51
|
Hossain MS, Rasiah PK, Seetharaman ATM, Alvarado D, Luo M, Wohlschlegel JA, Pentecost M, Gangaraju R. TNF-Stimulated Gene-6, Part of Extracellular Vesicles in Adipose Tissue-Derived Mesenchymal Stem Cell Concentrated Conditioned Medium, Affects Microglial Activity. J Neuroimmune Pharmacol 2025; 20:60. [PMID: 40439794 PMCID: PMC12122589 DOI: 10.1007/s11481-025-10216-3] [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: 09/10/2024] [Accepted: 04/30/2025] [Indexed: 06/02/2025]
Abstract
Identifying the specific bioactive molecules produced by mesenchymal stem cells (MSCs) and the signaling pathways and cell types upon which they act is critical to developing MSC-based therapeutics for inflammatory diseases with high unmet needs. Our study aimed to investigate the impact of extracellular vesicle (EV)-derived TNF-Stimulated Gene-6 (TSG-6, from adipose tissue-derived mesenchymal stem cell concentrated conditioned medium, ASC-CCM or TSG-6 overexpression in ASC using ORF expression-ready clone) on microglia and its potential anti-inflammatory effects. EV but not non-vesicular secretome prepared by ultracentrifugation confirmed the expression of TSG-6 exclusively in the small EV (sEV) fraction. sEV ranged from 50-150 nm as determined by Zetasizer, demonstrated bilipid membrane evidenced by transmission electron microscopy, expressed positive exosomal (e.g. CD63) markers, and were endocytosed by BV2 cells confirmed by DiI fluorescently labeled exosomes. BV2 microglia cultured under serum-free conditions stimulated with TLR4 agonists (LPS and IFNγ) for 12 h in the presence of p-ASC-EV (sEV derived from ASC after cytokine stimulation) and TSG-6-ORF-EV significantly reduced nitrite release (p < 0.001), phagocytic activity (p < 0.001) and reduced CD44 expression (p < 0.05). CD44 knockdown in BV2 cells ablated TSG-6-ORF-EV mediated nitrite release, IL1β downregulation, and phagocytosis with TLR4 agonists. Our results revealed that under cytokine stimulation, the EV portion of ASC-CCM becomes enriched with TSG-6. Overexpressing TSG-6 in ASC leads to an increased concentration of TSG-6 in sEVs. This enriched EV fraction, containing TSG-6, regulates microglial dynamics through a feedback loop with CD44. EV-associated TSG-6 can influence immune cell behavior and signaling, mitigating excessive inflammation or immune dysfunction.
Collapse
Affiliation(s)
| | - Pratheepa Kumari Rasiah
- Biomedical Engineering, Vanderbilt University, Vanderbilt Biophotonics Center, Nashville, TN, USA
| | - Amritha T M Seetharaman
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Megan Luo
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Rajashekhar Gangaraju
- Department of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
52
|
Samanta A, Yoo MJ, Koh J, Lufkin SC, Lufkin T, Kraus P. Proteomic profiling of small extracellular vesicles from bovine nucleus pulposus cells. PLoS One 2025; 20:e0324179. [PMID: 40440285 PMCID: PMC12121814 DOI: 10.1371/journal.pone.0324179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/21/2025] [Indexed: 06/02/2025] Open
Abstract
Small extracellular vesicles (small EV) are a conserved means of communication across the domains of life and lately gained more interest in mammalian non-cancerous work as non-cellular, biological therapeutic with encouraging results in recent studies of chronic degenerative diseases. The nucleus pulposus (NP) is the avascular and aneural center of an intervertebral disc (IVD), home to unique niche conditions and affected in IVD degeneration. We investigated autologous and mesenchymal stem cell (MSC) small EVs for their potential to contribute to cell and tissue homeostasis in the NP niche via mass spectrometric proteome and functional enrichment analysis using adult and fetal donors. We compared these findings to published small EV databases and MSC small EV data. We propose several mechanisms associated with NP small EVs: Membrane receptor trafficking to modify signal responses promoting niche homeostasis; Redox and energy homeostasis via metabolic enzymes delivery; Cell homeostasis via proteasome delivery and immunomodulation beyond an association with a serum protein corona. The proteome signature of small EVs generated by NP parent cells is similar to previously published small EV data, yet with a focus on supplementing anaerobic metabolism and redox balance while contributing to the maintenance of an aneural and avascular microniche.
Collapse
Affiliation(s)
- Ankita Samanta
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Mi-Jeong Yoo
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Jin Koh
- The Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States of America
| | - Sina Charlotte Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| |
Collapse
|
53
|
Castro-Manrreza M, Romano LE, López-García L, Medina-Contreras O, Montesinos J. Persistent Stimulation of Human Mesenchymal Stem/Stromal Cells with TNF-α and IFN-γ Affects the Release of Large Extracellular Vesicles with Immunoregulatory Phenotype. Stem Cells Dev 2025. [PMID: 40432595 DOI: 10.1089/scd.2025.0064] [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: 05/29/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) possess immunoregulatory capacity, which is enhanced in an inflammatory environment. Participation of extracellular vesicles (EVs) in this function is proposed, as they can transport various immunoregulatory molecules. However, the impact of the inflammatory microenvironment on the load of the different types of EVs released by these cells is not fully known. Therefore, this work analyzes in detail the temporal effect of IFN-γ, alone or in combination with TNF-α (TNF-α + IFN-γ), on the cargo of immunoregulatory molecules (programmed cell death ligand 1 [PD-L1], CD73, and intercellular adhesion molecule 1 [ICAM-1]) in large extracellular vesicles (L-EVs) released by human bone marrow mesenchymal stem cells (BM-MSCs). The presence of these molecules on the surface of L-EVs was determined by flow cytometry. Our results demonstrate that exposing BM-MSCs to TNF-α + IFN-γ for 24 h increases the percentage of PD-L1+ and CD73+ L-EVs. However, if this stimulus persists, the release of L-EVs with an immunoregulatory phenotype (PD-L1+, CD73+, and PD-L1+CD73+) decreases. The impact of pro-inflammatory cytokines on the transport of ICAM-1 by L-EVs is late, since up to 72 h of treatment with IFN-γ or TNF-α + IFN-γ, the percentage of ICAM-1+ L-EVs increases. In contrast, stimulation with IFN-γ for 72 h favors the release of CD73high and ICAM-1high L-EVs, but this effect also decreases in the presence of TNF-α. Our study generates novel knowledge about the impact of the inflammatory microenvironment on the cargo composition of L-EVs released by BM-MSCs and demonstrates, for the first time, that the prolonged presence of TNF-α reduces the cargo of immunoregulatory molecules in these structures.
Collapse
Affiliation(s)
- Marta Castro-Manrreza
- Immunology and Stem Cells Laboratory, FES Zaragoza, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Leslie Erika Romano
- Immunology and Stem Cells Laboratory, FES Zaragoza, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Lucero López-García
- Immunology and Stem Cells Laboratory, FES Zaragoza, National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Mexico Children's Hospital, Mexico City, Mexico
| | - Juan Montesinos
- Mesenchymal Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center (IMSS), Mexico City, Mexico
| |
Collapse
|
54
|
Putri PHL, Alamudi SH, Dong X, Fu Y. Extracellular vesicles in age-related diseases: disease pathogenesis, intervention, and biomarker. Stem Cell Res Ther 2025; 16:263. [PMID: 40437603 PMCID: PMC12121224 DOI: 10.1186/s13287-025-04374-7] [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: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 06/01/2025] Open
Abstract
Aging is a multifactorial biological process characterized by the irreversible accumulation of molecular damage, leading to an increased risk of age-related diseases. With the global prominent rise in aging populations, elucidating the mechanisms underlying the aging process and developing strategies to combat age-related diseases have become a pressing priority. Extracellular vesicles (EVs) have gained significant attention due to their role in intercellular communication. EVs are known for their ability to deliver biocargoes, such as miRNA, proteins, and lipids, implicating their involvement in disease pathogenesis and intervention. In this review article, we explore the dual role of EVs in age-related diseases: contributing to the pathogenesis of diseases by transferring deleterious molecules, while also offering therapeutic ability by transferring beneficial molecules. We also highlight the application of EVs as biomarkers for early diagnosis of age-related diseases, paving the way for early intervention and precision medicine. Additionally, we discuss how analysing the composition of EVs cargo can provide insights into disease progression. Finally, we address the challenges and future perspectives of EV-based-therapy in clinical translation, including standardization of EVs isolation methods and improving cargo specificity.
Collapse
Affiliation(s)
- Puan Haliza Lintang Putri
- BGI Research, Hangzhou, 310030, China
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16242, Indonesia
| | - Samira Husen Alamudi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok, 16242, Indonesia
| | - Xuan Dong
- BGI Research, Hangzhou, 310030, China
| | - Ying Fu
- BGI Research, Hangzhou, 310030, China.
- Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
55
|
Izco M, Sola C, Schleef M, Schmeer M, de Toro M, Verona G, Carlos E, Reinares-Sebastian A, Colina S, Marzo-Sola ME, Garcia-Sanmartin J, Fernández-Irigoyen J, Santamaría E, Mugica-Vidal R, Blesa J, Alvarez-Erviti L. Development of human targeted extracellular vesicles loaded with shRNA minicircles to prevent parkinsonian pathology. Transl Neurodegener 2025; 14:26. [PMID: 40420149 DOI: 10.1186/s40035-025-00484-7] [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: 11/15/2024] [Accepted: 04/09/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Neurological disorders are the second leading cause of death and the leading cause of disability in the world. Thus, the development of novel disease-modifying strategies is clearly warranted. We have previously developed a therapeutic approach using mouse targeted rabies virus glycoprotein (RVG) extracellular vesicles (EVs) to deliver minicircles (MCs) expressing shRNA (shRNA-MCs) to induce long-term α-synuclein down-regulation. Although the previous therapy successfully reduced the pathology, the clinical translation was extremely unlikely since they were mouse extracellular vesicles. METHODS To overcome this limitation, we developed a source of human RVG-EVs compatible with a personalized therapy using immature dendritic cells. Human peripheral blood monocytes were differentiated in vitro into immature dendritic cells, which were transfected to express the RVG peptide. RVG-EVs containing shRNA-MCs, loaded by electroporation, were injected intravenously in the α-synuclein performed fibril (PFF) mouse model. Level of α-synuclein, phosphorylated α-synuclein aggregates, dopaminergic neurons and motor function were evaluated 90 days after the treatment. To confirm that EVs derived from patients were suitable as a vehicle, proteomic analysis of EVs derived from control, initial and advanced Parkinson's disease was performed. RESULTS The shRNA-MCs could be successfully loaded into human RVG-EVs and downregulate α-synuclein in SH-SY5Y cells. Intravenous injection of the shRNA-MC-loaded RVG-EVs induced long-term downregulation of α-synuclein mRNA expression and protein level, decreased α-synuclein aggregates, prevented dopaminergic cell death and ameliorated motor impairment in the α-synuclein PFF mouse model. Moreover, we confirmed that the EVs from PD patients are suitable as a personalized therapeutic vehicle. CONCLUSION Our study confirmed the therapeutic potential of shRNA-MCs delivered by human RVG-EVs for long-term treatment of neurodegenerative diseases. These results pave the way for clinical use of this approach.
Collapse
Affiliation(s)
- Maria Izco
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain.
| | - Carlos Sola
- Transfusion Center and Blood Bank of La Rioja, 26006, Logroño, Spain
| | | | | | - María de Toro
- Genomics and Bioinformatics Core Facility, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain
| | - Guglielmo Verona
- Centre for Amyloidosis, UCL Medical School, Rowland Hill Street, London, NW3 2PF, UK
| | - Estefania Carlos
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain
| | - Alejandro Reinares-Sebastian
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Sandra Colina
- Servicio de Neurología, Hospital San Pedro, Piqueras 98, 26006, Logroño, Spain
| | | | - Josune Garcia-Sanmartin
- Angiogenesis Group, Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario, Universitario de Navarra (HUN), 31008, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario, Universitario de Navarra (HUN), 31008, Pamplona, Spain
| | - Rodolfo Mugica-Vidal
- Department of Mechanical Engineering, University of La Rioja, 26004, Logroño, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Lydia Alvarez-Erviti
- Laboratory of Molecular Neurobiology, Center for Biomedical Research of La Rioja (CIBIR), 26006, Logroño, Spain.
| |
Collapse
|
56
|
Niu Z, Zhou H, Zheng W, Hayes OG, Hou VWQ, Görgens A, Roudi S, Zhou G, Wiklander RJ, Sych T, Sezgin E, Nordin JZ, Zhao Y, Liang X, Andaloussi SEL. Screening scaffold proteins for improved functional delivery of luminal proteins using engineered extracellular vesicles. J Control Release 2025; 384:113882. [PMID: 40425092 DOI: 10.1016/j.jconrel.2025.113882] [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] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
The potential for engineered extracellular vesicles (EVs) to efficiently deliver biotherapeutics is still largely untapped. One of the key structures in determining cargo loading and subsequent functional delivery efficiency of engineered EVs is the sorting protein (scaffold). To determine the role of scaffold protein identity, a functional screen of scaffold proteins for efficient cargo delivery is required. Here, we applied the VEDIC (VSV-G plus EV-sorting Domain-Intein-Cargo) system, previously developed by our group, for the functional screen of 55 different scaffold proteins. Three tetraspanins (TSPAN2, TSPAN4 and TSPAN9) were identified that demonstrate enhanced intracellular delivery of cargo when compared to traditionally used CD63. We further explored the in vivo and ex vivo protein delivery performance of the best performing engineered EVs (TSPAN2) using melanoma xenografts and isolated primary cells from Cre-LoxP R26-LSL-tdTomato reporter mice, respectively. Finally, we report successful treatment of LPS-induced systemic inflammation by delivering a super-repressor inhibitor of NF-ĸB using TSPAN2 engineered EVs. This work highlights the importance of screening critical EV engineering elements, such as the scaffold protein, to modulate EV properties.
Collapse
Affiliation(s)
- Zheyu Niu
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Houze Zhou
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Wenyi Zheng
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Oliver G Hayes
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Vicky W Q Hou
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - André Görgens
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Samantha Roudi
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Guannan Zhou
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rim Jawad Wiklander
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Taras Sych
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Joel Z Nordin
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden; Cancer Research Laboratory, Shandong University-Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong, PR China.
| | - Samir E L Andaloussi
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden; Evox Therapeutics Limited, Oxford, United Kingdom
| |
Collapse
|
57
|
Xu K, Wu Q, Lingyun Z, Nguyen R, Safri F, Yang W, Xu Y, Ye Y, Kwan HY, Wang Q, Liang X, Shiddiky MJA, Warkiani ME, George J, Bao J, Qiao L. Extracellular vesicles as a promising platform of precision medicine in liver cancer. Pharmacol Res 2025:107800. [PMID: 40419123 DOI: 10.1016/j.phrs.2025.107800] [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: 02/19/2025] [Revised: 05/19/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025]
Abstract
Extracellular vesicles (EVs) are natural carriers of biological information and play pivotal roles in intercellular communication. EVs are biocompatible, have low immunogenicity, and are capable of traversing biological barriers, making them ideal tools for disease diagnosis and therapy. Despite their promising prospects, the full realization of EVs potential faces several challenges. This article aims to comprehensively review the biological and molecular features of EVs, their applications in liver cancer and possible underlying mechanisms, and the critical challenges affecting the clinical translation of EVs-based therapies in liver cancer.
Collapse
Affiliation(s)
- Keyang Xu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Zhao Lingyun
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Romario Nguyen
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Fatema Safri
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - William Yang
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yikun Xu
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yun Ye
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong, China
| | - Hiu Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong, China
| | - Qiang Wang
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine Karolinska Institute, Stockholm, Sweden
| | - Muhammad J A Shiddiky
- Rural Health Research Institute (RHRI), Charles Sturt University, Orange NSW 2800, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, the University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Jianfeng Bao
- Hangzhou Xixi Hospital affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
58
|
Dobra G, Gyukity-Sebestyen E, Bukva M, Boroczky T, Nyiraty S, Bordacs B, Varkonyi T, Kocsis A, Szabo Z, Kecskemeti G, Polgar TF, Szell M, Buzas K. Proteomic profiling of serum small extracellular vesicles predicts post-COVID syndrome development. Clin Immunol 2025:110532. [PMID: 40419011 DOI: 10.1016/j.clim.2025.110532] [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: 04/03/2025] [Revised: 05/20/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
Post-COVID syndrome affects 10-35 % of COVID-19 patients, and up to 85 % of hospitalized individuals, underscoring the need for early identification of high-risk cases. We hypothesized that the proteomic profile of serum small extracellular vesicles (sEVs) obtained during acute SARS-CoV-2 infection could predict post-COVID syndrome. Serum samples from 59 patients, stratified as asymptomatic, moderate, or severe, were analyzed. sEVs were isolated, characterized by electron microscopy, nanoparticle tracking, and flow cytometry, then profiled via LC-MS. Classification models integrating comorbidities, acute symptoms, and sEV proteomics distinguished the three groups, with sEV data outperforming conventional measures. Of 620 identified proteins, 30 showed significant differences between symptomatic and asymptomatic patients, including 12 linked to complement activation. ELISA confirmed LC-MS results that serum sEVs of post-COVID patients had altered C1 inhibitor, C3, and C5 levels. These results suggest that sEV-based proteomics can enable earlier detection and more targeted follow-up for individuals at risk of post-COVID syndrome.
Collapse
Affiliation(s)
- Gabriella Dobra
- Institute of Biochemistry, Biological Research Centre HUN-REN, Szeged, Hungary
| | | | - Matyas Bukva
- Department of Immunology, University of Szeged, Szeged, Hungary
| | - Timea Boroczky
- Institute of Biochemistry, Biological Research Centre HUN-REN, Szeged, Hungary; Department of Immunology, University of Szeged, Szeged, Hungary; Doctoral School of Interdisciplinary Medicine, University of Szeged, Szeged, Hungary
| | - Szabolcs Nyiraty
- Department of Internal Medicine, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Barbara Bordacs
- Department of Internal Medicine, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamas Varkonyi
- Department of Internal Medicine, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Andrea Kocsis
- Institute of Molecular Life Sciences, Research Centre for Natural Sciences HUN-REN, Budapest, Hungary
| | - Zoltan Szabo
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Gabor Kecskemeti
- Department of Medical Chemistry, University of Szeged, Szeged, Hungary
| | - Tamas Ferenc Polgar
- Transmission Electron Microscope Laboratory, Core Facility, Biological Research Centre HUN-REN, Szeged, Hungary; Doctoral School of Theoretical Medicine, University of Szeged, Szeged, Hungary
| | - Marta Szell
- Institute of Medical Genetics, University of Szeged, Szeged, Hungary
| | - Krisztina Buzas
- Institute of Biochemistry, Biological Research Centre HUN-REN, Szeged, Hungary; Department of Immunology, University of Szeged, Szeged, Hungary.
| |
Collapse
|
59
|
Ai J, Zhang S, Dai M, Jiang P, Huang J, Xiao H, Lin Y, Tang X, Tong W, He J, Mao Q, Wang Y, Ye Z, Wang T, Chai R. Small Extracellular Vesicles Orchestrate Cisplatin-Induced Ototoxicity: Potential Biomarker and Targets Discovery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02627. [PMID: 40411396 DOI: 10.1002/advs.202502627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/12/2025] [Indexed: 05/26/2025]
Abstract
Cisplatin-induced ototoxicity remains a clinical dilemma with limited mechanistic understanding and no food and drug administration (FDA)-approved therapies. Despite emerging roles of small extracellular vesicles (sEV) in drug ototoxicity, their molecular cargo profiles and causal roles to cisplatin-induced ototoxicity are unexplored. This study systematically investigates sEV derived from cochlear explants treated with cisplatin (Cis-sEV) and controls (Ctrl-sEV) using multi-omics profiling. Through small RNA sequencing, 83 differentially expressed microRNAs (miRNAs) are identified in Cis-sEV compared to Ctrl-sEV. Notably, mmu-miR-34a-5p, mmu-miR-140-5p, mmu-miR-15b-5p, mmu-miR-25-3p, and mmu-miR-339-5p are significantly upregulation in Cis-sEVs. Predicted target pathways of these differentially expressed miRNAs are enriched in apoptosis, inflammation, and cellular damage, indicating their potential involvement in cisplatin-induced cochlear damage. LC-MS/MS analysis reveals 90 upregulated and 150 downregulated proteins in Cis-sEV, with many involved in damage response. Specifically, CLTC, CCT2, ANXA6, and HSPA8 are uniquely upregulated proteins in Cis-sEV, and CLTC and ANXA6 are exclusively co-localized in hair cells (HCs) post-cisplatin exposure, suggesting that Cis-sEV originate primarily from damaged HCs. Moreover, CLTC in sEV may serve as a potential biomarker for cisplatin-induced ototoxicity as verified in both in vitro and in vivo models. This study provides novel insights into the molecular mechanisms of cisplatin-induced ototoxicity and identifies potential biomarker and therapeutic targets.
Collapse
Affiliation(s)
- Jingru Ai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Shasha Zhang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
| | - Mingchen Dai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Pei Jiang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jingyuan Huang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Hairong Xiao
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Yanqin Lin
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Xujun Tang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Wei Tong
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Jun He
- Department of Otolaryngology - Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Qiuyue Mao
- Department of Otolaryngology - Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yintao Wang
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Zixuan Ye
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
| | - Tian Wang
- Department of Otolaryngology - Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518063, China
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, School of Medicine, The University of Electronic Science and Technology of China, Chengdu, 610072, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
- Institute for Stem Cells and Regeneration, Chinese Academy of Science, Beijing, 100081, China
| |
Collapse
|
60
|
Ziyafati Kafi F, Eslami N, Shekari F, Bazrgar M. Theranostic potential of extracellular vesicles in reproductive tracts: implications for recurrent implantation failure. Mol Biol Rep 2025; 52:502. [PMID: 40411714 DOI: 10.1007/s11033-025-10619-8] [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: 03/14/2025] [Accepted: 05/15/2025] [Indexed: 05/26/2025]
Abstract
Embryo implantation is a critical step at the beginning of pregnancy, occurring during a specific and limited period known as the "implantation window". Successful implantation involves various signaling pathways and molecular interactions. Recent studies have highlighted the importance of extracellular vesicles (EVs) in mediating these complex interactions. Different cell types release EVs to transfer signals to other cells or tissues. Additionally, emerging evidence suggests that EVs regulate signaling between the developing embryo and endometrium. In this review, we summarize current findings that highlight the role of EVs in the reproductive tract, gamete production, and their potential roles in embryo development and implantation. We then examine studies emphasizing the role of EVs in embryo-maternal interactions and implantation. Finally, we will explore the theranostic potential of EVs in various aspects of assisted reproductive technology (ART), including modulation of embryo-maternal interactions, enhancement of embryo quality, and improvement of endometrial receptivity. A more comprehensive understanding of EVs in the pathology of recurrent implantation failure could support the development of personalized treatments.
Collapse
Affiliation(s)
- Fatemeh Ziyafati Kafi
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Nasim Eslami
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masood Bazrgar
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
61
|
Caller T, Moore KJ, Lehmann LH, Wu SM, Leor J. Insights Into Heart-Tumor Interactions in Heart Failure. Circ Res 2025; 136:1262-1285. [PMID: 40403117 DOI: 10.1161/circresaha.124.325490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/10/2025] [Accepted: 02/27/2025] [Indexed: 05/24/2025]
Abstract
Heart failure (HF) often coexists with cancer. Beyond the known cardiotoxicity of some cancer treatments, HF itself has been associated with increased cancer incidence. The 2 conditions share common risk factors, mechanisms, and interactions that can worsen patient outcomes. The bidirectional relationship between HF and cancer presents a complex interplay of factors that are not fully understood. Recent preclinical evidence suggests that HF may promote tumor growth via the release of protumorigenic factors from the injured heart, revealing HF as a potentially protumorigenic condition. Our review discusses the biological crosstalk between HF and cancer, emphasizing the impact of HF on tumor growth, with inflammation, and modulating the immune system as central mechanisms. We further explore the clinical implications of this connection and propose future research directions. Understanding the mechanistic overlap and interactions between HF and cancer could lead to new biomarkers and therapies, addressing the growing prevalence of both conditions and enhancing approaches to diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Tal Caller
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| | - Kathryn J Moore
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine (K.J.M.)
| | - Lorenz H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Germany (L.H.L.)
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg/Mannheim, Germany (L.H.L.)
- German Cancer Research Center (DKFZ), Heidelberg, Germany (L.H.L.)
| | - Sean M Wu
- Stanford Cardiovascular Institute (S.M.W.), Stanford University School of Medicine, CA
- Division of Cardiovascular Medicine, Department of Medicine (S.M.W.), Stanford University School of Medicine, CA
| | - Jonathan Leor
- Neufeld and Tamman Cardiovascular Research Institutes, Faculty of Medical and Health Sciences, Tel Aviv University, Israel (T.C., J.L.)
- Lev Leviev Cardiovascular and Thoracic Center, Sheba Medical Center, Tel Hashomer, Israel (T.C., J.L.)
| |
Collapse
|
62
|
Pantel K, Alix-Panabières C, Hofman P, Stoecklein NH, Lu YJ, Lianidou E, Giacomini P, Koch C, de Jager V, Deans ZC, Fairley J, Patton SJ, Toledo RA, Schuuring E, Heitzer E, Hendrix A, Ricklefs FL, Thakur BK, von Bubnoff N, Pierga JY, Gebhardt C, Andersen CL, Fijneman R, Malats N, Hallermayr A, Chelala C, Joosse SA, Ciliberto G. Fostering the implementation of liquid biopsy in clinical practice: meeting report 2024 of the European Liquid Biopsy Society (ELBS). J Exp Clin Cancer Res 2025; 44:156. [PMID: 40410806 PMCID: PMC12100835 DOI: 10.1186/s13046-025-03398-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/21/2025] [Indexed: 05/25/2025] Open
Affiliation(s)
- Klaus Pantel
- ELBS, Hamburg, Germany.
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany.
| | - Catherine Alix-Panabières
- ELBS, Hamburg, Germany
- Laboratory of Rare Human Circulating Cells (LCCRH) and Liquid Biopsy, University Medical Centre of Montpellier, Montpellier, France
- CREEC (CREES), Unité Mixte de Recherches, IRD 224-CNRS 5290-Université de Montpellier, Montpellier, France
| | - Paul Hofman
- IHU RespirERA, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Biobank 0033-00025, Côte d'Azur University, Nice, France
| | - Nikolas H Stoecklein
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of Heinrich-Heine University Düsseldorf, Moorenstr. 5, Düsseldorf, 40225, Germany
| | - Yong-Jie Lu
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Evi Lianidou
- Department of Chemistry, Analysis of Circulating Tumor Cells Lab, Laboratory of Analytical Chemistry, University of Athens, Athens, Greece
| | - Patrizio Giacomini
- Precision Medicine Unit in Senology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli, 8, Roma, 00168, Italy
| | - Claudia Koch
- ELBS, Hamburg, Germany
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Vincent de Jager
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Zandra C Deans
- Genomics Unit, NHS England, London, UK
- GenQA, Nine, 9 Little France Road, Edinburgh BioquarterEdinburgh, EH16 4SA, UK
| | - Jennifer Fairley
- GenQA, Nine, 9 Little France Road, Edinburgh BioquarterEdinburgh, EH16 4SA, UK
| | - Simon J Patton
- EMQN CIC, Unit 4, Enterprise House, Manchester Science Park, Pencroft Way, Manchester, M15 6SE, UK
| | - Rodrigo A Toledo
- Biomarkers and Clonal Dynamics Group, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, 08035, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Institute of Health Carlos III (ISCIII), Madrid, 28029, Spain
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
- Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Medical University of Graz, Graz, Austria
| | - An Hendrix
- Department of Human Structure and Repair, Laboratory of Experimental Cancer Research,, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Franz Lennard Ricklefs
- Department of Neurosurgery, University Medical-Center Hamburg-Eppendorf, Hamburg, Germany
| | - Basant Kumar Thakur
- Department of Pediatrics III, University Hospital Essen and University Duisburg, Essen, Germany
| | - Nikolas von Bubnoff
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, 23538, Germany
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, Lübeck, 23538, Germany
| | - Jean-Yves Pierga
- Department of Medical Oncology, Université Paris Cité, Institut Curie, Paris, 75005, France
| | - Christoffer Gebhardt
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Remond Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO) and CIBERONC, Madrid, Spain
| | | | - Claude Chelala
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Simon A Joosse
- ELBS, Hamburg, Germany
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg, 20246, Germany
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
63
|
Li Y, Qian L, Liu F, Xu S, Zhou L, Wei C, Zhang Y, Zhai Y, Gu Y, Li S. Light-Controlled Small Extracellular Vesicle-Based Spherical Nucleic Acid Nanomotor for Enhanced Transdermal Delivery against Skin Aging. NANO LETTERS 2025. [PMID: 40408545 DOI: 10.1021/acs.nanolett.5c01747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Small extracellular vesicles (sEV) derived from mesenchymal stem cells hold promise for anti-skin aging, yet their clinical application is hindered by poor transdermal permeability. Herein, we report an innovative light-controlled sEV-based spherical nucleic acid nanomotor (NM-ESNA). This nanosystem was composed of an sEV core and an MMP1-targeting siRNA shell, forming a 3D penetrative nanostructure. In addition, asymmetrically modified light-responsive gas-generating molecules were integrated into the nanomotor, enabling efficient dermal delivery. The light-controlled and enhanced transdermal delivery guaranteed synergistic anti-skin aging therapy through sEV-mediated paracrine effects and gene therapy targeting MMP1 in the dermis. On the basis of this deep transdermal delivery technology and the synergistic therapy strategy, NM-ESNA demonstrated outstanding anti-skin aging effects in a mouse model. This biocompatible nanosystem (NM-ESNA) enabled light-controlled and deep transdermal delivery, establishing a therapeutic platform with significant potential for sEV-based noninvasive anti-skin aging therapy.
Collapse
Affiliation(s)
- Yu Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Lu Qian
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, No. 29 Jiangjun Avenue, Jiangning District, Nanjing, 211100, Jiangsu Province, China
| | - Fei Liu
- Zhejiang Zhebei Pharmaceutical Co., Ltd., Hangzhou, 310000, Zhejiang Province, China
| | - Shilong Xu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Lian Zhou
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Chenxi Wei
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Yanqin Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Yuewen Zhai
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Yueqing Gu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| | - Siwen Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, 211198, Jiangsu Province, China
| |
Collapse
|
64
|
Samuels M, Karakostas C, Besta S, Lauer Betrán A, Tsilingiri K, Turner C, Shirazi Nia R, Poudine N, Goodyear R, Jones W, Klinakis A, Giamas G. LMTK3 regulation of EV biogenesis and cargo sorting promotes tumour growth by reducing monocyte infiltration and driving pro-tumourigenic macrophage polarisation in breast cancer. Mol Cancer 2025; 24:149. [PMID: 40405280 PMCID: PMC12100856 DOI: 10.1186/s12943-025-02346-2] [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: 01/09/2025] [Accepted: 04/28/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Lemur Tail Kinase 3 (LMTK3) promotes cell proliferation, invasiveness and therapy resistance, and its expression correlates with poor survival in several different malignancies, including breast cancer. Crosstalk through extracellular vesicles (EVs) is an increasingly appreciated mechanism of cell communication within the tumour immune microenvironment, which contributes to different aspects of cancer progression and plays a pivotal role in shaping tumour fate. METHODS Nanoparticle tracking analysis and transmission electron microscopy were used to study the effects of LMTK3 on EV size, while single particle interferometry allowed us to examine LMTK3-dependent effects on the subpopulation distribution of EVs. Quantitative mass spectrometry was used to profile LMTK3-dependent proteomics changes in breast cancer-derived EVs. Bioinformatics analysis of clinical data along with in vitro and cell-based assays were implemented to explore the effects of LMTK3-dependent EV protein cargo on the tumour immune microenvironment. To elucidate the mechanism through which LMTK3 impacts endosomal trafficking and regulates EV biogenesis, we used a variety of approaches, including in vitro kinase assays, confocal and electron microscopy, as well as in vivo subcutaneous and orthotopic breast cancer mouse models. RESULTS Here, we report that LMTK3 increases the average size of EVs, modulates immunoregulatory EV proteomic cargo and alters the subpopulation distribution of EVs released by breast cancer cells. Mechanistically, we provide evidence that LMTK3 phosphorylates Rab7, a key regulator of multivesicular body (MVB) trafficking, thereby reducing the fusion of MVBs with lysosomes and subsequent degradation of intralumenal vesicles, resulting in altered EV release. Moreover, LMTK3 causes increased packaging of phosphoserine aminotransferase 1 (PSAT1) in EVs, leading to a paracrine upregulation of phosphoglycerate dehydrogenase (PHGDH) in monocytes when these EVs are taken up. PSAT1 and PHGDH play key roles in the serine biosynthesis pathway, which is closely linked to cancer progression and regulation of monocyte behaviour. LMTK3 EV-induced elevated PHGDH expression in monocytes reduces their infiltration into breast cancer 3D spheroids and in vivo breast cancer mouse models. Furthermore, these infiltrating monocytes preferentially differentiate into pro-tumourigenic M2-like macrophages. Additional breast cancer mouse studies highlight the contribution of LMTK3-dependent EVs in the observed immunosuppressive macrophage phenotype. Finally, in vitro experiments show that pharmacological inhibition of LMTK3 reverses the pro-tumourigenic and immunomodulatory effects mediated by EVs derived from LMTK3 overexpressing cells. CONCLUSION Overall, this study advances our knowledge on the mechanisms of EV biogenesis and highlights a novel oncogenic role of LMTK3 in the breast TME, further supporting it as a target for cancer therapy.
Collapse
Affiliation(s)
- Mark Samuels
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Christos Karakostas
- Center of Basic Research Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Simoni Besta
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Andrea Lauer Betrán
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Katerina Tsilingiri
- Center of Basic Research Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Charlotte Turner
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Reza Shirazi Nia
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Niloufar Poudine
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Richard Goodyear
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, UK
| | - William Jones
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Apostolos Klinakis
- Center of Basic Research Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Georgios Giamas
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK.
| |
Collapse
|
65
|
Nadeau A, Tsering T, Abdouh M, Kienzle L, Cleyle J, Taylor L, Douanne N, Dickinson K, Siegel PM, Burnier JV. Characterization of extracellular vesicle-associated DNA and proteins derived from organotropic metastatic breast cancer cells. J Exp Clin Cancer Res 2025; 44:157. [PMID: 40410902 PMCID: PMC12100931 DOI: 10.1186/s13046-025-03418-3] [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: 02/01/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND While primary breast cancer (BC) is often effectively managed, metastasis remains the primary cause of BC-related fatalities. Gaps remain in our understanding of the mechanisms regulating cancer cell organotropism with predilection to specific organs. Unraveling mediators of site-specific metastasis could enhance early detection and enable more tailored interventions. Liquid biopsy represents an innovative approach in cancer involving the analysis of biological materials such as circulating tumor DNA and tumor-derived extracellular vesicles (EV) found in body fluids like blood or urine. This offers valuable insights for characterizing and monitoring tumor genomes to advance personalized medicine in metastatic cancers. METHODS We performed in-depth analyses of EV cargo associated with BC metastasis using eight murine cell line models with distinct metastatic potentials and organotropism to the lung, the bone, the liver, and the brain. We characterized the secretome of these cells to identify unique biomarkers specific to metastatic sites. RESULTS Small EVs isolated from all cell lines were quantified and validated for established EV markers. Tracking analysis and electron microscopy revealed EV secretion patterns that differed according to cell line. Cell-free (cf)DNA and EV-associated DNA (EV-DNA) were detected from all cell lines with varying concentrations. We detected a TP53 mutation in both EV-DNA and cfDNA. Mass spectrometry-based proteomics analyses identified 698 EV-associated proteins, which clustered according to metastatic site. This analysis highlighted both common EV signatures and proteins involved in cancer progression and organotropism unique to metastatic cell lines. Among these, 327 significantly differentially enriched proteins were quantified with high confidence levels across BC and metastatic BC cells. We found enrichment of specific integrin receptors in metastatic cancer EVs compared to EVs secreted from non-transformed epithelial cells and matched tumorigenic non-metastatic cells. Pathway analyses revealed that EVs derived from parental cancer cells display a cell adhesion signature and are enriched with proteins involved in cancer signaling pathways. CONCLUSION Taken together, the characterization of EV cargo in a unique model of BC organotropism demonstrated that EV-DNA and EV proteomes were informative of normal and cancer states. This work could help to identify BC biomarkers associated with site-specific metastasis and new therapeutic targets.
Collapse
Affiliation(s)
- Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Mohamed Abdouh
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Laura Kienzle
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Jenna Cleyle
- Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Lorne Taylor
- Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Peter M Siegel
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
66
|
Le Lay S, Scherer PE. Exploring adipose tissue-derived extracellular vesicles in inter-organ crosstalk: Implications for metabolic regulation and adipose tissue function. Cell Rep 2025; 44:115732. [PMID: 40408250 DOI: 10.1016/j.celrep.2025.115732] [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: 10/15/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/25/2025] Open
Abstract
Intercellular and inter-organ communication systems are vital for tissue homeostasis and disease development, utilizing soluble bioactive molecules for signaling. The field of extracellular vesicle (EV) biology has rapidly expanded in recent decades, highlighting EVs as effective bioactive nanovectors for cell-to-cell communication in various physiological and pathological contexts. Numerous studies indicate that adipocyte-derived EVs are crucial components of the adipose secretome, playing a key role in autocrine and paracrine interactions within adipose tissue, as well as in endocrine signaling. This review aims to present an updated perspective on EVs as mediators of communication between adipose tissue and other organs, while also examining their therapeutic potential in the light of recent advancements in EV biology research.
Collapse
Affiliation(s)
- Soazig Le Lay
- Nantes Université, CNRS, INSERM, l'institut du thorax, 44000 Nantes, France; Université Angers, SFR ICAT, 49000 Angers, France.
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
67
|
Jin J, Nolte PA. Mitochondrial Distribution and Osteocyte Mechanosensitivity. Curr Osteoporos Rep 2025; 23:22. [PMID: 40402395 PMCID: PMC12098195 DOI: 10.1007/s11914-025-00918-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE OF REVIEW Mechanical loading of bone is an important physical stimulus for bone tissue remodeling and adaptation. It is transmitted from the extracellular matrix all the way to the osteocyte nucleus via the extracellular matrix-integrin-cytoskeleton-nucleus system. Mitochondria are integral in sensing of mechanical loads to allow the cell to adapt to its environment. This review provides a background of mitochondrial distribution in osteocytes especially during mechanical loading, discussing the importance of mitochondrial distribution in osteocyte mechanosensitivity and mechanotransduction. RECENT FINDINGS Mitochondria throughout the osteocyte are highly dynamic and provide essential metabolic and signal functions to regulate osteocyte morphology and function. They undergo the processes of fission and fusion accompanied by mitochondrial DNA distribution. The mitochondrial network structure and function in osteocytes can be regulated by mechanical loading. Interestingly, mitochondria can be transmitted by osteocytes into adjacent cells to communicate with them via tunneling nanotubes, migrasomes, and blebbisomes, causing changes in cell morphology and/or function. Mitochondrial distribution in or out osteocytes can be rearranged by physical and (bio)chemical signals via fission and fusion, as well as tunneling nanotubes, migrasomes, and blebbisomes. Mechanical loading-induced changes in mitochondria may drive signaling pathways of cell function in aging and diseases. More insights into interactions between neighbouring osteocytes and between osteocytes and other cell types would facilitate the development of new strategies to apply mitochondrial therapy for bone-related diseases.
Collapse
Affiliation(s)
- Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, Amsterdam, 1081 LA, The Netherlands
| | - Peter A Nolte
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, Amsterdam, 1081 LA, The Netherlands.
- Department of Orthopedic Surgery, Spaarne Gasthuis, Spaarnepoort 1, 2134 TM, Hoofddorp, The Netherlands.
| |
Collapse
|
68
|
Mahapatra S, Sharma Y, Kashyap S, Mohanty S. Bioinspired Silk and Human Amniotic Membrane-Based MSC-sEV-Functionalized Wound Dressing Enhances Skin Regeneration: A Cell-Free Therapeutic Modality for Wound Care. ACS Biomater Sci Eng 2025. [PMID: 40401413 DOI: 10.1021/acsbiomaterials.5c00353] [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: 05/23/2025]
Abstract
Full-thickness wounds pose significant healing challenges due to their impaired regenerative capacity, persistent inflammation, and oxidative stress. Enhancing the bioactivity of silk fibroin (SF) and the mechanical strength of the human amniotic membrane (hAM) can improve wound healing outcomes. Mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEVs) offer promising anti-inflammatory and antioxidant benefits, but their poor retention and painful application limits their clinical utility. To overcome these challenges, we developed a composite scaffold of SF and hAM (Sh), loaded with sEVs (ShE), designed to accelerate wound healing by modulating inflammation, oxidative stress, and tissue regeneration. ShE exhibited excellent physical stability, optimal swelling, degradation kinetics, hemocompatibility, and sustained sEV release. In vitro, it enhanced keratinocyte and fibroblast proliferation and migration, reduced oxidative stress, and provided immunomodulatory and pro-angiogenic effects. ShE significantly lowered ROS levels, suppressed PHA-activated PBMNC proliferation, facilitated macrophage polarization from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, and promoted angiogenesis. In vivo, ShE accelerated wound closure within 21 days, outperforming DuoDERM, a commercial dressing. Histopathological analysis demonstrated improved epidermal maturation, dermal regeneration, and reduced scarring in ShE-treated wounds, confirming the superior tissue regeneration capacity. Additionally, its fabrication from medical waste and indigenous raw materials ensures cost-effectiveness and sustainability in healthcare applications. By synergistically regulating cell physiology for skin regeneration, ShE emerges as a promising, clinically viable, and affordable wound dressing for enhanced wound care management.
Collapse
Affiliation(s)
- Shruti Mahapatra
- Stem Cell Facility-DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Yashvi Sharma
- Stem Cell Facility-DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Seema Kashyap
- Department of Ocular Pathology, Dr. R. P. Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Mohanty
- Stem Cell Facility-DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi 110029, India
| |
Collapse
|
69
|
Catalán J, Martínez-Díaz P, Parra A, Bonet S, Yeste M, Roca J, Barranco I, Miró J. Isolation and characterization of extracellular vesicle subsets in donkey seminal plasma. Theriogenology 2025; 244:117501. [PMID: 40412157 DOI: 10.1016/j.theriogenology.2025.117501] [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: 04/22/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Seminal plasma (SP), a fluid composed of secretions from the male genital tract, is rich in seminal extracellular vesicles (sEVs), nano-sized particles surrounded by a lipid bilayer membrane and loaded with functionally active molecules. Seminal EVs are secreted by functional cells of the male genital tract and play a key role in modulating reproductive processes, including sperm function and immune response in the female genital tract. The aim of this study was to isolate and characterize sEVs from donkey SP for the first time. Nine SP samples were collected from nine healthy and reproductive active donkeys. The SP samples were randomly pooled to create three pools (three SP samples per pool). The SP pools were subjected to differential centrifugation and size-exclusion chromatography to separately isolate two subsets of sEVs: small (S-) and large (L-). Orthogonal characterization of sEV samples was performed according to MISEV 2023 guidelines, including morphology (by cryogenic electron microscopy), concentration (by total protein concentration and total and CFSE positive particles by flow cytometry [FC]), particle size distribution (by dynamic light scattering), purity (by albumin assessment by FC), and specific EV protein markers (tetraspanins CD9, CD63, and CD81, and HSP70 by FC). The results showed that donkey SP is highly enriched in sEVs. Size differences were found between both sEV subsets, with S-sEVs being smaller (∼160 nm) and L-sEVs larger (∼290 nm). Both sEV subsets were positive for the four EV protein markers. However, the percentage of CD81-positive events was higher in S-sEV samples than in L-sEV samples (P < 0.05). This study is the first to isolate and characterize sEVs in donkey SP, demonstrating their heterogeneity and suggesting differences in biogenesis and function between S-sEVs and L-sEVs.
Collapse
Affiliation(s)
- Jaime Catalán
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain; Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Pablo Martínez-Díaz
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain; International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Spain; EV-lab, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
| | - Ana Parra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain; International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Spain; EV-lab, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
| | - Sergi Bonet
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain; Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain; International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Spain; EV-lab, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
| | - Isabel Barranco
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain; International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Spain; EV-lab, Faculty of Veterinary Science, University of Murcia, Murcia, Spain.
| | - Jordi Miró
- Equine Reproduction Service, Department of Animal Medicine and Surgery, Faculty of Veterinary Sciences, Autonomous University of Barcelona, Bellaterra, Cerdanyola del Vallès, Spain.
| |
Collapse
|
70
|
Wang H, Wang Y, Ling M, Wang S, Luo J, Sun J, Xi Q, Chen T, Zhang Y. Comparison of the Immunomodulatory Roles of Porcine Milk-Derived Extracellular Vesicles from Different Stages of Lactation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12247-12260. [PMID: 40327365 DOI: 10.1021/acs.jafc.4c11983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Milk-derived extracellular vesicles (MEV) have received increasing attention due to their physiological benefits for newborn growth and development. However, their physiological characteristics and immunomodulatory capacities during lactation remain unclear. In this study, we isolated MEV from porcine milk at day 1, week 1, week 2, and week 3 after parturition and compared their differences by common EV characterization, showing the higher concentration and smaller mean size of MEV from colostrum. Porcine MEV could be internalized by RAW264.7 macrophages and functionally deliver immune-related microRNAs (miRNAs). Furthermore, MEV promoted macrophage polarization toward the M2-like phenotype and exerted anti-inflammatory effects in vitro. Mice models with sepsis also displayed that MEV suppressed the secretion of pro-inflammatory cytokines in serum and drove an anti-inflammatory M2-like phenotype of the spleen, ultimately weakening immune responses. Especially, MEV from colostrum exhibited the most robust effects on immunoregulation. Overall, this study provides valuable information for MEV secretion patterns and their differences in immunomodulatory bioactivities.
Collapse
Affiliation(s)
- Hailong Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuxuan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Mingwang Ling
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shumeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
71
|
Jiang J, Wang Z, Bao Q, Chen S, Xu W, Jiang J. Extracellular Vesicles as Emerging Therapeutic Strategies in Spinal Cord Injury: Ready to Go. Biomedicines 2025; 13:1262. [PMID: 40427089 PMCID: PMC12109484 DOI: 10.3390/biomedicines13051262] [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: 04/08/2025] [Revised: 05/17/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
Spinal cord injury (SCI) is a prevalent central nervous system disorder that causes significant disability and mortality. Unfortunately, due to the complex pathophysiological mechanisms involved, there remains a critical paucity of effective therapeutic interventions capable of achieving neural tissue regeneration and functional recovery enhancement in SCI patients. The advancements in extracellular vesicles (EVs) as a cell-free therapy for SCI have displayed notable benefits. These include their small size, low immunogenicity, capacity to target specific areas, and ability to cross the blood‒brain barrier (BBB). EVs offer the potential to not only repair tissue damage and stimulate regeneration but also effectively deliver and release them at the site of SCI when combined with diverse biomaterials. This review explores the biological role and importance of EVs in treating SCI, highlighting the combined use of modified EVs with different biomaterials and their potential for future applications. It presents new and hopeful treatment approaches for individuals afflicted with SCI.
Collapse
Affiliation(s)
- Jiali Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China; (J.J.); (Z.W.); (Q.B.); (S.C.)
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| | - Ziyi Wang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China; (J.J.); (Z.W.); (Q.B.); (S.C.)
| | - Qinghua Bao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China; (J.J.); (Z.W.); (Q.B.); (S.C.)
| | - Shenyuan Chen
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China; (J.J.); (Z.W.); (Q.B.); (S.C.)
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China; (J.J.); (Z.W.); (Q.B.); (S.C.)
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou 215600, China; (J.J.); (Z.W.); (Q.B.); (S.C.)
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| |
Collapse
|
72
|
Guerrero J, Maevskaia E, Pfister P, Dominguez AP, Ghayor C, Bhattacharya I, Scherberich A, Weber FE. Mineralized Osteoblast-Derived Exosomes and 3D-printed Ceramic-based Scaffolds for Enhanced Bone Healing: A Preclinical Exploration. Acta Biomater 2025:S1742-7061(25)00376-9. [PMID: 40409510 DOI: 10.1016/j.actbio.2025.05.051] [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: 04/25/2025] [Accepted: 05/21/2025] [Indexed: 05/25/2025]
Abstract
In regenerative medicine, addressing the complex challenge of bone tissue regeneration demands innovative strategies. Exosomes, nanoscale vesicles rich in bioactive molecules, have shown great promise in tissue repair. This study focuses on exosomes derived from mineralized osteoblasts (MOBs), which play a pivotal role in bone formation. We investigated the therapeutic potential of exosomes isolated from osteoblasts cultured in osteogenic medium for 21 days, delivered via 3D-printed gyroid scaffolds composed of hydroxyapatite (HA) and tricalcium phosphate (TCP). The exosomes were characterized through nanoparticle tracking analysis to determine size, morphology, and concentration, while proteomics revealed their cargo contents. In vitro, rabbit bone marrow mesenchymal stromal cells (rBMSCs) were cultured as monolayers and within ceramic scaffolds, where MOB-derived exosomes were shown to promote osteogenic differentiation. In vivo, their osteoconductive and bone augmentation capabilities were evaluated in two rabbit calvarial models, while the osteoinductive potential was further tested in a heterotopic mouse model. Neo-bone formation was assessed using µCT and histological analysis. Our findings demonstrated that MOB-derived exosomes upregulated bone-related gene expression and promoted mineralization in rBMSCs, even in the absence of osteogenic medium. Proteomics confirmed the presence of bone-associated proteins in these exosomes. In rabbit models, however, exosomes did not significantly enhance bone formation. In contrast, in the heterotopic mouse model, exosomes functionalized onto ceramic scaffolds exhibited strong osteoinductive activity. This study highlights the potential of MOB-derived exosomes to enhance 3D-printed ceramic scaffolds for bone regeneration, offering a promising avenue for bone healing without the need for additional growth factors or stem cells. STATEMENT OF SIGNIFICANCE: The here presented report of our project not only advances our understanding of the role of exosome-functionalized scaffolds in bone regeneration but also proposes a promising alternative to traditional growth factor- or cell-based approaches. We are confident that this study represents a novel and impactful contribution to the field.
Collapse
Affiliation(s)
- Julien Guerrero
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Ekaterina Maevskaia
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Pablo Pfister
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Ana Pérez Dominguez
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Chafik Ghayor
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Indranil Bhattacharya
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland
| | - Arnaud Scherberich
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Franz E Weber
- University of Zurich, Center of Dental Medicine, Oral Biotechnology & Bioengineering, Zürich, Switzerland; CABMM, Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
73
|
Xu R, Lu Y, Cai L, Zhang L. Utilizing Extracellular Vesicles from Phaeodactylum tricornutum as a Novel Approach for Protecting the Skin from Oxidative Damage. ACS Biomater Sci Eng 2025. [PMID: 40396567 DOI: 10.1021/acsbiomaterials.4c02346] [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: 05/22/2025]
Abstract
Oxidative stress is a principal factor contributing to skin damage induced by deleterious stimuli, including ultraviolet (UV) radiation. Microalgae-derived extracellular vesicles (EVs), particularly those from Phaeodactylum tricornutum (PTEV), are gaining recognition as a potential therapeutic avenue for restoring skin homeostasis, owing to their scalable production and multifaceted biological activities. This study evaluates the therapeutic effects of PTEV on oxidative damage in H2O2-stimulated HaCaT cells and UV-exposed KM mouse models, based on the extraction and characterization of PTEV. Subsequently, the oxidative stress injury model of HaCaT cells induced by H2O2 and the acute photodamage model of KM mice skin induced by UV were established. The results show that HaCaT cells exhibit a time-dependent uptake of PTEV, confirming that PTEV is nontoxic and has the potential for intercellular cross-boundary regulation. Treatment with PTEV can enhance the vitality of H2O2-stimulated HaCaT cells, reduce intracellular ROS levels, and increase antioxidant enzyme activity in the cells. Further evaluation revealed that PTEV can inhibit UV-induced thickening of the epidermis and degradation of collagen fibers in mice by suppressing the overexpression of matrix metalloproteinase (MMP-3) induced by UV. It enhances the expression of type I collagen (COL1A1) and increases the activity of antioxidant enzymes, as well as the overall antioxidant capacity of tissues. Additionally, PTEV reduces the increase in malondialdehyde levels and lowers the expression levels of inflammatory factors TNF-α and IL-6, thereby protecting the skin barrier and function in mice with acute photodamage. Continuous production of PTEV offers promising applications in therapeutic strategies.
Collapse
Affiliation(s)
- Ran Xu
- Ningbo Innovation Center, Zhejiang University, Ningbo 315100, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Ying Lu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Luyun Cai
- Ningbo Innovation Center, Zhejiang University, Ningbo 315100, China
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China
| | - Litao Zhang
- CAS and Shandong Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
74
|
Duffield C, Rey Gomez LM, Hnit SST, Zhang W, Tsao SCH, Inglis DW, Wang Y. Multi-line lateral flow immunoassay for the detection and subtyping of breast cancer-derived small extracellular vesicles. Chem Commun (Camb) 2025. [PMID: 40395113 DOI: 10.1039/d5cc01038a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Cancer cell-derived small extracellular vesicles (sEVs) are new promising biomarkers for improved cancer diagnosis. A multi-line lateral flow immunoassay (LFIA) has been proposed for sensitive detection and subtyping of breast cancer-derived sEVs. The expression of cancer surface biomarkers (EpCAM) was confirmed through colorimetric analysis of cell line-derived sEVs (p = 0.076) and patient samples, highlighting the potential for their use as a clinical aid.
Collapse
Affiliation(s)
- Chloe Duffield
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Laura M Rey Gomez
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Su Su Thae Hnit
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Wei Zhang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - David W Inglis
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Australia
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Australia.
| |
Collapse
|
75
|
Luo J, Yang A, Wang S, Zhu Q. Integrated Single Nanoparticle Analysis for Rapid Quantification of Spatiotemporal Crosstalk between Herpes Simplex Virus-1 and Extracellular Vesicles. NANO LETTERS 2025; 25:8151-8158. [PMID: 40340482 DOI: 10.1021/acs.nanolett.5c00794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Cells secrete extracellular vesicles (EVs) to mediate precise communication during viral infections, yet the spatiotemporal regulation of EV composition by herpes simplex virus 1 (HSV-1) remains poorly understood. Here, we develop an integrated single-nanoparticle analysis platform combining nanoporous membrane-based EV isolation with an on-chip immunoassay to quantitatively probe EV-HSV-1 interplay throughout infection. A dual-membrane filter design significantly enhances nanoparticle recovery, enabling high-sensitivity single-particle detection. We reveal that HSV-1-infected neural stem cells display viral glycoprotein B on EV surfaces at an early stage (<8 hpi), while intact virions are selectively packaged into EVs later (24-48 hpi). Proteomic profiling indicates infected cell-derived EVs facilitate antigen processing and presentation, potentially amplifying antiviral responses. Functional studies further demonstrate EVs promote viral entry at late stages (48 hpi), likely via EV-virion encapsulation. These findings elucidate a dynamic EV-virus interplay, offering insights into HSV-1 pathogenesis and EV-mediated immune modulation. Our platform provides a transformative approach for advancing infection diagnostics and therapeutics.
Collapse
Affiliation(s)
- Jiaxin Luo
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Ai Yang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Siyao Wang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qingfu Zhu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Zhejiang Key Laboratory of Key Technologies for Visual Pathway Reconstruction, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
76
|
Pan L, Li C. Exosome Therapy for Androgenetic Alopecia: Balancing Potential with Standardization Challenges. Aesthetic Plast Surg 2025:10.1007/s00266-025-04981-y. [PMID: 40394314 DOI: 10.1007/s00266-025-04981-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025]
Affiliation(s)
- Lingfeng Pan
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Caihong Li
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Hartmannstr, 14, 91054, Erlangen, Germany.
| |
Collapse
|
77
|
Cufaro MC, Lanuti P, De Bellis D, Veschi S, Piro A, Fontana A, Di Sebastiano A, Brocco D, Simeone P, Pilato S, Khorooshi RMH, Tomassini V, Rispoli MG, Federici L, Cicalini I, Pieragostino D, Del Boccio P. FACS-Proteomics strategy toward extracellular vesicles single-phenotype characterization in biological fluids: exploring the role of leukocyte-derived EVs in multiple sclerosis. J Transl Med 2025; 23:565. [PMID: 40394633 PMCID: PMC12093873 DOI: 10.1186/s12967-025-06558-4] [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: 01/30/2025] [Accepted: 04/30/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND The isolation and proteomics characterization of extracellular vesicles (EVs) from body fluids is challenging due to their vast heterogeneity. We have recently demonstrated that Fluorescence-activated Cell Sorting (FACS) efficiently isolates the whole EV circulating compartment directly from untouched body fluids enabling a comprehensive EV proteomics analysis. RESULTS Here, we characterized, for the first time, a single-phenotype EV subset by sorting leukocyte-derived EVs (Leuko EVs) from peripheral blood and tears of healthy volunteers. Using an optimized and patented staining protocol of the whole EV compartment we identified and excluded non-EV particles, debris and damaged EVs. We further isolated, using an anti-CD45 antibody, Leuko EVs (CD45+ EVs), reaching a high level of purity (> 90%). Purified Leuko EVs were characterized using atomic force microscopy, nanoparticle tracking, and shotgun proteomics analysis revealing a similar coded protein cargo in both biological fluids. Subsequently, the same workflow was applied to tears from Relapsing-Remitting Multiple Sclerosis (RRMS) patients, revealing a Leuko EVs protein cargo enrichment that reflects the neuroinflammatory condition characteristics of RRMS. This enrichment was evidenced by the activation of upstream regulators TGFB1 and NFE2L2, which are associated with inflammatory responses. Additionally, the analysis identified markers indicative of endothelial cell proliferation and the development of enhanced vascular networks, with AGNPT2 and VEGF emerging as activated upstream regulators. These findings indicate the complex interplay between inflammation and angiogenesis in RRMS. CONCLUSIONS In conclusion, our combined FACS-Proteomics strategy offers a promising approach for biomarker discovery, analysing cell-specific EV phenotypes directly from untouched body fluids, advancing the clinical value of tears EVs and improving the understanding of EV-mediated processes in vivo. Data are available via ProteomeXchange with the identifier PXD049036 and in EV-TRACK knowledgebase with ID: EV240150.
Collapse
Affiliation(s)
- Maria Concetta Cufaro
- Department of Innovative Technologies in Medicine and Dentistry, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Paola Lanuti
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Domenico De Bellis
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Serena Veschi
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Anna Piro
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Antonella Fontana
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Alice Di Sebastiano
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Science, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Davide Brocco
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Pasquale Simeone
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Serena Pilato
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Reza M H Khorooshi
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
| | - Valentina Tomassini
- Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Marianna Gabriella Rispoli
- Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara, Italy
| | - Luca Federici
- Department of Innovative Technologies in Medicine and Dentistry, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Ilaria Cicalini
- Department of Innovative Technologies in Medicine and Dentistry, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine and Dentistry, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST), G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy.
- Department of Science, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
78
|
Dai X, Huang H, Liu F. Rapid Identification of Esophageal Squamous Cell Carcinoma Biomarkers by MALDI-TOF MS Fingerprinting of Extracellular Vesicles. Anal Chem 2025; 97:10180-10189. [PMID: 40326690 DOI: 10.1021/acs.analchem.4c06273] [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: 05/07/2025]
Abstract
Esophageal cancer is a major global health challenge, with high incidence and mortality due to the lack of rapid and sensitive diagnostic tools and specific biomarkers. Cancer-cell-derived extracellular vesicles (EVs) carry unique proteins and nucleic acids, making them valuable sources of cancer biomarkers. We report an integrated method that combines an ultrafast exosome isolation system (EXODUS) with matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) to detect EVs and identify protein biomarkers for diagnosing and monitoring esophageal squamous cell carcinoma (ESCC). EVs derived from 20 mL culture medium supernatant of ESCC cells with varying degrees of differentiation serve as analysis models. We use EXODUS to isolate EVs rapidly. We then analyze the intact EVs using MALDI-TOF MS, which provides cell line-specific EV fingerprints in minutes. These protein fingerprints allow the discrimination of ESCC from normal control cells and enable the classification of ESCC based on the degree of cell differentiation. We explore critical EV biomarker peaks for ESCC diagnosis (5555 m/z, 8603 m/z, etc.) and monitoring (2268 m/z, etc.). Potential EV biomarker candidates, including YBX1, DIRAS2, HIST1H2AH, and MYBBP1A, are identified through tandem mass tag (TMT) proteomics. We tentatively assign the protein identities of EV marker peaks by correlation with the TMT proteomics. Applying this method to plasma-derived EVs shows promise for rapid, minimally invasive diagnosis and monitoring of ESCC.
Collapse
Affiliation(s)
- Xiaodan Dai
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Huiying Huang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Fei Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
79
|
Wu Y, Chen W, Deng J, Cao X, Yang Z, Chen J, Tan Q, Zhou E, Li M, Liu J, Guo M, Jin Y. Tumour-derived microparticles obtained through microwave irradiation induce immunogenic cell death in lung adenocarcinoma. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01922-3. [PMID: 40389640 DOI: 10.1038/s41565-025-01922-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/31/2025] [Indexed: 05/21/2025]
Abstract
Tumour-derived microparticles (TMPs), extracellular vesicles traditionally obtained upon ultraviolet (UV) radiation of tumour cells, hold promise in tumour immunotherapies and vaccines and have demonstrated potential as drug delivery systems for tumour treatment. However, concerns remain regarding the limited efficacy and safety of UV-derived TMPs. Here we introduce a microwave (MW)-assisted method for preparing TMPs, termed MW-TMPs. Brief exposure of tumour cells to short-wavelength MW radiation promotes the release of TMPs showing superior in vivo antitumour activity and safety compared with UV-TMPs. MW-TMPs induce immunogenic cell death and reprogramme suppressive tumour immune microenvironments in different lung tumour models, enabling dual targeting of tumour cells by natural killer and T cells. We show that they can efficiently deliver methotrexate to tumours, synergistically boosting the efficacy of PD-L1 blockade. This MW-TMP development strategy is simpler, more efficient and safer than traditional UV-TMP methods.
Collapse
Affiliation(s)
- Yali Wu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjuan Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Deng
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinghui Cao
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zimo Yang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Tan
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - E Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minglei Li
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiatong Liu
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Diseases, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
80
|
Ghoshal B, Patkulkar PA, Bhatt P, Rana S, Sinharay S, Jhunjhunwala S. Neutrophil-Derived Extracellular Vesicles for Facile Delivery of Diagnostic Agents to Tumor Microenvironments. ACS APPLIED BIO MATERIALS 2025; 8:4132-4139. [PMID: 40276969 DOI: 10.1021/acsabm.5c00266] [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] [Indexed: 04/26/2025]
Abstract
Targeted delivery of diagnostic and therapeutic agents to tumor microenvironments using nanoparticles improves the efficacy of these agents and reduces their unwarranted side effects. Numerous synthetic nanoparticle systems have been designed for this very purpose, but few have translated clinically due to poor efficacy-to-cost and efficacy-to-toxicity ratios. Biological nanoparticles such as modified extracellular vesicles (EVs) that are likely to have lower toxicities have also been developed but face challenges in clinical translation as they have primarily been produced from cancerous cells/cell lines and have high batch-to-batch variability. To overcome these issues, herein, we demonstrate that EVs isolated from neutrophils may be loaded with a specific diagnostic agent (indocyanine green) in a facile manner within a few hours, that these agent-loaded EVs are retained in vivo in mouse tumors for longer with a ∼5-fold increase in retention when compared to free diagnostic agents and hence facilitate short-term longitudinal imaging of the tumor microenvironments. The use of an individual's own cell-derived EVs requiring minimal ex vivo manipulations increases the likelihood of translating such a system into the clinic.
Collapse
Affiliation(s)
- Bartika Ghoshal
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | | | - Preeti Bhatt
- Materials Research Centre, Indian Institute of Science, Bengaluru 560012, India
| | - Subinoy Rana
- Materials Research Centre, Indian Institute of Science, Bengaluru 560012, India
| | - Sanhita Sinharay
- Department of Bioengineering, Indian Institute of Science, Bengaluru 560012, India
| | | |
Collapse
|
81
|
Ruan T, Han J, Xue C, Wang F, Lin J. Mesenchymal stem cells protect the integrity of the alveolar epithelial barrier through extracellular vesicles by inhibiting MAPK-mediated necroptosis. Stem Cell Res Ther 2025; 16:250. [PMID: 40390004 PMCID: PMC12090679 DOI: 10.1186/s13287-025-04388-1] [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: 09/11/2024] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Alveolar‒capillary barrier disruption is a hallmark of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). The contribution of necroptosis to the compromised alveolar-barrier in ALI remains unclear. Mesenchymal stem cells (MSCs) may contribute to tissue repair in ALI and ARDS. Here we evaluated the efficacy and explored the molecular mechanisms of menstrual blood-derived endometrial stem cells (MenSCs) and MenSC-derived extracellular vesicles (MenSC-EVs) in ALI-induced alveolar epithelial barrier dysfunction. METHODS Human lung epithelial cells were stimulated with endotoxin and treated with MenSCs or MenSC-EVs, and their barrier properties were evaluated. Lipopolysaccharide (LPS)-injured mice were treated with MenSCs or MSC-EVs, and the degree of lung injury and the alveolar epithelial barrier of the lung tissue were assessed. RESULTS We found that MenSCs reduced lung injury and restored alveolar-barrier integrity in lung tissue. In vitro, MenSCs reduced paracellular permeability and restored barrier integrity in human lung epithelial cells. MenSC-EVs replicated all these MenSC-mediated changes. Mechanistic research revealed that MenSCs inhibited MAPK signaling and necroptosis. JNK inhibition SP600125, and ERK inhibition U0126 or inhibition of necroptosis with Nec-1 or GSK872 diminished the beneficial anti-epithelial barrier dysfunction effects of MenSCs or MenSC-EVs. CONCLUSIONS Our results suggest that human menstrual blood-derived endometrial stem cells mitigate lung injury and improve alveolar barrier properties by inhibiting MAPK-mediated necroptosis through extracellular vesicles, supporting the application of MenSCs or MenSC-derived extracellular vesicles to treat ALI or ARDS.
Collapse
Affiliation(s)
- Tao Ruan
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jiaming Han
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chengxu Xue
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Fengyuan Wang
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
82
|
Margiotta A. Coupling of Intracellular Calcium Homeostasis and Formation and Secretion of Matrix Vesicles: Their Role in the Mechanism of Biomineralization. Cells 2025; 14:733. [PMID: 40422236 DOI: 10.3390/cells14100733] [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: 03/29/2025] [Revised: 05/07/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
The human bone is a dynamic, highly vascularized tissue composed of 60-70% minerals, which include mainly calcium phosphate (CaP) in the form of hydroxyapatite (HA) crystals, 30% organic matrix composed of type I collagen fibers, and less than 5% water and lipids. The crystals are formed inside the matrix vesicles (MVs) and are then released in the organic collagen-based fibrous matrix. Extracellular matrix (ECM) formation and mineralization processes, named osteogenesis, are associated with human mesenchymal stem cells (hMSCs) undergoing differentiation into osteoblasts (osteoblastogenesis). Osteogenesis is regulated by multiple intracellular signaling and genetic pathways and by environmental factors. Calcium flow is finely regulated and plays a key role in both osteoblastogenesis and osteogenesis. The formation and accumulation of CaP, the biogenesis of MVs, their secretion, and the deposition of HA crystals to fill the organic bone matrix are the fundamental events in the biomineralization process. In this paper, I will describe and discuss the recent findings and hypothesis on the molecular mechanism regulating this process.
Collapse
Affiliation(s)
- Azzurra Margiotta
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway
| |
Collapse
|
83
|
Yan Q, Liu M, Mao J, Zhao Z, Wang B. Extracellular Vesicles in Acute Kidney Injury: Mechanisms, Biomarkers, and Therapeutic Potential. Int J Nanomedicine 2025; 20:6271-6288. [PMID: 40400780 PMCID: PMC12094478 DOI: 10.2147/ijn.s519345] [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: 01/24/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025] Open
Abstract
Acute kidney injury (AKI) has a high morbidity and mortality rate but can only be treated with supportive therapy in most cases. The diagnosis of AKI is mainly based on serum creatinine level and urine volume, which cannot detect kidney injury sensitive and timely. Therefore, new diagnostic and therapeutic molecules of AKI are being actively explored. Extracellular vesicles (EVs), secreted by almost all cells, can originate from different parts of the kidney and mediate intercellular communication between various cell types of nephrons. At present, numerous successful EV-based biomarker discoveries and treatments for AKI have been made, such as the confirmed diagnostic role of urine-derived EVs in AKI and the established therapeutic role of mesenchymal stem cell-derived EVs in AKI have been confirmed; however, these related studies lack a full discussion. In this review, we summarize the latest progression in the profound understanding of the functional role of EVs in AKI caused by various etiologies in recent years and provide new insights into EVs as viable biomarkers and therapeutic molecules for AKI patients. Furthermore, the current challenges and prospects of this research area are briefly discussed, presenting a comprehensive overview of the growing foregrounds of EVs in AKI.
Collapse
Affiliation(s)
- Qianqian Yan
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Mengyuan Liu
- Department of Anesthesiology, Air Force Hospital of Western Theater Command, PLA, Chengdu, 610011, People’s Republic of China
| | - Jinyan Mao
- Department of Anesthesiology, Air Force Hospital of Western Theater Command, PLA, Chengdu, 610011, People’s Republic of China
| | - Zihao Zhao
- Nephrology Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Department of Radiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Bo Wang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
84
|
Di Febo R, Saeed Z, Serafini F, Brocco D, D'Ascanio F, Pizzi AD, Tinari N, Crescitelli R, Lanuti P, Renda G. Diagnostic and prognostic roles of endothelial- and platelet-derived extracellular vesicles in cardiovascular diseases. J Transl Med 2025; 23:553. [PMID: 40380176 PMCID: PMC12085008 DOI: 10.1186/s12967-025-06522-2] [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: 01/31/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound structures released by all cell types. They play a critical role in intercellular communication by transferring their cargo, comprising proteins, lipids, metabolites, RNAs, miRNAs, and DNA fragments, to recipient cells. This transfer influences gene expression, signaling pathways, and cellular behavior. Due to their ability to alter the physiology of recipient cells, EVs hold significant therapeutic potential. Additionally, EVs are implicated in various physiological and pathological processes, including immune regulation, cancer progression, and cardiovascular diseases. EVs have been detected in many biological fluids, such as peripheral blood, saliva, urine, cerebrospinal fluid, and breast milk. The cargo of EVs dynamically reflects the physiological and pathological state of their parent cells, making them promising candidates for liquid biopsies in various clinical conditions. Specifically, different EV subtypes in cardiovascular diseases have been studied, with both endothelial and platelet-derived EVs playing significant roles in cardiovascular pathologies. This review focuses on the diagnostic and prognostic potential of endothelial and platelet-derived EVs in cardiovascular diseases, highlighting the role of EV subpopulations.
Collapse
Affiliation(s)
- Riccardo Di Febo
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Zeeba Saeed
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francesco Serafini
- Department of Neuroscience, Imaging and Clinical Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Davide Brocco
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Francesca D'Ascanio
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Humanities, Law and Economics, Leonardo da Vinci University, 66010, Torrevecchia Teatina (CH), Italy
| | - Andrea Delli Pizzi
- Department of Innovative Technologies in Medicine & Dentistry, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Institute for Advanced Biomedical Technologies, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Paola Lanuti
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
- Department of Medicine and Aging Science, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy
| | - Giulia Renda
- Center for Advanced Studies and Technology, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy.
- Department of Neuroscience, Imaging and Clinical Sciences, G. d'Annunzio University of Chieti-Pescara, 66100, Chieti, Italy.
| |
Collapse
|
85
|
Liu X, Ding L, Zhang A, Feng F, Zhou F, Wu Y. Dynamic characteristics of metabolism and small extracellular vesicles during malignant transformation of BEAS-2B cells induced by coal tar pitch extract. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126108. [PMID: 40154873 DOI: 10.1016/j.envpol.2025.126108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Lung cancer poses a significant global burden with rising morbidity and mortality. Coal tar pitch-induced lung cancer is an occupational disease where early detection is crucial but challenging due to unclear pathogenesis. We established a malignant transformation model using BEAS-2B cells treated with coal tar pitch extract (CTPE). Macro- and micro-observations showed CTPE-induced alterations, including changes in cell morphology, enhanced proliferation and migration abilities, upregulated EGFR expression, modified levels of CYP1A1 and GSTM1 metabolizing enzymes, and a transition towards a mesenchymal phenotype. These findings strongly suggest that the cells have undergone malignant transformation. Metabolomics analysis revealed changes in 1120 metabolites, with 31 co-expressed, mainly in energy and amino acid metabolism. Small extracellular vesicles (SEVs) concentrations and EGFR levels were significantly altered. Correlation analysis identified a relationship between these biomarkers, implying their potential significance as early events in the initiation and progression of lung cancer. These findings provide valuable insights and a rationale for lung cancer screening and mechanistic investigations, thereby contributing to a deeper understanding of the disease.
Collapse
Affiliation(s)
- Xia Liu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Aiai Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Fang Zhou
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
86
|
Gummadi S, Chitti SV, Kang T, Shahi S, Mathivanan S, Fonseka P. ExoCarta 2024: A Web-based Repository of Small Extracellular Vesicles Cargo. J Mol Biol 2025:169218. [PMID: 40381982 DOI: 10.1016/j.jmb.2025.169218] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/06/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Small extracellular vesicles (sEVs) are small membrane vesicles of endocytic origin secreted into the extracellular environment by all cell types and are known to play a crucial role in intercellular and intracellular communication. These vesicles contain proteins, nucleic acids and lipids and their molecular content reflect the normal and pathophysiological conditions of the host cells. Hence, there is a significant interest in cataloguing the molecular content of sEVs in various conditions as this would aid researchers in understanding the biological roles and altered cellular processes under various diseases and healthy state. Here we report ExoCarta (https://www.exocarta.org), a freely accessible web-based compendium of studies that encompasses DNA, RNA, proteins, and lipids that are detected in sEVs. ExoCarta catalogues both published and unpublished sEV studies. Current version of ExoCarta contains data from 1249 sEV studies, 119,489 protein entries, 15,868 RNA entries, 3,946 lipid entries and quantitative data for 24,073 entries. QUANT, a quantitative plugin, enables users to compare protein abundance between conditions in a study for gene/protein of interest in real time. ExoCarta provides Gene Ontology (GO) annotations and reactome pathways along with dynamic protein-protein interaction networks, for sEV proteins. A tab delimited file containing the most identified sEV proteins is available for users to download. The integration of sEV DNA studies, QUANT and regularly updated dataset of the ExoCarta database makes it an invaluable resource for small extracellular vesicles researchers across the globe.
Collapse
Affiliation(s)
- Sriram Gummadi
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Sai V Chitti
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Taeyoung Kang
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Sanjay Shahi
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Suresh Mathivanan
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Pamali Fonseka
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
87
|
Gu X, Fan Z, Lu L, Xu H, He L, Shen H, Huang R, Li Z. Machine learning-assisted washing-free detection of extracellular vesicles by target recycling amplification based fluorescent aptasensor for accurate diagnosis of gastric cancer. Talanta 2025; 287:127506. [PMID: 39837204 DOI: 10.1016/j.talanta.2024.127506] [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: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/23/2025]
Abstract
Extracellular vesicles (EVs) are promising non-invasive biomarkers for cancer diagnosis. EVs proteins play a critical role in tumor progress and metastasis. However, accurately and reliably diagnosing cancers is greatly limited by single protein marker on EVs. Here, we reported an accurate diagnosis model of gastric cancer by analyzing five types of EVs surface proteins using machine learning in a retrospective study design. A washing-free detection method based on aptasensor and exonuclease Ⅰ was used to profile EVs surface proteins. The aptamer was designed as hairpin structure. The presence of target protein positive EVs converted the conformation of hairpin probes, which subsequently degraded by exonuclease Ⅰ. The exposed target protein could bind with and then open new hairpin probes, thus forming an amplification cycle. The lengths of different detection probes were optimized for detection. With the combination of five target proteins, five machine learning algorithms were compared to achieve a higher diagnostic accuracy. The best model, XGBoost, validated with 20 % of detection results could reach an accuracy of 0.8421. Furthermore, the XGBoost-based surface protein analysis could precisely identify gastric cancer patients with the area under the curve value of 0.9347 (95 % confidential interval (CI) = 0.8590 to 1.000). Since our method utilized a simple and versatile design of detection probes, its diagnostic scope could potentially be expanded to include different protein markers and accurately diagnose other diseases in the future.
Collapse
Affiliation(s)
- Xinrui Gu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Zeyu Fan
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China
| | - Luying Lu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Hongpan Xu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Lei He
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China.
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China.
| | - Zhiyang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| |
Collapse
|
88
|
Frisk NLS, Jørgensen MM, Bæk R, Atic AI, Brodersen TR, Ostrowski SR, Larsen MH, Posselt D, Høgdall E, Høgdall C, Pedersen OBV, Dalgaard LT. Characterization of small extracellular vesicles from ovarian cancer patients and pre-diagnostic patient samples: Evidence from the Danish blood donor study. PLoS One 2025; 20:e0323529. [PMID: 40372993 PMCID: PMC12080785 DOI: 10.1371/journal.pone.0323529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/08/2025] [Indexed: 05/17/2025] Open
Abstract
AIM Ovarian cancer (OC) is the leading cause of gynecological cancer deaths. Current biomarkers of OC are not specific or sensitive enough. Extracellular vesicles (EVs), EV surface proteins and their cargo microRNA (miRNA) show potential as biomarkers. This study aimed to characterize the ability of EVs to identify early OC-biomarkers among blood donors six months before their diagnosis. METHODS Study groups of OC patients, benign tumor patients (B), healthy blood donors (Control), and blood donors with incident OC diagnosis within six months of the last blood draw (Pre-diagnostic; PD) were established. Small EVs were enriched from plasma using ultracentrifugation. EVs were characterized by Dynamic Light Scattering (DLS), EV Array, NanoFlow Cytometry, Nanoparticle Tracking Analysis, and Western blots. RNA from EVs was isolated. A discovery study was performed on OC and B patients using the TaqMan Array Human MicroRNA A card. A validation study of 9 specific miRNAs was performed using RT-qPCR. RESULTS With DLS, it was identified that the OC patients' EVs were more heterogeneous in size compared to the other groups. Western blot identified CD63 and TSG101 in the EV enrichments. EV Array assessed 22 known protein biomarkers. TaqMan MicroRNA Array cards indicated a differential miRNA abundance between OC and B; however, technical replication and validation could not validate this pattern. CONCLUSION This study has analyzed EVs in OC, B, Control, and PD women. More extensive investigations of EV CD9, CD151, and CD81 in conjunction with other risk factors and well-known biomarkers like CA125 or HE4 should be the main objectives of future research.
Collapse
Affiliation(s)
- Nanna Lond Skov Frisk
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Malene Møller Jørgensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Rikke Bæk
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Amila Iriskic Atic
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Novo Nordisk A/S, Måløv, Copenhagen, Denmark
| | | | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Dorthe Posselt
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Claus Høgdall
- Department of Gynaecology, Rigshospitalet, Copenhagen, Denmark
| | - Ole Birger Vesterager Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
89
|
Jayabalan N, Nair S, Lai A, Scholz-Romero K, Razo-Azamar M, Ormazabal V, Lim R, Carrion F, Guanzon D, Rice GE, McIntyre HD, Lappas M, Salomon C. Extracellular vesicle-associated miR-515-5p from adipose tissue regulates placental metabolism and fetal growth in gestational diabetes mellitus. Cardiovasc Diabetol 2025; 24:205. [PMID: 40369565 PMCID: PMC12080180 DOI: 10.1186/s12933-025-02739-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) affects 2-20% of pregnant women worldwide and is linked to fetal overgrowth, increased perinatal morbidity, and mortality, as well as a higher risk of developing cardiovascular disease later in life for mother and child. MicroRNAs (miRNAs), which regulate gene expression, can be transported within extracellular vesicles (EVs). Adipose tissue-derived EVs have been associated with changes in placental metabolism in GDM, potentially influencing cardiovascular health outcomes. This study aimed to evaluate the miRNA profile in EVs from omental adipose tissue in GDM and their effect on placental nutrient uptake and fetal growth. METHODS This case-control study included patients with normal glucose tolerance (NGT) and GDM. We conducted a miRNA expression profiling on omental adipose tissue and its derived EVs from women with NGT (n = 20) and GDM (n = 36). Trophoblast cells were utilized to assess the effect of EVs on glucose and fatty acid uptake, pro-inflammatory cytokine, and chemokine release. Double-stranded miRNA mimics were used to investigate the effect of selected miRNAs on trophoblast cells. Subsequently, the impact of EVs from NGT and GDM, as well as miR-515-5p, on in vivo glucose tolerance and fetal growth was assessed in pregnant mice. RESULTS Fifty-four miRNAs showed significant differences between EVs from the adipose tissue of NGT and GDM groups. EVs from GDM increased glucose uptake in trophoblast cells, whereas EVs from NGT increased the secretion of CXCL8, IL-6, CXCL1, CXCL4, and CXCL5 from trophoblasts compared to the effect without EVs. Specifically, miR-515-5p increased glucose uptake and abolished TNF-α-dependent increase in pro-inflammatory cytokines and chemokines from trophoblast cells. Injection of pregnant mice with EVs from NGT adipose tissue loaded with miR-515-5p resulted in increased fetal weight and glucose levels. CONCLUSION miR-515-5p, specifically encapsulated within EVs from omental adipose tissue in GDM, regulates placental nutrient uptake, glucose homeostasis, and fetal growth.
Collapse
Affiliation(s)
- Nanthini Jayabalan
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia.
| | - Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- UQ Centre for Extracellular Vesicle nanomedicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- UQ Centre for Extracellular Vesicle nanomedicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Katherin Scholz-Romero
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- UQ Centre for Extracellular Vesicle nanomedicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Melissa Razo-Azamar
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- UQ Centre for Extracellular Vesicle nanomedicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Valeska Ormazabal
- Faculty of Biological Sciences, Pharmacology Department, University of Concepcion, Concepción, Chile
| | - Ratana Lim
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile
| | - Dominic Guanzon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- UQ Centre for Extracellular Vesicle nanomedicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | - Gregory E Rice
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- INOVIQ Ltd, Notting Hill, VIC, 3168, Australia
| | - Harold David McIntyre
- Mater Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia.
- UQ Centre for Extracellular Vesicle nanomedicine, The University of Queensland, Brisbane, QLD, 4029, Australia.
| |
Collapse
|
90
|
Zhang H, Liang Z, Zhuang H, Wang M, Huang Y, Cao X, Chen H, Shen L, Feng C. Proteomic study of plasma and L1CAM-captured exosomal proteins in children with autism spectrum disorders. J Pharm Biomed Anal 2025; 264:116965. [PMID: 40393167 DOI: 10.1016/j.jpba.2025.116965] [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: 03/09/2025] [Revised: 04/28/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
Autism spectrum disorder (ASD) has become a neurodevelopmental disorder that seriously endangers the health of infants and children. In order to explore the pathogenesis of the disease and search for early diagnostic biomarkers. In this study, plasma exosomes (PEs) and neural cell adhesion molecule L1 (L1CAM)-captured exosomes (LCEs) of ASD and controls were extracted and lysed to obtain proteins. Isobaric tags for relative and absolute quantitation (iTRAQ) proteomics were applied to investigate the differences in the expression of PEs and LCEs proteins between the two groups. Twenty-eight plasma exosomal differentially expressed proteins (DEPs) were identified, which were mainly associated with immunity, inflammation, complement and coagulation, and lipoprotein metabolism and transport. Twenty L1CAM-captured exosomal DEPs were identified, which were mainly involved in cytoskeleton, tight junctions, focal adhesion, and platelet-associated pathways. Meanwhile, our results suggested that processes or signaling pathways associated with the DEPs from plasma exosomes may be activated, whereas those associated with L1CAM-captured exosome may be inhibited. These processes or signaling pathways have been reported to be associated with ASD in previous studies. These DEPs have the potential to be diagnostic markers. This study provides new insights into disease mechanisms and diagnostic markers of ASD.
Collapse
Affiliation(s)
- Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Zhiyuan Liang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Hongbin Zhuang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Mingxian Wang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Yuhan Huang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Haiyi Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, PR China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, PR China.
| | - Chengyun Feng
- Shenzhen Baoan Women's and Children's Hospital, Shenzhen 518100, PR China.
| |
Collapse
|
91
|
Wang M, Wang W, Chopp M, Zhang ZG, Zhang Y. Therapeutic and diagnostic potential of extracellular vesicle (EV)-mediated intercellular transfer of mitochondria and mitochondrial components. J Cereb Blood Flow Metab 2025:271678X251338971. [PMID: 40367392 PMCID: PMC12078269 DOI: 10.1177/0271678x251338971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/20/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
Extracellular vesicles (EVs) facilitate the transfer of biological materials between cells throughout the body. Mitochondria, membrane-bound organelles present in the cytoplasm of nearly all eukaryotic cells, are vital for energy production and cellular homeostasis. Recent studies highlight the critical role of the transport of diverse mitochondrial content, such as mitochondrial DNA (mt-DNA), mitochondrial RNA (mt-RNA), mitochondrial proteins (mt-Prots), and intact mitochondria by small EVs (<200 nm) and large EVs (>200 nm) to recipient cells, where these cargos contribute to cellular and mitochondrial homeostasis. The interplay between EVs and mitochondrial components has significant implications for health, metabolic regulation, and potential as biomarkers. Despite advancements, the mechanisms governing EV-mitochondria crosstalk and the regulatory effect of mitochondrial EVs remain poorly understood. This review explores the roles of EVs and their mitochondrial cargos in health and disease, examines potential mechanisms underlying their interactions, and emphasizes the therapeutic potential of EVs for neurological and systemic conditions associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Mingjin Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Weida Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | | | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
92
|
Pirolli NH, Raufman JP, Jay SM. Therapeutic Potential and Translational Challenges for Bacterial Extracellular Vesicles in Inflammatory Bowel Disease. Inflamm Bowel Dis 2025:izaf107. [PMID: 40357729 DOI: 10.1093/ibd/izaf107] [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: 02/19/2025] [Indexed: 05/15/2025]
Abstract
Despite the availability of numerous new immune-directed therapeutics, the major constituents of inflammatory bowel disease (IBD)-ulcerative colitis (UC) and Crohn's disease (CD)-continue to afflict millions worldwide, resulting in significant morbidity and long-term health risks. IBD results from a triad of immune, environmental (eg, gut microbiome), and genetic (including epigenetic) mechanisms, and therefore has been subject to a wide variety of therapeutic strategies. Among these, the administration of probiotics, particularly Gram-positive lactic acid bacteria (LAB), targeting both immune and environmental factors, has shown promising potential for efficacy in selected populations in early clinical trials. However, knowledge gaps and inconsistent efficacy currently prevent recommendations for the use of probiotics in larger IBD patient populations. The inconsistent efficacy of probiotics is likely due to variable cell viability and potency after administration, further exacerbated by IBD patient heterogeneity. Thus, an alternative to live probiotics for IBD has emerged in the form of bacterial extracellular vesicles (BEVs)-cell-secreted nanovesicles containing abundant bioactive cargo that, like live probiotics, can regulate immune and environmental factors but with fewer viability limitations and safety concerns. In this review, we summarize the work done to date establishing the potential of BEVs to provide the therapeutic benefits in IBD and discuss the hurdles BEVs must overcome to achieve clinical translation. We also consider future directions for BEV therapeutics, especially treatment potential for necrotizing enterocolitis (NEC), which shares similarities in pathophysiology with IBD.
Collapse
Affiliation(s)
- Nicholas H Pirolli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Biomedical Laboratory Research and Development Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
- Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
93
|
Philippon M, Labib R, Ley MBRG, Kaplan LD, Mendez AJ, Best TM, Kouroupis D. Characterization of Extracellular Vesicles from Infrapatellar Fat Pad Mesenchymal Stem/Stromal Cells Expanded Using Regulatory-Compliant Media and Inflammatory/Hormonal Priming. Cells 2025; 14:706. [PMID: 40422209 PMCID: PMC12109853 DOI: 10.3390/cells14100706] [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: 03/22/2025] [Revised: 04/23/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Osteoarthritis (OA) remains a leading cause of disability worldwide, with no disease-modifying therapies currently available for treatment. The infrapatellar fat pad (IFP) harbors mesenchymal stem/stromal cells (MSC) with potent immunomodulatory and regenerative properties, making them a promising candidate for OA treatment. A growing body of evidence suggests that the therapeutic effects of MSC are largely mediated by their extracellular vesicles (EVs), which carry bioactive cargo that modulates inflammation and tissue repair. However, optimizing MSC-derived EVs as a cell-free therapeutic approach requires an in-depth understanding of how culture conditions and inflammatory/hormonal priming influence their functional properties. In this study, IFP-MSC were expanded in regulatory-compliant human platelet lysate (HPL) and xeno-/serum-free (XFSF) media and primed with an inflammatory/fibrotic cocktail (TIC) with oxytocin (OXT) to assess the impact on their immunophenotypic profile and EV cargo. The immunophenotype confirmed that TIC+OXT-primed MSC retained key immunomodulatory surface markers, while EV characterization verified the successful isolation of CD63+/CD9+ vesicles. Pathway enrichment analysis of both HPL- and XFSF- TIC+OXT EVs cargo identified key miRNAs associated with immune regulation, tissue repair, and anabolic signaling. Functional assays revealed that TIC+OXT EVs promoted M2-like anti-inflammatory macrophage polarization and exhibited chondroprotective properties in chondrocytes/synoviocytes inflammatory osteoarthritic assay. These findings highlight the therapeutic potential of TIC+OXT-primed IFP-MSC-derived EVs as immunomodulatory and chondroprotective agents, offering a promising strategy for OA treatment through a clinically viable, cell-free approach.
Collapse
Affiliation(s)
- Marc Philippon
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Ramy Labib
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Michelle Bellas Romariz Gaudie Ley
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Lee D. Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
| | - Armando J. Mendez
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Thomas M. Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
| | - Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| |
Collapse
|
94
|
Ngo CHL, Tukova A, Zhang W, Tsao SCH, Wang Y. Sensitive detection of small extracellular vesicles using a gold nanostar-based SERS assay. Analyst 2025; 150:2108-2117. [PMID: 40202797 DOI: 10.1039/d5an00110b] [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: 04/11/2025]
Abstract
Small extracellular vesicles (sEVs) are lipid bilayer-bound vesicles that carry critical biomarkers for disease detection. However, the inherent heterogeneity and complexity of sEV molecular characteristics pose significant challenges for accurate and comprehensive molecular profiling. Traditional analytical methods, including immunoblotting, enzyme-linked immunosorbent assay (ELISA), and flow cytometry, exhibit several limitations, such as being time-consuming, requiring large sample volumes, and demonstrating relatively low sensitivity. Therefore, there is an urgent need to develop a highly sensitive and specific assay for the reliable detection of sEVs. Surface-enhanced Raman scattering (SERS) assays have emerged as a promising approach for sEV detection, offering advantages including high sensitivity and specificity. In the proposed SERS assay, SERS nanotags - comprising nanoparticles coated with Raman-active molecules and conjugated with antibodies - are employed to label surface-bound molecules on sEVs. This approach facilitates the generation of a high-intensity signal from molecules present in low abundance. Recently, anisotropic nanoparticles, such as star-shaped nanostructures, have garnered interest due to their ability to significantly amplify generated SERS signals for ultra-sensitive biomarker detection. In this study, we explore the application of gold nanostars (AuNSs) as SERS nanotags for the detection of sEVs. In principle, AuNS-based SERS nanotags were used to label the EpCAM protein, which can be found on the surface of cancer cell derived sEVs, and then sEV labelled SERS nanotags were captured by CD9-conjugated magnetic beads to form an immunocomplex, which exhibits a SERS signal. Our results demonstrate that the proposed SERS assay utilizing AuNSs provides high specificity and sensitivity, with a detection limit as low as 2.47 × 103 particles per μL. Furthermore, the assay was tested with spiked plasma samples (cancer cell-derived sEVs spiked into healthy plasma), showing that its specificity remains unaffected by the presence of plasma. These findings suggest that the SERS assay incorporating AuNSs holds significant promise as an effective and reliable detection method for potential clinical applications.
Collapse
Affiliation(s)
| | - Anastasiia Tukova
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Macquarie University, Sydney, Australia.
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
95
|
Yeh H, Gupta K, Lu YH, Srinivasan A, Delila L, Yen NTH, Nyam-Erdene A, Burnouf T. Platelet Extracellular Vesicles as Natural Delivery Vehicles for Mitochondrial Dysfunction Therapy? ACS Biomater Sci Eng 2025; 11:2601-2621. [PMID: 40280866 PMCID: PMC12076291 DOI: 10.1021/acsbiomaterials.5c00473] [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: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Mitochondria are vital for energy production, metabolic regulation, and cellular signaling. Their dysfunction is strongly implicated in neurological, cardiovascular, and muscular degenerative diseases, where energy deficits and oxidative stress accelerate disease progression. Platelet extracellular vesicles (PEVs), once called "platelet dust", have emerged as promising agents for mitigating mitochondrial dysfunction. Like other extracellular vesicles (EVs), PEVs carry diverse molecular cargo and surface markers implicated in disease processes and therapeutic efficacy. Notably, they may possibly contain intact or partially functional mitochondrial components, making them tentatively attractive for targeting mitochondrial damage. Although direct research on PEVs-mediated mitochondrial rescue remains limited, current evidence suggests that PEVs can modulate diseases associated with mitochondrial dysfunction and potentially enhance mitochondrial health. This review explores the therapeutic potential of PEVs in neurodegenerative and cardiovascular disorders, highlighting their role in restoring mitochondrial health. By examining recent advancements in PEVs research, we aim to shed light on novel strategies for utilizing PEVs as therapeutic agents. Our goal is to underscore the importance of further fundamental and applied research into PEVs-based interventions, as innovative tools for combating a wide range of diseases linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hsien
Chang Yeh
- School
of Medicine, College of Medicine, Taipei
Medical University, Xin-Yi
Campus, Taipei City 110, Taiwan
| | - Kirti Gupta
- International
Graduate Program in Medicine, College of Medicine, Taipei Medical University, Xin-Yi Campus, Taipei 110, Taiwan
| | - Ya-Hsuan Lu
- School
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
| | - Abinaya Srinivasan
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei
City 110, Taiwan
| | - Liling Delila
- Graduate
Institute of Biomedical Materials and Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
| | - Nguyen Tran Hai Yen
- Graduate
Institute of Biomedical Materials and Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
| | - Ariunjargal Nyam-Erdene
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei
City 110, Taiwan
| | - Thierry Burnouf
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei
City 110, Taiwan
- Graduate
Institute of Biomedical Materials and Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
- International
PhD Program in Cell Therapy and Regeneration Medicine, College of
Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
96
|
Suresh PS, Zhang Q. Comprehensive Comparison of Methods for Isolation of Extracellular Vesicles from Human Plasma. J Proteome Res 2025. [PMID: 40356199 DOI: 10.1021/acs.jproteome.5c00149] [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: 05/15/2025]
Abstract
Extracellular vesicles (EVs) are a vital component in cell-cell communication and hold significant potential as biomarkers and therapeutic carriers. Having a reproducible and simple EV isolation method for small volumes of human plasma is essential for biomarker discovery. Although combining multiple methods has been a recent trend in its ability to minimize contamination, it is not ideal for clinical specimens due to the large sample number and small sample volume. This study compared EVs isolated from 100 μL of plasma by nine commonly used methods based on different principles, including centrifugation, polymer precipitation, size exclusion, electrostatic interaction, and affinity enrichment. The isolated EVs were characterized by particle size and number using nanoparticle tracking analysis, purity, and contaminants using Simple Western and overall proteomic profiles using bottom-up proteomics. Despite the same EV enrichment principle, individual methods isolated EVs exhibited distinct characteristics, likely due to variations in the physicochemical properties of materials used and specific protocols. Overall, all of the methods evaluated are reproducible. MagNet and MagCap methods result in purer EVs with the narrowest size distribution and the highest proteome coverage but modest yield. This is the first report on isolating EVs from 100 μL of plasma using nine different methods with detailed characterization.
Collapse
Affiliation(s)
- Patil Shivprasad Suresh
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
| | - Qibin Zhang
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
- Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27402, United States
| |
Collapse
|
97
|
Arthur P, Kandoi S, Kalvala A, Boirie B, Nathani A, Aare M, Bhattacharya S, Kulkarni T, Sun L, Lamba DA, Li Y, Singh M. Cannabidiol-Loaded Retinal Organoid-Derived Extracellular Vesicles Protect Oxidatively Stressed ARPE-19 Cells. Biomedicines 2025; 13:1167. [PMID: 40426994 PMCID: PMC12108686 DOI: 10.3390/biomedicines13051167] [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: 04/01/2025] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Age-related macular degeneration (AMD) is the third leading cause of irreversible blindness in elderly individuals aged over 50 years old. Oxidative stress plays a crucial role in the etiopathogenesis of multifactorial AMD disease. The phospholipid bilayer EVs derived from the culture-conditioned medium of human induced pluripotent stem cell (hiPSC) differentiated retinal organoids aid in cell-to-cell communication, signaling, and extracellular matrix remodeling. The goal of the current study is to establish and evaluate the encapsulation of a hydrophobic compound, cannabidiol (CBD), into retinal organoid-derived extracellular vesicles (EVs) for potential therapeutic use in AMD. Methods: hiPSC-derived retinal organoid EVs were encapsulated with CBD via sonication (CBD-EVs), and structural features were elucidated using atomic force microscopy, nanoparticle tracking analysis, and small/microRNA (miRNA) sequencing. ARPE-19 cells and oxidative-stressed (H2O2) ARPE-19 cells treated with CBD-EVs were assessed for cytotoxicity, apoptosis (MTT assay), reactive oxygen species (DCFDA), and antioxidant proteins (immunohistochemistry and Western blot). Results: Distinct miRNA cargo were identified in early and late retinal organoid-derived EVs, implicating their roles in retinal development, differentiation, and functionality. The therapeutic effects of CBD-loaded EVs on oxidative-stressed ARPE-19 cells showed greater viability, decreased ROS production, downregulated expression of inflammation- and apoptosis-related proteins, and upregulated expression of antioxidants by Western blot and immunocytochemistry. Conclusions: miRNAs are both prognostic and predictive biomarkers and can be a target for developing therapy since they regulate RPE physiology and diseases. Our findings indicate that CBD-EVs could potentially alleviate the course of AMD by activating the targeted proteins linked to the adenosine monophosphate kinase (AMPK) pathway. Implicating the use of CBD-EVs represents a novel frontline to promote long-term abstinence from drugs and pharmacotherapy development in treating AMD.
Collapse
Affiliation(s)
- Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Sangeetha Kandoi
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; (S.K.); (D.A.L.)
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anil Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Breana Boirie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA; (S.B.); (T.K.)
- Department of Physiology and Biomedical Engineering, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA
| | - Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA; (S.B.); (T.K.)
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Deepak A. Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; (S.K.); (D.A.L.)
- Immunology and Regenerative Medicine, Genentech, South San Francisco, CA 94080, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA;
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA; (P.A.); (A.K.); (B.B.); (A.N.); (M.A.)
| |
Collapse
|
98
|
Sun X, Feng Y, Xie L, Wu Y, Mao J, Zhang Y, Zhang L, Yuan X, Ni J, Xiao X, Liu H, Dai L, Ma F. Maternal plasma extracellular vesicles tsRNA as potential biomarkers for assessing preterm labor risk. BMC Pregnancy Childbirth 2025; 25:553. [PMID: 40348952 PMCID: PMC12065249 DOI: 10.1186/s12884-025-07672-3] [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: 01/15/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Spontaneous preterm labor (PTL) accounts for approximately 70% of preterm births, posing significant risks to both maternal and neonatal health. Current predictive biomarkers lack sufficient reliability, underscoring the need for non-invasive and dependable indicators. Emerging research indicates that tRNA-derived small RNAs (tsRNAs) are involved in various diseases; however, their potential association with PTL remains underexplored. METHODS Bioinformatics analyses of public GEO datasets (PRJNA415953 and PRJNA428989) were conducted to identify tsRNAs associated with PTL. Validation was performed using plasma extracellular vesicles samples collected at 12 weeks of gestation from PTL patients (n = 45) and healthy controls (n = 38). Functional assays were used to assess the impact of tsRNA1 (tRNA-Gly-GCC-5p-tRF-921) on extravillous trophoblast (EVT) function, including apoptosis, migration, invasion, and endothelial-like tube formation in HTR8/SVneo cells. Transcriptomic sequencing was conducted to identify tsRNA1-mediated pathways, and DNA methylation patterns were predicted based on the transcriptomic data. Statistical significance was determined using Student's t-test. RESULTS Two tsRNAs, tsRNA1 and tsRNA3 (tRNA-Gly-GCC-5p-tR-half-368), were significantly upregulated in PTL patient samples compared to controls. Overexpression of tsRNA1 impaired EVT function, increased apoptosis, and altered DNA methylation profiles, implicating its critical role in PTL mechanisms. CONCLUSIONS This study identifies tsRNA1 as a key regulator of EVT dysfunction and placental pathology in PTL. The findings provide novel insights into the mechanistic role of tsRNAs in PTL and highlight tsRNA1 as a promising biomarker for early risk stratification and prediction of the condition. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xinrui Sun
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying Feng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liang Xie
- The Joint Laboratory for Pulmonary Development and Related Diseases, West China Second University Hospital, West China Institute of Women and Children's Health, Sichuan University, Chengdu, Sichuan, China
| | - Yilun Wu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jia Mao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, State Key Laboratory of Biotherapy and Cancer Center, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yi Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Linyu Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Yuan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiangping Ni
- College of Horticulture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xue Xiao
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| | - Hanmin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Li Dai
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Health Commission Key Laboratory of Chronobiology (Sichuan University), Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
99
|
Vader P. Extracellular vesicles for drug delivery: A major interest for the Journal of Controlled Release. J Control Release 2025; 381:113599. [PMID: 40049519 DOI: 10.1016/j.jconrel.2025.113599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Affiliation(s)
- Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
100
|
Coelho MO, Quintas ST, Sarmento B, De Wever O, Castro F. Engineered dendritic cells-derived extracellular vesicles for cancer immunotherapy. J Control Release 2025; 381:113620. [PMID: 40088976 DOI: 10.1016/j.jconrel.2025.113620] [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: 12/10/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
Extracellular vesicles (EVs) have emerged as a cell-free therapeutic approach, garnering increasing attention for their potential to enhance the safety and efficacy of immunotherapy. This interest is primarily driven by the biocompatibility and cell/tissue tropism inherent to EVs, but also due to their reconfigurable content. This, termed as cargo, may comprise bioactive molecules as proteins, lipids, and nucleic acids that play a pivotal role in mediating intercellular communication. In particular, dendritic cells-derived extracellular vesicles (DC-EVs) facilitate the transfer of critical components, like antigens and immune-regulatory factors, and due to the expression of major histocompatibility complexes and co-stimulatory molecules on their surface can activate T cells, thereby modulating the immune response. Additionally, DC-EVs can be engineered to transport tumor-specific antigens, cytokines, or other agents in order to strength their immunotherapeutic potential, and even be used in vaccines formulation. In this review, the latest advancements in engineering DC-EVs to improve their immunotherapeutic potential is discussed in detail, while also addressing current challenges associated with DC-EVs therapies.
Collapse
Affiliation(s)
- Margarida Oliveira Coelho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sofia Torres Quintas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; IUCS-CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Olivier De Wever
- CRIG - Cancer Research Institute Ghent, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium; LECR - Laboratory Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal.
| |
Collapse
|