bims-engexo Biomed News
on Engineered exosomes
Issue of 2025–11–09
fourteen papers selected by
Ravindran Jaganathan, Universiti Kuala Lumpur



  1. Curr Mol Med. 2025 Oct 31.
      Osteoporosis is a chronic and progressive skeletal disease characterized by decreased bone mass and microarchitectural deterioration, leading to increased fracture susceptibility. Emerging evidence suggests that extracellular vesicles, particularly exosomes, play a pivotal role as mediators of intercellular communication within the bone microenvironment. This review focuses on the functional roles of exosomes derived from bone marrow mesenchymal stem cells (BMSCs) and other bone-resident cells in modulating bone remodeling. We detail the mechanisms by which exosome-encapsulated non-coding RNAs influence osteoblast and osteoclast activity through key regulatory pathways. Furthermore, we examine how pathological conditions, such as chronic inflammation and diabetes, reshape exosomal cargo, contributing to dysregulated bone homeostasis through mechanisms involving ferroptosis and oxidative stress. Advances in therapeutic applications are discussed, including engineered exosomes, bone-targeting delivery strategies, and hydrogelbased scaffolds. We also highlight the diagnostic potential of exosome-derived RNAs as biomarkers for disease progression and treatment response. Although exosomebased approaches represent a promising frontier in osteoporosis therapy and diagnosis, challenges, such as cargo heterogeneity, targeting specificity, and scalable manufacturing, remain to be addressed before clinical implementation.
    Keywords:  Osteoporosis; biomarkers.; bone marrow mesenchymal stem cells (BMSCs); bonetargeted drug delivery; exosomes; non-coding RNAs
    DOI:  https://doi.org/10.2174/0115665240429059251024093744
  2. Biomol Biomed. 2025 Nov 05.
      The transfer of molecular cargo in exosomes plays a crucial role in cancer progression, influencing metabolic processes, angiogenesis, immune interactions, and invasive capabilities. This review synthesizes current evidence on how exosomes modulate tumor metabolism and drive drug resistance, and outlines therapeutic opportunities. We searched PubMed, Scopus, Web of Science, and Google Scholar for English-language studies using terms related to exosomes/extracellular vesicles, glycolysis, oxidative phosphorylation (OXPHOS), lipid metabolism, and drug resistance/chemoresistance, and integrated the literature qualitatively. Evidence indicates that exosomes reprogram tumor and stromal metabolism by delivering enzymes and non-coding RNAs that boost glycolysis and dampen OXPHOS, activate cancer-associated fibroblasts and extracellular matrix (ECM) remodeling, and modulate ferroptosis. They stimulate angiogenesis (e.g., via vascular endothelial growth factor (VEGF)/Wnt pathways) and promote immune escape through programmed death-ligand 1 (PD-L1), transforming growth factor beta (TGF-β), and macrophage reprogramming. Exosomal integrins and proteases contribute to epithelial-mesenchymal transition (EMT), organotropism, and pre-metastatic niche formation. Critically, exosomes propagate chemoresistance by exporting drugs and spreading determinants-including P-gp/BCRP/MRP-1, anti-apoptotic proteins, and regulatory RNAs-to previously sensitive cells; adipose-derived vesicles and lipid cargos further reinforce metabolic plasticity and therapy resistance. Given their stability, nanoscale dimensions, and ability to cross the blood-brain barrier, exosomes are promising vectors for targeted delivery; engineered vesicles can enhance chemotherapy responsiveness and counteract resistance, particularly alongside immunotherapy. In summary, interventions that disrupt exosome biogenesis, cargo loading, or uptake-paired with engineered exosomes for precision delivery-could mitigate drug resistance, metastasis, and immune evasion and advance more effective cancer treatment.
    DOI:  https://doi.org/10.17305/bb.2025.13295
  3. Discov Oncol. 2025 Nov 05. 16(1): 2045
      Exosomes are small extracellular vesicles produced by most cell types. They carry specific markers and biomolecular cargo that reflect the characteristics of cancer cells. These vesicles contain oncoproteins, mutated DNA fragments, and cancer-specific RNA profiles, which can serve as biomarkers for tumor detection, disease progression, and treatment resistance analysis. As a non-invasive diagnostic tool, exosome biomarkers can be obtained from liquid biopsies, including blood, urine, and saliva samples. The cargo of exosomes, such as miRNAs and proteins, is often associated with patient prognosis, providing insights into tumor aggressiveness and survival rates, which helps in patient risk assessment. Due to their natural biocompatibility and minimal immune response, exosomes are promising drug delivery vehicles for anticancer treatments, including chemotherapeutic agents and RNA-based therapies. This review highlights the dual role of exosomes in cancer, focusing on their diagnostic and therapeutic potential.
    Keywords:  Cancer; Dendritic cell-derived exosomes; Diagnostic biomarkers; Exosomes; Therapeutic delivery; Tumor-derived exosomes
    DOI:  https://doi.org/10.1007/s12672-025-03916-y
  4. Oncol Lett. 2025 Dec;30(6): 606
      Pancreatic cancer is a highly aggressive digestive malignancy and gemcitabine (GEM)-based chemotherapy remains a cornerstone of its treatment despite widespread resistance. The present study employed human pancreatic cancer cell lines to construct a hypoxia model, exosomes were isolated and characterized via ultracentrifugation, microRNA (miR)-301a expression was detected using reverse transcription-quantitative PCR, GEM resistance was evaluated via the Cell Counting Kit-8 assay and the molecular mechanisms were validated by western blotting. The present study results demonstrated that under hypoxic stress, pancreatic cancer cells displayed morphological adaptations typical of cellular stress responses, upregulated miR-301a expression and markedly enhanced GEM resistance. Hypoxia increases the number of exosomes, which can transfer miR-301a to normoxic pancreatic cancer cells. Under normoxic conditions, inhibiting miR-301a expression markedly increased the sensitivity of pancreatic cancer cells to GEM. Furthermore, miR-301a promoted the development of drug resistance by regulating acyl-CoA synthetase long-chain family member 4 (ACSL4) expression. In conclusion, the present study revealed a novel mechanism by which hypoxia-derived exosomal miR-301a promotes GEM resistance in pancreatic cancer by regulating ACSL4. These findings provide a novel potential target to reverse GEM resistance, with theoretical and practical implications for future studies.
    Keywords:  exosomes; gemcitabine resistance; hypoxic microenvironment; microRNA-301a; pancreatic cancer
    DOI:  https://doi.org/10.3892/ol.2025.15352
  5. J Cell Mol Med. 2025 Nov;29(21): e70896
      Extracellular vesicles (EVs), including exosomes, play a pivotal role in intercellular communication by facilitating the transfer of bioactive molecules between cells. These vesicles, which encompass a variety of subtypes such as exosomes, microvesicles and apoptotic bodies, carry functional proteins, mRNAs, miRNAs and other molecular cargo that influence various physiological processes. In particular, skeletal muscle-derived EVs have recently emerged as a novel category of myokines, contributing to muscle homeostasis through paracrine signalling and exerting systemic endocrine effects on metabolic tissues, including the pancreas, adipose tissue and liver. This review systematically examines the regulatory mechanisms of skeletal muscle-derived EVs, with particular focus on exosomes, in mediating inter-organ crosstalk. Additionally, it examines the factors that influence the release of skeletal muscle-derived EVs, particularly exosomes and their subsequent effects on metabolism.
    Keywords:  adipose tissue; extracellular vesicles; liver; pancreas; skeletal muscle
    DOI:  https://doi.org/10.1111/jcmm.70896
  6. Eur Respir J. 2025 Nov 06. pii: 2402060. [Epub ahead of print]
       BACKGROUND: Pneumonia caused by viral or bacterial pathogens such as SARS-CoV2 or Pseudomonas aeruginosa may result in life-threatening disease with a strong contribution of proteases' dysregulation. The present study aimed to systematically characterize the contribution of ADAM10 and ADAM17 on leukocytes and circulating exosomes to viral and bacterial pneumonia.
    METHODS: The analysis of COVID-19 and bacterial pneumonia patient samples was combined with in vivo experiments in conditional knockout animals lacking either ADAM10 or ADAM17 in leukocytes and cell culture experiments for mechanistic studies.
    RESULTS: Hospitalized bacterial pneumonia and COVID-19 patients displayed a severity dependent increase of ADAM10 and ADAM17 activity on exosomes. These exosomes caused pathophysiological changes of cardiomyocytes and the endothelial barrier. In a preclinical murine pneumonia model, we observed that leukocytes contributed to this increase in exosomal proteolytic activity. In the local environment of the lung, ADAM10 orchestrated a pro-inflammatory response with M1 macrophage polarization, increased reactive oxygen species (ROS) generation, cytokine release, tissue damage and oedema formation, whereas ADAM17 seemed to dampen the initial inflammatory response to an anti-infective, ROS-balanced level.
    CONCLUSION: Leukocytic ADAM10 and ADAM17 and their release on exosomes may constitute relevant regulatory elements in bacterial and viral pneumonia, with a potential contribution of exosomes to disease progression and systemic inflammatory responses. Therefore, the diagnostic, prognostic, and therapeutic value of ADAM10 and ADAM17 should be evaluated in further preclinical and translational studies, addressing the changes of the immune response and exosomes as cargo vehicles both at local site and for the prevention of systemic effects.
    DOI:  https://doi.org/10.1183/13993003.02060-2024
  7. Sci Rep. 2025 Nov 05. 15(1): 38701
      Osteomyelitis (OM) is a severe bone infection characterized by inflammation and tissue damage. Macrophages play a crucial role in the inflammatory response during OM, and exosomes derived from bone marrow mesenchymal stem cells (BMSCs) have been proposed as potential therapeutic agents. Previous studies suggest that miR-223, a microRNA involved in inflammatory processes, is dysregulated in OM. This study investigates the role of BMSCs-derived exosomes carrying miR-223 in regulating macrophage pyroptosis, a form of programmed cell death triggered by inflammation. Blood samples were collected from OM patients and control subjects to assess miR-223 expression. BMSCs were treated with LPS to simulate the OM environment. Exosomes were extracted from miR-223 overexpressing BMSCs and characterized. The effects of these exosomes on macrophage survival, apoptosis, and pyroptosis were assessed through CCK-8 assays, flow cytometry, TUNEL staining, ELISA, and western blotting. The miR-223-mediated regulation of Caspase-1 and LACC1 expression was evaluated using specific inhibitors and gene expression analysis. miR-223 expression was significantly reduced in OM patients and in LPS-treated BMSCs. BMSCs-derived exosomes carrying miR-223 (miR-223 exo) enhanced macrophage viability, reduced apoptosis, and mitigated LPS-induced pyroptosis by targeting the NLRP3 inflammasome and Caspase-1 expression. Co-treatment with miR-223 inhibitors and Caspase-1 inhibitors showed that miR-223 regulated macrophage survival and inflammation through Caspase-1 modulation. Further investigation revealed that miR-223 targeted LACC1 to alleviate macrophage pyroptosis, with LACC1 overexpression reversing the protective effects of miR-223. BMSCs-derived exosomes carrying miR-223 play a protective role in OM by regulating macrophage pyroptosis and inflammation. This effect is mediated through the modulation of Caspase-1 and LACC1 expression, highlighting the potential of miR-223-based therapies for OM treatment.
    Keywords:  BMSCs; Exosomes; Macrophages; OM; Pyroptosis
    DOI:  https://doi.org/10.1038/s41598-025-22511-3
  8. Front Immunol. 2025 ;16 1687953
      Cancer remains the leading cause of death worldwide. Despite decades of continuous research, limitations persist in existing therapeutic approaches. Conventional strategies such as surgery, chemotherapy, and radiotherapy, though advanced, face challenges including poor bioavailability, toxic side effects, inadequate targeting of cancer cells, and limited survival benefits. The major issue lies in the inability of improved drug formulations to effectively reach cancer cells. Emerging approaches such as photodynamic therapy (PDT) and immunotherapy have shown greater promise, offering reduced side effects and higher treatment efficiency compared to traditional methods. Various natural and synthetic nanocarriers, including exosomes, liposomes, solid lipid nanoparticles (SLNs) and micelles have been explored as drug delivery vehicles in these therapies. Among them, exosomes, being natural secretory vesicles, have shown unique potential as independent delivery systems. However, challenges and limitations remain in their application for precise cancer targeting. A combinational strategy, integrating exosomes with other lipid-based drug delivery systems (LBDDS), while preserving their intrinsic properties and engineering their surface to carry photosensitizers (PS) or immune modulators, could overcome these barriers. Such well-designed natural cargos may enhance therapeutic efficacy, modulate the tumor microenvironment, and address current shortcomings in cancer therapy. This review highlights the individual applications of PDT and immunotherapy using exosomes and LBDDS, and explores their potential synergistic use for more effective and targeted cancer treatment.
    Keywords:  exosomes; immunotherapy and cancer; liposomes; nanocarriers; photodynamic therapy
    DOI:  https://doi.org/10.3389/fimmu.2025.1687953
  9. Cell Cycle. 2025 Nov 08. 1-23
      Hepatocellular carcinoma (HCC) is a highly aggressive liver cancer, and its progression is significantly influenced by the tumor microenvironment (TME). Tumor-derived exosomes (TEXs), an important component of the TME, significantly influence tumor growth by regulating immune responses, facilitating metastasis, and enhancing resistance to therapy. These extracellular vesicles (EVs) transport bioactive substances, such as proteins, lipids, and nucleic acids that promote interaction between cells in the TME. Recent research indicates that HCC-derived exosomes can inhibit immune cell activity, specifically in T cells, thus creating an immunosuppressive TME that facilitates tumor immune escape. They also augment metastatic capability by restructuring the extracellular matrix and promoting angiogenesis. Moreover, HCC-derived exosomes have been associated with developing resistance to drug therapy by transferring molecules such as apoptotic signals and oncogenic microRNAs, circRNAs and lncRNA. Understanding how HCC-derived exosomes affect immune modulation, metastasis, and drug resistance could yield innovative therapeutic targets to enhance therapy outcomes. This review focuses on recent research on the diverse functions of TEXs in HCC progression.
    Keywords:  Hepatocellular carcinoma; drug resistance; exosome; immune modulation; metastasis
    DOI:  https://doi.org/10.1080/15384101.2025.2583289
  10. Anal Chem. 2025 Nov 06.
      Exosomes, a subset of extracellular vesicles (EVs) ranging in size from 30 to 150 nm, are important, emergent tools in clinical diagnostics and biochemical studies. Exosomes are excreted from cells and participate in many biological processes, including intercellular communication and disease propagation. Challenges in quantification stem from problems in isolating pure and morphologically intact exosomes from complex biomatrices. This study presents a novel method for exosome quantification using capillary channeled polymer (C-CP) fiber spin-down tips for isolation and removal of unbound metal tags, ytterbium chloride (YbCl3) for labeling, and inductively coupled plasma mass spectrometry (ICP-MS) for quantification. Use of polyester (PET) C-CP fiber spin-down tips enables a rapid and cost-effective exosome isolation that preserves the morphological integrity of the exosomes. A well-known chelation reaction between trivalent lanthanide ions (Yb3+ here) and phosphate groups in the exosome phospholipid bilayer allows for sensitive quantification, with labeling optimized at 10 mM YbCl3 for 30 min at 37 °C. Results showed a linear relationship (R2 > 0.95) between Yb ICP-MS response and exosome particle counts, with a detection limit of 1.16 × 102 exosomes mL-1. This integrated workflow has the potential to be combined with targeted biomarker labeling as a comprehensive quantification and identification technique for clinical diagnostics.
    DOI:  https://doi.org/10.1021/acs.analchem.5c04640
  11. Nanomedicine (Lond). 2025 Nov 05. 1-15
       BACKGROUND: Transient receptor potential melastatin 8 (TRPM8) is a cold-sensing cation channel that regulates calcium (Ca2+) levels in cells. Its overexpression is linked to tumor development and progression. TRPM8 activation by specific agonists leads to increased Ca2+ influx, causing stress and apoptosis. This stress can enhance the production and release of exosomes, which have antitumor immunity properties. We hypothesize that activating TRPM8 with nano-icilin can stimulate immune responses when administered peritumorally.
    METHOD: 4T1 cancer cells were treated with icilin nanoparticles and hypothermia to evaluate cytotoxicity, apoptosis, calcium flux, and exosome extraction. Isolated exosomes were characterized and tested in vivo for antitumor immune response in a mouse model. Tumor growth, cytokines (IL-2, IL-12, IL-10, and IL-1β), and immunohistochemistry (IHC) were assessed. Data were analyzed using ANOVA and Duncan's test (P ≤ 0.05).
    RESULTS: TRPM8 activation by icilin nanoparticles triggers apoptosis and calcium influx in 4T1 cells. Exosomes from treated cells exhibited altered size, charge, and increased levels of DAMPs (HMGB1, HSP70). Administering these exosomes significantly inhibited tumor growth, increased CD4+/CD8+ T cells, and elevated IL-2 and IL-12, while reducing IL-10 and PD-L1, thus preventing lung metastasis.
    CONCLUSIONS: Activation of TRPM8 by icilin or cold can induce immunogenic exosomes, enhancing T cell infiltration, proinflammatory cytokines, and tumor suppression, offering a new strategy to boost immune responses against cancer.
    Keywords:  Cancer immunotherapy; TRPM8 channel; cold stress; exosomes; immunogenic cell death; nano-icilin
    DOI:  https://doi.org/10.1080/17435889.2025.2583209
  12. PLoS One. 2025 ;20(11): e0336322
      Exosomes are cell-derived vesicles that play a crucial role in intracellular communication and are promising biomarkers for therapeutic applications. Despite their significant potential, the application of exosomes as biological therapeutics is limited by their low yield and inconsistent production quality. Ionizing radiation is known to enhance exosome release; however, this effect has been primarily studied in cancer cells. Given the critical role of macrophages in immune regulation and their potential for exosome-based therapies, we investigated the impact of gamma radiation on the secretion of macrophage-derived exosomes. This study demonstrated that gamma radiation significantly enhanced exosome release by both naïve and polarized macrophages. This effect was associated with the overexpression of Myh10 and Myo5b, the motor proteins that play crucial roles in exosome biogenesis and secretion. Furthermore, RNA sequencing and western blot analyses identified the EGFR/IGFR-MYC signaling axis as a key upstream pathway regulating the expression of Myh10 and Myo5b, thereby accelerating exosome secretion. These findings provide a deeper understanding of the molecular mechanisms underlying radiation-induced exosome secretion from macrophages and offer a novel strategy for optimizing exosome production to advance exosome-based therapeutic applications.
    DOI:  https://doi.org/10.1371/journal.pone.0336322
  13. World J Stem Cells. 2025 Oct 26. 17(10): 111241
      Mesenchymal stem cells (MSCs) are known for their ability to differentiate into various cell lineages, including osteoblasts (bone-forming cells), and for their significant paracrine effects. Among their secreted products, exosomes have gained considerable attention as nanoscale carriers of bioactive molecules such as non-coding RNAs (ncRNAs). These ncRNAs, including microRNAs, long ncRNAs, and circular ncRNAs, are critical regulators of gene expression and cellular functions. Moreover, MSC-derived exosomes not only offer advantages such as targeted delivery, reduced immunogenicity, and protection of cargo material, but also carry ncRNAs that have therapeutic and diagnostic potential in bone-related disorders. Emerging evidence has highlighted the role of MSC-derived exosomal ncRNAs in osteogenesis, bone remodeling, and intercellular signaling in the bone microenvironment. This review consolidates recent research on the role of MSC-derived exosomal ncRNAs in maintaining bone homeostasis and bone-related disorders via various signaling pathways and epigenetic modifications. Furthermore, we explore the therapeutic potential of MSC-derived exosomal ncRNAs as biomarkers and therapeutic targets. This comprehensive review offers key insights into the regulatory roles of MSC-derived exosomal ncRNAs in bone biology and their clinical significance in bone-related diseases.
    Keywords:  Bone disorders; Bone homeostasis; Mesenchymal stem cell-derived exosomes; Non-coding RNAs; Signaling pathway
    DOI:  https://doi.org/10.4252/wjsc.v17.i10.111241
  14. Respir Med. 2025 Nov 05. pii: S0954-6111(25)00538-4. [Epub ahead of print] 108475
       INTRODUCTION: COVID-19 has become a global pandemic, potentially leading to conditions like ARDS. The infection triggers severe inflammation and cytokine storms, damaging the lungs and other organs. MSCs and their derived exosomes are suggested as therapeutic options for reducing inflammation and modulating the immune response. This study evaluates the safety and efficacy of exosomes from umbilical MSCs in ARDS patients.
    MATERIALS AND METHODS: This randomized, double-blind, placebo-controlled pilot trial, conducted at Razi Hospital, Ahvaz, Iran, between October and December 2022, enrolled thirty hospitalized patients with COVID-19 and ARDS. Group A (n=15) received a single intravenous dose of hUCMSC-Exos (5×1010 particles in 2-5 mL PBS), and Group B (n=15) received a placebo (PBS), in addition to standard-of-care treatments. Clinical outcomes, inflammatory biomarkers (TNF-α, IL-6, CRP), and immune cell subsets (CD3+, CD4+, CD8+) were assessed at baseline and seven days after treatment. Categorical variables were reported as percentages, continuous variables as mean ± SD. Non-parametric tests and Quade ANCOVA were used for group comparisons and adjustment for baseline differences. Subgroup analyses based on Actemra use were performed to account for potential confounding effects on inflammatory and immune parameters.
    RESULTS: The intervention group showed a significant reduction in inflammatory markers (TNF-α and CRP) and changes in immune cells, particularly CD3+ and CD4+. While no significant differences were found in mortality and hospitalisation duration, the intervention group demonstrated improvements in inflammatory profiles and immune responses. The PRIEST score, assessing disease severity, was influenced by the intervention-time interaction, suggesting the therapeutic potential of hUCMSC-Exos in modulating disease progression. Subgroup analyses revealed that Actemra influenced IL-6 and lymphocyte counts, while TNF-α, CRP, and adaptive immune trends persisted in the exosome group, indicating effects beyond Actemra. Unequal Actemra distribution was accounted for to reduce bias.
    CONCLUSION: The administration of hUCMSC-Exos was safe and associated with favorable trends, including reductions in TNF-α and CRP, as well as improvements in immune cell subsets (CD3+, CD4+, and CD8+). However, the independent effects of exosomes cannot be fully determined due to the concomitant use of therapies, including Actemra. The findings highlight the potential of exosome therapy in modulating inflammation and adaptive immune responses; however, larger, controlled trials stratified by concomitant treatments are required to confirm these preliminary results and clarify the therapeutic role of exosomes in COVID-19-associated ARDS.
    Keywords:  COVID-19; Cell-Derived Microparticles; Clinical Trial; Mesenchymal Stem Cells; Respiratory Distress Syndrome
    DOI:  https://doi.org/10.1016/j.rmed.2025.108475