bims-mitmed Biomed News
on Mitochondrial medicine
Issue of 2026–01–11
sixteen papers selected by
Dario Brunetti, Fondazione IRCCS Istituto Neurologico



  1. 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
  2. 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
  3. 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
  4. 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
  5. 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
  6. 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
  7. STAR Protoc. 2026 Jan 03. pii: S2666-1667(25)00715-4. [Epub ahead of print]7(1): 104309
      Here, we present a protocol to assess the lipogenic phenotype of induced neural stem cells (iNSCs) using stable isotopic tracing. We describe steps for the culture and preparation of iNSCs, labeling with [13C6]-glucose and [13C5, 15N2]-glutamine, and the subsequent extraction of metabolites, lipids, and proteins from the same sample. This protocol supports single-specimen, mass spectrometry-based multi-omics workflows and is applicable to steady-state analyses, stable isotope tracing, and characterization of protein post-translational modifications. For complete details on the use and execution of this protocol, please refer to Ionescu et al.1.
    Keywords:  metabolism; neuroscience; proteomics; protocols in metabolomics and lipidomics; stem cells
    DOI:  https://doi.org/10.1016/j.xpro.2025.104309
  8. 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
  9. 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
  10. 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
  11. 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
  12. 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
  13. Pharmacol Rep. 2026 Jan 05.
      Perinatal hypoxia, also known as neonatal hypoxia-ischemia (HI), can pose a significant threat to the life and health of newborns, leading to hypoxic-ischemic encephalopathy (HIE) and numerous organ dysfunctions, including the nervous system. Mitochondria, which are key organelles in maintaining cellular homeostasis, adenosine triphosphate (ATP) production, regulation of apoptosis, and the cell's response to oxidative stress, appear to be particularly vulnerable to hypoxic damage. In the course of HIE, a number of mitochondrial functions may be impaired, including inhibition of the respiratory chain, increased production of reactive oxygen species (ROS), loss of mitochondrial membrane potential, or activation of apoptotic pathways. As a result of HI, mitochondrial dynamics related to the processes of mitochondrial fusion, division, and autophagy are also changed, which contributes to exacerbating neuralcell damage. Because of the significant role of mitochondria in the pathophysiology of HIE, they represent a promising therapeutic target. This article presents the current state of knowledge on mitochondrial damage mechanisms in HI and discusses potential therapeutic strategies, such as modulators of mitochondrial dynamics, antioxidant compounds, or inhibitors of specific mitochondrial pathways. A better understanding of these mechanisms may contribute to the development of more effective treatments for neurodegeneration associated with perinatal hypoxia.
    Keywords:  Hypoxic-ischemic injury; Mitochondria; Neural cells; Neuroregeneration; Therapeutic strategies
    DOI:  https://doi.org/10.1007/s43440-025-00819-1
  14. J Biol Rhythms. 2026 Jan 04. 7487304251386926
      Circadian clocks present throughout the brain and body coordinate diverse physiological processes to support daily homeostasis, yet the specific interorgan signaling axes involved are not well defined. We previously demonstrated that the skeletal muscle clock controls transcript oscillations of genes involved in fatty acid metabolism in the liver, yet the impact of the liver clock on the muscle remained unknown. Here, we use male hepatocyte-specific Bmal1 KO mice (Bmal1hep-/-) to reveal that approximately one-third of transcript rhythms in skeletal muscle are influenced by the liver clock in vivo. Treatment of myotubes with serum harvested from Bmal1hep-/- mice inhibits expression of genes involved in metabolic pathways, including oxidative phosphorylation. Only small transcriptional changes were induced by liver clock-driven endocrine communication in vitro, leading us to surmise that the liver clock acts to fine-tune metabolic gene expression in muscle. Consistent with functional tuning, treatment of myotubes with serum collected from Bmal1hep-/- mice during the dark phase lowers mitochondrial ATP production compared with serum from wild-type mice. Overall, our results reveal communication between the liver clock and skeletal muscle, uncovering a bidirectional endocrine communication pathway that may contribute to the metabolic phenotypes of circadian disruption.
    Keywords:  Bmal1; circadian clocks; circadian rhythms; clock communication; interorgan crosstalk; liver metabolism; mitochondria; muscle-liver axis; peripheral clocks; skeletal muscle
    DOI:  https://doi.org/10.1177/07487304251386926
  15. Chem Biol Interact. 2026 Jan 06. pii: S0009-2797(26)00014-1. [Epub ahead of print]425 111906
      Cadmium (Cd), a widespread environmental pollutant, has toxic effects on spermatogenesis and sperm motility, but its role in uterine decidualization remains unknown. This study revealed that Cd exposure during early pregnancy might impair the uterine decidualization along with the disordered proliferation and apoptosis of stromal cells, resulting in the reduction of mouse neonatal number and birth weight. After disrupting the binding of Ca2+ and LNR-C, Cd exposure ligand-independently activated the NOTCH1 signaling and then restrained the expression of nuclear TAZ via RBPJ-targeting LATS. Further analysis suggested that Cd exposure contributed to the depletion of glutathione via Gclc that was identified as a direct downstream target of TAZ/TEAD, resulting in intracellular ROS accumulation and subsequent mitochondrial dysfunction. Meanwhile, Cd exposure prevented mitochondrial fusion and facilitated mitochondrial fission together with the fragmented mitochondria, whereas attenuation of intracellular ROS alleviated the imbalance between mitochondrial fusion and fission by Cd exposure. Moreover, improvement of mitochondrial fusion insufficiency and excessive fission rescued the impairment of Cd on decidualization. Collectively, Cd exposure impaired the uterine decidualization through inducing the imbalance of mitochondrial fusion and fission. These findings reveal the toxic effect of Cd on female reproduction and serve as a risk factor for adverse pregnancy outcomes.
    Keywords:  Cadmium; Mitochondria; NOTCH1 signaling; TAZ; Uterine decidualization
    DOI:  https://doi.org/10.1016/j.cbi.2026.111906
  16. Toxicol Rep. 2026 Jun;16 102183
      2,4-Dinitrophenol (DNP) is a potent mitochondrial uncoupler briefly marketed in the 1930s as a weight-reducing agent before being banned by the FDA after reports of severe toxicity. Since the early 2000s, DNP has reemerged as an illicit "fat-burner", causing characteristic metabolic disturbances with a high risk of fatal outcome. We describe two Swedish cases of DNP poisoning: one fatal after suicidal ingestion and one non-fatal after use for weight reduction. Clinical data, mitochondrial respirometry, and analysis of gas exchange and ventilatory dynamics were used to characterize the metabolic disturbances under intensive care. The fatal case progressed within hours to respiratory acidosis, hyperthermia, severe hyperkalemia, and peri-mortem rigidity consistent with catastrophic ATP depletion. The non-fatal case showed similar but reversible toxicity, with sustained yet manageable hypermetabolism lasting more than a week. Serial platelet respirometry demonstrated a marked initial increase in uncoupled respiration, followed by a progressive decline with a functional half-life of 4.9 days. Together, these cases suggest a self-amplifying feedback loop central to DNP toxicity, in which excessive CO₂ production from mitochondrial uncoupling causes local acidosis that enhances mitochondrial DNP uptake. Glucose supplementation and hyperkalemia management are essential supportive measures, whereas active cooling and high minute ventilation may blunt this self-reinforcing metabolic acceleration. Severe poisoning may result in a state of "runaway uncoupling," a term we propose for the catastrophic progression to death observed in numerous DNP poisonings. This feedback loop illustrates the unpredictable toxicokinetics of DNP and reinforces the FDA's early conclusion: DNP is "unfit for human consumption".
    Keywords:  DNP; Hyperthermia; Mitochondrial uncoupling; Postmortem redistribution
    DOI:  https://doi.org/10.1016/j.toxrep.2025.102183