bims-mithem Biomed News
on Mitochondria in Hematopoiesis
Issue of 2025–11–09
eight papers selected by
Tim van Tienhoven, Erasmus Medical Center



  1. Cell Death Dis. 2025 Nov 04. 16(1): 792
      Acute myeloid leukemia (AML) remains difficult to cure despite recent advances. Off-target side effects of drugs currently used lead to significant morbidity and mortality. There is recognition that in AML, there is an increased dependence on OXPHOS metabolism, especially in the leukemia stem cell compartment (AML-LSC). It is also recognized that there is potential to exploit this vulnerability to treat AML. Drug re-purposing screens have suggested the potential use of artesunate (ART) to inhibit mitochondrial respiration. We have explored the potential role of ART as an additive agent in treating AML in combination with conventional therapy. Through in-vitro and in-vivo mouse model studies, we demonstrate the mechanism and efficacy of these combinations and their potential to overcome venetoclax resistance. We further demonstrate the specificity of these combinations with minimal off-target effects on normal hematopoietic stem cells (HSC). These observations warrant exploration of the additive role of ART in clinical trials.
    DOI:  https://doi.org/10.1038/s41419-025-08129-3
  2. Ann Med Surg (Lond). 2025 Nov;87(11): 7386-7395
      Myeloproliferative neoplasms (MPNs) are a group of blood cancers characterized by the clonal proliferation of hematopoietic stem cells. These diseases are driven by acquired genetic mutations that activate the JAK-STAT signaling pathway, leading to abnormal cell proliferation and differentiation. Dysfunction of mitochondria, involving increased production of reactive oxygen species (ROS), mutations within its DNA, and alterations in its dynamics, significantly contributes to the development and progression of MPNs. This dysfunction promotes clonal expansion of abnormal blood cells and hinders their programmed cell death (apoptosis). However, targeting mitochondrial pathways offers a promising therapeutic approach. For instance, strategies that include the use of mitochondria-targeted antioxidants to counteract oxidative stress, inhibitors of mitochondrial metabolism to disrupt energy production in MPN cells, and interventions that modulate mitophagy (the process of removing damaged mitochondria) and mitochondrial dynamics. Indeed, achieving specific drug delivery, overcoming therapeutic resistance, and minimizing side effects are at the top of the list. This mini-review highlights the promise of innovative therapeutic approaches, such as gene editing technologies and RNA-based therapies, in addressing mitochondrial dysfunction. Furthermore, it underscores the critical importance of personalized medicine strategies in optimizing the efficacy and safety of mitochondrial-targeted treatments. Future research directions encompass further refinement of these strategies, validation of biomarkers for patient selection, and the development of effective combination therapies. By emphasizing the critical role of mitochondrial dysfunction in the pathogenesis of MPNs, this review provides a framework for improving clinical outcomes and developing novel therapeutic strategies that target these underlying metabolic abnormalities.
    Keywords:  combination drug therapy; drug delivery systems; mitochondrial DNA; mitochondrial dysfunction; myeloproliferative disorders; signal transduction
    DOI:  https://doi.org/10.1097/MS9.0000000000003984
  3. J Intensive Care. 2025 Nov 04. 13(1): 61
      Heatstroke is characterized by systemic inflammation, immune dysregulation, and multiorgan failure, in which mitochondrial damage in leukocytes plays a pivotal role. This review examines the mechanisms by which heat stress induces leukocyte mitochondrial dysfunction and its downstream effects on immunity, coagulation, and organ integrity. Exposure to heat stress activates leukocytes through damage-associated molecular patterns (DAMPs), triggering the release of proinflammatory cytokines, reactive oxygen species (ROS), and neutrophil extracellular traps (NETs). These responses disrupt endothelial integrity, promote microvascular thrombosis, and contribute to the development of disseminated intravascular coagulation (DIC). Prolonged heat exposure further shifts the immune landscape toward immunosuppression, marked by monocyte deactivation and lymphocyte apoptosis. Mitochondrial dysfunction is central to this biphasic immune response. Heat stress reduces mitochondrial membrane potential, increases ROS production, and promotes the release of mitochondrial DNA and cytochrome c, amplifying inflammation and initiating cell death pathways, including apoptosis, pyroptosis, and ferroptosis. Biomarkers such as reduced mitochondrial membrane potential (ΔΨm), elevated mitochondrial ROS, cytochrome c, circulating mitochondrial DNA (mtDNA), and altered expression of mitophagy regulators (e.g., PINK1 and Parkin) provide insights into mitochondrial integrity and function in leukocytes. In addition to immune disruption, mitochondrial injury exacerbates coagulation abnormalities by promoting platelet activation and endothelial dysfunction, fostering a prothrombotic environment. In the microcirculation, leukocyte adhesion, NET formation, and endothelial damage create a self-amplifying cycle of ischemia and inflammation, ultimately leading to organ dysfunction, including hepatic failure, acute kidney injury, acute lung injury, and gastrointestinal barrier breakdown. Therapeutic strategies aimed at preserving mitochondrial function include antioxidants (e.g., N-acetylcysteine and MitoQ), mitochondrial biogenesis inducers (e.g., PGC-1α activators), and mitophagy enhancers. Understanding the central role of leukocyte mitochondrial damage in heat stress provides a foundation for the development of targeted diagnostics and interventions to prevent organ failure and improve clinical outcomes.
    Keywords:  Cell death; Heat stress; Leukocyte; Mitochondria; Organ dysfunction
    DOI:  https://doi.org/10.1186/s40560-025-00832-9
  4. Front Immunol. 2025 ;16 1660709
      Age-related accumulation of somatic mutations in hematopoietic stem and progenitor cells (HSPCs), causing clonal hematopoiesis (CH), often precedes the development of hematologic malignancies. Chronic inflammation and aberrant cytokine expression that are common in aging, contribute to clonal expansion and genomic instability. Acute myeloid leukemia (AML) is an (epi)genetically and physiologically diverse malignancy, characterized by clonal proliferation and incomplete differentiation of HSPCs. The innate immune system, with pattern recognition receptors (PRRs), plays a pivotal role in maintaining hematopoietic homeostasis. Dysregulated signaling through PRRs disrupts hematopoiesis, fostering malignant cell proliferation. In addition, cytokines and interferons exert multifaceted effects on HSPCs, impacting their proliferation, differentiation, and survival. Therapeutic approaches targeting innate immune pathways, offer promising avenues for treating hematologic malignancies. Understanding the intricate crosstalk between innate immunity and hematopoiesis would provide insights into novel therapeutic strategies for combating hematologic malignancies, offering hope for improved patient outcomes and survival. In this review, we discuss about the malfunctioning innate immune-inflammatory axes in the context of abnormal hematopoiesis and the therapeutic approaches that are utilizing/targeting these pathways with efficacy. This review delves into the derangements of innate immune and inflammatory pathways implicated in the development of AML and myelodysplastic syndromes (MDS), shedding light on the therapeutic strategies targeting these pathways.
    Keywords:  acute myeloid leukemia; cytokines; hematopoiesis; inflammation; innate immunity; interferon signaling; mutation; pattern recognition receptors
    DOI:  https://doi.org/10.3389/fimmu.2025.1660709
  5. Platelets. 2025 Dec;36(1): 2577969
      Hematopoietic stem and progenitor cells (HSPCs) hold significant promise for various diseases and gene therapy, highlighting the need for improved in vitro expansion while maintaining their properties. Efficient HSPC expansion requires an environment that preserves self-renewal and homing capabilities. Human Platelet Lysates (HPL) contain bioactive molecules and growth factors that may enhance HSPC functionality. This study investigates the effects of HPL on peripheral blood HSPC proliferation, self-renewal capacity, and homing. We observed that HPL significantly promoted HSPC proliferation, resulting in a 1.7-fold increase in final cell count and reduced doubling time, without affecting colony-forming capacity. Flow cytometry analysis revealed no significant changes in the percentages of CD34+, CD34-CD38+, CD34+CD38+, and CD34+CD38- cells, though CXCR4 marker expression was notably higher in the HPL-treated group. Furthermore, real-time analysis of self-renewal genes (GFI1, HOXB4, and TAL1) indicated a significant increase in GFI1 expression, while HOXB4 and TAL1 remained unchanged. Among homing-related genes (CXCR4, VLA-4, and LFA-1), CXCR4 expression increased significantly, while VLA-4 and LFA-1 levels showed no significant alterations. These findings suggest that HPL enhances HSPC proliferation while preserving their self-renewal and homing abilities, providing a promising approach for optimizing HSPC culture conditions for both research and clinical use.
    Keywords:  Homing; human platelet lysate; in vitro culture; peripheral blood hematopoietic stem and progenitor cells; self-renewal potential
    DOI:  https://doi.org/10.1080/09537104.2025.2577969
  6. Immunity. 2025 Oct 31. pii: S1074-7613(25)00464-9. [Epub ahead of print]
      Aging is an essential aspect of human life, and studying its mechanisms is crucial for extending lifespan and improving quality of life. The immune system plays a central role in the onset of age-related diseases. Understanding the differences between healthy and dysfunctional aging provides key insights into the fundamental immune alternations that occur prior to the point where the system begins to fail. In this review, we explore current perspectives on human immune aging. We focus on changes in the composition of, and consequential functional effects within, the major immune compartments in both circulation and tissues. We discuss earlier findings obtained through flow cytometry, alongside more recent studies utilizing single-cell and advanced cytometry techniques. We highlight here how these methods complement each other and explore potential sources of discrepancies. Finally, we address the challenges that persist in the field of human immune aging.
    Keywords:  adaptive immunity; flow cytometry; human aging; immune repertoire; innate immune cells; single cell
    DOI:  https://doi.org/10.1016/j.immuni.2025.10.009
  7. Adv Sci (Weinh). 2025 Nov 03. e15097
      Strategies to enhance the anti-tumor immune response through the regulation of cellular metabolism are under intensive investigation. Herein, a T cell membrane (TCM)-coated biomimetic magnesium carbonate (MgCO3)/Fe-CD hybrid nanozyme system loaded with the Pyruvate kinase M2 (PKM2) activator TEPP-46 (TFMP) is developed, designed to simultaneously induce mitochondrial metabolic reprogramming in both T cells and tumor cells following radiotherapy (RT). The TCM coating enables TFMP to specifically target tumor tissues that highly express PD-L1, where it competitively binds to PD-L1 and thereby alleviates immune checkpoint-mediated T cell suppression. Upon X-ray irradiation, TFMP continuously catalyzes the conversion of radiotherapy-generated hydrogen peroxide into hydroxyl radicals, thereby sustaining reactive oxygen species production, which leads to mitochondrial damage and immunogenic cell death in tumor cells. Moreover, TFMP can neutralize the acidic tumor microenvironment, while the released Mg2+ and TEPP-46 further augment T cell activation and mitochondrial function, thereby increasing the production of ATP and granzyme B, which effectively eliminate residual tumor cells. Experimental results demonstrate that the combination of TFMP and RT can significantly inhibit tumor progression and activate anti-tumor immunotherapy. This TFMP enhances the efficacy of breast cancer radioimmunotherapy, offering a foundation for developing more comprehensive therapeutic approaches of breast cancer to achieve clinical benefits.
    Keywords:  PKM2 activator; biomimetic nanozyme; mitochondrial metabolism; radioimmunotherapy; sustaining ROS production
    DOI:  https://doi.org/10.1002/advs.202515097