201
|
Zhang T, Nunes ADC, Lee J, Larocca D, Camussi G, Lim SK, Bascones VU, Angelini L, O'Kelly RD, Dong X, Niedernhofer LJ, Robbins PD. Identification of Senomorphic miRNAs in Embryonic Progenitor and Adult Stem Cell-Derived Extracellular Vesicles. Aging Cell 2025:e70071. [PMID: 40275616 DOI: 10.1111/acel.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Extracellular vesicles (EVs) are secreted by most cell types, transmitting crucial signaling molecules like proteins, small RNAs, and DNA. We previously demonstrated that EVs from murine and human mesenchymal stem cells (MSCs) functioned as senomorphics to suppress markers of senescence and the inflammatory senescence-associated secretory phenotype (SASP) in cell culture and in aged mice. Here we demonstrate that EVs from additional types of human adult stem cells and embryonic progenitor cells have a senomorphic activity. Based on their miRNA profiles showing prevalence in stem cell EVs versus nonstem cell EVs and the number of age-related genes targeted, we identified eight miRNAs as potential senomorphic miRNAs. Analysis of these miRNAs by transfection into etoposide-induced senescent IMR90 human fibroblasts revealed that each of the miRNAs alone regulated specific senescence and SASP markers, but none had complete senomorphic activity. Evaluation of ~300 combinations of miRNAs for senotherapeutic activity identified a senomorphic cocktail of miR-181a-5p, miR-92a-3p, miR-21-5p, and miR-186-5p that markedly reduced the expression of p16INK4a, p21Cip1, IL-1β, and IL-6 and the percentage of SA-ß-gal-positive cells. Transcriptome analysis identified multiple pathways affected by the miRNA cocktail, including cellular senescence and inhibition of PCAF and HIPK2 in the p53 signaling pathway. Finally, treatment of aged mice with liposomes containing the four miRNA cocktail suppressed markers of senescence and inflammation in multiple tissues. These studies suggest that EVs derived from stem cells suppress senescence and inflammation, at least in part, through miRNAs and that a senomorphic miRNA cocktail could be used to target senescence and inflammation to extend health span.
Collapse
Affiliation(s)
- Tianpeng Zhang
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Allancer D C Nunes
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jieun Lee
- AgeX Therapeutics, Inc., Alameda, California, USA
| | - Dana Larocca
- AgeX Therapeutics, Inc., Alameda, California, USA
| | - Giovanni Camussi
- Department of Medical Science, University of Torino, Turin, Italy
| | - Sai Kiang Lim
- Institute of Medical Biology, ASTAR, Singapore, Singapore
| | - Vicky U Bascones
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Luise Angelini
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ryan D O'Kelly
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xiao Dong
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul D Robbins
- Masonic Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
202
|
Taghdi MH, Al-Masawa ME, Muttiah B, Fauzi MB, Law JX, Zainuddin AA, Lokanathan Y. Three-Dimensional Bioprinted Gelatin-Genipin Hydrogels Enriched with hUCMSC-Derived Small Extracellular Vesicles for Regenerative Wound Dressings. Polymers (Basel) 2025; 17:1163. [PMID: 40362948 PMCID: PMC12073717 DOI: 10.3390/polym17091163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEVs) have shown great promise in promoting tissue repair, including skin wound healing, but challenges like rapid degradation and short retention have limited their clinical application. Hydrogels have emerged as effective carriers for sustained EV release. Three-dimensional printing enables the development of personalized skin substitutes tailored to the wound size and shape. This study aimed to develop 3D bioprinted gelatin-genipin hydrogels incorporating human umbilical cord MSC-sEVs (hUCMSC-sEVs) for future skin wound healing applications. Gelatin hydrogels (8% and 10% w/v) were crosslinked with 0.3% genipin (GECL) to improve stability. The hydrogels were evaluated for their suitability for extrusion-based 3D bioprinting and physicochemical properties, such as the swelling ratio, hydrophilicity, enzymatic degradation, and water vapor transmission rate (WVTR). Chemical characterization was performed using EDX, XRD, and FTIR. The hUCMSC-sEVs were isolated via centrifugation and tangential flow filtration (TFF) and characterized. The crosslinked hydrogels were successfully 3D bioprinted and demonstrated superior properties, including high hydrophilicity, a swelling ratio of ~500%, slower degradation, and optimal WVTR. hUCMSC-sEVs, ranging from 50 to 200 nm, were positive for surface and cytosolic markers. Adding 75 μg/mL of hUCMSC-EVs into 10% GECL hydrogels significantly improved the biocompatibility. These hydrogels offer ideal properties for 3D bioprinting and wound healing, demonstrating their potential as biomaterial scaffolds for skin tissue regeneration applications.
Collapse
Affiliation(s)
- Manal Hussein Taghdi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
- Department of Anesthesia and Intensive Care, Faculty of Medical Technology, University of Tripoli, Tripoli P.O. Box 13932, Libya
| | - Maimonah Eissa Al-Masawa
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
| | - Barathan Muttiah
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
| | - Ani Amelia Zainuddin
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (M.H.T.); (M.E.A.-M.); (B.M.); (M.B.F.); (J.X.L.)
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| |
Collapse
|
203
|
Zhdanova DY, Bobkova NV, Chaplygina AV, Svirshchevskaya EV, Poltavtseva RA, Vodennikova AA, Chernyshev VS, Sukhikh GT. Effect of Small Extracellular Vesicles Produced by Mesenchymal Stem Cells on 5xFAD Mice Hippocampal Cultures. Int J Mol Sci 2025; 26:4026. [PMID: 40362265 PMCID: PMC12071690 DOI: 10.3390/ijms26094026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is one of the most common progressive neurodegenerative diseases leading to impairments in memory, orientation, and behavior. However, significant work is still needed to fully understand the progression of such disease and develop novel therapeutic agents for AD prevention and treatment. Small extracellular vesicles (sEVs) have received attention in recent years due to their potential therapeutic effects on AD. The aim of this study was to determine the potential effect of sEVs in an in vitro model of AD. sEVs were isolated from human Wharton's jelly mesenchymal stem cells (MSCs) by asymmetric depth filtration, a method developed recently by us. AD was modeled in vitro using cells obtained from the hippocampi of newborn 5xFAD transgenic mice carrying mutations involved in familial AD. After isolation, sEVs underwent detailed characterization that included scanning electron microscopy, nanoparticle tracking analysis, confocal microscopy, Western blotting, and Luminex assay. When added to 5xFAD hippocampal cells, sEVs were nontoxic, colocalized with neurons and astrocytes, decreased the level of Aβ peptide, and increased the synaptic density. These results support the possibility that sEVs can improve brain cell function during aging, decrease the risk of AD, and potentially be used for AD therapeutics.
Collapse
Affiliation(s)
- Daria Y. Zhdanova
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Natalia V. Bobkova
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Alina V. Chaplygina
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Elena V. Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ulitsa M0iklukho-Maklaya 16/10, 117997 Moscow, Russia;
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| | - Rimma A. Poltavtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| | - Anastasia A. Vodennikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ulitsa M0iklukho-Maklaya 16/10, 117997 Moscow, Russia;
- Institute of Bioorganic Chemistry, National Research Nuclear University “MEPhI”, Kashirskoe Shosse 31, 115409 Moscow, Russia
| | - Vasiliy S. Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, 121205 Moscow, Russia
| | - Gennadiy T. Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| |
Collapse
|
204
|
Abyadeh M, Kaya A. Multiomics from Alzheimer's Brains and Mesenchymal Stem Cell-Derived Extracellular Vesicles Identifies Therapeutic Potential of Specific Subpopulations to Target Mitochondrial Proteostasis. J Cent Nerv Syst Dis 2025; 17:11795735251336302. [PMID: 40297324 PMCID: PMC12035200 DOI: 10.1177/11795735251336302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Background Alzheimer's disease (AD) is characterized by complex molecular alterations that complicate its pathogenesis and contribute to the lack of effective treatments. Mesenchymal stem cell-derived extracellular vesicles (EVs) have shown promise in AD models, but results across different EV subpopulations remain inconsistent. Objectives This study investigates proteomic and transcriptomic data from publicly available postmortem AD brain datasets to identify molecular changes at both the gene and protein levels. These findings are then compared with the proteomes of various EV subpopulations, differing in size and distribution, to determine the most promising subtype for compensating molecular degeneration in AD. Design We conducted a comprehensive analysis of 788 brain samples, including 481 AD cases and 307 healthy controls, examining protein and mRNA levels to uncover AD-associated molecular changes. These findings were then compared with the proteomes of different EV subpopulations to identify potential therapeutic candidates. Methods A multi-omics approach was employed, integrating proteomic and transcriptomic data analysis, miRNA and transcription factor profiling, protein-protein network construction, hub gene identification, and enrichment analyses. This approach aimed to explore molecular changes in AD brains and pinpoint the most relevant EV subpopulations for therapeutic intervention. Results We identified common alterations in the cAMP signaling pathway and coagulation cascade at both the protein and mRNA levels. Distinct changes in energy metabolism were observed at the protein level but not at the mRNA level. A specific EV subtype, characterized by a broader size distribution obtained through high-speed centrifugation, was identified as capable of compensating for dysregulated mitochondrial proteostasis in AD brains. Network biology analyses further highlighted potential regulators of key therapeutic proteins within this EV subtype. Conclusion This study underscores the critical role of proteomic alterations in AD and identifies a promising EV subpopulation, enriched with proteins targeting mitochondrial proteostasis, as a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Alaattin Kaya
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
205
|
Guo MK, Scipione CA, Breda LCD, Prajapati K, Raju S, Botts SR, Abdul-Samad M, Patel S, Yu G, Dudley AC, Fish JE, Howe KL. Tracking Endothelial Extracellular Vesicles in a Mouse Model of Atherosclerosis. Circ Res 2025. [PMID: 40265255 DOI: 10.1161/circresaha.124.326024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Affiliation(s)
- Mandy Kunze Guo
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
| | - Corey A Scipione
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Leandro C D Breda
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Kamalben Prajapati
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
| | - Sneha Raju
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
- Division of Vascular Surgery, Department of Surgery, University of Toronto, ON, Canada. (S.R., K.L.H.)
| | - Steven R Botts
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
| | - Majed Abdul-Samad
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
| | - Sarvatit Patel
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Garry Yu
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
| | - Andrew C Dudley
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville (A.C.D.)
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
- Peter Munk Cardiac Centre, Toronto General Hospital, ON, Canada (J.E.F., K.L.H.)
| | - Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, ON, Canada (M.K.G., C.A.S., L.C.D.B., K.P., S.R., S.R.B., M.A.-S., S.P., G.Y., J.E.F., K.L.H.)
- Institute of Medical Sciences, University of Toronto, ON, Canada. (M.K.G., S.R., S.R.B., J.E.F., K.L.H.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada. (K.P., M.A.-S., J.E.F., K.L.H.)
- Division of Vascular Surgery, Department of Surgery, University of Toronto, ON, Canada. (S.R., K.L.H.)
- Peter Munk Cardiac Centre, Toronto General Hospital, ON, Canada (J.E.F., K.L.H.)
| |
Collapse
|
206
|
Ielo C, Breccia M. Extracellular vesicles as source of biomarkers in hematological malignancies: looking towards clinical applications. Expert Rev Mol Diagn 2025:1-12. [PMID: 40178353 DOI: 10.1080/14737159.2025.2488919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/01/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION Extracellular vesicles are membranous particles released by cells in physiological and pathological conditions. Their cargo is heterogeneous since it includes different biomolecules such as nucleic acids and proteins. Oncogenic alterations affect the composition of extracellular vesicles and model their content during cancer evolution. AREAS COVERED This review provides an overview of the studies focused on extracellular vesicles as source of biomarkers in hematological malignancies. A special insight into extracellular vesicles-derived biomarkers as tools for evaluating the prognosis of hematological malignancies and their response to treatment is given. EXPERT OPINION Extracellular vesicles are a valuable source of biomarkers in hematological malignancies. However, the translation from the bench to the bedside is challenged by the lack of standardization of the preanalytical variables of the experimental workflow. The release of standard operating procedures and the validation of the extracellular vesicles-derived biomarkers in large cohort of patients will help in exploiting the potential of extracellular vesicles in the clinical setting.
Collapse
Affiliation(s)
- Claudia Ielo
- Department of Translational and Precision Medicine, Sapienza University of Rome - Azienda Policlinico Umberto I, Rome, Italy
| | - Massimo Breccia
- Department of Translational and Precision Medicine, Sapienza University of Rome - Azienda Policlinico Umberto I, Rome, Italy
| |
Collapse
|
207
|
Gilboa T, Ter-Ovanesyan D, Babila CM, Whiteman S, Morton S, Kalish D, Johnston J, Tesin D, Davies M, Tam JM, Church GM, Walt DR. High-Throughput Extracellular Vesicle Isolation Using Plate-Based Size Exclusion Chromatography and Automation. J Am Chem Soc 2025; 147:13258-13263. [PMID: 40198271 DOI: 10.1021/jacs.4c17948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Extracellular vesicles (EVs) are natural, cell-derived nanoparticles released into biofluids, such as plasma, and hold great potential as a new class of biomarkers. However, the utility of analyzing EVs in clinical samples has been hampered by a lack of suitable EV isolation methods that can be performed reproducibly in a scalable manner. The current method of choice for isolating EVs, size exclusion chromatography (SEC), is performed manually one column at a time, and thus does not have the throughput for isolating EVs from clinical samples. In this work, we adapt SEC to a plate-based format to increase its throughput. We show that SEC can be performed using plates containing frits packed with resin, where each well of a 24-well plate can be used for a different sample. By measuring EV markers CD63 and CD81, as well as Albumin as a representative free protein, we optimize the separation of EVs from free proteins in the 24-well format. We also demonstrate that performing SEC in these plates can be automated using liquid handling platforms with the use of custom adapters. We quantify the high reproducibility of this automated platform and then apply the platform to analyze the tetraspanins CD63 and CD81 across individuals. Our work represents a solution to the long-standing challenge in the EV biomarker field of reproducible high-throughput EV isolation from plasma and other biofluids. We envision that the automated methods we have developed will scale SEC to hundreds of samples per day, enabling the use of EVs for biomarker discovery and diagnostics.
Collapse
Affiliation(s)
- Tal Gilboa
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
| | - Dmitry Ter-Ovanesyan
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - Clarissa May Babila
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - Sara Whiteman
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - Shad Morton
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | - David Kalish
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
| | | | | | | | - Jenny M Tam
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| | - George M Church
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| | - David R Walt
- Wyss Institute for Biologically Inspired Engineering, 201 Brookline Ave., Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
208
|
Padinharayil H, Varghese J, Varghese PR, Wilson CM, George A. Small Extracellular Vesicle (sEV) Uptake from Lung Adenocarcinoma and Squamous Cell Carcinoma Alters T-Cell Cytokine Expression and Modulates Protein Profiles in sEV Biogenesis. Proteomes 2025; 13:15. [PMID: 40407494 PMCID: PMC12101295 DOI: 10.3390/proteomes13020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Despite advances in immunotherapy, non-small-cell lung carcinoma (NSCLC)'s clinical success is limited, possibly due to substantial immunological alterations in advanced cancer patients. This study examines the immunomodulatory effects of sEVs derived from lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC) on T cells. METHODS SEVs were isolated from lung cancer cell lines and Jurkat-E6.1. SEV size and morphology were analyzed by NTA and TEM, respectively, while Western blotting confirmed sEV markers. SEV uptake was assessed, followed by resazurin assay, RNA isolation, quantification, cDNA preparation, RT-PCR, nano LC-MS, and bioinformatic analysis, before and after treating Jurkat-E6.1 cells with sEVs from A549 and SKMES1. RESULTS Cancer-derived sEVs were efficiently internalized by immune cells, reducing T-cell viability. The real-time PCR analysis showed downregulation of KI67, BCL2, BAX, TNFA, IL6, TGFβ, and IL10, suggesting reduced proliferation, dysregulated apoptosis, and impaired inflammatory and immunosuppressive signaling, and the upregulation of GZMB and IL2 suggests retained cytotoxic potential but possibly dysfunctional T-cell activation. Proteomic analysis revealed 39 differentially abundant proteins (DAPs) in ADC-treated T cells and 276 in SCC-treated T cells, with 19 shared DAPs. Gene Ontology (GO) analysis of these DAPs highlighted processes such as sEV biogenesis, metabolic pathways, and regulatory functions, with ADC sEVs influencing NAD metabolism, ECM binding, and oxidoreductase activity, while SCC sEVs affected mRNA stability, amino acid metabolism, and cadherin binding. The cytoplasmic colocalization suggests the presence of these proteins in the cellular and extracellular lumen, indicating the potential of further release of these proteins in the vesicles by T cells. CONCLUSION Lung cancer-derived sEVs regulate T-cell activities through immunoregulatory signaling. The molecular interactions between sEVs and immune cells can reveal novel tumor immune regulatory mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Mission Medical College & Research Institute, Thrissur 680005, Kerala, India; (H.P.); (P.R.V.)
- Department of Zoology, St. Thomas College, Kozhencherry, Pathanamthitta 689641, Kerala, India;
| | - Jinsu Varghese
- Department of Zoology, St. Thomas College, Kozhencherry, Pathanamthitta 689641, Kerala, India;
| | | | - Cornelia M. Wilson
- School of Psychology and Life Sciences, Canterbury Christ Church University, Kent CT1 1QU, UK
| | - Alex George
- Jubilee Mission Medical College & Research Institute, Thrissur 680005, Kerala, India; (H.P.); (P.R.V.)
| |
Collapse
|
209
|
Rasool GS, Shihab EM, Al-Bahrani MH, Al-Musawi MH, Malek Mohammadi Nouri K, Mehdinezhad Roshan M, Hajipour H. Enhanced endometrial receptivity via epigallocatechin gallate (EGCG)-loaded menstrual blood-derived exosomes. J Pharm Sci 2025; 114:103801. [PMID: 40280485 DOI: 10.1016/j.xphs.2025.103801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Embryo implantation failure is a significant challenge in infertility treatment, accounting for a substantial number of treatment failures. Increasing endometrial receptivity can potentially overcome this issue. This study aims to introduce a novel approach for enhancing endometrial receptivity by preparing epigallocatechin gallate (EGCG)-loaded menstrual blood-derived exosomes. Menstrual blood was used to isolate exosomes, which were then characterized for their size, zeta potential, morphology, and surface markers. EGCG was loaded into the isolated exosomes using sonication. The effects of EGCG-loaded exosomes on the adhesion ability of endometrial cells and the expression of endometrial receptivity-related genes were evaluated using in vitro implantation assays and real-time PCR, respectively. As results, exosomes with an average size of 86.2 nm, a surface charge of -11.8 mV, spherical morphology, and positive for CD9 and CD81 surface markers were successfully isolated. EGCG was loaded into exosomes with an encapsulation efficiency of 65.18 %. The in vitro implantation assay confirmed that EGCG-loaded exosomes had a greater potential to enhance the adhesion ability of endometrial cells compared to free EGCG. Furthermore, EGCG-loaded exosomes upregulated the expression of Leukemia inhibitory factor, homeobox A10, and integrin beta 3 genes more potently compared to free EGCG. These findings suggest that EGCG-loaded exosomes could be a therapeutic option for low endometrial receptivity. Moreover, menstrual blood-derived exosomes appear to be a promising drug delivery system for endometrial cells, offering a potential solution to the therapeutic limitations of the payload.
Collapse
Affiliation(s)
- Ghada S Rasool
- Department of Anatomy, Faculty of Medicine, Nineveh University, Mosul, Iraq
| | - Elaf Mahmood Shihab
- Department of Pharmacology and Toxicology Collage of Pharmacy, Al-Esraa university, Baghdad, Iraq
| | - Maha Hameed Al-Bahrani
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Baghdad, Iraq
| | - Mastafa H Al-Musawi
- Department of Biology, College of Science, Mustansiriyah University, Baghdad, Iraq
| | | | - Mehdi Mehdinezhad Roshan
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Hajipour
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
210
|
Pham QN, Milanova V, Tung TT, Losic D, Thierry B, Winter MA. Affinity enrichment of placental extracellular vesicles from minimally processed maternal plasma with magnetic nanowires. Analyst 2025; 150:1908-1919. [PMID: 40172922 DOI: 10.1039/d4an01414f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Affinity based enrichment of cell/tissue specific extracellular vesicles (EVs) with magnetic materials and analysis of their molecular cargo has the potential to improve assay sensitivity/specificity compared to whole plasma analysis. For example, syncytiotrophoblast EVs (STBEVs) shed from the placenta during pregnancy carry placental diagnostic markers relevant to pregnancy complications linked to placental insufficiency such as placental alkaline phosphatase (PLAP), Neprilysin (NEP) and Placental Protein 13 (PP13). However, the need for sample pre-enrichment of EVs from plasma adds significant complexity, time and cost. We report an affinity-based cell/tissue specific EV enrichment direct from plasma based on iron-oxide magnetic nanowires (NWs) coated with reversible-addition-fragmentation-chain-transfer (RAFT) polymers and conjugated with anti-PLAP antibodies. As anticipated the complex protein environment of minimally processed plasma significantly decreased STBEV enrichment yield. However, an optimized RAFT polymeric coating successfully mitigated the detrimental effect of the protein corona, yielding significantly improved STBEV recovery compared to Dynabeads™ in unenriched diluted plasma. Despite the presence of significant soluble PLAP protein, STBEV enrichment could be performed directly from the plasma of pregnant women (including preeclamptic samples) within 1.5 hours, enabling quantification of two placental protein markers PP13 and NEP with known diagnostic relevance to preeclampsia. Direct affinity-enrichment of STBEVs with high performance magnetic materials has the potential to underpin rapid clinical diagnostic assays for preeclampsia and related pregnancy complications.
Collapse
Affiliation(s)
- Quang Nghia Pham
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Valentina Milanova
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Tran Thanh Tung
- School of Chemical Engineering, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Dusan Losic
- School of Chemical Engineering, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| | - Marnie A Winter
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, Adelaide, South Australia 5095, Australia.
| |
Collapse
|
211
|
Zhu LL, Li LD, Lin XY, Hu J, Wang C, Wang YJ, Zhou QG, Zhang J. Plasma-Derived Small Extracellular Vesicles miR- 182 - 5p Is a Potential Biomarker for Diagnosing Major Depressive Disorder. Mol Neurobiol 2025:10.1007/s12035-025-04948-9. [PMID: 40261603 DOI: 10.1007/s12035-025-04948-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/12/2025] [Indexed: 04/24/2025]
Abstract
Depression, particularly major depressive disorder (MDD), is a debilitating neuropsychiatric condition characterized by high disability rates, primarily driven by chronic stress and genetic predispositions. Emerging evidence highlights the critical role of microRNAs (miRNAs) in the pathogenesis of depression, with plasma-derived small extracellular vesicles (sEVs) emerging as promising biomarkers. In this study, we collected peripheral blood plasma samples from patients diagnosed with MDD, as assessed by the Hamilton Depression Rating scale, alongside healthy individuals serving as controls. Plasma-derived sEVs were isolated via ultracentrifugation, followed by high-throughput sequencing of miRNAs encapsulated within sEVs, and finally image acquisition and differential expression analysis. Our results revealed a significant elevation of miR-182-5p in plasma-derived sEVs from MDD patients compared to healthy controls, a finding further validated in chronic mild stress (CMS) models. Further analysis suggested that miRNAs encapsulated within sEVs may influence depression onset and progression by modulating hypothalamic-pituitary-adrenal (HPA) axis activity. These findings underscore the potential of miRNAs and their target genes as novel biomarkers, offering improved diagnostic accuracy and therapeutic efficacy for MDD.
Collapse
Affiliation(s)
- Lin-Lin Zhu
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
| | - Lian-Di Li
- Anhui Institute for Food and Drug Control, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Xuan-Yu Lin
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
| | - Jian Hu
- The Second Affiliated Hospital of Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Chun Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yi-Jun Wang
- The Second Affiliated Hospital of Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210009, Jiangsu, China
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211167, Jiangsu Province, China.
| | - Jing Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
| |
Collapse
|
212
|
Lee J, Kwon K, Cho MJ, Son T, Roh Y, Lee S, Kim DS, Lee MS, Ban HS, Kim JS, Lim EK, Lee SH, Oh GT, Park JG, Han TS. Fusogenic Nanoreactor-Based Detection of Extracellular Vesicle-derived miRNAs for Diagnosing Atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501789. [PMID: 40254988 DOI: 10.1002/smll.202501789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Extracellular vesicle (EV) microRNAs (miRNAs) are critical liquid-biopsy biomarkers that facilitate noninvasive clinical diagnosis and disease monitoring. However, conventional methods for detecting these miRNAs require EV lysis, which is expensive, labor-intensive, and time-consuming. Inspired by natural viral infection mechanisms, a novel strategy is developed for detecting EV miRNAs in situ via vesicle fusion mediated by viral fusion proteins. A padlock probe encapsulated within fusogenic liposomes is activated by target miRNAs, thereby initiating a highly sensitive and specific rolling circle amplification (RCA) reaction. Three EV miRNAs associated with atherosclerosis are successfully analyzed using this method, thereby enabling clear differentiation of healthy and diseased mice at several disease stages. Overall, the developed platform offers a simple approach for detecting EV miRNAs and demonstrates significant potential for broad use in applications involving disease diagnosis and monitoring.
Collapse
Affiliation(s)
- Jiyoon Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Kiyoon Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Min Ji Cho
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Taesang Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Yuna Roh
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Sugi Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dae-Soo Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Moo-Seung Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Hyun Seung Ban
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jang-Seong Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Eun-Kyung Lim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Sang-Hak Lee
- Division of Cardiology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Goo Taeg Oh
- Heart-Immune-Brain Network Research Center, Department of Life Science and College of Natural Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Jong-Gil Park
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Tae-Su Han
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- KRIBB School, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- School of Medicine, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
213
|
Ozerklig B, Turkel I, Yilmaz M, Vaizoglu RD, Akan HS, Dikmen ZG, Saleem A, Kosar SN. Exercise-induced extracellular vesicles mediate apoptosis in human colon cancer cells in an exercise intensity-dependent manner. Eur J Appl Physiol 2025:10.1007/s00421-025-05787-1. [PMID: 40253655 DOI: 10.1007/s00421-025-05787-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/26/2025] [Indexed: 04/22/2025]
Abstract
Regular exercise reduces the incidence and improves the prognosis of many cancer types, but the underlying mechanisms remain elusive. Evidence suggests that exercise exerts its therapeutic effects through extracellular vesicles (EVs), which are essential for cellular communication. Here, we hypothesized that exercise-induced EVs from serum of healthy individuals would exert anti-tumorigenic effects on human colon cancer HT-29 cells, in an exercise intensity-dependent manner. Ten healthy young active males participated in a randomized crossover trial, completing two workload-matched acute exercise bouts, moderate-intensity continuous exercise (MICE) and high-intensity interval exercise (HIIE), on a cycle ergometer. A control session of rest (PRE) was included. EVs were isolated from serum samples collected during PRE and immediately after each exercise session. EVs were co-incubated with HT-29 colon cancer cells, and the effects on cell viability, migration, and apoptosis were measured. EV treatment reduced cell viability in all groups (PRE, MICE, and HIIE) by 35%, 43% and 47%, respectively, vs. PBS. HIIE-EVs showed a significantly greater reduction in cell viability vs. PRE; therefore, only these groups were used for further analysis. PRE EVs reduced migration by 27%, and HIIE-EVs by 39%. HIIE-EVs increased expression of pro-apoptotic markers: Bax/Bcl-2 ratio by 56% and Caspase 3 by 30% vs. PBS, with no change observed in the PRE group. Further, 16% of cells in PRE and 28% of cells in HIIE were TUNEL-positive, indicating DNA fragmentation. To our knowledge, this is the first human study that illustrates the therapeutic potential of exercise-induced EVs in cancer treatment.
Collapse
Affiliation(s)
- Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye.
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada.
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada.
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| | - Merve Yilmaz
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Refika Dilara Vaizoglu
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Handan Sevim Akan
- Department of Biology, Faculty of Science, Hacettepe University, Ankara, Türkiye
| | - Z Gunnur Dikmen
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Türkiye
| | - Ayesha Saleem
- Faculty of Kinesiology and Recreation Management, University of Manitoba, Winnipeg, Canada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, Canada
| | - Sukran Nazan Kosar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
214
|
Lei X, Ring S, Jin S, Singh S, Mahnke K. Extracellular Vesicles and Their Role in Skin Inflammatory Diseases: From Pathogenesis to Therapy. Int J Mol Sci 2025; 26:3827. [PMID: 40332512 PMCID: PMC12027629 DOI: 10.3390/ijms26083827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, are released into the extracellular space by almost all known cell types. They facilitate communication between cells by transferring bioactive molecules, which impact both physiological processes and the development of diseases. EVs play a crucial role in the pathogenesis of various diseases by participating in multiple pathological processes. They contribute to disease progression by triggering cytokine release, modulating immune cell activity, and inducing inflammatory and immune responses. Beyond their pathological implications, EVs also offer significant therapeutic potential. Both natural and engineered EVs show great potential in the fields of targeted therapy, drug delivery, and immune modulation in dermatological applications. The development of EV-based treatments is showing promise in advancing patient outcomes, particularly in chronic inflammatory and immune-mediated skin conditions. This review comprehensively examined the biogenesis, classification, and functional roles of EVs, including advanced methods for their isolation and characterization. Furthermore, we summarized recent studies highlighting the involvement of EVs in four major inflammatory skin diseases: psoriasis, atopic dermatitis, systemic lupus erythematosus, and wound healing.
Collapse
Affiliation(s)
| | | | | | | | - Karsten Mahnke
- Department of Dermatology, University Hospital Heidelberg, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany; (X.L.); (S.R.); (S.J.); (S.S.)
| |
Collapse
|
215
|
Panzetta G, Schirizzi A, Balestra F, De Luca M, Depalo N, Rizzi F, Ricci AD, De Leonardis G, Lotesoriere C, Giannelli G, D’Alessandro R, Scavo MP. Unravelling Paclitaxel Resistance in Gastric Cancer: The Role of Small Extracellular Vesicles in Epithelial Mesenchymal Transition and Extracellular Matrix Remodelling. Cancers (Basel) 2025; 17:1360. [PMID: 40282535 PMCID: PMC12025963 DOI: 10.3390/cancers17081360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Gastric cancer (GC) is a highly aggressive disease often complicated by resistance to chemotherapy agents like paclitaxel (PTX), which targets microtubules to induce apoptosis. Resistance arises through complex molecular mechanisms, including the overexpression of pro-angiogenic factors (VEGFA, ANG-2), activation of survival pathways (PDGFRβ, PPARγ), and epithelial-mesenchymal transition (EMT) driven by proteins such as VIM, E-CAD, N-CAD, and FLOT-1. The extracellular matrix (ECM), regulated by COL1A1 and influenced by PPARγ, acts as a physical barrier to drug penetration. Small extracellular vesicles (sEVs) have emerged as critical mediators of intercellular communication and may influence these resistance pathways. Methods: This study investigated the role of sEVs isolated from metastatic GC patients treated with Ramucirumab and PTX. Patients were stratified by progression-free survival (PFS) into rapidly progressing (RP) and controlled disease (CD) groups. sEVs from these patients were applied to HCEC-1CT and HEPA-RG cell lines. Cell viability assays, gene and protein expression analyses, and bioinformatic studies were conducted to assess the impact of sEVs on resistance-related markers. Results: Results showed that sEVs from CD patients reduced the expression of markers associated with drug resistance, while sEVs from RP patients increased these markers, promoting angiogenesis, EMT, and ECM remodeling. These changes correlated with enhanced cell survival and resistance phenotypes. Bioinformatic analyses confirmed that sEVs modulate inflammation, ECM dynamics, and EMT pathways. Conclusions: In conclusion, sEVs from metastatic GC patients significantly influence chemoresistance and tumor progression. Targeting sEV-mediated signaling may offer novel therapeutic strategies to overcome resistance and improve treatment outcomes in gastric cancer.
Collapse
Affiliation(s)
- Giorgia Panzetta
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.S.); (G.D.L.)
| | - Francesco Balestra
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| | - Maria De Luca
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| | - Nicoletta Depalo
- Institute for Chemical-Physical Processes, Italian National Research Council (IPCF)-CNR SS Bari, Via Orabona, 70125 Bari, Italy; (N.D.); (F.R.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Bari Research Unit, Via Orabona 4, 70126 Bari, Italy
| | - Federica Rizzi
- Institute for Chemical-Physical Processes, Italian National Research Council (IPCF)-CNR SS Bari, Via Orabona, 70125 Bari, Italy; (N.D.); (F.R.)
- National Interuniversity Consortium of Materials Science and Technology (INSTM), Bari Research Unit, Via Orabona 4, 70126 Bari, Italy
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.D.R.); (C.L.)
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.S.); (G.D.L.)
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.D.R.); (C.L.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy;
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (A.S.); (G.D.L.)
| | - Maria Principia Scavo
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy; (G.P.); (F.B.); (M.D.L.)
| |
Collapse
|
216
|
Chen W, Zhang Y, Chen J, Dong S, Wu X, Wu Y, Du Z, Yang Y, Gong L, Yu J. Heme Oxygenase-1 Modulates Macrophage Polarization Through Endothelial Exosomal miR-184-3p and Reduces Sepsis-Induce Lung Injury. Int J Nanomedicine 2025; 20:5039-5057. [PMID: 40264818 PMCID: PMC12013636 DOI: 10.2147/ijn.s506830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Pulmonary microvascular endothelial cells (PMVECs) are notably implicated in the pathogenesis of sepsis-induced lung injury. Exosomes derived from PMVECs facilitate intercellular communication among various cell types, especially crosstalk with macrophages. Heme oxygenase-1 (HO-1), an early stress-responsive enzyme with inherent protective functions, has been implicated in acute lung injury (ALI) mitigation. But research on the mechanism of HO-1 in macrophage polarization via PMVEC exosomes in sepsis-induced lung injury is lacking. Methods To investigate the role of HO-1 in the interaction between endothelial cells and macrophages, HO-1 knockout mouse model were established. Exosomes from PMVECs were isolated, and differential expression of microRNA (miRNA) was determined by sequencing. An in vitro co-culture system involving Murine Alveolar Macrophage Cell Line (MH-S cells) and HO-1/ PMVECs-derived exosomes (HP-exos) was used to investigate the underlying mechanisms. To further verify the involvement of HO-1 in intercellular communication through exosomal miRNA in vivo, the level of pulmonary inflammation was evaluated, and the polarization of pulmonary macrophages was analyzed. Results The results showed that miR-184-3p was significantly downregulated in HP-exos, and supplementation of miR-184-3p enhanced the polarization of M1 macrophages, thus intensifying lung inflammation. HO-1 regulates the polarization of macrophages by regulating endothelial exosomes. Overexpression of HO-1 downregulates miR-184-3p, which negatively regulates Semaphorin 7A (Sema7a), which attenuated M1 type macrophages (M1) polarization and augmented M2 type macrophages (M2) polarization, thereby partially mitigating lung injury and inflammation. Conclusion Collectively, we elucidated a novel potential therapeutic mechanism that HO-1 alleviate inflammation by modulating the M1/M2 ratio in sepsis-induced ALI by regulating miR-184-3p/Sema7a expression.
Collapse
Affiliation(s)
- Wei Chen
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Yuan Zhang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| | - Jinkun Chen
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Shuan Dong
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| | - Xiaoyang Wu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Ya Wu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Zhuo Du
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Yibo Yang
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
| | - Lirong Gong
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| | - Jianbo Yu
- Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, People’s Republic of China
- Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin, 300100, People’s Republic of China
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin, 300100, People’s Republic of China
| |
Collapse
|
217
|
Kim H, Lee J, Qian A, Ji YR, Zhang R, Hu Q, Williams CK, Chuang HY, Smalley MD, Xu Y, Gao L, Mayo MC, Zhang T, Posadas EM, Tan ZS, Vinters HV, Vossel K, Magaki S, Zhu Y, Tseng HR. Noninvasive Assessment of β-Secretase Activity Through Click Chemistry-Mediated Enrichment of Neuronal Extracellular Vesicles to Detect Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415289. [PMID: 40245252 DOI: 10.1002/advs.202415289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/27/2025] [Indexed: 04/19/2025]
Abstract
Alzheimer's disease (AD), the most prevalent type of dementia, is characterized by a biological process that begins with the development of AD neuropathologic change (ADNPC) while individuals remain asymptomatic. A key molecular hallmark of ADNPC is the accumulation of amyloid-β plaques. β-secretase plays a critical role in the upstream pathological cleavage of amyloid precursor protein (APP), producing amyloid-β peptides that are prone to misfolding, ultimately contributing to plaque formation. Neuronal extracellular vesicles (NEVs) in the blood transport β-secretase and preserve its activity, allowing for noninvasive profiling of β-secretase activity for detecting early onset of ADNPC. In this study, a novel approach is approached for noninvasive assessment of β-secretase activity in AD patients using an NEV β-secretase activity assay. This assay identifies NEVs exhibiting colocalization of NEV markers with AD-associated β-secretase, generating a β-secretase activity profile for each patient. The NEV β-secretase activity assay represents a significant advancement in leveraging the diagnostic potential of NEVs, offering a noninvasive, quantitative method for reliably assessing β-secretase activity to detect the early onset of ADNPC.
Collapse
Affiliation(s)
- Hyoyong Kim
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Junseok Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Audrey Qian
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - You-Ren Ji
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Ryan Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Qixin Hu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Christopher Kazu Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Han-Yu Chuang
- Eximius Diagnostics Corp, Magnify Incubator, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Matthew D Smalley
- Eximius Diagnostics Corp, Magnify Incubator, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Yaya Xu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Liang Gao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Mary C Mayo
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ting Zhang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Edwin M Posadas
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Zaldy S Tan
- Departments of Neurology and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Keith Vossel
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| |
Collapse
|
218
|
Zhou X, Huang J, Zhang D, Qian Z, Zuo X, Sun Y. Small extracellular vesicles: the origins, current status, future prospects, and applications. Stem Cell Res Ther 2025; 16:184. [PMID: 40247402 PMCID: PMC12004682 DOI: 10.1186/s13287-025-04330-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Small extracellular vesicles (sEVs) are membrane-bound vesicles with a size of less than 200 nm, released by cells. Due to their relatively small molecular weight and ability to participate in intercellular communication, sEVs can serve not only as carriers of biomarkers for disease diagnosis but also as effective drug delivery agents. Furthermore, these vesicles are involved in regulating the onset and progression of various diseases, reflecting the physiological and functional states of cells. This paper introduces the classification of extracellular vesicles, with a focus on the extraction and identification of sEVs and their significant role in repair, diagnosis, and intercellular communication. Additionally, the paper addresses the engineering modification of sEVs to provide a reference for enhanced understanding and application.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Clinical Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Jin Huang
- Department of Geriatrics, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dianqi Zhang
- Department of Central Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Zhenyu Qian
- Department of Neurology, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Xin Zuo
- Department of Geriatrics, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China.
| | - Yaoxiang Sun
- Department of Clinical Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China.
- Department of Central Laboratory, the Affiliated Yixing Hospital of Jiangsu University, Yixing, China.
| |
Collapse
|
219
|
Shefer A, Yanshole L, Proskura K, Tutanov O, Yunusova N, Grigor’eva A, Tsentalovich Y, Tamkovich S. From Cell Lines to Patients: Dissecting the Proteomic Landscape of Exosomes in Breast Cancer. Diagnostics (Basel) 2025; 15:1028. [PMID: 40310419 PMCID: PMC12026271 DOI: 10.3390/diagnostics15081028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Breast cancer (BC) is the most common cancer among women worldwide; therefore, the efforts of many scientists are aimed at finding effective biomarkers for this disease. It is known that exosomes are nanosized extracellular vesicles (EVs) that are released from various cell types, including cancer cells. Exosomes are directly involved in governing the physiological and pathological processes of an organism through the horizontal transfer of functional molecules (proteins, microRNA, etc.) from producing to receiving cells. Since the diagnosis and treatment of BC have been improved substantially with exosomes, in this study, we isolated breast carcinoma cell-derived exosomes, primary endotheliocyte-derived exosomes, and blood exosomes from BC patients (BCPs) in the first stage of disease and investigated their proteomic profiles. Methods: Exosomes were isolated from the samples by ultrafiltration and ultracentrifugation, followed by mass spectrometric and bioinformatics analyses of the data. The exosomal nature of vesicles was verified using transmission electron microscopy and flow cytometry. Results: Exosome proteins secreted by MCF-7 and BT-474 cells were found to form two clusters, one of which enhanced the malignant potential of cancer cells, while the other coincided with a cluster of HUVEC-derived exosome proteins. Despite the different ensembles of proteins in exosomes from the MCF-7 and BT-474 lines, the relevant portions of these proteins are involved in similar biological pathways. Comparison analysis revealed that more BC-associated proteins were found in the exosomal fraction of blood from BCPs than in the exosomal fraction of conditioned medium from cells mimicking the corresponding cancer subtype (89% and 81% for luminal A BC and MCF-7 cells and 86% and 80% for triple-positive BC and BT-474 cells, respectively). Conclusions: Tumor-associated proteins should be sought not in exosomes secreted by cell lines but in the composition of blood exosomes from cancer patients, while the contribution of endotheliocyte exosomes to the total pool of blood exosomes can be neglected.
Collapse
Affiliation(s)
- Aleksei Shefer
- Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.)
- Institute of Medicine and Medical Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Lyudmila Yanshole
- Laboratory of Proteomics and Metabolomics, International Tomography Center, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (L.Y.); (Y.T.)
| | - Ksenia Proskura
- Department of Mammology, Novosibirsk Regional Clinical Oncological Dispensary, 630108 Novosibirsk, Russia
| | - Oleg Tutanov
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA;
| | - Natalia Yunusova
- Laboratory of Tumor Biochemistry, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Science, 634028 Tomsk, Russia;
| | - Alina Grigor’eva
- Laboratory of Molecular Medicine, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (A.S.)
| | - Yuri Tsentalovich
- Laboratory of Proteomics and Metabolomics, International Tomography Center, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (L.Y.); (Y.T.)
| | - Svetlana Tamkovich
- Institute of Medicine and Medical Technologies, Novosibirsk State University, 630090 Novosibirsk, Russia
- Institute of Oncology and Neurosurgery, E. Meshalkin National Medical Research Center of the Ministry of Health of the Russian Federation, 630090 Novosibirsk, Russia
| |
Collapse
|
220
|
Yun JH, Noh YR, Yoo S, Park S, Choi Y, An J, Kim I. Harnessing Extracellular Vesicles for Targeted Drug Delivery in Ovarian Cancer. Pharmaceutics 2025; 17:528. [PMID: 40284522 PMCID: PMC12030366 DOI: 10.3390/pharmaceutics17040528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Ovarian cancer remains one of the most lethal gynecologic malignancies, primarily due to late-stage diagnosis, high recurrence rates, and the development of chemoresistance. Although targeted therapies have improved patient outcomes, their efficacy is often limited by off-target toxicity and acquired drug resistance. Extracellular vesicles (EVs), nanoscale vesicles naturally released by cells, have emerged as promising carriers for precision drug delivery. This review provides a comprehensive overview of recent advances in EV-based therapeutic strategies for ovarian cancer, including the delivery of chemotherapeutic agents, nucleic acid therapeutics, and immunomodulatory molecules. We further explore innovative engineering approaches to enhance targeting specificity, such as surface modification, cell source selection, biomaterial integration, and magnetic nanoparticle-assisted delivery. Key translational challenges in bringing EV-based therapies to clinical application are also addressed. Collectively, these insights underscore the transformative potential of EV-based platforms in advancing targeted and personalized treatment for ovarian cancer.
Collapse
Affiliation(s)
- Jang-Hyuk Yun
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yoo Rim Noh
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Seongkyeong Yoo
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Soohyun Park
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Yunsup Choi
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Jiyeon An
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Iljin Kim
- Department of Pharmacology and Program in Biomedical Science and Engineering, Inha University College of Medicine, Incheon 22212, Republic of Korea
- Research Center for Controlling Intercellular Communication, Inha University College of Medicine, Incheon 22212, Republic of Korea
| |
Collapse
|
221
|
Lin M, Alimerzaloo F, Wang X, Alhalabi O, Krieg SM, Skutella T, Younsi A. Harnessing stem cell-derived exosomes: a promising cell-free approach for spinal cord injury. Stem Cell Res Ther 2025; 16:182. [PMID: 40247394 PMCID: PMC12004558 DOI: 10.1186/s13287-025-04296-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/25/2025] [Indexed: 04/19/2025] Open
Abstract
Spinal cord injury (SCI) is a severe injury to the central nervous system that often results in permanent neurological dysfunction. Current treatments have limited efficacy and face challenges in restoring neurological function after injury. Recently, stem cell-derived exosomes have gained attention as an experimental treatment for SCI due to their unique properties, including superior biocompatibility, minimal immunogenicity and non-tumorigenicity. With their potential as a cell-free therapy, exosomes promote SCI repair by enhancing nerve regeneration, reducing inflammation and stabilizing the blood-spinal cord barrier. This review summarizes advances in stem cell-derived exosome research for SCI over the past years, focusing on their mechanisms and future prospects. Despite their promising therapeutic potential, clinical translation remains challenging due to standardization of exosome isolation protocols, compositional consistency and long-term safety profiles that require further investigation.
Collapse
Affiliation(s)
- Miaoman Lin
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Farzaneh Alimerzaloo
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Xingjin Wang
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Obada Alhalabi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Sandro M Krieg
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Thomas Skutella
- Medical Faculty, Heidelberg University, Heidelberg, Germany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
222
|
Korutla L, Hu R, Liu Y, Romano C, Habertheuer A, Abedi P, Wang H, Molugu S, Rostami S, Naji A, Nuqali A, Beasley M, Maulion C, Hahn S, Ahmad T, Wang Z, Sen S, Vallabhajosyula P. Circulating Tissue Specific Extracellular Vesicles for Noninvasive Monitoring of Acute Cellular Rejection in Clinical Heart Transplantation. Transplantation 2025:00007890-990000000-01061. [PMID: 40238644 DOI: 10.1097/tp.0000000000005369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BACKGROUND There remains a critical need for biomarkers of acute cellular rejection (ACR) in heart transplantation. We hypothesized that immunopathophysiology of ACR is reflected via dynamic changes in the protein and RNA cargoes of small extracellular vesicles (sEVs) released by cardiac allograft and T cells into circulation, thus enabling noninvasive window into ACR. METHODS T-cell sEVs were enriched using anti-CD3 antibody beads, and antidonor HLA I antibody beads for donor sEVs. Cargoes of donor sEVs (cardiac troponin T [cTnT] protein and mRNA) and T-cell sEVs (CD4, CD8, T-cell receptor proteins, miRNAs [miRs] let 7i, 101b, 21a) were compared with time-matched endomyocardial biopsy samples (n = 70) in 12 patients to postoperative day 120. RESULTS Six patients had 11 moderate ACR (15.7%) episodes, 1 had antibody-mediated rejection, and 5 had ≤ mild ACR. By Wilcoxon rank-sum tests, cTnT protein (P = 6.04 × 10-5) and mRNA (P = 6.87 × 10-7) were decreased with moderate ACR compared with grades 0/1 ACR. T-cell sEV CD4, CD8, and TCR protein cargoes (P ≤ 3.92 × 10-5) and miRs let 7i, 101b, and 21a (P ≤ 9.05 × 10-5) were increased with moderate ACR. Successful treatment of moderate ACR led to dynamic reversal in sEV profiles, especially donor heart sEV cTnT mRNA (Spearman coefficient 0.87) and miR 21a (coefficient 0.85). CONCLUSIONS Our first investigation in heart transplant patients demonstrated that circulating T cell-sEV and donor heart-sEV profiles enable diagnosis of moderate ACR with high diagnostic accuracy. A large sample cohort external validation study is warranted to better understand diagnostic potential of this platform for ACR monitoring in heart transplantation.
Collapse
Affiliation(s)
| | - Robert Hu
- Department of Surgery, Creighton University School of Medicine, Omaha, NE
| | - Yihan Liu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
| | - Connie Romano
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Andreas Habertheuer
- Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Parisa Abedi
- Division of Cardiac Surgery, Yale University School of Medicine, New Haven, CT
| | - He Wang
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | - Sudheer Molugu
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Susan Rostami
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Abdulelah Nuqali
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | - Michael Beasley
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | | | - Samuel Hahn
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | - Tariq Ahmad
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | - Zuoheng Wang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
| | - Sounok Sen
- Division of Cardiology, Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
223
|
Senesi G, Lodrini AM, Mohammed S, Mosole S, Hjortnaes J, Veltrop RJA, Kubat B, Ceresa D, Bolis S, Raimondi A, Torre T, Malatesta P, Goumans MJ, Paneni F, Camici GG, Barile L, Balbi C, Vassalli G. miR-24-3p secreted as extracellular vesicle cargo by cardiomyocytes inhibits fibrosis in human cardiac microtissues. Cardiovasc Res 2025; 121:143-156. [PMID: 39527589 PMCID: PMC11998913 DOI: 10.1093/cvr/cvae243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
AIMS Cardiac fibrosis in response to injury leads to myocardial stiffness and heart failure. At the cellular level, fibrosis is triggered by the conversion of cardiac fibroblasts (CF) into extracellular matrix-producing myofibroblasts. miR-24-3p regulates this process in animal models. Here, we investigated whether miR-24-3p plays similar roles in human models. METHODS AND RESULTS Gain- and loss-of-function experiments were performed using human induced pluripotent stem cell-derived cardiomyocytes (hCM) and primary hCF under normoxic or ischaemia-simulating conditions. hCM-derived extracellular vesicles (EVs) were added to hCF. Similar experiments were performed using three-dimensional human cardiac microtissues and ex vivo cultured human cardiac slices. hCF transfection with miR-24-3p mimic prevented TGFβ1-mediated induction of FURIN, CCND1, and SMAD4-miR-24-3p target genes participating in TGFβ1-dependent fibrogenesis-regulating hCF-to-myofibroblast conversion. hCM secreted miR-24-3p as EV cargo. hCM-derived EVs modulated hCF activation. Ischaemia-simulating conditions induced miR-24-3p depletion in hCM-EVs and microtissues. Similarly, hypoxia down-regulated miR-24-3p in cardiac slices. Analyses of clinical samples revealed decreased miR-24-3p levels in circulating EVs in patients with acute myocardial infarction (AMI), compared with healthy subjects. Post-mortem RNAScope analysis showed miR-24-3p down-regulation in myocardium from patients with AMI, compared with patients who died from non-cardiac diseases. Berberine, a plant-derived agent with miR-24-3p-stimulatory activity, increased miR-24-3p contents in hCM-EVs, down-regulated FURIN, CCND1, and SMAD4, and inhibited fibrosis in cardiac microtissues. CONCLUSION These findings suggest that hCM may control hCF activation through miR-24-3p secreted as EV cargo. Ischaemia impairs this mechanism, favouring fibrosis.
Collapse
Affiliation(s)
- Giorgia Senesi
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Alessandra M Lodrini
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Shafeeq Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Simone Mosole
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Switzerland
| | - Jesper Hjortnaes
- Department of Thoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Rogier J A Veltrop
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, The Netherlands
| | - Bela Kubat
- Department of Pathology, Maastricht University Medical Center, The Netherlands
| | - Davide Ceresa
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Sara Bolis
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
| | - Andrea Raimondi
- Institute of Biomedical Research, IRB, Bellinzona, Switzerland
| | - Tiziano Torre
- Heart Surgery Unit, Cardiocentro Ticino Institute, EOC, Lugano, Switzerland
| | - Paolo Malatesta
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Experimental Medicine (DIMES), Experimental Biology Unit, University of Genova, Genova, Italy
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Zurich, Switzerland
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Lucio Barile
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Faculty of Biomedical Sciences, Euler Institute, Università della Svizzera italiana, Lugano, Switzerland
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Medicine, Baden Cantonal Hospital, Baden, Switzerland
| | - Giuseppe Vassalli
- Laboratories for Translational Research, Ente Ospedaliero Cantonale, Istituto Cardiocentro Ticino, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
224
|
Du G, He J, Zhan Y, Chen L, Hu Y, Qian J, Huang H, Meng F, Shan L, Chen Z, Hu D, Zhu C, Yue M, Qi Y, Tan W. Changes and application prospects of biomolecular materials in small extracellular vesicles (sEVs) after flavivirus infection. Eur J Med Res 2025; 30:275. [PMID: 40229861 PMCID: PMC11998145 DOI: 10.1186/s40001-025-02539-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Small extracellular vesicles (sEVs), also known as exosomes, are membranous vesicles filled with various proteins and nucleic acids, serving as a communication vector between cells. Recent research has highlighted their role in viral diseases. This review synthesizes current understanding of viral sEVs and includes recent findings on sEVs infected with flaviviruses. It discusses the implications of viral sEVs research for advancing arbovirus sEVs research and anticipates the potential applications of sEVs in flavivirus infections.
Collapse
Affiliation(s)
- Gengting Du
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Junhua He
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Yan Zhan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Leru Chen
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Yue Hu
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Jiaojiao Qian
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Huan Huang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fanjin Meng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Laiyou Shan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Zhiyu Chen
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | | | - Changqiang Zhu
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Ming Yue
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Qi
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Weilong Tan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China.
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
225
|
Delshad M, Sanaei MJ, Mohammadi MH, Sadeghi A, Bashash D. Exosomal Biomarkers: A Comprehensive Overview of Diagnostic and Prognostic Applications in Malignant and Non-Malignant Disorders. Biomolecules 2025; 15:587. [PMID: 40305328 PMCID: PMC12024574 DOI: 10.3390/biom15040587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Exosomes are small extracellular vesicles, ranging from 30 to 150 nm, that are essential in cell biology, mediating intercellular communication and serving as biomarkers due to their origin from cells. Exosomes as biomarkers for diagnosing various illnesses have gained significant investigation due to the high cost and invasive nature of current diagnostic procedures. Exosomes have a clear advantage in the diagnosis of diseases because they include certain signals that are indicative of the genetic and proteomic profile of the ailment. This feature gives them the potential to be useful liquid biopsies for real-time, noninvasive monitoring, enabling early cancer identification for the creation of individualized treatment plans. According to our analysis, the trend toward utilizing exosomes as diagnostic and prognostic tools has raised since 2012. In this regard, the proportion of malignant indications is higher compared with non-malignant ones. To be precise, exosomes have been used the most in gastrointestinal, thoracic, and urogenital cancers, along with cardiovascular, diabetic, breathing, infectious, and brain disorders. To the best of our knowledge, this is the first research to examine all registered clinical trials that look at exosomes as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Mahda Delshad
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
- Department of Laboratory Sciences, School of Allied Medical Sciences, Zanjan University of Medical Sciences, Zanjan 1411718541, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717411, Iran;
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran; (M.D.); (M.-J.S.); (M.H.M.)
| |
Collapse
|
226
|
Preet R, Islam MA, Shim J, Rajendran G, Mitra A, Vishwakarma V, Kutz C, Choudhury S, Pathak H, Dai Q, Sun W, Madan R, Zhong C, Markiewicz MA, Zhang J. Gut commensal Bifidobacterium-derived extracellular vesicles modulate the therapeutic effects of anti-PD-1 in lung cancer. Nat Commun 2025; 16:3500. [PMID: 40221398 PMCID: PMC11993705 DOI: 10.1038/s41467-025-58553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although immunotherapy such as anti-programmed death-1 and its ligand 1 (PD-1/L1) is a standard treatment for advanced non-small cell lung cancer (NSCLC), many patients do not derive benefit directly. Several studies have elucidated new strategies to improve the antitumor immune response through gut microbiota modulation. However, it remains largely debatable regarding how gut microbiota remotely affect lung cancer microenvironment and subsequently modulate immunotherapy response. Here we show that commensal Bifidobacterium-derived extracellular vesicles (Bif.BEVs) can modulate the therapeutic effect of anti-PD-1 therapy in NSCLC. These Bif.BEVs are up-taken by lung cancer cells predominantly via dynamin-dependent endocytosis and upregulate PD-L1 expression through TLR4-NF-κB pathway. They also efficiently penetrate murine intestinal and patient-derived lung cancer organoids. Oral gavage of these Bif.BEVs result in their accumulation in tumors in mice. Using a syngeneic mouse model, Bif.BEVs are found to synergize the anti-tumor effect of anti-PD-1 via modulation of key cytokines, immune response and oncogenic pathways, and increase in tumor-infiltrating CD8+ T cells. Our study therefore identifies a link between Bif.BEVs and the tumor microenvironment, providing an alternative mechanism to explain how gut microbiota can influence immunotherapy response, particularly in tumors located anatomically distant from the gut.
Collapse
Affiliation(s)
- Ranjan Preet
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Md Atiqul Islam
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jiyoung Shim
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ganeshkumar Rajendran
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Amrita Mitra
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Vikalp Vishwakarma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Caleb Kutz
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, 66045, USA
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
227
|
Acevedo-Sánchez V, Rodríguez-Hernández RM, Aguilar-Ruíz SR, Torres-Aguilar H, Pina-Canseco S, Chávez-Olmos P, Garrido E, Baltiérrez-Hoyos R, Romero-Tlalolini MA. Keratinocyte-derived extracellular vesicles induce macrophage polarization toward an M1-like phenotype. Biochem Biophys Res Commun 2025; 758:151659. [PMID: 40121968 DOI: 10.1016/j.bbrc.2025.151659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Multiple reports have shown an effect of keratinocyte-derived extracellular vesicles (EVs) on keratinocytes and other cell types. However, the contribution of keratinocyte-derived EVs under physiological and pathological conditions is not fully elucidated. Therefore, whether there is an effect of EVs on macrophages in cervical cancer (CC) is also unknown. Here, we evaluated the effect of tumor and non-tumor keratinocyte-derived EVs on the polarization of peripheral blood mononuclear cells (PBMCs)-derived macrophages and THP-1 cell line. Flow cytometric evaluation of macrophages cultured in the presence of keratinocyte-derived EVs mainly indicated an increase in classical activation markers CD80 and CD86 (M1 phenotype) and little or no modification of alternative activation markers (M2 phenotype). ELISA evaluation of macrophage supernatants revealed an increase in the secretion of proinflammatory cytokines such as IL-1β and IL-6. On the other hand, TGF-β was not significantly modified and only EVs derived from non-cancerous keratinocytes induced a significant increase in IL-10. The expression levels of transcripts associated with the M1 phenotype were also evaluated by qRT-PCR with similar results to ELISA for TGF-β and IL-10; but also an increase in the expression of HLA-DRα and TNF-α was observed, and no statistically significant changes in ARG1. The ROS production was also evaluated and this increase mainly in macrophages treated with CC keratinocytes-derived EVs. So, our results suggest that the uptake of EVs derived from released by non-tumor and cervical cancer keratinocytes promotes in macrophages their polarization to an M1-like phenotype.
Collapse
Affiliation(s)
- V Acevedo-Sánchez
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - R M Rodríguez-Hernández
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - S R Aguilar-Ruíz
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - H Torres-Aguilar
- Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez de Oaxaca, Av. Universidad S/N, Cinco Señores, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - S Pina-Canseco
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - P Chávez-Olmos
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación, Gustavo A. Madero, 07360, Mexico City, Mexico.
| | - E Garrido
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación, Gustavo A. Madero, 07360, Mexico City, Mexico.
| | - R Baltiérrez-Hoyos
- Facultad de Medicina y Cirugía, CONAHCYT-Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - M A Romero-Tlalolini
- Facultad de Medicina y Cirugía, CONAHCYT-Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| |
Collapse
|
228
|
Wei J, Xie Z, Kuang X. Extracellular Vesicles in Renal Inflammatory Diseases: Revealing Mechanisms of Extracellular Vesicle-Mediated Macrophage Regulation. Int J Mol Sci 2025; 26:3646. [PMID: 40332144 PMCID: PMC12027779 DOI: 10.3390/ijms26083646] [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: 03/12/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Renal inflammatory diseases are a group of severe conditions marked by significant morbidity and mortality. Extracellular vesicles (EVs), as facilitators of intercellular communication, have been recognized as pivotal regulators of renal inflammatory diseases, significantly contributing to these conditions by modulating immune responses among other mechanisms. This review highlights the intricate mechanisms through which EVs modulate macrophage-kidney cell interactions by regulating macrophages, the principal immune cells within the renal milieu. This regulation subsequently influences the pathophysiology of renal inflammatory diseases such as acute kidney injury and chronic kidney disease. Furthermore, understanding these mechanisms offers novel opportunities to alleviate the severe consequences associated with renal inflammatory diseases. In addition, we summarize the therapeutic landscape based on EV-mediated macrophage regulatory mechanisms, highlighting the potential of EVs as biomarkers and therapeutic targets as well as the challenges and limitations of translating therapies into clinical practice.
Collapse
Affiliation(s)
- Jiatai Wei
- The Second Clinical Medical College, Nanchang University, Nanchang 330031, China; (J.W.); (Z.X.)
| | - Zijie Xie
- The Second Clinical Medical College, Nanchang University, Nanchang 330031, China; (J.W.); (Z.X.)
| | - Xiaodong Kuang
- Pathology Teaching and Research Office, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
229
|
Jacob V, de Berny Q, Brazier F, Presne C, Lion J, Ouled-Haddou H, Metzinger-Le Meuth V, Choukroun G, Metzinger L, Guillaume N. Quantification of Urine and Plasma Levels of Extracellular Vesicles in a Cohort of Kidney Transplant Recipients and Chronic Kidney Disease Patients. Int J Mol Sci 2025; 26:3635. [PMID: 40332150 PMCID: PMC12027010 DOI: 10.3390/ijms26083635] [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: 03/06/2025] [Revised: 04/02/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs) have a key role in intercellular communication. We hypothesized that EVs are biomarkers of nephropathy or kidney allograft rejection. We screened patients with chronic kidney disease (CKD) and kidney transplant (KT) recipients. We measured the urine and plasma levels of total EVs overall and EV subpopulations (positive for podocalyxin, aquaporin-1, CD133, CD144, CD19, CD3, CD16, CD56, or CD41). We included 92 patients with CKD, 70 KT recipients, and 33 healthy volunteers. In CKD, the total urine EV concentration was correlated positively with the estimated glomerular filtration rate (eGFR), but none of the subpopulations was identified as a potential biomarker of nephropathy. Among the KT recipients, 30 had good allograft function and 40 had allograft disease (13 with antibody-mediated rejections (ABMR), 12 with T-cell-mediated rejection (TCMR), and 15 with allograft dysfunction). Patients with ABMR had low plasma levels of EVs derived from B-cells, T-cells, and endothelium (p = 0.003, 0.009, and 0.005, respectively). Patients with TCMR had a low urine level of EVs derived from endothelium (p = 0.05). EVs derived from B-cells, T-cells, and endothelium might be biomarkers of kidney allograft rejection. However, we did not identify biomarkers of nephropathy in CKD.
Collapse
Affiliation(s)
- Valentine Jacob
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, F-80000 Amiens, France; (V.J.); (J.L.); (H.O.-H.); (V.M.-L.M.); (L.M.)
- Laboratory of Histocompatibility, Amiens University Hospital, F-80000 Amiens, France
| | - Quentin de Berny
- Department of Nephrology Dialysis Transplantation, Amiens University Hospital, F-80000 Amiens, France; (Q.d.B.); (F.B.); (C.P.); (G.C.)
| | - François Brazier
- Department of Nephrology Dialysis Transplantation, Amiens University Hospital, F-80000 Amiens, France; (Q.d.B.); (F.B.); (C.P.); (G.C.)
| | - Claire Presne
- Department of Nephrology Dialysis Transplantation, Amiens University Hospital, F-80000 Amiens, France; (Q.d.B.); (F.B.); (C.P.); (G.C.)
| | - Julien Lion
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, F-80000 Amiens, France; (V.J.); (J.L.); (H.O.-H.); (V.M.-L.M.); (L.M.)
- Laboratory of Histocompatibility, Amiens University Hospital, F-80000 Amiens, France
| | - Hakim Ouled-Haddou
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, F-80000 Amiens, France; (V.J.); (J.L.); (H.O.-H.); (V.M.-L.M.); (L.M.)
| | - Valérie Metzinger-Le Meuth
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, F-80000 Amiens, France; (V.J.); (J.L.); (H.O.-H.); (V.M.-L.M.); (L.M.)
- INSERM UMRS 1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, University of Sorbonne Paris Nord, F-93000 Bobigny, France
| | - Gabriel Choukroun
- Department of Nephrology Dialysis Transplantation, Amiens University Hospital, F-80000 Amiens, France; (Q.d.B.); (F.B.); (C.P.); (G.C.)
| | - Laurent Metzinger
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, F-80000 Amiens, France; (V.J.); (J.L.); (H.O.-H.); (V.M.-L.M.); (L.M.)
| | - Nicolas Guillaume
- HEMATIM UR-UPJV 4666, C.U.R.S, University of Picardie Jules Verne, F-80000 Amiens, France; (V.J.); (J.L.); (H.O.-H.); (V.M.-L.M.); (L.M.)
- Laboratory of Histocompatibility, Amiens University Hospital, F-80000 Amiens, France
| |
Collapse
|
230
|
Chen IP, Henning S, Bender M, Degenhardt S, Mhamdi Ghodbani M, Bergmann AK, Volkmer B, Brockhoff G, Wege AK, Greinert R. Detection of Human Circulating and Extracellular Vesicle-Derived miRNAs in Serum of Humanized Mice Transplanted with Human Breast Cancer (HER2 + and TNBC) Cells-A Proof of Principle Investigation. Int J Mol Sci 2025; 26:3629. [PMID: 40332177 PMCID: PMC12026515 DOI: 10.3390/ijms26083629] [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: 02/10/2025] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Humanized tumor mice (HTM) allow for preclinical cancer treatment studies of breast cancer (BC) under human-like conditions. This study utilized HTM for the first time to investigate potential miRNA biomarker candidates for treatment response in sera and extracellular vesicles (EVs), following X-irradiation and atezolizumab (anti-PD-L1) treatment. We identified the changes of human-specific miRNAs (miR-23b-3p and miR-155-5p) after irradiation and anti-PD-L1 treatment in HTMs with human epidermal growth factor receptor 2 positive (HER2+ BC) and triple-negative breast cancer (TNBC). The high degree of conserved, circulating free miRNA in mice and men represents a challenge of our assay; however, miRNAs with ≥2 nucleotide mismatches can be employed for human-specific analysis, and even conserved miRNAs may be utilized under clearly defined conditions of human tumor growth in HTM. A comparative analysis of extracellular vesicle miRNA cargo and free-circulating serum miRNAs revealed several exosome-specific miRNAs (miR-29b-3p, miR-34c-5p, miR-203a-3p, miR-378g, and miR-382-5p) in HTMs, which are known to play roles in BC. Our findings demonstrate that HTMs are a suitable model to identify treatment-induced changes in free-circulating and exosomal miRNAs that influence tumor progression and immunological tumor defense, both locally and at distant sites. This study presents a proof-of-principle approach to analyzing cell-free nucleotides and exosomes in a human-like, preclinical in vivo setting. Further refinements are necessary to enhance the sensitivity and the specificity of the HTM-based approach.
Collapse
Affiliation(s)
- I-Peng Chen
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Stefan Henning
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Marc Bender
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Sarah Degenhardt
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Mouna Mhamdi Ghodbani
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Ann Kathrin Bergmann
- Core Facility of Electron Microscopy, University Clinics Duesseldorf, 40225 Duesseldorf, Germany;
| | - Beate Volkmer
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, Medical Center Regensburg, 93053 Regensburg, Germany; (G.B.); (A.K.W.)
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Anja K. Wege
- Department of Gynecology and Obstetrics, Medical Center Regensburg, 93053 Regensburg, Germany; (G.B.); (A.K.W.)
- Bavarian Cancer Research Center (BZKF), 93053 Regensburg, Germany
| | - Rüdiger Greinert
- Department of Molecular Cell Biology, Skin Cancer Center Buxtehude, Elbekliniken Stade-Buxtehude, 21614 Buxtehude, Germany; (I.-P.C.); (S.H.); (M.B.); (M.M.G.); (B.V.)
| |
Collapse
|
231
|
Rey-Cadilhac F, Rachenne F, Marquant A, Kee Him JL, Ancelin A, Foisor V, Morille M, Lyonnais S, Cazevieille C, Missé D, Pompon J. Characterization of size distribution and markers for mosquito extracellular vesicles. Front Cell Dev Biol 2025; 13:1497795. [PMID: 40292329 PMCID: PMC12021844 DOI: 10.3389/fcell.2025.1497795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
Extracellular vesicles (EVs) are non-replicative, cell-derived membranous structures secreted by potentially all eukaryotic cells, playing a crucial role in intercellular communication. The study of EVs requires approaches and tools, which have predominantly been developed for mammalian models. Here, we undertook a multimodal characterization of mosquito EVs to provide a technical and knowledge foundation for their study. First, using a cell line model from Aedes aegypti and applying multiple analytical technologies (i.e., NTA, TEM, cryo-EM, and AFM), we observed that mosquito EVs range from 20 to 500 nm in diameter and that a majority are smaller than 100 nm. Second, we showed that smaller EVs are secreted in mosquito saliva. Third, we evaluated the capacity of differential centrifugation and size exclusion chromatography to separate mosquito EVs, revealing the strengths and weaknesses of each technology. Finally, we identified a mosquito homolog of CD63 as an extravesicular marker and the mosquito syntenin as a putative luminal marker. Overall, our results promote the development of tools and approaches for the study of mosquito EVs.
Collapse
Affiliation(s)
| | | | | | - Josephine Lai Kee Him
- CBS (Centre de Biologie Structurale), Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Aurélie Ancelin
- CBS (Centre de Biologie Structurale), Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | | | - Marie Morille
- ICGM, Univ. Montpellier, CNRS, ENSCM, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| | | | - Chantal Cazevieille
- INM (Institut de Neuroscience de Montpellier), Electronic Microscopy Plateform, Saint Eloi Hospital, Montpellier, France
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | - Julien Pompon
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| |
Collapse
|
232
|
Sun Y, Zhao M, Cheng L, He X, Shen S, Lv J, Zhang J, Shao Q, Yin W, Zhao F, Sun R, Lu P, Ji Y, Wang XW, Ji J. Reduction of alternative polarization of macrophages by short-term activated hepatic stellate cell-derived small extracellular vesicles. J Exp Clin Cancer Res 2025; 44:117. [PMID: 40211350 PMCID: PMC11983935 DOI: 10.1186/s13046-025-03380-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Activated hepatic stellate cells (HSCs) induce alternative (M2) polarization of macrophages and contribute to the progression of fibrosis and hepatocellular carcinoma (HCC). However, the effects of small extracellular vesicles released by HSCs (HSC-sEVs) during activation remain largely unknown. METHODS The aim of this study was to investigate the role of extracellular vesicles released by HSCs (HSC-sEVs) at different stages of activation in macrophage polarization. The effects of sEVs from short-term activated and long-term activated HSCs on liver macrophages was studied. Small RNA sequencing analyses were performed to obtain differential miRNAs transported by the short-term and long-term activated HSC- sEVs. The in vivo effects of short-term activated HSC-sEV-specific miRNA on liver macrophage and liver fibrosis were confirmed in a CCl4-induced liver injury mouse model. To study the tumor suppressive effects of the macrophages educated by short-term activated HSC-sEV-specific miRNA, human hepatoma cells were mixed and subcutaneously cotransplanted with miR-99a-5p mimic-pretreated macrophages. RESULTS We found that consistent with activated HSCs, long-term activated HSC-sEVs (14dHSC-sEVs) induce bone marrow-derived monocytes (MOs) toward an M2 phenotype, but short-term activated HSC-sEVs (3dHSC-sEVs) induce the resident macrophages (Kupffer cells, KCs) toward a classically activated (M1) phenotype. We identified five 3dHSC-sEV-specific miRNAs, including miR-99a-5p. In vitro and in vivo experiments support that miR-99a-5p negatively regulates alternative polarization of macrophages, decreases collagen deposition in chronic liver injury model, and suppresses the progression of hepatoma in a xenograft model partially by targeting CD93. CONCLUSION Collectively, our work reveals an unexpected proinflammatory role of 3dHSC-sEVs, preliminarily explores the underlying mechanism, and evaluates the therapeutic potential of 3dHSC-sEV-specific miR-99a-5p for liver fibrosis and tumorigenesis.
Collapse
Affiliation(s)
- Yufeng Sun
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, 226001, China
| | - Min Zhao
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Li Cheng
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Xiaoqian He
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Shiqi Shen
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Jiaying Lv
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Junyu Zhang
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Qian Shao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226001, China
| | - Wenxuan Yin
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
| | - Fengbo Zhao
- Basic Medical Research Center, Medical School of Nantong University, Nantong, 226001, China
| | - Rui Sun
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, 226001, China
| | - Peng Lu
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, 226001, China
| | - Yuhua Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, 226001, China.
- Department of Immunology and Microbiology, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA.
| | - Juling Ji
- Department of Pathology, Medical School of Nantong University, Nantong, 226001, China.
- Key Laboratory of Microenvironment and Translational Cancer Research, Nantong, 226001, China.
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, 226001, China.
| |
Collapse
|
233
|
Salih DJ, Reiners KS, Alfieri R, Salih AM, Percario ZA, Di Stefano M, Francesco S, Affabris E, Hartmann G, Santantonio T. Isolation and characterization of extracellular vesicles from EGFR mutated lung cancer cells. Clin Exp Med 2025; 25:114. [PMID: 40210802 PMCID: PMC11985682 DOI: 10.1007/s10238-025-01643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025]
Abstract
The epidermal growth factor receptor (EGFR) signaling pathway is essential for cellular processes such as proliferation, survival, and migration. Dysregulation of EGFR signaling is frequently observed in non-small cell lung cancer (NSCLC) and is associated with poor prognosis. This study aims to isolate and characterize extracellular vesicles (EVs) released by mutant EGFR lung cancer cell line PC9 and compare them with wild-type EGFR lung cancer cell line A549, while also evaluating the effect of gefitinib treatment on EV secretion and cargo composition. The two lung cancer cell lines were cultured with 2% EV-free serum, and EVs were subsequently isolated by differential ultra centrifugation. EVs were characterized by nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM) for quantification size and shape determination. Western blot analysis confirmed the enrichment and purity of isolated EVs. Results showed that EGFR mutation significantly increased EV release and altered their size, compared to EVs released by wild-type EGFR cells. In addition to classical EV markers such as CD81, Flotillin- 1, and TSG101, Western blot analysis also detected phosphorylated EGFR (p-EGFR) selectively packaged into EVs from PC9 cells. Gefitinib treatment significantly reduced EV secretion in PC9 cells and led to a marked decrease in p-EGFR incorporation into EVs, indicating that EV biogenesis and compostion are modulated by active EGFR signaling. In conclusion, this study highlights the significant influence of EGFR activation on EV secretion and cargo composition while demonstrating that EGFR inhibition via gefitinib alters EV-mediated signaling in lung cancer cells. These findings provide insights into tumor behavior, EV-mediated oncogenic communication, and the potential use of EVs as biomarkers and therapeutic targets in NSCLC.
Collapse
Affiliation(s)
- Dian Jamel Salih
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli, 121, 71122, Foggia, Italy.
- Department of Anatomy, Biology and Histology, College of Medicine, University of Duhok, Duhok, Iraq.
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.
| | - Katrin S Reiners
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | | | - Mariantonietta Di Stefano
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli, 121, 71122, Foggia, Italy
| | - Sollitto Francesco
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli, 121, 71122, Foggia, Italy
| | | | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Teresa Santantonio
- Department of Medical and Surgical Sciences, University of Foggia, Via Napoli, 121, 71122, Foggia, Italy
| |
Collapse
|
234
|
Ren T, Zhang Y, Tong Y, Zhang Q, Wang T, Wang Y, Yang C, Xu Z. FRET imaging of glycoRNA on small extracellular vesicles enabling sensitive cancer diagnostics. Nat Commun 2025; 16:3391. [PMID: 40210865 PMCID: PMC11985951 DOI: 10.1038/s41467-025-58490-2] [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: 07/12/2024] [Accepted: 03/24/2025] [Indexed: 04/12/2025] Open
Abstract
Glycosylated RNAs (glycoRNAs), a recently discovered class of membrane-associated glyco-molecules, remain poorly understood in function and clinical value due to limited detection methods. Here, we show a dual recognition Förster resonance energy transfer (drFRET) strategy using nucleic acid probes to detect N-acetylneuraminic acid-modified RNAs, enabling sensitive, selective profiling of glycoRNAs on small extracellular vesicles (sEVs) from minimal biofluids (10 μl initial biofluid). Using drFRET, we identify 5 prevalent sEV glycoRNAs derived from 7 cancer cell lines. In a 100-patient cohort (6 cancer types and non-cancer controls), sEV glycoRNA profiles achieve 100% accuracy (95% confidence interval) in distinguishing cancers from non-cancer cases and 89% accuracy in classifying specific cancer types. Furthermore, drFRET reveal that sEV glycoRNAs specifically interact with Siglec proteins and P-selectin, which is critical for sEV cellular internalization. The drFRET strategy provides a versatile and sensitive platform for the imaging and functional analysis of sEV glycoRNAs, with promising implications for clinical applications.
Collapse
Affiliation(s)
- Tingju Ren
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Yingzhi Zhang
- National Clinical Research Center for Laboratory Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yuxiao Tong
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Qi Zhang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Tianhao Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Yue Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Chunguang Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang, Liaoning Province, China.
| |
Collapse
|
235
|
Kadam V, Wacker M, Oeckl P, Korneck M, Dannenmann B, Skokowa J, Hauser S, Otto M, Synofzik M, Mengel D. Most L1CAM Is not Associated with Extracellular Vesicles in Human Biofluids and iPSC-Derived Neurons. Mol Neurobiol 2025:10.1007/s12035-025-04909-2. [PMID: 40210837 DOI: 10.1007/s12035-025-04909-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Transmembrane L1 cell adhesion molecule (L1CAM) is widely used as a marker to enrich for neuron-derived extracellular vesicles (EVs), especially in plasma. However, this approach lacks sufficient robust validation. This study aimed to assess whether human biofluids are indeed enriched for EVs, particularly neuron-derived EVs, by L1CAM immunoaffinity, utilizing multiple sources (plasma, CSF, conditioned media from iPSC-derived neurons [iNCM]) and different methods (mass spectrometry [MS], nanoparticle tracking analysis [NTA]). Following a systematic multi-step validation approach, we confirmed isolation of generic EV populations using size-exclusion chromatography (SEC) and polymer-aided precipitation (PPT)-two most commonly applied EV isolation methods-from all sources. Neurofilament light (NfL) was detected in both CSF and blood-derived EVs, indicating their neuronal origin. However, L1CAM immunoprecipitation did not yield enrichment of L1CAM in EV fractions. Instead, it was predominantly found in its free-floating form. Additionally, MS-based proteomic analysis of CSF-derived EVs also did not show L1CAM enrichment. Our study validates EV isolation from diverse biofluid sources by several isolation approaches and confirms that some EV subpopulations in human biofluids are of neuronal origin. Thorough testing across multiple sources by different orthogonal methods, however, does not support L1CAM as a marker to reliably enrich for a specific subpopulation of EVs, particularly of neuronal origin.
Collapse
Affiliation(s)
- Vaibhavi Kadam
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tuebingen, Tuebingen, Germany
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tuebingen, Tuebingen, Germany
| | - Madeleine Wacker
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tuebingen, Tuebingen, Germany
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany
| | - Patrick Oeckl
- Department of Neurology, Ulm University Hospital, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE) Ulm, Ulm, Germany
| | - Milena Korneck
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany
- Graduate School of Cellular and Molecular Neuroscience, University of Tuebingen, Tuebingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tuebingen, Tuebingen, Germany
| | - Benjamin Dannenmann
- Department of Oncology, Hematology, Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stefan Hauser
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tuebingen, Tuebingen, Germany
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Matthis Synofzik
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tuebingen, Tuebingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany.
| | - David Mengel
- Division Translational Genomics of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tuebingen, Tuebingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Tuebingen, Tuebingen, Germany.
| |
Collapse
|
236
|
Wasserman AH, Abolibdeh B, Hamdan R, Hong CC. Stem-Cell Derived Exosomal microRNAs as Biomarkers and Therapeutics for Pediatric Cardiovascular Disease. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2025; 27:32. [PMID: 40224357 PMCID: PMC11982073 DOI: 10.1007/s11936-025-01088-0] [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] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
Purpose of Review In recent years, several pre-clinical studies have demonstrated the therapeutic potential of stem cell-derived exosomes in the treatment of cardiovascular disease (CVD). Here, we evaluate their potential as biomarkers for the detection and monitoring of CVD, with a particular focus on pediatric heart disease. Recent Findings Exosomes isolated from stem cell sources, including mesenchymal stem cells (MSCs) and pluripotent stem cells (PSCs), benefit cardiovascular function, inflammatory responses, and angiogenesis in injured and diseased hearts. These exosomes carry a variety of cargo, such as proteins, lipids, and nucleic acids. However, the majority contain non-coding RNA molecules. Summary Review of the existing literature for several non-coding RNAs and their relationship to CVD suggests that exosomes containing microRNAs (miRNAs) can serve as promising biomarkers for CVD due to their presence in circulation, ease of isolation, and therapeutic potential. These biomarkers are especially promising as screening and diagnostic tools for the early detection of pediatric and congenital heart disease.
Collapse
Affiliation(s)
- Aaron H. Wasserman
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI USA
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI USA
| | - Bana Abolibdeh
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI USA
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI USA
| | - Reema Hamdan
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI USA
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI USA
| | - Charles C. Hong
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI USA
- Henry Ford Health + Michigan State Health Sciences, Detroit, MI USA
| |
Collapse
|
237
|
Jiang Q, Ning F, Jia Q, Wang H, Xue W, Wang J, Wang Y, Zhu Z, Tian L. MiR-148a-3p Loaded Human Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles Alleviates Silica-Induced Pulmonary Fibrosis by Inhibiting β-Catenin Signaling. Int J Nanomedicine 2025; 20:4319-4336. [PMID: 40230541 PMCID: PMC11994480 DOI: 10.2147/ijn.s506542] [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: 11/26/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Background In clinical practice, due to the lack of typical symptoms and specific diagnostic biomarkers, silicotic patients often having already developed pulmonary fibrosis by the time of clinical diagnosis. Studies have demonstrated that human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hucMSC-EVs) could moderate silicosis fibrosis, which may be related to the microRNAs (miRNAs) in hucMSC-EVs. While the full extent of their antifibrotic effects and the underlying mechanisms remain to be elucidated. Methods HucMSC-EVs were administered from day 28 to day 56 after silica exposure in mice, which in a therapeutic manner. In addition, the antifibrotic abilities of engineered hucMSC-EVs with varying levels of miR-148a-3p, a miRNA with antifibrotic properties, were evaluated. Heat shock protein 90 beta family member 1 (Hsp90b1) is reported to be a target of miR-148a-3p, the protein-protein interaction analysis was used to explore its regulated downstream factors in lung fibrosis. The underlying mechanisms were also investigated by using miR-148a-3p mimics and small interfering RNA (siRNA) targeting Hsp90b1 in vitro. Results HucMSC-EVs could reduce the histopathological changes and the levels of fibrotic proteins in the mouse lung tissues when administered in a therapeutic manner. Meanwhile, miR-148a-3p-overexpressed hucMSC-EVs intervention exhibited the enhanced anti-fibrotic effect compared with the negative control intervention group. In vitro, the elevated level of miR-148a-3p in hucMSC-EVs was shown to enhance hucMSC-EVs' inhibition of fibroblast collagen hypersecretion, whereas a depressed level of miR-148a-3p in hucMSC-EVs partially counteracted the inhibitory effect. Moreover, the mechanistic investigations revealed that miR-148a-3p could blunt β-catenin signaling via targeting Hsp90b1 in fibroblasts. Conclusion This study demonstrated that hucMSC-EVs retain their antifibrotic properties in silicotic mice when administered in a therapeutic manner. Further, miR-148a-3p was confirmed to be an essential component within hucMSC-EVs, mediating their inhibition of silica-induced pulmonary fibrosis by reducing β-catenin signaling via targeting of Hsp90b1 in fibroblasts.
Collapse
Affiliation(s)
- Qiyue Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Fuao Ning
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Qiyue Jia
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Hongwei Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Wenming Xue
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Jiaxin Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, People’s Republic of China
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing, 100069, People’s Republic of China
| |
Collapse
|
238
|
Lundy DJ, Liao CT. Extracellular Vesicles in Aging and Age-Related Diseases. How Important Are They? Adv Biol (Weinh) 2025:e2400656. [PMID: 40202045 DOI: 10.1002/adbi.202400656] [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: 10/26/2024] [Revised: 03/25/2025] [Indexed: 04/10/2025]
Abstract
Extracellular vesicles (EVs), lipid bilayer-bound particles secreted by cells, have attracted significant research attention for their roles in aging-related disorders, including cardiovascular disease, metabolic dysfunction, cancer, and neurodegeneration. Research shows that EV cargo and function are influenced by factors including age, disease state, exercise, nutrition and sleep, and they may modulate various aging-associated processes such as stem cell renewal, nutrient sensing, cell senescence, mitochondrial function, and insulin resistance. This perspective highlights, for a general audience, a selection of studies of EVs in aging and age-related diseases, and their diverse roles in organ crosstalk. While current evidence indicates that EVs play multiple roles in aging, there are numerous challenges including methodological challenges and limitations, heterogeneous reports of EV cargo, limited reproducibility, and apparent context-dependent effects of EVs and their cargo. Together, this limits the interpretation of these studies. This is proposed that EVs may act as fine-tuners of cellular communication within the broader aging-associated secretome and the importance of standardizing methods are emphasized. Last, future directions for EV research are suggested.
Collapse
Affiliation(s)
- David J Lundy
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, 301 Yuantong Road, Taipei, 235603, Taiwan
- Graduate Institute of Biomedical Materials & Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 301 Yuantong Road, Taipei, 235603, Taiwan
- Cell Therapy Center, Taipei Medical University Hospital, 250 Wuxing Street, Taipei, 110, Taiwan
| | - Chia-Te Liao
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, 235603, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 110, Taiwan
- Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei, 110, Taiwan
| |
Collapse
|
239
|
Wang L, Liu R, Wang Y. The roles of extracellular vesicles in mental disorders: information carriers, biomarkers, therapeutic agents. Front Pharmacol 2025; 16:1591469. [PMID: 40271072 PMCID: PMC12014780 DOI: 10.3389/fphar.2025.1591469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Mental disorders are complex conditions that encompass various symptoms and types, affecting approximately 1 in 8 people globally. They place a significant burden on both families and society as a whole. So far, the etiology of mental disorders remains poorly understood, making diagnosis and treatment particularly challenging. Extracellular vesicles (EVs) are nanoscale particles produced by cells and released into the extracellular space. They contain bioactive molecules including nucleotides, proteins, lipids, and metabolites, which can mediate intercellular communication and are involved in various physiological and pathological processes. Recent studies have shown that EVs are closely linked to mental disorders like schizophrenia, major depressive disorder, and bipolar disorder, playing a key role in their development, diagnosis, prognosis, and treatment. Therefore, based on recent research findings, this paper aims to describe the roles of EVs in mental disorders and summarize their potential applications in diagnosis and treatment, providing new ideas for the future clinical transformation and application of EVs.
Collapse
Affiliation(s)
| | | | - Ying Wang
- Department of Pharmacy, Tianjin Anding Hospital, Tianjin, China
| |
Collapse
|
240
|
Islam MK, Mahmud I, Leivo J. Designing nanoparticle-based bioaffinity assays for the detection of extracellular vesicles. Nanomedicine (Lond) 2025:1-4. [PMID: 40200739 DOI: 10.1080/17435889.2025.2488726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Affiliation(s)
- Md Khirul Islam
- Biotechnology Division, Department of Life Technologies and FICAN West Cancer Centre, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Imran Mahmud
- Biotechnology Division, Department of Life Technologies and FICAN West Cancer Centre, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Janne Leivo
- Biotechnology Division, Department of Life Technologies and FICAN West Cancer Centre, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| |
Collapse
|
241
|
Kawiková I, Špička V, Lai JCK, Askenase PW, Wen L, Kejík Z, Jakubek M, Valeš K, Španiel F. Extracellular vesicles as precision therapeutics for psychiatric conditions: targeting interactions among neuronal, glial, and immune networks. Front Immunol 2025; 16:1454306. [PMID: 40264776 PMCID: PMC12011847 DOI: 10.3389/fimmu.2025.1454306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/14/2025] [Indexed: 04/24/2025] Open
Abstract
The critical role of the immune system in brain function and dysfunction is well recognized, yet development of immune therapies for psychiatric diseases has been slow due to concerns about iatrogenic immune deficiencies. These concerns are emphasized by the lack of objective diagnostic tools in psychiatry. A promise to resolve this conundrum lies in the exploitation of extracellular vesicles (EVs) that are physiologically produced or can be synthetized. EVs regulate recipient cell functions and offer potential for EVs-based therapies. Intranasal EVs administration enables the targeting of specific brain regions and functions, thereby facilitating the design of precise treatments for psychiatric diseases. The development of such therapies requires navigating four dynamically interacting networks: neuronal, glial, immune, and EVs. These networks are profoundly influenced by brain fluid distribution. They are crucial for homeostasis, cellular functions, and intercellular communication. Fluid abnormalities, like edema or altered cerebrospinal fluid (CSF) dynamics, disrupt these networks, thereby negatively impacting brain health. A deeper understanding of the above-mentioned four dynamically interacting networks is vital for creating diagnostic biomarker panels to identify distinct patient subsets with similar neuro-behavioral symptoms. Testing the functional pathways of these biomarkers could lead to new therapeutic tools. Regulatory approval will depend on robust preclinical data reflecting progress in these interdisciplinary areas, which could pave the way for the design of innovative and precise treatments. Highly collaborative interdisciplinary teams will be needed to achieve these ambitious goals.
Collapse
Affiliation(s)
- Ivana Kawiková
- National Institute of Mental Health, Klecany, Czechia
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
- Department of Biology, Hartford University, West Hartford, CT, United States
| | - Václav Špička
- Institute of Physics of the Czech Academy of Sciences, Prague, Czechia
| | - James C. K. Lai
- Department of Biomedical and Pharmaceutical Sciences, Idaho State University College of Pharmacy, Pocatello, ID, United States
- Department of Diagnostic Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Philip W. Askenase
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Li Wen
- Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Zdeněk Kejík
- Biotechnology and Biomedical Center in Vestec (BIOCEV) , First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Milan Jakubek
- Biotechnology and Biomedical Center in Vestec (BIOCEV) , First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Karel Valeš
- National Institute of Mental Health, Klecany, Czechia
- 3rd Medical Faculty, Charles University, Prague, Czechia
| | - Filip Španiel
- National Institute of Mental Health, Klecany, Czechia
- 3rd Medical Faculty, Charles University, Prague, Czechia
| |
Collapse
|
242
|
Jang J, Shin J, Ahn Y, Kim K, Cho J, Lee WJ, Nam C, Baek MC, Seo D, Yea K. Modular and Nondisturbing Chimeric Adaptor Protein for Surface Chemistry of Small Extracellular Vesicles. ACS NANO 2025; 19:12839-12852. [PMID: 40119814 DOI: 10.1021/acsnano.4c15441] [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: 03/24/2025]
Abstract
Current chemical strategies for modifying the surface of extracellular vesicles (sEVs) often struggle to balance efficient functionalization with preserving structural integrity. Here, we present a modular approach for the surface modification of sEVs using a chimeric adaptor protein (CAP). The CAP was designed with three key features: a SNAP-tag for stable and modular binding, long and rigid linker to enhance spatial accessibility and conjugation efficiency, and the N-terminal sorting domain derived from syntenin to improve CAP expression on the sEV. We established a postsynthetic method to introduce diverse functional molecules onto sEVs, creating a versatile system termed "sEV-X" (where X represents an organic molecule, protein, or nanoparticle). Quantitative analyses at the single-molecule level revealed a linear relationship between CAP expression and the number of conjugated functional molecules, underscoring the importance of steric hindrance mitigation in sEV surface engineering. Moreover, antibody-conjugated sEVs as drug carriers, demonstrated significant tumor-specific delivery and therapeutic efficacy in a tumor-bearing mouse model, underscoring the potential of CAP-expressing sEVs as a customizable therapeutic vesicle. Overall, the CAP technology may serve as a universal platform for advancing the development of sEV-based therapeutics.
Collapse
Affiliation(s)
- Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Jiwon Shin
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Yongdeok Ahn
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Kiwook Kim
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Juhyeong Cho
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Wonhee John Lee
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Chaerin Nam
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Republic of Korea
| |
Collapse
|
243
|
Krause-Hauch M, Patel RS, Wang B, Osborne B, Jones B, Albear P, Patel NA. lncRNAs GAS5 and MALAT1 Contained in Human Adipose Stem Cell (hASC)-Derived Exosomes Drive the Cell-Free Repair and Regeneration of Wounds In Vivo. Int J Mol Sci 2025; 26:3479. [PMID: 40331955 PMCID: PMC12027045 DOI: 10.3390/ijms26083479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Wound healing progresses through four phases: hemostasis, inflammation, proliferation, and remodeling. Wounds may become chronic if this process is disrupted. The use of small extracellular vesicle (sEV; EVs < 200 nm) exosomes (exo; ~40-120 nm) derived from human adipose stem cells (hASCs) as a treatment for wounds is well studied. The cargo of these exosomes is of great interest as this accelerates wound healing. Our previous studies identified lncRNAs GAS5 and MALAT1 as packaged and enriched in hASC exosomes. In this study, we use a rat model to examine the effects on wound healing when hASC exosomes are depleted of GAS5 and MALAT1. Rats were wounded and wounds were treated with 100 μg hASCexo or hASCexo-G-M every 2 days for 1 week. qPCR was completed to evaluate the molecular effects of depletion of GAS5 and MALAT1 from hASCexo. RNAseq was performed on wound tissue to evaluate the molecular mechanisms changed by hASCexo-G-M in wound healing. While hASCexo-G-M significantly improved wound healing rate compared to control wounds, healing occurred slower than in wounds treated with hASCexo that were not depleted of GAS5 and MALAT1. Overall, this study reveals that molecular functions associated with healing are reduced in the absence of GAS5 and MALAT1, highlighting the importance of these lncRNAs.
Collapse
Affiliation(s)
- Meredith Krause-Hauch
- Research Service, James A. Haley Veterans’ Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (M.K.-H.); (R.S.P.); (B.W.); (P.A.)
| | - Rekha S. Patel
- Research Service, James A. Haley Veterans’ Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (M.K.-H.); (R.S.P.); (B.W.); (P.A.)
| | - Bangmei Wang
- Research Service, James A. Haley Veterans’ Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (M.K.-H.); (R.S.P.); (B.W.); (P.A.)
| | - Brenna Osborne
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Brianna Jones
- Department of Chemistry, University of South Florida, Tampa, FL 33612, USA;
| | - Paul Albear
- Research Service, James A. Haley Veterans’ Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (M.K.-H.); (R.S.P.); (B.W.); (P.A.)
| | - Niketa A. Patel
- Research Service, James A. Haley Veterans’ Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; (M.K.-H.); (R.S.P.); (B.W.); (P.A.)
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA;
| |
Collapse
|
244
|
Castedo N, Alfonso A, Alvariño R, Vieytes MR, Botana LM. Cyclophilin A and C are the Main Components of Extracellular Vesicles in Response to Hyperglycemia in BV2 Microglial Cells. Mol Neurobiol 2025:10.1007/s12035-025-04921-6. [PMID: 40199808 DOI: 10.1007/s12035-025-04921-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Cyclophilins (Cyps) and CD147 receptor play a crucial role in the inflammatory responses. Chronic inflammation causes tissue damage and is a common condition of several inflammation-based pathologies as diabetes or Alzheimer´s disease. Under high glucose (HG) conditions, microglia is activated and releases inflammatory mediators. In this process the role of Cyps is unknown, so this study was aimed to investigate the profile of Cyps in microglia and their release through extracellular vesicles (EVs) under hyperglycemia. An increase in reactive oxygen species (ROS) and nitric oxide (NO) levels was observed when BV2 glia cells were incubated with HG concentration. These effects were mitigated by the Cyps inhibitor cyclosporine A (CsA), suggesting the implication of Cyps in BV2 activation. In these conditions the intracellular expression of CypA, B, C and D, as well as the membrane expression of CD147 receptor was increased. In addition, only CypA and CypC were detected in the extracellular medium. Then, the presence of Cyps inside EVs was explored as an alternative secretion route. Interestingly, under HG treatment, an increase in the levels of the four Cyps in EVs was observed. When neurons were treated with EVs derived from HG-treated glia cells, their viability was reduced and EVs were detected in cytosol neurons pointing to an EVs-Cyps neurotoxic effect. These findings provide novel insights into the relationship between Cyps and EVs in neuroinflammation in hyperglycemia conditions. The current results strengthen the role of Cyps in cell communication and its potential role in brain function under pathological conditions.
Collapse
Affiliation(s)
- Noelia Castedo
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Rebeca Alvariño
- Departamento de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España.
| | - Mercedes R Vieytes
- Departamento de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España.
| |
Collapse
|
245
|
Sun J, Li Z, Chen Y, Chang Y, Yang M, Zhong W. Enhancing Analysis of Extracellular Vesicles by Microfluidics. Anal Chem 2025; 97:6922-6937. [PMID: 40133233 DOI: 10.1021/acs.analchem.4c07016] [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: 03/27/2025]
Abstract
Extracellular vesicles (EVs) play crucial roles in intercellular communication and hold great promise as biomarkers for noninvasive disease diagnosis. Intensive research efforts have been devoted to discovering the EV subpopulations responsible for specific functions or with enhanced effectiveness as disease markers, through extensive EV purification and content analysis. However, their high heterogeneity in size and cargo composition poses significant challenges for reaching such goals. Isolation methods like ultracentrifugation and size-exclusion chromatography, as well as content analysis approaches like polymerase chain reaction and enzyme-linked immunosorbent assay, have made significant contributions to improving our understanding of EV biology. Nonetheless, these methods face limitations in isolation efficiency, EV purity, and detection sensitivity and specificity due to issues like large sample consumption, unsatisfactory purity, and insufficient resolution in EV subtyping. Microfluidic technology presents promising solutions to these challenges, leveraging their intrinsic capabilities in precise flow and external energy field manipulation, sample compartmentalization, and signal enhancement at the micro- and nanoscale. Hence, this review summarizes the recent developments in microfluidics-enabled EV analysis, paying special attention to the unique microfluidic features exploited. Strategies such as viscoelastic and inertial flow, fluid mixing, and external-field-assisted approaches in improving EV purification, as well as compartmentalization and micro/nanostructures for enhancing EV detection, are examined. Furthermore, the current limitations and potential future directions are discussed to inspire advancements in this rapidly developing field.
Collapse
Affiliation(s)
- Jiayu Sun
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518057, P. R. China
| | | | | | | | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518057, P. R. China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, P. R. China
| | | |
Collapse
|
246
|
Brennan K, Vaiciuleviciute R, Uzieliene I, Pachaleva J, Kasilovskiene Z, Piesiniene L, Bernotiene E, Mc Gee MM. Menstrual blood serum extracellular vesicles reveal novel molecular biomarkers and potential endotypes of unexplained infertility. Sci Rep 2025; 15:11974. [PMID: 40199990 PMCID: PMC11978918 DOI: 10.1038/s41598-025-95818-w] [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: 11/04/2024] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
Several biomolecules have been previously associated with unexplained infertility (uIF) in blood and uterine samples, immune cells and their secreted factors, endometrial tissue, menstrual blood, serum, and stromal cells, however they do not comprehensively represent different uIF endotypes and their isolation/detection involves invasive diagnostic methods and lacks precision. This ex-vivo study was performed on extracellular vesicles (EVs) from menstrual blood collected on cycle day 2 from 9 fertile volunteers and 8 women with uIF. Menstrual blood serum (MBS) EVs were isolated from fertile and uIF MBS using Iodixanol Density Gradient Centrifugation and quantified by flow cytometry. EVs were characterized according to MISEV2023 guidelines. Comprehensive proteomic analysis of MBS EVs and EV-depleted MBS showed significant changes in uIF proteome, mostly affecting cell adhesion, immune response, apoptosis, response to oxidative stress and lipid metabolism. These processes were previously linked to pathologies of the female reproductive system but never investigated in uIF and were used to stratify patients into distinct molecular endotypes. Area under the curve (AUC) analysis was used to determine the optimum set of biomarkers for each of the uIF molecular endotypes. These findings provide new insights into uIF that could facilitate personalised treatment approaches.
Collapse
Affiliation(s)
- Kieran Brennan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Raminta Vaiciuleviciute
- Department of Regenerative Medicine, Innovative Medicine Centre, Santariskiu str 5, LT-08406, Vilnius, Lithuania
| | - Ilona Uzieliene
- Department of Regenerative Medicine, Innovative Medicine Centre, Santariskiu str 5, LT-08406, Vilnius, Lithuania
| | - Jolita Pachaleva
- Department of Regenerative Medicine, Innovative Medicine Centre, Santariskiu str 5, LT-08406, Vilnius, Lithuania
| | | | | | - Eiva Bernotiene
- Department of Regenerative Medicine, Innovative Medicine Centre, Santariskiu str 5, LT-08406, Vilnius, Lithuania
- Faculty of Fundamental Sciences, VilniusTech, Sauletekio al. 11, LT-10223, Vilnius, Lithuania
| | - Margaret M Mc Gee
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin 4, Ireland.
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
247
|
Cascabulho CM, Horita SIM, Beghini DG, Menna-Barreto RFS, Monsores ACHMG, Bertho AL, Henriques-Pons A. Plasma Microvesicles May Contribute to Muscle Damage in the mdx Mouse Model of Duchenne Muscular Dystrophy. Int J Mol Sci 2025; 26:3499. [PMID: 40331939 PMCID: PMC12026684 DOI: 10.3390/ijms26083499] [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: 03/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 05/08/2025] Open
Abstract
Extracellular vesicles (EVs) are cell-derived lipid-bound vesicles divided into apoptotic bodies, microvesicles (MVs), and exosomes based on their biogenesis, release pathway, size, content, and functions. EVs are intercellular mediators that significantly affect muscle diseases such as Duchenne muscular dystrophy (DMD). DMD is a fatal X-linked disorder caused by mutations in the dystrophin gene, leading to muscle degeneration. Mdx mice are the most commonly used model to study the disease, and in this study, we phenotypically characterized plasma MVs from mdx mice by flow cytometry. Furthermore, we assessed the ability of plasma MVs to modulate muscle inflammation, damage, and/or regeneration by intramuscular injection of MVs from mdx mice into mdx or DBA/2 mice as a control. In both mouse lineages, platelets and erythrocytes were the primary sources of MVs, and CD3+ CD4+ MVs were observed only in mdx mice. We also observed that plasma MVs from mdx mice induced muscle damage in mdx mice but not in DBA/2 mice, while plasma MVs from DBA/2 mice did not induce muscle damage in either mouse lineage. These results indicate that plasma MVs from mdx are potentially pathogenic. However, this condition also depends on the muscular tissue status, which must be responsive due to active inflammatory or regenerative responses.
Collapse
Affiliation(s)
- Cynthia Machado Cascabulho
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil; (S.I.M.H.); (D.G.B.); (A.C.H.M.G.M.); (A.H.-P.)
| | - Samuel Iwao Maia Horita
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil; (S.I.M.H.); (D.G.B.); (A.C.H.M.G.M.); (A.H.-P.)
- Laboratório de Pesquisas Sobre o Timo, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil
| | - Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil; (S.I.M.H.); (D.G.B.); (A.C.H.M.G.M.); (A.H.-P.)
| | | | - Ana Carolina Heber Max Guimarães Monsores
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil; (S.I.M.H.); (D.G.B.); (A.C.H.M.G.M.); (A.H.-P.)
| | - Alvaro Luiz Bertho
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil; (S.I.M.H.); (D.G.B.); (A.C.H.M.G.M.); (A.H.-P.)
| |
Collapse
|
248
|
Xing Q, Xu Y, Luo Y, Li C, Wang P, Kang B, Lu C. MiR-122-5p inhibits the epithelial mesenchymal transition of liver cancer cells by inducing hiPSCs to differentiate into hepatocyte-like cells. Eur J Histochem 2025; 69:4190. [PMID: 40336362 PMCID: PMC12086357 DOI: 10.4081/ejh.2025.4190] [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: 01/24/2025] [Accepted: 04/02/2025] [Indexed: 05/09/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is closely linked to liver cancer prognosis, invasiveness, and aggressiveness. One promising treatment for liver cancer is cell therapy, where stem cells are stimulated to develop into functional liver cells. This study aimed to investigate the effect of miR-122-5p on the differentiation of human induced pluripotent stem cells (hiPSCs) into hepatocyte-like cells and its impact on the EMT process in liver cancer cells. MiR-122-5p was overexpressed or silenced in hiPSCs to analyze the expression of liver-specific markers, including AFP, ALB and ASGPR, to confirm hepatocyte-like differentiation. A co-culture system with HepG2 liver cancer cells was also used to evaluate the effect of miR-122-5p-overexpressing hiPSCs or miR-122-5p-silencing hiPSCs on the expression of EMT markers. Results revealed that overexpression of miR-122-5p in hiPSCs induced hepatocyte-like characteristics, as evidenced by increased levels of AFP, ALB, and ASGPR. However, knockdown of miR-122-5p had the opposite effect. In the co-culture system, hiPSCs overexpressing miR-122-5p inhibited the EMT process of HepG2 cells, resulting in increased levels of mesenchymal markers and decreased levels of epithelial markers. Taken together, miR-122-5p promotes the differentiation of hiPSCs into hepatocyte-like cells and inhibits EMT process of liver cancer cells. Targeting miR-122-5p may be a novel approach to prevent liver cancer progression through cell therapy.
Collapse
Affiliation(s)
- Qianzhe Xing
- Department of Hepatobiliary Surgery
- Tianjin Institute of Hepatobiliary Disease
| | - Yanjie Xu
- Department of Hepatobiliary Surgery
- Tianjin Institute of Hepatobiliary Disease
| | - Ying Luo
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, China
| | - Chenglong Li
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, China
| | - Peng Wang
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, China
| | - Bin Kang
- Department of Hepatobiliary Surgery
- Tianjin Institute of Hepatobiliary Disease
| | - Chengjun Lu
- Department of Hepatobiliary Surgery
- Tianjin Institute of Hepatobiliary Disease
| |
Collapse
|
249
|
Nueraihemaiti N, Dilimulati D, Baishan A, Hailati S, Maihemuti N, Aikebaier A, Paerhati Y, Zhou W. Advances in Plant-Derived Extracellular Vesicle Extraction Methods and Pharmacological Effects. BIOLOGY 2025; 14:377. [PMID: 40282242 PMCID: PMC12024818 DOI: 10.3390/biology14040377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Abstract
Extracellular vesicles (EVs) are those with a double-membrane structure that contains proteins, lipids, nucleic acids, and other biologically active substances that play an important role in cell-cell and cell-environment communication. They have also become an important mechanism for exchanging biologically active substances for cellular molecules. As many studies on EVs have been conducted, plant-derived extracellular vesicles (PDEVs) have also started attracting attention. The biological activity and stability of PDEVs are closely related to the extraction and separation methods, and choosing a separation method that meets the requirements of PDEVs is important. The extraction methods of PDEVs include ultracentrifugation, ultrafiltration, size-exclusion chromatography, etc. In recent years, it has been found through research that PDEVs possess biological properties, such as anti-inflammatory, anti-cancer, and anti-infective properties, and that they show unique advantages as therapeutic agents and drug carriers. Therefore, we have collected the scientific literature related to EVs derived from more than a dozen fruits and vegetables, and summarized and analyzed their extraction, separation, and roles in disease treatment, aiming to provide reference and inspiration for the in-depth study of the efficacy of new drugs.
Collapse
Affiliation(s)
- Nuerbiye Nueraihemaiti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Dilihuma Dilimulati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alhar Baishan
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Sendaer Hailati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Nulibiya Maihemuti
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Alifeiye Aikebaier
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Yipaerguli Paerhati
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| | - Wenting Zhou
- Department of Pharmacology, School of Pharmacy, Xinjiang Medical University, Urumqi 830017, China; (N.N.); (D.D.); (A.B.); (S.H.); (N.M.); (A.A.); (Y.P.)
- Xinjiang Key Laboratory of Natural Medicines Active Components and Drug Release Technology, Urumqi 830017, China
- Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Urumqi 830017, China
- Engineering Research Center of Xinjiang and Central Asian Medicine Resources, Ministry of Education, Urumqi 830017, China
| |
Collapse
|
250
|
Chakraborty A, Mitra J, Malojirao VH, Kodavati M, Mandal SM, Gill SK, Sreenivasmurthy SG, Vasquez V, Mankevich M, Krishnan B, Ghosh G, Hegde M, Hazra T. Fructose-2,6-bisphosphate restores TDP-43 pathology-driven genome repair deficiency in motor neuron diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.13.623464. [PMID: 39990425 PMCID: PMC11844424 DOI: 10.1101/2024.11.13.623464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
TAR DNA-binding protein 43 (TDP-43) proteinopathy plays a critical role in neurodegenerative diseases, including amyotrophic lateral sclerosis and frontotemporal dementia (FTD). In our recent discovery, we identified that TDP-43 plays an essential role in DNA double-strand break (DSB) repair via the non-homologous end joining (NHEJ) pathway. Here, we found persistent DNA damage in the brains of ALS/FTD patients, primarily in the transcribed regions of the genome. We further investigated the underlying mechanism and found that polynucleotide kinase 3'-phosphatase (PNKP) activity was severely impaired in the nuclear extracts of both patient brains and TDP-43-depleted cells. PNKP is a key player in DSB repair within the transcribed genome, where its 3'-P termini processing activity is crucial for preventing persistent DNA damage and neuronal death. The inactivation of PNKP in ALS/FTD was due to reduced levels of its interacting partner, phosphofructo-2-kinase fructose 2,6 bisphosphatase (PFKFB3), and its biosynthetic product, fructose-2,6-bisphosphate (F2,6BP), an allosteric modulator of glycolysis. Recent work from our group has shown that F2,6BP acts as a positive modulator of PNKP activity in vivo. Notably, exogenous supplementation with F2,6BP restored PNKP activity in nuclear extracts from ALS/FTD brain samples and patient-derived induced pluripotent stem (iPS) cells harboring pathological mutations. Furthermore, we demonstrate that supplementation of F2,6BP restores genome integrity and partially rescues motor phenotype in a Drosophila model of ALS. Our findings underscore the possibility of exploring the therapeutic potential of F2,6BP or its analogs in TDP-43 pathology-associated motor neuron diseases.
Collapse
|