; life style; micronutrients; phytoestrogens; postmenopause
    DOI:  https://doi.org/10.3389/fragi.2025.1706117
  16. Geroscience. 2025 Dec 13.
      Women experience more pronounced lipidomic changes with aging than men, which may contribute to the higher rates of Alzheimer's disease seen in postmenopausal women. Our earlier findings showed that transplantation of young ovarian somatic tissues or cells produced positive health-enhancing results in postreproductive females. In the current experiments, we looked to find key health-enhancing ovarian cells and pathways involved in this phenomenon. We conducted physiological and molecular analysis on animals/samples from old, postreproductive mice that received young ovarian tissue/cell transplants. Our analysis revealed a loss with age and a restoration with ovarian tissue/cell exposure, of serum biomarkers of lipid signaling and histological and behavioral markers of cognitive function. We further found, with single-cell transcriptomics and Raman spectroscopy, two candidate ovarian somatic cell types implicated in the restoration of health through a lipid signaling-based process. These results have identified key factors toward the determination of how germ cell-independent ovarian somatic tissues restore health through regulation of lipid signaling and dementia in postreproductive female mice.
    Keywords:  Alzheimer’s; Apolipoproteins; Health span; Menopause; Ovarian aging
    DOI:  https://doi.org/10.1007/s11357-025-02019-4
  17. Metabolism. 2025 Dec 08. pii: S0026-0495(25)00337-3. [Epub ahead of print] 156467
      Current metabolomic aging clocks inadequately capture individual heterogeneity in biological aging trajectories, constraining their clinical utility. Here, we developed a metabolomic age clock in the UK Biobank (n = 196,790) using a comprehensive panel of 249 plasma metabolites. This framework was trained to predict phenotypic age (PhenoAge), a validated composite biomarker that integrates clinical chemistry across multiple systems, and was evaluated for its utility to predict incident cardiovascular diseases (CVDs) and dementia. We found that this new measure accurately predicted actual PhenoAge (Pearson's r = 0.90) and was significantly associated with the incidence of seven CVDs, including major adverse cardiovascular events, atherosclerotic cardiovascular disease, myocardial infarction, stroke, aortic stenosis, heart failure, and abdominal aortic aneurysm, but not dementia. Furthermore, metabolomic aging was associated with biological, physical, and cognitive age-related phenotypes, comprising telomere length, frailty index, and reaction time. Incorporating the metabolomic age clock with PREVENT (Predicting Risk of CVD Events) risk score modestly improved the performance, as measured by C-statistic and net reclassification index. Genetic analyses revealed 91 genomic loci and 168 genes (e.g., SERPINA1, FADS cluster), with tissue-enrichment analysis highlighting the liver's significant role in metabolic aging. By bridging metabolomic profiles with multisystem aging information, this framework provides a measure of biological aging that is associated with age-related functional status and cardiovascular risk.
    Keywords:  Biological age; Cardiovascular disease; Genetic determinant; Metabolomics; Risk
    DOI:  https://doi.org/10.1016/j.metabol.2025.156467
  18. BMC Sports Sci Med Rehabil. 2025 Dec 09.
       BACKGROUND: Coenzyme Q10 (CoQ10), a vital mitochondrial antioxidant, may enhance cardiovascular recovery and delay fatigue in athletes. This double-blind, randomized, placebo-controlled trial investigated CoQ10's effects on cardiac stress markers and exercise performance in 16 professional soccer players.
    METHODS: Participants received either CoQ10 (n = 8) or placebo (n = 8) for 30 days and completed exhaustive aerobic cycling tests pre- and post-supplementation. Blood samples were collected at six time points to analyze GDF-15, NT-proBNP, and hs-TnT levels.
    RESULTS: In the CoQ10 group, GDF-15 rose post-exercise (PreEx1-PostEx1: +8.84%, p = 0.003; PreEx2-PostEx2: +5.37%, p = 0.001) but declined by 24 h post-Ex1 (- 5.28%, p = 0.003) and 24 h post-Ex2 (-6.62%, p = 0.001) compared to post-Ex1 & post-Ex2, respectively. NT-proBNP increased post-Ex1 (19.66%, p = 0.001), post-Ex2 (12.09%, p = 0.001) but decreased in 24 h-Ex1 (- 20.28%, p = 0.001), 24 h-Ex2 (-20.83%, p = 0.001) compared to post-Ex1&Ex2, respectively. Following a 31.18% increase post-Ex1, a smaller 23.65% increase was observed post-Ex2 in hs-TnT levels, however, decreases of -19.57 and - 32.20% were observed 24 h- postEx1&Ex2 compared to post-Ex1&Ex2, (p = 0.001 for all). Time to fatigue (TTF) improved post-supplementation (- 0.5%, p = 0.002), whereas the placebo showed no change (- 0.08%, p = 0.793).
    CONCLUSIONS: CoQ10 supplementation significantly reduced post-exercise cardiac stress markers (GDF-15, NT-proBNP, hs-TnT) and improved TTF in athletes. These findings suggest potential associations with reduced cardiac stress responses to exercise, but further studies are required to confirm the clinical significance and long-term effects of CoQ10 supplementation on cardiovascular health in athletes.
    TRIAL REGISTRATION: not applicable.
    Keywords:  Aerobic exercise and soccer; GDF-15; Hs-TnT; NT-proBNP; Supplement CoQ10
    DOI:  https://doi.org/10.1186/s13102-025-01456-0
  19. EClinicalMedicine. 2025 Nov;89 103633
       Background: Long-COVID often involves cognitive difficulties, immune dysregulation, and mitochondrial dysfunction. Studies suggest nicotinamide adenine dinucleotide (NAD+) precursors like nicotinamide riboside (NR) may reduce inflammation and support mitochondrial and neurological function. This double-blind, placebo (PBO)-controlled clinical trial with a placebo lead-in phase evaluated the effects of NR (2000 mg/day) on NAD+ and changes in cognitive and long-COVID symptoms.
    Methods: This was a 24-week, double-blind, placebo-controlled trial at a single center in Boston, USA, between August 2021 and September 2023. 58 community-dwelling participants with long-COVID were randomized 2:1 to the NR-NR group (NR for 20 weeks) or the PBO-NR group (PBO for 10 weeks, followed by NR for 10 weeks). The primary outcome was cognition, assessed using the Everyday Cognition scale (ECog), Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), and Trail Making Test-B (TMT-B). Secondary outcomes included the Fatigue Severity Scale (FSS), Beck Depression Inventory (BDI), Beck Anxiety Inventory (BAI), and Pittsburgh Sleep Quality Index. We conducted a mixed model for repeated measures to compare groups, then post-hoc and unadjusted for multiplicity, combined both groups to explore changes from baseline after 10 weeks of NR. This trial was registered with ClinicalTrials.gov (NCT04809974) in 2021.
    Findings: 37 participants (64%) were assigned to NR-NR, and 21 participants (36%) to PBO-NR. There was a 32.4% and 51.4% dropout in the NR-NR group at 10 weeks and 20 weeks, respectively, vs. 14.3% dropout at each timepoint in the PBO-NR group. In the NR-NR group, NAD+ levels increased by 2.6- to 3.1-fold after 5-10 weeks of supplementation, respectively, and remained elevated at 20 weeks. In the PBO-NR group, NAD+ levels remained close to baseline (0.93- to 1.0-fold change, 95% CI: 0.5-1.4) during the initial 5 and 10 weeks of PBO. After switching to NR, levels rose to a 2.6-fold and 2.1-fold increase after 5 and 10 weeks of NR, respectively. No significant between-group differences were observed for cognitive outcomes (ECog, RBANS, TMT-B; p-values = 0.47-0.74). There were no significant differences in fatigue severity (p = 0.59), sleep quality p = 0.69), and symptoms of anxiety (p = 0.84) or depression (p = 0.20) between PBO and NR groups. In post-hoc exploratory analysis, examining within-group changes during 5 and 10 weeks of NR intake by grouping all participants during the first 10 weeks of the NR phase, there were significant differences from baseline after 10 weeks of NR in executive functioning, fatigue severity, sleep quality, and symptoms of depression (compared with no significant changes in TMT-B, FSS, PSQI, BAI, or BDI scores during the PBO phase). One serious adverse event was reported, deemed unrelated to the study drug or trial.
    Interpretation: In long-COVID, NR increased NAD+ within 5 weeks but did not significantly improve cognition, fatigue, sleep, or mood vs. PBO. Exploratory analyses suggested within-group benefits after 10 weeks of NR, supporting the need for larger trials.
    Funding: This work was supported by Niagen Bioscience, the MGH McCance Center for Brain Health, Lavine Brain Health Innovation Fund, MGH ECOR CDI Physician-Scientist Development Award, and the Alzheimer's Association (grant no. AARGD-23-114103).
    Keywords:  Depression; Fatigue; Nicotinamide adenine dinucleotide; PASC; SARS-CoV-2; Sleep
    DOI:  https://doi.org/10.1016/j.eclinm.2025.103633
  20. Theranostics. 2026 ;16(4): 1701-1719
      Aims: Type 2 diabetes mellitus (T2DM) significantly elevates the likelihood of atrial fibrillation (AF); However, the precise mechanisms remain incompletely elucidated. Mitochondrial dysfunction is a hallmark of diabetic cardiomyopathy, and recent evidence suggests that activation of the cGAS-STING signaling pathway may contribute to metabolic inflammation in the atria. This study aims to investigate the role of mitochondrial DNA (mtDNA)-mediated cGAS-STING activation in promoting diabetes-associated atrial fibrillation (AF) through cardiomyocyte-macrophage crosstalk. Methods and results: Using a high-fat diet combined with streptozotocin through intraperitoneal injection, we induced a diabetic mouse model. We observed increased AF inducibility, oxidative stress, and mitochondrial ultrastructural abnormalities, along with elevated expression of STING pathway components and pro-inflammatory cytokines in atrial tissue. RNA sequencing and histological analyses confirmed dysregulation of mitochondrial quality control (MQC), including impaired mitophagy, imbalance in fusion and fission, and reduced mitochondrial biogenesis. In vitro, HL-1 atrial cardiomyocytes exposed to high glucose and palmitic acid showed excessive production of mtROS and cytosolic release of mitochondrial DNA (mtDNA), which in turn triggered cGAS-STING activation. A transwell co-culture system revealed that cardiomyocyte-derived mtDNA was engulfed by RAW 264.7 macrophages, promoting M1 polarization of macrophages and further amplifying inflammatory signaling. Importantly, pharmacological intervention with the mitochondrial antioxidant mito-TEMPO or cardiomyocyte-specific STING knockdown suppressed inflammatory responses, reversed atrial remodeling, and reduced AF susceptibility. Notably, STING overexpression sustained inflammatory pathways independently of suppressing oxidative stress, highlighting cGAS-STING signaling as a downstream effector of mitochondrial damage. Conclusion: Impairment of mitochondrial quality control promotes atrial inflammation and remodeling in diabetes through mtDNA-induced cGAS-STING activation and cardiomyocyte-macrophage communication. Targeting this pathway may offer a novel strategy for AF management in metabolically compromised hearts.
    Keywords:  Atrial fibrillation; Atrial myocytes-macrophage crosstalk.; Mitochondrial quality control; Type 2 diabetes; cGAS-STING signaling pathway
    DOI:  https://doi.org/10.7150/thno.124140
  21. Front Pharmacol. 2025 ;16 1636538
       Background: Curcumin is a natural polyphenolic compound that originates from turmeric (Curcuma longa L., Linnaeus, Zingiberaceae), a traditional medicinal herb. It is widely recognized for its strong antioxidant properties.
    Objective: This comprehensive review aims to delineate the recent progress in comprehending the role of curcumin in modulating oxidative stress and exerting an anti-fibrotic effect, with a particular focus on liver, renal, myocardial, and pulmonary fibrosis.
    Methods: A systematic review of the literature was conducted via the PubMed, Web of Science, Google Scholar, and China National Knowledge Infrastructure, covering 2000 until 2024. A systematic review identified studies examining curcumin's regulation of oxidative stress pathways in therapeutic strategies for multiple fibrotic disorders, which were analyzed to synthesize current evidence.
    Results: In recent years, the application of curcumin for the clinical management of fibrotic diseases in a variety of clinical applications has been extensively investigated. Accumulating evidence suggests that curcumin can exert antifibrotic effects by ameliorating oxidative stress through the modulation of various signaling pathways such as regulating reactive oxygen species (ROS), nuclear factor erythroid-2-related factor 2 (NRF2), peroxisome proliferator-activated receptors (PPAR), transforming growth factor- β1 (TGF-β1). In this review, we investigate the pharmacokinetics of curcumin, the relationship between oxidative stress and the pathogenesis of fibrosis, and summarize the related studies of curcumin in the treatment of fibrotic diseases by regulating oxidative stress.
    Conclusion: This comprehensive review elucidates curcumin's antifibrotic potential and explores its translational applications in developing novel therapeutic strategies to combat fibrotic pathologies, supported by mechanistic evidence that informs safer, more effective treatment paradigms.
    Keywords:  anti-fibrosis effects; curcuma; reactive nitrogen species; reactive oxygen species; signaling pathways; traditional chinese medicine
    DOI:  https://doi.org/10.3389/fphar.2025.1636538
  22. Nutr Rev. 2025 Dec 06. pii: nuaf206. [Epub ahead of print]
       CONTEXT: A dietary regimen that includes a fasting period of 16 hours coupled with a daily time-restricted eating (TRE) period of 8 hours (16/8 TRE) is gaining popularity; however, its effect on glycolipid metabolic markers remains unclear.
    OBJECTIVE: In this systematic review and meta-analysis of reported studies we aimed to examine the effects on glycolipid metabolism of the 16/8 TRE dietary regimen compared with a control diet in adult study participants.
    DATA SOURCES: We searched the PubMed, MEDLINE, Cochrane Library, Scopus, and Web of Science databases for relevant articles published from database inception until January 15, 2025.
    DATA EXTRACTION: We extracted publication details, including data on study participant characteristics, intervention durations, and outcomes.
    DATA ANALYSIS: All analyses were conducted using a random-effects model with the standardized mean difference (SMD). Subgroup analyses were performed for physical activity (PA) level, participant sex, and intervention duration.
    RESULTS: In total, 23 randomized controlled trials (RCTs)with a total of 1280 participants were included in this analysis. The 16/8 TRE intervention resulted in a slight reduction in levels of fasting glucose (SMD, -0.25; 95% CI, -0.42 to -0.08; P = .004), homeostasis model assessment for insulin resistance (HOMA-IR) (SMD, -0.16; 95% CI, -0.29 to -0.02; P = .03), and insulin levels (SMD, -0.22; 95% CI, -0.43 to -0.01; P = .04). Regarding lipid profiles, 16/8 TRE had a modest effect on high-density lipoprotein cholesterol (HDL-C) (SMD, 0.15; 95% CI: 0.01-0.29; P = .04). No significant effects were found for other markers compared to the control diet. Subgroup analysis suggested the presence of sex differences in triglycerides (TGs) and low-density lipoprotein cholesterol (LDL-C), with significant improvements in TGs (SMD, -0.52; 95% CI, -1.04 to -0.01; P = .05), and LDL-C (SMD, -0.41; 95% CI, -0.75 to -0.07; P = .02) found in studies with only male participants. Additionally, participants with higher PA levels demonstrated greater improvements in LDL-C (SMD, -0.43; 95% CI, -0.74 to -0.12; P = .007). Studies with durations exceeding 6 months showed a significant improvement in HbA1c (SMD, -0.31; 95% CI, -0.56 to -0.05; P = .02).
    CONCLUSION: Study participants who followed the 16/8 TRE showed an overall improvement in fasting glucose, HOMA-IR, insulin, and HDL-C levels compared to participants who followed the control diet. However, the effects of the 16/8 TRE in individuals were likely to have been moderated by participant sex, PA level, and intervention duration.
    SYSTEMATIC REVIEW REGISTRATION: PROSPERO registration No. CRD42025645789.
    Keywords:  intermittent fasting; meta-analysis; metabolic health; time-restricted eating
    DOI:  https://doi.org/10.1093/nutrit/nuaf206
  23. Brain Behav Immun. 2025 Dec 09. pii: S0889-1591(25)00462-3. [Epub ahead of print] 106220
      Western-style diets, high in saturated fats and refined carbohydrates and low in dietary fiber, are strongly linked to cognitive decline, particularly in aging. However, the specific macronutrient contributions and mechanisms underlying these effects remain unclear. Here, we investigated how short-term exposure to refined-ingredient diets (RDs) varying in fat and sugar content impacts memory, mitochondrial function, and metabolic signaling in young adult and aged male rats. A key finding was that amygdala-dependent memory was broadly impaired in aged rats across all RDs, regardless of fat or sugar content, suggesting a unique vulnerability of the aging amygdala to refined dietary ingredients. In contrast, hippocampal-dependent memory impairments were observed only in aged rats fed a high-fat, low-sugar RD. Functional mitochondrial assays revealed significant RD-induced reductions in oxygen consumption in amygdalar and hippocampal mitochondria isolated from aged rats. Cell-type-specific analyses identified aged microglia as particularly susceptible, showing widespread suppression of mitochondrial respiration with limited metabolic flexibility. Astrocytes and synaptic mitochondria exhibited more region- and age-specific effects. All RDs lacked dietary fiber, and consistent with prior findings, butyrate, a microbial-derived short-chain fatty acid, was rapidly and robustly depleted in both gut and circulation, especially in aged animals. Proteomic and phosphoproteomic analyses identified diet-induced disruptions in mitochondrial proteins and synaptic signaling pathways, including complex I subunits and glutamate receptor signaling. Together, these findings reveal that the aged amygdala is especially sensitive to refined diet exposure and highlight microbial, metabolic, and inflammatory pathways that may underlie diet-induced cognitive decline.
    Keywords:  Diet-induced memory deficits; Mitochondrial function; Proteomics; Refined diets; Short-chain fatty acids
    DOI:  https://doi.org/10.1016/j.bbi.2025.106220
  24. Endocr Regul. 2025 Jan 01. 59(1): 244-254
      Objective. Cardiovascular complications are highly prevalent in type 2 diabetes mellitus (T2DM) driven by obesity, dyslipidemia, hypertension, and hypercoagulability associated with insulin resistance. The purpose of this study was to elucidate the effects of combined treatment with acetyl-L-carnitine (ALC), alpha-lipoic acid (ALA), and nicotinamide (NAm) on diabetes-induced metabolic, hemostatic, and heart abnormalities. Methods. Male non-linear Wistar rats were fed with a high-calorie diet for 2 months followed by a single low-dose streptozotocin injection to induce T2DM. Two weeks later, the diabetic rats received ALC (100 mg/kg), ALA (50 mg/kg), and NAm (100 mg/kg) for 2 weeks in separate daily injections. Fasting blood glucose, glycated hemoglobin (HbA1c), and hemostatic parameters: fibrinogen, protein C, factor X, plasminogen activator inhibitor-1 (PAI-1), were measured. The NAD+ content and NAD+/NADH ratio were assessed in the heart tissue. Results. After 12 weeks, blood glucose and HbA1c levels in diabetic rats were 1.8-fold and 2-fold higher, respectively. Diabetes increased fibrinogen (1.5-fold) and PAI-1 (1.7-fold) levels, caused the appearance of soluble fibrin monomers complexes, while protein C and factor X levels were decreased by 18% and 19%, respectively, indicating hypercoagulability and impaired fibrinolysis. In diabetic rats, the cardiac NAD+ level was reduced by 48%. The NAD+/NADH ratio decreased by 2-fold. Combined treatment lowered the glucose levels by 1.3-fold and HbA1c by 1.7-fold and improved the NAD+ metabolism and partially corrected the hemostatic abnormalities. Conclusion. Co-treatment with ALC, ALA, and NAm improved the glycemic control, partially restored the cardiac NAD+ metabolism and reduced the hemostatic abnormalities in T2DM suggesting their potential as a safe adjunct therapy for diabetes-associated cardiovascular complications.
    Keywords:  NAD+; NAD+/NADH ratio; coagulation; heart; mitochondria-targeted compounds; type 2 diabetes mellitus
    DOI:  https://doi.org/10.2478/enr-2025-0028