bims-polgdi Biomed News
on POLG disease
Issue of 2025–12–14
fifty-one papers selected by
Luca Bolliger, lxBio



  1. Mitochondrion. 2025 Dec 04. pii: S1567-7249(25)00104-7. [Epub ahead of print]87 102107
      Mitochondrial protein import is necessary to ensure the proper functioning of the organelle of the cell as a whole. More than 1000 proteins are synthesized on cytosolic ribosomes and then imported into mitochondria through translocases such as TOMM and TIMM complexes. Upon entry, they can reach their final mitochondrial compartment, namely the outer mitochondrial membrane (OMM), the intermembrane space (IMS), the inner mitochondrial membrane (IMM), and the matrix. In this review, we will first explore the main mitochondrial protein import mechanisms. Then, we will focus on how import deficiencies may trigger stress paradigms. Stress response pathways are activated to restore correct cellular homeostasis. We will explore four interconnected pathways at the cellular or mitochondrial scale, which can compensate for import alterations. These are the DELE1-HRI axis combined with the ISR, the UPRam, the UPRmt, and mitophagy. Their activation depends on the extent of import alteration, with ISR and UPRmt pathways activated in conditions of low stress. If stress levels are too high, the elimination of dysfunctional mitochondria by mitophagy is triggered. Last, we will explore how mitochondrial import deficiencies are a feature common to multifaceted pathologies, such as neurodegenerative diseases and cancer. We will also present pharmacological compounds mimicking stress response mechanisms and that could be used as a therapeutic option in the near future to restore efficient mitochondrial protein import rates. Overall, this review highlights the critical role of mitochondrial protein import in cellular and mitochondrial stress response, and in disease pathogenesis. It also emphasizes the potential of mitochondrial protein import as a therapeutic target, despite the surprising absence of direct pharmacological treatments to date.
    Keywords:  DELE1/HRI; ISR; Mitochondrial protein import; Pharmacological modulation; UPRam; UPRmt
    DOI:  https://doi.org/10.1016/j.mito.2025.102107
  2. Mol Neurobiol. 2025 Dec 10. 63(1): 279
      Tauopathies are neurodegenerative diseases characterized by the abnormal accumulation of tau protein in neurons, leading to cognitive impairment. A common feature of these disorders is mitochondrial dysfunction, which leads to bioenergetic deficits and contributes to neuronal cell death. As neurons have high energy demands, impaired mitochondrial function directly affects their viability and function. Thus, mitochondria represent an attractive target for neuroprotective strategies in tauopathies. Mitochondrial transplantation (MT) is an emerging therapeutic approach to restoring cellular bioenergetics. Although MT has shown promise in various models of brain diseases, its efficacy has not been evaluated in the context of tau-induced mitochondrial dysfunction. This study examines the impact of MT on healthy cells and in a cellular model of tauopathy. Mitochondria were freshly isolated from astrocytic cells and transplanted into healthy SH-SY5Y neuroblastoma cells and SH-SY5Y cells overexpressing the P301L tau mutation, for 24 and 48 h. Our results demonstrate that MT enhances cell viability, ATP production, mitochondrial membrane potential, and respiration in both healthy and tau-mutant SH-SY5Y cells. In addition, MT reduced mitochondrial superoxide anion levels and promoted neurite outgrowth in both cell lines. Key bioenergetic outcomes were recapitulated in neurons derived from induced pluripotent stem cells (iPSCs) carrying the P301L tau mutation. These findings suggest that MT might be a promising therapeutic strategy to counteract mitochondrial deficits in tauopathies. Importantly, this approach positions mitochondria not as a target but as the therapeutic agent itself. Further studies are warranted to advance MT toward in vivo applications in tau-related neurodegenerative disorders.
    Keywords:  Bioenergetic; Mitochondria; Neurites; P301Ltau mutation; Tauopathies; Transplantation
    DOI:  https://doi.org/10.1007/s12035-025-05604-y
  3. Geroscience. 2025 Dec 12.
      Growing evidence shows that epigenetic modification and mitochondrial dysfunction are hallmarks of aging and are associated with the development of a wide range of age-related diseases. Mitochondrial biogenesis, which is marked by mitochondrial DNA copy number (mtDNAcn), is one of the major regulations of mitochondrial function by a set of transacting elements, including mitochondrial DNA polymerase gamma (POLG), working on the mtDNA control region. In this study, we investigated the mtDNAcn and the methylation status at both mtDNA control and POLGA promoter regions in human blood cells from individuals with a wide range of ages. A total of 119 blood samples were collected, including 24 umbilical cord blood samples from newborns and 95 peripheral blood samples from individuals aged 18 to 96 years. We observed an increase in mtDNAcn, as well as a rise in the methylation levels of the mtDNA control region during aging, particularly in subjects aged ≥ 45. In addition, a positive correlation was also found between the methylation levels of the 4th CpG site in the POLGA promoter region and mtDNAcn during aging. These results suggest epigenetic regulation at mitochondrial and nuclear genes for mitochondrial biogenesis during aging in human blood cells.
    Keywords:  Aging; DNA methylation; DNA polymerase gamma A (POLGA); Mitochondrial DNA D-loop region; Mitochondrial DNA copy number; Mitochondrial biogenesis
    DOI:  https://doi.org/10.1007/s11357-025-02037-2
  4. J Cereb Blood Flow Metab. 2025 Dec 08. 271678X251399018
      Extracellular vesicles (EVs) are naturally secreted as non-cell-autonomous signals involved in regulating immune responses, aging, angiogenesis, and tissue injury and repair within the central nervous system (CNS). Consequently, EVs have emerged as promising therapeutic targets in CNS-related diseases. More recently, a subset of these vesicles has been found to contain mitochondria (mitoEVs), suggesting that vesicles carrying mitochondrial signatures may also function as non-cell-autonomous signals and serve as potential biomarkers for injury or recovery in CNS pathophysiology. In this mini-review, we summarize current findings highlighting the critical roles of EVs and their therapeutic potential as demonstrated in cellular, animal, and human studies related to CNS injury and disease.
    Keywords:  CNS disease; Mitochondria; biomarker; extracellular vesicle; therapy
    DOI:  https://doi.org/10.1177/0271678X251399018
  5. J Cereb Blood Flow Metab. 2025 Dec 07. 271678X251400466
      Extracellular vesicles (EVs) have emerged as critical mediators of cell-to-cell communication. More recently, a subset of these vesicles has been found to contain mitochondria (EV-Mito). These mitochondria-bearing EVs may act as non-cell-autonomous signaling entities and serve as potential biomarkers for injury and recovery in central nervous system (CNS) pathophysiology. Mitochondria play a vital role in regulating cellular respiration, metabolism, and overall tissue function. In the context of CNS injury or disease, mitochondrial dysfunction can disrupt metabolic homeostasis, leading to cell death and inflammation. Consequently, restoring mitochondrial function represents a key therapeutic target with strong translational potential. This special issue of JCBFM presents a multidisciplinary collection of high-impact reviews and original research articles. These contributions cover a broad spectrum-from basic studies on EV-mediated mechanisms in CNS disorders and the molecular pathways underlying intercellular mitochondrial transfer, to therapeutic applications of EVs and mitochondrial transplantation in cellular and animal models. The issue also highlights the latest clinical trial developments assessing the feasibility of EV and mitochondrial transplantation in cerebral ischemia. Collectively, these articles offer valuable insights into emerging research directions and underscore the many unresolved questions that remain-particularly regarding the quantitative thresholds required for treatment efficacy and the molecular mechanisms driving beneficial tissue remodeling.
    Keywords:  CNS disorders; Extracellular mitochondria; Extracellular vesicles; Therapy
    DOI:  https://doi.org/10.1177/0271678X251400466
  6. Cells. 2025 Dec 03. pii: 1923. [Epub ahead of print]14(23):
      The absent in melanoma 2 (AIM2) inflammasome is a cytosolic DNA sensor that links genomic instability, mitochondrial dysfunction, and chronic inflammation. Unlike the nucleotide-binding domain, leucine-rich repeat (NLR) family pyrin domain-containing protein 3 (NLRP3) inflammasome, AIM2 is activated directly by double-stranded Deoxyribonucleic Acid (dsDNA), including mitochondrial DNA (mtDNA) released under stress conditions. This positions AIM2 at the intersection of oxidative stress, impaired mitophagy, and innate immune dysregulation. Current therapies for ankylosis spondylitis (AS), such as anti-tumor necrosis factor (TNF), anti-interleukin 17 (IL-17), and Janus kinase (JAK) inhibitors, improve clinical outcomes; however, they do not address upstream mitochondrial dysfunction or DNA-driven inflammasome activation. By contrast, other inflammasomes, such as AIM2, remain comparatively less studied. Since autoimmune diseases, including AS, are frequently accompanied by uncontrolled innate immune responses to self-DNA, these findings provide a framework for comprehending the mechanisms of AIM2 activation and its interaction with inflammation, mitophagy, and oxidative stress. Here, we review the current evidence on AIM2 inflammasome involvement in AS pathogenesis and its potential as a therapeutic target. This approach offers new insight into disease control through re-establishing the balance between mitochondrial dysfunction and autoimmunity.
    Keywords:  AIM2 inflammasome; ankylosing spondylitis; autoimmunity; mitochondrial dysfunction; mtDNA; targeted therapy
    DOI:  https://doi.org/10.3390/cells14231923
  7. ACS Biomater Sci Eng. 2025 Dec 10.
      Mitochondria are essential organelles that govern energy metabolism, redox balance, and cell survival; their dysfunction is implicated in a wide range of pathologies, including neurodegenerative disorders, cardiovascular diseases, metabolic syndromes, and cancer. Despite their significance as therapeutic targets, the unique structural and electrochemical properties of mitochondria, particularly the impermeable inner mitochondrial membrane and high membrane potential pose major challenges for the targeted delivery of therapeutic agents. Recent advances in biomaterials have spotlighted peptide-polymer conjugates as versatile platforms, capable of navigating intracellular barriers and achieving precise mitochondrial localization. These hybrid systems combine the physicochemical tunability of polymers with the biofunctionality of peptides, enhancing cellular uptake, endosomal escape, and suborganelle trafficking. The incorporation of stimuli-responsive elements further enables spatiotemporal control of cargo release in response to intracellular cues such as pH shifts, thermal fluctuations, redox gradients, or enzymatic activity. Such systems are especially promising for mitochondrial gene and protein delivery, offering improved selectivity, reduced systemic toxicity, and the potential to restore mitochondrial function under pathological conditions. This review showcases advanced strategies in stimuli-responsive peptide-polymer systems for mitochondria-targeted delivery, highlighting how their smart, responsive functions enable precise, controllable therapeutic interventions and drive the development of next-generation, transformative biomaterials in precision nanomedicine.
    Keywords:  gene therapy; mitochondria-targeted delivery; peptide–polymer conjugates; smart drug delivery systems; stimuli-responsive materials
    DOI:  https://doi.org/10.1021/acsbiomaterials.5c01513
  8. Adv Biol (Weinh). 2025 Dec 12. e00472
      HK1 and HK2 are increasingly recognized not only as glycolytic enzymes but also as key modulators of mitochondrial function and cell fate through dynamic interactions with VDAC. This review explores how HK-VDAC complexes support metabolic flexibility, regulate apoptosis, and coordinate glycolytic and mitochondrial activity across diverse physiological and pathological conditions. We incorporate recent reinterpretations of the Warburg effect, emphasizing how spatial and functional reorganization of HK supports proliferative metabolism beyond classical models of mitochondrial dysfunction. Importantly, the HK-VDAC interaction is dynamically regulated by post-translational modifications and signaling pathways that control its stability and mitochondrial anchoring. Disruption of these regulatory mechanisms can impair the balance between glycolytic and mitochondrial metabolism, contributing to disease progression. Emerging evidence links altered HK-VDAC interactions to the metabolic and apoptotic imbalances observed in cancer, neurodegeneration, and aging. By integrating insights from structural biology, bioenergetics, and disease models, we highlight mitochondrial HK anchoring as a central hub for metabolic adaptation and stress response.
    Keywords:  HK‐VDAC; Warburg effect; aging; apoptosis; cancer; metabolism; mitochondria
    DOI:  https://doi.org/10.1002/adbi.202500472
  9. Int J Mol Sci. 2025 Nov 25. pii: 11370. [Epub ahead of print]26(23):
      Mitochondria are highly dynamic and responsive organelles that perform multifaceted functions depending on the environmental conditions and cellular demands [...].
    DOI:  https://doi.org/10.3390/ijms262311370
  10. G3 (Bethesda). 2025 Dec 08. pii: jkaf298. [Epub ahead of print]
      Aminoacyl-tRNA synthetases (aaRSs) are essential for translation, as they charge tRNA molecules with their corresponding amino acids. Alterations in aaRSs can significantly disrupt both cytosolic and mitochondrial translation. Through a forward genetic screen for mitochondrial unfolded protein response (UPRmt) activators in C. elegans, we identified a missense mutation (P447V) in the previously uncharacterized gene Y105E8A.20, which encodes for a methionine tRNA synthetase (MetRS). Here, we characterize the UPRmt induction by Y105E8A.20, which we call mars-2, and demonstrate that the P447V allele is a loss-of-function mutation. Furthermore, we show that impaired mars-2 activity leads to reduced mitochondrial-encoded protein abundance, depletion of mitochondrial membrane potential, fragmented mitochondrial morphology, and mild developmental delay, although the animals remain viable. Hence, this hypomorphic mars-2(P447V) strain provides a valuable tool for studying mitochondrial translation and understanding how aaRSs are involved in mitochondrial homeostasis.
    Keywords:   Caenorhabditis elegans ; WormBase; mars-2; metionine tRNA-synthetase; mitochondria; mitochondrial unfolded protein response; mtDNA; tRNAs; translation
    DOI:  https://doi.org/10.1093/g3journal/jkaf298
  11. Ann Med Surg (Lond). 2025 Dec;87(12): 9163-9164
      Barth syndrome is a rare X-linked mitochondrial disorder caused by mutations in the Tafazzin (TAZ) gene. These mutations make it hard for cardiolipin to remodel and mitochondria to work properly. This condition is characterized by growth retardation, neutropenia, skeletal myopathy, and dilated cardiomyopathy, frequently leading to significant morbidity and mortality, with numerous patients necessitating heart transplants. There are no treatments available at this time to fix the genetic problem. Recent progress in gene editing, especially CRISPR-based methods, holds great promise for fixing TAZ mutations. Research utilizing patient-derived cardiomyocytes has demonstrated that the rectification of TAZ mutations reinstates mitochondrial efficiency and enhances cellular functionality. Animal models, including TAZ-knockout mice, have exhibited substantial enhancements in cardiac function, survival rates, and diminished fibrosis subsequent to gene replacement therapy.
    Keywords:  adeno-associated virus vectors; barth syndrome; cardiolipins; cardiomyopathy, dilated; crispr-cas systems; gene editing; gene therapy; induced pluripotent stem cells; mitochondrial diseases; myopathy; neutropenia
    DOI:  https://doi.org/10.1097/MS9.0000000000004188
  12. Transl Neurodegener. 2025 Dec 08. 14(1): 64
       BACKGROUND: Previous studies have shown that astrocytes can transfer healthy mitochondria to dopaminergic (DA) neurons, which may serve as an intrinsic neuroprotective mechanism in Parkinson's disease (PD). LRRK2 G2019S is the most common pathogenic mutation associated with PD. In this study, we explored whether mitochondrial transfer is influenced by genetic and environmental factors and whether dysfunction in this process is one of the mechanisms of the pathogenic LRRK2 G2019S mutation.
    METHODS: DA neurons and astrocytes were differentiated from induced pluripotent stem cells generated from the peripheral blood of a healthy individual and a PD patient carrying the LRRK2 G2019S mutation. A coculture system of astrocytes and DA neurons was established to explore the pathogenic mechanisms of LRRK2 G2019S.
    RESULTS: Exposure to the environmental toxin rotenone impaired mitochondrial transfer from astrocytes to DA neurons. Compared with the co-culture system from the healthy participant, the co-culture system harboring the LRRK2 G2019S mutation experienced more pronounced damage. Specifically, STX17 was colocalized with the mitochondrial outer membrane marker TOM20, and its knockdown caused damage to mitochondrial transfer. Drp1 interacted with STX17. LRRK2 G2019S-mutant astrocytes exhibited markedly increased phosphorylation of Drp1 at Ser616 upon rotenone exposure. Moreover, the degree of colocalization of STX17 with TOM20 decreased. The Drp1 phosphorylation inhibitor DUSP6 restored the colocalization of STX17 and TOM20, as well as the mitochondrial transfer efficiency and neuronal survival.
    CONCLUSIONS: The impairment of mitochondrial transfer is a potential pathogenic mechanism associated with LRRK2 G2019S mutation. The molecular mechanisms of mitochondrial transfer were observed to occur through a Drp1-STX17-dependent pathway. Notably, inhibitors for Drp1 Ser616 phosphorylation may offer neuroprotection through mitigating mitochondrial transfer impairments. This study provides novel insights into the pathogenesis of PD and the development of new therapeutic targets.
    Keywords:   LRRK2 G2019S mutation; Astrocyte; Dopaminergic neuron; Induced pluripotent stem cell; Membrane fusion-related protein STX17; Mitochondrial transfer; Parkinson’s disease
    DOI:  https://doi.org/10.1186/s40035-025-00525-1
  13. BMJ Case Rep. 2025 Dec 11. pii: e269373. [Epub ahead of print]18(12):
      DNA polymerase subunit gamma-1 (POLG)-related disease is a heterogeneous spectrum of mitochondrial disorders with neurologic and hepatic manifestations. We report a woman in her 20s who developed refractory seizures followed by fulminant hepatic failure after valproic acid exposure. Laboratory evaluation revealed low copper indices without evidence of Wilson disease, neuroimaging demonstrated evolving posterior-predominant abnormalities, and liver biopsy showed acute hepatitis with microvesicular change and 'two-toned' hepatocytes. Rapid whole-genome sequencing identified compound-heterozygous POLG variants c.1399G>A p.(Ala467Thr) and c.2243G>C p.(Trp748Ser), confirming a juvenile/adult-onset POLG-related disorder. This case highlights key diagnostic pitfalls, including potential misdirection of copper studies and risk of valproate hepatotoxicity in patients with unrecognised POLG variants. Supportive clues like occipital-predominant electroencephalogram/MRI changes, rapid neurologic-hepatic progression and hepatic microvesicular pathology can aid early suspicion but are not universally present. Prompt genetic testing and multidisciplinary follow-up are essential to guide management, avoid harmful therapies and anticipate the trajectory of this multisystem disease.
    Keywords:  Epilepsy and seizures; Genetics; Immunology; Liver disease
    DOI:  https://doi.org/10.1136/bcr-2025-269373
  14. Int J Mol Sci. 2025 Nov 30. pii: 11615. [Epub ahead of print]26(23):
      Parkinson's disease (PD) is a progressive neurodegenerative disorder marked by the gradual and irreversible loss of neurons, especially within the substantia nigra region of the midbrain. Early and accurate diagnosis remains a significant challenge in both research and clinical practice. This difficulty is further compounded by the substantial clinical and molecular heterogeneity of PD, emphasizing the urgent need for reliable biomarkers to enhance diagnostic precision and guide therapeutic strategies. One promising candidate biomarker is cell-free DNA (cfDNA), comprising short DNA fragments composed of mitochondrial (cf-mtDNA) and nucleus-derived (cf-ntDNA) DNA. cfDNA is released into body fluids through physiological or pathological processes such as apoptosis, necrosis, NETosis, or active secretion. The presence of cfDNA in human biological fluids has been utilized for years in oncology and prenatal medicine and, more recently, it has gained attention as a non-invasive diagnostic tool in the context of neurodegenerative diseases such as PD. This review article aims to provide a comprehensive overview of the current knowledge on the origin of cfDNA, highlighting the roles of the mitochondria and cf-mtDNA in PD, mitochondria quality control, and neuroinflammation in cfDNA biogenesis. The review collates available research on cfDNA types in human serum, plasma, and CSF, sequence analysis, and its potential application as a biomarker in the diagnosis and monitoring of PD, contributing to the ongoing search for non-invasive biomarkers of neurodegenerative diseases.
    Keywords:  Parkinson’s disease; cell-free DNA (cfDNA); cell-free mitochondrial DNA (cf-mtDNA); cell-free nuclear DNA (cf-ntDNA); cerebrospinal fluid; mitochondria; mitophagy; neuroinflammation; plasma; serum
    DOI:  https://doi.org/10.3390/ijms262311615
  15. Adv Sci (Weinh). 2025 Dec 08. e17721
      Mitochondrial dysfunction and impaired neurogenesis are central to mitochondrial DNA polymerase (POLG)-related disorders, yet therapeutic options remain limited. Here, patient-derived induced pluripotent stem cell (iPSC)-based cortical organoids are used to model POLG-associated neurodegeneration and assess the therapeutic potential of metformin. Single-cell RNA-seq reveals distinct vulnerabilities in dopaminergic, glutamatergic, and GABAergic neuronal subtypes, with dopaminergic neurons exhibiting the most severe loss and mitochondrial transcriptomic deficits. Metformin treatment (250 µm, 2 months) significantly restores neuronal identity, subtype-specific gene expression, and mitochondrial function. Functional assays demonstrate improved mitochondrial membrane potential (TMRE), increased mitochondrial mass (MTG, MTDR), and reduced oxidative stress (MitoSOX, BAX/cleaved caspase 3). Notably, mitochondrial DNA (mtDNA) copy number and the expression of mitochondrial replisome proteins (POLG, POLG2) are upregulated, indicating enhanced mitochondrial genome maintenance. Calcium measurement confirms improved neuronal excitability. Untargeted metabolomics further reveals metformin-induced metabolic reprogramming, including enrichment of the tricarboxylic acid (TCA) cycle, amino acid metabolism, and redox-related pathways. Together, these findings demonstrate that metformin enhances mitochondrial integrity and neural function across multiple neuronal subtypes and offer mechanistic insights into its potential as a treatment for POLG-related disorders.
    Keywords:  POLG‐related disorders; cortical organoids; iPSCs; mitochondrial dysfunction; neurogenesis impairment
    DOI:  https://doi.org/10.1002/advs.202417721
  16. Biol Chem. 2025 Dec 10.
      The diverse, and sometimes opposing, roles of mitochondria require sophisticated organizational and regulatory strategies. This review examines emerging evidence that mitochondria can solve this challenge through functional specialization - adopting distinct bioenergetic and metabolic programs based on location, contacts, and cellular conditions. We discuss both established principles and recent technological breakthroughs that reveal this hidden complexity. Ongoing advances promise to move the field from describing mitochondrial diversity to uncovering its regulatory mechanisms and therapeutic potential.
    Keywords:  heterogeneity; metabolic specialization; mitochondria
    DOI:  https://doi.org/10.1515/hsz-2025-0210
  17. FEBS J. 2025 Dec 07.
      The biogenesis of mitochondria relies on the import of newly synthesized precursor proteins from the cytosol. Tom70 is a mitochondrial surface receptor which recognizes precursors and serves as an interface between mitochondrial protein import and the cytosolic proteostasis network. Mitochondrial import defects trigger a complex stress response, in which compromised protein synthesis rates are a characteristic element. The molecular interplay that connects mitochondrial (dys)function to cytosolic translation rates in yeast cells is however poorly understood. Here, we show that the deletion of the two Tom70 paralogs of yeast (TOM70 and TOM71) leads to defects in mitochondrial biogenesis and slow cell growth. Surprisingly, upon heat stress, the deletion of ZUO1, a chaperone of the ribosome-associated complex (RAC), largely prevented the slow growth and the reduced translation rates in the tom70Δ/tom71Δ double deletion mutant. In contrast, the mitochondrial defects were not cured but even enhanced by ZUO1 deletion. Our study shows that Zuo1 is a critical component in the signaling pathway that mutes protein synthesis upon mitochondrial dysfunction. We propose a novel paradigm according to which RAC serves as a stress-controlled regulatory element of the cytosolic translation machinery.
    Keywords:  Tom70; mitochondria; protein import; proteostasis; ribosome‐associated complex
    DOI:  https://doi.org/10.1111/febs.70356
  18. J Genet Couns. 2025 Dec;34(6): e70154
      Genetic testing is often portrayed as a neutral tool for clinical clarification. However, in the context of rare diseases, it plays a far more complex role in the moral, bureaucratic, and social experiences of patients and families. Drawing on qualitative research conducted in Chile, this article explores how genetic testing is not only a diagnostic resource but also a symbolic and institutional technology that shapes how care, legitimacy, and identity are negotiated across multiple domains. We analyze 11 narrative interviews with patients and family members affected by rare diseases, alongside 10 interviews with healthcare professionals, including geneticists and clinicians, and two hybrid forums. Using a grounded, interdisciplinary approach informed by Science and Technology Studies (STS) and medical sociology, we identify four key themes: (1) the moral resolution offered by diagnosis after long periods of uncertainty; (2) structural barriers to accessing testing, including social capital and institutional discretion; (3) the epistemic ambiguity of genetic information, especially in cases of inconclusive results; and (4) the mobilization of diagnoses beyond the clinic, including in education, legal systems, and reproductive decisions. Our findings suggest that genetic testing functions as a moral and classificatory technology: it produces recognition, redistributes responsibility, and serves as a resource for navigating fragmented care systems. For genetic counselors, this research offers insights into how patients and families interpret, act on, and negotiate the consequences of genetic information. Understanding the broader moral and institutional landscape in which testing occurs can enhance counseling practices, particularly in under-resourced or structurally unequal contexts. This study contributes to growing efforts to embed genetic counseling in a wider social and ethical framework. By attending to how genetic testing is lived, contested, and deployed, we highlight the need for a counseling practice that is attuned to both genomic data and the realities of care.
    Keywords:  epistemic justice; genetic testing; health equity; patient experience; rare diseases
    DOI:  https://doi.org/10.1002/jgc4.70154
  19. Nat Commun. 2025 Dec 12. 16(1): 11088
      Mitochondria are central to cellular metabolism. Various fluorescence tools have been developed for imaging the mitochondrial environment. Yet, new reporters and imaging methods for directly reading the mitochondrial status are needed for high spatial-temporal resolution imaging. Here, we introduce PK Mito Deep Red (PKMDR), a low-phototoxicity mitochondrial probe for time-lapse imaging, whose fluorescence lifetime serves as a sensitive indicator of mitochondrial membrane potential (Δψm). The positively charged PKMDR accumulates within mitochondria under a higher Δψm, leading to concentration-induced quenching and a measurable decrease in fluorescence lifetime. Since mitochondrial respiration primarily regulates Δψm, PKMDR's fluorescence lifetime effectively reports on the status of oxidative phosphorylation. Using PKMDR with fluorescence lifetime imaging microscopy (FLIM), we visualize heterogeneous Δψm across individual cells, organoids, and tissues over time. This method reliably reveals the heterogeneity between metabolically active peripheral mitochondria and relatively inactive perinuclear mitochondria in various cell types. Overall, PKMDR-FLIM is a robust tool for directly visualizing Δψm with high spatiotemporal resolution.
    DOI:  https://doi.org/10.1038/s41467-025-66042-x
  20. J Neuropathol Exp Neurol. 2025 Dec 09. pii: nlaf136. [Epub ahead of print]
      Mitochondria are critical for cellular function. Their dysfunction contributes to cell degeneration and death, leading to disease progression. This study examined mitochondrial changes in idiopathic inflammatory myopathies (IIMs) including antisynthetase syndrome (ASyS), dermatomyositis (DM), and inclusion body myositis (IBM). Skeletal muscle biopsies were analyzed using histology, histochemistry, and electron microscopy from patients diagnosed with IIMs, according to the clinico-sero-morphological classification. There was no significant age difference between 16 ASyS and 16 DM patients, but 11 IBM patients were significantly older. The ASyS group had higher serum creatine kinase levels and showed prominent mitochondrial abnormalities similar to IBM and greater than the DM group. While all IIM groups displayed conventional mitochondrial changes, including ultrastructural abnormalities with cristae alterations, paracrystalline inclusions were exclusive to IBM and ASyS. There were significantly more rod-like filamentous inclusions adjacent to mitochondria in the IBM and ASyS groups, compared to the DM group. Intra-mitochondrial filament aggregates with focal formation of inclusions were also identified in individual ASyS and IBM patients, suggesting a link between the mitochondrial filamentous inclusions and nuclear and/or cytoplasmic filamentous inclusions. These findings suggest that mitochondrial abnormalities, particularly in ASyS and IBM, may contribute to the pathogenic process and clinical manifestations of the disease.
    Keywords:  antisynthetase syndrome; dermatomyositis; filamentous inclusions; inclusion body myositis; inflammatory myopathies; mitochondria; muscle biopsy
    DOI:  https://doi.org/10.1093/jnen/nlaf136
  21. Neuropsychobiology. 2025 Dec 12. 1-30
      Bipolar disorder (BD) is a severe, recurrent mood disorder associated with mitochondrial and bioenergetic dysfunction, which may contribute to both symptom expression and variability in treatment response. Although lithium remains the gold standard treatment, a significant proportion of patients fail to achieve full benefit, and reliable predictive biomarkers are still lacking. Increasing evidence suggests that lithium exerts part of its therapeutic effects through modulation of mitochondrial function, including enhanced oxidative phosphorylation, regulation of mitochondrial dynamics, and reduction of oxidative stress. In this state-of-the-art review, we synthesize the current literature on the relationship between lithium and mitochondrial function, with the aim of evaluating how this relationship may inform our understanding of lithium response in BD. We reviewed findings on mitochondrial bioenergetics, oxidative stress, and mitochondrial DNA alterations, and discussed the roles of key regulatory proteins such as Drp1, Opa1, Mfn2, and Nrf2. In addition, we explore peripheral and epigenetic biomarkers, including mitochondrial DNA D-loop methylation, microRNAs and a potential therapeutic target ⎻ mitochondrial transfer mechanism. In addition to synthesizing the existing literature, we identify key gaps that hinder progress, such as clinical studies being predominantly cross-sectional, lacking standardized mitochondrial assessments, and rarely employing longitudinal or genetically informed designs like mitochondrial twin studies. We highlight the need for unified protocols, integration of omics technologies, extracellular vesicle-based sampling strategies, and improved in vitro and in vivo models. A better understanding of mitochondrial signatures related to lithium may enable biomarker discovery and advance personalized treatment in BD.
    DOI:  https://doi.org/10.1159/000549993
  22. JCI Insight. 2025 Dec 08. pii: e177004. [Epub ahead of print]10(23):
      Mitochondrial dysfunction is a major mechanism of acute kidney injury (AKI), and increased circulating interleukin 6 (IL-6) is associated with systemic inflammation and death due to sepsis. We tested whether kidney mitochondrial DNA (mtDNA) contributes to IL-6 release in sepsis-associated AKI via Toll-like receptor 9 (TLR9). In a murine model of sepsis via cecal ligation and puncture (CLP), we used next-generation sequencing of plasma mtDNA to inform the design of optimal target sequences for quantification by droplet digital PCR, and to identify single-nucleotide polymorphisms (SNPs) to infer tissue origin. We found significantly higher concentrations of plasma mtDNA after CLP versus shams and that plasma mtDNA SNPs matched kidney SNPs more than other organs. Kidney mtDNA contributed directly to IL-6 and mtDNA release from dendritic cells in vitro and kidney mitochondria solution led to higher IL-6 concentrations in vivo. IL-6 release was mitigated by a TLR9 inhibitor. Finally, plasma mtDNA was significantly higher in septic patients with AKI compared with those without AKI and correlated significantly with plasma IL-6. We conclude that AKI contributes to increased circulating IL-6 in sepsis via mtDNA release. Targeting kidney mitochondria and mtDNA release are potential translational avenues to decrease mortality from sepsis-associated AKI.
    Keywords:  Inflammation; Innate immunity; Mitochondria; Nephrology
    DOI:  https://doi.org/10.1172/jci.insight.177004
  23. Gut Pathog. 2025 Dec 08. 17(1): 100
      The gut microbiota and its hypoxic host environment play a critical role in human health. Despite its importance, the mechanisms maintaining homeostasis and the characteristics defining dysbiosis remain largely undefined. In particular, the regulation of intestinal oxygen (IO) levels emerges as a critical factor in maintaining microbial balance. Host-driven factors, including epithelial oxygen consumption, mucosal perfusion, and luminal gas diffusion, establish a hypoxic gradient essential for the stable colonization by obligate anaerobes. Disruptions to this gradient, leading to pathological hyperoxia, are associated with overgrowth of facultative anaerobic bacteria and contribute to gastrointestinal diseases like ulcerative colitis, colorectal cancer, and irritable bowel syndrome. Emerging therapeutic approaches focus on modulating IO homeostasis to address dysbiosis. Compounds like sodium tungstate inhibit microbial respiratory pathways, while PPAR-γ agonists enhance mitochondrial efficiency in colonic epithelial cells, thereby restoring proper hypoxia. Dietary interventions and probiotic therapies also hold promise by promoting local anaerobic conditions and enhancing barrier functions, thus supporting the restoration of a healthy microbial community. This review highlights the role of IO in shaping host-microbe interactions, focusing on how host IO levels influence microbial homeostasis. We evaluate the potential for IO modulation to improve gut microbiota structure and explore its impact on microbial metabolism and disease pathogenesis. Additionally, we discuss the promise of dietary, probiotic, and pharmacological interventions in restoring the host's control over the IO microenvironment and microbiota, aiming to prevent and treat related diseases.
    Keywords:  Dysbiosis; Gut microbiome; Hypoxia; Mitochondria; Oxygen
    DOI:  https://doi.org/10.1186/s13099-025-00783-4
  24. Nat Commun. 2025 Dec 12. 16(1): 11103
      Microglial capacity to adapt to tissue needs is a hallmark feature of these cells. New studies show that mitochondria critically regulate the phenotypic adaptability of macrophages. To determine whether these organelles play similar roles in shaping microglial phenotypes, we generated transgenic mouse crosses to accurately visualize and manipulate microglial mitochondria. We find that brain-region differences in microglial attributes and responses to aging are accompanied by regional differences in mitochondrial mass and aging-associated mitochondrial remodeling. Microglial mitochondria are also altered within hours of LPS injections and microglial expression of inflammation-, trophic-, and phagocytosis-relevant genes is strongly correlated with expression of mitochondria-relevant genes. Finally, direct genetic manipulation of microglial mitochondria alters microglial morphology and leads to brain-region specific effects on microglial gene expression. Overall, this study advances our understanding of microglial mitochondria and supports the idea that mitochondria influence basal microglial phenotypes and phenotypic remodeling that takes place over hours to months.
    DOI:  https://doi.org/10.1038/s41467-025-66709-5
  25. Nat Commun. 2025 Dec 12. 16(1): 11104
      Microglia continually surveil the brain allowing for rapid detection of tissue damage or infection. Microglial metabolism is linked to tissue homeostasis, yet how mitochondria are subcellularly partitioned in microglia and dynamically reorganize during surveillance, injury responses, and phagocytic engulfment in the intact brain are not known. Here, we performed intravital imaging and ultrastructural analyses of microglia mitochondria in mice and human tissue, revealing that microglial processes diverge in their mitochondrial content, with some containing multiple mitochondria while others are completely void. Microtubules and hexokinase 2 mirror this uneven mitochondrial distribution indicating that these cytoskeletal and metabolic components are linked to mitochondrial organization in microglia. Microglial processes that engage in minute-to-minute surveillance typically do not have mitochondria. Moreover, unlike process surveillance, mitochondrial motility does not change with animal anesthesia. Likewise, the processes that acutely chemoattract to a lesion site or initially engage with a neuron undergoing programmed cell death do not contain mitochondria. Rather, microglia mitochondria have a delayed arrival into the responding cell processes. Thus, there is subcellular heterogeneity of mitochondrial partitioning. Moreover, microglial processes that surveil and acutely respond to damage do not contain mitochondria.
    DOI:  https://doi.org/10.1038/s41467-025-66708-6
  26. Front Cell Dev Biol. 2025 ;13 1679675
      Lowe syndrome (LS) is an X-linked, recessive disease with a characteristic clinical triad of eye, brain, and kidney defects. LS results from mutations in the OCRL gene that encodes for inositol polyphosphate 5-phosphatase enzyme. The OCRL protein has been localized to multiple subcellular organelles including the plasma membrane and endo-lysosomal system, but the relevance of these to disease phenotypes is unclear. Previous studies have reported severe hypotonia at birth in LS patients along with structural changes in the mitochondria in muscle biopsies. These mitochondrial changes have been proposed to be secondary to renal tubular acidosis seen in LS patients. In this study, we find that neural stem cells and neurons differentiated from OCRL-depleted induced pluripotent stem cells (iPSCs) show mild defects in mitochondrial structure and function, whereas such defects are not seen in the iPSCs themselves. These mitochondrial phenotypes in neural stem cells and neurons were associated with modest changes in the mitochondrial transcriptome. Overall, our results indicate that loss of OCRL leads to mild cell autonomous defects in mitochondrial structure and function that is cell type-dependent.
    Keywords:  Lowe syndrome; glia; iPSC; metabolism; mitochondria; neural stem cells; neurons
    DOI:  https://doi.org/10.3389/fcell.2025.1679675
  27. J Med Genet. 2025 Dec 11. pii: jmg-2025-111137. [Epub ahead of print]
       BACKGROUND: Whole genome sequencing (WGS) has recently been introduced as a diagnostic test for patients with particular rare diseases in the National Health Service (NHS) in England. Little is known about the process of communicating results from WGS to families in practice.
    METHODS: We audio-recorded clinicians and parents discussing the results of WGS for their child's rare disease diagnosis as part of a larger mixed-methods evaluation of the implementation of the NHS Genomic Medicine Service during its early years.
    RESULTS: 10 consultations were audio-recorded across four NHS Trusts. Clinical indications for WGS were related to neurological and developmental disorders. Seven parents received a genetic diagnosis for their child's condition, two received a variant of uncertain significance, and one received a no primary finding result. One parent also received an incidental finding for their child. Challenges in discussing results included (1) explaining a diagnosis when the genotype was established before detailed phenotyping, (2) navigating follow-up for an adult-onset condition identified in childhood, (3) disclosing an unexpected diagnosis for a parent from trio testing and (4) conveying a diagnosis with an uncertain prognosis.
    CONCLUSION: This study illustrates some of the issues that can arise from unexpected and uncertain information when returning results from broad-scope genomic testing for paediatric neurological and developmental disorders. Further study of actual interactions between clinicians and families discussing results from WGS across different specialities and conditions is needed to inform guidance on communication of results within this rapidly evolving area of medicine.
    Keywords:  Child Health; Genetic Counseling; Genetics, Medical; Genomics; Health Services Research
    DOI:  https://doi.org/10.1136/jmg-2025-111137
  28. Front Aging Neurosci. 2025 ;17 1678460
      Parkinson's disease (PD) is a common neurodegenerative disorder with a rising incidence in aging populations, substantially diminishing patients' quality of life. Mitochondria are central to neuronal energy metabolism, and mitophagy plays a pivotal role in maintaining mitochondrial quality by removing damaged organelles. In PD, impaired mitophagy leads to the accumulation of dysfunctional mitochondria, exacerbating oxidative stress and bioenergetic deficits and thereby accelerating disease progression. In recent years, exercise has emerged as a safe and cost-effective intervention that alleviates PD symptoms. Exercise can activate mitophagy through key signaling pathways-including AMP-activated protein kinase (AMPK)/Unc-51-like kinase 1 (ULK1) and PTEN-induced kinase 1 (PINK1)/Parkin-thereby enhancing mitochondrial function and antioxidant capacity. This review synthesizes current evidence on how exercise modulates mitophagy to confer neuroprotection in PD, providing conceptual and practical insights for non-pharmacological management strategies in neurodegenerative disease.
    Keywords:  AMPK signaling; PINK1/Parkin pathway; Parkinson’s disease; exercise intervention; mitophagy
    DOI:  https://doi.org/10.3389/fnagi.2025.1678460
  29. bioRxiv. 2025 Dec 01. pii: 2025.11.27.690007. [Epub ahead of print]
       Background and aims: Transfer RNA (tRNA) modifications determine translation fidelity and efficiency. It occurs through the action of specific enzymes that modify the nucleotides within the tRNA molecule. Our previous study demonstrated tRNA modopathies and altered queuine-related metabolites in inflammatory bowel diseases. Queuine tRNA-ribosyltransferase catalytic subunit 1 (QTRT1) and QTRT 2 co-localize in mitochondria and form a heterodimeric TGT participating in tRNA Queuosine (tRNA-Q) modification. Human body acquires Queuine/Vitamin Q from intestinal microbiota or from diet. However, the roles of tRNA-Q modifications in the maintenance of intestinal mitochondrial homeostasis and microbiome are still unclear.
    Methods: We used publicly available human IBD datasets, QTRT1 knockout (KO) mice, QTRT1 intestinal epithelial conditional KO (QTRT1 ΔIEC ) mice, cultured cell lines with QTRT1-specific siRNA, and organoids from patients with IBD to investigate the mechanism of tRNA-Q modifications in intestinal mitochondrial homeostasis and therapeutic potential in anti-inflammation.
    Results: In single cell RNA sequencing datasets of human IBD, we identified significant reduced intestinal epithelial QTRT1 expression in the patients with Crohn's Disease. Using publicly available datasets, we identified significantly changes of Vitamin Q-associated bacteria in human IBD, compared to the healthy control. Qtrt1 -/- mice had significant reduction of Q-associated bacteria, e.g., Bacteroides . Alcian Blue and Mucin-2 staining revealed mucosal barrier damage and disrupted homeostasis, with reduced colonic cell proliferation. Intestinal tight junction integrity was impaired in QTRT1-KO mice, as evidenced by reduced ZO-1 and increased Claudin-10 expression. QTRT1 ΔIEC mice also showed dysbiosis and disrupted TJs. ATP synthesis was significantly decreased in the colon of QTRT1-KO mice, accompanied by severe mitochondrial dysfunction: reduced mitochondrial quality, Cytochrome-C release, and mitochondrial DNA (mtDNA) leakage. Mitochondrial dysfunction contributed to colonic cell death, as shown by elevated expressions of Cleaved Caspase-3 and Cleaved Caspase-1, increased BAX/Bcl-2 ratio, and positive TUNEL signals. Elevated CDC42, CD14, and CD4 levels in QTRT1-KO colon suggested mucosal immune activation and tissue repair responses. QTRT1-deficient CaCO2-BBE cells showed mitochondrial dysfunction. Cytochrome-C and mito-DNA release leading to cell death characterized by elevated expressions of Cleaved Caspase-3 and Caspase-1, increased BAX/Bcl-2 ratio, and higher apoptosis rate. Organoids isolated from patients with IBD showed reduced levels of QTRT1 and dysfunctional mitochondria. Restoring mitochondrial function leads to enhanced QTRT1.
    Conclusions: These findings underscore the critical role of QTRT1/Q-tRNA modification in maintaining intestinal and microbial homeostasis. Mechanistically, QTRT1 loss impacts mitochondrial integrity and mucosal homeostasis. Our study highlights the novel roles of tRNA-Q modification in maintaining mucosal barriers and innate immunity in intestinal health.
    What is already known about this subject?: Eukaryotes acquire queuine (q), also known as Vitamin Q, as a micronutrient factor from intestinal microbiota or from diet.Vitamin Q is needed for queuosine (Q) modification of tRNAs for the protein translation rate and fidelity.Queuine tRNA-ribosyltransferase catalytic subunit 1 (QTRT1) is reduced in human IBD.However, health consequences of disturbed availability of queuine and altered Q-tRNA modification in digestive diseases remain to be investigated.
    What are the new findings?: QTRT1 deficiency leads to altered microbiome and reduced Vitamin Q-associated bacteria in human IBD and a QTRT1 KO animal model.QTRT1 protects the host against losing intestinal integrity during inflammation.QTRT1 localizes in mitochondria and plays novel functions by maintaining intestinal mitochondrial function. QTRT1 loss impacts tRNA modification in the intestine, linking to mitochondrial integrity and mucosal homeostasis.Human IBD showed reduced levels of QTRT1 and dysfunctional mitochondria. Restoring mitochondrial function leads to enhanced QTRT1.
    How might it impact on clinical practice in the foreseeable future?: Targeting tRNA-Q modification in enhancing mitochondrial function will be a novel method to maintain intestinal health.
    DOI:  https://doi.org/10.1101/2025.11.27.690007
  30. Health Technol Assess. 2025 Dec;29(65): 1-172
       Background: Newborn screening using whole genome sequencing is being evaluated in numerous projects across the world, including Genomics England Limited's Generation Study. It presents considerable challenges for policy advisors, not least, given the logistics of simultaneously evaluating the evidence for the suggested 200 rare genetic conditions. The 'genotype-first' approach has the potential for harms through overdiagnosis, and benefits are uncertain.
    Objective: To assess different approaches to evaluating whole genome sequencing for newborn screening to inform the development of a robust method of evaluation for informing policy decisions.
    Methods: We approached the objective with systematic review methods for a sample of five conditions (considering gene penetrance, expressivity, accuracy and effectiveness of whole genome sequencing and effect of earlier treatment) (search inception to November 2023), evaluated the National Institutes of Health [US] Clinical Genome Resource (ClinGen) as an alternative evidence source for the five conditions and we compared this to a review of genomic studies of newborn screening cohorts reporting penetrance for pathogenic variants of any paediatric condition (search inception to February 2024). We undertook a methodological review of economic evaluations of whole genome sequencing/whole exome sequencing (search inception to January 2024) and explored public views on evaluating whole genome sequencing.
    Data sources: MEDLINE (Ovid), EMBASE (Ovid), Web of Science, Science Citation Index (via Clarivate), the Cochrane Library (via Wiley), cost-effectiveness analysis registry and American Economic Association electronic bibliography. Actionability reports and scores from the Clinical Genome Resource website (downloaded 30 April 2024).
    Results: The traditional review approach identified 268 studies reporting the genetic spectrum of individuals with the five conditions or benefits of earlier, symptomatic treatment. No evidence on the penetrance and expressivity or the accuracy or effectiveness of whole genome sequencing in newborns was identified. A review of 200 conditions would take a team of five reviewers 23 years to complete. Clinical Genome Resource reviews were available for four or five conditions. All four 'actionability' ratings disagreed with the findings of our reviews. Our review of 14 genomic studies of newborn screening cohorts found insufficient information to allow individual highly penetrant pathogenic variants for any condition to be identified. None of the 86 economic evaluations of whole genome sequencing or whole exome sequencing were set in a screening context. Some micro-costing studies are available that could help understand the resource use and costs associated with whole genome sequencing. Following a series of patient and public involvement meetings, attendees appreciated the uncertainties of whole genome sequencing. A wider stakeholder perspective is needed to inform policy decisions.
    Limitations: Although we only examined five conditions in depth, the consistency in lack of data suggests that our conclusions are robust.
    Conclusions: The systematic review approach for evaluating whole genome sequencing of newborns identified a paucity of high-quality evidence. Extending the review to all 200 conditions is not feasible. Currently, the use of existing genome resources and review of genomic studies of newborn screening cohorts are not viable alternatives. The cost-effectiveness of whole genome sequencing in a newborn screening context is unknown.
    Future work: Large-scale collaborative research is required to evaluate the short- and long-term harms, benefits and economic implications of whole genome sequencing for screening newborns. We propose a staged approach to evaluation, considering only conditions with pathogenic variants with high penetrance to minimise harm from overdiagnosis.
    Study registration: This study is registered as PROSPERO CRD42023475529.
    Funding: This award was funded by the National Institute for Health and Care Research (NIHR) Evidence Synthesis Programme (NIHR award ref: NIHR159928) and is published in full in Health Technology Assessment; Vol. 29, No. 65. See the NIHR Funding and Awards website for further award information.
    Keywords:  HUMANS; INFANT; NEONATAL SCREENING; NEWBORN; PATIENT ADVOCACY; PENETRANCE; POLICY; WHOLE GENOME SEQUENCING
    DOI:  https://doi.org/10.3310/DJRF1124
  31. Front Med Technol. 2025 ;7 1654003
      Nanoparticle-based drug delivery systems, such as liposomes, polymeric micelles, dendrimers, and other nanosized carriers, have emerged as promising strategies to improve the targeted delivery of therapeutic agents to the brain. These nanoparticles can be engineered to encapsulate drugs, facilitating their passage across the BBB, enabling localized treatment of the regions affected by neurodegeneration. Nanoparticles are characterized by their small size, large surface area, and possibility of functionalization, which allows them to be useful in many areas, including improved bioavailability, decreased systemic side effects, and improved therapeutic efficacy. Additionally, nanoparticles may also be surface-modified with appropriate ligands like antibodies, peptides, or small molecules, which exhibit specific interactions with receptors or cellular targets associated with the disease process. Such targeting has the potential to make targeted drug delivery possible, allowing therapeutic factors that can damage the healthy part of the brain to be delivered only to the diseased region. Furthermore, probable treatments for neurodegenerative diseases are also reviewed with the potential for complexation of different therapeutic agents, including small molecules, proteins, RNA, lipid nanoparticles and gene therapies with nanoparticle-based systems.
    Keywords:  CNS therapeutics; blood-brain barrier; nanocrystals; nerve growth factor; neural stem cells; neurodegenerative diseases
    DOI:  https://doi.org/10.3389/fmedt.2025.1654003
  32. Neurobiol Dis. 2025 Dec 06. pii: S0969-9961(25)00436-X. [Epub ahead of print]218 107219
      Autism spectrum disorder (ASD) is heterogeneous at every level, from behavior to molecular pathways, limiting the value of subgrouping schemes built on surface phenotypes alone. We synthesize evidence that biologically anchored subtypes, defined by convergent genetics, developmental timing, and brain-body crosstalk, offer a tractable path to precision medicine. Leveraging advances in large-scale genomic resources and computational analytics, we propose a multi-axis framework: (i) genetic architecture spanning rare variants and polygenic load, (ii) developmental windows from mid-gestation to infancy divergence and regression, and (iii) brain-body interactions shaping plasticity and symptom expression. This framework enables mechanism-guided therapeutic strategies through biomarker-stratified enrollment, target-engagement readouts, and circuit-anchored outcomes. Preclinical platforms, genetically engineered mice and patient-derived induced pluripotent stem cells (iPSCs), demonstrate convergence onto limited synaptic and connectivity "neurotypes," enabling causal links from gene to circuit to behavior and proof-of-concept rescue. We close with priorities: standardized multi-platform characterization, decision tools linking subtype labels to interventions, and stratified trials that co-report clinical and biological endpoints, with ethical guardrails to ensure early stratification expands opportunity while advancing individualized care.
    Keywords:  Autism spectrum disorder; Genetic architecture; Heterogeneity; Neural circuits; Precision medicine; Translational neuroscience
    DOI:  https://doi.org/10.1016/j.nbd.2025.107219
  33. Int J Mol Sci. 2025 Nov 23. pii: 11322. [Epub ahead of print]26(23):
      Sleep disturbances and liver diseases have a bidirectional relationship. Unhealthy sleep habits promote liver diseases, such as steatotic liver disease, and impact the prognosis, promoting progression to liver cirrhosis and liver-related mortality. Sleep accounts for 20% of the association between lifestyle and steatotic liver disease, indirectly by promoting obesity and metabolic syndrome and through direct effects in the liver. Conversely, liver diseases can affect sleep. Patients with liver cirrhosis complain of sleep disturbances five times more than the general population, with a profound impact on their quality of life. Common drugs used to treat sleep disorders, such as hypnotics and benzodiazepines, must be used very carefully in patients with cirrhosis due to altered hepatic metabolism and the potential to induce hepatic encephalopathy, making sleep disorders particularly challenging to manage in these patients. This review summarizes the available knowledge on the interplay between sleep and liver diseases.
    Keywords:  MASLD; cirrhosis; liver disease; sleep disorders
    DOI:  https://doi.org/10.3390/ijms262311322
  34. FASEB J. 2025 Dec 31. 39(24): e71340
      Mitochondrial dysfunction is central to the pathogenesis of podocytopathies, yet the determinants of metabolic resilience versus failure remain elusive. We investigated how distinct disruptions of mitochondrial architecture, specifically hyperfusion via OMA1 deletion versus compromised inner mitochondrial membrane (IMM) integrity via PHB2 knockdown, influence the metabolic fate and insulin responsiveness of podocytes. To this end, we analyzed conditionally immortalized mouse podocytes with genetic OMA1 deletion or inducible PHB2 knockdown and employed an integrated approach combining bioenergetic studies, quantitative proteomics, phosphoproteomics, metabolomics, and stable isotope tracing studies with 13C6-glucose and 13C5-glutamine. We characterized metabolic remodeling at baseline and after insulin treatment and uncovered profoundly divergent metabolic states. OMA1 deficiency conferred robust metabolic resilience, characterized by a compensatory glycolytic shift and remodeling of TCA cycle flux through glutamine-driven anaplerosis while maintaining oxidative phosphorylation. OMA1-deficient podocytes sustained bioenergetic homeostasis upon insulin challenge by flexibly rerouting carbon flux, including the GABA shunt. In contrast, PHB2 deficiency led to metabolic failure, impaired respiration, and anaplerotic insufficiency. While maintaining basal ATP levels at baseline, PHB2-deficient podocytes exhibited energetic collapse upon insulin treatment, revealing profound metabolic inflexibility. Taken together, the structural integrity of the inner mitochondrial membrane, rather than mitochondrial morphology per se, is a driving determinant of metabolic competence and resilience in podocytes.
    Keywords:  OMA1; PHB2; anaplerosis; glycolysis; insulin signaling; metabolism; mitochondria; podocytes
    DOI:  https://doi.org/10.1096/fj.202502934R
  35. Saudi J Ophthalmol. 2025 Oct-Dec;39(4):39(4): 416-418
      Mitochondrial disorders, particularly those associated with the m.3243A>G mutation in the MT-TL1 gene, can manifest with diverse systemic and ocular features, including mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) and maternally inherited diabetes and deafness. Retinal involvement often presents as macular pattern dystrophy. A 65-year-old female with a known history of mitochondrial disease (m.3243A>G mutation) presented for evaluation of retinal findings. She had asymptomatic diabetes and deafness, with visual acuity measured at 0.12 bilaterally. Clinical examination revealed clear corneas, nonsignificant cataracts, and fundoscopic findings of patchy retinal and parafoveal atrophy with preserved foveal regions. Optical coherence tomography indicated a preserved fovea, but thinning of perifoveal layers. The findings suggest retinal dystrophy indicative of mitochondrial retinopathy, characterized by macular pattern dystrophy associated with the m.3243A>G mutation. Given the potential for varied clinical presentations linked to this mutation, multidisciplinary evaluations are essential to assess systemic involvement and facilitate appropriate management. This case underscores the importance of recognizing retinal manifestations in patients with mitochondrial disorders, particularly in those with the m.3243A>G mutation, and highlights the need for comprehensive monitoring and care.
    Keywords:  Deafness; diabetes; genetics; mitochondrial diseases; retinopathy
    DOI:  https://doi.org/10.4103/sjopt.sjopt_314_24
  36. eNeuro. 2025 Dec 12. pii: ENEURO.0436-25.2025. [Epub ahead of print]
      Autism spectrum disorder, schizophrenia, and bipolar disorder are neuropsychiatric conditions that manifest early in life with a wide range of phenotypes, including repetitive behavior, agitation, and anxiety (American Psychological Association, 2013). While the etiology of these disorders is incompletely understood, recent data implicate a role for mitochondrial dysfunction (Norkett et al., 2017; Khaliulin et al., 2025). Mitochondria dynamically translocate to intracellular compartments to support energetics and free-radical buffering; failure to achieve this localization results in cellular dysfunction (Picard et al., 2016). Mitochondrial Rho-GTPase 1 (Miro1) resides on the outer mitochondrial membrane and facilitates microtubule-mediated mitochondrial motility and homeostasis (Fransson et al., 2003). The loss of MIRO1 is reported to contribute to the onset/progression of neurodegenerative diseases, including amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease (Kay et al., 2018). We have hypothesized that MIRO1 also has a role in nervous system development and function (Lin-Hendel et al., 2016). To test this, we ablated Miro1 from cortical excitatory progenitors by crossing floxed Miro1 mice with Emx1-Cre mice and used mice of either sex for experiments. We found that mitochondrial mis-localization in migrating excitatory neurons was associated with reduced brain weight, decreased cortical volume, and subtle cortical disorganization. Adult Miro1 conditional mutants exhibit agitative-like behaviors, including decreased nesting behavior and abnormal home cage activity. The mice exhibited anxiety-like behavior and avoided confined spaces, features that have been linked to several human behavioral disorders. Our data link MIRO1 function with mitochondrial dynamics in the pathogenesis of several neuropsychiatric disorders and implicate intracellular mitochondrial dynamics to some anxiety-like behaviors.Significance Statement Neuropsychological disorders such as autism spectrum disorder, schizophrenia, and bipolar disorder have overlapping endophenotypes. While the mechanisms underlying these disorders are poorly understood, recent evidence implicates mitochondrial dysfunction and cellular mis-localization playing a role. Mitochondria support energy requirements and other physiological functions in cells. Previous research from our lab has shown distinct dynamic localization patterns within migrating excitatory and inhibitory neurons during development. To further examine the importance of mitochondrial localization, we ablated MIRO1, a protein important for coupling mitochondria to motor proteins, in excitatory neurons. The mis-localization of mitochondria in migrating excitatory neurons is associated with diminished motor skills and anxiety-like behavior in post-natal mice.
    DOI:  https://doi.org/10.1523/ENEURO.0436-25.2025
  37. NEJM AI. 2025 Nov 24. pii: AIp2500802. [Epub ahead of print]2(12):
      The Orphan Drug Act defines a rare disease as a condition affecting fewer than 200,000 people in the United States. However, most rare diseases are categorized as ultrarare or hyper-rare, impacting fewer than 100 individuals worldwide. Developing drugs for these conditions involves multiple challenges, such as geographically dispersed and small patient populations, limited natural history data, and poor disease characterization. Issues related to small patient numbers, scarce natural history information, and clinical heterogeneity within rare diseases can be addressed by various strategies, including using artificial intelligence and advanced analytical methods, leveraging detailed individual-level data, and exploring synthetic data generation to overcome the limitations of small datasets. Moreover, establishing centralized databases and promoting public-private partnerships can help build a more comprehensive repository of available data.
    DOI:  https://doi.org/10.1056/AIp2500802
  38. J Hematol Oncol. 2025 Dec 10.
      Mitochondria, the powerhouse of the cell, orchestrate a plethora of critical functions, including energy production, metabolic regulation, programmed cell death, and signal transduction. Their pivotal role in the pathogenesis of numerous diseases underscores their significance. Among the various regulatory mechanisms, RNA modifications emerge as a dominant posttranscriptional modulator of gene expression, increasingly recognized for their profound impact on mitochondrial functions. Groundbreaking discoveries have unveiled compelling links between RNA modifications and oxidative phosphorylation, regulated cell death-particularly cuproptosis-and antitumor immunity, underscoring RNA modifications' vital role and untapped potential in mitochondrial biology, cancers and aging-related diseases. In this Review, we comprehensively catalog the primary RNA modifications modifiers and their small-molecule inhibitors that influence mitochondrial functions. We explore the latest research delineating RNA modifications' involvement in mitochondria-related glucose metabolism, regulated cell death, and mitochondrial dynamics, presenting an intricate regulatory network. Furthermore, we investigate the intriguing intersection of RNA modifications and mitochondria-related antitumor immunity, highlighting prospective therapeutic targets to enhance immunotherapy outcomes. This review not only accentuates the critical importance of RNA modifications in mitochondrial function but also paves the way for novel therapeutic strategies in disease treatment.
    Keywords:  Aging-related disease; Cancer; Drug resistance; Glucose metabolism; Mitochondria; Mitochondrial dynamics; RNA modifications; Regulated cell death; Tumor microenvironment
    DOI:  https://doi.org/10.1186/s13045-025-01762-7
  39. Biochim Biophys Acta Rev Cancer. 2025 Dec 06. pii: S0304-419X(25)00252-5. [Epub ahead of print]1881(1): 189510
      Mitochondria continuously alternate between fragmented and fused states, a process known as mitochondrial dynamics, which plays a pivotal role in essential cellular functions, including metabolism, apoptosis, reactive oxygen species production, and signal transduction. Disruptions in this dynamic equilibrium, frequently observed in aggressive cancers, can promote malignant transformation and tumor progression. A growing body of evidence indicates that dysregulated mitochondrial dynamics contribute to resistance against both conventional and targeted anticancer therapies. In this review, we explore the regulatory mechanisms governing mitochondrial dynamics, with a focus on the genetic and epigenetic modulation of key drivers such as DRP1, MFN1/2 and OPA1. We also discuss how altered mitochondrial dynamics converge into diverse mechanisms of drug resistance in cancer. Overall, these insights underscore aberrant mitochondrial dynamics as a potential biomarker of therapeutic resistance, and position mitochondrial dynamics-related GTPases, particularly DRP1 and Mitofusins, as exploitable targets for novel treatments in advanced solid and hematologic malignancies.
    Keywords:  Cancer therapy; DRP1; Drug resistance; MFF; MFN2; Mitochondrial dynamics
    DOI:  https://doi.org/10.1016/j.bbcan.2025.189510
  40. Int J Biol Macromol. 2025 Dec 09. pii: S0141-8130(25)10149-9. [Epub ahead of print] 149592
      Human manganese superoxide dismutase (MnSOD2) is a critical mitochondrial antioxidant that catalyzes the conversion of highly reactive superoxide radicals into molecular oxygen and hydrogen peroxide. The peroxide molecules are subsequently neutralized by other antioxidant systems, positioning MnSOD2 as the primary defense against mitochondrial oxidative stress and diseases associated with disrupted in vivo redox balance. MnSOD2 has been studied since its discovery in the early 1960s, particularly in the context of cellular pathology and as a therapeutic target. Recent studies combining neutron protein crystallography (NPC), X-ray absorption spectroscopy (XAS), and quantum mechanical (QM) computations have uncovered previously uncharacterized protonation states and atypically short and strong hydrogen bonds within the active site of MnSOD2. Together, these drive the enzyme's exceptionally rapid turnover. This focused review summarizes emerging insights to generate an updated landscape of MnSOD2's structure-function relationship and to highlight remaining challenges. The primary bottleneck to a complete understanding of the structural mechanism of MnSOD2 catalysis is the lack of a superoxide-bound MnSOD2 structure that resolves all proton positions, defines the redox state of the catalytic metal, the metal ligands, and the position of superoxide. Additionally, another largely unexplored area is how Fe substitution converts MnSOD2 into a peroxidase, and how this metal promiscuity affects mitochondrial redox homeostasis. This review synthesizes current evidence and states an informed hypothesis for the catalytic mechanism of Fe-substituted SOD2 (FeSOD2). Clarifying these gaps will advance our understanding of the structural basis of SOD2 catalysis and how it shapes mitochondrial redox biology in health and disease.
    Keywords:  Iron superoxide dismutase (FeSOD2); Manganese superoxide dismutase (MnSOD2); Metalloenzyme; Mitochondrial antioxidants; Oxidative stress; Proton- coupled Electron transfer (PCET); Redox balance
    DOI:  https://doi.org/10.1016/j.ijbiomac.2025.149592
  41. Mol Neurobiol. 2025 Dec 09. 63(1): 273
      The central nervous system (CNS) relies on tightly regulated barriers to maintain homeostasis and protect neural tissue from blood-borne toxins, pathogens, and inflammatory mediators. Tight junctions (TJs) are critical components of the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB), forming selective paracellular seals that regulate molecular trafficking. These structures comprise transmembrane proteins and cytoplasmic scaffolding proteins, which anchor TJs to the actin cytoskeleton. The spatial organization and function of TJs are dynamically regulated by calcium-dependent signaling, phosphorylation events, and G-protein-mediated pathways, which govern their assembly, disassembly, and response to physiological and pathological stimuli. The integrity of TJ complexes is particularly vulnerable to disruption in neurological disorders. Dysregulation of key TJ proteins has been implicated in neurodegenerative diseases, neuroinflammation, and CNS injury, leading to barrier permeability defects that exacerbate disease progression. Emerging therapeutic strategies aim to modulate TJs to stabilize barrier integrity and to mitigate pathology. This review examines the molecular architecture and regulatory mechanisms of TJ complexes, their dysfunction in disease states, and the translational potential of targeting them for therapy. A detailed understanding of TJ dynamics is essential for developing strategies to restore barrier function in neurological disorders.
    Keywords:  Barrier permeability; Blood-brain barrier; Blood-spinal cord barrier; Neurovascular dysfunction; Tight junctions
    DOI:  https://doi.org/10.1007/s12035-025-05592-z
  42. Nanoscale. 2025 Dec 08.
      For therapeutics to reach the brain, the several administration routes available come with some disadvantages, with the primary biological obstacle being the blood-brain barrier (BBB), which is not easy to penetrate despite the sophisticated technologies which have been developed. In addition, reaching specific brain structures invokes additional challenges, entailing more complicated delivery strategies. Nose-to-brain (N2B) delivery or the intranasal (IN) administration route provides a less invasive alternative. With the wealth of knowledge available on N2B delivery of nanomedicines and biotherapeutics, there is an opportunity to synthesize the current literature, especially in terms of promising strategies to improve N2B delivery of nanomedicines, highlighting experimental evaluation and translational challenges. We also emphasized the latest advancements in experimental models for nasal delivery. Aiming to bridge the gap between bench research and clinical application, we reviewed the cases of insulin and oxytocin, two biotherapeutics with high clinical potential for CNS-related diseases, and explore how nanomedicine-based platforms can enhance their effectiveness. This review offers a roadmap for overcoming barriers and accelerating the clinical translation of N2B therapeutics.
    DOI:  https://doi.org/10.1039/d5nr02259b
  43. J Inflamm Res. 2025 ;18 16761-16771
      Schizophrenia, a complex psychiatric disorder, is increasingly understood to involve immune dysregulation intertwined with metabolic and mitochondrial dysfunction. Neuroinflammation, driven by microglial activation, aberrant cytokine signalling, and skewed T cell polarization, intersects with impaired cellular bioenergetics and oxidative stress. Metabolic and mitochondrial alterations, consistently observed in patients, may constitute both cause and consequence of immune imbalance, sustaining a pathological loop that links bioenergetic failure to neuroinflammation. The ketogenic diet (KD), a high-fat, very low-carbohydrate intervention has recently gained attention as a potential therapy for schizophrenia. Emerging clinical reports describe improvements in symptom burden, weight regulation, and sustained remission. However, this evidence remains preliminary and is limited to pilot studies and case series. Preclinical studies provide mechanistic evidence, demonstrating that KD and its primary ketone body, β-hydroxybutyrate, attenuate core pathological features including inflammation, synaptic pruning, mitochondrial dysfunction, T cell imbalances and epigenetic alterations. Mechanistically, KD reshapes immune balance by favoring regulatory T cell induction over T helper 17 cell polarization and dampening pro-inflammatory signalling. Further to this, it improves mitochondrial biogenesis, increases ATP yield and reduces reactive oxygens species through increased efficiency of ATP hydrolysis. Epigenetic regulation by multiple pathways provides an additional layer of transcriptional control that may sustain therapeutic benefits. By framing KD within the context of inflammation research, this review synthesises findings from clinical, preclinical and mechanistic studies to highlight its potential to address fundamental disease mechanisms.
    Keywords:  epigenetics; inflammation; ketogenic diet; metabolism; mitochondria; schizophrenia; β-hydroxybutyrate
    DOI:  https://doi.org/10.2147/JIR.S540859
  44. Int J Mol Sci. 2025 Dec 04. pii: 11736. [Epub ahead of print]26(23):
      Myasthenia gravis, chronic inflammatory demyelinating polyneuropathy, and idiopathic inflammatory myopathies are among the most widely recognized autoimmune neuromuscular disorders. Although they differ in clinical presentation, shared immunopathogenic mechanisms place them at a molecular crossroads. Evidence of overlapping pathways has led to the development of targeted strategies including complement inhibition, FcRn antagonism, B-cell depletion, and the CAR-T cell approach. In this review, we analyze current knowledge regarding pathogenic mechanisms and their link to immunotherapy, extensively outlining both similarities and distinctions. We further discuss existing challenges, including diagnostic limitations and refractory disease variants, how technological advances have already addressed some of these issues, and where further progress is still needed.
    Keywords:  B-cell depletion therapy; FcRn antagonists; autoantibodies; chronic inflammatory demyelinating polyneuropathy; complement inhibition; idiopathic inflammatory myopathies; myasthenia gravis; targeted immunotherapy
    DOI:  https://doi.org/10.3390/ijms262311736
  45. Nature. 2025 Dec 10.
      Mitochondrial protein import is required for maintaining organellar function1. Perturbations in this process are associated with various physiological and disease conditions2. Several stress responses, including the mitochondrial compromised protein import response (mitoCPR), combat damage caused by mitochondrial protein import defects2. However, how this defect is sensed remains largely unknown. Here we reveal that the conserved mitochondrial Hsp70 co-chaperone, Mge1, acts as a stress messenger in budding yeast. During mitochondrial stress, unimported Mge1 entered the nucleus and triggered the transcription of mitoCPR target genes. This was mediated by the interaction of Mge1 with the transcription factor Pdr3 on DNA regulatory elements. The mitochondrial targeting sequence of Mge1 was both sufficient and essential for mitoCPR induction, demonstrating that in addition to their roles in mitochondrial protein import, targeting sequences can also function as signalling molecules. As protein import defects are a common consequence of various types of mitochondrial damage3,4, these findings suggest a novel function for the targeting sequence of Mge1 as an indicator of mitochondrial health.
    DOI:  https://doi.org/10.1038/s41586-025-09834-x
  46. Cells. 2025 Dec 01. pii: 1900. [Epub ahead of print]14(23):
      Dendritic cells (DCs) are among the first immune cells to detect viral invasion and play a central role in initiating and shaping antiviral immune responses. Many innate and adaptive immune functions of DCs are regulated by cathepsins, proteolytic enzymes primarily found in acidic endolysosomal compartments. Different DC subsets exhibit distinct cathepsin expression patterns, influencing their functional capacities and interactions with viruses. In DCs, cathepsins contribute to virus sensing through innate receptors, regulate cytokine production and DC migration, and are essential for viral antigen degradation and loading onto MHC molecules for T-cell activation. Many viruses, however, have evolved mechanisms to alter cathepsin expression and activity, thereby subverting DC function and promoting their own persistence. Indeed, cathepsins can facilitate viral entry into DCs, promote viral replication, and support immune evasion strategies. In this review, we summarize recent advances in understanding the role of cathepsins in DC-virus interactions, emphasizing both how DCs exploit cathepsins to generate protective immune responses and how viruses manipulate cathepsin activity to their advantage. We particularly focus on clinically relevant viral pathogens, including HIV, influenza virus, hepatitis C virus, human cytomegalovirus, Ebola virus, and SARS-CoV-2, to illustrate the multifaceted influence of cathepsins on DC biology during viral infection.
    Keywords:  DC; DC-virus interaction; antigen presentation; cathepsins; cysteine cathepsins; dendritic cells; endolysosomal proteases; viral immune evasion
    DOI:  https://doi.org/10.3390/cells14231900
  47. Nat Rev Neurol. 2025 Dec 12.
      Vaccination is widely considered to be the pre-eminent public health achievement of modern history, but declining coverage resulting from vaccine hesitancy and from interruptions in immunization campaigns in geopolitically unstable regions threatens progress against vaccine-preventable diseases. The global burden of vaccine-preventable neurological diseases is substantial, resulting in acute and chronic complications as well as high case fatality rates. In recent years, outbreaks of dengue, poliomyelitis, measles, pertussis, meningococcal disease and Japanese encephalitis virus have been linked to lack of access to vaccines, overwhelmed health-care systems, misinformation and disinformation regarding vaccine safety, and gaps in vaccination coverage caused by environmental factors and geopolitical conflicts. Coordinated global strategies, including addressing barriers to vaccination and ensuring equitable access; targeted health education about vaccine benefits and risks; integration with other public services; and advances in next-generation vaccine technologies to tackle antimicrobial resistance and non-vaccine serotype replacement, will be crucial to prevent a resurgence of vaccine-preventable neurological diseases, especially in vulnerable populations, to maintain global health security.
    DOI:  https://doi.org/10.1038/s41582-025-01172-w
  48. Int J Mol Sci. 2025 Nov 22. pii: 11301. [Epub ahead of print]26(23):
      Bile acids, once considered mere digestive detergents, have emerged as multifaceted signaling molecules with systemic influence extending far beyond the gastrointestinal tract. Recent discoveries reveal their capacity to modulate immune responses, cross the blood-brain barrier, and interact with central nervous system (CNS) cells through their receptors. Neuroinflammation, a key driver of neurodegenerative and neuroimmune disorders, is increasingly linked to bile acid signaling pathways that regulate glial activation, cytokine production, and neuronal survival. This review compiles the current evidence connecting bile acids to CNS inflammation, highlighting mechanistic insights, disease-specific alterations, and the gut-microbiome-bile acid-brain axis. It also explores the therapeutic potential of bile acid derivatives and receptor modulators, as well as their emerging role as biomarkers in conditions such as Alzheimer's disease, multiple sclerosis, and hepatic encephalopathy. Despite promising advances, critical gaps remain, including the need for bile receptor mapping in human CNS cells, standardized CNS bile acid profiling, and longitudinal metabolomic studies. Bridging these gaps may unlock new strategies for targeting neuroinflammation through bile acid-immune crosstalk.
    Keywords:  Alzheimer’s disease; FXR; Huntington’s disease; Parkinson’s disease; TGR5; amyotrophic lateral sclerosis; bile acids; blood-brain barrier; multiple sclerosis; neurodegeneration; neuroinflammation
    DOI:  https://doi.org/10.3390/ijms262311301
  49. 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