bims-engexo Biomed News
on Engineered exosomes
Issue of 2025–03–30
nine papers selected by
Ravindran Jaganathan, Universiti Kuala Lumpur



  1. Histol Histopathol. 2025 Mar 12. 18903
      Extracellular vesicles (EVs) are lipid bilayer nanoparticles that encapsulate proteins, lipids, nucleic acids, and small molecules and display low immunogenicity, high stability, and cross-species transmission. By applying engineering technologies, the surface of EVs can be modified and loaded with cargo with therapeutic properties. Thus, engineered EVs can play important roles in preventing and treating various diseases. However, many challenges and uncertainties are faced in the clinical translation of EVs. In this review, we comprehensively analyzed the types of EVs derived from animal cells, plant cells, and microorganisms and summarized their biological properties and potential for engineering modifications. Furthermore, we also explored their therapeutic potential and discussed recent advancements in relevant clinical trials, aiming to provide scientific guidance for future research on the engineering of EVs and precision treatment of clinical diseases.
    DOI:  https://doi.org/10.14670/HH-18-903
  2. Pharmaceutics. 2025 Mar 13. pii: 366. [Epub ahead of print]17(3):
      Chronic wounds pose a significant healthcare challenge, impacting millions of patients worldwide and burdening healthcare systems substantially. These wounds often occur as comorbidities and are prone to infections. Such infections hinder the healing process, complicating clinical management and proving recalcitrant to therapy. The environment within the wound itself poses challenges such as lack of oxygen, restricted blood flow, oxidative stress, ongoing inflammation, and bacterial presence. Traditional systemic treatment for such chronic peripheral wounds may not be effective due to inadequate blood supply, resulting in unintended side effects. Furthermore, topical applications are often impervious to persistent biofilm infections. A growing clinical concern is the lack of effective therapeutic modalities for treating chronic wounds. Additionally, the chemically harsh wound microenvironment can reduce the effectiveness of treatments, highlighting the need for drug delivery systems that can deliver therapies precisely where needed with optimal dosages. Compared to cell-based therapies, exosome-based therapies offer distinct advantages as a cell-free approach for chronic wound treatment. Exosomes are of endosomal origin and enable cell-to-cell communications, and they possess benefits, including biocompatibility and decreased immunogenicity, making them ideal vehicles for efficient targeting and minimizing off-target damage. However, exosomes are rapidly cleared from the body, making it difficult to maintain optimal therapeutic concentrations at wound sites. The hydrogel-based approach and development of biocompatible scaffolds for exosome-based therapies can be beneficial for sustained release and prolong the presence of these therapeutic exosomes at chronic wound sites. Engineered exosomes have been shown to possess stability and effectiveness in promoting wound healing compared to their unmodified counterparts. Significant progress has been made in this field, but further research is essential to unlock their clinical potential. This review seeks to explore the benefits and opportunities of exosome-based therapies in chronic wounds, ensuring sustained efficacy and precise delivery despite the obstacles posed by the wound environment.
    Keywords:  biocompatible scaffolds; cell–cell communication; engineered exosomes; exosome-targeted therapies; functional wound closure
    DOI:  https://doi.org/10.3390/pharmaceutics17030366
  3. JACC Basic Transl Sci. 2025 Feb;pii: S2452-302X(24)00324-3. [Epub ahead of print]10(2): 131-147
      Circular RNAs are associated with cardiovascular disease, including coronary artery disease, but the mechanisms have not been completely elucidated. We found a new protein, circBTBD7-420aa, encoded by hsa_circ_0000563. Our results suggest that circBTBD7-420aa may inhibit the abnormal proliferation and migration of human coronary artery smooth muscle cells by promoting SLC3A2 degradation through the ubiquitin-proteasome pathway. In addition, we constructed engineered exosomes loaded with circBTBD7-420aa that can target vascular smooth muscle cells by modifying peptide fragments targeting osteopontin. This study suggests that circBTBD7-420aa may inhibit the progression of atherosclerosis and serve as a new target for the diagnosis and treatment of coronary artery disease.
    Keywords:  SLC3A2; circular RNA; coronary artery disease; engineered exosomes; translation; ubiquitination
    DOI:  https://doi.org/10.1016/j.jacbts.2024.09.003
  4. Brain Res. 2025 Mar 20. pii: S0006-8993(25)00149-0. [Epub ahead of print]1855 149590
      Exosomes are a subject of continuous investigation due to their function as extracellular vesicles (EVs) that significantly contribute to the pathophysiology of certain neurodegenerative disorders (NDD), including Alzheimer's disease (AD). Exosomes have shown the potential to carry both therapeutic and pathogenic materials; hence, researchers have used exosomes for medication delivery applications. Exosomes have reduced immunogenicity when used as natural drug delivery vehicles. This guarantees the efficient delivery of the medication without causing significant side reactions. Exosomes have lately enabled the potential for drug delivery in AD, along with promising future therapeutic uses for the detection of neurodegenerative disorders. Furthermore, exosomes have been examined for their prospective use in illness diagnosis and prediction before the manifestation of symptoms. This review will document prior studies and will concentrate on the rationale behind the substantial potential of exosomes in the treatment of AD and their prospective use as a diagnostic and predictive tool for this condition.
    Keywords:  Alzheimer’s disease; Blood-brain barrier; Exosome; Neuroinflammation; miRNA
    DOI:  https://doi.org/10.1016/j.brainres.2025.149590
  5. Cancers (Basel). 2025 Mar 10. pii: 940. [Epub ahead of print]17(6):
      Exosomes have emerged as pivotal players in precision oncology, offering innovative solutions to longstanding challenges such as metastasis, therapeutic resistance, and immune evasion. These nanoscale extracellular vesicles facilitate intercellular communication by transferring bioactive molecules that mirror the biological state of their parent cells, positioning them as transformative tools for cancer diagnostics and therapeutics. Recent advancements in exosome engineering, artificial intelligence (AI)-driven analytics, and isolation technologies are breaking barriers in scalability, reproducibility, and clinical application. Bioengineered exosomes are being leveraged for CRISPR-Cas9 delivery, while AI models are enhancing biomarker discovery and liquid biopsy accuracy. Despite these advancements, key obstacles such as heterogeneity in exosome populations and the lack of standardized isolation protocols persist. This review synthesizes pioneering research on exosome biology, molecular engineering, and clinical translation, emphasizing their dual roles as both mediators of tumor progression and tools for intervention. It also explores emerging areas, including microbiome-exosome interactions and the integration of machine learning in exosome-based precision medicine. By bridging innovation with translational strategies, this work charts a forward-looking path for integrating exosomes into next-generation cancer care, setting it apart as a comprehensive guide to overcoming clinical and technological hurdles in this rapidly evolving field.
    Keywords:  biomarkers; cancer diagnostics; drug delivery; exosomes; extracellular vesicles (EVs); immune modulation; liquid biopsies; precision medicine; therapeutic resistance; tumor microenvironment (TME); tumor-derived exosomes (TDEs)
    DOI:  https://doi.org/10.3390/cancers17060940
  6. Cell Rep Med. 2025 Mar 14. pii: S2666-3791(25)00106-5. [Epub ahead of print] 102033
      Drug-induced liver injury (DILI) involves multifaceted pathogenesis, necessitating effective therapeutic strategies. Wnt2, secreted by liver sinusoidal endothelial cell (LSEC), activates the Wnt/β-catenin signaling pathway to promote hepatocyte proliferation after injury. To address the dual challenges of targeted delivery and phagocytosis evasion, we develop a combined "eat me/don't eat me" strategy. RLTRKRGLK (RLTR) peptide-functionalized exosomes are engineered by inserting DMPE-PEG2000-CRLTRKRGLK into the lipid membrane of exosome derived from bEnd.3 cell. Surface-displayed RLTR mediates exosomal enrichment in LSEC, while CD47 engineering reduces macrophage clearance via "don't eat me" signaling. Then, lentiviral transfection enables stable encapsulation of functional Wnt2 mRNA into ExoCD47 (designated Wnt2@ExoCD47). In both acetaminophen (APAP) and dimethylnitrosamine (DMN)-induced murine liver injury models, RLTR-Wnt2@ExoCD47 demonstrates LSEC-specific targeting and significant hepatoprotection. This engineered exosome platform provides a therapeutic strategy for DILI.
    Keywords:  Wnt/β-catenin pathway; drug-induced liver injury; engineered exosome; liver regeneration; liver sinusoidal endothelial cell
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102033
  7. Cell Biochem Biophys. 2025 Mar 24.
      Exosomes are tiny extracellular vesicles that are essential for intercellular communication and have shown great promise in the detection and treatment of disease. They are especially useful in the treatment of cancer, cardiovascular conditions, and neurological diseases because of their capacity to transport bioactive substances including proteins, lipids, and nucleic acids. Because of their low immunogenicity, ability to traverse biological barriers, and biocompatibility, exosome-based medicines have benefits over conventional treatments. Large-scale production, standardization of separation methods, possible immunological reactions, and worries about unforeseen biological effects are some of the obstacles that still need to be overcome. Furthermore, there are major barriers to the clinical use of exosomes due to their complex cargo sorting mechanisms and heterogeneity. Future studies should concentrate on enhancing separation and purification procedures, optimizing exosome engineering techniques, and creating plans to reduce immune system modifications. This review examines the most recent developments in exosome-based diagnostics and treatments, identifies current issues, and suggests ways to improve their clinical translation in the future.
    Keywords:  Cancer; Drug delivery; Engineering Strategies; Exosomes; Therapeutics
    DOI:  https://doi.org/10.1007/s12013-025-01730-5
  8. J Nanobiotechnology. 2025 Mar 24. 23(1): 241
      Exosomes-loaded hydrogels have potential value in wound treatment. Current studies focus on improving hydrogels' biocompatibility and optimizing different stem cell-derived exosomes for better therapeutic effect. Herein, we present a novel biocompatible recombinant human collagen (RHC) hydrogel loading with different MSCs-derived exosomes for promoting wound healing. We modify the RHC with methacrylate anhydride (MA) at optimal concentration, generating collagen hydrogel (RHCMA) with ideal physiochemical properties for exosome delivery (MSC-exos@RHCMA). Exosomes derived from human adipose-derived MSCs (ADSC-exos), bone marrow-derived MSCs (BMSC-exos) and umbilical cord MSCs (ucMSC-exos) are harvested from the culture supernatants and are loaded into RHCMA, respectively. These three hydrogel systems exhibit desired sustained release features, and can significantly improve cell proliferation and migration. In addition, these MSC-exos@RHCMAs show excellent therapeutic performance in treating the wounds of rats. Notably, we have demonstrated that the healing effect occurs best under the treatment of ucMSC-exos@RHCMA, following inflammatory resolution, angiogenesis, and collagen formation. These results would supply important value for the clinical application of MSC-exos in wound treatment in the future.
    Keywords:  Drug delivery; Exosome; Hydrogel; MSC; Wound healing
    DOI:  https://doi.org/10.1186/s12951-025-03319-9
  9. Mol Genet Genomics. 2025 Mar 25. 300(1): 36
      Intrauterine adhesion (IUA) refer to persistent inflammation and fibrosis due to damaged or infected endometrium and eventually lead to dysfunction. This study aimed to explore the therapeutic effects of exosomes (Exos) derived from placental mesenchymal stem cells (PMSCs) on endometrial repair in a rat model of IUA and to elucidate the underlying molecular mechanisms. PMSCs were characterized using flow cytometry and differentiation assays (osteogenic, adipogenic, and chondrogenic). Exos were isolated via ultracentrifugation and validated through transmission electron microscopy, nanoparticle tracking analysis and Western blot. An IUA model was established via electrocoagulation, and endometrial repair was assessed using hematoxylin-eosin (HE) and Masson staining. RNA sequencing, differential expression analysis and protein-protein interaction (PPI) network construction were employed to investigate the molecular mechanisms of PMSC Exos mediated repair. The role of miR-143 in targeting MyD88 and modulating the NF-κB signaling pathway was confirmed using Dual-Luciferase Reporter Assay and qRT-PCR. PMSC Exos significantly improved endometrial thickness, increased glandular number and reduced fibrosis in the IUA model. RNA sequencing and differential expression analysis screened 3980 differentially expressed genes (DEGs) common to the IUA vs normal groups and Exo vs IUA groups. Enrichment analysis revealed significant involvement of immune system processes, natural killer cell-mediated cytotoxicity and NF-κB signaling. PMSC Exos delivered miR-143, which targeted MyD88, thereby regulating the NF-κB pathway. PMSC Exos effectively repaired endometrial damage in the IUA model by modulating the NF-κB signaling pathway through miR-143 delivery. These findings suggest that PMSC Exos hold promise as a novel therapeutic strategy for IUA, offering insights into the molecular mechanisms underlying endometrial repair.
    Keywords:  Exosomes; Immunity; Intrauterine adhesion; MyD88; NF-κB signaling; Placental mesenchymal stem cell
    DOI:  https://doi.org/10.1007/s00438-025-02241-x