bims-miptne Biomed News
on Mitochondrial permeability transition pore-dependent necrosis
Issue of 2025–09–28
nine papers selected by
Oluwatobi Samuel Adegbite, University of Liverpool



  1. Microorganisms. 2025 Sep 12. pii: 2139. [Epub ahead of print]13(9):
      Coccidiosis due to Eimeria tenella remains a major constraint on the poultry industry. Previous studies have revealed that E. tenella infection triggers apoptosis in host cells. The mitochondrial permeability transition pore (MPTP) plays a pivotal role in the apoptosis and necrosis observed in infected host cells. However, the effect of MPTP opening on mitochondrial apoptotic factors remains unclear. To elucidate the dynamic changes in apoptotic signals during MPTP-mediated apoptosis in host cells infected with E. tenella, we established a chicken embryo caecal epithelial cell infection model. Cyclosporin A (CsA) was used to inhibit the MPTP. The infection rate was assessed by Hematoxylin and eosin (H&E) staining, whereas MPTP opening and the abundances of the mitochondrial apoptotic factors Smac, Endo G, and AIF were determined by flow cytometry and ELISA, respectively. The results revealed that both the degree of MPTP opening was markedly reduced in the E. tenella+CsA group compared to the E. tenella group (p < 0.05). Between 24 and 120 h post-infection (hpi), the cytoplasmic levels of Smac, Endo G, and AIF were significantly elevated in the E. tenella group compared with the control group (p < 0.05), while their mitochondrial levels were markedly decreased (p < 0.05). In contrast, mitochondrial expression of these factors was restored in the E. tenella+CsA group (p < 0.05), accompanied by a reduction in their cytoplasmic abundance (p < 0.05). These findings indicate that E. tenella promotes MPTP-dependent release of mitochondrial pro-apoptotic factors into the cytosol during the mid-to-late stages of infection, whereas pharmacological inhibition of the MPTP limits this redistribution.
    Keywords:  Eimeria tenella; apoptotic; host cell; mitochondrial intermembrane protein
    DOI:  https://doi.org/10.3390/microorganisms13092139
  2. Metabolites. 2025 Sep 09. pii: 600. [Epub ahead of print]15(9):
      Background/Objectives: Sepsis is a life-threatening condition characterized by organ dysfunction due to a dysregulated host response to infection. Mitochondrial dysfunction is considered a key contributor to the pathogenesis of sepsis, but its molecular mechanisms remain unclear. Methods: In this study, we used a cecal ligation and puncture (CLP) model to induce sepsis in wild-type (WT) and cyclophilin D knockout (CypD KO) mice. Liver tissues were collected at 0, 6, and 18 h post-CLP and analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Results: Metabolomic profiling revealed that lactate levels significantly increased in the WT mice but remained stable in the KO mice. While AMP levels were preserved in the KO mice, these mice had significantly higher glutathione disulfide (GSSG) and spermidine concentrations than the WT mice at 18 h (p < 0.05). The levels of malondialdehyde (MDA), a marker of oxidative stress, were also significantly lower in the KO mice at 18 h (p < 0.05). These findings suggest that CypD deficiency preserves mitochondrial function, enhances resistance to oxidative stress, and mitigates septic liver injury. Conclusions: Our results highlight the potential of targeting mitochondrial permeability transition as a therapeutic strategy for sepsis.
    Keywords:  cyclophilin D; liver injury; metabolomics; mitochondrial dysfunction; mitochondrial permeability transition; mouse; sepsis
    DOI:  https://doi.org/10.3390/metabo15090600
  3. J Cell Physiol. 2025 Sep;240(9): e70093
      Cancer is a leading cause of death in developed countries, despite many breakthroughs in targeted small molecule and immunotherapeutic interventions. A deeper understanding of the characteristics and processes that underlie malignancy will enable us to develop more effective therapeutic options to improve patient outcomes. One particular area of interest is in cancer cell metabolism. Even as early as the 1920s, Otto Warburg recognized dysregulated metabolism in cancerous cells. Altered metabolism may provide targetable nutrient dependencies for further clinical development, either by nutrient restriction or pathway inhibition. More recently, researchers have observed an increasingly strong linkage between altered mitochondrial Ca2+ homeostasis and tumor cell metabolism, with strong implications for therapeutic targeting. In this review, we summarize the literature surrounding mitochondrial Ca2+ homeostasis, metabolism, and cancer, as well as providing a discussion of the potential for mitochondrial Ca2+ modulation as an anticancer therapeutic modality.
    Keywords:  Ca2+ signaling; cancer; metabolism; mitochondria
    DOI:  https://doi.org/10.1002/jcp.70093
  4. Front Genet. 2025 ;16 1632867
       Introduction: Reperfusion serves as an effective therapeutic strategy for myocardial infarction (MI), but it causes damage to the heart. Although many studies have investigated the mechanism of disparate forms of cell death in myocardial ischemia-reperfusion injury (I/R), there remains a paucity of studies focus on the direct comparison of the mode of cell death events resulting from different reperfusion periods.
    Methods: We conducted an analysis of different sequencing data available in public databases to investigate the relationship between the diverse patterns of cell death and different reperfusion times. Additionally, we evaluated the time window of multiple categories of cell death between cells and mice.
    Results: We explored the relationship between the various modes of cell death and different reperfusion times induced by 6h, 12h and 24 h reperfusion. Our findings revealed that apoptosis occurred in the early stage of I/R injury and continued to appear as the reperfusion time increased. Meanwhile, the changes in autophagy and cuproptosis were also more obvious in the early stage of reperfusion. Notably, ferroptosis and necrosis emerged as the predominant forms of cell death during the medium-to-long-term reperfusion period.
    Discussion: In summary, this study demonstrates that apoptosis takes place during the early stage of reperfusion. Besides, ferroptosis, necrosis and pyroptosis played a crucial role in the prolonged I/R injury period.
    Keywords:  apoptosis; ferroptosis; myocardial infarction; programmed cell death; reperfusion
    DOI:  https://doi.org/10.3389/fgene.2025.1632867
  5. Apoptosis. 2025 Sep 26.
      The mitochondrial voltage-dependent anion channel-1 (VDAC1) protein plays a central role in regulating mitochondrial metabolism, energy production, and apoptosis. VDAC1 interacts with over 100 proteins across the cytosol, endoplasmic reticulum, plasma membrane, and mitochondrial membranes. These interactions coordinate metabolism, cell death, and signal transduction, integrating mitochondrial and cellular functions. To identify VDAC1 binding sites, we designed a peptide array of 768 peptides from 19 selected VDAC1-interacting proteins. We focused on three partners: GAPDH, gelsolin, and actin. Their VDAC1-binding sequences as peptides interacted with purified VDAC1 and, as cell-penetrating peptides, induced cell death, and elevated intracellular Ca2⁺ and ROS levels. Despite sequence diversity, the peptides converged on enhancing transcription factors p53 and c-Jun, upregulating VDAC1, promoting its oligomerization, and triggering apoptosis. Other effects related to their originated protein's function include no significant effect of the GAPDH-derived peptide on its catalytic activity, indicating its effects are independent of glycolysis. The gelsolin-derived peptide altered actin organization, increasing filopodia and focal adhesion, and actin-derived peptides reduced actin, gelsolin, and tubulin expression. This study is the first to identify VDAC1 binding sites on 19 interacting partners and to demonstrate their use as cell-penetrating peptides to modulate the VDAC1 network. These findings highlight VDAC1's multifaceted regulatory role and offer a novel approach for targeting VDAC1-protein interactions for therapeutic purposes.
    Keywords:  Apoptosis; Mitochondria; Peptide array; Protein–protein interaction; VDAC1
    DOI:  https://doi.org/10.1007/s10495-025-02185-y
  6. Cell Calcium. 2025 Sep 14. pii: S0143-4160(25)00089-2. [Epub ahead of print]132 103080
       BACKGROUND: Subarachnoid hemorrhage (SAH) is a severe neurological emergency associated with substantial morbidity and mortality. Research into the mechanisms underlying neuronal injury following SAH has identified early brain injury (EBI) as a critical factor influencing clinical outcomes. Among the various pathological processes involved in EBI, calcium overload remains relatively understudied yet plays a pivotal role in neuronal damage. Excessive accumulation of calcium within mitochondria can initiate apoptotic and autophagic pathways, contributing to cell death. Mitochondrial calcium uptake 1 (MICU1), a regulatory protein located on the inner mitochondrial membrane, functions to modulate mitochondrial calcium ions by inhibiting calcium influx under conditions of low intracellular calcium concentration.
    METHODS: Mitochondria were extracted from the cerebrospinal fluid (CSF) of patients with SAH to evaluate the extent of mitochondrial damage. In vivo and in vitro SAH models were employed to assess mitochondrial damage and dynamic changes in both mitochondrial and cytosolic calcium levels. The interaction between MICU1 and mitochondria was further examined. To investigate the functional role of MICU1, lentivirus vectors were used to upregulate MICU1 expression, while siRNA was applied to knock down its expression in Neuron-2a (N2a) cells. Following hemoglobin (Hb) stimulation, mitochondrial damage and apoptosis were systematically evaluated.
    RESULTS: Analysis of CSF from SAH patients revealed decreased MICU1 expression and aggravated mitochondrial damage. Hb stimulation of primary neurons and N2a cells led to reduced MICU1 expression and mitochondrial calcium overload, which mediated mitochondrial damage and promoted the progression of neuronal apoptosis. Following upregulation of MICU1 expression in N2a cells, the cells exhibited enhanced tolerance to Hb-induced calcium overload, resulting in a significant reduction in mitochondrial damage. This protective effect was attenuated by MICU1 siRNA treatment. Moreover, MICU1 overexpression alleviated Hb-induced apoptosis in N2a cells, whereas siRNA-mediated knockdown of MICU1 exacerbated apoptotic responses.
    CONCLUSION: Mitochondrial calcium overload in neurons following SAH contributes to the development of EBI and neuronal damage. MICU1 exerts a neuroprotective role by mitigating mitochondrial calcium overload, thereby reducing mitochondrial damage and neuronal apoptosis.
    Keywords:  Apoptosis; Calcium overload; Mitochondrial calcium uptake 1 (MICU1); Mitochondrial damage; Subarachnoid hemorrhage (SAH)
    DOI:  https://doi.org/10.1016/j.ceca.2025.103080
  7. Inorg Chem. 2025 Sep 23.
      To address the challenges of precise calcium regulation and limited efficacy in MOF-based nanomedicine, we developed a biodegradable Ca-MOF platform (Ca-MOF@MnCO/HA) coated with hyaluronic acid (HA) and loaded with a H2O2-responsive CO prodrug, manganese carbonyl (MnCO). This system enables CD44-mediated tumor targeting, followed by acid-triggered biodegradation in the tumor microenvironment (TME) to release Ca2+ and MnCO. Intracellular H2O2 then promotes CO release, inducing mitochondrial dysfunction and impairing calcium efflux. The concurrent release of Ca2+ and CO causes sustained calcium overload, intensifying oxidative stress, activating apoptosis, and triggering tumor-specific calcification. This gas-ion synergy highlights the potential of programmable inorganic nanomedicines for improved anticancer therapy.
    DOI:  https://doi.org/10.1021/acs.inorgchem.5c04034
  8. Cell Rep. 2025 Sep 22. pii: S2211-1247(25)01079-4. [Epub ahead of print]44(10): 116308
      Colorectal cancer (CRC) cells exhibit high plasticity and transition between different cellular states during the development of metastasis. Lgr5-expressing cancer stem cells fuel the growth of the primary tumor and metastasis, yet disseminated tumor cells arriving at the metastatic site and seeding liver metastases are devoid of Lgr5 expression. Using CRC organoid models, we demonstrate that mechanical interactions with collagen I, a main constituent of the interstitial matrix, instruct the reprogramming of CRC cells. Collagen I-induced pulling forces are sensed by integrins and mechanosensitive calcium channels, which together direct the transition of CRC cells into a cellular state with transcriptional similarities to fetal intestinal cells. CRC cells infiltrating the interstitial stroma show upregulation of this fetal-like transcriptional program, which correlates with the ability of Lgr5-negative cells to initiate metastasis formation. Our findings indicate that mechanical interactions with collagen I regulate cell fate transitions associated with the metastatic cascade of CRC.
    Keywords:  CP: Cancer; CP: Cell biology; Lgr5; TRPV4; YAP1; cancer stem cell; colorectal cancer; fetal-like state; integrins; mechanosensitive calcium channels; mechanotransduction
    DOI:  https://doi.org/10.1016/j.celrep.2025.116308
  9. Biology (Basel). 2025 Aug 29. pii: 1140. [Epub ahead of print]14(9):
      Mitochondrial reverse electron transport (RET) represents a fundamental but potentially hazardous metabolic process in eukaryotic cells. This review systematically examines current understanding of RET mechanisms and their pathophysiological consequences. RET occurs when electrons flow inversely from reduced coenzyme Q (CoQH2) to complex I, driven by excessive reduction of the CoQ pool and elevated mitochondrial membrane potential, resulting in substantial superoxide production. While moderate RET contributes to physiological redox signaling, sustained RET activation leads to oxidative damage and activates regulated cell death pathways. Notably, RET demonstrates metabolic duality: it facilitates ATP generation through NAD+ reduction while simultaneously inducing mitochondrial dysfunction via reactive oxygen species overproduction. Pathologically, RET has been implicated in myocardial ischemia-reperfusion injury, neurodegenerative disorders including Alzheimer's diseases, and exhibits context-dependent roles in tumor progression. Emerging evidence also suggests RET involvement in microbial pathogenesis through modulation of host immune responses. These findings position RET as a critical regulatory node in cellular metabolism with broad implications for human diseases. Future investigations should focus on developing tissue-specific RET modulators and elucidating the molecular switches governing its activation threshold, which may yield novel therapeutic strategies for diverse pathological conditions.
    Keywords:  cancer; ischemia–reperfusion; mitochondrial reverse electron transport; neurodegenerative diseases; tuberculosis
    DOI:  https://doi.org/10.3390/biology14091140