1
|
Choi H, Kwak MJ, Choi Y, Kang AN, Mun D, Eor JY, Park MR, Oh S, Kim Y. Extracellular vesicles of Limosilactobacillus fermentum SLAM216 ameliorate skin symptoms of atopic dermatitis by regulating gut microbiome on serotonin metabolism. Gut Microbes 2025; 17:2474256. [PMID: 40028723 PMCID: PMC11881872 DOI: 10.1080/19490976.2025.2474256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 03/05/2025] Open
Abstract
Atopic dermatitis (AD) is a globally prevalent chronic inflammatory skin disorder. Its pathogenesis remains incompletely understood, resulting in considerable therapeutic challenges. Recent studies have highlighted the significance of the interaction between AD and gut microbiome. In this study, we investigated the effects of probiotic-derived extracellular vesicles on AD. Initially, we isolated and characterized extracellular vesicles from Limosilactobacillus fermentum SLAM 216 (LF216EV) and characterized their composition through multi-omics analysis. Gene ontology (GO) and pathway analysis classified LF216EV proteins into biological processes, molecular functions, and cellular components. Importantly, specific abundance in linoleic, oleic, palmitic, sebacic, and stearic acids indicating upregulated fatty acid metabolism were observed by metabolomic analysis. Furthermore, featured lipid profiling including AcylGlcADG and ceramide were observed in LF216EV. Importantly, in an atopic dermatitis-like cell model induced by TNFα/IFNγ, LF216EV significantly modulated the expression of immune regulatory genes (TSLP, TNFα, IL-6, IL-1β, and MDC), indicating its potential functionality in atopic dermatitis. LF216EV alleviated AD-like phenotypes, such as redness, scaling/dryness, and excoriation, induced by DNCB. Histopathological analysis revealed that LF216EV decreased epidermal thickness and mast cell infiltration in the dermis. Furthermore, LF216EV administration reduced mouse scratching and depression-related behaviors, with a faster onset than the classical treatment with dexamethasone. In the quantitative real-time polymerase chain reaction (qRT-PCR) analysis, we observed a significant increase in the expression levels of htrb2c, sert, and tph-1, genes associated with serotonin, in the skin and gut of the LF216EV-treated group, along with a significant increase in the total serum serotonin levels. Gut microbiome analysis of the LF216EV-treated group revealed an altered gut microbiota profile. Correlation analysis revealed that the genera Limosilactobacillus and Desulfovibrio were associated with differences in the intestinal metabolites, including serotonin. Our findings demonstrate that LF216EV mitigates AD-like symptoms by promoting serotonin synthesis through the modulation of gut microbiota and metabolome composition.
Collapse
Affiliation(s)
- Hyejin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Min-Jin Kwak
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Youbin Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - An Na Kang
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Daye Mun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Ju Young Eor
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| | - Mi Ri Park
- Food Functionality Research Division, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Korea
| | - Sangnam Oh
- Department of Functional Food and Biotechnology, Jeonju University, Jeonju, Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea
| |
Collapse
|
2
|
Li S, Li J, Chen G, Lin T, Zhang P, Tong K, Chen N, Liu S. Exosomes originating from neural stem cells undergoing necroptosis participate in cellular communication by inducing TSC2 upregulation of recipient cells following spinal cord injury. Neural Regen Res 2025; 20:3273-3286. [PMID: 38993124 PMCID: PMC11881710 DOI: 10.4103/nrr.nrr-d-24-00068] [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: 01/18/2024] [Revised: 04/09/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00030/figure1/v/2024-12-20T164640Z/r/image-tiff We previously demonstrated that inhibiting neural stem cells necroptosis enhances functional recovery after spinal cord injury. While exosomes are recognized as playing a pivotal role in neural stem cells exocrine function, their precise function in spinal cord injury remains unclear. To investigate the role of exosomes generated following neural stem cells necroptosis after spinal cord injury, we conducted single-cell RNA sequencing and validated that neural stem cells originate from ependymal cells and undergo necroptosis in response to spinal cord injury. Subsequently, we established an in vitro necroptosis model using neural stem cells isolated from embryonic mice aged 16-17 days and extracted exosomes. The results showed that necroptosis did not significantly impact the fundamental characteristics or number of exosomes. Transcriptome sequencing of exosomes in necroptosis group identified 108 differentially expressed messenger RNAs, 104 long non-coding RNAs, 720 circular RNAs, and 14 microRNAs compared with the control group. Construction of a competing endogenous RNA network identified the following hub genes: tuberous sclerosis 2 ( Tsc2 ), solute carrier family 16 member 3 ( Slc16a3 ), and forkhead box protein P1 ( Foxp1 ). Notably, a significant elevation in TSC2 expression was observed in spinal cord tissues following spinal cord injury. TSC2-positive cells were localized around SRY-box transcription factor 2-positive cells within the injury zone. Furthermore, in vitro analysis revealed increased TSC2 expression in exosomal receptor cells compared with other cells. Further assessment of cellular communication following spinal cord injury showed that Tsc2 was involved in ependymal cellular communication at 1 and 3 days post-injury through the epidermal growth factor and midkine signaling pathways. In addition, Slc16a3 participated in cellular communication in ependymal cells at 7 days post-injury via the vascular endothelial growth factor and macrophage migration inhibitory factor signaling pathways. Collectively, these findings confirm that exosomes derived from neural stem cells undergoing necroptosis play an important role in cellular communication after spinal cord injury and induce TSC2 upregulation in recipient cells.
Collapse
Affiliation(s)
- Shiming Li
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Jianfeng Li
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Guoliang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Lin
- Department of Orthopedics and Traumatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Penghui Zhang
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Kuileung Tong
- Department of Orthopedic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ningning Chen
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong Province, China
| | - Shaoyu Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
3
|
Chen J, Li Y, Quan X, Chen J, Han Y, Yang L, Zhou M, Mok GSP, Wang R, Zhao Y. Utilizing engineered extracellular vesicles as delivery vectors in the management of ischemic stroke: a special outlook on mitochondrial delivery. Neural Regen Res 2025; 20:2181-2198. [PMID: 39101653 PMCID: PMC11759020 DOI: 10.4103/nrr.nrr-d-24-00243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 08/06/2024] Open
Abstract
Ischemic stroke is a secondary cause of mortality worldwide, imposing considerable medical and economic burdens on society. Extracellular vesicles, serving as natural nano-carriers for drug delivery, exhibit excellent biocompatibility in vivo and have significant advantages in the management of ischemic stroke. However, the uncertain distribution and rapid clearance of extracellular vesicles impede their delivery efficiency. By utilizing membrane decoration or by encapsulating therapeutic cargo within extracellular vesicles, their delivery efficacy may be greatly improved. Furthermore, previous studies have indicated that microvesicles, a subset of large-sized extracellular vesicles, can transport mitochondria to neighboring cells, thereby aiding in the restoration of mitochondrial function post-ischemic stroke. Small extracellular vesicles have also demonstrated the capability to transfer mitochondrial components, such as proteins or deoxyribonucleic acid, or their sub-components, for extracellular vesicle-based ischemic stroke therapy. In this review, we undertake a comparative analysis of the isolation techniques employed for extracellular vesicles and present an overview of the current dominant extracellular vesicle modification methodologies. Given the complex facets of treating ischemic stroke, we also delineate various extracellular vesicle modification approaches which are suited to different facets of the treatment process. Moreover, given the burgeoning interest in mitochondrial delivery, we delved into the feasibility and existing research findings on the transportation of mitochondrial fractions or intact mitochondria through small extracellular vesicles and microvesicles to offer a fresh perspective on ischemic stroke therapy.
Collapse
Affiliation(s)
- Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Li Yang
- Department of Pharmacy, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Manfei Zhou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Greta Seng Peng Mok
- Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| |
Collapse
|
4
|
Tian L, Jin J, Lai F, Yao S, Zhang Y, Liu J, Zhang H, Lu Q, Liu C, Tian S, Lu Y, Liang Y, Zhao Y, Fan H, Ren W. Nebulized M2 macrophage-derived nanovesicles for the treatment of explosion-induced acute lung injury. J Colloid Interface Sci 2025; 691:137381. [PMID: 40187079 DOI: 10.1016/j.jcis.2025.137381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025]
Abstract
Gas explosion-induced acute lung injury (ALI) presents a significant clinical challenge in industrial and military contexts, yet current therapeutic interventions remain suboptimal. Extracellular vesicles (EVs) have emerged as promising nanoscale therapeutic agents, owing to their superior biocompatibility and inherent therapeutic potential. In this study, we engineered and isolated alternatively activated macrophages extracellular vesicles (M2-EVs) and administered them via nebulization to the injured lung tissue in an established murine model of blast-induced ALI. Administration of M2-EVs led to a significant amelioration of pulmonary function, characterized by decreased lung injury scores and attenuated inflammatory markers. Mechanistically, M2-EVs promoted the M1-to-M2 phenotypic transition of pulmonary macrophages, modulated the P62-Keap1-Nrf2 signaling pathway, and consequently mitigated excessive autophagy and oxidative stress. Collectively, these findings not only offer mechanistic insights into the pathogenesis of blast-induced ALI but also highlight M2-EVs as a promising cell-free therapeutic approach for explosion-related pulmonary injuries.
Collapse
Affiliation(s)
- Linqiang Tian
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Jin
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Feng Lai
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Sanqiao Yao
- Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China; School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Yue Zhang
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Jie Liu
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China
| | - Huajing Zhang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Qianying Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Chuanchuan Liu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Sijia Tian
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Yujia Lu
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Yangfan Liang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China
| | - Yanmei Zhao
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China.
| | - Haojun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Tianjin Key Laboratory of Disaster Medical Technology, Tianjin 300072, China.
| | - Wenjie Ren
- Henan Medical Key Laboratory for Research of Trauma and Orthopedics, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan Province 453003, China; Institutes of Health Central Plain, Xinxiang Medical University, Xinxiang, Henan Province 453003, China.
| |
Collapse
|
5
|
Ahn W, Han J, Kim N, Hwang YH, Kim W, Lee Y, Lee DY, Cheong IW, Han K, Nam GH, Kim IS, Lee EJ. Hierarchical protein nano-crystalline hydrogel with extracellular vesicles for ectopic lymphoid structure formation. Biomaterials 2025; 318:123166. [PMID: 39933315 DOI: 10.1016/j.biomaterials.2025.123166] [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: 10/17/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Among cancer therapies, immune checkpoint blockade (ICB) has emerged as a prominent approach, substantially enhancing anti-tumor immune responses. However, the efficacy of ICB is often limited in the absence of a pre-existing immune response within the tumor microenvironment. Here, we introduce a novel hierarchical protein hydrogel platform designed to facilitate the formation of artificial tertiary lymphoid structures (aTLS), thereby improving ICB efficacy. Through the integration of self-assembling ferritin protein nanocages, rec1-resilin protein, and CP05 peptide, our hierarchical hydrogels provide a structurally supportive and functionally adaptive scaffold capable of on-demand self-repair in response to mild thermal treatments. The effective encapsulation of extracellular vesicles (EVs) via the CP05 peptide ensures the formation of aTLS with germinal center-like structures within the hierarchical hydrogel. We demonstrate that, combined with ICB therapy, EV-loaded hierarchical hydrogels also induce the TLS within the tumor, markedly promoting immune responses against ICB-resistant tumor. This bioactive hydrogel platform offers a versatile tool for enhancing a broad range of immunotherapies, with potential applications extending beyond TLS to other frameworks that support complex tissue architectures.
Collapse
Affiliation(s)
- Wonkyung Ahn
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Jihoon Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeong Ha Hwang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Wonjun Kim
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dong Yun Lee
- Department of Polymer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - In Woo Cheong
- Department of Applied Chemistry, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Koohee Han
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Gi-Hoon Nam
- Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02842, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Chemical Engineering and Applied Chemistry, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
6
|
Bonizzi A, Signati L, Grimaldi M, Truffi M, Piccotti F, Gagliardi S, Dotti G, Mazzucchelli S, Albasini S, Cazzola R, Bhowmik D, Narayana C, Corsi F, Morasso C. Exploring breast cancer-related biochemical changes in circulating extracellular vesicles using Raman spectroscopy. Biosens Bioelectron 2025; 278:117287. [PMID: 40023908 DOI: 10.1016/j.bios.2025.117287] [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: 10/25/2024] [Revised: 12/31/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Extracellular vesicles (EVs) are a subgroup of the circulating particles, released by cells in both normal and diseased states, carrying active biomolecules. They have gained significant attention as potential cancer biomarkers, particularly in breast cancer (BC). Previous research showed variations in EVs content and quantity between BC patients and healthy controls (HC). However, studying the biochemical profile of EVs remains challenging due to their low abundance and complex composition. Additionally, EVs may interact with other plasma components, like lipoproteins (LPs), forming a so called "biomolecular corona" that further complicates their analysis. Here, Raman spectroscopy (RS) is proposed as a fast tool to obtain the biochemical profile of circulating EVs in the context of BC. RS was employed to differentiate various extracellular particles (EPs) in blood, including LPs and EVs. The study also evaluated RS's capability to quantify major classes of biomolecules and compared these results with those obtained by traditional biochemical assays. Finally, compositional differences in large EVs (lEVs) and small EVs (sEVs) were assessed between HC and BC patients. RS revealed the existence of distinct biochemical signatures associated with BC, highlighting increased levels of nucleic acids and lipids in the BC group.
Collapse
Affiliation(s)
- Arianna Bonizzi
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Via Giovanni Battista Grassi, 74, 20157, Milan, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Lorena Signati
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Via Giovanni Battista Grassi, 74, 20157, Milan, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Maria Grimaldi
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Francesca Piccotti
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics Unit, IRCCS Mondino Foundation, Via Mondino 2, Pavia, 27100, Italy
| | - Giulia Dotti
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Serena Mazzucchelli
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Via Giovanni Battista Grassi, 74, 20157, Milan, Italy
| | - Sara Albasini
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy
| | - Roberta Cazzola
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Via Giovanni Battista Grassi, 74, 20157, Milan, Italy
| | - Debanjan Bhowmik
- Transdisciplinary Biology Program, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O., Poojappura, Thiruvananthapuram, 695014, India
| | - Chandrabhas Narayana
- Transdisciplinary Biology Program, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O., Poojappura, Thiruvananthapuram, 695014, India; Chemistry and Physics of Materials Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bangalore, 560064, India
| | - Fabio Corsi
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Via Giovanni Battista Grassi, 74, 20157, Milan, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy.
| | - Carlo Morasso
- Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 4, Pavia, 27100, Italy.
| |
Collapse
|
7
|
Getachew H, Mehrotra S, Kaur T, Fernandez-Godino R, Pierce EA, Garita-Hernandez M. The RNA content of extracellular vesicles from gene-edited PRPF31 +/- hiPSC-RPE show potential as biomarkers of retinal degeneration. Mol Ther Methods Clin Dev 2025; 33:101452. [PMID: 40231248 PMCID: PMC11995067 DOI: 10.1016/j.omtm.2025.101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025]
Abstract
Retinitis pigmentosa (RP) is the most common inherited retinal degeneration (IRD), causing vision loss via the dysfunction and death of photoreceptors and retinal pigment epithelium (RPE). Mutations in the PRPF31 gene are associated with autosomal dominant RP, impairing RPE function. While adeno-associated virus (AAV)-mediated gene therapy shows promise for treating IRDs, the slow progression of these diseases often makes timely measurement of clinical efficacy challenging. Extracellular vesicles (EVs) are lipid enclosed vesicles secreted by cells, and their RNA contents are being explored as circulating biomarkers for other diseases. We hypothesize that EV RNAs could serve as biomarkers of the health status of the neural retina and RPE. To test this, we used PRPF31 +/+ and PRPF31 +/- human induced pluripotent stem cell (hiPSC)-derived RPE (hi-RPE) to investigate the RNAs contained in RPE-derived EVs and how they change in disease. We also compared the RNA contents of RPE-EVs with the RNAs of the hi-RPE cells themselves. We found that EVs from mutant PRPF31 hi-RPE cells have distinct RNA profiles compared to those from control cells, suggesting that EV RNA contents change during disease. Additionally, we identified 18 miRNAs and 865 poly(A) RNAs enriched in EVs from PRPF31 +/- hi-RPE, which could serve as biomarkers for RPE degeneration.
Collapse
Affiliation(s)
- Heran Getachew
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sudeep Mehrotra
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Tarandeep Kaur
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Rosario Fernandez-Godino
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Eric A. Pierce
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Marcela Garita-Hernandez
- Ocular Genomics Institute, Department of Ophthalmology, Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Saint-Pol J, Culot M. Minimum information for studies of extracellular vesicles (MISEV) as toolbox for rigorous, reproducible and homogeneous studies on extracellular vesicles. Toxicol In Vitro 2025; 106:106049. [PMID: 40074066 DOI: 10.1016/j.tiv.2025.106049] [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: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Studies based on extracellular vesicles (EVs) have been multiplying exponentially for almost two decades, since they were first identified as vectors of cell-cell communication. However, several of these studies display a lack of rigor in EVs characterization and isolation, without discriminating between the different EV populations, thus generating conflicting and unreproducible results. There is therefore a strong need for standardization and guidelines to conduct studies that are rigorous, transparent, reproducible and comply with certain nomenclatures concerning the type of EVs used. The International Society for Extracellular Vesicles (ISEV) published the Minimum Information for Studies of Extracellular Vesicles (MISEV) in 2014, updating it in 2018 and 2023 to reflect different study contexts and technical advancements. The primary objective of this review is to inform future authors about EVs, including their history, nomenclature, and technical recommendations for the for isolation and functionality analysis for conducing EV-based studies according to current standards. Additionally, it aims to inform reviewers about the key parameters required for characterizing EV preparations.
Collapse
Affiliation(s)
- Julien Saint-Pol
- Univ. Artois, UR 2465, Blood-Brain Barrier laboratory (LBHE), F-62300 Lens, France.
| | - Maxime Culot
- Univ. Artois, UR 2465, Blood-Brain Barrier laboratory (LBHE), F-62300 Lens, France
| |
Collapse
|
9
|
Tu M, Liu A, Huang W, Wang D, Chen H, Hu X. Macrophages-derived small extracellular vesicles regulate chondrocyte proliferation and affect osteoarthritis progression via upregulating Osteopontin expression. J Cell Commun Signal 2025; 19:e70008. [PMID: 40264984 PMCID: PMC12012988 DOI: 10.1002/ccs3.70008] [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: 06/19/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 04/24/2025] Open
Abstract
Small extracellular vesicles (sEVs) are considered promising gene-delivery vehicles for the treatment of osteoarthritis (OA). This study aimed to explore the molecular mechanism by which M2 macrophage-derived sEVs (M2-sEVs) modulate chondrocyte proliferation and apoptosis, thereby affecting OA progression. M2 macrophages were successfully induced, and M2-sEVs were successfully isolated. The sEVs were small vesicles with diameters ranging from 50 to 150 nm. The exosomal markers, including CD9, CD63, and CD81, were highly expressed, whereas the negative marker calnexin was absent in M2-sEVs. M2-sEVs effectively alleviated OA tissue and chondrocyte damage in both in vivo and in vitro models, evidenced by reduced rat knee joint injury, increased chondrocyte viability, and decreased chondrocyte apoptosis and extracellular matrix (ECM) degradation. Furthermore, M2-sEVs decreased the levels of pro-inflammatory cytokines IL-6 and TNF-α. Osteopontin (OPN) was upregulated within rats with OA and IL-1β-induced chondrocytes. Silencing of OPN exacerbated IL-1β-induced chondrocyte damage and partially abrogated the therapeutic effects of M2-sEVs. Additionally, M2-sEVs enhanced OPN expression and activated CD44 and the PI3K/AKT signaling pathway. In conclusion, M2-sEVs promoted OPN expression to improve knee joint tissue damage in rats with OA and chondrocyte damage. This protective effect of M2-sEVs might be associated with the activation of CD44 and the PI3K/AKT signaling.
Collapse
Affiliation(s)
- Min Tu
- Department of OrthopedicsThe People's Hospital of JingmenJingmenHubeiChina
- Jingmen People's Hospital Affiliated to Jingchu University of TechnologyJingmenHubeiChina
| | - An‐Min Liu
- Department of OrthopedicsThe People's Hospital of JingmenJingmenHubeiChina
- Jingmen People's Hospital Affiliated to Jingchu University of TechnologyJingmenHubeiChina
| | - Wei Huang
- Department of OrthopedicsThe People's Hospital of JingmenJingmenHubeiChina
- Jingmen People's Hospital Affiliated to Jingchu University of TechnologyJingmenHubeiChina
| | - Dan Wang
- Department of OrthopedicsThe People's Hospital of JingmenJingmenHubeiChina
- Jingmen People's Hospital Affiliated to Jingchu University of TechnologyJingmenHubeiChina
| | - Hou‐Qiong Chen
- Department of OrthopedicsThe People's Hospital of JingmenJingmenHubeiChina
- Jingmen People's Hospital Affiliated to Jingchu University of TechnologyJingmenHubeiChina
| | - Xiao‐Yuan Hu
- Department of OrthopedicsThe People's Hospital of JingmenJingmenHubeiChina
- Jingmen People's Hospital Affiliated to Jingchu University of TechnologyJingmenHubeiChina
| |
Collapse
|
10
|
Liu X, Ding L, Zhang A, Feng F, Zhou F, Wu Y. Dynamic characteristics of metabolism and small extracellular vesicles during malignant transformation of BEAS-2B cells induced by coal tar pitch extract. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126108. [PMID: 40154873 DOI: 10.1016/j.envpol.2025.126108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Lung cancer poses a significant global burden with rising morbidity and mortality. Coal tar pitch-induced lung cancer is an occupational disease where early detection is crucial but challenging due to unclear pathogenesis. We established a malignant transformation model using BEAS-2B cells treated with coal tar pitch extract (CTPE). Macro- and micro-observations showed CTPE-induced alterations, including changes in cell morphology, enhanced proliferation and migration abilities, upregulated EGFR expression, modified levels of CYP1A1 and GSTM1 metabolizing enzymes, and a transition towards a mesenchymal phenotype. These findings strongly suggest that the cells have undergone malignant transformation. Metabolomics analysis revealed changes in 1120 metabolites, with 31 co-expressed, mainly in energy and amino acid metabolism. Small extracellular vesicles (SEVs) concentrations and EGFR levels were significantly altered. Correlation analysis identified a relationship between these biomarkers, implying their potential significance as early events in the initiation and progression of lung cancer. These findings provide valuable insights and a rationale for lung cancer screening and mechanistic investigations, thereby contributing to a deeper understanding of the disease.
Collapse
Affiliation(s)
- Xia Liu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Lihua Ding
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Aiai Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Fang Zhou
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
11
|
Gu X, Fan Z, Lu L, Xu H, He L, Shen H, Huang R, Li Z. Machine learning-assisted washing-free detection of extracellular vesicles by target recycling amplification based fluorescent aptasensor for accurate diagnosis of gastric cancer. Talanta 2025; 287:127506. [PMID: 39837204 DOI: 10.1016/j.talanta.2024.127506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/23/2025]
Abstract
Extracellular vesicles (EVs) are promising non-invasive biomarkers for cancer diagnosis. EVs proteins play a critical role in tumor progress and metastasis. However, accurately and reliably diagnosing cancers is greatly limited by single protein marker on EVs. Here, we reported an accurate diagnosis model of gastric cancer by analyzing five types of EVs surface proteins using machine learning in a retrospective study design. A washing-free detection method based on aptasensor and exonuclease Ⅰ was used to profile EVs surface proteins. The aptamer was designed as hairpin structure. The presence of target protein positive EVs converted the conformation of hairpin probes, which subsequently degraded by exonuclease Ⅰ. The exposed target protein could bind with and then open new hairpin probes, thus forming an amplification cycle. The lengths of different detection probes were optimized for detection. With the combination of five target proteins, five machine learning algorithms were compared to achieve a higher diagnostic accuracy. The best model, XGBoost, validated with 20 % of detection results could reach an accuracy of 0.8421. Furthermore, the XGBoost-based surface protein analysis could precisely identify gastric cancer patients with the area under the curve value of 0.9347 (95 % confidential interval (CI) = 0.8590 to 1.000). Since our method utilized a simple and versatile design of detection probes, its diagnostic scope could potentially be expanded to include different protein markers and accurately diagnose other diseases in the future.
Collapse
Affiliation(s)
- Xinrui Gu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Zeyu Fan
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China
| | - Luying Lu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Hongpan Xu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Lei He
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China.
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China.
| | - Zhiyang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| |
Collapse
|
12
|
Vader P. Extracellular vesicles for drug delivery: A major interest for the Journal of Controlled Release. J Control Release 2025; 381:113599. [PMID: 40049519 DOI: 10.1016/j.jconrel.2025.113599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Affiliation(s)
- Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
13
|
Coelho MO, Quintas ST, Sarmento B, De Wever O, Castro F. Engineered dendritic cells-derived extracellular vesicles for cancer immunotherapy. J Control Release 2025; 381:113620. [PMID: 40088976 DOI: 10.1016/j.jconrel.2025.113620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
Extracellular vesicles (EVs) have emerged as a cell-free therapeutic approach, garnering increasing attention for their potential to enhance the safety and efficacy of immunotherapy. This interest is primarily driven by the biocompatibility and cell/tissue tropism inherent to EVs, but also due to their reconfigurable content. This, termed as cargo, may comprise bioactive molecules as proteins, lipids, and nucleic acids that play a pivotal role in mediating intercellular communication. In particular, dendritic cells-derived extracellular vesicles (DC-EVs) facilitate the transfer of critical components, like antigens and immune-regulatory factors, and due to the expression of major histocompatibility complexes and co-stimulatory molecules on their surface can activate T cells, thereby modulating the immune response. Additionally, DC-EVs can be engineered to transport tumor-specific antigens, cytokines, or other agents in order to strength their immunotherapeutic potential, and even be used in vaccines formulation. In this review, the latest advancements in engineering DC-EVs to improve their immunotherapeutic potential is discussed in detail, while also addressing current challenges associated with DC-EVs therapies.
Collapse
Affiliation(s)
- Margarida Oliveira Coelho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sofia Torres Quintas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; IUCS-CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Olivier De Wever
- CRIG - Cancer Research Institute Ghent, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium; LECR - Laboratory Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal.
| |
Collapse
|
14
|
Jing S, Liu Y, Wang B, Zhou H, Zhang H, Siwakoti P, Qu X, Ye P, He Y, Kumeria T, Ye Q. Microneedle-mediated hypoxic extracellular vesicle-encapsulated selenium nanoparticles delivery to treat androgenetic alopecia. J Control Release 2025; 381:113597. [PMID: 40043914 DOI: 10.1016/j.jconrel.2025.113597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/20/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
Androgenetic alopecia (AGA) is the most common type of hair loss, and there is a lack of ideal treatment options. The damage and shedding of hair follicles are closely associated with niche dysregulation, including reactive oxygen species (ROS) accumulation, microvascular damage, and persistent inflammation. In this study, a biocomposite microneedle system comprising hypoxic extracellular vesicle (EV)-encapsulated selenium nanozyme (Se-HEVs-AMN) was designed to create a favorable perifollicular microenvironment. The novel Se-HEVs-AMN biocomposite patch features microneedles with sufficient mechanical strength, tailored dissolution properties, and a convenient detachable backing layer. The microneedles are modified with Astragalus polysaccharide (APS) and loaded with hypoxia-induced EVs containing selenium nanozyme. When applied to the dorsal skin of AGA mice, the microneedles rapidly dissolve, releasing active ingredients that increase hair density and enlarge hair follicle diameter through regulating inflammation, promoting angiogenesis, scavenging ROS, and resisting androgen.
Collapse
Affiliation(s)
- Shuili Jing
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Stomatology, Linhai Second People's Hospital, Linhai 317000, China; Cell Therapy Center, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Yonghao Liu
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ben Wang
- Department of Stomatology, Linhai Second People's Hospital, Linhai 317000, China
| | - Heng Zhou
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hui Zhang
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Prakriti Siwakoti
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiangyu Qu
- School of Computer Science, Wuhan University, Wuhan 430072, China
| | - Peng Ye
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan 430064, China; Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston 02114, MA, USA.
| | - Tushar Kumeria
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW 2052, Australia; School of Pharmacy, University of Queensland, Brisbane, QLD 4102, Australia; Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Qingsong Ye
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Stomatology, Linhai Second People's Hospital, Linhai 317000, China.
| |
Collapse
|
15
|
Song LL, Tang YP, Qu YQ, Yun YX, Zhang RL, Wang CR, Wong VKW, Wang HM, Liu MH, Qu LQ, Wu JH, Lo HH, Law BYK. Exosomal delivery of rapamycin modulates blood-brain barrier penetration and VEGF axis in glioblastoma. J Control Release 2025; 381:113605. [PMID: 40058500 DOI: 10.1016/j.jconrel.2025.113605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/04/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Exosomes (Exos), nanosized membranous vesicles (30-160 nm), have been validated as an effective drug delivery system capable of traversing biological barriers. Mesenchymal stem cells (MSCs), due to their near-limitless self-renewal capabilities, provide a plentiful source of exosomes for clinical applications. In this study, we utilized an exosome-encapsulated rapamycin (Exo-Rapa) delivery strategy, which permits the use of smaller drug dosages to achieve effects typically seen with higher dosages, thus enhancing drug efficacy. Moreover, Exos can transport pharmaceuticals across the blood-brain barrier (BBB) to the brain, and further penetrate GL261 cells to exert their effects. Within the tumor microenvironment, Exo-Rapa is released more rapidly and efficiently at the tumor site. The acidic conditions in tumors accelerate the release of Exo-Rapa, a characteristic that may make it a promising targeted therapeutic in future cancer research. Additionally, a series of in vivo experiments have further demonstrated the permeability of Exo-Rapa across the BBB, enabling it to accumulate at tumor sites; it also ameliorates inflammatory responses in Glioblastoma multiforme (GBM) mouse models and enhances anti-tumor activity through the regulation of angiogenesis via the VEGF/VEGFRs axis. Our results indicate that MSC-derived exosomes are a potent therapeutic carrier for GBM, offering an effective strategy for enhancing drug delivery across the BBB and providing a scientific foundation for the use of exosomes in the treatment of GBM and other diseases.
Collapse
Affiliation(s)
- Lin Lin Song
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yong Pei Tang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuan Qing Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yun Xiao Yun
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Rui Long Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Cai Ren Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, China
| | - Hui Miao Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Meng Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Li Qun Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian Hui Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hang Hong Lo
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Betty Yuen Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, China.
| |
Collapse
|
16
|
Kaur J, Aggarwal J, Behera M, Dahiya TR, Kumar D, Dhania NK. Size exclusion chromatography: An efficient tool for hsEV isolation from insect Haemolymph. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1257:124570. [PMID: 40188567 DOI: 10.1016/j.jchromb.2025.124570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/08/2025]
Abstract
Extracellular vesicles (EV) serve an important role in biological system as they can change the phenotype and function of the cell. The discovery of EVs has led to the development of novel vaccines, associated with immunosuppressive and immunostimulatory effects during disease progression. To address the opportunities in EVs, here we used insect as model system. The insect also has medicinal properties as it produces antimicrobial peptides which are found circulating fluid called as hemolymph. Hemolymph consists of defensins, gloverins, proline-rich peptides, attacins, cecropins, moricins, anionic antimicrobial peptides, and several other components that create robust defense mechanisms against a broad spectrum of pathogens. Here, the insect hemolymph was used for EV isolation which encapsulates such kinds of proteins and acts as cargo transporters. As the biogenesis was unknown, we have defined it as hemolymph-derived small extracellular vesicles (hsEV). The present paper deals with the appropriate methodology for the isolation of hsEV. The hemolymph was processed for isolation via size exclusion chromatography. Molecular and physical characterization was performed using western blotting utilizing anti-CD63, anti-Flotillin-2, TEM, FESEM, confocal microscopy, and DLS. The isolated hsEV was used to assess the antibacterial property by measuring bacterial growth.
Collapse
Affiliation(s)
- Jaspreet Kaur
- Department of Zoology, Faculty of Sciences, University of Delhi, Delhi 110007, India
| | - Janvi Aggarwal
- Department of Zoology, Faculty of Sciences, University of Delhi, Delhi 110007, India
| | - Mukesh Behera
- Department of Zoology, Faculty of Sciences, University of Delhi, Delhi 110007, India
| | - Tannu Rachna Dahiya
- Department of Zoology, Faculty of Sciences, University of Delhi, Delhi 110007, India
| | - Dhiraj Kumar
- Centre for Skill Development and Seri technology, Department of Zoology, Hansraj College, University of Delhi, Delhi 110007, India
| | - Narender K Dhania
- Department of Zoology, Faculty of Sciences, University of Delhi, Delhi 110007, India.
| |
Collapse
|
17
|
Abida, Alhuthali HM, Alshehri JM, Alkathiri A, Almaghrabi ROM, Alsaeed SS, Albebi SAH, Almethn RM, Alfuraydi BA, Alharbi SB, Kamal M, Imran M. Exosomes in infectious diseases: insights into leishmaniasis pathogenesis, immune modulation, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4913-4931. [PMID: 39702600 DOI: 10.1007/s00210-024-03702-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Leishmaniasis continues to be a critical international health issue due to the scarcity of efficient treatment and the development of drug tolerance. New developments in the research of extracellular vesicles (EVs), especially exosomes, have revealed novel disease management approaches. Exosomes are small vesicles that transport lipids, nucleic acids, and proteins in cell signalling. Its biogenesis depends on several cellular processes, and their functions in immune response, encompassing innate and adaptive immunity, underline their function in the pathogen-host interface. Exosomes play a significant role in the pathogenesis of some parasitic infections, especially Leishmaniasis, by helping parasites escape host immunity and promote disease progression. This article explains that in the framework of parasitic diseases, exosomes can act as master regulators that define the pathogenesis of the disease, as illustrated by the engagement of exosomes in the Leishmaniasis parasite and immune escape processes. Based on many published articles on Leishmaniasis, this review aims to summarize the biogenesis of exosomes, the properties of the cargo in exosomes, and the modulation of immune responses. We delve deeper into the prospect of using exosomes for the therapy of Leishmaniasis based on the possibility of using these extracellular vesicles for drug delivery and as diagnostic and prognostic biomarkers. Lastly, we focus on the recent research perspectives and future developments, underlining the necessity to continue the investigation of exosome-mediated approaches in Leishmaniasis treatment. Thus, this review intends to draw attention to exosomes as a bright new perspective in the battle against this disabling affliction.
Collapse
Affiliation(s)
- Abida
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Jawaher Mohammad Alshehri
- Optometry Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Afnan Alkathiri
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | - Ruba Omar M Almaghrabi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Albaha University, 65431, Albaha, Saudi Arabia
| | | | | | | | | | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| |
Collapse
|
18
|
Kostevšek N. Erythrocyte membrane vesicles as drug delivery systems: A systematic review of preclinical studies on biodistribution and pharmacokinetics. BIOMATERIALS ADVANCES 2025; 170:214234. [PMID: 39961269 DOI: 10.1016/j.bioadv.2025.214234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 03/05/2025]
Abstract
This systematic review aims to summarize the development of erythrocyte membrane vesicles (EMVs) as drug delivery carriers, with a focus on elucidating their fate in terms of biodistribution and pharmacokinetics in preclinical studies. The PubMed database was systematically reviewed to search for original peer-reviewed published studies on the use of EMVs for drug delivery to summarize the preclinical findings, following the PRISMA guidelines. A total of 142 articles matched the selection criteria and were included in the review. For each study, the following parameters were extracted: type of active pharmaceutical ingredient (API) encapsulated into EMVs, EMVs-API formulation method and final particle size, EMVs surface modifications for active targeting, cell lines and animal models used in the study, crucial treatment data, biodistribution data and finally, where applicable, data about the EMVs circulation time and blood half-life. EMVs size did not vary significantly among the different formulation methods. A complete list of cell lines and animal models used is provided. Circulation times and data for blood half-life were grouped per animal type. For the most commonly used animal type, BALB/c mice, the average half-life of EMV-API was calculated to be 10.4 h, and in all cases, up to a 10-fold increase was observed compared with that of free API. Surface modifications did not drastically change the circulation time but did improve target tissue accumulation. The most critical weaknesses in the analysed studies were identified. Key points for future studies are provided to fill the current knowledge gaps and improve the quality of publications.
Collapse
Affiliation(s)
- Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, 1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, 1000 Ljubljana, Slovenia.
| |
Collapse
|
19
|
Nouri M, Nasiri F, Sharif S, Abbaszadegan MR. Unraveling extracellular vesicle DNA: Biogenesis, functions, and clinical implications. Pathol Res Pract 2025; 269:155937. [PMID: 40199015 DOI: 10.1016/j.prp.2025.155937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
Extracellular Vesicles (EVs) have emerged as essential carriers of molecular biomarkers and mediators of intercellular communication. While previous studies have predominantly focused on EV proteins, lipids, and RNA, the role of EV-derived DNA (EV-DNA) remains relatively unexplored. Understanding EV-DNA is crucial, given its association with nearly all EV populations. This review aims to comprehensively summarize existing EV-DNA research, emphasizing its functional significance and potential as a disease biomarker. By bridging the gap in our understanding, we shed light on the origins, structure, localization, and distribution of EV-DNA. We analyze a wide range of studies, investigating EV-DNA across various pathological conditions. Our review encompasses experimental methods, theoretical approaches, and clinical observations, providing a holistic view of EV-DNA research. We discuss the biogenesis mechanisms of different EV subtypes, the available isolation methods for these subtypes, and consider their origins and variability under different conditions. EV-DNA exhibits remarkable stability and reflects genomic alterations, making it a promising candidate for liquid biopsy applications. From cancer diagnostics to treatment monitoring, EV-DNA holds significant potential. The findings underscore the importance of EV-DNA as an innovative biomarker. As research continues, EV-DNA may revolutionize disease detection, prognosis, and therapeutic strategies.
Collapse
Affiliation(s)
- Mehraneh Nouri
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fateme Nasiri
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Sharif
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Jung D, Kim NE, Kim S, Bae JH, Jung IY, Doh KW, Lee B, Kim DK, Cho YE, Baek MC. Plant-derived nanovesicles and therapeutic application. Pharmacol Ther 2025; 269:108832. [PMID: 40023319 DOI: 10.1016/j.pharmthera.2025.108832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/27/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Plant-derived nanovesicles (PDNVs) are becoming more popular as promising therapeutic tools owing to their diversity, cost-effectiveness, and biocompatibility with very low toxicity. Therefore, this review aims to discuss the methods for isolating and characterizing PDNVs and emphasize their versatile roles in direct therapeutic applications and drug delivery systems. Their ability to effectively encapsulate and deliver large nucleic acids, proteins, and small-molecule drugs was highlighted. Moreover, advanced engineering strategies, such as surface modification and fusion with other vesicles, have been developed to enhance the therapeutic effects of PDNVs. Additionally, we describe key challenges related to this field, encouraging further research to optimize PDNVs for various clinical applications for prevention and therapeutic purposes. The distinctive properties and diverse applications of PDNVs could play a crucial role in the future of personalized medicine, fostering the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Dokyung Jung
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Na-Eun Kim
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Sua Kim
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ju-Hyun Bae
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Il-Young Jung
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Kyung-Won Doh
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Do-Kyun Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 54531, Republic of Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong 36729, 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.
| |
Collapse
|
21
|
Haizan I, Choi MY, Park DH, Choi JH. Dual-target magneto-immunoassay with bifunctional nanohybrids for breast cancer exosome detection. Talanta 2025; 286:127532. [PMID: 39788070 DOI: 10.1016/j.talanta.2025.127532] [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: 11/08/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Exosomes, crucial for intercellular communication, hold potential as noninvasive liquid biopsy biomarkers especially in early breast cancer detection benefitted from the distinctive "cancer signature" on their membrane surface. Yet, the present methodologies of exosomes for breast cancer detection have involved the implementation of only a single member from the tetraspanin protein group as a biomarker. Moreso, due to the high concentration of exosomes in complex body fluids, there is a compelling need to measure a small concentration of cancer-derived exosomes with a low background noise signal. In this study, we designed and characterized magnetic core-gold shell nanohybrids (mAuNHs) that function as detection and isolator probes, which were integrated in a simple colorimetric sandwich magneto-immunoassay (mLISA). The magnetic core of mAuNHs facilitates the separation of exosomes from complex samples of biological origin whereby amorphous structures were effectively removed, decreasing background signal. Meanwhile, the coalescence effect of pairing biologically abundance exosomal marker (CD9 antibody) with the cancer specific (CD24 antibody) offers a highly selective and sensitive detection of our target model, MCF7 exosomes. As a result, using our mLISA system, exosomes derived from MCF7 can be selectively recognized from other tested cancer cell lines, BT474 and PC3. Besides, as low as 37 particles/μL of limit of detection (LOD) was achieved using mLISA sensor, exhibiting a good sensitivity as compared to conventional ELISA. Overall, our proposed dual-target biosensor offers a great reduction on background noise from samples, simplicity for users as in exosome's lengthy preparation is reduced as well as good sensitivity.
Collapse
Affiliation(s)
- Izzati Haizan
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| | - Min Yu Choi
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| | - Dong Hyeok Park
- School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| | - Jin-Ha Choi
- Department of Bioprocess Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea; School of Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896, Republic of Korea.
| |
Collapse
|
22
|
Machimbirike VI, Onireti O, Chukwu-Osazuwa J, Cao T, Vasquez I, Rise ML, Kumar S, Santander J. Proteomics analysis of extracellular vesicles during Vibrio anguillarum infection in lumpfish (Cyclopterus lumpus). FISH & SHELLFISH IMMUNOLOGY 2025; 160:110205. [PMID: 39970972 DOI: 10.1016/j.fsi.2025.110205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 02/21/2025]
Abstract
Lumpfish (Cyclopterus lumpus) is a native fish of the North Atlantic Ocean used as sea lice biocontrol in Atlantic salmon farms. Lumpfish also has been used as model for marine infectious diseases and immunity. Lumpfish is susceptible to Vibrio anguillarum infection, and upon infection, lumpfish immunity is activated to preclude the disease progression. Extracellular vesicles (EVs) play an important role in early immune cellular communication. Lumpfish EVs and their potential role in immunity have not been explored. Herein, EVs where isolated from serum of naïve lumpfish and from lumpfish infected with V. anguillarum at 5 and 10 days post infection (dpi). EVs characteristics were studied by electron microscopy and nanoparticle tracking, and protein cargo was analysed by Western blot and proteomic analysis. The isolated EVs showed a spherical shape ranging from ∼30 nm to 300 nm in diameter, but at 5 dpi the size variation was higher. A total of 395 proteins were identified. Upregulated proteins were linked to complement pathway/innate immunity, heme/iron binding, defense response to bacterium, apoptotic signaling pathway, and actin binding. Downregulated proteins were associated with ribonucleoprotein/ribosomal protein, transport and translation elongation factor activity, acute phase, protein phosphorylation and apoptotic process. Upon infection V. anguillarum infection, lumpfish EVs cargo was modified, from transporting metabolic proteins to proteins related to immunity. Characterization of peripheral lumpfish EVs protein profile during V. anguillarum infection provided with potential biomarkers repertoire that could be utilised in the development of novel tools to diagnose and control of V. anguillarum infection in finfish aquaculture.
Collapse
Affiliation(s)
- V I Machimbirike
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, A1C 5S7, St. John's, Newfoundland and Labrador, Canada
| | - O Onireti
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, A1C 5S7, St. John's, Newfoundland and Labrador, Canada
| | - J Chukwu-Osazuwa
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, A1C 5S7, St. John's, Newfoundland and Labrador, Canada
| | - T Cao
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, A1C 5S7, St. John's, Newfoundland and Labrador, Canada
| | - I Vasquez
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, A1C 5S7, St. John's, Newfoundland and Labrador, Canada
| | - M L Rise
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1C 5S7, Canada
| | - S Kumar
- Department of Ocean Sciences, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, A1C 5S7, Canada
| | - J Santander
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, A1C 5S7, St. John's, Newfoundland and Labrador, Canada.
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
Wang K, Han X, Zhou W, Zhai J, Yuan Z, Li A, Du P. Integration of Macrogenomics and Metabolomics: Comprehensive Insights into the Effects of In Vitro Fermentation with Human Milk Exosomes on Infant Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40266004 DOI: 10.1021/acs.jafc.4c09971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
The role of human milk exosomes (HMEs) in maintaining infant intestinal health has attracted considerable attention, yet the mechanisms by which they regulate the infant gut microbiota remain to be elucidated. In this study, we constructed an in vitro fermentation model, combined with macrogenomics and nontargeted metabolomics technologies, to deeply analyze the effects of HMEs on the composition of intestinal microorganisms, the expression of functional genes, and the production of metabolites. It showed that HMEs significantly reduced the potential pathogenic bacteria like Escherichia coli, Klebsiella pneumoniae, Dysgonomonas capnocytophagoides, and Shigella flexneri, but increased Bacteroides fragilis and Bifidobacterium pseudocatenulatum. Moreover, HMEs promote key metabolic pathways including propionate and butyrate metabolism, glycolysis/glycogenesis, and amino acid metabolism. Consequently, beneficial intestinal metabolites such as short-chain fatty acids (SCFAs), amino acids, indoles, and secondary bile acids were elevated. It is speculated that HMEs may act as key signaling molecules or regulators to improve infant gut microecology.
Collapse
Affiliation(s)
- Kaili Wang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xueting Han
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Wenjia Zhou
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jianing Zhai
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Zekun Yuan
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Aili Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Green Food Research Institute, Harbin 150030, China
| | - Peng Du
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
27
|
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
|
28
|
Spesyvyi A, Cebecauer M, Žabka J, Olżyńska A, Malečková M, Johanovská Z, Polášek M, Charvat A, Abel B. Separation and Detection of Charged Unilamellar Vesicles in Vacuum by a Frequency-Controlled Quadrupole Mass Sensor. Anal Chem 2025. [PMID: 40260580 DOI: 10.1021/acs.analchem.4c05730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Extracellular vesicles (EVs) are membranous particles released by cells and are considered to be promising sources of biomarkers for various diseases. Mass spectrometry (MS) analysis of EVs requires a sample of purified and detergent-lysed EVs. Purification of EVs is laborious, based on size, density, or surface nature, and requires large amounts of the source material (e.g., blood, spinal fluid). We have employed synthetically produced large unilamellar lipid vesicles (LUVs) as analogs of EVs to demonstrate an alternative approach to vesicle separation for subsequent mass spectrometry analysis of their composition. Mass-to-charge ratio m/z separation by frequency-controlled quadrupole was employed to filter narrow-size distributions of LUVs from a water sample. Lipid vesicles were positively charged with nanoelectrospray and transferred into a vacuum using two wide m/z-range frequency-controlled quadrupoles. The m/z, charges, and masses of individual vesicles were obtained by the nondestructive single-pass charge detector. The resolving mode of the second quadrupole with m/z RSD < 10% allowed to separate size selected distributions of vesicles with modal diameters of 88, 112, 130, 162, and 190 nm at corresponding quadrupole m/z settings of 2.5 × 105, 5 × 105, 8 × 105, 1.5 × 106, and 2.5 × 106, respectively with a rate of 20-100 counts per minute. The distributions of bioparticles with masses between 108 and 1010 Da were separated from human blood serum in the pilot experiment. The presented approach for lipid vesicle separation encourages the development of new techniques for the direct mass-spectrometric analysis of biomarkers in MS-separated EVs in a vacuum.
Collapse
Affiliation(s)
- Anatolii Spesyvyi
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
| | - Marek Cebecauer
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
| | - Ján Žabka
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
| | - Agnieszka Olżyńska
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
| | - Michaela Malečková
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
| | - Zuzana Johanovská
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
- Faculty of Mathematics and Physics, Charles University, Prague 12116, Czechia
| | - Miroslav Polášek
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
| | - Ales Charvat
- Institute of Chemical Technology, Leipzig University, Leipzig 04103, Germany
- Leibniz Institute of Surface Engineering, Leipzig 04318, Germany
| | - Bernd Abel
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Prague 18223, Czechia
- Institute of Chemical Technology, Leipzig University, Leipzig 04103, Germany
- Leibniz Institute of Surface Engineering, Leipzig 04318, Germany
| |
Collapse
|
29
|
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
|
30
|
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
|
31
|
Hsu CW, Fang YC, Li JF, Cheng CA. Decoding Complex Biological Milieus: SHINER's Approach to Profiling and Functioning of Extracellular Vesicle Subpopulations. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503638. [PMID: 40255212 DOI: 10.1002/smll.202503638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Extracellular vesicles (EVs) are celebrated for their pivotal roles in cellular communication and their potential in disease diagnosis and therapeutic applications. However, their inherent heterogeneity acts as a double-edged sword, complicating the isolation of specific EV subpopulations. Conventional EV isolation methods often fall short, relying on biophysical properties, while affinity-based techniques may compromise EV integrity and utility with harsh recovery conditions. To address these limitations, the SHINER (subpopulation homogeneous isolation and nondestructive EV release) workflow is introduced, which redefines how EVs are isolated and recoverd, featuring the innovative SWITCHER (switchable extracellular vesicle releaser) tool. The SHINER workflow facilitates the precise purification and gentle recovery of target EV subpopulations from complex biological mixtures, preserving their structural integrity and biological functionality. Importantly, SHINER demonstrates exceptional adaptability to multiple markers and clinical applications. It not only enhances the ability to trace EV origins for accurate disease diagnosis but also advances fundamental EV research and provides standardized EV materials for therapeutic innovations. By improving the understanding of EVs and enabling the development of personalized diagnostics and treatments, SHINER propels EV-based science into new frontiers of advanced medicine, offering transformative potential for healthcare.
Collapse
Affiliation(s)
- Chen-Wei Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Yao-Ching Fang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Jhih-Fong Li
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Chi-An Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
Barhoum M, Brassart-Pasco S, Dupont-Deshorgue A, Thierry A, Kanagaratnam L, Brassart B, Ramaholimihaso F, Botsen D, Carlier C, Brugel M, Perrier M, Ramont L, Bouché O. Circulating Exosomal Proteins as New Diagnostic Biomarkers for Colorectal Cancer (EXOSCOL01): A Pilot Case-Controlled Study Focusing on MMP14 Potential. J Clin Lab Anal 2025:e70016. [PMID: 40244893 DOI: 10.1002/jcla.70016] [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: 09/05/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. The French CRC screening campaign is based on fecal immunochemical tests (FIT), confirmed by colonoscopy, an invasive procedure with a poor participation rate. This study aimed to compare the expression of circulating exosomal proteins (MMP14, β1-Integrin subunit, β3-Integrin subunit, and α1(I) Collagen chain) in patients with CRC or adenomas. METHODS A total of 71 patients were recruited, including 24 controls (normal colonoscopy), 11 patients with adenoma, and 36 with CRC. Plasmatic exosomal protein expression was measured by western blot analysis and reported to either protein or exosome content. RESULTS The three groups were comparable regarding clinical characteristics. A significant difference was observed for MMP14 relative expression (p = 0.0007), MMP14 expression reported to exosomal protein content (p = 0.0003), and MMP14 expression reported to exosome content (p = 0.0005). These three parameters were significantly higher in patients with adenoma vs. control patients (p = 0.0013, p = 0.0004, and p = 0.0003, respectively). Only MMP14 relative intensity was significantly higher in the CRC group vs. the control group (p = 0.0018). CONCLUSIONS Exosomal MMP14 is a promising early diagnostic biomarker for CRC and adenoma. These preliminary results warrant confirmation in larger studies using quantitative measurements such as ELISA or flow cytometry.
Collapse
Affiliation(s)
- Mickaël Barhoum
- Department of Gastroenterology and Digestive Oncology, Université de Reims Champagne-Ardenne, Reims, France
| | - Sylvie Brassart-Pasco
- Université de Reims Champagne-Ardenne, CNRS, MEDyC Research Unit, UMR CNRS/URCA 7369, Reims, France
| | - Aurélie Dupont-Deshorgue
- Université de Reims Champagne-Ardenne, CNRS, MEDyC Research Unit, UMR CNRS/URCA 7369, Reims, France
| | - Aurore Thierry
- CHU Reims, Unité d'Aide Méthodologique, Université de Reims Champagne-Ardenne, VieFra UR 3797, Reims, France
| | - Lukshe Kanagaratnam
- CHU Reims, Unité d'Aide Méthodologique, Université de Reims Champagne-Ardenne, VieFra UR 3797, Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne-Ardenne, CNRS, MEDyC Research Unit, UMR CNRS/URCA 7369, Reims, France
| | - Fidy Ramaholimihaso
- Department of Gastroenterology and Digestive Oncology, Université de Reims Champagne-Ardenne, Reims, France
| | - Damien Botsen
- Department of Medical Oncology, Godinot Cancer Institute, Reims, France
| | - Claire Carlier
- Department of Medical Oncology, Godinot Cancer Institute, Reims, France
| | - Mathias Brugel
- Gastroenterology and Digestive Oncology Department, Centre Hospitalier Côte Basque, Bayonne, France
| | - Marine Perrier
- Department of Gastroenterology and Digestive Oncology, Université de Reims Champagne-Ardenne, Reims, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, CNRS, MEDyC Research Unit, UMR CNRS/URCA 7369, Reims, France
- Biochemistry, Pharmacology and Toxicology Department, CHU Reims, Reims, France
| | - Olivier Bouché
- Department of Gastroenterology and Digestive Oncology, Université de Reims Champagne-Ardenne, Reims, France
- Université de Reims Champagne-Ardenne, CNRS, MEDyC Research Unit, UMR CNRS/URCA 7369, Reims, France
| |
Collapse
|
36
|
Tetta C, Camussi G. Challenges and Opportunities in Translating Extracellular Vesicles into Clinical Applications. Stem Cells Dev 2025. [PMID: 40242855 DOI: 10.1089/scd.2025.0051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Affiliation(s)
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
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
|
43
|
Xu S, Zhang Z, Zhou X, Liao Y, Peng Z, Meng Z, Nüssler AK, Ma L, Xia H, Liu L, Yang W. Gouqi-derived Nanovesicles (GqDNVs) promoted MC3T3-E1 cells proliferation and improve fracture healing. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156755. [PMID: 40252435 DOI: 10.1016/j.phymed.2025.156755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Lycium barbarum L., also known as Gouqi, a traditional Chinese herbal medicine, is widely utilized in health care products and clinical therapies. Its muscle and bone strengthening efficacy has been recorded in medical classics for a long time. In addition, plant exosome-like nanovesicles (PELNVs) have attracted more and more attention owing to their biological traits. Therefore, we intended to explore the functions, regulatory role, and underlying mechanism of Gouqi-derived Nanovesicles (GqDNVs) on fracture healing. METHODS In this study, we employed the sucrose density gradient differential ultracentrifugation to isolate GqDNVs. The effects of GqDNVs on the proliferation and differentiation of MC3T3-E1 cells were evaluated using the CCK-8 assay, ALP activity measurement, and cell scratch assay. Additionally, leveraging a fracture mouse model, we utilized Micro-CT, immunological staining, and histologic analyses to comprehensively assess the impact of GqDNVs on fracture healing in mice. RESULTS GqDNVs stimulated cell viability, increased ALP activity, and promoted cellular osteogenic protein expression (OPN, ALP, and RUNX2). Subsequently, in the mouse fracture model, trabecular thickness, and bone marrow density were increased in the GqDNVs treatment group after 28 days of injection. Meanwhile, the expressions of OPN and BGP were significantly elevated after both 14 and 28 days. Additionally, the expressions of p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR, p-4EBP1/4EBP1 and p-p70S6K/ p70S6K were also increased after14 days of treatment. CONCLUSIONS GqDNVs effectively promoted the proliferation and differentiation of MC3T3-E1 cells. Furthermore, GqDNVs could improve fracture healing, which is associated with PI3K/Akt/mTOR/p70S6K/4EBP1 signaling pathway.
Collapse
Affiliation(s)
- Shiyin Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zixuan Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, Tübingen 72076, Germany
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China.
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
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
|
47
|
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
|
48
|
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
|
49
|
Praveena G, Jayachandran A, Manda Venkata S, Asthana A. From bench to bedside: The evolution of extracellular vesicle diagnostics through microfluidic and paper-based technologies. Colloids Surf B Biointerfaces 2025; 252:114675. [PMID: 40222114 DOI: 10.1016/j.colsurfb.2025.114675] [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: 12/19/2024] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 04/15/2025]
Abstract
"Extracellular vesicles (EVs) have emerged as key mediators of intercellular communication and valuable biomarkers for various diseases. However, traditional EV isolation and detection methods often struggle with efficiency, scalability, and purity, limiting their clinical utility. Recent advances in microfluidic and paper-based technologies offer innovative solutions that enhance EV isolation and detection by reducing sample volume, accelerating processing times, and integrating multiple analytical steps into compact platforms. These technologies hold significant promise for advancing point-of-care diagnostics, enabling rapid disease detection, personalized treatment monitoring, and better patient outcomes. For example, early detection of cancer biomarkers through EVs can facilitate timely intervention, potentially improving survival rates, while rapid infectious disease diagnostics can support prompt treatment. Despite their potential, challenges such as standardization, scalability, and regulatory hurdles remain. This review discusses recent advancements in microfluidic and paper-based EV diagnostic technologies, their comparative advantages over traditional methods, and their transformative potential in clinical practice."
Collapse
Affiliation(s)
- Ganji Praveena
- Urvogelbio Private Limited, AHERF, Film Nagar, Hyderabad, Telangana 500033, India
| | - Arjun Jayachandran
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad (NIPER - Hyderabad), Balanagar, Hyderabad, Telangana 500037, India
| | - Sasidhar Manda Venkata
- Urvogelbio Private Limited, AHERF, Film Nagar, Hyderabad, Telangana 500033, India; Apollo Hospitals Educational and Research Foundation (AHERF), Cell and Molecular Biology Research Lab, Hyderabad, India.
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad (NIPER - Hyderabad), Balanagar, Hyderabad, Telangana 500037, India.
| |
Collapse
|
50
|
Ngo CHL, Tukova A, Zhang W, Tsao SCH, Wang Y. Sensitive detection of small extracellular vesicles using a gold nanostar-based SERS assay. Analyst 2025. [PMID: 40202797 DOI: 10.1039/d5an00110b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Small extracellular vesicles (sEVs) are lipid bilayer-bound vesicles that carry critical biomarkers for disease detection. However, the inherent heterogeneity and complexity of sEV molecular characteristics pose significant challenges for accurate and comprehensive molecular profiling. Traditional analytical methods, including immunoblotting, enzyme-linked immunosorbent assay (ELISA), and flow cytometry, exhibit several limitations, such as being time-consuming, requiring large sample volumes, and demonstrating relatively low sensitivity. Therefore, there is an urgent need to develop a highly sensitive and specific assay for the reliable detection of sEVs. Surface-enhanced Raman scattering (SERS) assays have emerged as a promising approach for sEV detection, offering advantages including high sensitivity and specificity. In the proposed SERS assay, SERS nanotags - comprising nanoparticles coated with Raman-active molecules and conjugated with antibodies - are employed to label surface-bound molecules on sEVs. This approach facilitates the generation of a high-intensity signal from molecules present in low abundance. Recently, anisotropic nanoparticles, such as star-shaped nanostructures, have garnered interest due to their ability to significantly amplify generated SERS signals for ultra-sensitive biomarker detection. In this study, we explore the application of gold nanostars (AuNSs) as SERS nanotags for the detection of sEVs. In principle, AuNS-based SERS nanotags were used to label the EpCAM protein, which can be found on the surface of cancer cell derived sEVs, and then sEV labelled SERS nanotags were captured by CD9-conjugated magnetic beads to form an immunocomplex, which exhibits a SERS signal. Our results demonstrate that the proposed SERS assay utilizing AuNSs provides high specificity and sensitivity, with a detection limit as low as 2.47 × 103 particles per μL. Furthermore, the assay was tested with spiked plasma samples (cancer cell-derived sEVs spiked into healthy plasma), showing that its specificity remains unaffected by the presence of plasma. These findings suggest that the SERS assay incorporating AuNSs holds significant promise as an effective and reliable detection method for potential clinical applications.
Collapse
Affiliation(s)
| | - Anastasiia Tukova
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Wei Zhang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| | - Simon Chang-Hao Tsao
- School of Natural Sciences, Macquarie University, Sydney, Australia.
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia
| | - Yuling Wang
- School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|