bims-mitdis Biomed News
on Mitochondrial disorders
Issue of 2026–01–11
73 papers selected by
Catalina Vasilescu, Helmholz Munich



  1. 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
  2. bioRxiv. 2025 Dec 31. pii: 2025.12.31.697091. [Epub ahead of print]
      Of the ∼1100 mitochondrial proteins, only a handful like PINK1 and ATFS-1 are known to stabilize and relocalize upon collapse of the proton motive force (PMF) to execute signaling roles. To systematically identify genes that increase exclusively at the protein level upon PMF collapse, we performed a joint proteomic and RNA-seq screen. The screen revealed 10 candidates (six mitochondrial), including the vitamin B12 chaperone MMADHC and cytosolic B12-dependent methionine synthase (MTR). MMADHC is short-lived across cell types and we show that its levels increase with PMF collapse. MMADHC stabilization precedes PINK1 activation in a time course of increasing mtDNA depletion, suggesting greater sensitivity to PMF collapse. MMADHC accumulates in mitochondria with LONP1 inhibition but in the cytosol upon PMF collapse, likely due to mitochondrial import failure. Cytosol-stabilized MMADHC increases MTR levels and activity. Altogether, the mitochondrial PMF regulates the cytosolic B12-dependent MTR, integral to one-carbon metabolism, by controlling the stability and compartmentalization of the B12 chaperone MMADHC.
    Significance Statement: Humans have only two vitamin B12-dependent enzymes - mitochondrial MMUT and cytosolic MTR - and both require a common B12 chaperone MMADHC. We discover that MMADHC is a low abundant, short-lived protein that is continuously imported and degraded by energized mitochondria. Upon collapse of the mitochondrial proton motive force, MMADHC accumulates in the cytosol and increases the levels and activity of MTR, critical for one-carbon metabolism. This PMF-dependent regulation of MMADHC stability and localization is important for understanding cofactor rationing and spatiotemporal compartmentalization of B12 metabolism.
    DOI:  https://doi.org/10.64898/2025.12.31.697091
  3. J Inherit Metab Dis. 2026 Jan;49(1): e70132
      Mitochondrial trifunctional protein (TFP) deficiency is an inherited disorder of long-chain fatty acid β-oxidation (FAO). TFP is a heteromeric enzyme composed of two α and two β-subunits. Despite early detection and dietary treatment, TFP deficiency patients often develop hypoglycemia, rhabdomyolysis, cardiomyopathy, and peripheral neuropathy. Degenerative retinopathy and milder peripheral neuropathy occur in patients with an isolated deficiency of the αTFP subunit of long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) activity. Triheptanoin treatment improves most complications, but not peripheral neuropathy and retinopathy. Notably, TFP also carries a fourth enzymatic function involved in cardiolipin remodeling, which we previously found to be impaired in TFP/LCHAD deficiency. We therefore tested whether elamipretide, a synthetic cardiolipin-binding peptide, could improve mitochondrial function and cardiolipin levels in βTFP-deficient mice and patient-derived fibroblasts. Mice were treated with elamipretide delivered by osmotic minipump and challenged with treadmill exercise or cold stress after fasting. βTFP-deficient mice treated with elamipretide showed improved exercise endurance, but cold tolerance was not altered. Liver mitochondria from male βTFP-deficient mice demonstrated improved FAO-ETC enzyme activities. However, cardiolipin content and composition were unchanged. In patient fibroblasts, elamipretide produced a possible genotype-dependent increase in mitochondrial bioenergetics and a reduction in ROS. These results support a mechanism in which elamipretide stabilizes between FAO enzymes and ETC complexes, thereby improving mitochondrial function independently of changes in cardiolipin levels. Elamipretide thus emerges as a potential therapeutic agent for TFP/LCHAD deficiency, warranting further preclinical studies.
    Keywords:  cardiolipin; elamipretide; electron transport chain complex proteins; fatty acid oxidation; mitochondria; mitochondrial trifunctional protein deficiency; multiprotein energy complex; oxidative phosphorylation
    DOI:  https://doi.org/10.1002/jimd.70132
  4. bioRxiv. 2025 Dec 23. pii: 2025.12.21.693523. [Epub ahead of print]
      Outer mitochondrial membranes (OMM) function as dynamic hubs for inter-organelle communication, integrating bidirectional signals, and coordinating organelle behavior in a context-dependent manner. However, tools for mapping mitochondrial surface proteomes with high spatial and temporal resolution remain limited. Here, we introduce an optogenetic proximity labeling strategy using LOV-Turbo, a light-activated biotin ligase, to profile mitochondrial surface proteomes with improved precision, temporal control, and reduced background. By fusing LOV-Turbo to a panel of variants of an OMM-anchored protein, Miro1, we generate spatially distinct baits that resolve modular architectures and regulatory states of the OMM proteomes across diverse conditions, a database we name MitoSurf. Building on this proteomic map, we present RiboLOOM, a platform that defines LOV-Turbo labeled ribosomes and their bound mRNAs at the mitochondrial surface. MitoSurf and RiboLOOM uncover a spatially distinct ribosome pool at the OMM that is maintained by Miro1, enabling local mRNA engagement and translation of mitochondria-related proteins. These findings establish Miro1 as a key organizer of mitochondrial protein biogenesis through spatial confinement of surface-associated ribosomes. Our platform reveals an uncharted layer of mitochondrial surface biology and provides a generalizable strategy to dissect dynamic RNA-protein-organelle interfaces in living cells.
    DOI:  https://doi.org/10.64898/2025.12.21.693523
  5. 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
  6. 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
  7. 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
  8. Essays Biochem. 2025 Dec 22. pii: EBC20253045. [Epub ahead of print]69(5):
      Through its various roles in protein quality control, membrane dynamics, and cellular survival pathways, the AAA+ ATPase p97/valosin-containing protein emerges as a significant regulator of mitochondrial homeosta sis. This review comprehensively examines the multifaceted functions of p97 in mitochondrial biology, spanning from mitochondria-associated degradation to newly discovered functions in organellar cross-talk and disease pathogenesis. Underlying its cellular importance, p97 mutations are found in amyotrophic lateral sclerosis and frontotemporal dementia. To elucidate its mechanistic contribution to these processes, we provide a detailed table (Table 1) listing all known mitochondrial Cdc48/p97 substrates and associ ated proteins, categorized by their respective pathways. Recruitment to most of these substrates occurs by specialized adaptors, including Doa1/phospholipase A-2-activating protein, UBXD8, and UBXN1. p97 orchestrates the extraction and proteasomal degradation of outer mitochondrial membrane proteins, which are essential for maintaining mitochondrial integrity. For example, by controlling the turnover of fusion factors MFN1/2 and fission machinery, p97 regulates mitochondrial dynamics. p97 also governs apoptotic signaling through the regulated degradation of anti-apoptotic factors, such as myeloid cell leukemia-1 and VDAC, thereby modulating mitochondrial permeability. In mitophagy, p97 enables the clearance of damaged organelles by extracting ubiquitinated substrates and recruiting autophagy machinery. Beyond proteolysis, p97 facilitates recycling of endoplasmic reticulum-mitochondria contact sites through regulation of UBXD8-dependent lipid metabolism. Recent discoveries have revealed p97's involvement in pathogen host interactions and circular RNA-mediated regulation, thereby expanding our understanding of its cellular functions. The emerging picture positions p97 as an integrative hub co-ordinating mitochondrial protein homeostasis, organellar dynamics, and cell fate decisions, with therapeutic potential for metabolic and neurodegenerative disorders.
    Keywords:  Cdc48; ERAD; MAD; P97; VCP; mitochondria; mitostasis; proteasome; ubiquitin
    DOI:  https://doi.org/10.1042/EBC20253045
  9. J Cell Biol. 2026 Apr 06. pii: e202507116. [Epub ahead of print]225(4):
      Hundreds of mitochondrial proteins rely on N-terminal presequences for organellar targeting and import. While generally described as positively charged amphiphilic helices, presequences lack a consensus motif and thus likely promote protein import into mitochondria with variable efficiencies. Indeed, the concept of presequence strength underlies biological models such as stress sensing, yet a quantitative analysis of what dictates strong versus weak presequences is lacking. Furthermore, the extent to which presequence strength affects mitochondrial function and cellular fitness remains unclear. Here, we capitalize on the MitoLuc protein import assay to define multiple aspects of presequence strength. We find that select presequences, including those that regulate the mitochondrial unfolded protein response (UPRmt), impart differential import efficiencies during mitochondrial uncoupling. Surprisingly, we find that presequences beyond those associated with stress signaling promote highly variable import efficiency in vitro, suggesting presequence strength may influence a broader array of processes than currently appreciated. We exploit this variability to demonstrate that only presequences that promote robust in vitro import can fully rescue defects in respiratory growth in complex IV-deficient yeast, suggesting that presequence strength dictates metabolic potential. Collectively, our findings demonstrate that presequence strength can describe numerous metrics, such as total imported protein, maximal import velocity, or sensitivity to uncoupling, suggesting that the annotation of presequences as weak or strong requires more nuanced characterization than typically performed. Importantly, we find that such variability in presequence strength meaningfully affects cellular fitness beyond stress signaling, suggesting that organisms may broadly exploit presequence strength to fine-tune mitochondrial import and thus organellar homeostasis.
    DOI:  https://doi.org/10.1083/jcb.202507116
  10. Science. 2026 Jan 08. eady5532
      Environmental adaptation often involves a shift in energy utilization toward mitochondrial fatty acid oxidation, which requires carnitine. Besides dietary sources of animal origin, carnitine biosynthesis from trimethyllysine (TML) is essential, particularly for those who consume plant-based diets; however, its molecular regulation and physiological role remain elusive. Here, we identify SLC25A45 as a mitochondrial TML carrier that controls carnitine biosynthesis and fuel switching. SLC25A45 deficiency decreased the carnitine pool and impaired mitochondrial fatty acid oxidation, shifting reliance to carbohydrate metabolism. Slc25a45-deficient mice were cold-intolerant and resistant to lipid mobilization by GLP1 receptor agonist (GLP-1RA), rendering them resistant to adipose tissue loss. Our study suggests that mitochondria serve as a regulatory checkpoint in fuel switching, with implications for metabolic adaptation and the efficacy of GLP-1RA-based anti-obesity therapy.
    DOI:  https://doi.org/10.1126/science.ady5532
  11. 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
  12. 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
  13. 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
  14. 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
  15. 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
  16. bioRxiv. 2025 Dec 29. pii: 2025.12.29.696888. [Epub ahead of print]
      Mutations in the mitochondrial protein CHCHD10 (D10) cause a spectrum of hereditary neurodegenerative disorders. Among these, the p.R15L variant is linked to a slowly progressive, late-onset familial form of amyotrophic lateral sclerosis (ALS) with unclear pathogenic mechanisms. To better understand this, we investigated a knock-in (KI) mouse model carrying the p.R15L mutation in the endogenous protein. Unlike previously described mutant D10 KI models, p.R15L KI mice exhibited normal D10 protein levels, with no evidence of large protein aggregates. Mitochondrial respiration and hydrogen peroxide emission in mitochondria isolated from muscle and brain were unaltered. Similarly, fibroblasts from human p.R15L carriers exhibited normal D10 levels and unchanged oxidative phosphorylation function. Histochemical analyses of p.R15L KI muscle revealed mild increases in mitochondrial enzymatic activity in a subset of muscle fibers and muscle transcriptomics showed elevated expression of PGC-1α, suggesting enhanced mitochondrial biogenesis. p.R15L KI mice developed subtle, late-onset phenotypes, including reduced body weight and motor activity and increased anxiety-like behavior. Importantly, in aged mice electrophysiological studies demonstrated decreased amplitude of the compound muscle action potential, commensurate with a moderate loss of spinal cord motor neurons and elevated serum neurofilament light levels, indicative of neurodegeneration. Together, these results indicate that the p.R15L mutation produces a mild, late-onset motor neuron phenotype in mice, partially recapitulating the human disease, without mitochondrial functional or morphological alterations. The findings indicate that p.R15L D10 selectively impairs mouse motor neurons through a gain-of-function mechanism, providing a genetically accurate yet mild in vivo model of familial ALS.
    DOI:  https://doi.org/10.64898/2025.12.29.696888
  17. J Cell Sci. 2026 Jan 01. pii: jcs263847. [Epub ahead of print]139(1):
      Mitochondrial dynamics are defined by the continuous processes of fusion and fission that regulate mitochondrial shape, distribution and activity. They are also involved in cellular functions of mitochondria, such as energy production, metabolic adaptation, apoptosis and cellular stress responses. Consequently, these organelle dynamics play a crucial role in development, growth, differentiation and disease. Mitochondrial morphology is controlled by Drp1 (also known as DNM1L) and Fis1, which drive fission, whereas Opa1, Mfn1 and Mfn2 mediate fusion. The transcription, activation and degradation of these proteins are often regulated by signaling cascades that are crucial for stem cell maintenance and differentiation. In turn, mitochondrial dynamics regulate key outcomes of these pathways. We explore the interplay between mitochondrial fusion and fission proteins and such signaling pathways, including Notch, receptor tyrosine kinase, JNK, Hippo and mTOR signaling, finding that stem cell renewal and differentiation states are dependent on the regulation of signaling pathways by mitochondrial morphology and activity. Overall, this Review highlights how mitochondrial morphology and activity crucially regulate stem cell division for renewal and differentiation, examining their impact across diverse systems.
    Keywords:  Drp1; Marf; Mfn; Mitochondria; Opa1; Signaling; Stem cells
    DOI:  https://doi.org/10.1242/jcs.263847
  18. J Clin Med. 2025 Dec 24. pii: 146. [Epub ahead of print]15(1):
      Neurometabolic diseases encompass a diverse group of rare and often progressive genetic disorders affecting the nervous system due to abnormalities in metabolic pathways. These conditions, including mitochondrial disorders, lysosomal storage diseases, and others, can manifest in adults with a range of neurological symptoms, which will be reviewed here. Given their complexity and chronic nature, comprehensive management is crucial for improving patients' quality of life. In this Invited Perspective, we review the neurological signs and symptoms of the most commonly encountered inherited metabolic disorders in adult neurology. Furthermore, drawing on our clinical experience, we demonstrate that an integrated local care approach is fundamental for these patients, as it enables continuous monitoring, early intervention, and coordinated multidisciplinary support.
    Keywords:  ataxia; epileptic encephalopathies; metabolic myopathies; mitochondrial diseases; movement disorders; spastic paraparesis; stroke; stroke-like episodes
    DOI:  https://doi.org/10.3390/jcm15010146
  19. Redox Biol. 2026 Jan 05. pii: S2213-2317(26)00001-7. [Epub ahead of print]89 104003
      A recent study published in Redox Biology (Volume 88, December 2025, 103952) demonstrates that molecular hydrogen (H2) rapidly suppresses mitochondrial Complex III activity through a mechanism involving the Rieske iron-sulfur protein (RISP) and subsequent LONP1-dependent proteolysis, challenging the long-standing view of H2 as merely a selective radical scavenger. While these findings compellingly identify RISP as a key mediator of mitochondrial responses to H2, its designation as the primary molecular target warrants broader consideration. From an evolutionary and structural standpoint, RISP belongs to a wider family of hydrogenase-like mitochondrial redox proteins that retain ancient iron-sulfur architectures. Proteins such as succinate dehydrogenase subunit B (SDHB), iron-sulfur subunits of Complex I, and CISD family [2Fe-2S] proteins share comparable redox logic and strategic positioning within mitochondrial bioenergetic networks. Here, these candidates are prioritized and placed into a hierarchical, testable framework, and specific comparative structural, biochemical, and proteostatic approaches are proposed to define the true molecular entry point of H2 signaling in human mitochondria.
    Keywords:  Bioenergetics; Electron transport chain; LONP1-Mediated proteostasis; Mitochondrial redox signaling; Molecular hydrogen; Rieske iron-sulfur protein
    DOI:  https://doi.org/10.1016/j.redox.2026.104003
  20. Aging Cell. 2026 Jan;25(1): e70355
      Age-associated degeneration of neuromuscular junctions (NMJs) contributes to sarcopenia and motor function decline, yet the mechanisms that drive this dysfunction in aging remain poorly defined. Here, we demonstrate that postsynaptic mitochondria are significantly diminished in quantity in old-aged skeletal muscle, correlating with increased denervation and delayed reinnervation following nerve injury. Single-nucleus RNA sequencing before and after sciatic nerve crush from young and old-aged muscles further revealed that sub-synaptic myonuclei in old-aged muscle exhibit attenuated expression of mitochondrial gene programs, including oxidative phosphorylation, biogenesis, and import. To test whether these deficits are causal, we developed a muscle-specific CRISPR genome editing approach and targeted CHCHD2 and CHCHD10-two nuclear-encoded mitochondrial proteins that localize to the intermembrane space and interact with the mitochondrial contact site and cristae organizing system. CRISPR knockout of CHCHD2 and CHCHD10 in young muscle recapitulated old-aged muscle phenotypes, including mitochondrial disorganization, reduced ATP production, NMJ fragmentation, and delayed reinnervation. Transcriptional profiling of sub-synaptic myonuclei using single-nuclei RNA sequencing from CHCHD2 and CHCHD10 knockout muscles revealed impairments in activation of mitochondrial remodeling programs and elevated stress signatures when compared with controls. These findings establish a critical role for postsynaptic mitochondrial integrity in sustaining NMJ stability and regenerative capacity and identify CHCH domain-containing proteins as key regulators of postsynaptic mitochondrial function during aging and injury.
    DOI:  https://doi.org/10.1111/acel.70355
  21. 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
  22. Nat Cardiovasc Res. 2026 Jan 06.
      The heart uses a muscle-specific ribosome in cardiomyocytes, where the ribosomal protein RPL3 is replaced by its paralog RPL3L. Rare biallelic RPL3L mutations cause fatal neonatal dilated cardiomyopathy, yet the mechanisms that link genotype to heart failure are unclear. Despite the recessive inheritance pattern in humans, Rpl3l knockout mice show no overt cardiac phenotype, probably because of compensatory RPL3 upregulation through unknown mechanisms. Here we report four additional cases and propose a unifying pathogenetic model by integrating human genetics, patient tissues and isogenic cell models. Affected individuals typically carry one of two recurrent hotspot missense variants paired with a private allele. Whereas non-hotspot variants phenocopy knockout and allow RPL3 compensation, hotspot variants induce nucleolar protein aggregation, disrupt rRNA processing and block compensation by preserving the role of RPL3L in repressing RPL3 via unproductive splicing. These findings establish combined loss-of-function and gain-of-function mechanisms for RPL3L-associated cardiomyopathy and inform genetic screening, diagnosis and therapeutic development.
    DOI:  https://doi.org/10.1038/s44161-025-00761-8
  23. Cell Death Dis. 2026 Jan 09. 17(1): 26
      Lipophagy is a form of selective autophagy that targets the lipid droplets for lysosomal decay and has been implicated in the onset and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Factors that augment lipophagy have been identified as targets for MASLD therapeutic development. TMEM55B is a key regulator of lysosomal positioning, which is critical for lysosome fusion with the autophagosome, but is less well studied. Here, we demonstrate that the absence of TMEM55B in murine models accelerates MASLD onset and progression to metabolic dysfunction-associated steatohepatitis (MASH). In cellular models, TMEM55B deficiency enhances incomplete lipophagy, whereby lysosome-lipid droplet interactions are increased, but lysosomal cargo is not fully degraded and/or released, leading to the development of lipid-filled lysosomes (lipolysosomes). Loss of TMEM55B also impairs mitophagy, causing an accumulation of dysfunctional mitochondria. This imbalance leads to increased lipid accumulation and oxidative stress, worsening MASLD. These findings underscore the importance of lysosomal positioning in lipid metabolism and suggest that targeting lipophagy for MASLD therapeutic development should be carefully considered to ensure promotion of the entire lipophagic flux pathway and whether it occurs in the context of mitochondrial dysfunction.
    DOI:  https://doi.org/10.1038/s41419-025-08210-x
  24. Mol Ther. 2026 Jan 02. pii: S1525-0016(25)01139-6. [Epub ahead of print]
      Mitochondrial dysfunction is a hallmark of myopathies and impaired skeletal muscle differentiation. Here, we demonstrate that C1q/TNF-related protein 1 (CTRP1) is essential for maintaining mitochondrial dynamics and supporting myogenic differentiation. Loss of CTRP1 in myoblasts and in skeletal muscle-specific knockout (CTRP1 KOΔACTA) mice led to impaired myotube formation, reduced muscle fiber cross-sectional area, and decreased muscle strength. CTRP1 deficiency also shifted the muscle fiber composition from oxidative Type IIA to glycolytic Type IIB fibers, indicating a compromised mitochondrial capacity. At the cellular level, CTRP1 loss resulted in elongated and disorganized mitochondria with diminished cristae density, membrane potential, and oxidative respiration. These mitochondrial abnormalities are associated with defective recruitment of dynamin-related protein 1 (DRP1), a central mediator of mitochondrial fission. Restoring CTRP1 expression or performing mitochondrial transplantation in CTRP1 KO myoblasts rescued mitochondrial function and re-established differentiation capacity. Furthermore, CTRP1 expression progressively decreased in accordance with disease severity in skeletal muscle biopsies from patients with polymyositis, dermatomyositis, and Duchenne muscular dystrophy, supporting its potential relevance to human myopathies. Together, these findings identify CTRP1 as a novel regulator of mitochondrial quality and myogenic differentiation, highlighting its potential as a therapeutic target for mitochondrial myopathies.
    DOI:  https://doi.org/10.1016/j.ymthe.2025.12.063
  25. Nat Commun. 2026 Jan 08.
      Healthy mitochondria are crucial for maintaining neuronal homeostasis. Their activity depends on a dynamic lipid and protein exchange through fusion, fission, and vesicular trafficking. Studying vesicles in neurons is challenging with conventional microscopy due to their small size, heterogeneity, and dynamics. We use multicolour stimulated emission depletion nanoscopy to uncover the ultrastructure of mitochondrial-derived vesicles (MDVs) in live neurons, biosensors to define their functional state, and a pulse-chase strategy to identify their turnover in situ. We identified three populations of vesicular structures: one transporting degradation products originating from oxidative stress, one shuttling cargo and newly translated proteins for local organelle biogenesis and one consisting of small, functional mitochondria. Furthermore, we provide evidence supporting that de novo peroxisomes biogenesis occurs via the fusion of endoplasmic reticulum and MDVs at mitochondrial sites. Our data provide mechanistic insight into organelle biogenesis driven by significant diversity in MDV morphology, functional state, and molecular composition.
    DOI:  https://doi.org/10.1038/s41467-025-68160-y
  26. bioRxiv. 2025 Dec 23. pii: 2025.12.19.695597. [Epub ahead of print]
      Metabolic enzymes can influence chromatin organization by modulating the availability of key metabolites, yet how specific metabolic reactions affect chromatin function remains poorly understood. Here, we show that in Caenorhabditis elegans, the methionine-cycle enzyme methionine synthase reductase (MTRR-1/MSR) regulates heterochromatin independently of methionine synthesis. Loss of MTRR-1, but not of the methionine synthase METR-1/MS, specifically reduces heterochromatic histone methylation, derepresses repetitive elements, and causes developmental delay. Multi-omics profiling revealed that mtrr-1 mutants activate transcriptional programs associated with mitochondrial stress and accumulate long-chain acylcarnitines, indicating disrupted mitochondrial homeostasis. Functional assays confirmed altered mitochondrial respiration in mtrr-1 mutants, while direct perturbation of mitochondrial function was sufficient to induce heterochromatin defects. Together, our results reveal a previously unrecognized mitochondria-to-chromatin axis controlled by the methionine-cycle enzyme MTRR-1/MSR.
    DOI:  https://doi.org/10.64898/2025.12.19.695597
  27. 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
  28. 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
  29. Am J Med Genet A. 2026 Jan 08.
      We report two siblings harboring a homozygous MGME1 variant, NM_052865.4:c.818 T>A; p.(Val273Glu), both presenting with ptosis, myopathy, scoliosis, and gastrointestinal symptoms. The index patient developed progressive, medically refractory dilated cardiomyopathy and underwent successful orthotopic heart transplantation (OHT). Reanalysis of previously negative WES identified the variant in the index case, and segregation by Sanger sequencing confirmed homozygosity in both siblings. Although several clinical findings overlap with previously described MGME1-related disease, the detected variant remains classified as a variant of uncertain significance (VUS); thus, functional evidence is needed to better understand its potential causal relevance. Additionally, this report underscores the importance of periodic genomic data reanalysis and highlights the variable expressivity that may occur even within the same family.
    Keywords:   MGME1 ; dilated cardiomyopathy; heart transplantation; mitochondrial DNA depletion syndrome 11; reanalyze; whole exome sequencing
    DOI:  https://doi.org/10.1002/ajmga.70049
  30. Cell Mol Immunol. 2026 Jan 06.
      Innate immunity in host cells must be rapidly activated to combat invading microbes. Upon RIG-I activation, the transcription of type I interferons is induced within one hour in virus-infected cells. Previous studies have shown that endogenous MAVS spreads signals via aggregation on the mitochondrial membrane, whereas truncated recombinant MAVS forms prion-like filaments in vitro. How MAVS transmits signals so quickly, and the molecular architecture of its membrane aggregates, remains elusive. Here, we report that activated MAVS forms fibrils encircling its resident mitochondrion or connecting neighboring mitochondria with a "ladder-like" structure, allowing the activation of dormant MAVS on encountered mitochondria. This "intermitochondrial activation" process promotes a rapid antiviral response in cells to overcome the immediate danger caused by viruses. Moreover, stuck MAVS fibrils between mitochondria have limited cytosolic protein access and thus relay signals poorly. This study demonstrated that prion-like MAVS fibrils cluster in mitochondria to ensure a rapid antiviral response.
    Keywords:  Innate immunity; MAVS; mitochondria; prion-like fibrillation; viral infection
    DOI:  https://doi.org/10.1038/s41423-025-01382-8
  31. 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
  32. medRxiv. 2025 Dec 29. pii: 2025.12.22.25342856. [Epub ahead of print]
      Leucine-rich repeat kinase 2 (LRRK2) variants are the most common cause of inherited Parkinson's disease (PD), and the hyperactivity of the LRRK2 variants represent a validated drug target for PD. The penetration of common LRRK2 variants is incomplete, underscoring the need for molecular biomarkers that predict disease onset and guide therapeutics development. Here, we analyzed large datasets of cerebrospinal fluid (CSF) and urinary proteomics from the Parkinson's Progression Markers Initiative (PPMI) and identified distinct lysosomal and immune protein signatures as potential biomarkers for LRRK2-linked PD (LRRK2 PD). Longitudinal analysis revealed that levels of specific lysosomal and immune proteins remained elevated in CSF during the prodromal phase but declined following clinical symptom onset. Furthermore, examination of multiple brain cell types from Lrrk2 mutant mice carrying disease variant (G2019S) showed heightened secretion of lysosomal proteins in microglia and astrocytes, but not neurons, supporting a glial origin and intrinsic LRRK2 mutant activity responsible for the elevated CSF lysosomal proteins. Furthermore, proteomics analysis of urine from humanized LRRK2 G2019S transgenic mice identified lysosome and glycosphingolipid protein signatures shared with human LRRK2 PD patients. Collectively, our integrated proteomics reveals dynamic changes of functional biofluid signatures for LRRK2 PD, which enables the determination of biomarkers for early disease onset. The humanized LRRK2 G2019S mice provide a valuable platform for biomarker refinement and therapeutic development.
    One Sentence Summary: Integrated human and mouse proteomic analyses identify dynamic lysosomal and immune biofluid signatures, possibly of glial origin, as functional biomarkers of LRRK2-linked Parkinson's disease progression, supported by a novel humanized LRRK2 G2019S mouse model that recapitulates key urinary biomarker profiles.
    DOI:  https://doi.org/10.64898/2025.12.22.25342856
  33. Redox Biol. 2025 Nov 26. pii: S2213-2317(25)00449-5. [Epub ahead of print]89 103936
      Coenzyme Q (CoQ) is a crucial lipid-soluble antioxidant and electron transporter vital for mitochondrial respiration and cellular redox balance. Despite the role of CoQ in oxidative phosphorylation being well established, the mechanisms by which CoQ is internalized, distributed among subcellular compartments, and trafficked to mitochondria remain poorly defined. Here, we present the development of a minimally modified, azide-tagged CoQ analogue that enables high-resolution visualization of CoQ localization using fluorescence-based imaging. Using this probe, we focus our investigation on brown adipose tissue (BAT), a mitochondria-rich, highly metabolically active tissue with elevated CoQ demand. On a cellular level, we demonstrate that CoQ is internalized via receptor-mediated endocytosis, predominantly localizing to lysosomes. Genetic knockdown and pharmacological studies identify CD36 and NPC1 as essential transporters in this process. Our work provides both a technical advance for the redox biology field, with the development and characterization of a CoQ probe, and the essential new biological insight that NPC1 is linked to CoQ homeostasis and thus provides a foundation for further dissection of CoQ biology in health and disease.
    DOI:  https://doi.org/10.1016/j.redox.2025.103936
  34. JCI Insight. 2026 Jan 06. pii: e183247. [Epub ahead of print]
      Iron regulatory protein 1 (IRP1) is a post-transcriptional regulator of cellular iron metabolism. In mice, loss of IRP1 causes polycythemia through translational de-repression of hypoxia-inducible factor 2α (HIF2α) mRNA, which increases renal erythropoietin production. Here we show that Irp1-/- mice develop fasting hypoglycemia and are protected against high-fat diet-induced hyperglycemia and hepatic steatosis. Discovery-based proteomics of Irp1-/- livers revealed a mitochondrial dysfunction signature. Seahorse flux analysis in primary hepatocytes and differentiated skeletal muscle myotubes confirmed impaired respiratory capacity, with a shift from oxidative phosphorylation to glycolytic ATP production. This metabolic rewiring was associated with enhanced insulin sensitivity and increased glucose uptake in skeletal muscle. Under metabolic stress, IRP1 deficiency altered the redox balance of mitochondrial iron, resulting in inefficient energy production and accumulation of amino acids and metabolites in skeletal muscle, rendering them unavailable for hepatic gluconeogenesis. These findings identify IRP1 as a critical regulator of systemic energy homeostasis.
    Keywords:  Diabetes; Glucose metabolism; Hepatology; Metabolism; Proteomics
    DOI:  https://doi.org/10.1172/jci.insight.183247
  35. Nature. 2026 Jan 07.
      Primary sensory neurons in dorsal root ganglia (DRG) have long axons and a high demand for mitochondria, and mitochondrial dysfunction has been implicated in peripheral neuropathy after diabetes and chemotherapy1,2. However, the mechanisms by which primary sensory neurons maintain their mitochondrial supply remain unclear. Satellite glial cells (SGCs) in DRG encircle sensory neurons and regulate neuronal activity and pain3. Here we show that SGCs are capable of transferring mitochondria to DRG sensory neurons in vitro, ex vivo and in vivo by the formation of tunnelling nanotubes with SGC-derived myosin 10 (MYO10). Scanning and transmission electron microscopy revealed tunnelling nanotube-like ultrastructures between SGCs and sensory neurons in mouse and human DRG. Blockade of mitochondrial transfer in naive mice leads to nerve degeneration and neuropathic pain. Single-nucleus RNA sequencing and in situ hybridization revealed that MYO10 is highly expressed in human SGCs. Furthermore, SGCs from DRG of people with diabetes exhibit reduced MYO10 expression and mitochondrial transfer to neurons. Adoptive transfer of human SGCs into the mouse DRG provides MYO10-dependent protection against peripheral neuropathy. This study uncovers a previously unrecognized role of peripheral glia and provides insights into small fibre neuropathy in diabetes, offering new therapeutic strategies for the management of neuropathic pain.
    DOI:  https://doi.org/10.1038/s41586-025-09896-x
  36. Stem Cell Res. 2025 Dec 22. pii: S1873-5061(25)00245-4. [Epub ahead of print]90 103895
      Sengers syndrome is a rare mitochondrial disorder caused by the loss of a nuclear encoded mitochondrial protein, acylglycerol kinase (AGK). Here, we describe the generation of a novel in vitro stem cell model of Sengers syndrome (AGKKO C10) using CRISPR/Cas9 gene editing. This cell line displayed normal characteristics of pluripotent stem cells, including colony morphology, expression of pluripotency markers, trilineage potential, and no karyotypic abnormalities. Together with the parental H9 hESC control line, the AGKKO C10 line can ultimately be used for investigation of disease mechanisms and drug testing.
    DOI:  https://doi.org/10.1016/j.scr.2025.103895
  37. 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
  38. 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
  39. 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
  40. J Biotechnol. 2026 Jan 06. pii: S0168-1656(26)00002-7. [Epub ahead of print]
      Metabolomics analyses of cell culture processes can provide valuable insight into cellular physiology that can be leveraged to develop more productive processes. In this work, we applied metabolomics to interrogate CHO cell behavior in two industrial chemically-defined media in cultures co-fed with glucose and lactic acid. We previously reported that secreted acylcarnitines are indicative of altered mitochondrial metabolism when cultures are fed lactic acid and serve to maintain homeostasis between free CoA, acetyl-CoA, free carnitines, and acylcarnitines (Vappiani et al., 2021). One of the two media ("Medium B") increased significantly viable-cell count and antibody titer than Medium A. Here, we report that CHO's mitochondrial dysfunctionality based on the secretion of acylcarnitines in lactic acid-fed cultures depends on the overall medium composition. We hypothesize that in order to achieve better growth and titer, Medium B exhibited an increased oxidative phosphorylation based on the lower secretion of acylcarnitines and a differential utilization of riboflavin and thiamine, precursors of coenzymes required to enhance mitochondrial pyruvate incorporation and TCA cycle function. Therefore, our data provides further evidence that non-obvious changes to medium composition can have substantial effects on CHO-based production processes by altering the activity of oxidative phosphorylation required for the proper functioning of mitochondria but also for better antibody production.
    Keywords:  Chinese hamster ovary (CHO) cells; Tricarboxylic acid (TCA) cycle; carnitines; media optimization; metabolomics; mitochondrial disfunction
    DOI:  https://doi.org/10.1016/j.jbiotec.2026.01.002
  41. 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
  42. Genome Med. 2026 Jan 09.
       BACKGROUND: Singleton short-read genome sequencing (GS) is increasingly used as a first-line genetic test for childhood neurological disorders (such as intellectual disability, neurodevelopmental delay, motor delay, and hypotonia) with diagnostic yields from 26 to 35%, typically involving a mix of single nucleotide variants and small insertions/deletions (SNV/INDELs), structural variants (SVs), and short tandem repeats (STRs). Long-read GS is emerging as an attractive alternative, offering a more comprehensive assessment of the genome, but its utility still needs to be systematically evaluated in a clinical diagnostic setting.
    METHODS: We prospectively included 100 children and adolescents (≤ 20 years) with neurological disorders, newly referred for genetic testing. Routine DNA was used for singleton standard clinical short-read GS in parallel with long-read GS (Oxford Nanopore Technologies). In addition to comprehensive variant calling, long-read GS data was also phased and underwent methylation analysis. Variant interpretation was restricted to in-silico gene panels targeting either intellectual disability (1,568 genes) or neuromuscular disorders (1,035 genes) depending on the clinical presentation.
    RESULTS: The long-read GS generated an average of 111 GB data per sample, with a median read-length of 5 kb and average N50 of 16 kb; resulting in an average coverage of 34X. Short-read and long-read GS identified the same 29% diagnostic yield, including SNV/INDELs (n = 18), SVs (n = 9), STRs (n = 1), and uniparental disomy (n = 1). Long-read GS provided additional diagnostic value in 13 cases involving 17 distinct variants, including phasing of SMN1 and biallelic SNVs/INDELs in autosomal recessive genes, accurate determination of STR length and sequence as well as detailed structural characterization of SVs. Of note, an unbalanced translocation, der(14)t(8;14)(p11.2;p23.1), required de novo assembly and T2T-CHM13 alignment to resolve the breakpoint junctions. Furthermore, long-read GS detected disease-associated aberrant methylation patterns in the Prader-Willi region and across an FMR1 expansion.
    CONCLUSIONS: In a clinical diagnostic setting, long-read GS proved to be a streamlined, first-line test, capturing the full spectrum of disease-causing variants, reducing the need for follow-up testing and enabling more precise interpretation. While the overall diagnostic yield may be comparable to that of short-read approaches, long-read GS offers significant added value across multiple variant types.
    Keywords:  Chromosomal rearrangements; Clinical diagnostics; Long-read sequencing; Methylation analysis; Rare diseases; Short tandem repeat expansions; Short-read sequencing; Single nucleotide variants; Structural variants; Whole genome sequencing
    DOI:  https://doi.org/10.1186/s13073-025-01596-5
  43. 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
  44. Mol Genet Metab. 2026 Jan 02. pii: S1096-7192(25)00707-3. [Epub ahead of print]147(3): 109715
      Mutations in the Ndufs4 gene encoding the accessory subunit of complex I (CI) of the mitochondrial oxidative phosphorylation (OXPHOS) system, are the most common causes of Leigh Syndrome (LS). LS is a severe infantile neurodegenerative disorder characterised by various clinical phenotypes ranging from ataxia, cardiomyopathy, swallowing difficulties, visual problems, psychomotor regression to fatal respiratory failure. The mechanistic processes contributing to the onset and progression of these clinical manifestations remain poorly understood. This study investigates tissue-specific proteomic changes in a mouse model of LS using quantitative proteomics as a hypothesis-generating technique. Six distinct tissues, namely three brain regions (brainstem, cerebellum, olfactory bulb), heart, kidney, and liver, were collected from the LS mouse model (Ndufs4 KO mice) and compared to wild type (WT) controls using SWATH-MS analysis as a data acquisition method. Functional enrichment analysis revealed distinct tissue-specific cellular responses which include a shift toward amino acid metabolism in the heart, increased mitochondrial translation in the kidney, and alterations in phase II detoxification pathways in the liver. Our results unravel candidate mechanisms for tissue-specific vulnerability and highlight the regulation of PTEN gene transcription as potential driver of neurodegeneration. These findings provide data-driven hypotheses for tissue-specific vulnerability in LS, highlighting potential mechanisms and therapeutic targets. This study established a foundation for future hypothesis-driven research into the tissue-specific pathophysiology of mitochondrial disease.
    Keywords:  Leigh syndrome; Mitochondrial disease; Ndufs4 KO; Proteomics; SWATH-MS; Tissue specificity
    DOI:  https://doi.org/10.1016/j.ymgme.2025.109715
  45. J Drug Target. 2026 Jan 06. 1-32
      Mitochondria-targeted antioxidants can selectively accumulate within mitochondria at low doses, thereby significantly enhancing therapeutic efficiency while minimizing potential side effects. SKQ1, a novel mitochondria-targeted antioxidant, operates through a well-defined mechanism: a lipophilic cation enables mitochondrial targeting, while plastoquinone exerts antioxidant activity. SKQ1 primarily exerts its potent antioxidative effects by directly neutralizing reactive oxygen species (ROS), thereby protecting mitochondrial function. Numerous studies have explored the biological functions of SKQ1, identifying its significant potential in anti-aging, immune regulation, and antimicrobial activity. In this review, we summarize all available therapeutic evidence of SKQ1. We propose that SKQ1 represents a promising candidate for treating mitochondrial dysfunction-related diseases; however, its safety profile warrants further investigation.
    Keywords:  Aging; MitoQ; Mitochondria-targeted antioxidants; ROS; SKQ1
    DOI:  https://doi.org/10.1080/1061186X.2026.2613054
  46. bioRxiv. 2025 Dec 26. pii: 2025.12.24.696399. [Epub ahead of print]
      Mitochondria and nucleotide metabolism are critical for cellular and developmental homeostasis, yet their potential interdependence and role in neurodevelopmental disease remain unclear. In MECP2 Duplication Syndrome (MDS), we identify a conserved correlation between mitochondrial function and purine metabolism that is disrupted across human, organoid, and mouse models. Multiomics integration reveals Complex III as the focal point of mitochondrial collapse, leading to redox stress, DNA damage, and hyperactivation of the de novo purine biosynthesis via purinosome assembly. The breakdown of mitochondria-purinosome coupling compromises genome stability, impairs radial glia proliferation, and delays neuronal maturation. By linking a defined genetic dosage imbalance to metabolic network failure, our study positions the mitochondria-purinosome coordination as a fundamental control axis for neurodevelopment and a therapeutic entry point across metabolic and neurodevelopmental disorders.
    Keywords:  MeCP2; brain organoids; metabolome; mitochondria; multiomics; neuroprogenitors; purine
    DOI:  https://doi.org/10.64898/2025.12.24.696399
  47. Mol Ther Nucleic Acids. 2026 Mar 12. 37(1): 102786
      The gene therapy landscape has evolved substantially in recent years, beginning with the approval of the first adeno-associated virus-based gene therapy, Luxterna, in 2017. Since then, the US FDA has approved nearly 30 new viral gene therapy programs, with notable examples including Zolgensma, Spinraza, Hemgenix, Zynteglo, Lyfgenia, Kymriah, Skysona, and Tecelra. Remarkably, all these products rely on delivery via adeno-associated vectors (AAVs) and lentiviral vectors (LVs). Improvements in viral-mediated gene transfer efficiency and clinical-scale manufacturing, together with immense commercial interest, have greatly propelled the clinical adoption of gene therapy products. In recent years, clustered regularly interspaced short palindromic repeats (CRISPR) and its related Cas proteins (CRISPR-Cas) have made significant advances in gene therapy, offering next-generation approaches for curative gene editing to treat genetic diseases and disorders. In this review, we examine the range of these therapeutics and their viral carriers, focusing primarily on LVs and AAVs. We provide a snapshot of the current status of the field and highlight some of the current challenges in the clinical application of gene therapy, with particular emphasis on viral CRISPR-Cas-based technologies and their future potential.
    Keywords:  AAV; CRISPR-Cas9; CRISPR-associated protein 9; MT: Delivery Strategies; adeno-associated vectors; all-in-one delivery system; base editing; clinical trials; clustered regularly-interspaced short palindromic repeats; epigenome-based editing; gene delivery; gene therapy; lentiviral vectors; prime editing
    DOI:  https://doi.org/10.1016/j.omtn.2025.102786
  48. Anal Chem. 2026 Jan 09.
      Mitochondrial peroxynitrite (ONOO-), a highly reactive nitrogen species, is critically involved in oxidative-stress-induced cell damage, playing vital roles in the pathogenesis of numerous diseases. However, the real-time detection of ONOO- in mitochondria is still challenging due to the reliance of existing probes on mitochondrial membrane potential (MMP), often disrupted under disease conditions. Therefore, we attempted to develop and investigate an MMP-independent, near-infrared fluorescent probe (Mito-ONOO-). To achieve this, a series of fluorophores Flu-(1-5) was rationally constructed by introducing alkyl chains of varying lengths (C2, C4, C8, C12, and C16) to fine-tune their lipophilicity and mitochondrial retention potential. Flu-(1-5) were initially screened for their lipophilicity and mitochondria-targeting capability under ONOO--stimulated stress. Flu-5 was finally selected for its optimal combination of notable lipophilicity and mitochondrial retention to construct the Mito-ONOO-. Upon evaluation, Mito-ONOO- displayed a reputable Stokes shift (88 nm), fast response (around 1 min), and excellent mitochondrial localization potential (PC = 0.96). In addition, the designed probe demonstrated excellent selectivity and sensitivity, enabling precise visualization of mitochondrial ONOO- in living HepG2 cells. Furthermore, the probe was successfully employed to monitor ONOO- dynamics during a drug-induced liver injury (DILI) mice model. Collectively, the findings of this study underscore the potential of Mito-ONOO- as a powerful tool for exploring mitochondrial redox homeostasis and its perturbation under pathological conditions.
    DOI:  https://doi.org/10.1021/acs.analchem.5c06669
  49. Science. 2026 Jan 08. eadv7953
      Conformational biasing (CB) is a rapid and streamlined computational method that uses contrastive scoring by inverse folding models to predict protein variants biased toward desired conformational states. We successfully validated CB across seven diverse datasets, identifying variants of K-Ras, SARS-CoV-2 spike, β2 adrenergic receptor, and Src kinase with improved conformation-specific functions, such as enhanced binding or enzymatic activity. Applying CB to the enzyme lipoic acid ligase (LplA), we uncovered a previously unknown mechanism controlling its promiscuous activity. Variants biased toward an "open" conformation state became more promiscuous, whereas "closed"-biased variants were more selective, enhancing LplA's utility for site-specific protein labeling with fluorophores in living cells. The speed and simplicity of CB make it a versatile tool for engineering protein dynamics with broad applications in basic research, biotechnology, and medicine.
    DOI:  https://doi.org/10.1126/science.adv7953
  50. bioRxiv. 2025 Dec 30. pii: 2025.12.30.693754. [Epub ahead of print]
      Mitochondria are dynamic organelles that can fragment or fuse to support different bioenergetic demands (e.g., glycolysis vs. oxidative phosphorylation) and distinct cell behaviors (e.g., mitosis or migration). While the role of mitochondrial dynamics in wound healing and metabolic disorders has received significant attention, the role of mitochondrial fission and fusion during normal embryonic development is less well understood--in part due to the difficulty of studying such processes in vivo . Combined with the depth-resolved imaging capabilities of multiphoton microscopy, fluorescence lifetime imaging (FLIM) of the mitochondrial cofactor NADH can be used to simultaneously visualize mitochondrial network morphology and infer certain aspects of cellular bioenergetics (e.g., glycolysis vs. oxidative phosphorylation) in a label-free, non-invasive manner. Here we demonstrate that NADH FLIM can be used to accurately track the subcellular localization and topology of mitochondrial networks in live Drosophila embryos. We used this technique to assess whether cells show changes in NADH lifetime during convergent extension (CE)--a conserved process of tissue remodeling in which thousands of germband cells undergo coordinated intercalation to drive elongation of the head-to-tail axis. Contrary to our expectations, we did not observe significant changes in NADH lifetime or network appearance during CE in wild-type embryos, suggesting that germband cells do not need to alter their baseline metabolism to fuel cell intercalation during normal development. To directly assess the role of mitochondrial fission and fusion during CE, we used RNA interference to disrupt the fission mediator Drp1 and the fusion mediator Opa1 . Consistent with expectations, inhibiting mitochondrial fission in Drp1- knockdown embryos led to hyper-fused networks and significantly longer NADH lifetimes, indicating a shift towards oxidative phosphorylation. Conversely, inhibiting mitochondrial fusion in Opa1- knockdown embryos led to more hyper-fragmented networks and significantly shorter NADH lifetimes, indicating a shift towards glycolysis. Interestingly, inhibiting either fission or fusion altered tissue elongation and greatly increased the rate of cell intercalation errors, suggesting that a precise network topology is required for proper CE. We hypothesize that the CE defects in Drp1 -knockdown embryos are primarily due to incorrect basal subcellular localization of mitochondria, whereas the CE defects in Opa1 -knockdown embryos are due to deficient ATP and/or ROS production. These experiments demonstrate the utility of FLIM-based applications for characterizing the role of mitochondria during normal embryonic development, which could yield a better understanding of the metabolic underpinnings of various pathologies that involve epithelial remodeling, including spina bifida, defective wound healing, and cancer metastasis.
    DOI:  https://doi.org/10.64898/2025.12.30.693754
  51. Sci Rep. 2026 Jan 09.
      Dysfunctional cGMP signaling is implicated in multiple neurodegenerative diseases of the central nervous system (CNS), including glaucoma, an optic neuropathy and leading cause of irreversible blindness. Female mice lacking the alpha catalytic subunit of soluble guanylate cyclase (sGCα1-/-), the active site that binds nitric oxide (NO) to produce cGMP, exhibit progressive retinal ganglion cell (RGC) degeneration with age. Yet, the role of sGC in age- and sex-dependent RGC function remains uncharacterized. We investigated how preventing NO binding to sGC influences RGC function in the context of aging and sex by combining bulk and single-cell RNA sequencing, Western blotting, mitochondrial ultrastructural analysis, visual acuity measurements, and in vivo measurements of retinal oxidative metabolism. We found that global sGCα1 deletion impairs visual function and RGC health in aging female mice, while male mice remained unaffected. Glucose uptake was significantly disrupted in female sGCα1-/-retinas with age, and accompanied by reduced retinal expression of the glucose transporter, GLUT1. Aged sGCα1-/- females also exhibited dysregulated retinal mitochondrial gene and protein expression and increased nitrosative stress localized to the RGC layer. RGC mitochondria in male mice increased in size with age, while female mitochondria did not. Furthermore, retinal metabolic analysis showed decreased oxygen consumption rate in aged female but not male sGCα1-/-retinas, suggesting impaired oxidative metabolism. These findings reveal a potential sex-specific role for cGMP signaling in maintaining retinal metabolic integrity and RGC function with age. Our results point to a possible mechanistic link between impaired cGMP signaling and age-related retinal neurodegeneration in females, highlighting the sGC-cGMP signaling pathway as a promising therapeutic target for glaucoma and other CNS neurodegenerative diseases.
    Keywords:  CGMP; Glucose; Mitochondria; Neurodegeneration; Retinal ganglion cell; Retinal metabolism
    DOI:  https://doi.org/10.1038/s41598-025-34243-5
  52. Int J Mol Sci. 2025 Dec 23. pii: 156. [Epub ahead of print]27(1):
      Precision medicine starts with a precision diagnosis. Yet up to 80% of cases of monogenic diabetes, a form of diabetes characterized by mutations in a single gene, are either overlooked or misdiagnosed. A genetic test for monogenic diabetes does not always lead to a precise diagnosis, as novel variants are often classified as variants of unknown significance. Variant interpretation requires collation of a framework of evidence, including population, computational, and segregation data, and can be assisted by functional analysis. The inclusion of functional data can be challenging, depending on the number of benign and pathogenic variants available for benchmarking assays. Glucokinase is the rate-limiting step for glucose metabolism in the pancreatic beta-cell and governs the threshold for glucose-stimulated insulin release. Loss-of-function alleles in the glucokinase (GCK) gene are a cause of stable fasting hyperglycemia from birth and/or diabetes. In this study, we functionally characterized 25 variants identified during diagnostic testing or in exome sequencing studies. We assessed their kinetic characteristics, stability, and interaction with pharmacological and physiological regulators. We integrated our functional data with existing data from the ClinGen Monogenic Diabetes Variant Curation Expert Review panel using a gene-specific framework to assist variant classification. We show how functional evidence can aid variant classification, thus enabling diagnostic certainty.
    Keywords:  MODY; VUS; glucokinase; hypoglycemia; monogenic diabetes
    DOI:  https://doi.org/10.3390/ijms27010156
  53. Nat Genet. 2026 Jan 09.
    Mathieu Quinodoz, Kim Rodenburg, Zuzana Cvackova, Karolina Kaminska, Suzanne E de Bruijn, Ana Belén Iglesias-Romero, Erica G M Boonen, Mukhtar Ullah, Nick Zomer, Marc Folcher, Jacques Bijon, Lara K Holtes, Stephen H Tsang, Zelia Corradi, K Bailey Freund, Stefanida Shliaga, Daan M Panneman, Rebekkah J Hitti-Malin, Manir Ali, Ala'a AlTalbishi, Sten Andréasson, Georg Ansari, Gavin Arno, Galuh D N Astuti, Carmen Ayuso, Radha Ayyagari, Sandro Banfi, Eyal Banin, Tahsin Stefan Barakat, Mirella T S Barboni, Miriam Bauwens, Tamar Ben-Yosef, Virginie Bernard, David G Birch, Pooja Biswas, Fiona Blanco-Kelly, Beatrice Bocquet, Camiel J F Boon, Kari Branham, Dominique Bremond-Gignac, Alexis Ceecee Britten-Jones, Kinga M Bujakowska, Cyril Burin des Roziers, Elizabeth L Cadena, Giacomo Calzetti, Francesca Cancellieri, Luca Cattaneo, Naomi Chadderton, Peter Charbel Issa, Luísa Coutinho-Santos, Stephen P Daiger, Elfride De Baere, Marieke De Bruyne, Berta de la Cerda, John N De Roach, Julie De Zaeytijd, Ronny Derks, Claire-Marie Dhaenens, Lubica Dudakova, Jacque L Duncan, G Jane Farrar, Nicolas Feltgen, Beau J Fenner, Lidia Fernández-Caballero, Juliana M Ferraz Sallum, Simone Gana, Alejandro Garanto, Jessica C Gardner, Christian Gilissen, Roser Gonzàlez-Duarte, Kensuke Goto, Sam Griffiths-Jones, Tobias B Haack, Lonneke Haer-Wigman, Alison J Hardcastle, Takaaki Hayashi, Elise Héon, Lies H Hoefsloot, Alexander Hoischen, Josephine P Holtan, Carel B Hoyng, Manuel Benjamin B Ibanez, Chris F Inglehearn, Takeshi Iwata, Brynjar O Jensson, Kaylie Jones, Vasiliki Kalatzis, Smaragda Kamakari, Marianthi Karali, Ulrich Kellner, Caroline C W Klaver, Krisztina Knézy, Robert K Koenekoop, Susanne Kohl, Taro Kominami, Laura Kühlewein, Tina M Lamey, Rina Leibu, Bart P Leroy, Petra Liskova, Irma Lopez, Victor R de J López-Rodríguez, Quinten Mahieu, Omar A Mahroo, Gaël Manes, Luke Mansard, M Pilar Martín-Gutiérrez, Nelson Martins, Laura Mauring, Martin McKibbin, Terri L McLaren, Isabelle Meunier, Michel Michaelides, José M Millán, Kei Mizobuchi, Rajarshi Mukherjee, Zoltán Zsolt Nagy, Kornelia Neveling, Monika Ołdak, Michiel Oorsprong, Yang Pan, Anastasia Papachristou, Antonio Percesepe, Maximilian Pfau, Eric A Pierce, Emily Place, Raj Ramesar, Francis Ramond, Florence Andrée Rasquin, Gillian I Rice, Lisa Roberts, María Rodríguez-Hidalgo, Javier Ruiz-Ederra, Ataf H Sabir, Ai Fujita Sajiki, Ana Isabel Sánchez-Barbero, Asodu Sandeep Sarma, Riccardo Sangermano, Cristina M Santos, Margherita Scarpato, Hendrik P N Scholl, Dror Sharon, Sabrina G Signorini, Francesca Simonelli, Ana Berta Sousa, Maria Stefaniotou, Kari Stefansson, Katarina Stingl, Akiko Suga, Patrick Sulem, Lori S Sullivan, Viktória Szabó, Jacek P Szaflik, Gita Taurina, Alberta A H J Thiadens, Carmel Toomes, Viet H Tran, Miltiadis K Tsilimbaris, Pavlina Tsoka, Veronika Vaclavik, Marie Vajter, Sandra Valeina, Enza Maria Valente, Casey Valentine, Rebeca Valero, Sophie Valleix, Joseph van Aerschot, L Ingeborgh van den Born, Mattias Van Heetvelde, Virginie J M Verhoeven, Andrea L Vincent, Andrew R Webster, Laura Whelan, Bernd Wissinger, Georgia G Yioti, Kazutoshi Yoshitake, Juan C Zenteno, Roberta Zeuli, Theresia Zuleger, Chaim Landau, Allan I Jacob, Siying Lin, Frans P M Cremers, Winston Lee, Jamie M Ellingford, David Stanek, Susanne Roosing, Carlo Rivolta.
      Small nuclear RNAs (snRNAs) combine with specific proteins to generate small nuclear ribonucleoproteins (snRNPs), the building blocks of the spliceosome. U4 snRNA forms a duplex with U6 and, together with U5, contributes to the tri-snRNP spliceosomal complex. Variants in RNU4-2, which encodes U4, have recently been implicated in neurodevelopmental disorders. Here we show that heterozygous inherited and de novo variants in RNU4-2 and in four RNU6 paralogs (RNU6-1, RNU6-2, RNU6-8 and RNU6-9), which encode U6, recur in individuals with nonsyndromic retinitis pigmentosa (RP), a genetic disorder causing progressive blindness. These variants cluster within the three-way junction of the U4/U6 duplex, a site that interacts with tri-snRNP splicing factors also known to cause RP (PRPF3, PRPF8, PRPF31), and seem to affect snRNP biogenesis. Based on our cohort, deleterious variants in RNU4-2 and RNU6 paralogs may explain up to ~1.4% of otherwise undiagnosed RP cases. This study highlights the contribution of noncoding RNA genes to Mendelian disease and reveals pleiotropy in RNU4-2, where distinct variants underlie neurodevelopmental disorder and retinal degeneration.
    DOI:  https://doi.org/10.1038/s41588-025-02451-4
  54. bioRxiv. 2025 Dec 23. pii: 2025.12.21.695848. [Epub ahead of print]
      Ferroptosis is an iron-catalyzed lipid peroxidation (LP)-dependent cell death. Induction of mitochondrial ROS (mtROS) is crucial in the execution of ferroptosis, but the underlying mechanism remains unclear. Through utilizing the hepatocyte model and RNA-seq analysis, we determined mtROS-dependent metabolic changes that modulate ferroptosis sensitivity. Elevated mtROS production and LP suppressed glycolysis, fatty acid oxidation, and citric acid cycle activity, representing adaptive responses that protect cells from ferroptosis. On the other hand, mtROS-driven signaling impaired glutathione biosynthesis and downregulated genes involved in coenzyme Q10 (CoQ) biosynthesis, including those in the mevalonate pathway and CoQ8A, a key stabilizer of the CoQ biosynthetic complex. Importantly, silencing CoQ8A expression enhanced, whereas overexpression of CoQ8A reduced, ferroptosis susceptibility of hepatocytes and various cancer cell types. The mtROS-mediated downregulation of CoQ8A was dependent on farnesoid X receptor (FXR) and retinoid X receptors (RXRs). Collectively, our findings highlight that mtROS promotes ferroptosis, at least in part, by suppressing glutathione and CoQ biosynthesis.
    DOI:  https://doi.org/10.64898/2025.12.21.695848
  55. 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
  56. Stem Cell Res Ther. 2026 Jan 09.
      Friedreich's ataxia (FRDA) is an inherited, autosomal recessive, multisystem disorder that primarily manifests in children and affects the nervous system and the heart. FRDA is caused by an expansion of GAA repeats in the first intron of the frataxin (FXN) gene. The expansion disrupts transcription of FXN, resulting in significantly decreased FXN expression in FRDA patients' tissues. Frataxin is involved in biosynthesis of iron-sulfur (Fe-S) clusters, which are critical for the function of the electron transport chain and many metabolic enzymes. Frataxin deficiency leads to reduced energy production and accumulation of iron in mitochondria that exacerbates oxidative stress. Despite significant advancements in the field, FXN cellular functions and underlying pathological mechanisms of FXN deficiency in cell-type specific contexts remain to be elucidated. Inaccessibility to the most vulnerable cell types in FRDA patients, including neurons, cardiomyocytes, and β-cells, largely accounts for these limitations. Significant progress in recent years regarding the derivation and differentiation of human pluripotent stem cells (hPSCs), along with breakthroughs in gene editing technologies, enables the generation of patient-derived and isogenic control disease-relevant cell types and organoid-like structures as platforms for studying disease mechanisms and for drug discovery. Herein, we first provide an overview of hPSC derivation and intrinsic properties of these cells. We then discuss current advances and limitations of hiPSC-based cell models for FRDA. We also highlight the need to further refine and develop these in vitro cell models for pre-clinical advancement of therapeutic approaches for FRDA.
    Keywords:   In vitro differentiation; Astrocytes; Cardiomyocytes; Disease modeling; Friedreich’s ataxia; Neurons; Organoids; Pluripotent stem cells; β-cells
    DOI:  https://doi.org/10.1186/s13287-025-04861-x
  57. Science. 2026 Jan 08. 391(6781): eads9530
      Recent breakthroughs in protein structure prediction have opened new avenues for genome-wide drug discovery, yet existing virtual screening methods remain computationally prohibitive. We present DrugCLIP, a contrastive learning framework that achieves ultrafast and accurate virtual screening, up to 10 million times faster than docking, while consistently outperforming various baselines on in silico benchmarks. In wet-lab validations, DrugCLIP achieved a 15% hit rate for norepinephrine transporter, and structures of two identified inhibitors were determined in complex with the target protein. For thyroid hormone receptor interactor 12, a target that lacks holo structures and small-molecule binders, DrugCLIP achieved a 17.5% hit rate using only AlphaFold2-predicted structures. Finally, we released GenomeScreenDB, an open-access database providing precomputed results for ~10,000 human proteins screened against 500 million compounds, pioneering a drug discovery paradigm in the post-AlphaFold era.
    DOI:  https://doi.org/10.1126/science.ads9530
  58. bioRxiv. 2026 Jan 02. pii: 2025.12.23.696269. [Epub ahead of print]
      Substance use disorders and other mental health conditions often co-occur with metabolic disorders, suggesting shared biological underpinnings 1 . These heightened behavioral and physiological responses may have evolved to promote survival during resource scarcity but can become maladaptive in modern environments 2 . The genetic mechanisms linking these traits have remained elusive. Here, we show that a variable gene enhancer in mice jointly regulates genes encoding an epigenetic factor ( Eed ) and a mitochondrial enzyme ( Me3 ) thereby influencing propensity to addiction-related behaviors and mitochondrial function. We further identify variation in a corresponding enhancer in humans regulating EED and ME3 associated with substance use, psychiatric and metabolic disorders. These findings reveal a convergent genetic regulatory network linking mitochondrial biology to behavioral and metabolic risk, offering insight into how genetic variation in beneficial regulatory pathways can predispose individuals to substance use disorders and related conditions.
    DOI:  https://doi.org/10.64898/2025.12.23.696269
  59. Nat Chem Biol. 2026 Jan 09.
      Lipid droplets (LDs) dynamically interact with other organelles, such as mitochondria, in surveillance of cellular metabolic homeostasis. The transient nature of LDs, however, poses technical challenges to snapshot molecular information underlying these interactions. Herein, we present a small-molecule-based photocatalytic protein proximity labeling method (named LipoID) to enable in situ labeling, capturing and profiling of the LD-interacting proteome. This method is enabled by a set of LD-targeting probes designed to catalyze protein modifications nearby LDs using nucleophilic substrates. Profiled by liquid chromatography-tandem mass spectrometry, LipoID identifies tethered interorganellar interactions, particularly with mitochondria, in addition to reliable capture of validated LD biomarkers (for example, perilipins (PLINs)). Coupled with comparative proteomics, LipoID discovers mitochondrial voltage-dependent anion channel 3 as a potential regulator of LD-mitochondria proximity through interacting with PLIN3 on LDs. Further metabolomics analysis suggested remodeled lipid metabolism in line with the LD-mitochondria interaction. Together, LipoID enables in situ profiling of the LD interactome and reveals interorganellar regulation.
    DOI:  https://doi.org/10.1038/s41589-025-02127-4
  60. Cell Death Dis. 2026 Jan 08. 17(1): 10
      As a pathological hallmark of Parkinson's disease (PD), a-synucleinopathy induces various cellular damages, including calcium overload, mitochondrial and autophagic dysfunction, ultimately resulting in dopaminergic neuron death. However, the hierarchy of these detrimental events remains unclear. It is well established that a-synuclein can induce calcium overload through diverse mechanisms. To assess whether calcium overload plays a crucial detrimental role, we established a calcium overload model in Drosophila and conducted genetic screening. Our findings indicate that calcium overload caused mitochondrial damage and lysosomal dysfunction, leading to cell death, and these cytotoxic processes were significantly mitigated by the loss of Tousled-like kinase (TLK). Notably, the loss of TLK also ameliorated defects induced by a-synuclein overexpression in Drosophila. This suggests that calcium overload is a critical event in a-synucleinopathy. In mammalian cells and mice, calcium overload activated TLK2 (the homologue of Drosophila TLK) by enhancing TLK2 phosphorylation, which increases TLK2 kinase activity. Increased TLK2 phosphorylation was detected in the brains of GluR1Lc and a-synuclein overexpression mice, suggesting that TLK2 is activated under these pathological conditions. Furthermore, TLK2 knockout mice exhibited rescue of multi-aspect cytotoxicity induced by calcium overload and a-synuclein overexpression. Our research demonstrates that TLK2 activation by calcium overload appears to be a pivotal step in the progression of PD. This finding provides a potential link between calcium overload, the subsequent mitochondrial and lysosomal dysfunction observed in the disease.
    DOI:  https://doi.org/10.1038/s41419-025-08213-8
  61. Front Biosci (Elite Ed). 2025 Dec 18. 17(4): 44344
       BACKGROUND: Parkinson's disease (PD) is a progressive neurodegenerative disorder with which the leucine-Rich repeat kinase 2 glycine 2019 serine (LRRK2 G2019S) mutation is strongly associated. This mutation elevates kinase activity, disrupts mitochondrial function, increases reactive oxygen species (ROS) production, and impairs DNA repair mechanisms, all of which contribute to the pathogenesis of PD. Thus, addressing these pathological features through targeted delivery systems holds promise for more effective therapies.
    METHODS: This study aimed to investigate the use of Ginkgo biloba leaf extract (EGB) to synthesize sphingomyelin-cholesterol solid lipid nanoparticles (SLNPs) functionalized with poly-L-lysine (EGB-PLL-SLNPs) for siRNA delivery targeting the LRRK2 G2019S mutation. SLNPs suspended in water (H₂O-PLL-SLNPs) served as the comparator. In vitro assays were conducted using either wild-type or LRRK2 G2019S-transformed SH-SY5Y and HEK293 cells. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was employed to evaluate the cytotoxicity of the SLNPs and nanocomplexes; meanwhile, flow cytometry was used to assess caspase 3/7 activity, mitochondrial membrane potential, DNA damage, and ROS levels.
    RESULTS: Treatment with EGB-PLL-SLNPs significantly improved mitochondrial health, reducing depolarized and dead cells and enhancing overall cell viability. ROS levels, DNA damage and kinase activity were significantly decreased compared to the control H₂O-PLL-SLNPs.
    CONCLUSION: The enhanced therapeutic outcomes observed with the EGB-PLL-SLNPs can be attributed to the bioactive compounds in EGB, particularly the flavonoids and terpenoids, such as quercetin and kaempferol. These molecules play crucial roles in stabilizing mitochondrial membranes, facilitating ATP synthesis, and regulating genes linked to mitochondrial biogenesis. The interaction between EGB and siRNA to mediate gene silencing provides a multifaceted approach to counteracting PD pathophysiology. This study demonstrates that EGB-PLL-SLNPs offer superior gene silencing and cytoprotective effects compared to conventional formulations. The integration of plant-based bioactives with nanomedicine enhances therapeutic delivery and efficacy, positioning biosynthesized PLL-SLNPs as a promising strategy for treating Parkinson's disease.
    Keywords:  Ginkgo biloba; LRRK2 G2019S ; Parkinson's disease; SLNPs; gene therapy; nanomedicine; siRNA
    DOI:  https://doi.org/10.31083/FBE44344
  62. Elife. 2026 Jan 08. pii: RP105418. [Epub ahead of print]14
      Mutations in the TANGO2 gene are associated with a severe neurometabolic disorder in humans, often presenting with life-threatening metabolic crisis. However, the function of TANGO2 protein remains unknown. It has recently been proposed that TANGO2 transports heme within and between cells, from areas with high heme concentrations to those with lower concentrations. Here, we demonstrate that prior heme-related observations in Caenorhabditis elegans lacking TANGO2 homologs HRG-9 and HRG-10 may be better explained by a previously unreported metabolic phenotype, characterized by reduced feeding, decreased lifespan and brood sizes, and poor motility. We also show that several genes not implicated in heme transport are upregulated in the low heme state and conversely demonstrate that hrg-9 in particular is highly responsive to oxidative stress, independent of heme status. Collectively, these data implicate bioenergetic failure and oxidative stress as potential factors in the pathophysiology of TANGO2 deficiency, in alignment with observations from human patients. Our group performed several experiments in yeast and zebrafish deficient in TANGO2 homologs and was unable to replicate prior findings from these models. Overall, we believe there is insufficient evidence to support heme transport as the primary function for TANGO2.
    Keywords:  C. elegans; S. cerevisiae; TANGO2 deficiency; cell biology; genetics; genomics; heme metabolism; metabolic dysfunction; mitochondria; oxidative stress; rare disease; zebrafish
    DOI:  https://doi.org/10.7554/eLife.105418
  63. Transl Pediatr. 2025 Dec 31. 14(12): 3420-3428
       Background: There was limited research comparing retinal nerve fibre layer (RNFL) involvement among different mitochondrial DNA (mtDNA) mutations associated with varying visual prognoses. This study aimed to observe and compare the thickness changes of RNFL among Leber's hereditary optic neuropathy (LHON) patients with G11778A, T14484C and G3460A mtDNA mutations.
    Methods: A retrospective cross-sectional analysis was conducted on LHON patients with G11778A (189 eyes of 121 patients), T14484C (20 eyes of 10 patients), or G3460A (28 eyes of 15 patients) mutations, enrolled between July 2017 and December 2020. We also recruited age-matched healthy individuals as the healthy control group. Patients were grouped based on their mutation type and disease duration (<6, 6-12, and >12 months). RNFL thickness measurements were obtained for all quadrants and compared among the three mutation groups.
    Results: During the subacute phase, the temporal quadrant RNFL thickness in LHON patients with G11778A, T14484C, or G3460A mutations was significantly reduced compared to healthy controls. In the dynamic phase, the RNFL thickness in all quadrants of G11778A or G3460A LHON patients was significantly thinned, except for the nasal quadrant in LHON patients with G3460A mtDNA mutation (P=0.29). As the disease progressed, RNFL thickness in all quadrants and average RNFL thickness were significantly thinner in all three mutation groups relative to controls, except for the nasal quadrant in T14484C mutation patients (P=0.10). Furthermore, during both the subacute and dynamic phases, RNFL thickness in all quadrants was thinner in G11778A and G3460A mutation patients compared to T14484C mutation patients.
    Conclusions: The papillomacular bundle was the initial and preferential site of involvement in LHON patients across all mutation types. The pattern of RNFL involvement was similar among the three mutations: temporal quadrant thinning occurred first, followed by the inferior and superior quadrants, and finally the nasal quadrant. Patients with G11778A and G3460A mutations exhibited earlier and more pronounced RNFL atrophy compared to those with T14484C mutations.
    Keywords:  Leber’s hereditary optic neuropathy (LHON); optical coherence tomography (OCT); retinal nerve fibre layer thickness (RNFL thickness)
    DOI:  https://doi.org/10.21037/tp-2025-589
  64. Rev Neurol. 2025 Dec 18. 80(11): 44110
       BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a rare neurodegenerative disease that mostly presents as sporadic cases. Currently, no mitochondrial-related gene mutations have been identified as the cause of ALS. Mitochondrial gene mutations cause rare hereditary diseases, and the symptoms of pure muscle weakness and muscle atrophy are rarely observed.
    CASE REPORT: We report the case of a young patient clinically diagnosed with ALS concurrently associated with a pathogenic mutation in the mitochondrially encoded nicotinamide adenine dinucleotide: ubiquinone oxidoreductase core subunit 6 (MT-ND6) gene. However, the pathogenic relationship between the MT-ND6 gene and ALS has not been confirmed.
    CONCLUSION: We provide a case report and a literature review aimed at increasing the understanding of the connection between the two. It is essential to consider the potential modifying role of mitochondrial pathogenic genes in ALS.
    Keywords:  Leber’s hereditary optic neuropathy; MT-ND6 gene; amyotrophic lateral sclerosis; m.14484T>C; muscle weakness
    DOI:  https://doi.org/10.31083/RN44110
  65. Nat Biotechnol. 2026 Jan 05.
      Transcript diversity including splicing and alternative 3' end usage is crucial for cellular identity and adaptation, yet its spatial coordination remains poorly understood. Here we present SPLISOSM (spatial isoform statistical modeling), a method for detecting isoform-resolution patterns from spatial transcriptomics data. SPLISOSM uses multivariate testing with nonparametric kernels to account for spot-level and isoform-level dependencies, achieving high statistical power on sparse data. In the mouse brain, we identify over 1,000 spatially variable transcript diversity events, primarily in synaptic signaling pathways linked to neuropsychiatric disorders, and uncover both known and previously unknown regulatory relationships with region-specific RNA binding proteins. We further show that these patterns are evolutionarily conserved between mouse and human prefrontal cortex. Analysis of human glioblastoma highlights pervasive transcript diversity in antigen presentation and adhesion genes associated with specific microenvironmental conditions. Together, we present a comprehensive spatial splicing analysis in the brain under normal and neoplastic conditions.
    DOI:  https://doi.org/10.1038/s41587-025-02965-6
  66. FEBS Lett. 2026 Jan 09.
      Mitochondrial protein Slm35 is linked to TOR1 signaling, mitophagy, and stress response in Saccharomyces cerevisiae. Nonetheless, little is known about its regulation or how it affects stress adaptation. In this work, we identified stress-related transcription factor binding sites and two upstream open reading frames (uORFs) in the 5'-UTR of SLM35. Using transcriptional reporters, we showed that the transcription factor Gis1 represses SLM35 transcription; however, Slm35 protein levels increased under oxidative stress and in early stationary phase, suggesting post-transcriptional regulation. Site-directed mutagenesis revealed that one uORF negatively regulates translation, with its disruption leading to altered Slm35 levels and a reproducible increase in mitophagy flux. These findings reveal multilayered control of SLM35 expression and underscore the role of uORF-mediated translation in mitochondrial stress responses. Impact statement This study shows that SLM35, encoding a mitochondrial protein, is controlled through multiple regulatory layers, combining transcriptional repression by stress-responsive factors with uORF-mediated translational regulation. By linking these mechanisms to mitophagy, the work provides new insight into mitochondrial quality control under stress.
    Keywords:  SLM35; Saccharomyces cerevisiae; gene expression; mitochondria; mitophagy; stress‐response; upstream open reading frame
    DOI:  https://doi.org/10.1002/1873-3468.70269
  67. Commun Biol. 2026 Jan 08.
      Mitochondria respond to various stresses. Nevertheless, the regulation of this response while considering coordination between mitochondrial (mtDNA)- and nuclear DNA (nDNA)-encoded gene expression has been overlooked. Our RNA-seq analysis of 18 human cell lines grown in hypoxia (0.2-2% oxygen, 16-24 h) reveals a significant and coordinated reduction of mito-nuclear oxidative phosphorylation (OXPHOS) genes' expression in most (N = 11) cell lines. mtDNA copy number assessment in U87, HCT-116, MCF-7, and HeLa cells reveals non-significant changes, suggesting that the overall reduced mito-nuclear gene expression (MNGE) in hypoxia occurs at the RNA level. Analysis of HIF1α ChIP-seq experiments from cells exposed to hypoxia reveals increased binding to upstream regulatory elements of certain regulators of mitochondrial gene expression. Furthermore, RNA-seq analysis of HIF1α knockout HCT-116 cells grown in hypoxia reveals reduced mtDNA gene expression, yet no change in nDNA OXPHOS genes, suggesting that HIF1α knockout led to departure from coordination of MNGE. Finally, nascent RNA transcripts analysis (PRO-seq) in HeLa, U87, and D407 cells grown in hypoxia shows increased intensity of pausing sites throughout the mtDNA. This finding suggests an important role for transcriptional pausing in the regulation of mtDNA gene expression. Taken together, coordinated reduction of MNGE in hypoxia underlines MNGE as a pivotal player in general mitochondrial function, and particularly in response to stress.
    DOI:  https://doi.org/10.1038/s42003-025-09457-y
  68. Nature. 2026 Jan 07.
      Mammalian genomes contain millions of regulatory elements that control the complex patterns of gene expression1. Previously, the ENCODE consortium mapped biochemical signals across hundreds of cell types and tissues and integrated these data to develop a registry containing 0.9 million human and 300,000 mouse candidate cis-regulatory elements (cCREs) annotated with potential functions2. Here we have expanded the registry to include 2.37 million human and 967,000 mouse cCREs, leveraging new ENCODE datasets and enhanced computational methods. This expanded registry covers hundreds of unique cell and tissue types, providing a comprehensive understanding of gene regulation. Functional characterization data from assays such as STARR-seq3, massively parallel reporter assay4, CRISPR perturbation5,6 and transgenic mouse assays7 have profiled more than 90% of human cCREs, revealing complex regulatory functions. We identified thousands of novel silencer cCREs and demonstrated their dual enhancer and silencer roles in different cellular contexts. Integrating the registry with other ENCODE annotations facilitates genetic variation interpretation and trait-associated gene identification, exemplified by the identification of KLF1 as a novel causal gene for red blood cell traits. This expanded registry is a valuable resource for studying the regulatory genome and its impact on health and disease.
    DOI:  https://doi.org/10.1038/s41586-025-09909-9