bims-mithem Biomed News
on Mitochondria in Hematopoiesis
Issue of 2025–08–17
nineteen papers selected by
Tim van Tienhoven, Erasmus Medical Center



  1. Aging Dis. 2025 Jul 31. 16(5): 2495-2497
      Mitochondrial dysfunction is increasingly recognized as a unifying mechanism underlying aging and a wide range of age-related diseases. This special issue brings together recent advances that elucidate how impaired mitochondrial function contributes to neurodegenerative, cardiovascular, and metabolic disorders. The featured articles highlight molecular pathways of mitochondrial decline, its systemic consequences, and potential interventions aimed at restoring mitochondrial health. Collectively, these studies reinforce the concept that targeting mitochondrial integrity holds significant promise for promoting healthy aging and preventing chronic disease.
    DOI:  https://doi.org/10.14336/AD.2025.10731
  2. Cell Mol Neurobiol. 2025 Aug 14. 45(1): 79
      Mitochondrial dysfunction has been identified as a key factor in the pathophysiological changes associated with intracerebral hemorrhage (ICH). As the core of intracellular energy metabolism, mitochondrial homeostasis is highly dependent on the precise regulation of its mitochondrial quality control (MtQC) system. After ICH, dysfunctional mitochondria lead to impaired oxidative phosphorylation and cellular bioenergetic stress, inducing oxidative stress, inflammatory responses, and programmed cell death, further exacerbating cellular damage. To counteract this injury, cells activate a series of MtQC mechanisms for compensatory repair, including mitochondrial dynamics, mitochondrial biogenesis, mitophagy, and intercellular mitochondrial transfer. These stringent mechanisms help maintain the mitochondrial network, restore the integrity of mitochondrial structural and functional integrity, improve neural function, and mitigate brain injury. In this review, we discuss key evidence regarding the role of mitochondrial dysfunction in ICH, focusing on the MtQC mechanisms involved in ICH. We also summarize potential therapeutic strategies targeting MtQC to intervene in ICH, providing valuable insights for clinical applications.
    Keywords:  Intercellular mitochondrial transfer; Intracerebral hemorrhage; Mitochondrial dynamics; Mitochondrial dysfunction; Mitochondrial quality control; Mitophagy
    DOI:  https://doi.org/10.1007/s10571-025-01599-1
  3. J Biochem Mol Toxicol. 2025 Aug;39(8): e70441
      Proton pump inhibitors (PPIs), including esomeprazole, impact the acidic tumor microenvironment, potentially influencing cancer cell behavior. By examining the combined effects of esomeprazole and cisplatin on SNU-1 gastric carcinoma cells, this study sought to elucidate the mechanisms through which esomeprazole enhances cisplatin's cytotoxicity, potentially allowing for effective treatment with reduced cisplatin dosages. SNU-1 cells were treated with varying doses of esomeprazole and cisplatin, alone and in combination. Cell viability was assessed using the XTT assay. Oxidative stress (TAS/TOS), apoptosis (Annexin V, cleaved PARP), mitochondrial membrane potential, and DNA damage (8-oxo-dG, γH2AX, ATM) were evaluated using flow cytometry and ELISA. Statistical significance was determined by ANOVA. Esomeprazole alone showed no significant effect on SNU-1 cell viability, oxidative stress (TAS/TOS), apoptosis, mitochondrial membrane potential, or DNA damage. Cisplatin, however, significantly reduced cell viability (IC50 = 3.024 µg/mL), increased oxidative stress (decreased TAS, increased TOS), diminished apoptosis (increased Annexin V binding and cleaved PARP levels), disrupted mitochondrial membrane potential, and caused significant DNA damage (increased H2AX and ATM phosphorylation, and elevated 8-oxo-dG) (p < 0.001). Notably, the combination of esomeprazole and cisplatin synergistically enhanced cisplatin's effects. The combination resulted in a significantly greater reduction in cell viability (CI < 1), a further increase in oxidative stress, a higher level of apoptosis, amplified mitochondrial depolarization, and potentiated DNA damage compared to cisplatin alone (p < 0.001). Esomeprazole potentiates cisplatin-induced cytotoxicity in SNU-1 gastric cancer cells by enhancing oxidative stress, apoptosis, mitochondrial dysfunction, and DNA damage. This suggests a potential therapeutic strategy to improve cisplatin efficacy and overcome resistance in gastric cancer.
    Keywords:  DNA damage; apoptosis; cisplatin; combination; esomeprazole; mitochondrial membrane potential
    DOI:  https://doi.org/10.1002/jbt.70441
  4. Int J Mol Sci. 2025 Jul 31. pii: 7400. [Epub ahead of print]26(15):
      T cells play a vital role in resisting pathogen invasion and maintaining immune homeostasis. However, T cells gradually become exhausted under chronic antigenic stimulation, and this exhaustion is closely related to mitochondrial dysfunction in T cells. Mitochondria play a crucial role in the metabolic reprogramming of T cells to achieve the desired immune response. Here, we compiled the latest research on how mitochondrial metabolism determines T cell function and differentiation, with the mechanisms mainly including mitochondrial biogenesis, fission, fusion, mitophagy, and mitochondrial transfer. In addition, the alterations in mitochondrial metabolism in T-cell exhaustion were also reviewed. Furthermore, we discussed intervention strategies targeting mitochondrial metabolism to reverse T cell exhaustion in detail, including inducing PGC-1α expression, alleviating reactive oxygen species (ROS) production or hypoxia, enhancing ATP production, and utilizing mitochondrial transfer. Targeting mitochondrial metabolism in exhausted T cells may achieve the goal of reversing and preventing T cell exhaustion.
    Keywords:  T-cell exhaustion; metabolic reprogramming; metabolism; mitochondria; mitochondrial dynamics
    DOI:  https://doi.org/10.3390/ijms26157400
  5. Mol Ther. 2025 Aug 11. pii: S1525-0016(25)00649-5. [Epub ahead of print]
      Hematopoietic stem cells are essential for sustaining the hematopoietic system throughout an individual's life. Within the HSC hierarchy, long-term hematopoietic stem cells are at the apex, maintaining the system by their unique capacity for self-renewal and differentiation into all blood cell lineages. Traditionally, it was believed that long-term hematopoietic stem cell homeostasis is solely regulated within the bone marrow niche. However, our innovative single cell exosomal tracking analysis and in vivo transplantation studies revealed that lung cells play a significant role in this regulation through exosomal communication. Our in vivo primary and secondary engraftment studies demonstrate that exosomes derived from human primary small airway epithelial cells significantly enhance hematopoietic stem cell engraftment. We developed a single-cell exosome tracking pipeline to track these exosomes and elucidated their effects on target cell populations. By integrating single-cell RNA sequencing, exosomal miRNA profiling, and proteomics, we have identified specific genes, miRNAs, and proteins that regulate hematopoietic stem cell function, providing a comprehensive understanding of the molecular mechanisms involved in homeostasis. This insight suggests potential utility in modulating HSC function under experimental conditions, especially in conditions associated with low stem cell fitness including sickle cell disease and bone marrow failure.
    DOI:  https://doi.org/10.1016/j.ymthe.2025.08.020
  6. Int J Mol Sci. 2025 Jul 24. pii: 7149. [Epub ahead of print]26(15):
      Glycolysis and oxidative phosphorylation are the main pathways of cellular energy production. Glucose is metabolized via glycolysis to generate pyruvate, which, under anaerobic conditions, is converted into lactate, while, under aerobic conditions, pyruvate enters mitochondria for oxidative phosphorylation to produce more energy. Accordingly, mitochondrial dysfunction disrupts the energy balance. Lactate, historically perceived as a harmful metabolic byproduct. However, emerging research indicates that lactate has diverse biological functions, encompassing energy regulation, epigenetic remodeling, and signaling activities. Notably, the 2019 study revealed the role of lactate in regulating gene expression through histone and non-histone lactylation, thereby influencing critical biological processes. Metabolic reprogramming is a key adaptive mechanism of cells responding to stresses. The Warburg effect in tumor cells exemplifies this, with glucose preferentially converted to lactate for rapid energy, accompanied by metabolic imbalances, characterized by exacerbated aerobic glycolysis, lactate accumulation, suppressed mitochondrial oxidative phosphorylation, and compromised mitochondrial function, ultimately resulting in a vicious cycle of metabolic dysregulation. As molecular bridges connecting metabolism and epigenetics, lactate and lactylation offer novel therapeutic targets for diseases like cancer and neurodegenerative diseases. This review summarizes the interplay between metabolic reprogramming and mitochondrial dysfunction, while discussing lactate and lactylation's mechanistic in the pathogenesis of related diseases.
    Keywords:  epigenetics; lactate; lactylation; metabolic reprogramming; mitochondrial dysfunction
    DOI:  https://doi.org/10.3390/ijms26157149
  7. Life Med. 2025 Aug;4(4): lnaf019
      Mitochondrial dysfunction is a hallmark of aging, characterized by a decline in mitochondrial biogenesis and quality control, compromised membrane integrity, elevated ROS production, damaged mitochondrial DNA (mtDNA), impaired mitochondrial-nuclear crosstalk, and deregulated metabolic balance. Among the key longevity regulators, sirtuin family members SIRT3, SIRT4, and SIRT5 are predominantly localized to mitochondria and play crucial roles in maintaining mitochondrial function and homeostasis. This review explores how mitochondrial sirtuins mitigate aging-related mitochondrial dysfunctions and their broader implications in aging-related diseases. By elucidating the intricate interplay between mitochondrial dysfunction and mitochondrial sirtuins, we aim to provide insights into therapeutic strategies for promoting healthy aging and combating age-related pathologies.
    Keywords:  aging; mitochondrial dysfunction; mitochondrial sirtuins
    DOI:  https://doi.org/10.1093/lifemedi/lnaf019
  8. DNA Cell Biol. 2025 Jul 25.
      Megakaryocytes (MKs), which are differentiated from megakaryocytic-erythrocytic progenitors, generate platelets (PLTs) by expanding and branching their cellular fragments under the influence of cytokines and intercellular mechanisms such as autophagy and release of reactive oxygen species (ROS) in the bone marrow. Autophagy is a self-destructive process that plays a significant role in cell growth and energy maintenance of the cells. In contrast, ROS are toxic products of cellular metabolism that are harmful to human stem cells but have a crucial role in determining lineage commitment at the common myeloid progenitor stage and deriving further maturation progression toward MKs and PLTs production, with an interconnected relationship in the onset and deriving of autophagy. This review summarizes and discusses what has been discovered about the current state of knowledge regarding autophagy effects on MK differentiation, ROS effects on megakaryopoiesis (MKp), and the relationship between these molecules and autophagy initiation. Furthermore, in vitro applications of controlling these external factors on MKp are reviewed according to pertinent studies. Utilizing these regulatory mechanisms supports an improved design of in vitro MKp for introducing artificial PLT sources and might be beneficial in creating novel treatments of PLT-related coagulation disorders.
    Keywords:  ROS; autophagy; megakaryocytes; megakaryopoiesis; platelets
    DOI:  https://doi.org/10.1177/10445498251361641
  9. Free Radic Res. 2025 Aug 14. 1-20
      Mitochondrial function and redox regulatory processes are crucial aspects of cellular metabolism and energy production. Cancers, including gliomas, largely exhibit altered mitochondrial function, which can lead to changes in cellular signaling pathways and redox homeostasis. Aberrant redox signaling can promote glioma progression by influencing cell proliferation, metastasis and therapeutic response. Several cancer-associated driver mutations - genetic alterations that confer survival and growth advantage to cancer cells, are associated with gliomas and affect mitochondrial function and redox states. Here is an overview of the crucial intersection between mitochondrial function and driver genes in glioma, highlighting some of the recent advances that augment our understanding of this intersection.
    Keywords:  Cancer drivers; Driver mutations; Glioma; Mitochondria; Redox homeostasis
    DOI:  https://doi.org/10.1080/10715762.2025.2548479
  10. Cardiovasc Diabetol. 2025 Aug 13. 24(1): 331
       BACKGROUND: Mitochondrial dysfunction plays a pivotal role in the onset and progression of diabetic cardiomyopathy (DCM). It is hypothesized that ultrastructural mitochondrial abnormalities, molecular dynamics imbalance, and bioenergetic impairments collectively contribute significantly to cardiac dysfunction. Consequently, investigating mitochondrial ultrastructural changes and metabolic disturbances is crucial for elucidating the mechanistic underpinnings of DCM.
    OBJECTIVE: This study aims to comprehensively characterize alterations in mitochondrial ultrastructure and energy metabolism in DCM and examine the interplay between these two factors.
    METHODS: High glucose-treated H9c2 cardiomyocytes and DCM model mice were analyzed via scanning electron microscopy (SEM) and 3D imaging. Three-dimensional morphometric parameters-such as Length3D, Thickness3D, Width3D, Area3D, Volume3D, Anisotropy, Flatness, and Elongation-were quantified to evaluate mitochondrial architecture. At the two-dimensional (2D) level, mitochondria-associated membrane (MAM) parameters were analyzed. Further, detailed statistical analysis was conducted on mitochondrial cristae, including cristae scores, count, width, gap size, and junction widths in myocardial tissues. Mitochondrial dynamics and autophagy-related protein expression (Mfn1, Mfn2, Opa1, p-Drp1(ser616), PINK1, Parkin1) in myocardial tissues were assessed by Western blot. Mitochondrial bioenergetics were measured by ATP content, membrane potential, mtDNA copy number, SOD levels, mitochondrial Ca2+ levels, and oxidative phosphorylation (OXPHOS) activity across Complexes I-V in myocardial tissue. Additionally, the oxygen consumption rate (OCR) of viable H9c2 cells was measured using the O2k system.
    RESULTS: 3D reconstruction revealed key myocardial ultrastructures-including T-tubules, sarcoplasmic reticulum (SR), intercalated discs, and mitochondria-and demonstrated significant differences in mitochondrial morphology and 3D morphometric parameters across subpopulations. Under high glucose (HG) conditions, in vitro analysis showed a reduction in mitochondrial Length3D and Anisotropy in H9c2 cells, accompanied by increases in Thickness3D, Width3D, Flatness, and Elongation. HG exposure also led to an increase in the length of MAM contact sites and the MAM-to-mitochondria perimeter ratio. In vivo, the DCM group exhibited decreased 2D morphometric parameters (length, width, area, perimeter, and shape AP), as well as reductions in 3D measurements (Thickness3D, Width3D, and Volume3D) compared to controls. No significant differences were observed in Length3D, Area3D, Anisotropy, Flatness, and Elongation between groups. 3D surface analysis revealed rough mitochondrial surfaces in the DCM group, while controls displayed smooth surfaces. Control mitochondria exhibited well-aligned, well-defined cristae, whereas DCM mitochondria showed cristae dissolution, disorganized arrangements, and vacuolization within the cristae. The DCM group also had increased cristae junction width and spacing. Additionally, megamitochondria were observed in DCM samples. The DCM group showed a significant increase in MAM contact site length and MAM-to-mitochondria perimeter ratio in myocardial tissue. Molecular analysis revealed decreased expression of fusion proteins (Mfn1, Opa1) and increased levels of p-Drp1(ser616), alongside elevated autophagy markers (PINK1, Parkin1). Bioenergetic dysfunction was evident through decreased ATP production, mitochondrial membrane potential collapse (ΔΨm), reduced mtDNA copy number, decreased SOD levels, impaired activities of complexes I/III/IV/V, and diminished basal/maximal respiration, ATP-linked respiration, and spare respiratory capacity. Conversely, mitochondrial Ca2+ levels were elevated in the DCM group, along with increased proton leakage in H9c2 cells.
    CONCLUSIONS: This study establishes a comprehensive framework linking "3D morphology-molecular regulation-metabolic dysfunction," highlighting the synergistic interplay between mitochondrial ultrastructural remodeling and bioenergetic failure as key drivers of DCM progression. These findings provide valuable insights into DCM pathogenesis and suggest potential therapeutic targets.
    Keywords:  3D visualization imaging; Diabetic cardiomyopathy; Energy metabolism; Mitochondria; Scanning electron microscopy; Ultrastructure
    DOI:  https://doi.org/10.1186/s12933-025-02884-5
  11. Ann Med. 2025 Dec;57(1): 2528167
       BACKGROUND: The Long COVID syndrome is a major global health problem, affecting approximately 10-20% of individuals infected with SARS-CoV-2 virus with many remaining symptomatic beyond one year. Fatigue, reduced exercise tolerance and hyperlactataemia on minimal exertion have led to the suggestion of a bioenergetic defect. We hypothesised that mitochondrial dysfunction is a pathological feature in Long COVID cases and would correlate with clinical outcome.
    METHODS: This prospective, case-controlled, observational study recruited 27 participants with an established diagnosis of Long COVID syndrome from a single tertiary clinic together with 16 age-matched controls aged 25-65 years. Seahorse-based mitochondrial flux analysis and bioenergetics profile of isolated peripheral blood mononuclear cells (PBMCs) was performed and correlated with clinical phenotype.
    FINDINGS: Long COVID cases had an increased baseline and ATP-induced oxygen consumption rate with a significant attenuation in tetramethylrhodamine methyl ester perchlorate fluorescence response to oligomycin. Correlations were observed between mitochondrial function and autonomic health, quality of life and time from index infection. Sex-specific differences were also observed.
    INTERPRETATION: PBMCs from Long COVID subjects exhibit an exceptional and distinctive change in ATP synthase, as it contributes to the mitochondrial membrane potential rather than using it exclusively to generate ATP. The findings suggest that the enzyme runs both forward and reverse reactions, synthesising and hydrolysing ATP. The correlation of mitochondrial function with clinical phenotype in Long COVID may indicate a causal relationship and warrants further validation in larger scale studies.
    Keywords:  ATP synthase; long COVID; mitochondrial dysfunction; peripheral blood mononuclear cells (PBMCs)
    DOI:  https://doi.org/10.1080/07853890.2025.2528167
  12. Biochem Biophys Res Commun. 2025 Aug 08. pii: S0006-291X(25)01156-8. [Epub ahead of print]780 152441
      Monitoring active mitochondria is crucial for gaining insights into essential cellular processes, including energy production, apoptosis, cancer, and neurodegenerative diseases. However, current commercial tools have limitations in detecting subtle changes in mitochondrial membrane potential and tracking the dynamics of these changes in live cells. Here, we report a novel application of LDS 698, a hemicyanine solid-state laser dye, for staining functional mitochondria based on their membrane potential. LDS 698 exhibits high sensitivity and specificity in detecting subtle changes in mitochondrial membrane potential, making it suitable for various analytical techniques, including fluorescence microscopy, flow cytometry, and plate reader assays. Its robustness, photostability, and non-toxicity enable prolonged live-cell imaging for the detection and quantification of mitochondrial morphology and membrane potential. The use of LDS 698 can be extended to study cellular mitochondrial homeostasis and membrane potential dynamics, offering new opportunities for biological research.
    Keywords:  LDS 698; Live cell imaging; Mitochondrial membrane potential
    DOI:  https://doi.org/10.1016/j.bbrc.2025.152441
  13. Autophagy. 2025 Aug 11.
      Ferroptosis remains an underexamined iron- and lipid peroxides-driven cell death modality despite its importance to several human and plant diseases and to immunity thereof. Here, we utilized chemical cell biology, molecular genetics and biochemical analyses to gain insights into how the fungal pathogen Magnaporthe oryzae undergoes ferroptosis strictly in the spore cells to successfully transit to infectious development. We reveal a complex functional interdependency and crosstalk between intrinsic ferroptosis and autophagy-mediated mitochondrial degradation. Mechanistically, the requirement of mitophagy for ferroptotic cell death was attributed to its ability to maintain a pool of metabolically active mitochondria. Pharmacological disruption of the electron transport chain or membrane potential led to complete inhibition of ferroptosis, thus simulating the loss of mitophagy phenotypes. Conversely, increased mitochondrial membrane potential in a mitophagy-defective mutant alleviated the ferroptosis defects therein. Graded inhibition of mitochondrial coenzyme Q biosynthesis with or without ferroptosis inhibitor liproxstatin-1 distinguished its antioxidant function in such regulated cell death. Membrane potential-dependent regulation of ATP synthesis and iron homeostasis, as well as dynamics of tricarboxylic acid cycle enzyme AcoA (aconitase A) in the presence or absence of mitophagy, mitochondrial poisoning or iron chelation further linked mitochondrial metabolism to ferroptosis. Last, we present an important bioenergetics- and redox-based mitochondrial regulon essential for intrinsic ferroptosis and its precise role in fungal pathogenesis leading up to the establishment of the devastating rice blast disease.
    Keywords:  Cell death; coenzyme Q; iron; mitochondrial metabolism; mitophagy; rice blast
    DOI:  https://doi.org/10.1080/15548627.2025.2546944
  14. bioRxiv. 2024 Apr 25. pii: 2024.04.25.590465. [Epub ahead of print]
      MYC is a potent oncogene that is frequently overexpressed in human tumors arising in different tissues. To date there are no approved therapies to directly antagonize oncogenic MYC and its role in driving tumorigenesis. As an alternative approach we employed genetic screens using CRISPR and shRNA to identify the genes that are required for the survival and growth of cells harboring high levels of MYC expression. We find that cells with elevated MYC require the expression of many pro-growth and metabolic pathways including genes involved in mitochondrial citrate production and transport. This citrate producing pathway is critical for cells with elevated MYC to generate the necessary acetyl-CoA to drive the lipid synthesis required for increased proliferation. Inhibition of this pathway results in reduced proliferation and in vivo tumor growth providing a potential therapeutic strategy to target MYC-driven cancers.
    HIGHLIGHTS: CRISPR and shRNA screens identify synthetic lethal interactions with overexpressed MYCMYC overexpressing cells are more sensitive to disruption of citrate production and transportInhibition of SLC25A1 reduces growth of MYC driven tumors.
    DOI:  https://doi.org/10.1101/2024.04.25.590465
  15. Cell Death Discov. 2025 Aug 09. 11(1): 375
      Colorectal cancer (CRC) is a leading cause of global cancer mortality, with therapeutic resistance constituting a major barrier to sustained clinical benefit. Mitochondrial metabolic reprogramming has emerged as a central adaptive mechanism that enables CRC cells to withstand hypoxia and therapeutic pressure, while concurrently driving resistance to chemotherapy, targeted agents, and immunotherapy. In this Review, we explore how mitochondrial metabolism contributes to therapeutic resistance, with particular emphasis on metabolic plasticity, redox balance, and organelle quality control. We also assess enabling technologies such as spatial transcriptomics, proteomics, and patient-derived organoids, and discuss their translational relevance in stratifying metabolic vulnerabilities and informing individualized therapies. Targeting mitochondrial rewiring represents a compelling strategy to overcome resistance and drive progress toward personalized CRC therapy.
    DOI:  https://doi.org/10.1038/s41420-025-02670-y
  16. Cells. 2025 Jul 25. pii: 1153. [Epub ahead of print]14(15):
      The growing global elderly population underscores the escalating importance of anti-aging interventions to combat age-related diseases and extend both health span and lifespan. Over the past decades, various anti-aging interventions have gained recognition, each with its unique set of advantages and limitations. Notably, the transplantation of rejuvenated autologous adult stem cells is standing out as a powerful strategy that holds significant promise in combating age-related functional decline and diseases. This review delves into our current biological insights into cellular rejuvenation and provides an overview of both pre-clinical and clinical experiences with autologous and allogeneic adult stem cell transplantations. It reinforces the concept that rejuvenated adult stem cells constitute a pivotal element in the quest for the fountain of youth. Additionally, we examine the technical challenges involved in obtaining and utilizing these rejuvenated adult stem cells.
    Keywords:  adult stem cells; autologous transplantation; cellular rejuvenation; cellular reprogramming
    DOI:  https://doi.org/10.3390/cells14151153
  17. Nat Aging. 2025 Aug;5(8): 1415-1424
      Senescent cells are intrinsically immunogenic and can be eliminated by the immune system to facilitate tissue repair and regeneration. However, immune-mediated elimination is compromised with age, causing senescent cell accumulation in tissues, thus limiting healthspan and lifespan and promoting age-related diseases such as cancer. Here, we review how different components of the innate and adaptive immune systems, including natural killer cells, macrophages, neutrophils, dendritic cells, T cells and B cells, target senescent cells and how the intrinsic properties of senescent cells can lead to their escape from surveillance. We also discuss the phenomenon of senescence in immune cells themselves and how this affects the surveillance of senescent and cancerous cells. Finally, we touch on emerging therapeutic strategies to enhance the immunosurveillance of senescent cells, as understanding the molecular basis of senescence immunosurveillance and why its potency fails during aging may offer opportunities to treat senescence-mediated age-associated diseases and tissue dysfunction.
    DOI:  https://doi.org/10.1038/s43587-025-00910-5
  18. Hemasphere. 2025 Aug;9(8): e70188
      Acute myeloid leukemia (AML) is a heterogeneous clonal disorder of hematopoietic stem and progenitor cells and the most common malignant myeloid disorder in adults. Extensive research has elucidated the broad spectrum of biological mechanisms contributing to the development of AML and specifically characterized a variety of genetic alterations initiating and defining the disease. However, the role of inflammation in the pathogenesis of AML remains relatively unexplored; indeed, studies on the contribution of inflammatory signaling to disease initiation in myeloid malignancies have only recently gained attention, marking an emerging area of research. AML has the highest incidence in the elderly, where inflammation plays an even greater role. A granular understanding of inflammatory processes driving leukemogenesis thus promises to guide therapeutic strategies for a patient population where outcomes remain dismal. This review offers a comprehensive synthesis of the current knowledge on the role of inflammatory signaling in AML pathogenesis. It discusses the role of inflammation from premalignant states through malignant transformation, dissecting phenotypic, correlative studies from mechanistic evidence. We thereby highlight questions requiring further research to exploit the translational potential of therapies targeting inflammatory signaling and to address challenges with current immune-modulating treatments. A particular focus is placed on assessing the role of inflammation in the interplay with genetic events as established factors in disease initiation and progression to clarify the current understanding of inflammatory signaling in AML pathogenesis.
    DOI:  https://doi.org/10.1002/hem3.70188
  19. Tissue Barriers. 2025 Aug 14. 2537991
       BACKGROUND: Parkinson's disease (PD) is the second most common neurodegenerative disorder, characterized by motor symptoms and progressive degeneration of dopaminergic neurons. Accumulating evidence indicates that mitochondrial dysfunction and oxidative stress are major contributors to PD pathogenesis.
    OBJECTIVES: This review explores the molecular mechanisms underlying PD, emphasizing mitochondrial dysfunction and oxidative stress. It also examines genetic and environmental contributors, emerging biomarkers, and future treatment strategies.
    METHODS: An extensive literature review was conducted, focusing on mitochondrial biology, oxidative stress, genetic mutations, and environmental toxins relevant to PD. Investigations into treatment options - including redox therapies, gene therapies, and lifestyle approaches - were also examined.
    RESULTS: Mitochondrial dysfunction in PD includes disrupted oxidative phosphorylation and elevated reactive oxygen species (ROS). This also affects calcium homeostasis, especially in substantia nigra neurons. Genetic mutations (PINK1, Parkin, DJ-1, LRRK2, GBA) impair mitophagy and antioxidant defenses. Environmental toxins (e.g. MPTP, rotenone) further damage mitochondrial function and contribute to dopaminergic neuron loss. Emerging biomarkers involve measurements of lipid peroxidation and mitochondrial DNA damage. Promising therapeutic strategies include mitochondria-targeted antioxidants (e.g. MitoQ), PINK1-based gene therapy, Parkin activation, ketogenic diet, and exercise-induced mitochondrial biogenesis.
    CONCLUSIONS: Mitochondrial dysfunction and oxidative stress are central to PD pathophysiology. Strategies targeting these mechanisms may slow disease progression. Future research should emphasize combination therapies and early intervention trials, alongside biomarker integration, to enhance clinical outcomes.
    Keywords:  Mitochondrial dysfunction; Parkinson’s disease; oxidative stress
    DOI:  https://doi.org/10.1080/21688370.2025.2537991