bims-polgdi Biomed News
on POLG disease
Issue of 2026–01–11
thirty-two papers selected by
Luca Bolliger, lxBio



  1. Pharm Res. 2026 Jan 08.
       BACKGROUND: Mitochondria besides being the powerhouse of the cell are also involved in performing a multitude of critical cellular functions. Any failure in maintenance of these organelles is implicated in multiple human pathologies, including neurodegenerative disorders. Over the past two decades, significant efforts have been made to investigate the pharmacodynamic propensity of various potential compounds, which could be engaged as efficient therapeutic approach in modulating mitochondrial dynamics during neuronal dysfunctions.
    METHOD: This review comprehensively overviews the contribution of potential compounds that could be employed as mitochondrial medicine in reversing neurological pathologies, with special focus on their significant roles as: metabolic antioxidants, conjugated molecules for mitochondrial function modulation, mitochondrial targeted peptides, optogenetic based induction of the mitochondria, potential mitochondrial biomarkers and other advanced transportation systems for mitochondrial delivery to brain.
    RESULTS AND DISCUSSION: The manuscript discusses the mechanism of action of potential compounds (natural and pharmacologically synthesized), and other advance approaches that could efficiently modulate mitochondrial machinery in terms of regulating mitochondrial biogenesis, mitophagy, bioenergetics pathways, oxidative stress, calcium homeostasis and mitochondrial DNA stability.
    CONCLUSION: The optimal maintenance of mitochondrial dynamics offered by variety of mitochondria targeting compounds highlights their prospective value for considering them as futuristic neurotherapeutic agents, which could be considered in managing neurodegenerative conditions.
    Keywords:  antioxidants; mitochondria; mitochondrial dynamics; neuroprotection; pharmaceutics
    DOI:  https://doi.org/10.1007/s11095-025-04004-0
  2. Front Biosci (Schol Ed). 2025 Dec 18. 17(4): 45427
      Leigh syndrome (LS), first reported in 1951, is the most common primary mitochondrial disease. The overarching term, Leigh Syndrome Spectrum (LSS) was proposed by a ClinGen Expert Panel to encompass the wide continuum of neurodegenerative and non-neurologic manifestations which were associated with classic LS and Leigh-Like Syndrome (LLS). Notably, LSS typically presents developmental regression or delay by two years of age, with about 20% of cases presenting as late-/adult-onset forms after 2 years. Historically defined by clinical, biochemical, and neuropathological findings, the genetic basis of LSS has been elucidated through the use of Sanger and next-generation sequencing (NGS), resulting in the discovery of over 120 causative genes. Moreover, LSS can be caused by mutations in both nuclear-encoded genes and mitochondrial DNA (mtDNA), with overlapping clinical characteristics that occur at similar frequencies. This review aims to summarize the clinical and onset characteristics of LSS, genetic testing-aided diagnosis criteria, and the development of treatments. Furthermore, this review organizes the years since the first reports of gene and mutation discoveries into four consecutive eras: Clinical-Biochemical Era (1990-1999), Early Genomics Era (2000-2009), NGS Revolution Era (2010-2019), and Modern Era (2020-Present). Thus, using this framework, this review chronicles the evolution of LSS molecular genetics and treatment development, highlighting the shift from supportive care to targeted therapies driven by modern technologies. Cornerstone experimental models, such as the Ndufs4 -/-knockout mouse and patient-derived induced pluripotent stem cells (iPSCs), have facilitated mechanistic studies and drug repurposing screens, including the identification of sildenafil as a potential therapeutic agent, which has led to medical improvements in patients. Current advances in gene editing, including mitochondrial single-base editors such as eTd-mtABE and mitoBEs, are enabling gene therapy with precise introduction and correction of LS-causing variants in rat and mouse models. On the preventative front, Mitochondrial Replacement Therapy (MRT), guided by precise maternal mtDNA genotyping, has been successfully applied in clinical practice, allowing mothers carrying LSS-causing mtDNA variants to have healthy babies free of the LS manifestation. Collectively, these advances in gene discovery, genetic diagnosis, sophisticated disease modeling, rapid screening of small molecule drugs, precise gene editing for gene therapy, and innovative treatment strategies, such as MRT, are ushering in an era of precision medicine for LSS.
    Keywords:  Leigh Syndrome (LS); Leigh Syndrome Spectrum (LSS); Ndufs4-/- knockout mouse; gene therapy; mitochondrial DNA (mtDNA); mitochondrial replacement therapy (MRT); precision medicine; targeted therapy
    DOI:  https://doi.org/10.31083/FBS45427
  3. Ann Indian Acad Neurol. 2026 Jan 08.
       ABSTRACT: Mitochondrial DNA maintenance defects (MDMD) are rare genetic disorders that typically present in infancy but can manifest later with multi-organ involvement. We describe four MDMD cases (age 19-25) with distinct clinical and genetic profiles and delayed diagnosis. Two patients with mitochondrial neurogastrointestinal encephalomyopathy (homozygous TYMP variants: c.454G>T, c.866A>C) exhibited cachexia, ptosis, neuropathy, and confluent white matter hyperintensities leukodystrophy. Two others with MPV17 (c.293C>T) presented with neuromyopathy and hepatosplenomegaly; one showed novel concentric ring lesions on magnetic resonance imaging (MRI). Despite severe white matter changes/leukodystrophy, cognition was preserved. Diagnoses were delayed due to atypical gastrointestinal or neuromuscular symptoms. This series highlights the diagnostic challenge of MDMD and underscores that it should be considered in adolescents or young adults with unexplained neuropathy, white matter hyperintensities/leukodystrophy, or cachexia, even without classic hepatic or encephalopathic features. Genetic testing is essential for diagnosis, as phenotypic variability often obscures underlying MDMD. Our findings underscore the need for increased awareness of this delayed-diagnosis presentation to enable timely intervention.
    Keywords:  MPV17-related mitochondrial disease; Mitochondrial DNA maintenance defects (MDMD); mitochondrial neurogastrointestinal encephalomyopathy
    DOI:  https://doi.org/10.4103/aian.aian_843_25
  4. Biogerontology. 2026 Jan 06. 27(1): 33
      Chronic stress has been linked to mitochondrial dysfunction and impaired telomere maintenance, yet the mechanistic relationships connecting these pathways in humans remain poorly resolved. Using longitudinal findings from the Guillén-Parra cohort as a motivating human example, this Perspective offers a reinterpreted framework that proposes a unifying energetic interpretation in which bioenergetic insufficiency-defined as a mismatch between stress-induced energetic demand and mitochondrial throughout-rather than accumulated molecular damage, forms the upstream constraint linking stress physiology, mitochondrial performance, and telomerase regulation. In this cohort, lower baseline mitochondrial energetic capacity predicted greater longitudinal declines in telomerase activity, while telomere length remained stable across the short observation window, supporting the view that telomerase activity represents an early, energy-sensitive marker of unresolved stress adaptation, whereas telomere shortening is a delayed structural consequence. Interpreted within the Exposure-Related Malnutrition (ERM) framework, these patterns suggest that repeated activation of stress-response pathways without adequate metabolic recovery limits mitochondrial throughput and progressively compromises genome maintenance. In contrast, repeated exposure to mild stressors followed by sufficient recovery promotes adaptive strengthening of mitochondrial function and telomeric maintenance, consistent with physiological hormesis. We outline a roadmap integrating telomerase activity with dynamic indices of mitochondrial and redox function, including NAD⁺ availability, and emerging biomarkers of systemic energetic strain, such as circulating cell-free mitochondrial DNA and GDF15. By reframing aging phenotypes as early-stage failures of energetic resolution, this model highlights modifiable windows of vulnerability and hormesis-informed strategies-including exercise-induced adaptive stress, circadian alignment, and nutritional sufficiency-as actionable pathways for preserving mitochondrial resilience and telomere maintenance.
    Keywords:  Bioenergetic stress; Cellular senescence; Mitochondrial energetics; Psychological stress; Telomerase activity
    DOI:  https://doi.org/10.1007/s10522-025-10377-x
  5. Ann Med Surg (Lond). 2026 Jan;88(1): 1019-1020
      Kearns-Sayre syndrome (KSS) is a rare mitochondrial disorder defined by a combination of ophthalmoplegia, pigmentary retinopathy, and cardiac conduction defects. KSS arises from mitochondrial DNA (mtDNA) deletions and heteroplasmic imbalance, where there is a variation in levels of normal versus abnormal mtDNA. Current therapies offer symptomatic relief at most; they do not address the primary issue of correcting the genetic mutation. Innovative methods employing CRISPR Prime Editing (PE), an accurate and RNA-less technology, allow for unique correction of pathogenic mtDNA variants and errors. By fixing the wild type to variant ratio, PE could directly correct ocular- and cardiac-related signs and symptoms in KSS, in two tissue types that are entirely dependent on mitochondrial bioenergetics for their energy needs. Furthermore, the use of tissue-specific delivery methods, such as AAV2 vectors or cardiomyocyte promoters, would further enhance the targeting of the corrective approach to more specifically correct disease processes. This represents a completely innovative approach to genomic correction in the field of mitochondrial medicine, and there is important to translate this research to the clinic.
    DOI:  https://doi.org/10.1097/MS9.0000000000004366
  6. Cytoskeleton (Hoboken). 2026 Jan 08.
      Mitochondrial dysfunction and cytoskeletal disorganization are widely recognized hallmarks of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). Although these disorders differ in clinical presentation and etiology, accumulating evidence points to a shared cellular vulnerability at the intersection of mitochondrial dynamics and actin cytoskeletal regulation. In this review, we examine the emerging role of actin-mitochondria crosstalk as a convergent mechanism in neurodegeneration. We discuss how disruptions in actin filament remodeling, mitochondrial fission and fusion, organelle transport, and mitophagy contribute to neuronal dysfunction and loss across these diseases. Particular attention is given to disease-specific pathways, including cofilin-actin rod formation in AD, α-synuclein-driven actin disruption in PD, mutant huntingtin's effects on mitochondrial fragmentation in HD, and profilin-1-associated mitochondrial defects in ALS. By synthesizing findings from diverse models, we highlight how perturbations in the cytoskeleton-mitochondria interface may act as an upstream trigger and amplifier of neurodegenerative cascades. We also outline key knowledge gaps and propose future directions for research, with an emphasis on targeting actin-mitochondrial interactions as a potential therapeutic strategy across multiple neurodegenerative conditions.
    Keywords:  actin cytoskeleton; mitochondria dysfunction; mitochondria‐cytoskeleton crosstalk; neurodegeneration
    DOI:  https://doi.org/10.1002/cm.70095
  7. Neurol Genet. 2026 Feb;12(1): e200343
       Background and Objectives: The aim of this study was to profile immune cell mitochondrial phenotypes in mitochondrial diseases (MitoD) and evaluate how these phenotypes relate to disease manifestations or biomarkers.
    Methods: We profiled mitochondrial content and oxidative phosphorylation (OxPhos) enzymatic activities in isolated monocytes, lymphocytes, neutrophils, platelets, and mixed peripheral blood mononuclear cells (PBMCs) from 37 individuals with MitoD (m.3243A > G, n = 23; single, large-scale mitochondrial DNA (mtDNA) deletions, n = 14) and 68 healthy women and men from the Mitochondrial Stress, Brain Imaging, and Epigenetics study.
    Results: We first confirmed and quantified robust cell type differences in mitochondrial content; activities of OxPhos complexes I, II, and IV; and the mitochondrial respiratory capacity (MRC) index. In relation to MitoD, neither mitochondrial content nor OxPhos capacity was consistently affected, other than a mild monocyte-specific reduction in complex I (partially mtDNA encoded) relative to complex II (entirely nDNA encoded), consistent with the mtDNA defects examined. Relative to the large differences in cell type-specific mitochondrial phenotypes, differences in MitoD relative to controls were generally small (<25%) across mitochondrial measures. MitoD biomarkers growth differentiation factor 15 and fibroblast growth factor 21, as well as clinical disease severity measures, were most strongly related to mitochondrial abnormalities in platelets, and most weakly related to mitochondrial OxPhos capacity in lymphocytes, which are known to eliminate mtDNA defects. Finally, comparing PBMCs collected in the morning/fasted state with those in the afternoon/fed state after a stressful experience, we report significant time-dependent changes in mitochondrial biology over hours.
    Conclusions: Overall, these results demonstrate that the dynamic and cell type-specific mitochondrial phenotypes are preserved in MitoD and are generally unrelated to symptom severity.
    DOI:  https://doi.org/10.1212/NXG.0000000000200343
  8. Biosci Rep. 2026 Jan 09. pii: BSR20254097. [Epub ahead of print]46(1):
      The maintenance of proteostasis is essential for cellular function and organismal health. Its decline with age is a key contributor to neurodegenerative diseases, metabolic disorders, and other chronic conditions. Eukaryotic cells respond to proteotoxic stress through compartment-specific pathways, including the heat shock response (HSR), the unfolded protein response of the endoplasmic reticulum (UPRER), and the mitochondrial UPR (UPRmt). While these pathways have been extensively studied in cell-autonomous contexts, recent evidence reveals that neurons and glial cells can co-ordinate these responses across tissues through cell-non-autonomous mechanisms. Neuronal signals, including neuropeptides, biogenic amines, and possibly extracellular vesicles, can activate stress responses in distal cells, modulating lipid metabolism and impacting longevity. Emerging data also suggest a role for glial cells in systemic proteostasis regulation, though their mechanisms remain relatively uncharacterized. This review discusses both classical and emerging concepts of proteostasis stress-response pathways, their integration with neural signaling, and how their modulation influences aging and disease. Understanding how intercellular communication governs proteostasis could open new avenues for therapeutic interventions in age-related and neurodegenerative disorders.
    Keywords:  aging; cell-non-autonomous signaling; heat shock response (HSR); proteostasis network; unfolded protein response (UPR)
    DOI:  https://doi.org/10.1042/BSR20254097
  9. J Mol Biol. 2026 Jan 02. pii: S0022-2836(25)00684-9. [Epub ahead of print] 169618
      As a professor of biomedicine in the School of Life Sciences at East China Normal University (ECNU), I am dedicated to developing advanced genome editing technologies for disease modeling and precise gene therapy. My foundational training at Hunan Normal University and Texas A&M University cultivated a deep interest in using engineered cellular and animal models to understand human diseases. Since 2013, my laboratory at ECNU has pioneered the use of TALEN and CRISPR/Cas9 for the rapid generation of knockout mouse and rat models for disease studies. Once stepped in genome editing field, I shifted my focus to advancing editing tools and developing gene therapy strategies for genetic disorders and cancer. My team has developed a suite of high-performance base editors for nuclear DNA, mitochondrial DNA, and RNA, broadening editing capabilities while enhancing precision and safety. Leveraging these technologies, we have designed several therapeutic strategies that have shown efficacy in cellular and animal models of genetic diseases. Through collaborative efforts, we have successfully translated genome editing into clinical applications, contributing to the treatment of patients with β-thalassemia. Additionally, we have developed a non-viral, site-specific CAR-T platform for lymphoma therapy. Looking forward, I aim to develop the next generation of long-fragment, site-specific integration technologies and accelerate clinical translation to bring transformative cures to more patients.
    Keywords:  CRISPR/Cas9; Cell Therapy; Gene Editing; Gene Therapy; TALEN
    DOI:  https://doi.org/10.1016/j.jmb.2025.169618
  10. Int J Mol Sci. 2026 Jan 02. pii: 485. [Epub ahead of print]27(1):
      Mitochondrial dysfunction represents a central hallmark of aging and a broad spectrum of chronic diseases, ranging from metabolic to neurodegenerative and ocular disorders. Nicotinamide riboside (NR), a vitamin B3 derivative and efficient precursor of NAD+ (nicotinamide adenine dinucleotide), and berberine (BBR), an isoquinoline alkaloid widely investigated in metabolic regulation, have independently emerged as promising mitochondrial modulators. NR enhances cellular NAD+ pools, thereby activating sirtuin-dependent pathways, stimulating PGC-1α-mediated mitochondrial biogenesis, and triggering the mitochondrial unfolded protein response (UPRmt). BBR, by contrast, primarily activates AMPK (AMP-activated protein kinase) and interacts with respiratory complex I, improving bioenergetics, reducing mitochondrial reactive oxygen species, and promoting mitophagy and organelle quality control. Importantly, despite distinct upstream mechanisms, NR and BBR converge on shared signaling pathways that support mitochondrial health, including redox balance, metabolic flexibility, and immunometabolic regulation. Unlike previous reviews addressing these compounds separately, this article integrates current preclinical and clinical findings to provide a unified perspective on their converging actions. We critically discuss translational opportunities as well as limitations, including heterogeneous clinical outcomes and the need for robust biomarkers of mitochondrial function. By outlining overlapping and complementary mechanisms, we highlight NR and BBR as rational combinatorial strategies to restore mitochondrial resilience. This integrative perspective may guide the design of next-generation clinical trials and advance precision approaches in mitochondrial medicine.
    Keywords:  NAD+ metabolism; berberine; cardiometabolic disease; mitochondrial dysfunction; neuroprotection; nicotinamide; nicotinamide riboside; oxidative stress; retinal ganglion cells
    DOI:  https://doi.org/10.3390/ijms27010485
  11. PLoS One. 2026 ;21(1): e0339326
      Mitochondrial bioenergetics are vital for ATP production and are associated with several diseases, including Parkinson's Disease (PD). Here, we simulated a computational model of mitochondrial ATP production to interrogate mitochondrial bioenergetics under physiological and pathophysiological conditions, and provide a data resource that can be used to interpret mitochondrial bioenergetics experiments. We first characterised the impact of several common electron transport chain (ETC) impairments on experimentally-observable bioenergetic parameters. We then established an analysis pipeline to integrate simulations with experimental data and predict the molecular defects underlying experimental bioenergetic phenotypes. We applied the pipeline to data from PD models. We verified that the impaired bioenergetic profile previously measured in Parkin knockout (KO) neurons can be explained by increased mitochondrial uncoupling. We then generated primary cortical neurons from a Pink1 KO mouse model of PD, and measured reduced oxygen consumption rate (OCR) capacity and increased resistance to Complex III inhibition. Here, our pipeline predicted that multiple impairments are required to explain this bioenergetic phenotype. Finally, we provide all simulated data as a user-friendly resource that can be used to interpret mitochondrial bioenergetics experiments, predict underlying molecular defects, and inform experimental design.
    DOI:  https://doi.org/10.1371/journal.pone.0339326
  12. J Inherit Metab Dis. 2026 Jan;49(1): e70139
      MRPS genes, which encode components of the small mitoribosomal subunit, have not been previously linked to adult-onset neurological diseases. These genes play a critical role in mitochondrial translation and the biogenesis of the oxidative phosphorylation system. Whole Genome Sequencing was performed on adult patients presenting with an unexplained neurological picture. In parallel, functional studies were carried out in patient-derived fibroblasts to assess mitochondrial translation and the status of oxidative phosphorylation pathways. Bi-allelic pathogenic variants in MRPS22, MRPS23, and MRPS34 were identified in four patients from unrelated families. All patients presented a similar complex neurological phenotype, including cerebellar ataxia, distal motor neuropathy, pyramidal syndrome, and a distinctive leukoencephalopathy on brain MRI. Additional findings included elevated cerebrospinal fluid (CSF) protein levels and profound cerebral folate deficiency. Functional analyses revealed impaired mitochondrial translation and multiple defects in oxidative phosphorylation. Treatment with oral folinic acid resulted in clinical stabilization, radiological improvement, and normalization of CSF 5-methyltetrahydrofolate levels. Our findings expand the spectrum of mitochondrial diseases caused by defects in mitoribosomal proteins, highlighting their role in adult-onset neurological disorders with distinctive brain imaging features, high CSF protein levels, and cerebral folate deficiency.
    Keywords:  cerebral folate deficiency; distal motor neuropathy; leukoencephalopathy; mitoribosome
    DOI:  https://doi.org/10.1002/jimd.70139
  13. Drug Dev Res. 2026 Feb;87(1): e70221
      Mitochondrial dysfunction critically underpins the pathogenesis of inflammatory skin diseases such as psoriasis, vitiligo, atopic dermatitis, and impaired wound healing. This comprehensive review synthesizes recent evidence to elucidate mechanisms, including compromised bioenergetics, excessive reactive oxygen species (ROS), mitochondrial DNA (mtDNA) damage, and aberrant mitochondrial dynamics. Distinct from prior work, this analysis uncovers novel findings: mtDNA acts as a damage-associated molecular pattern, activating cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathways to drive type I interferon in vitiligo and IL-17A in psoriasis; succinate-mediated immune-metabolic signaling amplifies type 2 inflammation in atopic dermatitis; and subclinical mitochondrial impairments in non-lesional skin serve as early indicators of disease susceptibility across these conditions. Preclinical studies have shown that emerging therapies, including antioxidants (e.g., NMN), mitochondrial modulators (e.g., SS31), senotherapeutics, and mitochondrial transplantation, are promising strategies for restoring cellular function. Future research should focus on multi-omics to dissect mitochondrial-epigenetic interactions, validate mitochondrial metabolites like succinate as diagnostic biomarkers, and explore synergistic combination therapies. This integrative framework of mitochondrial-driven pathology provides fresh perspectives to advance diagnostic and therapeutic innovation in dermatology.
    Keywords:  atopic dermatitis; emerging therapies; mitochondrial DNA (mtDNA); mitochondrial dysfunction; psoriasis; vitiligo
    DOI:  https://doi.org/10.1002/ddr.70221
  14. J Biomed Semantics. 2026 Jan 06.
       BACKGROUND: Around 30 million people in Europe are affected by a rare (or orphan) disease, defined as a condition occurring in fewer than 1 in 2,000 individuals. The primary challenge is to automatically and efficiently identify scientific articles and guidelines that address a particular rare disease. We present a novel methodology to annotate and index scientific text with taxonomical concepts describing rare diseases from the OrphaNet taxonomy. This task is complicated by several technical challenges, including the lack of sufficiently large, human-annotated datasets for supervised training and the polysemy/synonymy and surface-form variation of rare disease names, which can hinder any annotation engine.
    RESULTS: We introduce a framework that operationalizes OrphaNet for large-scale literature annotation by integrating the TERMite engine with curated synonym expansion, label normalization (including deprecated/renamed concepts), and fuzzy matching. On benchmark datasets, the approach achieves precision = 92%, recall = 75%, and F1 = 83%, outperforming an string-matching baseline. Applying the pipeline to Scopus produces disease-specific corpora suitable for bibliometric and scientometric analyses (e.g., institution, country, and subject-area profiles). These outputs power the Rare Diseases Monitor dashboard for exploring national and global research activity.
    CONCLUSION: To our knowledge, this is the first systematic, scalable semantic framework for annotating and indexing rare disease literature at scale. By operationalizing OrphaNet in an automated, reproducible pipeline and addressing data scarcity and lexical variability, the work advances biomedical semantics for rare diseases and enables disease-centric monitoring, evaluation, and discovery across the research landscape.
    Keywords:  Annotation; Bibliographic databases; Health sciences; Indexing; Natural language processing; Rare diseases; Research applications; Scientometrics
    DOI:  https://doi.org/10.1186/s13326-025-00346-1
  15. Mol Cell. 2026 Jan 08. pii: S1097-2765(25)01013-5. [Epub ahead of print]86(1): 6-8
      In this issue of Molecular Cell, Zhu et al.1 show that mitochondria of cancer cells rely on the import of glutamine not only to fuel metabolite synthesis via the tricarboxylic acid cycle but also to charge mt-tRNAGln to allow mitochondrial protein synthesis and respiration.
    DOI:  https://doi.org/10.1016/j.molcel.2025.12.014
  16. PLoS Genet. 2026 Jan 09. 22(1): e1011836
      The mitochondrial genome (mtDNA) encodes essential subunits of the electron transport chain and ATP synthase. Mutations in these genes impair oxidative phosphorylation, compromise mitochondrial ATP production and cellular energy supply, and can cause mitochondrial diseases. These consequences highlight the importance of mtDNA quality control (mtDNA-QC), the process by which cells selectively maintain intact mtDNA to preserve respiratory function. Here, we developed a high-throughput flow cytometry assay for Saccharomyces cerevisiae to track mtDNA segregation in cell populations derived from heteroplasmic zygotes, in which wild-type (WT) mtDNA is fluorescently labeled and mutant mtDNA remains unlabeled. Using this approach, we observe purifying selection against mtDNA lacking subunits of complex III (COB), complex IV (COX2) or the ATP synthase (ATP6), under fermentative conditions that do not require respiratory activity. By integrating cytometric data with growth assays, qPCR-based mtDNA copy-number measurements, and simulations, we find that the decline of mtDNAΔatp6 in populations derived from heteroplasmic zygotes is largely explained by the combination of its reduced mtDNA copy number-biasing zygotes toward higher contributions of intact mtDNA-and the proliferative disadvantage of cells carrying this variant. In contrast, the loss of mtDNAΔcob and mtDNAΔcox2 cannot be explained by growth defects and copy-number asymmetries alone, indicating an additional intracellular selection against these mutant genomes when intact mtDNA is present. In heteroplasmic cells containing both intact and mutant mtDNA, fluorescent reporters revealed local reductions in ATP levels and membrane potential ([Formula: see text]) near mutant genomes, indicating spatial heterogeneity in mitochondrial physiology that reflects local mtDNA quality. Disruption of the respiratory chain by deletion of nuclear-encoded subunits (RIP1, COX4) abolished these physiological gradients and impaired mtDNA-QC, suggesting that local bioenergetic differences are required for selective recognition. Together, our findings support a model in which yeast cells assess local respiratory function as a proxy for mtDNA integrity, enabling intracellular selection for functional mitochondrial genomes.
    DOI:  https://doi.org/10.1371/journal.pgen.1011836
  17. Ageing Res Rev. 2026 Jan 03. pii: S1568-1637(26)00001-2. [Epub ahead of print]115 103009
      Reprogramming of somatic cells into induced pluripotent stem cells through the introduction of transcription factors Oct3/4, Sox2, Klf4, and c-Myc (OSKM) represents a landmark advance in regenerative biology. Building on this foundation, partial reprogramming can help reset epigenetic age. It further opens opportunities to treat degenerative diseases without the tumorigenic risks associated with full pluripotency. The review advances the field in three ways: it links lineage-preserving partial reprogramming to quantifiable rejuvenation endpoints; defines mesenchymal drift as an age- and disease-associated trajectory amenable to reversal; and maps strategies beyond OSKM, including small-molecule programs and CRISPR-based control circuits. Convergent phenotypes are surveyed in nervous, metabolic, musculoskeletal, and craniofacial systems, with emphasis on improved tissue repair and regeneration. A translational checklist is proposed that emphasizes schedule, delivery, and safety pharmacology to guide rigorous preclinical studies and de-risk early clinical entry points for partial reprogramming therapies.
    Keywords:  Aging; Cellular plasticity; Epigenetic rejuvenation; Partial reprogramming; Regenerative medicine
    DOI:  https://doi.org/10.1016/j.arr.2026.103009
  18. iScience. 2026 Jan 16. 29(1): 114279
      Mitochondrial dysfunction has emerged as a key contributor to the pathogenesis of steroid-resistant nephrotic syndrome (SRNS) and genetic focal-segmental glomerulosclerosis (FSGS). This study explores the role of mitochondrial integrity in podocyte biology, focusing on the impact of OMA1, a critical regulator of mitochondrial morphology. Using a model of disrupted mitochondrial homeostasis, we show that mitochondrial dysfunction sensitizes podocytes to insulin, triggering the overactivation of mTOR signaling. Disruption of OMA1 function was achieved through the deletion of Oma1 or a podocyte-specific knockout of its regulator Phb2. Remarkably, simultaneous Oma1 deletion extended the lifespan of severely affected Phb2 pko mice, alleviated proteinuria, and restored mitochondrial morphology. Increased mTOR activity was observed in Phb2 pko , Oma1 del , and Phb2/Oma1 double-knockout mice. Our findings highlight the critical role of mitochondrial integrity in podocyte function and disease mitigation, providing potential therapeutic insights for mitochondrial dysfunction-associated nephropathies.
    Keywords:  cell biology
    DOI:  https://doi.org/10.1016/j.isci.2025.114279
  19. Platelets. 2026 Dec;37(1): 2610264
      Platelets, traditionally known for their role in hemostasis, also contribute to inflammation, cancer, and intercellular communication through the release of platelet-derived extracellular vesicles (or platelet-derived microparticles; PMPs). Among these vesicles, a subpopulation containing functional mitochondria, known as mitoMPs, can be transferred to recipient cells, thereby modulating their metabolism and biological responses. This mitochondrial transfer plays a key role in various pathological processes, where it may either restore metabolic functions or enhance cancer cell proliferation, survival, and metabolic plasticity. In this study, we developed a permeabilization protocol combined with high-resolution respirometry to assess mitochondrial respiration in both platelets and PMPs. First, we found that saponin was a more effective permeabilizing agent than digitonin to measure mitochondrial respiration in these models. Moreover, our analysis revealed distinct respiratory profiles between platelets and PMPs and demonstrated that freeze-thaw cycles severely compromise mitochondrial functions in PMPs. Additionally, we performed proteomic profiling of PMPs to characterize their protein cargo, which associate with specific molecular pathways, particularly those associated with mitochondrial metabolism. These results provide novel insights into the biological functions of PMPs and their potential involvement in disease processes. Together, these findings advance the understanding of PMP-mediated mitochondrial transfer and intercellular communication and establish a foundation for future biomedical and therapeutic investigations.
    Keywords:  High-resolution respirometry; OXPHOS; microparticles; mitochondria; platelets; proteomics
    DOI:  https://doi.org/10.1080/09537104.2025.2610264
  20. Int J Mol Sci. 2026 Jan 04. pii: 522. [Epub ahead of print]27(1):
      Aging is a multifactorial process marked by a progressive decline in physiological function and increased vulnerability to diseases such as neurodegeneration, cancer, cardiovascular disorders, and infections. A central feature of aging is inflammaging, a state of chronic low-grade inflammation driven by cellular senescence, mitochondrial dysfunction, and oxidative stress. Recently, two regulated forms of non-apoptotic cell death-ferroptosis and cuproptosis-have emerged as critical mechanisms linking redox imbalance, mitochondrial stress, and disrupted metal homeostasis to age-related pathology. Ferroptosis, an iron-dependent process characterized by lipid peroxidation and impaired glutathione peroxidase 4 (GPX4) activity, and cuproptosis, a copper-dependent mechanism associated with protein lipoylation stress, both intersect with aging-related changes in mitochondrial and metabolic function. Importantly, these two forms of cell death should not be viewed as entirely separate pathways but rather as interconnected axes within a broader metal-redox-metabolic network. Disturbances in copper or iron homeostasis, glutathione (GSH)/GPX4 dysfunction, mitochondrial and iron-sulfur (Fe-S) cluster compromise, and enhanced lipid peroxidation may converge to lower cellular survival thresholds, thereby exacerbating oxidative damage, immune dysfunction, and tissue degeneration and ultimately fueling aging and inflammaging. This review offers a unique integrated perspective that situates ferroptosis and cuproptosis within a unified framework of aging biology, emphasizing their roles in age-related diseases and the therapeutic potential of targeting these pathways through nutritional, pharmacological, and lifestyle interventions.
    Keywords:  Alzheimer’s disease; Parkinson’s disease; aging; cancer; cardiovascular disease; cuproptosis; curcumin; diet; ferroptosis; hormesis; infection; inflammaging; neurodegeneration; oxidative stress; polyphenols; polyunsaturated fatty acids (PUFAs)
    DOI:  https://doi.org/10.3390/ijms27010522
  21. Precis Nutr. 2025 Mar;pii: e00098. [Epub ahead of print]4(1):
       Background: Mitochondrial dysfunction has been linked with metabolic disorders, given the central role of mitochondria in multiple crucial metabolic processes. However, little is known about how maternal metabolic conditions may affect mitochondrial function in newborns, although several other prenatal exposures have been associated with impaired mitochondrial function in either the placenta or cord blood. This study aimed to assess the association between maternal metabolic conditions and mitochondrial DNA copy number (mtDNA-CN, as a biomarker for mitochondrial function) in newborns.
    Methods: Among 999 mother-newborn pairs from the Boston Birth Cohort, mtDNA-CN in newborn umbilical cord blood and maternal blood collected at delivery were measured by a targeted genome sequencing approach. Linear regressions were used to evaluate the association between maternal metabolic conditions (diabetes mellitus, pre-pregnancy obesity, and the combination of the two conditions) and mtDNA-CN Z-scores in cord blood. The models were initially adjusted for maternal and child demographic and lifestyle characteristics, followed by additional adjustment for maternal mtDNA-CN to explore its potential influence on the results.
    Results: In this sample, 23.6% of mothers had pre-pregnancy obesity, and 13.6% had diabetes. In models examining the two conditions separately, both maternal diabetes (Beta = 0.18, 95% CI: 0.00, 0.36, p = 0.054) and obesity (Beta = 0.10, 95% CI: -0.04, 0.25, p = 0.166) were associated with increased cord blood mtDNA-CN Z scores, but neither association reached statistical significance. When examined together, newborns whose mothers had both diabetes and pre-pregnancy obesity had statistically significantly higher cord mtDNA-CN Z scores (Beta = 0.35, 95% CI: 0.08, 0.63, p = 0.012) compared to newborns whose mothers did not have the two conditions. Further adjustment for maternal blood mtDNA-CN did not substantially alter the above associations.
    Conclusion: Maternal diabetes and obesity jointly were associated with higher levels of cord blood mtDNA-CN. Future studies should further assess whether cord blood mtDNA-CN explains the link between maternal metabolic conditions and offspring health outcomes.
  22. ACS Nano. 2026 Jan 08.
      Mitochondria play a central role in cellular energy metabolism, survival, and apoptosis, with their dysfunction implicated in numerous diseases, including neurodegenerative and age-related disorders. Modulating mitochondrial function therefore represents a promising therapeutic strategy. In this study, we demonstrate that high-frequency terahertz (THz) irradiation elicits frequency-specific effects on mitochondrial biogenesis. Through MitoTimer and MitoTracker assays, we observed that irradiation at 34.5 THz significantly enhanced mitochondrial biogenesis, an effect not observed at 36.1 THz. Electrophysiological and molecular analyses revealed that 34.5 THz irradiation elevates intracellular calcium flux and activates the calcium-mediated PGC-1α-NRF1/2-TFAM pathway, leading to increased cellular energy production and oxygen consumption. Computational modeling suggested a resonant coupling mechanism in which 34.5 THz irradiation interacts with the bending vibration of the glutamate C-C-C bond at the narrowest region of the calcium ion channel pore, thereby lowering the energy barrier for calcium influx. Our findings reveal a noninvasive, frequency-specific mitochondrial modulation by THz irradiation, which may offer a promising therapeutic avenue for addressing mitochondrial dysfunction.
    Keywords:  PGC-1α pathway; calcium signaling; frequency-selective modulation; mitochondrial biogenesis; terahertz irradiation
    DOI:  https://doi.org/10.1021/acsnano.5c16791
  23. Cell Res. 2026 Jan;36(1): 11-37
      Mitophagy, an evolutionarily conserved quality-control process, selectively removes damaged mitochondria to maintain cellular homeostasis. Recent advances in our understanding of the molecular machinery underlying mitophagy - from receptors and stress-responsive triggers to lysosomal degradation - illustrate its key role in maintaining mitochondrial integrity and adapting mitochondrial function to ever-changing physiological demands. In this review, we outline the fundamental mechanisms of mitophagy and discuss how dysregulation of this pathway disrupts mitochondrial function and metabolic balance, driving a wide range of disorders, including neurodegenerative, cardiovascular, metabolic, and immune-related diseases, as well as cancer. We explore the dual role of mitophagy as both a disease driver and a therapeutic target, highlighting the efforts and challenges of translating mechanistic insights into precision therapies. Targeting mitophagy to restore mitochondrial homeostasis may be at the center of a large range of translational opportunities for improving human health.
    DOI:  https://doi.org/10.1038/s41422-025-01203-7
  24. Postepy Biochem. 2025 12 16. 71(4): 369-382
      Epigenetic regulation of gene expression is an intensively studied area of molecular biology. It includes cytosine methylation, whose mechanism of action in nuclear DNA is relatively well understood. This process is mediated by enzymes from the DNA methyltransferase family. Hydroxymethylation is, considered both an intermediate step in cytosine demethylation and a potentially independent mechanism of regulation of gene expression. Functional significance—and even the presence—of methylation within mitochondrial DNA (mtDNA) remains a matter of debate. Accumulated evidence indicates that methylation and hydroxymethylation may play important role in mitochondria. Although epigenetic regulation of gene expression in mitochondria is not yet fully understood, the current state of knowledge suggests that it may influence proper cellular function and the pathogenesis of numerous diseases.
    DOI:  https://doi.org/10.18388/pb.2021_632
  25. Am J Med Genet C Semin Med Genet. 2026 Jan 07.
      Rare diseases collectively affect millions of Americans, but less than 5% have approved treatments, and new drug development remains limited. For such diseases, drug repurposing may be an effective strategy to find new treatment options. In the rare genetic disorder community, drugs are frequently prescribed off-label. This information is rarely available for research, but if captured, could be leveraged to accelerate the identification of candidate drugs to be evaluated for safety and efficacy of the treatment of rare diseases. CURE ID is a publicly available treatment registry that collects real-world treatment data directly from healthcare providers, patients, and care partners in a consistent format. By aggregating this information, CURE ID can generate hypotheses for follow-up targeted research of repurposed drugs, potentially leading to the approval of these drugs for new indications. The success of the platform is predicated on its adoption in the rare disease community and routinely reporting treatment experiences to CURE ID.
    Keywords:  CURE ID; drug repurposing; rare diseases; rare genetic disorders; real‐world data
    DOI:  https://doi.org/10.1002/ajmg.c.32153
  26. Mol Neurobiol. 2026 Jan 07. 63(1): 349
      The gut-brain axis represents a dynamic two-way signaling network whose dysregulation has been implicated in a wide range of neurogastrointestinal disorders. In this context, extracellular vesicles (EVs) have emerged as critical mediators of intercellular signaling and as promising non-invasive biomarkers. Derived from host and microbial cells, EVs carry bioactive cargo-including proteins, lipids, nucleic acids, and metabolites-that reflect the physiological or pathological state of their cells of origin. Their ability to cross biological barriers, such as the blood-brain barrier, underscores their potential for diagnosing and monitoring gut-brain axis dysfunctions. In this mini-review, we integrate microbial and brain-derived EVs within the framework of gut-brain axis disorders and propose three translational "diagnostic niches": microbial EVs as systemic markers of dysbiosis and immune activation, brain-derived EVs as liquid biopsies of the central nervous system (CNS) pathology, and engineered or technologically captured EVs as platforms for point-of-care testing. We summarize recent mechanistic insights, highlight disease-specific evidence in irritable bowel syndrome, inflammatory bowel disease, neurodegenerative, and psychiatric conditions, and critically appraise emerging isolation and analytical technologies in light of MISEV2023 recommendations. Finally, we discuss current limitations and translational hurdles, outlining how standardized EV-based diagnostics may be incorporated into precision medicine strategies targeting neurogastrointestinal diseases.
    Keywords:  Biomarkers; Diagnostics; Extracellular vesicles; Gut–brain axis; Isolation methods; Precision medicine
    DOI:  https://doi.org/10.1007/s12035-025-05645-3
  27. Int J Mol Sci. 2026 Jan 03. pii: 486. [Epub ahead of print]27(1):
      Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra and pathological α-synuclein aggregation. Growing evidence identifies chronic neuroinflammation-particularly NLRP3 inflammasome activation in microglia-as a central driver for PD onset and progression. Misfolded α-synuclein, mitochondrial dysfunction, and environmental toxins act as endogenous danger signals that prime and activate NLRP3 inflammasome, leading to caspase-1-mediated maturation of IL-1β and IL-18 and subsequent pyroptotic cell death. Impaired mitophagy, due to defects in PINK1/Parkin pathways or receptor-mediated mechanisms, permits accumulation of dysfunctional mitochondria and release DAMPs, thereby amplifying NLRP3 activity. Studies demonstrate that promoting mitophagy or directly inhibiting NLRP3 attenuates neuroinflammation and protects dopaminergic neurons in PD models. Autophagy-inducing compounds, along with NLRP3 inhibitors, demonstrate neuroprotective potential, though their clinical translation remains limited due to poor blood-brain barrier penetration, off-target effects, and insufficient clinical data. Additionally, the context-dependent nature of mitophagy underscores the need for precise therapeutic modulation. This review summarizes current understanding of inflammasome-mitophagy crosstalk in PD, highlights major pharmacological strategies under investigation, and outlines its limitations. Future progress requires development of specific modulators, targeted delivery systems, and robust biomarkers of mitochondrial dynamics and inflammasome activity for slowing PD progression.
    Keywords:  NLRP3 inflammasomes; PINK1/Parkin pathway; Parkinson’s disease; mitophagy; neuroinflammation; neuroprotection
    DOI:  https://doi.org/10.3390/ijms27010486
  28. Yakugaku Zasshi. 2026 ;146(1): 25-34
      Patients with rare diseases worldwide face substantial unmet therapeutic needs. In Japan, drug lag-delays in drug approval compared to other countries-has resurfaced as a pressing public health issue. This study analyzes orphan drugs (ODs) approved in the United States (U.S.) from 2005 to 2021, examining OD lag trends, and research and development (R&D) models to streamline OD development in Japan. Despite increased OD approval in the U.S. since 2018, the number of unapproved ODs in Japan has substantially increased. Although OD lag had once decreased, it has shown a tendency to resurge since 2017. This is largely due to changes in the R&D strategies of pharmaceutical companies, which are driven by the growing presence of U.S.- and Europe-based small- to mid-sized enterprises (SMEs) and the evolving industry landscape. Large foreign pharmaceutical companies have shifted toward a global development strategy for Japan, aiming for more efficient development and competitive advantage. This has been propelled by a move toward in-licensing earlier-stage drug candidates with global exclusivity from SMEs. Japanese pharmaceutical companies have focused on in-licensing late-stage drug candidates for the Japanese market from SMEs without a business presence in Japan, which have not been developed locally, thereby employing a bridging strategy for Japan. With the increase in ODs developed in the U.S. by these SMEs, this practice has substantially exacerbated the OD lag. As these SMEs are unlikely to enter the Japanese market, it is crucial for Japanese pharmaceutical companies to proactively pursue earlier, more proactive global partnerships with SMEs.
    Keywords:  development strategy; drug lag; rare disease; startup
    DOI:  https://doi.org/10.1248/yakushi.25-00140-2
  29. Int J Mol Sci. 2025 Dec 28. pii: 332. [Epub ahead of print]27(1):
      Sepsis induces severe immune and metabolic dysfunction driven by mitochondrial failure. Mitochondrial transplantation (MT) has emerged as a promising strategy to restore mitochondrial bioenergetics, but its metabolic impact on immune cells remains unclear. Here, we used gas chromatography-time-of-flight mass spectrometry (GC-TOF-MS)-based metabolomics to evaluate metabolic alterations in peripheral blood mononuclear cells (PBMCs) and splenocytes from a rat polymicrobial sepsis model treated with MT. Principal component and partial least-squares discriminant analyses revealed distinct clustering between sham, sepsis, and MT groups. Sepsis markedly suppressed metabolites related to amino acid, carbohydrate, and lipid metabolism, including aspartic acid, glutamic acid, AMP, and myo-inositol, reflecting mitochondrial metabolic paralysis. MT partially restored these metabolites toward sham levels, reactivating tricarboxylic acid (TCA) cycle, nucleotide, and lipid pathways. Pathway analysis confirmed that exogenous mitochondria reversed sepsis-induced metabolic suppression and promoted bioenergetic recovery in immune cells. These findings provide direct metabolomic evidence that MT reprograms immune metabolism and restores oxidative and biosynthetic function during sepsis, supporting its potential as a mitochondrial-based metabolic therapy.
    Keywords:  energy metabolism; gas chromatography–mass spectrometry; immune cells; metabolomics; mitochondrial dysfunction; mitochondrial transplantation; oxidative phosphorylation; peripheral blood mononuclear cells; sepsis; splenocytes
    DOI:  https://doi.org/10.3390/ijms27010332
  30. Redox Biol. 2025 Dec 24. pii: S2213-2317(25)00492-6. [Epub ahead of print]89 103979
      The mechanism of selecting dysfunctional mitochondria for mitophagy is only partially understood. Evidence suggests the mechanism involves reactions of superoxide (O2-•), hydrogen peroxide (H2O2), nitric oxide (NO•), peroxynitrite (ONOO-), carbonate radicals (•CO3-), nitrogen dioxide radicals (•NO2), hydroxyl radicals (•OH), oxygen (•O2• or O2), and carbon dioxide (CO2). However, the larger picture of how these reactions are organized to induce mitophagy is unclear. Extensive evidence suggests that increased mitochondrial matrix O2-• is associated with the mitophagy of dysfunctional organelles. In most cells, mitochondrial O2-• is mainly produced by the reaction of O2 with free radical intermediate forms of coenzyme Q (CoQ) and flavins, which are generated in substantial amounts in the inner membrane and matrix space of dysfunctional mitochondria. Mitochondrial O2-• plays two key roles in orchestrating mitophagy. First, it is dismutated by mitochondrial matrix superoxide dismutase 2 (SOD2) to H2O2. This diffusible messenger directs the nuclear and cytoplasmic compartments to prepare for mitophagy, including the generation of cytoplasmic NADPH and glutathione and the increased synthesis of membrane-diffusible NO•. Second, mitochondrial matrix space O2-• readily reacts with NO• to form ONOO-, which initiates a cascade of free radical reactions culminating in mitochondrial membrane depolarization and PINK1 and Parkin-driven mitophagy. Compelling observations that support the proposed mechanism are given. This mechanism could be targeted for the treatment of diseases characterized by dysfunctional mitophagy, such as Parkinson's disease. Because of the central role of mitochondrial O2-• as a sentinel for selective mitophagy, we have named this hypothesis the superoxide sentinel hypothesis of mitochondrial quality control.
    Keywords:  DJ-1; Mitophagy; NADPH; Nitric oxide synthase; Parkinson's disease; Superoxide sentinel hypothesis
    DOI:  https://doi.org/10.1016/j.redox.2025.103979
  31. Yakugaku Zasshi. 2026 ;146(1): 15-24
      Approximately 7000 rare diseases have been identified globally; however, effective treatments are available for only 5% of them, highlighting substantial unmet medical needs worldwide. In Japan, drug loss-the lack of domestic approval or development of drugs already approved overseas-has become a pressing public health issue. We analyzed orphan drugs (ODs) approved in the United States (U.S.) between 2005 and 2021 to examine trends in OD development in the U.S., drug loss and associated research and development (R&D) models, with the aim of identifying factors that could facilitate more seamless OD development in Japan. Despite a marked increase in OD approvals in the U.S. since 2018, the number of ODs not approved in Japan have increased rapidly, reaching 120 drugs (49%), of which approximately 70% had not entered development, indicating greater OD loss in Japan. U.S./Europe-based startups have become key players in rare disease drug R&D, significantly contributing to this drug loss trend. They successfully develop drugs in the U.S. by combining in-licensing with in-house drug discovery. Out-licensing to Japanese companies or large pharma is critical for expansion into Japan, with successes attributed to drug innovation, target indications, and transactional capabilities. Underlying this trend were Japan's perceived low market potential, its complex clinical trial environment and regulatory requirements, and the financial limitations of startups. These findings highlight the need to foster partnerships with startups and cultivate an ecosystem in Japan that nurtures local startups, to address drug loss and ensure access to promising drugs.
    Keywords:  development; drug lag; drug loss; orphan designation; rare disease; research
    DOI:  https://doi.org/10.1248/yakushi.25-00140-1
  32. Cell Death Dis. 2026 Jan 08. 17(1): 9
      Heat shock protein 60 (HSP60) plays a vital role in maintaining mitochondrial homeostasis and essential functions and requires ATP for its assembly into chaperone complexes. This study aimed to investigate the long-term effects of HSP60 induction on mitochondrial homeostasis at varying doses and durations using HSP60 transgenic mice. In this study, we generated transgenic mice with elevated levels of native HSP60 using the LoxP-Cre system. These mice exhibited impaired postnatal development, skeletal muscle dystrophy, and increased mortality. Initially, excess HSP60 enhanced the mitochondrial oxidative respiratory capacity, which was later compensated for by increased glycolysis. Surplus HSP60 primarily accumulated in the mitochondria, likely due to insufficient ATP availability, leading to the buildup of HSP60 heptamers. Consequently, mitochondrial number and morphology were altered, protein levels in electron transport chain complexes were reduced, and oxidative phosphorylation was impaired. Additionally, reactive oxygen species accumulated, contributing to mitochondrial dysfunction in skeletal muscles. The upregulation of Pink-1/Parkin triggered enhanced autophagy, while increased Bax and poly (ADP-ribose) polymerase (PARP) cleavage mediated heightened apoptosis; both mechanisms aimed at eliminating damaged mitochondria. However, prolonged HSP60 accumulation overwhelmed these protective processes, ultimately leading to skeletal muscle dystrophy and premature death. Our findings demonstrated that excessive mitochondrial HSP60 initially boosts oxidative respiration; however, over time, it contributes to mitochondrial dysregulation and myopathy. This study provides novel insights into how excessive HSP60 affects mitochondrial oxidative respiration and glycolysis, with potential links to certain mitochondria-related diseases.
    DOI:  https://doi.org/10.1038/s41419-025-08260-1