bims-mistre Biomed News
on Mito stress
Issue of 2025–11–16
fifteen papers selected by
Ellen Siobhan Mitchell, MitoQ



  1. Nutrients. 2025 Oct 22. pii: 3316. [Epub ahead of print]17(21):
      Background/Objectives: Atherosclerosis remains a leading cause of morbidity and mortality worldwide, with an urgent need for novel preventive and therapeutic strategies. Sirtuin 1 (SIRT1), an NAD+-dependent deacetylase, has emerged as a central regulator of vascular homeostasis, modulating oxidative stress, inflammation, lipid metabolism, and endothelial function. Increasing evidence highlights that some natural activators of SIRT1 may be interesting in mitigating the development of cardiovascular diseases. Methods: Searching in the main databases PubMed and Scopus, we made a literature revision, including studies from January 2000 to June 2025, of the major natural SIRT1 activators involved in vascular impairment in order to investigate their potential therapeutic use in atherosclerosis. Results: Among them, resveratrol, quercetin, naringenin, and hydroxytyrosol show the strongest evidence in activating SIRT1 and modulating the essential molecular pathways involved in atherosclerotic disease. These findings span from preclinical to clinical studies, with limited randomized clinical trial data for hard cardiovascular outcomes. Conclusions: This review synthesizes current knowledge on natural SIRT1 activators in the context of atherosclerosis, emphasizing their molecular mechanisms and clinical perspectives. The concept of using nutraceuticals-based interventions targeting SIRT1 may pave the way for innovative strategies in cardiovascular diseases.
    Keywords:  SIRT1; atherosclerosis; inflammation; natural compounds; oxidative stress; vascular protection
    DOI:  https://doi.org/10.3390/nu17213316
  2. J Neurochem. 2025 Nov;169(11): e70294
      Alzheimer's disease (AD) is more than just amyloid and tau. While often described as a disease of metabolic dysfunction, AD can more accurately be described as a disorder of metabolic inflexibility that leads to bioenergetic failure. In the healthy brain, neurons, glia, and vascular cells dynamically share and switch between different fuel sources (e.g., glucose, lactate, ketones, and fatty acids) to match functional demand. In AD, this adaptability is progressively lost because cellular metabolism is actively reprogrammed to support neuroinflammatory and disease-associated processes at the cost of neuronal function. Microglia, in particular, upregulate glycolytic metabolism, alter lipid handling, and prioritize immune functions, which actively depletes the brain's energy supply. These adaptations are initially compensatory but ultimately trap the brain in a rigid metabolic program that deprioritizes neuronal support. This metabolic shift unfolds along a biphasic trajectory: early, glia-driven hypermetabolism aligned with inflammation, followed by late-stage brain hypometabolism and energy collapse that leads to neuronal dysfunction. System-level consequences include altered excitability, decreased network connectivity, sleep disruption, and cognitive decline. Critically, these changes feed forward to accelerate AD pathogenesis: glycolytically biased microglia and stressed neurons promote amyloid-β production, tau release, and protein aggregation, adding to metabolic rigidity. Evidence from human neuroimaging studies, brain/cerebral spinal fluid (CSF) multi-omic studies, and preclinical studies demonstrate that shifts in glycolytic flux, tricarboxylic acid cycle (TCA) intermediates, and lipid metabolism parallel amyloid and tau pathology and cognition decline. We hypothesize that these metabolic programs, while initially protective, are chronically maladaptive yet reversible. We propose that restoring metabolic flexibility can mitigate amyloid and tau pathology, neuronal loss, and functional decline. Ongoing preclinical studies and clinical trials are actively exploring metabolism as a therapeutic target in AD. Collectively, these findings define AD as a disorder of metabolic inflexibility, where adaptive shifts in cellular metabolism become pathologically rigid and drive disease progression, while offering a promising target for therapeutic intervention in AD.
    Keywords:  Alzheimer's disease; glia; metabolism
    DOI:  https://doi.org/10.1111/jnc.70294
  3. Front Immunol. 2025 ;16 1682406
      Aging involves a gradual decline in physiological integrity, and rapamycin (RAPA) has demonstrated potential as an anti-aging agent. Nonetheless, its effects on glucose metabolism and immune function may vary based on dosage and administration regimen. This study investigates the impact of intermittent low-dose RAPA on glucose metabolism and immune function in Senescence-Accelerated Mouse Prone 8 (SAMP8) and Senescence-Accelerated Mouse Resistant 1 (SAMR1) mice. Twelve-week-old male SAMP8 and SAMR1 mice were treated with RAPA (0.78 µg/kg) every five days for six months. Glucose uptake, mitochondrial respiratory capacity, spleen and thymus immunophenotype, lymphoproliferation, and cytokine profiles were evaluated. Our findings indicate that RAPA reduced glucose uptake in the bladder and the percentage of FoxP3+ lymphocytes in the spleen of SAMP8 mice, while enhancing mitochondrial respiratory control and ATP production in liver. In SAMR1 mice, RAPA administration led to a decrease in CD3+ thymocytes and splenic lymphoproliferative capacity, while also enhanced mitochondrial performance. Comparisons between Control groups revealed that SAMP8 mice exhibited higher glucose uptake in several tissues, lower lymphocyte populations in spleen and thymus, altered CD4+/CD8+ ratios, and reduced IL-4 expression compared with SAMR1 mice. The findings reinforce the potential of RAPA to modulate aging-related processes, highlighting improvements in mitochondrial function and energy metabolism across strains with different aging processes. However, the immunosuppressive effects of RAPA remain evident, even at low doses administered intermittently, in an age- and strain-specific manner. These findings emphasize the therapeutic potential of RAPA while underscoring the need for customized dosing strategies to balance efficacy and safety. These data highlight mitochondrial metabolic improvements as the primary benefit of intermittent low-dose RAPA and suggest potential clinical relevance for conditions involving compromised mitochondrial energy metabolism.
    Keywords:  SAMP8; SAMR1; immunosenescence; mitochondria; rapamycin
    DOI:  https://doi.org/10.3389/fimmu.2025.1682406
  4. Nutr Metab (Lond). 2025 Nov 13. 22(1): 140
       BACKGROUND: The prevalence of metabolic disorders such as obesity, type 2 diabetes, and dyslipidemia has increased globally. Postbiotics as non-viable microbial products or metabolites, have recently emerged as potential modulators of metabolic health due to their anti-inflammatory and insulin-sensitizing properties. In order to obtain a better viewpoint from them, this study aimed to comprehensively investigate the effects of postbiotics on metabolic health.
    METHODS: This systematic review and meta-analysis, adhering to PRISMA 2020 guidelines, synthesized data from 25 RCTs assessing the effects of postbiotic supplementation on metabolic parameters. Databases including PubMed, Embase, Web of Science, and Scopus were searched up to June 2025. Outcomes included glycemic indices, anthropometric measures, lipid profiles, inflammatory markers, and blood pressure.
    RESULTS: Postbiotic supplementation significantly reduced serum insulin levels (WMD: - 2.76 µU/mL), triglycerides (TG) (-8.46 mg/dL), waist circumference (WC) (-1.47 cm), and C-reactive protein (CRP) (-0.99 mg/L). However, changes in fasting blood glucose (FBG), homeostatic model assessment for insulin resistance (HOMA-IR), HbA1c, other profile lipids, blood pressure as well as weight and body mass index (BMI) were not statistically significant. Subgroup analyses revealed more pronounced benefits in younger participants, bacterial-based formulations, and interventions longer than 8 weeks. Risk of bias was low to moderate, and no major publication bias was detected.
    CONCLUSION: Postbiotic supplementation demonstrates modest but clinically relevant benefits on insulin sensitivity, central adiposity, TG, and systemic inflammation. These effects suggest a promising adjunctive role for postbiotics in metabolic health interventions, though further trials with standardized formulations and longer durations are warranted.
    Keywords:  Inflammation; Insulin resistance; Meta-analysis; Metabolic health; Postbiotics; SCFAs
    DOI:  https://doi.org/10.1186/s12986-025-01037-5
  5. J Nutr Biochem. 2025 Nov 12. pii: S0955-2863(25)00340-7. [Epub ahead of print] 110178
      Pyrroloquinoline quinone (PQQ) is an enzyme belonging to the family of quinone cofactors (or quinoproteins) naturally found in soil and food. PQQ was initially discovered as an essential cofactor for bacterial dehydrogenases and has since been reported to be involved in several biological processes important for mammalian growth and development. Animal studies have demonstrated that insufficient dietary intake of PQQ can lead to notable deficits, including impaired growth and compromised reproductive outcomes. In more recent years, PQQ has been recognized for its neuroprotective effects in several in vitro and in vivo models of brain injury and disease, rendering it an attractive compound to be incorporated into treatment strategies for various neurological disorders. More specifically, PQQ has been reported to enhance mitochondrial function and mitigate inflammatory and oxidative stress responses in the central nervous system (CNS) through the activation of several signaling pathways. Additionally, PQQ has emerged as a promising compound that could be incorporated in treatment strategies for cerebral folate deficiency, a pediatric neurological condition characterized by suboptimal folate levels in the cerebrospinal fluid, leading to developmental delays, epilepsy, and other neurological symptoms. This review will address the biochemical properties, mechanism of action, and physiological roles of PQQ with a specific focus on its antioxidant and anti-inflammatory effects as well as its role in enhancing mitochondrial function. The therapeutic implications of these findings will be discussed, emphasizing PQQ's potential as a novel pharmacological approach for the management of neurological disorders, including its emerging role in cerebral folate deficiency.
    Keywords:  Antioxidant Mechanisms; Cerebral Folate Deficiency; Folate Transport; Mitochondrial Function; Neuroinflammation; Neuroprotection; PQQ
    DOI:  https://doi.org/10.1016/j.jnutbio.2025.110178
  6. Int J Mol Sci. 2025 Oct 29. pii: 10497. [Epub ahead of print]26(21):
      This perspective piece extrapolates knowledge of mitochondriology to the clinical aspects of cardiovascular disease (CVDs) development. The aim was to deepen the understanding of the etiopathogenesis of CVDs by conceptualizing the systemic involvement of mitochondrial dysfunction mechanisms in their follow-up. A theoretical comparison of mitochondrial status and mitochondrial dysfunction across stages of the cardiovascular continuum was performed based on a systematic analysis of the scientific literature data using general scientific, theoretical, and logical methods and normative rules. Conceptual aspects of the involvement of mitochondrial dysfunction (MD) mechanisms at each stage of the CVDs continuum were identified. MD is a dynamic, complex, multifactorial process that is characterized by quantitative and qualitative changes in the mitochondrial pool of human body cells during the development of CVDs. MD is a fundamental participant in the pathogenesis of CVDs, predetermining the nature and features of the clinical manifestation and course of the disease in each patient. MD has distinctive features at each stage of the catamnesis of CVDs and can be classified according to this principle. The development of objective methods for assessing the degree of MD and its classification criteria is a promising task for future scientific research.
    Keywords:  cardiovascular continuum; cardiovascular diseases; mitochondrial dysfunction
    DOI:  https://doi.org/10.3390/ijms262110497
  7. Nutrients. 2025 Oct 25. pii: 3358. [Epub ahead of print]17(21):
      Background/Objectives: Prediabetes markedly increases the risk of progression to type 2 diabetes. While exercise and dietary polyphenols independently enhance metabolic health, their combined and synergistic effects remain unclear. This randomized, single-blind, placebo-controlled trial investigated the synergistic effects of concurrent training and a microencapsulated persimmon-karonda polyphenol formulation on glycemic control and inflammatory outcomes in adults with prediabetes and who are overweight/obese. Methods: Forty-three participants completed the intervention and were assigned to placebo, concurrent training (CBT), supplementation (EATME), or the combined intervention (CBT + EATME) for 8 weeks. Primary outcomes included fasting blood glucose (FBG), glycated hemoglobin (HbA1c), homeostatic model assessment for insulin resistance (HOMA-IR), high-sensitivity C-reactive protein (hs-CRP), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), adiponectin, physical fitness, and quality of life (QoL). Results: All intervention groups (CBT, EATME, and CBT + EATME) showed improvements in glycemic indices, with the greatest reductions in FBG (p < 0.01), HbA1c (p < 0.05), and HOMA-IR (p < 0.01) observed in the CBT + EATME group compared with placebo. All interventions significantly reduced hs-CRP (p < 0.01) and IL-6 (p < 0.01), accompanied by marked increases in adiponectin (p < 0.01), compared with placebo. In the CBT + EATME group, reductions in hs-CRP were positively correlated with improvements in HOMA-IR (r = 0.627, p < 0.05). Both CBT and CBT + EATME improved muscular strength and maximal oxygen consumption (V̇O2max), with the combined intervention producing greater gains in upper- and lower-body strength (p < 0.05), V̇O2max (p < 0.05), and the psychological well-being domain of QoL (p < 0.05) compared with placebo. Conclusions: These findings highlight that combining concurrent training with microencapsulated polyphenol supplementation produced the most consistent improvements across metabolic, inflammatory, and fitness outcomes, supporting this combined approach as an integrated and synergistic strategy to reduce diabetes risk and promote overall health in at-risk adults. The trial was registered at the Thai Clinical Trials Registry (TCTR20250512003).
    Keywords:  anthocyanin; hyperglycemia; inflammation; insulin; oxygen consumption; polyphenols
    DOI:  https://doi.org/10.3390/nu17213358
  8. Int J Mol Sci. 2025 Nov 05. pii: 10743. [Epub ahead of print]26(21):
      Neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), stroke, and depression, are marked by progressive neuronal dysfunction and loss, yet current treatments remain largely symptomatic with limited disease-modifying efficacy. Glucagon-like peptide-1 (GLP-1), an incretin hormone traditionally associated with metabolic regulation, has emerged as a promising neuroprotective agent. Its receptor, GLP-1R, is expressed in key brain regions implicated in cognition, emotion, and motor control, including the hippocampus, frontal cortex, and substantia nigra. GLP-1R agonists (GLP-1RAs) activate multiple intracellular signaling cascades-cAMP/PKA, PI3K/Akt, and MAPK pathways-that collectively promote neuronal survival, enhance synaptic plasticity, reduce oxidative stress, inhibit apoptosis, and modulate neuroinflammation. These agents also regulate autophagy, promote remyelination, and reprogram microglial phenotypes toward anti-inflammatory states. Preclinical models have shown that GLP-1RAs reduce amyloid-β and tau pathology in AD, preserve dopaminergic neurons in PD, protect astrocytes and neural progenitors after ischemic stroke, and alleviate depressive behaviors. Notably, GLP-1RAs such as liraglutide, exenatide, and dulaglutide can cross the blood-brain barrier and have demonstrated safety and potential efficacy in early-phase clinical trials. These studies report attenuation of cortical atrophy, preservation of cerebral glucose metabolism, and improvements in quality of life, though changes in core AD biomarkers remain inconclusive. Ongoing large-scale trials (e.g., EVOKE, ELAD) are further exploring their therapeutic impact. This review consolidates the mechanistic basis and translational potential of GLP-1RAs in age-related neurodegenerative diseases, highlighting both their promise and the challenges that must be addressed in future clinical applications.
    Keywords:  Alzheimer’s disease; GLP-1 receptor agonists (GLP-1RAs); Parkinson’s disease; clinical trials; neurodegeneration; neuroinflammation; stroke; synaptic plasticity
    DOI:  https://doi.org/10.3390/ijms262110743
  9. Aging Dis. 2025 Oct 28.
      Neurodegenerative diseases represent an escalating global health crisis affecting more than 55 million people worldwide; however, underlying mechanisms remain unclear, and therapeutic breakthroughs are elusive. Emerging evidence indicates that hexokinase (HK), the rate-limiting glycolytic enzyme, functions as a master regulator orchestrating neuronal survival through metabolic‒mitochondrial coupling. This review consolidates emerging paradigms revealing that HK maintains neuronal viability through its obligate interaction with mitochondrial VDAC1, forming a metabolic checkpoint that integrates energy status with survival signaling. Disease-specific HK dysfunction patterns precede clinical manifestations and drive pathological cascades across primary neurodegenerative conditions. Pathological proteins characteristic of neurodegeneration-amyloid-β in AD, α-synuclein in PD, mutant SOD1 in ALS, and huntingtin in HD-converge to disrupt the HK-VDAC1 axis through distinct molecular mechanisms, triggering mitochondrial permeabilization, bioenergetic collapse, and inflammatory activation. This uncoupling event promotes VDAC1 oligomerization, enabling the cytosolic release of mtDNA, which in turn activates the NLRP3 inflammasome while depleting antioxidant capacity, establishing self-perpetuating neuroinflammatory cycles. The literature reveals that HK functions as a molecular rheostat, determining neuronal fate through glucose-6-phosphate-mediated feedback control, modulation of growth factor signaling, and regulation of apoptosis/survival pathways. Therapeutic targeting of HK through peptide interventions, enzymatic modulation, and gene therapy demonstrates robust neuroprotective effects across multiple disease models. Meanwhile, combination strategies addressing metabolic-inflammatory networks show synergistic efficacy. These insights position HK as a convergent therapeutic nexus offering unprecedented opportunities for precision intervention in neurodegeneration, with potential for early diagnostic applications and preventive strategies that could transform treatment paradigms for conditions affecting millions worldwide.
    DOI:  https://doi.org/10.14336/AD.2025.0891
  10. Gen Hosp Psychiatry. 2025 Nov 11. pii: S0163-8343(25)00214-2. [Epub ahead of print]97 284-293
       BACKGROUND: Cognitive impairment, encompassing mild cognitive impairment (MCI) and Alzheimer's disease (AD), poses a significant public health challenge worldwide. Emerging evidences suggest that probiotics, through modulation of the gut-brain axis, may improve cognitive function. However, their efficacy in individuals with cognitive impairment remains unclear.
    METHODS: This meta-analysis adhered to PRISMA guidelines, systematically reviewing randomized controlled trials (RCTs) investigating the effects of probiotics for individuals with cognitive impairment. Data were extracted from the PubMed, Embase, and Cochrane databases. Subgroup analyses were conducted based on different measurement scales (MMSE, MoCA, and RBANS), population types (cognitive impairment vs. no cognitive impairment), multiple or single strain and intervention duration. Effect sizes were expressed as standardized mean differences (SMDs) using random-effects models.
    RESULTS: Fifteen RCTs involving 994 participants were included. Probiotic supplementation significantly improved cognitive function compared to placebo (SMD = 0.57; 95 % CI, 0.19-0.94; P = 0.003). Subgroup analyses revealed that both single-strain and multi-strain probiotics were effective; however, multi-strain probiotics demonstrated greater heterogeneity and variability in efficacy. Cognitive improvements were most pronounced with supplementation durations of at least 12 weeks (SMD = 0.73; 95 % CI, 0.30-1.16; P = 0.0009) and higher probiotic doses (>1 × 109 CFU/g; SMD = 0.93; 95 % CI, 0.36-1.49; P < 0.01).
    CONCLUSION: Probiotic supplementations significantly enhance cognitive function in individuals with cognitive impairment, particularly with single-strain formulations, higher doses, and a minimum duration of 12 weeks.
    Keywords:  Alzheimer's disease; Cognitive impairment; Gut-brain axis; Meta-analysis; Mild cognitive impairment; Probiotics; Randomized controlled trials
    DOI:  https://doi.org/10.1016/j.genhosppsych.2025.11.004
  11. Front Nutr. 2025 ;12 1705284
       Introduction: Polycystic ovary syndrome (PCOS) affects 5-15% of reproductive-aged women and involves significant metabolic dysregulation, for which nutritional interventions show therapeutic potential. Methods: This umbrella meta-analysis synthesizes evidence from 46 randomized trials (n = 30,133) to evaluate dietary supplements targeting core PCOS pathways.
    Methods: This umbrella meta-analysis synthesizes evidence from 46 randomized trials (n = 30,133) to evaluate dietary supplements targeting core PCOS pathways.
    Results: Key nutraceuticals demonstrate clinically relevant benefits: myo-inositol significantly improves insulin sensitivity (HOMA-IR SMD = -0.81) and SHBG levels (SMD = 9.65) by enhancing glucose transporter activity; probiotics reduce systemic inflammation (CRP SMD = -0.82) via gut-microbiota modulation; omega-3 fatty acids ameliorate dyslipidemia (LDL-C SMD = -9.57; HDL-C SMD = 2.31) through anti-inflammatory mechanisms. Plant-derived compounds like curcumin lower fasting glucose (SMD = -3.43) via NF-ĸB pathway inhibition, while green tea catechins reduce adiposity. Significant heterogeneity arises from variations in supplement bioavailability, dosing protocols, and patient metabolic phenotypes. Nevertheless, consistent evidence confirms that targeted nutrient supplementation modulates insulin signaling, lipid metabolism, and hormonal balance in PCOS. Emerging research priorities include personalized nutrition protocols leveraging nutrigenomic interactions and antioxidant-rich formulations (e.g., vitamin E, selenium).
    Discussion: This work establishes a mechanistic foundation for integrating evidence-based nutraceuticals-particularly myo-inositol, probiotics, and omega-3s-into PCOS management, offering clinically actionable strategies while highlighting needs for standardized dosing and bioavailability studies.
    Systematic review registration: https://www.crd.york.ac.uk/PROSPERO/view/CRD42024602681.
    Keywords:  bibliometrics; dietary supplements; future trend; polycystic ovary syndrome; umbrella meta-analysis
    DOI:  https://doi.org/10.3389/fnut.2025.1705284
  12. Cells. 2025 Oct 31. pii: 1711. [Epub ahead of print]14(21):
       BACKGROUND: Cognitive, emotional, and social impairments are pervasive across neuropsychiatric conditions, where alterations in the tryptophan (Trp)-kynurenine pathway and its product kynurenic acid (KYNA) from kynurenine aminotransferases (KATs) have been linked to Alzheimer's disease, Parkinson's disease, depression, and post-traumatic stress disorder. In novel CRISPR/Cas9-engineered KAT II knockout (aadat-/- also known as kat2-/-) mice, we observed despair-linked depression-like behavior with peripheral excitotoxicity and oxidative stress. KAT II's role and its crosstalk with serotonin, indole-pyruvate, and tyrosine-dopamine remain unclear. It is unknown whether deficits extend to cognitive, emotional, motor, and social domains or whether brain tissues mirror peripheral stress.
    OBJECTIVES: Delineate domain-wide behaviors, brain oxidative/excitotoxic profiles, and pathway interactions attributable to KAT II.
    RESULTS: Behavior was unchanged across strains. kat2-/- deletion remodeled Trp metabolic pathways: 3-hydroxykynurenine increased, xanthurenic acid decreased, KYNA fell in cortex and hippocampus but rose in striatum, quinaldic acid decreased in cerebellum and brainstem. These region-specific changes indicate metabolic stress across the brain and align with higher oxidative load and signs of excitotoxic pressure.
    CONCLUSIONS: Here, we show that KAT II deletion reshapes regional Trp metabolism and amplifies oxidative and excitotoxic imbalance. Although domain-wide behavioral measures, spanning cognition, sociability, and motor coordination, remained largely unchanged, these neurochemical alterations signify a latent emotional bias rather than overt depressive-like behavior. This work, therefore, refines prior findings by delineating KAT II-linked biochemical vulnerability as a potential substrate for stress-reactive affective dysregulation.
    Keywords:  affective vulnerability; behavioral test; dopamine; emotional bias; excitotoxicity; gut microbiota; kynurenine; kynurenine aminotransferase (KAT); oxidative stress; serotonin; transgenic mice; tryptophan metabolism
    DOI:  https://doi.org/10.3390/cells14211711
  13. Endocrinology. 2025 Nov 14. pii: bqaf163. [Epub ahead of print]
      Preliminary studies suggest a link between shortened telomeres, infertility, and poorer in vitro fertilization (IVF) outcomes. Infertility patients often experience high levels of psychological stress during treatment. Whether there is a link between stress and telomere length in infertility patients has not previously been studied, thus our goal was to examine differences in telomere length in infertile versus non-infertile women, and to determine if telomere length correlates with psychological stress and IVF laboratory outcomes. We conducted a case-control study comparing nulliparous women aged 35 to 42 years with unexplained infertility or diminished ovarian reserve undergoing IVF to non-infertile age-matched controls. Average telomere length was measured in peripheral blood mononuclear cells. Psychological stress was measured by the Perceived Stress Scale and the Center for Epidemiologic Studies Depression Scale. The infertile cases had significantly shorter telomeres than the fertile controls; stress did not account for this difference. Associations were observed between telomere length, ovarian reserve measures and quantitative IVF outcomes, independent of age, suggesting that telomere attrition in somatic cells may relate to the underlying pathophysiology of low ovarian reserve and fertility status.
    Keywords:  in vitro fertilization; infertility; perceived stress; psychological stress; telomeres
    DOI:  https://doi.org/10.1210/endocr/bqaf163
  14. EBioMedicine. 2025 Nov 11. pii: S2352-3964(25)00451-7. [Epub ahead of print]122 106007
       BACKGROUND: Reduced mitochondrial respiratory function has been implicated in metabolic disorders like type 2 diabetes (T2D), obesity, and metabolic dysfunction-associated steatotic liver disease (MASLD), which are tightly linked to insulin resistance and impaired metabolic flexibility. However, the contribution of the ketone bodies (KBs) β-hydroxybutyrate (HBA) and acetoacetate (ACA) as substrates for mitochondrial oxidative phosphorylation (OXPHOS) in these insulin resistant states remains unclear.
    METHODS: Targeted high-resolution respirometry protocols were applied to detect the differential contribution of HBA and ACA to OXPHOS capacity in heart, skeletal muscle, kidney, and liver of distinct human or murine cohorts with T2D, obesity, and MASLD.
    FINDINGS: In humans with T2D, KB-driven mitochondrial OXPHOS capacity was ∼30% lower in the heart (p < 0.05) and skeletal muscle (p < 0.05) compared to non-diabetic controls. The relative contribution of KBs to maximal OXPHOS capacity in T2D was also lower in both the heart (∼25%, p < 0.05) and skeletal muscle (∼50%, p < 0.05). Similarly, in kidney cortex from high-fat diet-induced obese mice, both the absolute and relative contribution of KBs to OXPHOS capacity was ∼15% lower (p < 0.05). Finally, hepatic HBA-driven mitochondrial OXPHOS capacity was 29% lower (p < 0.05) in obese humans with hepatic steatosis compared to humans without.
    INTERPRETATION: Mitochondrial KB-driven OXPHOS capacity is impaired in insulin resistant states in various organs in absolute and relative terms, likely reflecting impaired mitochondrial metabolic flexibility. Our data suggest that KB respirometry can provide a sensitive readout of impaired mitochondrial function in diabetes, obesity, and MASLD.
    FUNDING: German Research Foundation, German Diabetes Center, German Federal Ministry of Health, Ministry of Culture and Science of the state of North Rhine-Westphalia, German Federal Ministry of Education and Research, German Center for Diabetes Research, German Heart Foundation, German Diabetes Society, Christiane-and-Claudia Hempel Foundation, European Community and Schmutzler Stiftung.
    Keywords:  Diabetes mellitus; Ketone bodies; MASLD; Mitochondrial respiration; Obesity
    DOI:  https://doi.org/10.1016/j.ebiom.2025.106007
  15. Molecules. 2025 Nov 04. pii: 4286. [Epub ahead of print]30(21):
      Oxidative and glycation stress are interrelated pathological processes that significantly contribute to the development and progression of chronic diseases, including diabetes, chronic kidney disease, cardiovascular disorders, and neurodegenerative conditions. These processes alter biomolecules by generating reactive oxygen species (ROS), reactive nitrogen species (RNS), and advanced glycation end products (AGEs), thereby amplifying cellular dysfunction. Therefore, precise monitoring of these biomarkers is essential for understanding disease mechanisms and for clinical assessments. Conventional methods, such as chromatography, mass spectrometry, and immunoassays, provide high sensitivity and specificity; however, their extensive clinical application is restricted owing to their high cost, labor intensity, and equipment requirements. In contrast, emerging electrochemical and optical biosensor technologies offer advantages in terms of rapidity, portability, and real-time analysis and hold promise for point-of-care (POC) testing and integration into wearable devices. This review systematically summarizes the detection principles and clinical applications of oxidative and glycation stress-related biomarkers and highlights the need for integrated monitoring systems that can simultaneously capture both processes. Advances in these technologies are expected to contribute significantly to early diagnosis, risk stratification, and implementation of personalized medicine.
    Keywords:  AGEs; POC testing; biomarkers; biosensors; glycation stress; oxidative stress
    DOI:  https://doi.org/10.3390/molecules30214286