bims-mithem Biomed News
on Mitochondria in Hematopoiesis
Issue of 2025–08–03
thirteen papers selected by
Tim van Tienhoven, Erasmus Medical Center



  1. Gene. 2025 Jul 25. pii: S0378-1119(25)00474-3. [Epub ahead of print]966 149685
      AML (Acute myeloid leukemia) is an aggressive cancer of the blood and bone marrow characterized by the excessive proliferation of immature white blood cells, that disrupt normal hematopoiesis. LSCs (Leukemic stem cells) represent a subpopulation of AML cells with stem cell-like properties that drive AML initiation, progression, and relapse by evading conventional therapies and sustaining leukemic growth. Despite advances in understanding AML biology, particularly their metabolic alternations, remain poorly understood. Indeed, recent studies have shown that mitochondrial metabolism plays a pivotal role in the regulation of both normal HSCs (hematopoietic stem cells) and LSCs. In this review, we delve into the mitochondrial metabolic characteristics of normal HSCs to provide comprehensive background knowledge. Subsequently, we thoroughly analyze how the distinctive metabolic features of LSCs, highlighting the impact of these differences on cell function and survival. We also investigate the unique mechanisms of drug resistance in LSCs, explaining how these mechanisms enhance the survival of LSCs in the face of conventional treatments. Finally, we discuss emerging therapeutic strategies targeting the mitochondrial metabolism of LSCs in AML, and we discuss prospective therapeutic strategies and future research directions.
    Keywords:  Acute myeloid leukemia; Hematopoietic stem cell; Leukemia stem cell; Metabolism; Mitochondria; Mitochondrial quality control
    DOI:  https://doi.org/10.1016/j.gene.2025.149685
  2. Stem Cells. 2025 Jul 29. pii: sxaf053. [Epub ahead of print]
      In steady state, hematopoietic stem cells (HSCs) reside quiescently in their hypoxic niche with minimal mitochondrial activity, maintaining characteristically low levels of reactive oxygen species (ROS) and instead favoring glycolysis to meet their low energy requirements. However, stress, such as acute infection, triggers a state of emergency hematopoiesis during which HSCs expand more rapidly to produce up to ten-fold more downstream differentiated immune cells. To cope with this demand, HSCs increase their energy production by switching from low ATP-yielding glycolysis to high ATP-yielding mitochondrial oxidative phosphorylation. It is this metabolic switch that enables rapid HSC expansion and differentiation into downstream progeny to increase the immune cell pool and effectively clear the infection. This metabolic switch relies on the sufficient availability of healthy mitochondria as well as fuel in the form of free fatty acids to drive the necessary production of cellular components. This concise review aims to focus on how HSCs increase their mitochondrial content and fuel ATP production via fatty acid oxidation and the impact of HSC dysfunction during aging and other metabolic diseases.
    Keywords:  Acute myelogenous leukemia (AML); Adult haematopoietic stem cells; Bone marrow; Stem cell expansion; adipose
    DOI:  https://doi.org/10.1093/stmcls/sxaf053
  3. Oncol Res. 2025 ;33(8): 2161-2174
       Background: Acute Myeloid Leukemia (AML) is a highly aggressive clonal hematological malignancy with limited treatment options. This study aimed to evaluate the therapeutic potential of nigericin, a polyether ionophore derived from Streptomyces DASNCL-29, as a mitochondrial-targeted agent for AML treatment.
    Methods: Nigericin was isolated from Streptomyces DASNCL-29 and characterized via chromatography and NMR. Its cytotoxicity was tested in MOLM13 (sensitive and venetoclax-resistant) and HL60 (sensitive and cytarabine-resistant) cells using the MTT assay. Mitochondrial dysfunction was assessed by measuring reactive oxygen species (ROS), mitochondrial membrane potential (Δψm), and mitochondrial mass. Apoptosis was evaluated with Annexin V/PI assays and immunoblotting, while proteomic analysis was conducted using Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) to identify differentially regulated proteins.
    Results: Nigericin demonstrated potent cytotoxicity with IC50 values of 57.02 nM in MOLM13-sensitive, 35.29 nM in MOLM13-resistant, 20.49 nM in HL60-sensitive, and 1.197 nM in HL60-cytarabine-resistant cells. Apoptosis was confirmed by Annexin V/PI staining and caspase-3/PARP cleavage, along with MCL-1 downregulation. Mitochondrial dysfunction was evident from increased ROS, reduced Δψm, and decreased mitochondrial mass. Proteomic profiling identified 264 dysregulated proteins, including a 3.8-fold upregulation of Succinate Dehydrogenase [Ubiquinone] Flavoprotein Subunit A (SDHA).
    Conclusion: Nigericin induces apoptosis in AML cells by disrupting mitochondrial function and enhancing oxidative stress. Its nanomolar potency highlights the need for further mechanistic studies and in vivo evaluations to explore its potential in AML treatment.
    Keywords:  Acute myeloid leukemia (AML); Antineoplastic agents; Apoptosis; Mitochondrial dysfunction; Nigericin
    DOI:  https://doi.org/10.32604/or.2025.062951
  4. Stem Cell Res Ther. 2025 Jul 28. 16(1): 408
       BACKGROUND: G-protein coupled receptor 68 (Gpr68) was enriched in long-term hematopoietic stem cells, indicating a potential role of Gpr68 in the HSC function. However, there is no significant phenotype in the HSC biology of Gpr68 whole-body KO mice, which may be counteracted by compensation. To study an intrinsic function of Gpr68 in hematopoiesis, Gpr68flox/flox;Vav-cre+ mouse model where the Gpr68 gene was specifically deleted in hematopoietic cells was generated and monitored here (C57BL/6 J genetic background).
    METHODS: We used complete blood counting and flow cytometry to determine the number and frequency of mature cells in normal hematopoiesis. We evaluated the number and function of stem cells after competitive bone marrow transplantation using cell surface immune markers. Biological functional experiments were used to explore the related cellular mechanisms.
    RESULTS: Apart from a slightly increased megakaryocyte erythroid progenitor subpopulation, the number of hematopoietic stem and progenitor cells was unaltered in young and mid-aged Gpr68flox/flox;Vav-cre+ mice compared with age-matched Vav-cre+ mice. However, the stem cell function was enhanced in mid-aged Gpr68flox/flox;Vav-cre+ mice, represented by increased donor-derived chimerism compared with age-matched Vav-cre+ mice. As enhanced chimerism was traced to LT-HSC, it revealed an increased LT-HSC activation due to loss of Gpr68 in hematopoietic cells upon aging. Consistently, reduced Gpr68 expression was observed in LT-HSC of old C57BL/6 WT mice compared with young WT mice, validating the specific role of Gpr68 in responding to aging. Besides, the Annexin V staining and active caspases in Gpr68 down-expression mice, i.e., Gpr68flox/flox;Vav-cre+ mice and old C57BL/6 WT mice, were decreased when compared with their control mice, respectively.
    CONCLUSION: Loss of Gpr68 in hematopoietic tissues enhances LT-HSC function upon aging by inhibiting a cell apoptosis.
    Keywords:  Aging; Calcium pathway; Erythrocyte parameters; Gpr68; Long-term hematopoietic stem cell
    DOI:  https://doi.org/10.1186/s13287-025-04506-z
  5. Int J Hematol. 2025 Jul 31.
      Clonal hematopoiesis (CH) has emerged as a common age-related phenomenon and a central concept linking somatic mutations in hematopoietic stem cells to both malignant and non-malignant diseases. While initially recognized in the context of hematologic neoplasms, CH is now known to contribute to increased all-cause mortality, particularly through heightened risk of cardiovascular and inflammatory diseases. Frequent mutations in genes such as DNMT3A, TET2, and ASXL1 alter epigenetic regulation and immune signaling, thereby promoting clonal expansion and systemic consequences. Longitudinal studies have illuminated the dynamics of clonal growth and revealed how germline variants influence somatic selection. VEXAS syndrome, driven by UBA1-mutated CH, exemplifies the broader clinical reach of clonal expansion beyond malignancy. CH occupies an intermediate biological state with far-reaching implications. In this Progress in Hematology series, contributors explore the natural history, genetic underpinnings, and inflammatory manifestations of CH, offering insights into its role as both a biomarker and a potential therapeutic target in aging populations.
    Keywords:  Clonal hematopoiesis; Hematopoietic stem cell; Mutation
    DOI:  https://doi.org/10.1007/s12185-025-04034-9
  6. Trends Cell Biol. 2025 Jul 29. pii: S0962-8924(25)00151-5. [Epub ahead of print]
      Blood stem cells are among the body's longest-living cells despite being highly vulnerable to proteotoxic damage, which accelerates their aging. To maintain protein homeostasis (proteostasis), hematopoietic stem cells (HSCs) employ mechanisms such as reduced translation rates, high chaperone activity, autophagy, and selective protein degradation. These strategies mitigate protein misfolding, maintain quiescence, and preserve regenerative potential. Disruptions in proteostasis can lead to the elimination of impaired HSCs through differentiation or apoptosis, ensuring the integrity of the stem cell pool. Due to the systemic impact of the blood on aging and its experimental and clinical accessibility, investigating HSC proteostasis provides insights into longevity and potential therapeutic strategies. This review examines emerging mechanistic links between proteostasis and HSC fate, concluding with unresolved questions and challenges of the current research.
    Keywords:  aging; autophagy; hematopoietic stem cells (HSCs); proteasome; protein homeostasis (proteostasis); translation control
    DOI:  https://doi.org/10.1016/j.tcb.2025.06.006
  7. Pestic Biochem Physiol. 2025 Sep;pii: S0048-3575(25)00233-0. [Epub ahead of print]213 106520
      Dimethoate (DM), a widely used organophosphate pesticide, induces significant alterations in mitochondrial-related proteomes of SH-SY5Y cells without directly affecting cell viability. After, cells were exposed to 100 μM DM for 48 h, proteomic analysis revealed that 27 proteins associated with cellular metabolism and mitochondrial function were notably altered, affecting pathways such as oxidative phosphorylation, electron transport chain, and ATP synthesis. At sublethal concentrations, DM reduced mitochondrial ATP production, oxygen consumption rates (OCR), basal and maximal respiration, while preserving spare respiratory capacity (SRC) and proton leak, indicating maintained mitochondrial membrane integrity. Despite this, DM exposure caused mitochondrial membrane depolarization and increased mitochondrial superoxide production. These mitochondrial alterations were accompanied by enhanced cellular senescence, marked by p53-independent p21 activation, p38 MAPK activation, increased senescence-associated β-galactosidase (SA-β-gal) activity, and disrupted cell cycle progression. Additionally, DM treatment led to upregulation of DNA damage response (DDR) proteins and downregulation of proteins involved in DNA repair and genome stability. Although early-stage apoptosis was observed, elevated Bcl-2 expression suggested a shift toward apoptosis resistance and senescence. DM also disrupted energy-sensing pathways by increasing AMPK subunit expression, yet suppressed autophagy, as indicated by decreased p-mTOR, p-Beclin-1, and LC3-II/I ratios. Collectively, these findings highlight a complex interplay between mitochondrial dysfunction, cellular senescence, and survival mechanisms, suggesting potential long-term effects of DM exposure on cellular health and aging processes.
    Keywords:  Dimethoate; Mitochondria; Neurotoxicity; Pesticide; Senescence
    DOI:  https://doi.org/10.1016/j.pestbp.2025.106520
  8. Mol Ther. 2025 Jul 29. pii: S1525-0016(25)00575-1. [Epub ahead of print]
      GATA2 deficiency is a rare inborn error of immunity caused by monoallelic variants in the GATA2 gene, leading to dysfunction of hematopoietic stem and progenitor cells (HSPCs). Here, we investigate a potential therapeutic strategy for GATA2 deficiency based on CRISPR/Cas9-based gene correction, utilizing recombinant adeno-associated virus serotype 6 (rAAV6) as a template for homology-directed repair (HDR). For a 7-bp deletion giving rise to GATA2 deficiency, we identify a single guide RNA (sgRNA) supporting allele-specific cleavage in the disease allele. Initially, we observe high cytotoxicity in HSPCs upon Cas9/sgRNA ribonucleoprotein nucleofection and rAAV6 transduction, but this is mitigated by co-administering mRNA-based modulators of the DNA-damage response combined with a 10-fold reduction in rAAV6 dose. Using this protocol, we achieve efficient HDR (>80%) in HSPCs derived from a patient carrying the 7-bp deletion and show increased engraftment potential after GATA2 correction. Using DISCOVER-seq, we find limited off-target activity. However, with PCR-free long-read sequencing, we detect frequent large aberrations at the on-target site in HSPCs, primarily attributed to the integration of AAV concatemers identified in 15% of the targeted alleles. Our findings describe the effect of gene correction on GATA2 deficiency and highlight potential on-target aberrations following HDR-mediated gene correction.
    DOI:  https://doi.org/10.1016/j.ymthe.2025.07.038
  9. Adv Pharmacol. 2025 ;pii: S1054-3589(25)00038-9. [Epub ahead of print]104 259-287
      Senescent cells are attributed to aging and age-related diseases. Clearance of senescent cells can delay the aging process and treat age-related diseases. Senescent cells have typical phenotypes including permanent cell cycle arrest, metabolic changes, senescence-associated secretory phenotype, and other structural and functional changes. Senescent cells-targeted therapeutics containing senolytics and senomorphics have been widely investigated but still insufficient, and the internal processes are still unclear, leaving a large gap between preclinical and clinical usage for aging and age-related disease management. Thus, it is urgently demanded to discover many more drugs or new targets with in vivo pharmacodynamics and pharmacokinetics evaluation and monitoring, promoting clinical translation. As a revolutionizing approach, molecular imaging exhibited great potential in exploring the in vivo pathophysiological mechanisms and further promoting the diagnosis and therapies of diseases. It can realize the visualization of complex biochemical processes from living cells, tissues, and organs, to subjects. Benefiting from the numerous imaging probes designed and synthesized with specificity and sensitivity, molecular imaging can vigorously facilitate exploring underlying in vivo mechanisms of senescent cells and senotherapeutics for aging and age-related diseases. Moreover, conjugating the senolytics and senomorphics with imaging probes can realize in vivo image-guided therapy for senescent cells, reversing the dysfunction of aging and treating age-related diseases. Molecular imaging exhibits great potential in visualizing and monitoring senescent cells-targeted therapeutics for aging and age-related diseases, and can forcefully contribute to the clinical translation of gerophamocology.
    Keywords:  Aging; Fluorescence imaging; Molecular imaging; Near-infrared imaging; Senescent cells; Senolytics
    DOI:  https://doi.org/10.1016/bs.apha.2025.01.020
  10. Life Sci. 2025 Jul 30. pii: S0024-3205(25)00525-9. [Epub ahead of print] 123890
      High mortality rates due to cardiovascular diseases (CVDs) fascinate the scientists worldwide in the past few decades to discover potent therapeutic strategies to save the victims. The myocardium being a highly active tissue, mitochondrial homeostasis and mitochondrial quality control system are crucial for maintaining optimal cardiac performance. Mitochondrial quality control mechanism is a finely tuned regulatory network encompassing mitochondrial biogenesis, mitochondrial dynamics and mitophagy and is an integral component of the mitochondrial response to stressor stimuli. Mitochondrial dynamics including the fusion and fission of mitochondrial membranes is regulated by an extensively conserved mechanism comprising a group of mitochondrial membrane proteins belonging to the dynamin family of GTPases. Emerging evidences indicate that defects in mitochondrial fusion or fission are intrinsically correlated with the pathophysiology of CVDs. Mitophagy is a kind of selective autophagy which removes damaged or redundant mitochondria. Experimental findings demonstrated that impairment of mitophagy in cardiomyocytes induces the accumulation of dysfunctional mitochondria, leading to the disruption of cellular homeostasis and consequently precipitating various CVDs. These findings speculate that pharmacological modulation of mitochondrial homeostasis including mitochondrial dynamics and mitophagy may represent a potential therapeutic approach in restoring cardiac physiology. This review summarizes the prevailing insight into the impact of disturbed mitochondrial dynamics and mitophagy in the pathogenesis of CVDs and also delineates the therapeutic potential of several relevant regulatory drugs that target mitochondrial function and quality control in alleviating mitochondrial impairment-related cardiac dysfunction.
    Keywords:  Cardiomyocytes; Cardiovascular diseases; Mitochondrial dynamics; Mitochondrial dysfunction; Mitophagy
    DOI:  https://doi.org/10.1016/j.lfs.2025.123890
  11. Int J Mol Sci. 2025 Jul 15. pii: 6768. [Epub ahead of print]26(14):
      The aging process is associated with the emergence of low-grade, sterile inflammation, called inflammaging, which can accelerate aging-related diseases, such as neurodegenerative, cardiovascular, and musculoskeletal diseases. Recent studies have focused on the novel concept that inflammasomes represent a key innate immune pathway, mechanistically participating in aging-induced stress recognition. This review summarizes the advancements in inflammasome research related to aging. Particular attention is given to the close relationship between aging and inflammasomes and how these processes impact the health of the elderly. Inflammaging has various causes, such as metabolic disorders, changes in the gut microbiota, and immunosenescence. Hence, the connection between inflammasomes and these causes must be explored. This paper describes inflammasomes as a significant contributing factor among the mechanisms that make individuals susceptible to aging-related diseases and discusses the potential role of inflammasome regulation in effectively counteracting aging.
    Keywords:  NLRP3; aging; immunosenescence; inflammaging; inflammasome
    DOI:  https://doi.org/10.3390/ijms26146768
  12. Neurochem Res. 2025 Jul 28. 50(4): 251
      Alzheimer's disease (AD) is a neurodegenerative disorder that causes progressive neurodegeneration and a variety of cognitive deficits. Of note, mitochondrial malfunctions occur early in the disease's development. Mitophagy impairment leads to the build-up of damaged mitochondria inside the cells, causing malfunction and eventual death of the cells. This review summarizes the mechanisms linking mitochondrial damage and autophagy dysregulation to AD and highlights potential therapeutic opportunities. We summarize how mitochondrial dysfunction contributes to AD, including defects in mitochondrial biogenesis, impaired dynamics, the impact of AD-related protein aggregates on mitochondrial integrity, and defective axonal transport. We also explore the roles of mitophagy in AD, including its function in the removal of harmed proteins and organelles. Finally, we highlight the therapeutic strategies for the treatment of AD, targeting molecular components involved in mitochondrial damage and autophagy dysregulation in AD, i.e., antioxidants, mitochondrial modulators, and mitophagy enhancers.
    Keywords:  Alzheimer’s disease; Aβ; Mitochondrial dysfunction; Mitophagy; p-tau
    DOI:  https://doi.org/10.1007/s11064-025-04490-z
  13. Semin Thromb Hemost. 2025 Aug 01.
      0. Abstract Thrombocytopenia following hematopoietic stem cell transplantation (HSCT) is a common complication that is associated with a remarkable increase in morbidity and mortality. Post-HSCT thrombocytopenia is a multifactorial condition with several mechanisms, including reduced platelet production in bone marrow, immune-mediated platelet destruction, and consumptive thrombocytopenia. Graft-versus-host disease (GVHD), medications, infections, and autoimmune mechanisms are potential risk factors for post-HSCT thrombocytopenia. Management of post-HSCT thrombocytopenia primarily focuses on supportive care through platelet transfusions. Moreover, immunosuppressive agents are used to target immune-mediated mechanisms. Thrombopoietin receptor agonists and complement inhibitors are novel treatment options with promising results and fewer side effects. However, further research is essential to establish treatment protocols and improve patient care. In this review, we provide a better understanding of the pathophysiology and risk factors associated with post-HSCT thrombocytopenia for early detection and intervention, ultimately aiming to reduce complications.
    DOI:  https://doi.org/10.1055/a-2673-4266