151
|
Weiser T, Hoch CC, Petry J, Shoykhet M, Schmidl B, Yazdi M, Hachani K, Mergner J, Theodoraki MN, Azimzadeh O, Multhoff G, Bashiri Dezfouli A, Wollenberg B. Head and neck squamous cell carcinoma-derived extracellular vesicles mediate Ca²⁺-dependent platelet activation and aggregation through tissue factor. Cell Commun Signal 2025; 23:210. [PMID: 40312345 PMCID: PMC12044835 DOI: 10.1186/s12964-025-02215-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy, characterized by poor clinical outcomes, primarily driven by high rate of locoregional recurrence and metastasis. Extensive heterogeneity among the tumor cells as well as modulation of a highly immunosuppressive tumor microenvironment shape cancer progression. Shedding of extracellular vesicles (EVs) derived from tumor cells is a critical mediator of the disease initiating horizontal transfer of tumor components into platelets. This triggers platelet activation and thromboinflammation fueling tumor progression through multiple mechanisms. METHODS HNSCC-derived EVs isolated from HNSCC cell lines (SAS, UD-SCC 5) using size exclusion chromatography and characterized via flow cytometry, electron microscopy, nanoparticle tracking analysis and Western blotting, were used to induce platelet activation and aggregation, measured by aggregometry, flow cytometry, as well as the release of chemokines and Adenosine triphosphate, which were quantified using enzyme-linked immunosorbent assays (ELISA). Mechanistic investigations included inhibitor assays, thrombin activity measurements, and proteomic analyses. RESULTS We could show that EVs do not activate platelets through the FcγRIIa-IgG axis but platelet activation and aggregation is induced in a calcium-dependent manner, primarily mediated by EV-associated tissue factor. Proteomic analysis confirmed the presence of tissue factor in these vesicles, implicating its involvement in initiating the coagulation cascade, that leads to platelet activation and aggregation. This process was characterized by delayed aggregation kinetics and relied on thrombin activation, as the inhibition of thrombin and its receptors reduced platelet aggregation. HNSCC-derived EVs are pivotal in establishing a prothrombotic environment by promoting platelet activation and aggregation through tissue factor-dependent thrombin generation. CONCLUSION These findings indicate a therapeutic potential of targeting EV-mediated pathways as a therapeutic approach to alleviate thrombotic complications in HNSCC patients. Subsequent animal studies will be crucial to validate and extend these observations, providing deeper insight into their clinical implications.
Collapse
Affiliation(s)
- Tobias Weiser
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Cosima C Hoch
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Julie Petry
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Maria Shoykhet
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Benedikt Schmidl
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Khouloud Hachani
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at Klinikum rechts der Isar (BayBioMS@MRI), TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Marie-Nicole Theodoraki
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Omid Azimzadeh
- Section Radiation Biology, Federal Office for Radiation Protection (BfS), Neuherberg, Germany
| | - Gabriele Multhoff
- Central Institute for Translational Cancer Research, Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Ali Bashiri Dezfouli
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
- Central Institute for Translational Cancer Research, Department of Radiation Oncology, TUM School of Medicine and Health, Technical University of Munich (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Barbara Wollenberg
- Department of Otolaryngology, Head and Neck Surgery, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany.
| |
Collapse
|
152
|
Patel N, Avery E, Huang Y, Chung EJ. Developing Therapeutically Enhanced Extracellular Vesicles for Atherosclerosis Therapy. Adv Healthc Mater 2025; 14:e2404398. [PMID: 40192440 DOI: 10.1002/adhm.202404398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/19/2025] [Indexed: 05/17/2025]
Abstract
Atherosclerosis is a chronic condition and the leading cause of death worldwide. While statin therapy is the clinical standard, many patients still experience acute cardiovascular events. To develop better therapies, the group previously delivered microRNA-145 (miR-145) via micellar nanoparticles to vascular smooth muscle cells (VSMCs) to inhibit atherosclerosis. However, for chronic diseases requiring repeat dosing, synthetic nanoparticles have drawbacks such as immunogenic response and low delivery efficiency. To meet this challenge, therapeutically enhanced extracellular vesicles (EVs) are engineered as a biologically-derived nanoparticle modality to mitigate atherosclerosis. A novel strategy is employed to load miR-145 into EVs using ExoMotifs-short miRNA sequences that facilitate miR cargo loading. EVs are further functionalized with a monocyte chemoattractant 1 (MCP-1) peptide, which binds to C-C chemokine receptor 2 upregulated in pathogenic VSMCs. Mouse aortic smooth muscle cell MCP-1-miR-145 EVs restored VSMC gene expression and function in vitro. Moreover, compared to miR-145-loaded synthetic nanoparticles, MCP-1-miR-145 EVs exerted similar therapeutic effects but with 25,000x less miR-145 cargo. Lastly, MCP-1-miR-145 EVs inhibited plaque growth in mid-stage ApoE-/- atherosclerotic mice at a miR-145 dose 5000x less than synthetic nanoparticles. Collectively, it is demonstrated that genetic engineering of VSMCs with miR-145 produces therapeutically boosted EVs that reduce atherosclerosis plaque burden.
Collapse
Affiliation(s)
- Neil Patel
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Elijah Avery
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Yi Huang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Bridge Institute, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
153
|
Fatima A, Sanyal S, Jha GK, Kaliki S, Pallavi R. The enigmatic world of tear extracellular vesicles (EVs)-exploring their role in ocular health and beyond. FEBS Lett 2025; 599:1346-1372. [PMID: 39961136 DOI: 10.1002/1873-3468.70004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/29/2024] [Accepted: 01/10/2025] [Indexed: 05/27/2025]
Abstract
Extracellular vesicles (EVs) are released by all kind of cells into the extracellular space, where they shuttle parental cell-derived molecular cargoes (DNA, RNA, proteins) to both adjacent and distant cells, influencing the physiology of target cells. Their specific cargo content and abundance in liquid biopsies make them excellent candidates for biomarker studies. Indeed, EVs isolated from various body fluids, including blood, pleural fluid, urine, cerebrospinal fluid, saliva, milk, ascites, and tears, have been recognized for their potential as biomarkers in diagnosis, monitoring treatment, and predicting outcomes for various diseases. Increasing studies suggest that tears have great promise as a noninvasive liquid biopsy source for EVs. Our aim here is to provide a comprehensive review of the exploration of tears as a noninvasive reservoir of EVs and their contents, evaluating their accessibility and potential utility as a liquid biopsy method. Additionally, the potential of tear EVs in various cancers, including ocular cancer, is discussed. Finally, the advantages and challenges of employing tear-based liquid biopsy for EVs for the disease's biomarker studies are evaluated.
Collapse
Affiliation(s)
- Azima Fatima
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, 500034, Telangana State, India
| | - Shalini Sanyal
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, 500034, Telangana State, India
| | - Gaurab Kumar Jha
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, 500034, Telangana State, India
| | - Swathi Kaliki
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, 500034, Telangana State, India
| | - Rani Pallavi
- The Operation Eyesight Universal Institute for Eye Cancer, LV Prasad Eye Institute, Hyderabad, India
- Prof. Brien Holden Eye Research Centre, LV Prasad Eye Institute, Hyderabad, 500034, Telangana State, India
| |
Collapse
|
154
|
Liu C, Luo Y, Zhou H, Lin M, Zang D, Chen J. Immune cell-derived exosomal non-coding RNAs in tumor microenvironment: Biological functions and potential clinical applications. Chin J Cancer Res 2025; 37:250-267. [PMID: 40353080 PMCID: PMC12062983 DOI: 10.21147/j.issn.1000-9604.2025.02.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/25/2025] [Indexed: 05/14/2025] Open
Abstract
The intricate interactions between immune cells and tumors exert a profound influence on cancer progression and therapeutic efficacy. Within the tumor microenvironment, exosomes have emerged as pivotal mediators of intercellular communication, with their cargo of non-coding RNAs (ncRNAs) serving as key regulatory elements. This review examines the multifaceted roles of immune cell-derived exosomal ncRNAs in tumor biology. The involvement of various immune cells, including T cells, B cells, natural killer cells, macrophages, neutrophils, and myeloid-derived suppressor cells, in utilizing exosomal ncRNAs to regulate tumor initiation and progression is explored. Additionally, the biogenesis and delivery mechanisms of these immune cell-derived exosomal ncRNAs are discussed, alongside their potential clinical applications in cancer.
Collapse
Affiliation(s)
- Chenguang Liu
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Yawen Luo
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Huan Zhou
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Meixi Lin
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Dan Zang
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Jun Chen
- Department of Oncology, the Second Hospital of Dalian Medical University, Dalian 116023, China
| |
Collapse
|
155
|
Wu L, Wei J, Zhan Y, Xiao X, Xu X, Huang C, Long T, Li Y, Fu B, Wang M, Gao C. Comparative evaluation of methods for isolating extracellular vesicles from ICC cell culture supernatants: Insights into proteomic and glycomic analysis. Cell Commun Signal 2025; 23:207. [PMID: 40301937 PMCID: PMC12042569 DOI: 10.1186/s12964-025-02207-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are nanoscale structures involved in intercellular communication and play a key role in cancer pathology. Intrahepatic cholangiocarcinoma (ICC) is a highly invasive malignancy marked by abnormal sialylated glycosylation. Analyzing proteins and glycans in EVs provides insights into ICC molecular subtyping and mechanisms. Optimizing EV isolation methods for ICC-derived EVs enables comprehensive proteomic and glycomic analysis. METHODS We systematically evaluated five EV isolation methods-Ultracentrifugation (UC), exoEasy, Total Exosome Isolation (TEI), EVtrap, and ÄKTA-by analyzing the biophysical properties, proteomic profiles, and glycomic structures of EVs. Subsequently, we applied TMT-based quantitative proteome and light/heavy methylamine labeling for the quantification of sialylated N-glycan linkage isomers to investigate alterations in proteins and N-glycans within EVs secreted by HuCCT1 and HCCC-9810 cells with overexpressing ST6 β‑galactoside α2,6‑sialyltransferase 1 (ST6GAL1). RESULTS By evaluating the biophysical properties, proteome, and N-glycome of EVs extracted using five different methods, UC was identified as the optimal approach for this study, as it offered a balance between operational complexity, cost-effectiveness, and the preservation of EVs activity. In this study, a total of 1,928 high-confidence proteins and over 84 high-confidence glycans were quantified. EVs secreted by HuCCT1 and HCCC-9810 cells overexpressing ST6GAL1 exhibited consistent upregulation of 16 proteins, consistent downregulation of 10 proteins, as well as consistent upregulation of 3 glycans and consistent downregulation of 3 glycans. CONCLUSIONS Quantitative proteomic and glycomic analysis of ICC-derived EVs revealed that ST6GAL1 overexpression led to significant alterations in proteins involved in cancer cell adhesion and glycosylation pathways, along with specific changes in N-glycan structures. Notably, these modifications extended beyond α2,6-sialylation, suggesting that interactions between glycosyltransferases and glycans may drive these alterations.
Collapse
Affiliation(s)
- Linlin Wu
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Jiao Wei
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yueping Zhan
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xiao Xiao
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Xuewen Xu
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Chenjun Huang
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Tian Long
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Yueyue Li
- Shanghai Cancer Center and Institutes of Biomedical Sciences and Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200030, China
| | - Bin Fu
- Shanghai Cancer Center and Institutes of Biomedical Sciences and Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, 200030, China
| | - Mengmeng Wang
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Chunfang Gao
- Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| |
Collapse
|
156
|
Dauphin T, de Beaurepaire L, Salama A, Pruvost Q, Claire C, Haurogné K, Sourice S, Dupont A, Bach JM, Hervé J, Olmos E, Bosch S, Lieubeau B, Mosser M. Scalability of spheroid-derived small extracellular vesicles production in stirred systems. Front Bioeng Biotechnol 2025; 13:1516482. [PMID: 40365014 PMCID: PMC12069995 DOI: 10.3389/fbioe.2025.1516482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Small extracellular vesicle (sEV)-based therapies have gained widespread interest, but challenges persist to ensure standardization and high-scale production. Implementing upstream processes in a chemically defined media in stirred-tank bioreactors (STBr) is mandatory to closely control the cell environment, and to scale-up production, but it remains a significant challenge for anchorage-dependent cells. Methods We used a human β cell line, grown as monolayer or in suspension as spheroid in stirred systems. We assessed the consequences of culturing these cells in 3D with, or without fetal bovine serum in a chemically defined medium, for cell growth, viability and metabolism. We next explored how different scale-up strategies might influence cell and spheroid formation in spinner flask, with the aim to transfer the process in instrumented Ambr®250 STBr. Lastly, we analyzed and characterized sEV production in monolayer, spinner flask and STBr. Results and discussion Generation of spheroids in a chemically defined medium allowed the culture of highly viable cells in suspension in stirred systems. Spheroid size depended on the system's volumetric power input (P/V), and maintaining this parameter constant during scale-up proved to be the optimal strategy for standardizing the process. However, transferring the spinner flask (SpF) process to the Ambr®250 STBr at constant P/V modified spheroid size, due to important geometric differences and impeller design. Compared to a monolayer reference process, sEV yield decreased two-fold in SpF, but increased two-fold in STBr. Additionally, a lower expression of the CD63 tetraspanin was observed in sEV produced in both stirred systems, suggesting a reduced release of exosomes compared to ectosomes. This study addresses the main issues encountered in spheroid culture scale-up in stirred systems, rather conducive for the production of ectosomes.
Collapse
Affiliation(s)
| | | | | | | | - Clémentine Claire
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
- Oniris VetAgroBio, B-FHIT, Nantes, France
| | | | | | - Aurélien Dupont
- CNRS, INSERM, BIOSIT_UAR 3480, Univ Rennes, Inserm 018, Rennes, France
| | - Jean-Marie Bach
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
- Oniris VetAgroBio, B-FHIT, Nantes, France
| | - Julie Hervé
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
| | - Eric Olmos
- University of Lorraine, CNRS, LRGP, Nancy, France
| | | | | | - Mathilde Mosser
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
- Oniris VetAgroBio, B-FHIT, Nantes, France
| |
Collapse
|
157
|
Mas-Bargues C, Huete-Acevedo J, Arnal-Forné M, Sireno L, Pérez V, Borrás C. Extracellular Vesicles as Epigenetic Regulators of Redox Homeostasis: A Systematic Review and Meta-Analysis. Antioxidants (Basel) 2025; 14:532. [PMID: 40427414 PMCID: PMC12108429 DOI: 10.3390/antiox14050532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Extracellular vesicles (EVs) are emerging as key regulators of cellular communication, with increasing evidence supporting their role in oxidative stress (OS) modulation. In particular, the miRNA cargo of EVs plays a crucial role in mitigating OS and promoting redox balance through both direct antioxidant effects and epigenetic regulation. This study aimed to evaluate the impact of EVs on OS markers, influenced by their miRNA-mediated effects and potential epigenetic modifications in target cells. A systematic literature search was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines to identify studies reporting the effects of EVs on OS parameters. A meta-analysis was performed on key OS biomarkers, including reactive oxygen species (ROS), superoxide dismutase (SOD), glutathione (GSH), and malondialdehyde (MDA). The heterogeneity of EV isolation and characterization techniques was also analyzed. The included studies demonstrated that EVs exert significant antioxidant effects by reducing ROS levels, increasing SOD activity and GSH levels, and lowering MDA levels. These effects were largely attributed to EV-miRNAs, which induce epigenetic modifications that modulate redox-related signaling pathways. However, the variability in EV isolation methods and characterization approaches highlights the need for standardization to improve data comparability. Despite their therapeutic potential, this significant heterogeneity in EV research remains a barrier to translation. Moreover, further exploration of epigenetic mechanisms is essential to fully harness their benefits for OS-related diseases.
Collapse
Grants
- : This research was funded by Grants PID2020-113839RB-I00 funded by Ministerio de Ciencia, Innovación y Universidades and CIAICO/2022/190 funded by Conselleria de Educación, Cultura, Universidades, PI-2023-004 funded by VCL-Bioclinic, and AP2024VLC-08 fun : This research was funded by Grants PID2020-113839RB-I00 funded by Ministerio de Ciencia, Innovación y Universidades and CIAICO/2022/190 funded by Conselleria de Educación, Cultura, Universidades, PI-2023-004 funded by VCL-Bioclinic, and AP2024VLC-08 fun
Collapse
Affiliation(s)
- Cristina Mas-Bargues
- MiniAging/Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.M.-B.); (J.H.-A.); (M.A.-F.)
| | - Javier Huete-Acevedo
- MiniAging/Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.M.-B.); (J.H.-A.); (M.A.-F.)
| | - Marta Arnal-Forné
- MiniAging/Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.M.-B.); (J.H.-A.); (M.A.-F.)
| | - Laura Sireno
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy;
| | - Virgilio Pérez
- Department of Applied Economics (Quantitative Methods), Faculty of Economics, University of Valencia, 46022 Valencia, Spain;
| | - Consuelo Borrás
- MiniAging/Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), Institute of Health Research-INCLIVA, 46010 Valencia, Spain; (C.M.-B.); (J.H.-A.); (M.A.-F.)
| |
Collapse
|
158
|
Ramezani A, Rahnama M, Mahmoudian F, Shirazi F, Ganji M, Bakhshi S, Khalesi B, Hashemi ZS, Khalili S. Current Understanding of the Exosomes and Their Associated Biomolecules in the Glioblastoma Biology, Clinical Treatment, and Diagnosis. J Neuroimmune Pharmacol 2025; 20:48. [PMID: 40299204 DOI: 10.1007/s11481-025-10204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Glioblastoma is the most common and aggressive brain tumor with a low survival rate. Due to its heterogeneous composition, high invasiveness, and frequent recurrence after surgery, treatment success has been limited. In addition, due to the brain's unique immune status and the suppressor tumor microenvironment (TME), glioblastoma treatment has faced more challenges. Exosomes play a critical role in cancer metastasis by regulating cell-cell interactions that promote tumor growth, angiogenesis, metastasis, treatment resistance, and immunological regulation in the tumor microenvironment. This review explores the pivotal role of exosomes in the development of glioblastoma, with a focus on their potential as non-invasive biomarkers for prognosis, early detection and real-time monitoring of disease progression. Notably, exosome-based drug delivery methods hold promise for overcoming the blood-brain barrier (BBB) and developing targeted therapies for glioblastoma. Despite challenges in clinical translation, the potential for personalized exosome = -054321`therapies and the capacity to enhance therapeutic responses in glioblastoma, present intriguing opportunities for improving patient outcomes. It seems that getting a good and current grasp of the role of exosomes in the fight against glioblastoma would properly serve the scientific community to further their understanding of the related potentials of these biological moieties.
Collapse
Affiliation(s)
- Aghdas Ramezani
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Rahnama
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Shirazi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Mahmoud Ganji
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shohreh Bakhshi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Education and Extension Organization, Razi Vaccine and Serum Research Institute, Agricultural Research, Karaj, 3197619751, Iran
| | - Zahra Sadat Hashemi
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran.
| |
Collapse
|
159
|
Thapa HB, Passegger CA, Fleischhacker D, Kohl P, Chen YC, Kalawong R, Tam-Amersdorfer C, Gerstorfer MR, Strahlhofer J, Schild-Prüfert K, Zechner EL, Blesl A, Binder L, Busslinger GA, Eberl L, Gorkiewicz G, Strobl H, Högenauer C, Schild S. Enrichment of human IgA-coated bacterial vesicles in ulcerative colitis as a driver of inflammation. Nat Commun 2025; 16:3995. [PMID: 40301356 PMCID: PMC12041585 DOI: 10.1038/s41467-025-59354-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
The gut microbiome contributes to chronic inflammatory responses in ulcerative colitis (UC), but molecular mechanisms and disease-relevant effectors remain unclear. Here we analyze the pro-inflammatory properties of colonic fluid obtained during colonoscopy from UC and control patients. In patients with UC, we find that the pelletable effector fraction is composed mostly of bacterial extracellular vesicles (BEVs) that exhibit high IgA-levels and incite strong pro-inflammatory responses in IgA receptor-positive (CD89+) immune cells. Biopsy analyses reveal higher infiltration of CD89+ immune cells in the colonic mucosa from patients with UC than control individuals. Further studies show that IgA-coated BEVs, but not host-derived vesicles nor soluble IgA, are potent activators of pro-inflammatory responses in CD89+ cells. IgA-coated BEVs also exacerbate intestinal inflammation in a dextran sodium sulfate colitis model using transgenic mice expressing human CD89. Our data thus implicate a link between IgA-coated BEVs and intestinal inflammation via CD89+ immune cells, and also hint a potential new therapeutic target for UC.
Collapse
Affiliation(s)
- Himadri B Thapa
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Christina A Passegger
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | | | - Paul Kohl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Yi-Chi Chen
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Ratchara Kalawong
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Carmen Tam-Amersdorfer
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Michael R Gerstorfer
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Jana Strahlhofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
- Field of Excellence Biohealth - University of Graz, Graz, Austria
| | - Andreas Blesl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Lukas Binder
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Georg A Busslinger
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Gregor Gorkiewicz
- BioTechMed, Graz, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
| | - Christoph Högenauer
- BioTechMed, Graz, Austria.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed, Graz, Austria.
- Field of Excellence Biohealth - University of Graz, Graz, Austria.
- Austrian Agency for Health and Food Safety (AGES), Institute for Medical Microbiology and Hygiene, Graz, Austria.
| |
Collapse
|
160
|
Tiberti N, Castilletti C, Gobbi FG. Extracellular vesicles in arbovirus infections: from basic biology to potential clinical applications. Front Cell Infect Microbiol 2025; 15:1558520. [PMID: 40357393 PMCID: PMC12066795 DOI: 10.3389/fcimb.2025.1558520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Arthropod-borne viruses, or arboviruses, are currently considered a global health threat responsible for potentially severe human diseases. The increased population density, changes in land use and climate change are some of the factors that are contributing to the spread of these infections over the last years. The pathogenesis of these diseases and the mechanisms of interaction with the host, especially those leading to the development of severe forms, are yet to be fully understood. In recent years extracellular vesicles (EVs) have emerged as important players in the inter-cellular and host-pathogen interaction arising a lot of interest also in the field of vector-borne viruses. In this context, EVs seem to play a dual role, by either promoting, thus facilitating, or preventing infection. Many studies are showing how viruses can hijack the vesiculation machinery to escape the host immune response and exploit EVs to sustain their replication and propagation, even though EVs shed by immune cells seem essential to promote antiviral responses. In this manuscript we reviewed the current knowledge regarding the association between EVs and vector-borne viruses, paying particular attention to their possible role in disease transmission and dissemination, as well as to their potential as novel tools for clinical applications, spanning from biomarkers of clinical utility to novel therapeutic options.
Collapse
Affiliation(s)
- Natalia Tiberti
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Concetta Castilletti
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
| | - Federico Giovanni Gobbi
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar di Valpolicella, Verona, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
161
|
Chen C, Zhong W, Zheng H, Zhao W, Wang Y, Shen B. Current state of heart failure treatment: are mesenchymal stem cells and their exosomes a future therapy? Front Cardiovasc Med 2025; 12:1518036. [PMID: 40357434 PMCID: PMC12066684 DOI: 10.3389/fcvm.2025.1518036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Heart failure (HF) represents the terminal stage of cardiovascular disease and remains a leading cause of mortality. Epidemiological studies indicate a high prevalence and mortality rate of HF globally. Current treatment options primarily include pharmacological and non-pharmacological approaches. With the development of mesenchymal stem cell (MSC) transplantation technology, increasing research has shown that stem cell therapy and exosomes derived from these cells hold promise for repairing damaged myocardium and improving cardiac function, becoming a hot topic in clinical treatment for HF. However, this approach also presents certain limitations. This review summarizes the mechanisms of HF, current treatment strategies, and the latest progress in the application of MSCs and their exosomes in HF therapy.
Collapse
Affiliation(s)
- Chengqian Chen
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Wentao Zhong
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Hao Zheng
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, China
| | - Wei Zhao
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Yushi Wang
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| | - Botao Shen
- Department of Cardiology Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
162
|
Dipalo LL, Mikkelsen JG, Gijsbers R, Carlon MS. Trojan Horse-Like Vehicles for CRISPR-Cas Delivery: Engineering Extracellular Vesicles and Virus-Like Particles for Precision Gene Editing in Cystic Fibrosis. Hum Gene Ther 2025. [PMID: 40295092 DOI: 10.1089/hum.2024.258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
The advent of genome editing has kindled the hope to cure previously uncurable, life-threatening genetic diseases. However, whether this promise can be ultimately fulfilled depends on how efficiently gene editing agents can be delivered to therapeutically relevant cells. Over time, viruses have evolved into sophisticated, versatile, and biocompatible nanomachines that can be engineered to shuttle payloads to specific cell types. Despite the advances in safety and selectivity, the long-term expression of gene editing agents sustained by viral vectors remains a cause for concern. Cell-derived vesicles (CDVs) are gaining traction as elegant alternatives. CDVs encompass extracellular vesicles (EVs), a diverse set of intrinsically biocompatible and low-immunogenic membranous nanoparticles, and virus-like particles (VLPs), bioparticles with virus-like scaffold and envelope structures, but devoid of genetic material. Both EVs and VLPs can efficiently deliver ribonucleoprotein cargo to the target cell cytoplasm, ensuring that the editing machinery is only transiently active in the cell and thereby increasing its safety. In this review, we explore the natural diversity of CDVs and their potential as delivery vectors for the clustered regularly interspaced short palindromic repeats (CRISPR) machinery. We illustrate different strategies for the optimization of CDV cargo loading and retargeting, highlighting the versatility and tunability of these vehicles. Nonetheless, the lack of robust and standardized protocols for CDV production, purification, and quality assessment still hinders their widespread adoption to further CRISPR-based therapies as advanced "living drugs." We believe that a collective, multifaceted effort is urgently needed to address these critical issues and unlock the full potential of genome-editing technologies to yield safe, easy-to-manufacture, and pharmacologically well-defined therapies. Finally, we discuss the current clinical landscape of lung-directed gene therapies for cystic fibrosis and explore how CDVs could drive significant breakthroughs in in vivo gene editing for this disease.
Collapse
Affiliation(s)
- Laudonia Lidia Dipalo
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| | | | - Rik Gijsbers
- Department of Pharmaceutical and Pharmacological Sciences, Advanced Disease Modelling, Targeted Drug Discovery, and Gene Therapy (ADVANTAGE) labs, KU Leuven, Leuven, Belgium
- Leuven Viral Vector Core, group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Marianne S Carlon
- Department of Chronic Diseases and Metabolism, Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Leuven, Belgium
| |
Collapse
|
163
|
Zhao DZ, Wei HX, Yang YB, Yang K, Chen F, Zhang Q, Zhang T. Advances in the Research of Mesenchymal Stromal Cells in the Treatment of Maxillofacial Neurological Disorders and the Promotion of Facial Nerve Regeneration. Mol Neurobiol 2025:10.1007/s12035-025-04981-8. [PMID: 40295362 DOI: 10.1007/s12035-025-04981-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Maxillofacial neurological disorders include a range of disorders affecting the cranial nerves, which can be caused by a variety of reasons, including infection, trauma, tumor, and surgical complications, resulting in severe dysfunction, and the study of new approaches for the treatment of these disorders is crucial for the restoration of sensory and motor functions of the face. In recent years, due to the excellent tissue regenerative ability of mesenchymal stromal cells (MSCs), research on MSCs and MSC-derived exosomes has been progressively deepened, bringing many new perspectives to the therapeutic strategies for many diseases. Facial nerve regeneration is a complex process involving various pathophysiological mechanisms and therapeutic strategies to restore nerve function after injury. And the rapid development of stem cell tissue engineering has greatly facilitated the research process of facial nerve regeneration. In this paper, we review the characteristics of MSCs and neural stem cells (NSCs), the roles they play in the neural microenvironment and the mechanisms that promote nerve regeneration, summarize the research progress of MSCs in the treatment of maxillofacial neurological disorders, and highlight the promising directions for future development.
Collapse
Affiliation(s)
- De-Zhi Zhao
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Department of Prosthetics, Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Han-Xiao Wei
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yi-Bing Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kang Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Fang Chen
- Department of Prosthetics, Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
164
|
Liu S, Wang Z, Li Y, Pan Z, Huang L, Cui J, Zhang X, Yang M, Zhang Y, Li D, Sun H. Erythropoietin-Stimulated Macrophage-Derived Extracellular Vesicles in Chitosan Hydrogel Rescue BMSCs Fate by Targeting EGFR to Alleviate Inflammatory Bone Loss in Periodontitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500554. [PMID: 40289904 DOI: 10.1002/advs.202500554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/26/2025] [Indexed: 04/30/2025]
Abstract
Loss of periodontal tissue due to persistent inflammation in periodontitis is a major cause of tooth loss in adults. Overcoming osteogenic inhibition in the inflammatory periodontal environment and restoring the regenerative capacity of endogenous bone marrow mesenchymal stem cells (BMSCs) remain critical challenges in current treatment approaches. Macrophage-derived extracellular vesicles (EVs) are key regulators of osteogenesis in recipient cells, yet the role of erythropoietin (EPO) in modifying macrophages and the function of their EVs in bone regeneration remain unclear. In this study, EVs from EPO-stimulated macrophages (EPO-EVs) are isolated, and they are encapsulated in a chitosan/β-sodium glycerophosphate/gelatin (CS/β-GP/gelatin) hydrogel to create a controlled-release EVs delivery system for localized periodontal environment. EPO-EVs restore the osteogenic function of mouse BMSCs (mBMSCs) and mitigate inflammatory bone loss in a periodontitis mouse model. Mechanistically, miR-5107-5p, significantly enriched in EPO-EVs, is delivered to mBMSCs, where it suppresses epidermal growth factor receptor (EGFR) expression and alleviates EGFR's inhibitory effect on RhoA. This process counteracts osteogenic inhibition in inflammatory settings through the EGFR/RhoA axis. Overall, EVs from EPO pretreated macrophages restore the osteogenic capacity of mBMSCs under inflammation by inhibiting EGFR expression, providing new insight into therapeutic mechanisms and offering a promising approach for future periodontitis treatment.
Collapse
Affiliation(s)
- Shuchen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Zhuoran Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Yuhuan Li
- Department of Surgery, Experimental Surgery, CCM, CVK, Charité - Berlin University Medicine, Free University of Berlin and Humboldt University of Berlin, 10117, Berlin, Germany
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, 130012, China
| | - Ziyi Pan
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Jing Cui
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Xue Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Mingxi Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Yuan Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun, 130021, China
| |
Collapse
|
165
|
Luo X, Zhang X, Xu A, Yang Y, Xu W, Cai M, Xu P, Wang Z, Ying Y, Li K. Mechanistic Insights into the Anti-Glioma Effects of Exosome-Like Nanoparticles Derived from Garcinia Mangostana L.: A Metabolomics, Network Pharmacology, and Experimental Study. Int J Nanomedicine 2025; 20:5407-5427. [PMID: 40321800 PMCID: PMC12047050 DOI: 10.2147/ijn.s514930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Garcinia mangostana L. a widely used tropical fruit, has been historically valued as a medicinal plant. Modern pharmacological research has identified several compounds in its pericarp, such as alkaloids, flavonoids, and phenolic acids, which possess antioxidant and anticancer properties. Recent studies have shown that various medicinal and edible plant-derived nanovesicles (MEPNs) exhibit potent anti-tumor effects. However, the impact of Garcinia mangostana L.-derived exosome-like nanoparticles (GELNVs) in treating glioma remains insufficiently explored. Methods GELNVs were isolated from the rind of Garcinia mangostana L. using an environmentally sustainable method. The chemical composition of GELNVs was systematically characterized through both qualitative and quantitative analyses via UPLC-MS/MS. Network pharmacology was utilized to identify potential anti-glioma targets, with a focus on the PI3K-Akt signaling pathway. Subsequently, in vitro experiments were conducted to assess the uptake of GELNVs by glioma cells and their anti-tumor effects, including apoptosis induction, cell proliferation suppression, and effects on microglial polarization. Results Using a sustainable extraction method, GELNVs were successfully isolated and chemically characterized by UPLC-MS/MS. Network pharmacology and molecular docking identified key anti-glioma targets, particularly AKT1 within the PI3K-Akt signaling pathway. In vitro experiments demonstrated that GELNVs were effectively internalized by GL261 glioma and BV2 microglial cells, showing significant anti-glioma activity by inducing apoptosis and inhibiting cell proliferation. Additionally, GELNVs promoted the activation of M1-type microglia while inhibiting M2 polarization induced by IL-4 and glioma cells. This was evidenced by the upregulation of inflammatory mediators, including iNOS, TNF-α, IL-6, and IL-1β at the mRNA level. Conclusion Our findings demonstrate that GELNVs represent a potential therapeutic strategy for glioma by inducing apoptosis in tumor cells and inhibiting their proliferation through suppression of the PI3K-Akt signaling pathway. Furthermore, GELNVs show promise in modulating microglial polarization, further highlighting their potential as a glioma treatment.
Collapse
Affiliation(s)
- Xuling Luo
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xiaoting Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Aibo Xu
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| | - Yimin Yang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| | - Wei Xu
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| | - Min Cai
- Hangzhou Zhongmeihuadong Pharmaceutical Co., Ltd, Hangzhou, 310011, People’s Republic of China
| | - Peng Xu
- Department of Clinical laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310011, People’s Republic of China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| | - Youmin Ying
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Kaiqiang Li
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| |
Collapse
|
166
|
Simonassi-Paiva B, Luz JA, Ribeiro JH, da Silveira JC, de Souza CA, Pappas Jr GJ, de Carvalho JL, Lynch M, Pogue R, Rowan NJ. Identification of eccDNA in Extracellular Vesicles Derived from Human Dermal Fibroblasts Through Nanopore Sequencing. Int J Mol Sci 2025; 26:4144. [PMID: 40362382 PMCID: PMC12071958 DOI: 10.3390/ijms26094144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Extrachromosomal circular DNAs (eccDNAs) are heterogeneous circular DNA molecules derived from genomic DNA, and believed to be involved in intercellular communication and in natural biological processes. Extracellular vesicles (EVs) are membrane-bound particles released from all cells, and have been shown to contain various classes of nucleic acids. EVs can play a role in intercellular communication and may be used as biomarkers. This constitutes the first study to demonstrate that EVs derived from healthy human dermal fibroblasts carry eccDNA. eccDNA from EVs and their corresponding donor cells were isolated and sequenced on the Oxford Nanopore MinIon platform, followed by the identification of potential eccDNAs through four different bioinformatic pipelines, namely ecc_Finder, cyrcular-calling, CReSIL, and Flec. Our main findings demonstrate that EVs derived from human dermal fibroblasts carry eccDNA; there is variability in the number of eccDNAs identified in the same sample through different pipelines; and there is variability in the identified eccDNAs across biological replicates. Additionally, eccDNAs characterized in this research had (a) sequences as small as 306 base pairs and as large as 28,958 base pairs across all samples, (b) uneven chromosomal distribution, and (c) an average of 49.7% of the identified eccDNAs harboring gene fragments. Future implications for this novel research include using this framework method to elucidate factors and conditions that may influence the skin aging process and related biogenesis in human dermal cells.
Collapse
Affiliation(s)
- Bianca Simonassi-Paiva
- Faculty of Science & Health, Technological University of the Shannon, Athlone Campus, N37HD68 Athlone, Ireland; (B.S.-P.); (M.L.)
| | - Julia Alves Luz
- Department of Cell Biology, University of Brasilia, Brasilia 70910-900, DF, Brazil; (J.A.L.); (G.J.P.J.)
| | - Julia Hellena Ribeiro
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Brasilia 71966-700, DF, Brazil;
| | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (J.C.d.S.); (C.A.d.S.)
| | - Camila Azzolin de Souza
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (J.C.d.S.); (C.A.d.S.)
| | - Georgios Joannis Pappas Jr
- Department of Cell Biology, University of Brasilia, Brasilia 70910-900, DF, Brazil; (J.A.L.); (G.J.P.J.)
| | | | - Mark Lynch
- Faculty of Science & Health, Technological University of the Shannon, Athlone Campus, N37HD68 Athlone, Ireland; (B.S.-P.); (M.L.)
| | - Robert Pogue
- Genomic Sciences and Biotechnology Program, Catholic University of Brasília, Brasilia 71966-700, DF, Brazil;
| | - Neil J. Rowan
- Faculty of Science & Health, Technological University of the Shannon, Athlone Campus, N37HD68 Athlone, Ireland; (B.S.-P.); (M.L.)
| |
Collapse
|
167
|
Alexandrova M, Manchorova D, Vangelov I, Terzieva A, Dimitrova V, Mor G, Dimova T. First trimester extravillous trophoblast secretes HLA class I molecules via small extracellular vesicles. Placenta 2025; 167:11-21. [PMID: 40300266 DOI: 10.1016/j.placenta.2025.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION Human pregnancy requires acceptance and support for the semi-allogeneic embryo and effective protection of both mother and fetus. A failure to adapt, from either side, may cause abortion. The placenta-derived extracellular vesicles (EVs) have a crucial role in human implantation and pregnancy. These are lipid bilayer membrane-delimited, nano-to-micro sized extracellular microvesicles of endosomal origin, containing diverse signaling molecules, and functioning as short and long-distance messengers. We have already shown that first-trimester placenta releases the soluble HLA-C and HLA-G KIR ligands to modulate maternal cytotoxicity via the KIR/HLA axis. This study is to find whether extravillous trophoblast (EVT) secretes these HLA class I molecules via small EVs. METHODS sEVs were isolated by ultrafiltration or precipitation from serum-free conditioned media from primary trophoblast-derived EVT, and non-tumor EVT-like model Sw71 cell line, cultured as monolayer and spheroids. sEVs from cultured placental explants served as a positive control. Combined data from several methods was used for their characterization including BCA, DLS, TEM, IEM, Dot blot, and FACS. RESULTS Primary trophoblast-derived EVT and Sw71 EVT-like cells produced intact and well-visible CD63+, HLA-G- and HLA-C-bearing sEVs, regardless of culture mode and type of isolation. Both methods yielded sEVs sized 30-100 nm. DISCUSSION We show original data on the HLA-C secretion via sEVs by early pregnancy EVT and confirm the production of HLA-G-positive sEVs. A new asset to the usefulness of the Sw71 spheroid model as an implanting blastocyst surrogate is added as a tool to elucidate the sEV-based signalization in the implantation.
Collapse
Affiliation(s)
- Marina Alexandrova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diana Manchorova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ivaylo Vangelov
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Antonia Terzieva
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Violeta Dimitrova
- University Obstetrics and Gynecology Hospital "Maichin dom", Medical University, Sofia, Bulgaria
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Tanya Dimova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria.
| |
Collapse
|
168
|
Dwarshuis G, Kroon LL, Brandsma D, Noske DP, Best MG, Sol N. Liquid biopsies for the monitoring of gliomas and brain metastases in adults. Acta Neuropathol 2025; 149:37. [PMID: 40285800 PMCID: PMC12033197 DOI: 10.1007/s00401-025-02880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025]
Abstract
Clinical evaluation and MR imaging are currently the cornerstone of brain tumor progression monitoring. However, this is complicated by the occurrence of treatment effects such as pseudoprogression and radionecrosis. While essential for patient management, the distinction from true progression remains a significant challenge. Moreover, MR imaging provides limited real-time insights into tumor heterogeneity, genetic divergence, and treatment resistance. Although surgical histopathological biopsies can yield additional valuable information, they are not always conclusive, invasive, and therefore, not suitable for longitudinal measurements. In the era of precision medicine, there is a critical need for minimally invasive, accurate, and cost-effective monitoring methods for both primary brain tumors and brain metastases. Liquid biopsies have emerged as a potential candidate. Various analytes, including circulating nucleic acids, extracellular vesicles, platelet RNAs, and circulating tumor cells, can be obtained from whole blood and its derivatives, as well as other body fluids such as cerebrospinal fluid. In this narrative review, we outline the potential of liquid biopsies for the management of gliomas and brain metastases in adults and emphasize their utility in monitoring disease progression and treatment response. We discuss the most studied biofluids and analytes, along with their respective advantages and downsides. Furthermore, we address key considerations for future research and biobanking to pave the way for clinical implementation.
Collapse
Affiliation(s)
- Govert Dwarshuis
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Lente L Kroon
- Department of Neurology, Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - Dieta Brandsma
- Department of Neurology, Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Amsterdam, The Netherlands
| | - David P Noske
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Myron G Best
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nik Sol
- Department of Neurology, Netherlands Cancer Institute-Antoni Van Leeuwenhoek, Amsterdam, The Netherlands.
| |
Collapse
|
169
|
Yao Y, Yin Y, Shuai F, Lam W, Zhou T, Xie Y, He X, Han X. M2 Macrophage-Derived Extracellular Vesicles Reprogram Immature Neutrophils into Anxa1 hi Neutrophils to Enhance Inflamed Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416159. [PMID: 40277454 DOI: 10.1002/advs.202416159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/22/2025] [Indexed: 04/26/2025]
Abstract
Periodontitis is a microbiome-related inflammation that can lead to irreversible bone reduction and even tooth loss. This study reveals that macrophage polarization states significantly influence periodontal homeostasis, with M2 macrophage-derived extracellular vesicles (M2-EVs) playing a pivotal role in mitigating periodontitis-induced bone loss. Single-cell RNA sequencing of periodontal tissues treated with M2-EVs uncovered a unique Anxa1hi neutrophil subpopulation exhibiting pro-reparative properties. This subpopulation is characterized by immaturity and demonstrated osteogenic and angiogenic capabilities in vivo, partially mediated through the secretion of oncostatin M (OSM) signals. The findings suggest that this functional heterogeneity arises from M2-EVs disrupting the neutrophil maturation trajectory, with pivotal reprogramming genes, such as Acvrl1 and Fpr2, driving the differentiation of the Anxa1hi reparative subpopulation. This work underscores the potential of targeting M2 macrophage-neutrophil interactions to promote the regeneration of inflamed bone tissues.
Collapse
Affiliation(s)
- Yufei Yao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yijia Yin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fangyuan Shuai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Waishan Lam
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tao Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yaxin Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xuesong He
- The ADA Forsyth Institute, 100 Chestnut Street, Somerville, MA, 02143, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, 02115, USA
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
170
|
Tiszbein K, Koss-Mikołajczyk I, Martysiak-Żurowska D. Unlocking the Secrets of Human Milk: Isolation and Characterization of Extracellular Vesicles. Adv Nutr 2025; 16:100430. [PMID: 40288493 DOI: 10.1016/j.advnut.2025.100430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Extracellular vesicles from human milk (HMEVs) are crucial for neonatal development, immune modulation, and protection against pathogens. However, the lack of standardized isolation and characterization protocols poses significant challenges. This review aims to evaluate and compare various methods for the isolation and characterization of HMEVs, highlighting their effectiveness and potential applications. Preliminary purification steps, including the removal of cells, fat globules, and casein micelles, enhance the purity of isolated HMEVs. We categorized isolation methods into density-based, size-based, and affinity-based techniques. Density-based methods include differential and density gradient ultracentrifugation. Size-based methods encompass polymer precipitation, membrane filtration, electrophoretic filtration, size exclusion chromatography, and microfluidics. Affinity-based methods involve immunoisolation using antibodies specific to HMEV surface proteins. Characterization techniques discussed include flow cytometry, dynamic light scattering, nanoparticle tracking analysis, tunable resistive pulse sensing, electron microscopy, atomic force microscopy, confocal microscopy, western blotting, ELISA, and lateral flow immunoassay systems. Differential ultracentrifugation, considered the "gold standard," provides high purity but is time-consuming. Density gradient ultracentrifugation offers precise separation. Size-based methods like polyethylene glycol precipitation and membrane filtration are simple and fast. Electrophoretic filtration and microfluidics provide precise control of sample flow. Affinity-based methods are highly specific but costly. Advanced characterization techniques provide comprehensive insights into HMEV properties and functions. Standardizing isolation protocols and employing advanced characterization techniques are essential for advancing HMEV research. Future studies should focus on understanding the molecular mechanisms of HMEVs, exploring the impact of maternal health, and developing targeted delivery technologies. These efforts will enhance the therapeutic potential of HMEVs in neonatal care and contribute to personalized nutritional interventions.
Collapse
|
171
|
Lorico A, Santos MF, Karbanová J, Corbeil D. Extracellular membrane particles en route to the nucleus - exploring the VOR complex. Biochem Soc Trans 2025:BST20253005. [PMID: 40366329 DOI: 10.1042/bst20253005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
Intercellular communication is an essential hallmark of multicellular organisms for their development and adult tissue homeostasis. Over the past two decades, attention has been focused on communication mechanisms based on various membrane structures, as illustrated by the burst of scientific literature in the field of extracellular vesicles (EVs). These lipid bilayer-bound nano- or microparticles, as vehicle-like devices, act as regulators in various biological and physiological processes. When EVs are internalized by recipient cells, their membrane and cytoplasmic cargoes can interfere with cellular activities, affecting pathways that regulate cell proliferation, differentiation, and migration. In cancer, EVs can transfer oncogenic factors, stimulate neo-angiogenesis and immunosuppression, reprogram stromal cells, and confer drug resistance traits, thereby remodeling the surrounding microenvironment. Although the mechanisms underlying EV biogenesis and uptake are now better understood, little is known about the spatiotemporal mechanism(s) of their actions after internalization. In this respect, we have shown that a fraction of endocytosed EVs reaches the nuclear compartment via the VOR (VAP-A-ORP3-Rab7) complex-mediated docking of late endosomes to the outer nuclear membrane in the nucleoplasmic reticulum, positioning and facilitating the transfer of EV cargoes into the nucleoplasm via nuclear pores. Here, we highlight the EV heterogeneity, the cellular pathways governing EV release and uptake by donor and recipient cells, respectively, and focus on a novel intracellular pathway leading to the nuclear transfer of EV cargoes. We will discuss how to intercept it, which could open up new avenues for clinical applications in which EVs and other small extracellular particles (e.g., retroviruses) are implicated.
Collapse
Affiliation(s)
- Aurelio Lorico
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV 89014, U.S.A
| | - Mark F Santos
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV 89014, U.S.A
| | - Jana Karbanová
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Saxony, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Dresden, Saxony, Germany
| | - Denis Corbeil
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Saxony, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Dresden, Saxony, Germany
| |
Collapse
|
172
|
Li C, Wen Y, Wang J, Li L, He Y, Cheng Y, Chen J, Huang J, Ouyang C, Liu Y, Zhou R, Chen H, Li F, Guo Q, Chen Y, Chen C, Zhang Q. Human Mesenchymal Stem Cell-Derived Exosomes as Engineering Vehicles of Daunorubicin for Targeted c-Mpl+ AML Therapy. Int J Nanomedicine 2025; 20:5267-5289. [PMID: 40297403 PMCID: PMC12036622 DOI: 10.2147/ijn.s511713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Background Acute myeloid leukemia (AML) is a highly heterogeneous disease with poor therapeutic outcomes and overall prognosis, particularly in c-Mpl+ AML. c-Mpl, a proto-oncogene, is expressed at significantly higher levels in AML compared to normal human tissue cells. This study aimed to develop a type of targeted exosomes (Exos) capable of delivering anticancer drugs directly to c-Mpl+ AML cells. Methods Human umbilical cord mesenchymal stem cells (hUCMSCs) were isolated as the source of Exos. Fusion CD63 proteins with varying numbers of thrombopoietin (TPO)-mimic peptides, designed to target c-Mpl, were bioengineered to be expressed on the membranes of hUCMSCs and their derived Exos. The targeting capability of the fusion proteins was assessed using the DUAL membrane system, fluorescence resonance energy transfer efficiency, and endocytosis assays. After encapsulating the anticancer drug daunorubicin (DNR), these targeted Exos were evaluated for their ability to eliminate c-Mpl+ AML cells. Safety and efficacy were further tested in a mouse AML model. Results Our findings showed that the engineered hUCMSCs-derived Exos demonstrated excellent targeting ability to c-Mpl and a strong propensity for endocytic uptake by c-Mpl+ AML cells. Among the engineered Exos, those with the fusion protein containing three TPO-mimic peptides (CD63-mTPO3), named as m3Exos, exhibited the highest binding affinity for c-Mpl. When loaded with DNR, these engineered Exos (m3Exos@DNR) effectively eliminated c-Mpl+ AML cells in both in vitro and in vivo experiments. Furthermore, safety assessments revealed that therapy-related toxicities were within acceptable limits and associated with manageable side effects. Conclusion In summary, our results suggest engineered Exos as a highly effective targeted drug delivery vehicle for eliminating c-Mpl+ AML cells while maintaining a favorable safety profile. These findings also provide valuable insights for developing therapeutic strategies for AML and other tumors characterized by specific membrane protein expression.
Collapse
Affiliation(s)
- Chunmou Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Yuchen Wen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Jiasheng Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| | - Lindi Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Yue He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| | - Yucai Cheng
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Junru Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| | - Junbin Huang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Cheng Ouyang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Yong Liu
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Ruizhi Zhou
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Haisheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| | - Fei Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| | - Qiqi Guo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| | - Yun Chen
- Department of Pediatrics, Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Chun Chen
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, 518107, People’s Republic of China
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, People’s Republic of China
| |
Collapse
|
173
|
Zhang T, Nunes ADC, Lee J, Larocca D, Camussi G, Lim SK, Bascones VU, Angelini L, O'Kelly RD, Dong X, Niedernhofer LJ, Robbins PD. Identification of Senomorphic miRNAs in Embryonic Progenitor and Adult Stem Cell-Derived Extracellular Vesicles. Aging Cell 2025:e70071. [PMID: 40275616 DOI: 10.1111/acel.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Extracellular vesicles (EVs) are secreted by most cell types, transmitting crucial signaling molecules like proteins, small RNAs, and DNA. We previously demonstrated that EVs from murine and human mesenchymal stem cells (MSCs) functioned as senomorphics to suppress markers of senescence and the inflammatory senescence-associated secretory phenotype (SASP) in cell culture and in aged mice. Here we demonstrate that EVs from additional types of human adult stem cells and embryonic progenitor cells have a senomorphic activity. Based on their miRNA profiles showing prevalence in stem cell EVs versus nonstem cell EVs and the number of age-related genes targeted, we identified eight miRNAs as potential senomorphic miRNAs. Analysis of these miRNAs by transfection into etoposide-induced senescent IMR90 human fibroblasts revealed that each of the miRNAs alone regulated specific senescence and SASP markers, but none had complete senomorphic activity. Evaluation of ~300 combinations of miRNAs for senotherapeutic activity identified a senomorphic cocktail of miR-181a-5p, miR-92a-3p, miR-21-5p, and miR-186-5p that markedly reduced the expression of p16INK4a, p21Cip1, IL-1β, and IL-6 and the percentage of SA-ß-gal-positive cells. Transcriptome analysis identified multiple pathways affected by the miRNA cocktail, including cellular senescence and inhibition of PCAF and HIPK2 in the p53 signaling pathway. Finally, treatment of aged mice with liposomes containing the four miRNA cocktail suppressed markers of senescence and inflammation in multiple tissues. These studies suggest that EVs derived from stem cells suppress senescence and inflammation, at least in part, through miRNAs and that a senomorphic miRNA cocktail could be used to target senescence and inflammation to extend health span.
Collapse
Affiliation(s)
- Tianpeng Zhang
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Allancer D C Nunes
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jieun Lee
- AgeX Therapeutics, Inc., Alameda, California, USA
| | - Dana Larocca
- AgeX Therapeutics, Inc., Alameda, California, USA
| | - Giovanni Camussi
- Department of Medical Science, University of Torino, Turin, Italy
| | - Sai Kiang Lim
- Institute of Medical Biology, ASTAR, Singapore, Singapore
| | - Vicky U Bascones
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Luise Angelini
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan D O'Kelly
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xiao Dong
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul D Robbins
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
174
|
Taghdi MH, Al-Masawa ME, Muttiah B, Fauzi MB, Law JX, Zainuddin AA, Lokanathan Y. Three-Dimensional Bioprinted Gelatin-Genipin Hydrogels Enriched with hUCMSC-Derived Small Extracellular Vesicles for Regenerative Wound Dressings. Polymers (Basel) 2025; 17:1163. [PMID: 40362948 PMCID: PMC12073717 DOI: 10.3390/polym17091163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) have shown great promise in promoting tissue repair, including skin wound healing, but challenges like rapid degradation and short retention have limited their clinical application. Hydrogels have emerged as effective carriers for sustained EV release. Three-dimensional printing enables the development of personalized skin substitutes tailored to the wound size and shape. This study aimed to develop 3D bioprinted gelatin-genipin hydrogels incorporating human umbilical cord MSC-sEVs (hUCMSC-sEVs) for future skin wound healing applications. Gelatin hydrogels (8% and 10% w/v) were crosslinked with 0.3% genipin (GECL) to improve stability. The hydrogels were evaluated for their suitability for extrusion-based 3D bioprinting and physicochemical properties, such as the swelling ratio, hydrophilicity, enzymatic degradation, and water vapor transmission rate (WVTR). Chemical characterization was performed using EDX, XRD, and FTIR. The hUCMSC-sEVs were isolated via centrifugation and tangential flow filtration (TFF) and characterized. The crosslinked hydrogels were successfully 3D bioprinted and demonstrated superior properties, including high hydrophilicity, a swelling ratio of ~500%, slower degradation, and optimal WVTR. hUCMSC-sEVs, ranging from 50 to 200 nm, were positive for surface and cytosolic markers. Adding 75 μg/mL of hUCMSC-EVs into 10% GECL hydrogels significantly improved the biocompatibility. These hydrogels offer ideal properties for 3D bioprinting and wound healing, demonstrating their potential as biomaterial scaffolds for skin tissue regeneration applications.
Collapse
Affiliation(s)
- Manal Hussein Taghdi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
- Department of Anesthesia and Intensive Care, Faculty of Medical Technology, University of Tripoli, Tripoli P.O. Box 13932, Libya
| | - Maimonah Eissa Al-Masawa
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
| | - Barathan Muttiah
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
| | - Ani Amelia Zainuddin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
175
|
Zhdanova DY, Bobkova NV, Chaplygina AV, Svirshchevskaya EV, Poltavtseva RA, Vodennikova AA, Chernyshev VS, Sukhikh GT. Effect of Small Extracellular Vesicles Produced by Mesenchymal Stem Cells on 5xFAD Mice Hippocampal Cultures. Int J Mol Sci 2025; 26:4026. [PMID: 40362265 PMCID: PMC12071690 DOI: 10.3390/ijms26094026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is one of the most common progressive neurodegenerative diseases leading to impairments in memory, orientation, and behavior. However, significant work is still needed to fully understand the progression of such disease and develop novel therapeutic agents for AD prevention and treatment. Small extracellular vesicles (sEVs) have received attention in recent years due to their potential therapeutic effects on AD. The aim of this study was to determine the potential effect of sEVs in an in vitro model of AD. sEVs were isolated from human Wharton's jelly mesenchymal stem cells (MSCs) by asymmetric depth filtration, a method developed recently by us. AD was modeled in vitro using cells obtained from the hippocampi of newborn 5xFAD transgenic mice carrying mutations involved in familial AD. After isolation, sEVs underwent detailed characterization that included scanning electron microscopy, nanoparticle tracking analysis, confocal microscopy, Western blotting, and Luminex assay. When added to 5xFAD hippocampal cells, sEVs were nontoxic, colocalized with neurons and astrocytes, decreased the level of Aβ peptide, and increased the synaptic density. These results support the possibility that sEVs can improve brain cell function during aging, decrease the risk of AD, and potentially be used for AD therapeutics.
Collapse
Affiliation(s)
- Daria Y. Zhdanova
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Natalia V. Bobkova
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Alina V. Chaplygina
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Elena V. Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ulitsa M0iklukho-Maklaya 16/10, 117997 Moscow, Russia;
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| | - Rimma A. Poltavtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| | - Anastasia A. Vodennikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ulitsa M0iklukho-Maklaya 16/10, 117997 Moscow, Russia;
- Institute of Bioorganic Chemistry, National Research Nuclear University “MEPhI”, Kashirskoe Shosse 31, 115409 Moscow, Russia
| | - Vasiliy S. Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, 121205 Moscow, Russia
| | - Gennadiy T. Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| |
Collapse
|
176
|
Abyadeh M, Kaya A. Multiomics from Alzheimer's Brains and Mesenchymal Stem Cell-Derived Extracellular Vesicles Identifies Therapeutic Potential of Specific Subpopulations to Target Mitochondrial Proteostasis. J Cent Nerv Syst Dis 2025; 17:11795735251336302. [PMID: 40297324 PMCID: PMC12035200 DOI: 10.1177/11795735251336302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Background Alzheimer's disease (AD) is characterized by complex molecular alterations that complicate its pathogenesis and contribute to the lack of effective treatments. Mesenchymal stem cell-derived extracellular vesicles (EVs) have shown promise in AD models, but results across different EV subpopulations remain inconsistent. Objectives This study investigates proteomic and transcriptomic data from publicly available postmortem AD brain datasets to identify molecular changes at both the gene and protein levels. These findings are then compared with the proteomes of various EV subpopulations, differing in size and distribution, to determine the most promising subtype for compensating molecular degeneration in AD. Design We conducted a comprehensive analysis of 788 brain samples, including 481 AD cases and 307 healthy controls, examining protein and mRNA levels to uncover AD-associated molecular changes. These findings were then compared with the proteomes of different EV subpopulations to identify potential therapeutic candidates. Methods A multi-omics approach was employed, integrating proteomic and transcriptomic data analysis, miRNA and transcription factor profiling, protein-protein network construction, hub gene identification, and enrichment analyses. This approach aimed to explore molecular changes in AD brains and pinpoint the most relevant EV subpopulations for therapeutic intervention. Results We identified common alterations in the cAMP signaling pathway and coagulation cascade at both the protein and mRNA levels. Distinct changes in energy metabolism were observed at the protein level but not at the mRNA level. A specific EV subtype, characterized by a broader size distribution obtained through high-speed centrifugation, was identified as capable of compensating for dysregulated mitochondrial proteostasis in AD brains. Network biology analyses further highlighted potential regulators of key therapeutic proteins within this EV subtype. Conclusion This study underscores the critical role of proteomic alterations in AD and identifies a promising EV subpopulation, enriched with proteins targeting mitochondrial proteostasis, as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Alaattin Kaya
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
177
|
Guo MK, Scipione CA, Breda LCD, Prajapati K, Raju S, Botts SR, Abdul-Samad M, Patel S, Yu G, Dudley AC, Fish JE, Howe KL. Tracking Endothelial Extracellular Vesicles in a Mouse Model of Atherosclerosis. Circ Res 2025. [PMID: 40265255 DOI: 10.1161/circresaha.124.326024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Affiliation(s)
- Mandy Kunze Guo
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
| | - Corey A Scipione
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Leandro C D Breda
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Kamalben Prajapati
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
| | - Sneha Raju
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
- Division of Vascular Surgery, Department of Surgery, University of Toronto, ON, Canada. (S.R., K.L.H.)
| | - Steven R Botts
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
| | - Majed Abdul-Samad
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
| | - Sarvatit Patel
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Garry Yu
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Andrew C Dudley
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville (A.C.D.)
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
- Peter Munk Cardiac Centre, Toronto General Hospital, ON, Canada (J.E.F., K.L.H.)
| | - Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
- Division of Vascular Surgery, Department of Surgery, University of Toronto, ON, Canada. (S.R., K.L.H.)
- Peter Munk Cardiac Centre, Toronto General Hospital, ON, Canada (J.E.F., K.L.H.)
| |
Collapse
|
178
|
Ielo C, Breccia M. Extracellular vesicles as source of biomarkers in hematological malignancies: looking towards clinical applications. Expert Rev Mol Diagn 2025:1-12. [PMID: 40178353 DOI: 10.1080/14737159.2025.2488919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION Extracellular vesicles are membranous particles released by cells in physiological and pathological conditions. Their cargo is heterogeneous since it includes different biomolecules such as nucleic acids and proteins. Oncogenic alterations affect the composition of extracellular vesicles and model their content during cancer evolution. AREAS COVERED This review provides an overview of the studies focused on extracellular vesicles as source of biomarkers in hematological malignancies. A special insight into extracellular vesicles-derived biomarkers as tools for evaluating the prognosis of hematological malignancies and their response to treatment is given. EXPERT OPINION Extracellular vesicles are a valuable source of biomarkers in hematological malignancies. However, the translation from the bench to the bedside is challenged by the lack of standardization of the preanalytical variables of the experimental workflow. The release of standard operating procedures and the validation of the extracellular vesicles-derived biomarkers in large cohort of patients will help in exploiting the potential of extracellular vesicles in the clinical setting.
Collapse
Affiliation(s)
- Claudia Ielo
- Department of Translational and Precision Medicine, Sapienza University of Rome - Azienda Policlinico Umberto I, Rome, Italy
| | - Massimo Breccia
- Department of Translational and Precision Medicine, Sapienza University of Rome - Azienda Policlinico Umberto I, Rome, Italy
| |
Collapse
|
179
|
Gilboa T, Ter-Ovanesyan D, Babila CM, Whiteman S, Morton S, Kalish D, Johnston J, Tesin D, Davies M, Tam JM, Church GM, Walt DR. High-Throughput Extracellular Vesicle Isolation Using Plate-Based Size Exclusion Chromatography and Automation. J Am Chem Soc 2025; 147:13258-13263. [PMID: 40198271 DOI: 10.1021/jacs.4c17948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Extracellular vesicles (EVs) are natural, cell-derived nanoparticles released into biofluids, such as plasma, and hold great potential as a new class of biomarkers. However, the utility of analyzing EVs in clinical samples has been hampered by a lack of suitable EV isolation methods that can be performed reproducibly in a scalable manner. The current method of choice for isolating EVs, size exclusion chromatography (SEC), is performed manually one column at a time, and thus does not have the throughput for isolating EVs from clinical samples. In this work, we adapt SEC to a plate-based format to increase its throughput. We show that SEC can be performed using plates containing frits packed with resin, where each well of a 24-well plate can be used for a different sample. By measuring EV markers CD63 and CD81, as well as Albumin as a representative free protein, we optimize the separation of EVs from free proteins in the 24-well format. We also demonstrate that performing SEC in these plates can be automated using liquid handling platforms with the use of custom adapters. We quantify the high reproducibility of this automated platform and then apply the platform to analyze the tetraspanins CD63 and CD81 across individuals. Our work represents a solution to the long-standing challenge in the EV biomarker field of reproducible high-throughput EV isolation from plasma and other biofluids. We envision that the automated methods we have developed will scale SEC to hundreds of samples per day, enabling the use of EVs for biomarker discovery and diagnostics.
Collapse
Affiliation(s)
- Tal Gilboa
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
| | - Dmitry Ter-Ovanesyan
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - Clarissa May Babila
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - Sara Whiteman
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - Shad Morton
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - David Kalish
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | | | | | | | - Jenny M Tam
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| | - David R Walt
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
180
|
Padinharayil H, Varghese J, Varghese PR, Wilson CM, George A. Small Extracellular Vesicle (sEV) Uptake from Lung Adenocarcinoma and Squamous Cell Carcinoma Alters T-Cell Cytokine Expression and Modulates Protein Profiles in sEV Biogenesis. Proteomes 2025; 13:15. [PMID: 40407494 PMCID: PMC12101295 DOI: 10.3390/proteomes13020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Despite advances in immunotherapy, non-small-cell lung carcinoma (NSCLC)'s clinical success is limited, possibly due to substantial immunological alterations in advanced cancer patients. This study examines the immunomodulatory effects of sEVs derived from lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC) on T cells. METHODS SEVs were isolated from lung cancer cell lines and Jurkat-E6.1. SEV size and morphology were analyzed by NTA and TEM, respectively, while Western blotting confirmed sEV markers. SEV uptake was assessed, followed by resazurin assay, RNA isolation, quantification, cDNA preparation, RT-PCR, nano LC-MS, and bioinformatic analysis, before and after treating Jurkat-E6.1 cells with sEVs from A549 and SKMES1. RESULTS Cancer-derived sEVs were efficiently internalized by immune cells, reducing T-cell viability. The real-time PCR analysis showed downregulation of KI67, BCL2, BAX, TNFA, IL6, TGFβ, and IL10, suggesting reduced proliferation, dysregulated apoptosis, and impaired inflammatory and immunosuppressive signaling, and the upregulation of GZMB and IL2 suggests retained cytotoxic potential but possibly dysfunctional T-cell activation. Proteomic analysis revealed 39 differentially abundant proteins (DAPs) in ADC-treated T cells and 276 in SCC-treated T cells, with 19 shared DAPs. Gene Ontology (GO) analysis of these DAPs highlighted processes such as sEV biogenesis, metabolic pathways, and regulatory functions, with ADC sEVs influencing NAD metabolism, ECM binding, and oxidoreductase activity, while SCC sEVs affected mRNA stability, amino acid metabolism, and cadherin binding. The cytoplasmic colocalization suggests the presence of these proteins in the cellular and extracellular lumen, indicating the potential of further release of these proteins in the vesicles by T cells. CONCLUSION Lung cancer-derived sEVs regulate T-cell activities through immunoregulatory signaling. The molecular interactions between sEVs and immune cells can reveal novel tumor immune regulatory mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Mission Medical College & Research Institute, Thrissur 680005, Kerala, India; (H.P.); (P.R.V.)
- Department of Zoology, St. Thomas College, Kozhencherry, Pathanamthitta 689641, Kerala, India;
| | - Jinsu Varghese
- Department of Zoology, St. Thomas College, Kozhencherry, Pathanamthitta 689641, Kerala, India;
| | | | - Cornelia M. Wilson
- School of Psychology and Life Sciences, Canterbury Christ Church University, Kent CT1 1QU, UK
| | - Alex George
- Jubilee Mission Medical College & Research Institute, Thrissur 680005, Kerala, India; (H.P.); (P.R.V.)
| |
Collapse
|
181
|
Rasool GS, Shihab EM, Al-Bahrani MH, Al-Musawi MH, Malek Mohammadi Nouri K, Mehdinezhad Roshan M, Hajipour H. Enhanced endometrial receptivity via epigallocatechin gallate (EGCG)-loaded menstrual blood-derived exosomes. J Pharm Sci 2025; 114:103801. [PMID: 40280485 DOI: 10.1016/j.xphs.2025.103801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Embryo implantation failure is a significant challenge in infertility treatment, accounting for a substantial number of treatment failures. Increasing endometrial receptivity can potentially overcome this issue. This study aims to introduce a novel approach for enhancing endometrial receptivity by preparing epigallocatechin gallate (EGCG)-loaded menstrual blood-derived exosomes. Menstrual blood was used to isolate exosomes, which were then characterized for their size, zeta potential, morphology, and surface markers. EGCG was loaded into the isolated exosomes using sonication. The effects of EGCG-loaded exosomes on the adhesion ability of endometrial cells and the expression of endometrial receptivity-related genes were evaluated using in vitro implantation assays and real-time PCR, respectively. As results, exosomes with an average size of 86.2 nm, a surface charge of -11.8 mV, spherical morphology, and positive for CD9 and CD81 surface markers were successfully isolated. EGCG was loaded into exosomes with an encapsulation efficiency of 65.18 %. The in vitro implantation assay confirmed that EGCG-loaded exosomes had a greater potential to enhance the adhesion ability of endometrial cells compared to free EGCG. Furthermore, EGCG-loaded exosomes upregulated the expression of Leukemia inhibitory factor, homeobox A10, and integrin beta 3 genes more potently compared to free EGCG. These findings suggest that EGCG-loaded exosomes could be a therapeutic option for low endometrial receptivity. Moreover, menstrual blood-derived exosomes appear to be a promising drug delivery system for endometrial cells, offering a potential solution to the therapeutic limitations of the payload.
Collapse
Affiliation(s)
- Ghada S Rasool
- Department of Anatomy, Faculty of Medicine, Nineveh University, Mosul, Iraq
| | - Elaf Mahmood Shihab
- Department of Pharmacology and Toxicology Collage of Pharmacy, Al-Esraa university, Baghdad, Iraq
| | - Maha Hameed Al-Bahrani
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Mastafa H Al-Musawi
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq
| | | | - Mehdi Mehdinezhad Roshan
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hajipour
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
182
|
Pham QN, Milanova V, Tung TT, Losic D, Thierry B, Winter MA. Affinity enrichment of placental extracellular vesicles from minimally processed maternal plasma with magnetic nanowires. Analyst 2025; 150:1908-1919. [PMID: 40172922 DOI: 10.1039/d4an01414f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Affinity based enrichment of cell/tissue specific extracellular vesicles (EVs) with magnetic materials and analysis of their molecular cargo has the potential to improve assay sensitivity/specificity compared to whole plasma analysis. For example, syncytiotrophoblast EVs (STBEVs) shed from the placenta during pregnancy carry placental diagnostic markers relevant to pregnancy complications linked to placental insufficiency such as placental alkaline phosphatase (PLAP), Neprilysin (NEP) and Placental Protein 13 (PP13). However, the need for sample pre-enrichment of EVs from plasma adds significant complexity, time and cost. We report an affinity-based cell/tissue specific EV enrichment direct from plasma based on iron-oxide magnetic nanowires (NWs) coated with reversible-addition-fragmentation-chain-transfer (RAFT) polymers and conjugated with anti-PLAP antibodies. As anticipated the complex protein environment of minimally processed plasma significantly decreased STBEV enrichment yield. However, an optimized RAFT polymeric coating successfully mitigated the detrimental effect of the protein corona, yielding significantly improved STBEV recovery compared to Dynabeads™ in unenriched diluted plasma. Despite the presence of significant soluble PLAP protein, STBEV enrichment could be performed directly from the plasma of pregnant women (including preeclamptic samples) within 1.5 hours, enabling quantification of two placental protein markers PP13 and NEP with known diagnostic relevance to preeclampsia. Direct affinity-enrichment of STBEVs with high performance magnetic materials has the potential to underpin rapid clinical diagnostic assays for preeclampsia and related pregnancy complications.
Collapse
Affiliation(s)
- Quang Nghia Pham
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Valentina Milanova
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Tran Thanh Tung
- School of Chemical Engineering, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Dusan Losic
- School of Chemical Engineering, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Marnie A Winter
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| |
Collapse
|
183
|
Zhu LL, Li LD, Lin XY, Hu J, Wang C, Wang YJ, Zhou QG, Zhang J. Plasma-Derived Small Extracellular Vesicles miR- 182 - 5p Is a Potential Biomarker for Diagnosing Major Depressive Disorder. Mol Neurobiol 2025:10.1007/s12035-025-04948-9. [PMID: 40261603 DOI: 10.1007/s12035-025-04948-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Depression, particularly major depressive disorder (MDD), is a debilitating neuropsychiatric condition characterized by high disability rates, primarily driven by chronic stress and genetic predispositions. Emerging evidence highlights the critical role of microRNAs (miRNAs) in the pathogenesis of depression, with plasma-derived small extracellular vesicles (sEVs) emerging as promising biomarkers. In this study, we collected peripheral blood plasma samples from patients diagnosed with MDD, as assessed by the Hamilton Depression Rating scale, alongside healthy individuals serving as controls. Plasma-derived sEVs were isolated via ultracentrifugation, followed by high-throughput sequencing of miRNAs encapsulated within sEVs, and finally image acquisition and differential expression analysis. Our results revealed a significant elevation of miR-182-5p in plasma-derived sEVs from MDD patients compared to healthy controls, a finding further validated in chronic mild stress (CMS) models. Further analysis suggested that miRNAs encapsulated within sEVs may influence depression onset and progression by modulating hypothalamic-pituitary-adrenal (HPA) axis activity. These findings underscore the potential of miRNAs and their target genes as novel biomarkers, offering improved diagnostic accuracy and therapeutic efficacy for MDD.
Collapse
Affiliation(s)
- Lin-Lin Zhu
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
| | - Lian-Di Li
- Anhui Institute for Food and Drug Control, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Xuan-Yu Lin
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
| | - Jian Hu
- The Second Affiliated Hospital of Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Chun Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yi-Jun Wang
- The Second Affiliated Hospital of Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211167, Jiangsu Province, China.
| | - Jing Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
| |
Collapse
|
184
|
Lee J, Kwon K, Cho MJ, Son T, Roh Y, Lee S, Kim DS, Lee MS, Ban HS, Kim JS, Lim EK, Lee SH, Oh GT, Park JG, Han TS. Fusogenic Nanoreactor-Based Detection of Extracellular Vesicle-derived miRNAs for Diagnosing Atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501789. [PMID: 40254988 DOI: 10.1002/smll.202501789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Extracellular vesicle (EV) microRNAs (miRNAs) are critical liquid-biopsy biomarkers that facilitate noninvasive clinical diagnosis and disease monitoring. However, conventional methods for detecting these miRNAs require EV lysis, which is expensive, labor-intensive, and time-consuming. Inspired by natural viral infection mechanisms, a novel strategy is developed for detecting EV miRNAs in situ via vesicle fusion mediated by viral fusion proteins. A padlock probe encapsulated within fusogenic liposomes is activated by target miRNAs, thereby initiating a highly sensitive and specific rolling circle amplification (RCA) reaction. Three EV miRNAs associated with atherosclerosis are successfully analyzed using this method, thereby enabling clear differentiation of healthy and diseased mice at several disease stages. Overall, the developed platform offers a simple approach for detecting EV miRNAs and demonstrates significant potential for broad use in applications involving disease diagnosis and monitoring.
Collapse
Affiliation(s)
- Jiyoon Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Kiyoon Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Min Ji Cho
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Taesang Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Yuna Roh
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Sugi Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dae-Soo Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Moo-Seung Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Hyun Seung Ban
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jang-Seong Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Eun-Kyung Lim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jong-Gil Park
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Tae-Su Han
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
185
|
Hsu CW, Fang YC, Li JF, Cheng CA. Decoding Complex Biological Milieus: SHINER's Approach to Profiling and Functioning of Extracellular Vesicle Subpopulations. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503638. [PMID: 40255212 DOI: 10.1002/smll.202503638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Extracellular vesicles (EVs) are celebrated for their pivotal roles in cellular communication and their potential in disease diagnosis and therapeutic applications. However, their inherent heterogeneity acts as a double-edged sword, complicating the isolation of specific EV subpopulations. Conventional EV isolation methods often fall short, relying on biophysical properties, while affinity-based techniques may compromise EV integrity and utility with harsh recovery conditions. To address these limitations, the SHINER (subpopulation homogeneous isolation and nondestructive EV release) workflow is introduced, which redefines how EVs are isolated and recoverd, featuring the innovative SWITCHER (switchable extracellular vesicle releaser) tool. The SHINER workflow facilitates the precise purification and gentle recovery of target EV subpopulations from complex biological mixtures, preserving their structural integrity and biological functionality. Importantly, SHINER demonstrates exceptional adaptability to multiple markers and clinical applications. It not only enhances the ability to trace EV origins for accurate disease diagnosis but also advances fundamental EV research and provides standardized EV materials for therapeutic innovations. By improving the understanding of EVs and enabling the development of personalized diagnostics and treatments, SHINER propels EV-based science into new frontiers of advanced medicine, offering transformative potential for healthcare.
Collapse
Affiliation(s)
- Chen-Wei Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Yao-Ching Fang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Jhih-Fong Li
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Chi-An Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| |
Collapse
|
186
|
Ozerklig B, Turkel I, Yilmaz M, Vaizoglu RD, Akan HS, Dikmen ZG, Saleem A, Kosar SN. Exercise-induced extracellular vesicles mediate apoptosis in human colon cancer cells in an exercise intensity-dependent manner. Eur J Appl Physiol 2025:10.1007/s00421-025-05787-1. [PMID: 40253655 DOI: 10.1007/s00421-025-05787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Regular exercise reduces the incidence and improves the prognosis of many cancer types, but the underlying mechanisms remain elusive. Evidence suggests that exercise exerts its therapeutic effects through extracellular vesicles (EVs), which are essential for cellular communication. Here, we hypothesized that exercise-induced EVs from serum of healthy individuals would exert anti-tumorigenic effects on human colon cancer HT-29 cells, in an exercise intensity-dependent manner. Ten healthy young active males participated in a randomized crossover trial, completing two workload-matched acute exercise bouts, moderate-intensity continuous exercise (MICE) and high-intensity interval exercise (HIIE), on a cycle ergometer. A control session of rest (PRE) was included. EVs were isolated from serum samples collected during PRE and immediately after each exercise session. EVs were co-incubated with HT-29 colon cancer cells, and the effects on cell viability, migration, and apoptosis were measured. EV treatment reduced cell viability in all groups (PRE, MICE, and HIIE) by 35%, 43% and 47%, respectively, vs. PBS. HIIE-EVs showed a significantly greater reduction in cell viability vs. PRE; therefore, only these groups were used for further analysis. PRE EVs reduced migration by 27%, and HIIE-EVs by 39%. HIIE-EVs increased expression of pro-apoptotic markers: Bax/Bcl-2 ratio by 56% and Caspase 3 by 30% vs. PBS, with no change observed in the PRE group. Further, 16% of cells in PRE and 28% of cells in HIIE were TUNEL-positive, indicating DNA fragmentation. To our knowledge, this is the first human study that illustrates the therapeutic potential of exercise-induced EVs in cancer treatment.
Collapse
Affiliation(s)
- Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye.
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada.
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada.
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| | - Merve Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Refika Dilara Vaizoglu
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Handan Sevim Akan
- Department of Biology, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Z Gunnur Dikmen
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ayesha Saleem
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada
| | - Sukran Nazan Kosar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
187
|
Panzetta G, Schirizzi A, Balestra F, De Luca M, Depalo N, Rizzi F, Ricci AD, De Leonardis G, Lotesoriere C, Giannelli G, D’Alessandro R, Scavo MP. Unravelling Paclitaxel Resistance in Gastric Cancer: The Role of Small Extracellular Vesicles in Epithelial Mesenchymal Transition and Extracellular Matrix Remodelling. Cancers (Basel) 2025; 17:1360. [PMID: 40282535 PMCID: PMC12025963 DOI: 10.3390/cancers17081360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Gastric cancer (GC) is a highly aggressive disease often complicated by resistance to chemotherapy agents like paclitaxel (PTX), which targets microtubules to induce apoptosis. Resistance arises through complex molecular mechanisms, including the overexpression of pro-angiogenic factors (VEGFA, ANG-2), activation of survival pathways (PDGFRβ, PPARγ), and epithelial-mesenchymal transition (EMT) driven by proteins such as VIM, E-CAD, N-CAD, and FLOT-1. The extracellular matrix (ECM), regulated by COL1A1 and influenced by PPARγ, acts as a physical barrier to drug penetration. Small extracellular vesicles (sEVs) have emerged as critical mediators of intercellular communication and may influence these resistance pathways. Methods: This study investigated the role of sEVs isolated from metastatic GC patients treated with Ramucirumab and PTX. Patients were stratified by progression-free survival (PFS) into rapidly progressing (RP) and controlled disease (CD) groups. sEVs from these patients were applied to HCEC-1CT and HEPA-RG cell lines. Cell viability assays, gene and protein expression analyses, and bioinformatic studies were conducted to assess the impact of sEVs on resistance-related markers. Results: Results showed that sEVs from CD patients reduced the expression of markers associated with drug resistance, while sEVs from RP patients increased these markers, promoting angiogenesis, EMT, and ECM remodeling. These changes correlated with enhanced cell survival and resistance phenotypes. Bioinformatic analyses confirmed that sEVs modulate inflammation, ECM dynamics, and EMT pathways. Conclusions: In conclusion, sEVs from metastatic GC patients significantly influence chemoresistance and tumor progression. Targeting sEV-mediated signaling may offer novel therapeutic strategies to overcome resistance and improve treatment outcomes in gastric cancer.
Collapse
Affiliation(s)
- Giorgia Panzetta
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.S.); (G.D.L.)
| | - Francesco Balestra
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| | - Maria De Luca
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| | - Nicoletta Depalo
- Institute for Chemical-Physical Processes, Italian National Research Council (IPCF)-CNR SS Bari, Via Orabona, 70125 Bari, Italy; (N.D.); (F.R.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Bari Research Unit, Via Orabona 4, 70126 Bari, Italy
| | - Federica Rizzi
- Institute for Chemical-Physical Processes, Italian National Research Council (IPCF)-CNR SS Bari, Via Orabona, 70125 Bari, Italy; (N.D.); (F.R.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Bari Research Unit, Via Orabona 4, 70126 Bari, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.D.R.); (C.L.)
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.S.); (G.D.L.)
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.D.R.); (C.L.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy;
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.S.); (G.D.L.)
| | - Maria Principia Scavo
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| |
Collapse
|
188
|
Lei X, Ring S, Jin S, Singh S, Mahnke K. Extracellular Vesicles and Their Role in Skin Inflammatory Diseases: From Pathogenesis to Therapy. Int J Mol Sci 2025; 26:3827. [PMID: 40332512 PMCID: PMC12027629 DOI: 10.3390/ijms26083827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, are released into the extracellular space by almost all known cell types. They facilitate communication between cells by transferring bioactive molecules, which impact both physiological processes and the development of diseases. EVs play a crucial role in the pathogenesis of various diseases by participating in multiple pathological processes. They contribute to disease progression by triggering cytokine release, modulating immune cell activity, and inducing inflammatory and immune responses. Beyond their pathological implications, EVs also offer significant therapeutic potential. Both natural and engineered EVs show great potential in the fields of targeted therapy, drug delivery, and immune modulation in dermatological applications. The development of EV-based treatments is showing promise in advancing patient outcomes, particularly in chronic inflammatory and immune-mediated skin conditions. This review comprehensively examined the biogenesis, classification, and functional roles of EVs, including advanced methods for their isolation and characterization. Furthermore, we summarized recent studies highlighting the involvement of EVs in four major inflammatory skin diseases: psoriasis, atopic dermatitis, systemic lupus erythematosus, and wound healing.
Collapse
Affiliation(s)
| | | | | | | | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany; (X.L.); (S.R.); (S.J.); (S.S.)
| |
Collapse
|
189
|
Chen W, Zhang Y, Chen J, Dong S, Wu X, Wu Y, Du Z, Yang Y, Gong L, Yu J. Heme Oxygenase-1 Modulates Macrophage Polarization Through Endothelial Exosomal miR-184-3p and Reduces Sepsis-Induce Lung Injury. Int J Nanomedicine 2025; 20:5039-5057. [PMID: 40264818 PMCID: PMC12013636 DOI: 10.2147/ijn.s506830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Pulmonary microvascular endothelial cells (PMVECs) are notably implicated in the pathogenesis of sepsis-induced lung injury. Exosomes derived from PMVECs facilitate intercellular communication among various cell types, especially crosstalk with macrophages. Heme oxygenase-1 (HO-1), an early stress-responsive enzyme with inherent protective functions, has been implicated in acute lung injury (ALI) mitigation. But research on the mechanism of HO-1 in macrophage polarization via PMVEC exosomes in sepsis-induced lung injury is lacking. Methods To investigate the role of HO-1 in the interaction between endothelial cells and macrophages, HO-1 knockout mouse model were established. Exosomes from PMVECs were isolated, and differential expression of microRNA (miRNA) was determined by sequencing. An in vitro co-culture system involving Murine Alveolar Macrophage Cell Line (MH-S cells) and HO-1/ PMVECs-derived exosomes (HP-exos) was used to investigate the underlying mechanisms. To further verify the involvement of HO-1 in intercellular communication through exosomal miRNA in vivo, the level of pulmonary inflammation was evaluated, and the polarization of pulmonary macrophages was analyzed. Results The results showed that miR-184-3p was significantly downregulated in HP-exos, and supplementation of miR-184-3p enhanced the polarization of M1 macrophages, thus intensifying lung inflammation. HO-1 regulates the polarization of macrophages by regulating endothelial exosomes. Overexpression of HO-1 downregulates miR-184-3p, which negatively regulates Semaphorin 7A (Sema7a), which attenuated M1 type macrophages (M1) polarization and augmented M2 type macrophages (M2) polarization, thereby partially mitigating lung injury and inflammation. Conclusion Collectively, we elucidated a novel potential therapeutic mechanism that HO-1 alleviate inflammation by modulating the M1/M2 ratio in sepsis-induced ALI by regulating miR-184-3p/Sema7a expression.
Collapse
Affiliation(s)
- Wei Chen
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Yuan Zhang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| | - Jinkun Chen
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Shuan Dong
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| | - Xiaoyang Wu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Ya Wu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Zhuo Du
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Yibo Yang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Lirong Gong
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| | - Jianbo Yu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| |
Collapse
|
190
|
Kim H, Lee J, Qian A, Ji YR, Zhang R, Hu Q, Williams CK, Chuang HY, Smalley MD, Xu Y, Gao L, Mayo MC, Zhang T, Posadas EM, Tan ZS, Vinters HV, Vossel K, Magaki S, Zhu Y, Tseng HR. Noninvasive Assessment of β-Secretase Activity Through Click Chemistry-Mediated Enrichment of Neuronal Extracellular Vesicles to Detect Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415289. [PMID: 40245252 DOI: 10.1002/advs.202415289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/27/2025] [Indexed: 04/19/2025]
Abstract
Alzheimer's disease (AD), the most prevalent type of dementia, is characterized by a biological process that begins with the development of AD neuropathologic change (ADNPC) while individuals remain asymptomatic. A key molecular hallmark of ADNPC is the accumulation of amyloid-β plaques. β-secretase plays a critical role in the upstream pathological cleavage of amyloid precursor protein (APP), producing amyloid-β peptides that are prone to misfolding, ultimately contributing to plaque formation. Neuronal extracellular vesicles (NEVs) in the blood transport β-secretase and preserve its activity, allowing for noninvasive profiling of β-secretase activity for detecting early onset of ADNPC. In this study, a novel approach is approached for noninvasive assessment of β-secretase activity in AD patients using an NEV β-secretase activity assay. This assay identifies NEVs exhibiting colocalization of NEV markers with AD-associated β-secretase, generating a β-secretase activity profile for each patient. The NEV β-secretase activity assay represents a significant advancement in leveraging the diagnostic potential of NEVs, offering a noninvasive, quantitative method for reliably assessing β-secretase activity to detect the early onset of ADNPC.
Collapse
Affiliation(s)
- Hyoyong Kim
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Junseok Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Audrey Qian
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - You-Ren Ji
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Ryan Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Qixin Hu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Christopher Kazu Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Han-Yu Chuang
- Eximius Diagnostics Corp, Magnify Incubator, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Matthew D Smalley
- Eximius Diagnostics Corp, Magnify Incubator, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Yaya Xu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Liang Gao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Mary C Mayo
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ting Zhang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Edwin M Posadas
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Zaldy S Tan
- Departments of Neurology and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Keith Vossel
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| |
Collapse
|
191
|
Zhou X, Huang J, Zhang D, Qian Z, Zuo X, Sun Y. Small extracellular vesicles: the origins, current status, future prospects, and applications. Stem Cell Res Ther 2025; 16:184. [PMID: 40247402 PMCID: PMC12004682 DOI: 10.1186/s13287-025-04330-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Small extracellular vesicles (sEVs) are membrane-bound vesicles with a size of less than 200 nm, released by cells. Due to their relatively small molecular weight and ability to participate in intercellular communication, sEVs can serve not only as carriers of biomarkers for disease diagnosis but also as effective drug delivery agents. Furthermore, these vesicles are involved in regulating the onset and progression of various diseases, reflecting the physiological and functional states of cells. This paper introduces the classification of extracellular vesicles, with a focus on the extraction and identification of sEVs and their significant role in repair, diagnosis, and intercellular communication. Additionally, the paper addresses the engineering modification of sEVs to provide a reference for enhanced understanding and application.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Clinical Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Jin Huang
- Department of Geriatrics, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dianqi Zhang
- Department of Central Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Zhenyu Qian
- Department of Neurology, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Xin Zuo
- Department of Geriatrics, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China.
| | - Yaoxiang Sun
- Department of Clinical Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China.
- Department of Central Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China.
| |
Collapse
|
192
|
Shefer A, Yanshole L, Proskura K, Tutanov O, Yunusova N, Grigor’eva A, Tsentalovich Y, Tamkovich S. From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer. Diagnostics (Basel) 2025; 15:1028. [PMID: 40310419 PMCID: PMC12026271 DOI: 10.3390/diagnostics15081028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Breast cancer (BC) is the most common cancer among women worldwide; therefore, the efforts of many scientists are aimed at finding effective biomarkers for this disease. It is known that exosomes are nanosized extracellular vesicles (EVs) that are released from various cell types, including cancer cells. Exosomes are directly involved in governing the physiological and pathological processes of an organism through the horizontal transfer of functional molecules (proteins, microRNA, etc.) from producing to receiving cells. Since the diagnosis and treatment of BC have been improved substantially with exosomes, in this study, we isolated breast carcinoma cell-derived exosomes, primary endotheliocyte-derived exosomes, and blood exosomes from BC patients (BCPs) in the first stage of disease and investigated their proteomic profiles. Methods: Exosomes were isolated from the samples by ultrafiltration and ultracentrifugation, followed by mass spectrometric and bioinformatics analyses of the data. The exosomal nature of vesicles was verified using transmission electron microscopy and flow cytometry. Results: Exosome proteins secreted by MCF-7 and BT-474 cells were found to form two clusters, one of which enhanced the malignant potential of cancer cells, while the other coincided with a cluster of HUVEC-derived exosome proteins. Despite the different ensembles of proteins in exosomes from the MCF-7 and BT-474 lines, the relevant portions of these proteins are involved in similar biological pathways. Comparison analysis revealed that more BC-associated proteins were found in the exosomal fraction of blood from BCPs than in the exosomal fraction of conditioned medium from cells mimicking the corresponding cancer subtype (89% and 81% for luminal A BC and MCF-7 cells and 86% and 80% for triple-positive BC and BT-474 cells, respectively). Conclusions: Tumor-associated proteins should be sought not in exosomes secreted by cell lines but in the composition of blood exosomes from cancer patients, while the contribution of endotheliocyte exosomes to the total pool of blood exosomes can be neglected.
Collapse
Affiliation(s)
- Aleksei Shefer
- Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.)
- Institute of Medicine and Medical Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Lyudmila Yanshole
- Laboratory of Proteomics and Metabolomics, International Tomography Center, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (L.Y.); (Y.T.)
| | - Ksenia Proskura
- Department of Mammology, Novosibirsk Regional Clinical Oncological Dispensary, 630108 Novosibirsk, Russia
| | - Oleg Tutanov
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA;
| | - Natalia Yunusova
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Science, 634028 Tomsk, Russia;
| | - Alina Grigor’eva
- Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.)
| | - Yuri Tsentalovich
- Laboratory of Proteomics and Metabolomics, International Tomography Center, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (L.Y.); (Y.T.)
| | - Svetlana Tamkovich
- Institute of Medicine and Medical Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
- Institute of Oncology and Neurosurgery, E. Meshalkin National Medical Research Center of the Ministry of Health of the Russian Federation, 630090 Novosibirsk, Russia
| |
Collapse
|
193
|
Yun JH, Noh YR, Yoo S, Park S, Choi Y, An J, Kim I. Harnessing Extracellular Vesicles for Targeted Drug Delivery in Ovarian Cancer. Pharmaceutics 2025; 17:528. [PMID: 40284522 PMCID: PMC12030366 DOI: 10.3390/pharmaceutics17040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Ovarian cancer remains one of the most lethal gynecologic malignancies, primarily due to late-stage diagnosis, high recurrence rates, and the development of chemoresistance. Although targeted therapies have improved patient outcomes, their efficacy is often limited by off-target toxicity and acquired drug resistance. Extracellular vesicles (EVs), nanoscale vesicles naturally released by cells, have emerged as promising carriers for precision drug delivery. This review provides a comprehensive overview of recent advances in EV-based therapeutic strategies for ovarian cancer, including the delivery of chemotherapeutic agents, nucleic acid therapeutics, and immunomodulatory molecules. We further explore innovative engineering approaches to enhance targeting specificity, such as surface modification, cell source selection, biomaterial integration, and magnetic nanoparticle-assisted delivery. Key translational challenges in bringing EV-based therapies to clinical application are also addressed. Collectively, these insights underscore the transformative potential of EV-based platforms in advancing targeted and personalized treatment for ovarian cancer.
Collapse
Affiliation(s)
- Jang-Hyuk Yun
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yoo Rim Noh
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Soohyun Park
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Yunsup Choi
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Jiyeon An
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| |
Collapse
|
194
|
Lin M, Alimerzaloo F, Wang X, Alhalabi O, Krieg SM, Skutella T, Younsi A. Harnessing stem cell-derived exosomes: a promising cell-free approach for spinal cord injury. Stem Cell Res Ther 2025; 16:182. [PMID: 40247394 PMCID: PMC12004558 DOI: 10.1186/s13287-025-04296-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/25/2025] [Indexed: 04/19/2025] Open
Abstract
Spinal cord injury (SCI) is a severe injury to the central nervous system that often results in permanent neurological dysfunction. Current treatments have limited efficacy and face challenges in restoring neurological function after injury. Recently, stem cell-derived exosomes have gained attention as an experimental treatment for SCI due to their unique properties, including superior biocompatibility, minimal immunogenicity and non-tumorigenicity. With their potential as a cell-free therapy, exosomes promote SCI repair by enhancing nerve regeneration, reducing inflammation and stabilizing the blood-spinal cord barrier. This review summarizes advances in stem cell-derived exosome research for SCI over the past years, focusing on their mechanisms and future prospects. Despite their promising therapeutic potential, clinical translation remains challenging due to standardization of exosome isolation protocols, compositional consistency and long-term safety profiles that require further investigation.
Collapse
Affiliation(s)
- Miaoman Lin
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Farzaneh Alimerzaloo
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Xingjin Wang
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Obada Alhalabi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Sandro M Krieg
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Thomas Skutella
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
195
|
Korutla L, Hu R, Liu Y, Romano C, Habertheuer A, Abedi P, Wang H, Molugu S, Rostami S, Naji A, Nuqali A, Beasley M, Maulion C, Hahn S, Ahmad T, Wang Z, Sen S, Vallabhajosyula P. Circulating Tissue Specific Extracellular Vesicles for Noninvasive Monitoring of Acute Cellular Rejection in Clinical Heart Transplantation. Transplantation 2025:00007890-990000000-01061. [PMID: 40238644 DOI: 10.1097/tp.0000000000005369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BACKGROUND There remains a critical need for biomarkers of acute cellular rejection (ACR) in heart transplantation. We hypothesized that immunopathophysiology of ACR is reflected via dynamic changes in the protein and RNA cargoes of small extracellular vesicles (sEVs) released by cardiac allograft and T cells into circulation, thus enabling noninvasive window into ACR. METHODS T-cell sEVs were enriched using anti-CD3 antibody beads, and antidonor HLA I antibody beads for donor sEVs. Cargoes of donor sEVs (cardiac troponin T [cTnT] protein and mRNA) and T-cell sEVs (CD4, CD8, T-cell receptor proteins, miRNAs [miRs] let 7i, 101b, 21a) were compared with time-matched endomyocardial biopsy samples (n = 70) in 12 patients to postoperative day 120. RESULTS Six patients had 11 moderate ACR (15.7%) episodes, 1 had antibody-mediated rejection, and 5 had ≤ mild ACR. By Wilcoxon rank-sum tests, cTnT protein (P = 6.04 × 10-5) and mRNA (P = 6.87 × 10-7) were decreased with moderate ACR compared with grades 0/1 ACR. T-cell sEV CD4, CD8, and TCR protein cargoes (P ≤ 3.92 × 10-5) and miRs let 7i, 101b, and 21a (P ≤ 9.05 × 10-5) were increased with moderate ACR. Successful treatment of moderate ACR led to dynamic reversal in sEV profiles, especially donor heart sEV cTnT mRNA (Spearman coefficient 0.87) and miR 21a (coefficient 0.85). CONCLUSIONS Our first investigation in heart transplant patients demonstrated that circulating T cell-sEV and donor heart-sEV profiles enable diagnosis of moderate ACR with high diagnostic accuracy. A large sample cohort external validation study is warranted to better understand diagnostic potential of this platform for ACR monitoring in heart transplantation.
Collapse
Affiliation(s)
| | - Robert Hu
- Department of Surgery, Creighton University School of Medicine, Omaha, NE
| | - Yihan Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
| | - Connie Romano
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Andreas Habertheuer
- Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Parisa Abedi
- Division of Cardiac Surgery, Yale University School of Medicine, New Haven, CT
| | - He Wang
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Sudheer Molugu
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Susan Rostami
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Abdulelah Nuqali
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | - Michael Beasley
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | | | - Samuel Hahn
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | - Tariq Ahmad
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
| | - Sounok Sen
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
196
|
Senesi G, Lodrini AM, Mohammed S, Mosole S, Hjortnaes J, Veltrop RJA, Kubat B, Ceresa D, Bolis S, Raimondi A, Torre T, Malatesta P, Goumans MJ, Paneni F, Camici GG, Barile L, Balbi C, Vassalli G. miR-24-3p secreted as extracellular vesicle cargo by cardiomyocytes inhibits fibrosis in human cardiac microtissues. Cardiovasc Res 2025; 121:143-156. [PMID: 39527589 PMCID: PMC11998913 DOI: 10.1093/cvr/cvae243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
AIMS Cardiac fibrosis in response to injury leads to myocardial stiffness and heart failure. At the cellular level, fibrosis is triggered by the conversion of cardiac fibroblasts (CF) into extracellular matrix-producing myofibroblasts. miR-24-3p regulates this process in animal models. Here, we investigated whether miR-24-3p plays similar roles in human models. METHODS AND RESULTS Gain- and loss-of-function experiments were performed using human induced pluripotent stem cell-derived cardiomyocytes (hCM) and primary hCF under normoxic or ischaemia-simulating conditions. hCM-derived extracellular vesicles (EVs) were added to hCF. Similar experiments were performed using three-dimensional human cardiac microtissues and ex vivo cultured human cardiac slices. hCF transfection with miR-24-3p mimic prevented TGFβ1-mediated induction of FURIN, CCND1, and SMAD4-miR-24-3p target genes participating in TGFβ1-dependent fibrogenesis-regulating hCF-to-myofibroblast conversion. hCM secreted miR-24-3p as EV cargo. hCM-derived EVs modulated hCF activation. Ischaemia-simulating conditions induced miR-24-3p depletion in hCM-EVs and microtissues. Similarly, hypoxia down-regulated miR-24-3p in cardiac slices. Analyses of clinical samples revealed decreased miR-24-3p levels in circulating EVs in patients with acute myocardial infarction (AMI), compared with healthy subjects. Post-mortem RNAScope analysis showed miR-24-3p down-regulation in myocardium from patients with AMI, compared with patients who died from non-cardiac diseases. Berberine, a plant-derived agent with miR-24-3p-stimulatory activity, increased miR-24-3p contents in hCM-EVs, down-regulated FURIN, CCND1, and SMAD4, and inhibited fibrosis in cardiac microtissues. CONCLUSION These findings suggest that hCM may control hCF activation through miR-24-3p secreted as EV cargo. Ischaemia impairs this mechanism, favouring fibrosis.
Collapse
Affiliation(s)
- Giorgia Senesi
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Alessandra M Lodrini
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Shafeeq Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Switzerland
| | - Jesper Hjortnaes
- Department of Thoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Rogier J A Veltrop
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Bela Kubat
- Department of Pathology, Maastricht University Medical Center, The Netherlands
| | - Davide Ceresa
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Sara Bolis
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
| | - Andrea Raimondi
- Institute of Biomedical Research, IRB, Bellinzona, Switzerland
| | - Tiziano Torre
- Heart Surgery Unit, Cardiocentro Ticino Institute, EOC, Lugano, Switzerland
| | - Paolo Malatesta
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), Experimental Biology Unit, University of Genova, Genova, Italy
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Lucio Barile
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera italiana, Lugano, Switzerland
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Medicine, Baden Cantonal Hospital, Baden, Switzerland
| | - Giuseppe Vassalli
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
197
|
Du G, He J, Zhan Y, Chen L, Hu Y, Qian J, Huang H, Meng F, Shan L, Chen Z, Hu D, Zhu C, Yue M, Qi Y, Tan W. Changes and application prospects of biomolecular materials in small extracellular vesicles (sEVs) after flavivirus infection. Eur J Med Res 2025; 30:275. [PMID: 40229861 PMCID: PMC11998145 DOI: 10.1186/s40001-025-02539-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Small extracellular vesicles (sEVs), also known as exosomes, are membranous vesicles filled with various proteins and nucleic acids, serving as a communication vector between cells. Recent research has highlighted their role in viral diseases. This review synthesizes current understanding of viral sEVs and includes recent findings on sEVs infected with flaviviruses. It discusses the implications of viral sEVs research for advancing arbovirus sEVs research and anticipates the potential applications of sEVs in flavivirus infections.
Collapse
Affiliation(s)
- Gengting Du
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Junhua He
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Yan Zhan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Leru Chen
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Yue Hu
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Jiaojiao Qian
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Huan Huang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fanjin Meng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Laiyou Shan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Zhiyu Chen
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | | | - Changqiang Zhu
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Ming Yue
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Qi
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Weilong Tan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China.
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
198
|
Delshad M, Sanaei MJ, Mohammadi MH, Sadeghi A, Bashash D. Exosomal Biomarkers: A Comprehensive Overview of Diagnostic and Prognostic Applications in Malignant and Non-Malignant Disorders. Biomolecules 2025; 15:587. [PMID: 40305328 PMCID: PMC12024574 DOI: 10.3390/biom15040587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes are small extracellular vesicles, ranging from 30 to 150 nm, that are essential in cell biology, mediating intercellular communication and serving as biomarkers due to their origin from cells. Exosomes as biomarkers for diagnosing various illnesses have gained significant investigation due to the high cost and invasive nature of current diagnostic procedures. Exosomes have a clear advantage in the diagnosis of diseases because they include certain signals that are indicative of the genetic and proteomic profile of the ailment. This feature gives them the potential to be useful liquid biopsies for real-time, noninvasive monitoring, enabling early cancer identification for the creation of individualized treatment plans. According to our analysis, the trend toward utilizing exosomes as diagnostic and prognostic tools has raised since 2012. In this regard, the proportion of malignant indications is higher compared with non-malignant ones. To be precise, exosomes have been used the most in gastrointestinal, thoracic, and urogenital cancers, along with cardiovascular, diabetic, breathing, infectious, and brain disorders. To the best of our knowledge, this is the first research to examine all registered clinical trials that look at exosomes as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan 1411718541, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| |
Collapse
|
199
|
Preet R, Islam MA, Shim J, Rajendran G, Mitra A, Vishwakarma V, Kutz C, Choudhury S, Pathak H, Dai Q, Sun W, Madan R, Zhong C, Markiewicz MA, Zhang J. Gut commensal Bifidobacterium-derived extracellular vesicles modulate the therapeutic effects of anti-PD-1 in lung cancer. Nat Commun 2025; 16:3500. [PMID: 40221398 PMCID: PMC11993705 DOI: 10.1038/s41467-025-58553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although immunotherapy such as anti-programmed death-1 and its ligand 1 (PD-1/L1) is a standard treatment for advanced non-small cell lung cancer (NSCLC), many patients do not derive benefit directly. Several studies have elucidated new strategies to improve the antitumor immune response through gut microbiota modulation. However, it remains largely debatable regarding how gut microbiota remotely affect lung cancer microenvironment and subsequently modulate immunotherapy response. Here we show that commensal Bifidobacterium-derived extracellular vesicles (Bif.BEVs) can modulate the therapeutic effect of anti-PD-1 therapy in NSCLC. These Bif.BEVs are up-taken by lung cancer cells predominantly via dynamin-dependent endocytosis and upregulate PD-L1 expression through TLR4-NF-κB pathway. They also efficiently penetrate murine intestinal and patient-derived lung cancer organoids. Oral gavage of these Bif.BEVs result in their accumulation in tumors in mice. Using a syngeneic mouse model, Bif.BEVs are found to synergize the anti-tumor effect of anti-PD-1 via modulation of key cytokines, immune response and oncogenic pathways, and increase in tumor-infiltrating CD8+ T cells. Our study therefore identifies a link between Bif.BEVs and the tumor microenvironment, providing an alternative mechanism to explain how gut microbiota can influence immunotherapy response, particularly in tumors located anatomically distant from the gut.
Collapse
Affiliation(s)
- Ranjan Preet
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Md Atiqul Islam
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jiyoung Shim
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ganeshkumar Rajendran
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Amrita Mitra
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Vikalp Vishwakarma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Caleb Kutz
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, 66045, USA
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
200
|
Acevedo-Sánchez V, Rodríguez-Hernández RM, Aguilar-Ruíz SR, Torres-Aguilar H, Pina-Canseco S, Chávez-Olmos P, Garrido E, Baltiérrez-Hoyos R, Romero-Tlalolini MA. Keratinocyte-derived extracellular vesicles induce macrophage polarization toward an M1-like phenotype. Biochem Biophys Res Commun 2025; 758:151659. [PMID: 40121968 DOI: 10.1016/j.bbrc.2025.151659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Multiple reports have shown an effect of keratinocyte-derived extracellular vesicles (EVs) on keratinocytes and other cell types. However, the contribution of keratinocyte-derived EVs under physiological and pathological conditions is not fully elucidated. Therefore, whether there is an effect of EVs on macrophages in cervical cancer (CC) is also unknown. Here, we evaluated the effect of tumor and non-tumor keratinocyte-derived EVs on the polarization of peripheral blood mononuclear cells (PBMCs)-derived macrophages and THP-1 cell line. Flow cytometric evaluation of macrophages cultured in the presence of keratinocyte-derived EVs mainly indicated an increase in classical activation markers CD80 and CD86 (M1 phenotype) and little or no modification of alternative activation markers (M2 phenotype). ELISA evaluation of macrophage supernatants revealed an increase in the secretion of proinflammatory cytokines such as IL-1β and IL-6. On the other hand, TGF-β was not significantly modified and only EVs derived from non-cancerous keratinocytes induced a significant increase in IL-10. The expression levels of transcripts associated with the M1 phenotype were also evaluated by qRT-PCR with similar results to ELISA for TGF-β and IL-10; but also an increase in the expression of HLA-DRα and TNF-α was observed, and no statistically significant changes in ARG1. The ROS production was also evaluated and this increase mainly in macrophages treated with CC keratinocytes-derived EVs. So, our results suggest that the uptake of EVs derived from released by non-tumor and cervical cancer keratinocytes promotes in macrophages their polarization to an M1-like phenotype.
Collapse
Affiliation(s)
- V Acevedo-Sánchez
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - R M Rodríguez-Hernández
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - S R Aguilar-Ruíz
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - H Torres-Aguilar
- Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez de Oaxaca, Av. Universidad S/N, Cinco Señores, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - S Pina-Canseco
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - P Chávez-Olmos
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación, Gustavo A. Madero, 07360, Mexico City, Mexico.
| | - E Garrido
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación, Gustavo A. Madero, 07360, Mexico City, Mexico.
| | - R Baltiérrez-Hoyos
- Facultad de Medicina y Cirugía, CONAHCYT-Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - M A Romero-Tlalolini
- Facultad de Medicina y Cirugía, CONAHCYT-Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| |
Collapse
|