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] [Download PDF] [Figures] [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
|
Clerici M, Ciardulli MC, Lamparelli EP, Lovecchio J, Giordano E, Dale TP, Forsyth NR, Maffulli N, Della Porta G. Human tendon stem/progenitor cell-derived extracellular vesicle production promoted by dynamic culture. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2025; 53:1-16. [PMID: 40063517 DOI: 10.1080/21691401.2025.2475099] [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: 03/05/2024] [Revised: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025]
Abstract
Tendon injuries significantly impact quality of life, prompting the exploration of innovative solutions beyond conventional surgery. Extracellular Vesicles (EVs) have emerged as a promising strategy to enhance tendon regeneration. In this study, human Tendon Stem/Progenitor Cells (TSPCs) were isolated from surgical biopsies and cultured in a Growth-Differentiation Factor-5-supplemented medium to promote tenogenic differentiation under static and dynamic conditions using a custom-made perfusion bioreactor. Once at 80% confluence, cells were transitioned to a serum-free medium for conditioned media collection. Ultracentrifugation revealed the presence of vesicles with a 106 particles/mL concentration and sub-200nm diameter size. Dynamic culture yielded a 3-fold increase in EV protein content compared to static culture, as confirmed by Western-blot analysis. Differences in surface marker expression were also shown by flow cytometric analysis. Data suggest that we efficiently developed a protocol for extracting EVs from human TSPCs, particularly under dynamic conditions. This approach enhances EV protein content, offering potential therapeutic benefits for tendon regeneration. However, further research is needed to fully understand the role of EVs in tendon regeneration.
Collapse
Affiliation(s)
- Marta Clerici
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Salerno, Italy
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Salerno, Italy
| | - Erwin Pavel Lamparelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Salerno, Italy
| | - Joseph Lovecchio
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
- Institute of Biomedical and Neural Engineering, Reykjavik University, Reykjavík, Iceland
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Cesena, Italy
| | - Tina P Dale
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK
| | - Nicholas R Forsyth
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK
- Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen, UK
| | - Nicola Maffulli
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK
- Department of Trauma and Orthopaedics, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Salerno, Italy
- Interdepartmental Centre BIONAM, University of Salerno, Fisciano, Salerno, Italy
| |
Collapse
|
3
|
Cui Y, Zhu X, Qian L, Zhang S. Extracellular transfer of HuR promotes acquired cisplatin resistance in esophageal cancer cells. Cancer Biol Ther 2025; 26:2495999. [PMID: 40269355 PMCID: PMC12026070 DOI: 10.1080/15384047.2025.2495999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 03/10/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Cisplatin (DDP) resistance is a key factor hindering esophageal cancer (ESCA) treatment. Exosomes have been reported to confer resistance to DDP in various tumor cells. However, the effects of ESCA cell-derived exosomes and exosomal human antigen R (HuR) on DDP resistance in cancer cells have not been elucidated. In this study, isolated exosomes were identified by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. CCK-8 and flow cytometry were employed to assess the functional role of exosomes in ESCA DDP-resistant cells and their parental cells. Bioinformatics analysis was performed to identify molecules that were positively associated with HuR and validated using dual-luciferase reporter analysis and RNA immunoprecipitation assays. We found that exosomes from ESCA cells enhance the resistance of drug-resistant cells to DDP. Importantly, HuR protein, but not mRNA, was directly transferred into DDP-resistant cells via exosomes, thereby increasing the level of HuR protein. Mechanistically, HuR positively correlated with Lamin B2 (LMNB2) in ESCA cells, and ESCA DDP-resistant cells transfected with siRNA targeting LMNB2 exhibited reduced cell viability and elevated apoptosis rates. Moreover, the role of ESCA cell-derived exosomes in the transmission of DDP resistance in vivo was validated using a nude mouse model. Collectively, our results revealed that exosomes exposed to ESCA cells induced greater drug resistance in DDP-resistant ESCA cells via HuR delivery. Targeting HuR or its positively related target LMNB2 may present new therapeutic opportunities for treating patients with DDP-resistant ESCA.
Collapse
Affiliation(s)
- Yayun Cui
- Department of Oncology, Shandong University Cancer Center, Jinan, Shandong, China
| | - Xiaofeng Zhu
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (Anhui Provincial Cancer Hospital), Hefei, Anhui, China
| | - Liting Qian
- Department of Cancer Radiotherapy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (Anhui Provincial Cancer Hospital), Hefei, Anhui, China
| | - Shu Zhang
- Department of Gastroenterology, Shandong University Cancer Center, Jinan, Shandong, China
| |
Collapse
|
4
|
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
|
5
|
Yun HW, Kim M, Shin DI, Kwon HJ, Park IS, Park DY, Min BH. Matrix-bound nanovesicles recapitulate tissue-specific angiogenic properties of parent extracellular matrix with distinct miRNA profiles. BIOMATERIALS ADVANCES 2025; 175:214338. [PMID: 40378642 DOI: 10.1016/j.bioadv.2025.214338] [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: 02/11/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Decellularized extracellular matrix (dECM) exhibits tissue-specific pro- or anti-angiogenic effects. Previous studies have demonstrated that matrix-bound nanovesicles (MBVs) act as key bioactive components of dECM, replicating various biological functions such as anti-inflammatory and immunomodulatory effects. Building on this evidence, this study hypothesized that MBVs derived from cartilage and small intestinal submucosa (SIS) modulate angiogenesis through the selective packaging of miRNAs. Cartilage-derived MBVs (cMBVs) and SIS-derived MBVs (sMBVs) were isolated, characterized, and analyzed for their miRNA profiles using RNA sequencing and RT-qPCR validation. The interactions between MBVs and human umbilical vein endothelial cells (HUVECs) were assessed by examining proliferation, adhesion, migration, and tube formation in comparison to the parent ECM. Angiogenic modulation was further evaluated using a mouse Matrigel plug assay and a rabbit corneal neovascularization (NV) model. Our results demonstrated that anti-angiogenic miRNAs (e.g., miR-140-3p, miR-455-5p, and miR-148a-5p) were predominant in cMBVs, suppressing endothelial cell activity and angiogenesis, while pro-angiogenic miRNAs (e.g., miR-143-3p, miR-181a, and miR-21-5p) were prevalent in sMBVs, enhancing vessel formation. In vivo, cMBVs significantly inhibited vascular invasion and neovessel formation, whereas sMBVs promoted angiogenesis in both models. These findings confirm that MBVs reflect the tissue-specific angiogenic regulatory functions of their parent ECM, and highlight their potential as therapeutic tools for targeted modulation of angiogenesis in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Hee-Woong Yun
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Mijin Kim
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Dong Il Shin
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Hyeon Jae Kwon
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - In-Su Park
- Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Do Young Park
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea
| | - Byoung-Hyun Min
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Republic of Korea; Cell Therapy Center, Ajou Medical Center, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
6
|
Cho H, Ju H, Ahn Y, Jang J, Cho J, Park E, Kang SM, Lee J, Seo D, Baek MC, Yea K. Engineered extracellular vesicles with surface FGF21 and enclosed miR-223 for treating metabolic dysfunction-associated steatohepatitis. Biomaterials 2025; 321:123321. [PMID: 40209593 DOI: 10.1016/j.biomaterials.2025.123321] [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: 10/28/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disorder with a complex pathogenesis that requires combination therapies rather than monotherapies. Extracellular vesicles (EVs) exhibit inherently efficient delivery to the liver and can be engineered to carry various therapeutic substances, making them promising agents. In this study, EVs were engineered to display fibroblast growth factor 21 (FGF21) on their surface and encapsulate miR-223 (223/F-EVs), aiming to improve steatosis and alleviate inflammation and fibrosis, respectively. Introducing the 223/F-EVs into human liver cell lines significantly reduced both basal and induced levels of lipid storage, inflammation, and fibrosis markers. Furthermore, using an FGF21-blocking antibody or miR-223 inhibitor effectively diminished the efficacy of the 223/F-EVs, confirming the essential roles of FGF21 and miR-223 in these processes. In a Choline-Deficient, l-Amino acid-defined, High-Fat Diet (CDAHFD)-fed mouse model, intravenously administered 223/F-EVs demonstrated liver-preferential delivery and a marked reduction in the MASH phenotype without compromising bone density, unlike conventional FGF21 treatment. Collectively, 223/F-EVs convey FGF21 and miR-223 exclusively to the liver, offering strategic advantages by mitigating MASH progression via multiple pathways. This study lays a solid foundation for further investigation of engineered EVs as a transformative therapeutic approach for treating MASH.
Collapse
Affiliation(s)
- Hanchae Cho
- Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Hyunji Ju
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Yongdeok Ahn
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhyeong Cho
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Eunju Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Sung-Min Kang
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea.
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea; New Biology Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 43024, Republic of Korea.
| |
Collapse
|
7
|
Manzaneque-López MC, González-Arce A, Pérez-Bermúdez P, Soler C, Marcilla A, Sánchez-López CM. Pasteurization and lyophilization affect membrane proteins of pomegranate-derived nanovesicles reducing their functional properties and cellular uptake. Food Chem 2025; 483:144303. [PMID: 40250286 DOI: 10.1016/j.foodchem.2025.144303] [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: 01/03/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
Plant-derived nanovesicles (PDNVs) are promising therapeutic agents, valued for their bioactive compound content and as drug delivery systems. However, the effects of industrial food processing techniques on PDNVs remain underexplored. This study evaluated the impact of lyophilization and pasteurization on the properties and uptake efficiency of pomegranate-derived nanovesicles (PgNVs). PgNVs were isolated from pomegranate juice using tangential flow filtration and size exclusion chromatography. Treated PgNVs were analyzed via transmission electron microscopy, nanoparticle tracking analysis, and LC-MS/MS, assessing their functional properties and cellular uptake. High-purity PgNVs were obtained, although both treatments reduced their yield. Pre-treated PgNVs displayed diminished anti-inflammatory and wound-healing capacities, though antioxidant activity remained unaffected. Treated PgNVs also exhibited lower internalization by human macrophages compared to untreated PgNVs. Proteomic analyses revealed that the damage of membrane proteins, such as Tetraspanin-8, might be responsible for these effects. Our findings emphasize the need of optimizing processing to preserve PDNV therapeutic potential.
Collapse
Affiliation(s)
- Mari Cruz Manzaneque-López
- Dept. Medicina Preventiva i Salut Pública, F. Farmàcia i Ciències de l'Alimentació, Universitat de València, Burjassot, València, Spain; Instituto de Ciencia de los Materiales, Parque Científico, Universitat de València, Burjassot, València, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics UV-IIS La Fe, Valencia 46026, Spain
| | - Aránzazu González-Arce
- Parasites & Health Research Group, Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Valencia 46100, Spain
| | - Pedro Pérez-Bermúdez
- Dept. Biologia Vegetal, F. Ciències Biológiques, Universitat de València, Burjassot, Valencia, Spain
| | - Carla Soler
- Dept. Medicina Preventiva i Salut Pública, F. Farmàcia i Ciències de l'Alimentació, Universitat de València, Burjassot, València, Spain; Instituto de Ciencia de los Materiales, Parque Científico, Universitat de València, Burjassot, València, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics UV-IIS La Fe, Valencia 46026, Spain
| | - Antonio Marcilla
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics UV-IIS La Fe, Valencia 46026, Spain; Parasites & Health Research Group, Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Valencia 46100, Spain.
| | - Christian M Sánchez-López
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics UV-IIS La Fe, Valencia 46026, Spain; Parasites & Health Research Group, Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Burjassot, Valencia 46100, Spain.
| |
Collapse
|
8
|
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] [MESH Headings] [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
|
9
|
Malhotra P, Fyfe J, Emmanouilidi A, Casari I, Mellett NA, Huynh K, Pajic M, Greening DW, Meikle PJ, Falasca M. Oncogenic small extracellular vesicles enriched in sphingosine-1-phosphate play a crucial role in pancreatic cancer progression. Cell Signal 2025; 132:111775. [PMID: 40158707 DOI: 10.1016/j.cellsig.2025.111775] [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: 01/13/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Small extracellular vesicles (sEVs) from tumour cells mediate intercellular communication and signalling to regulate the progression of pancreatic ductal adenocarcinoma (PDAC). While we and others have shown that PDAC-derived sEVs comprise oncogenic protein and nucleic acid cargo, understanding the lipid landscape of these sEVs remains unknown. Lipids influence both the composition of sEVs and their roles in lipid metabolism and signalling pathways within the tumour microenvironment and tumorigenesis. We hypothesised that specific lipids in oncogenic sEVs might provide insights into PDAC. Comprehensive mass spectrometry-based lipidomic analysis was performed using liquid chromatography-electrospray ionisation-tandem mass spectrometry on sEVs isolated from PDAC and non-malignant pancreatic cell lines, patient-derived xenograft cell lines and plasma from the PDAC transgenic mouse model KPC (KRASWT/G12D/ TP53WT/R172H/Pdx1-Cre+/+). The sEV lipidomic analyses identified over 700 lipid species from 25 lipid classes and subclasses. Our results showed that, compared to non-malignant cells, PDAC-derived sEVs were enriched in specific lysophospholipids, particularly sphingosine-1-phosphate (S1P), a lipid known for its pivotal role in cancer pathogenesis. S1P enrichment was validated in plasma-derived sEVs from KPC mice compared to WT. To explore the functional implications of S1P enrichment, we conducted assays demonstrating that S1P in sEVs facilitated tubule formation in human microvascular endothelial cells and promoted cancer-associated fibroblast cell migration. We show that PDAC-derived sEVs are differentially enriched in specific lipids associated with cancer phenotype. Our findings highlight that PDAC-derived sEVs are enriched in specific lipids, particularly S1P, which plays a crucial role in promoting cancer progression.
Collapse
Affiliation(s)
- Pratibha Malhotra
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Jordan Fyfe
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Aikaterini Emmanouilidi
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Ilaria Casari
- Curtin Medical Research Institute, Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Natalie A Mellett
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Kevin Huynh
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Marina Pajic
- Translational Oncology Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2010, Australia
| | - David W Greening
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC 3086, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marco Falasca
- University of Parma, Department of Medicine and Surgery, Via Volturno 39, 43125 Parma, Italy.
| |
Collapse
|
10
|
Zhang J, Weng S, Fan Z, Hu D, Le J, Sheng K. Migrasomes: Critical players in intercellular nanovesicle communication. Cell Signal 2025; 132:111796. [PMID: 40209968 DOI: 10.1016/j.cellsig.2025.111796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/24/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Migrasomes are vesicular structures that form on elongated tethers originating from the tips or junctions of cellular tails during migration. These organelles, named for their vesicle rich lumen and release during cell movement, have gained attention for their role in intercellular communication and signal transduction. Migrasome formation is closely associated with the dynamic and active movement of cells, as well as with the intrinsic properties of cells and the extracellular microenvironment under various pathophysiological conditions. This review provides a comprehensive overview of migrasome dynamics, examining the mechanisms and distinct features of nanoscale vesicle-mediated intercellular signaling. It also highlights the influence of microscopic secretory factors on migrasome generation and formation. By comparing migrasomes with other active extracellular vesicles, this review highlights the advantages of migrasomes and addresses future challenges.
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of ECG, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, China
| | - Shoutao Weng
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zaiwei Fan
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi 330006, China
| | - Dongyang Hu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiadi Le
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kongsheng Sheng
- Department of Pharmacy, Taizhou Municipal Hospital, Taizhou 318000, China.
| |
Collapse
|
11
|
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
|
12
|
Yu L, Shi H, Gao T, Xu W, Qian H, Jiang J, Yang X, Zhang X. Exomeres and supermeres: Current advances and perspectives. Bioact Mater 2025; 50:322-343. [PMID: 40276541 PMCID: PMC12020890 DOI: 10.1016/j.bioactmat.2025.04.012] [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: 01/26/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Recent studies have revealed a great diversity and complexity in extracellular vesicles and particles (EVPs). The developments in techniques and the growing awareness of the particle heterogeneity have spurred active research on new particle subsets. Latest discoveries highlighted unique features and roles of non-vesicular extracellular nanoparticles (NVEPs) as promising biomarkers and targets for diseases. These nanoparticles are distinct from extracellular vesicles (EVs) in terms of their smaller particle sizes and lack of a bilayer membrane structure and they are enriched with diverse bioactive molecules particularly proteins and RNAs, which are widely reported to be delivered and packaged in exosomes. This review is focused on the two recently identified membraneless NVEPs, exomeres and supermeres, to provide an overview of their biogenesis and contents, particularly those bioactive substances linked to their bio-properties. This review also explains the concepts and characteristics of these nanoparticles, to compare them with other EVPs, especially EVs, as well as to discuss their isolation and identification methods, research interests, potential clinical applications and open questions.
Collapse
Affiliation(s)
- Li Yu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Tingxin Gao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
13
|
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
|
14
|
Cao LM, Qiu YZ, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Wu Q, Bu LL. Extracellular Vesicles: Hermes between cancers and lymph nodes. Cancer Lett 2025; 623:217735. [PMID: 40268131 DOI: 10.1016/j.canlet.2025.217735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Cancer is one of the main causes of death and a major obstacle to increasing life expectancy in all countries of the world. Lymph node metastasis (LNM) of in cancer patients indicates poor prognosis and it is an important indication to determine the therapeutic regime. Therefore, more attention should be given to the molecular mechanics of tumor lymphangiogenesis and LNM. Extracellular vesicles (EVs) are nanoscale cargo-bearing membrane vesicles that can serve as key mediators for the intercellular communication. Like Hermes, the messenger of the Greek gods, EVs can be secreted by tumor cells to regulate the LNM process. Many evidence has proved the clinical correlation between EVs and LNM in various cancer types. EVs plays an active role in the process of metastasis by expressing its connotative molecules, including proteins, nucleic acids, and metabolites. However, the clear role of EVs in the process of cancer LNM has not been thoroughly studied yet. In this review, we will summarize the clinical and mechanical findings of EVs regulating role on cancer LNM, and discuss the advanced modification of the research proposal. We propose the "PUMP" principle of EVs in LNM, including Preparation, Unleash, Migration, and Planting.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yu-Zhong Qiu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Han-Yue Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
15
|
Chen C, Wu Y, Wu J, Sun R, Li Y, Yao Y, Li D. Identification of a panel of lncRNAs derived from urinary extracellular vesicles as non-invasive diagnostic biomarkers for bladder cancer. Clin Chim Acta 2025; 575:120376. [PMID: 40393569 DOI: 10.1016/j.cca.2025.120376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/02/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025]
Abstract
Bladder cancer (BLCA) is a common malignant tumor of the urinary system and is histopathologically divided into high-grade and low-grade BLCA. Accurate diagnosis of BLCA and high-grade BLCA are critical for clinical treatment and early intervention. High-throughput RNA-seq was performed to explore dysregulated long non-coding RNAs (lncRNAs) in urinary extracellular vesicles (uEVs) from BLCA patients, and their expression levelswereexamined inalarge cohort of uEVs samples using qRT-PCR. Weexaminedthe expressionlevels and subcellular localization of the lncRNAs in BLCA tissues andcelllines. We analyzed the correlation between the expression levels of lncRNAs in uEVs and clinical parameters and assessed their clinical value as diagnostic biomarkers for BLCA and high-grade BLCA using receiver operating characteristic (ROC) curve. Through high-throughput RNA-seq, we identified several dysregulated lncRNAs (MALAT1, SCARNA10, LINC00963 and LINC01578) in uEVs from BLCA patients. The lncRNAs were significantly upregulated in uEVs of BLCA patients, however with varying expression levels in tissues and cell lines. The lncRNAsarepredominantlylocalizedinthe nucleus of BLCA cell lines. Elevated expression levels of the lncRNAs were associated with adverse factors, including higher tumor grade and larger tumor diameter. ROCcurve analysis showed thatthe combination of four lncRNAs in uEVs and the existing marker nuclear matrix protein 22 provided substantial diagnostic value for BLCA and high-grade BLCA, with area under curve values of 0.900 and 0.917, respectively. The lncRNA panel derived from uEVs may serve as a promising non-invasive biomarker for diagnosing BLCA and high-grade BLCA.
Collapse
Affiliation(s)
- Chen Chen
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ying Wu
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Junlu Wu
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Ruixin Sun
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yaran Li
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yiwen Yao
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Dong Li
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| |
Collapse
|
16
|
Kim MG, Ryu SM, Shin Y. Recent advances in bioreceptor-based sensing for extracellular vesicle analysis. Biosens Bioelectron 2025; 280:117432. [PMID: 40187151 DOI: 10.1016/j.bios.2025.117432] [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: 11/18/2024] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Extracellular vesicles (EVs) are nanoscale, membrane-bound structures secreted by various cell types into biofluids. They show great potential as biomarkers for disease diagnostics, owing to their ability to carry molecular cargo that reflects their cellular origin. However, the inherent heterogeneity of EVs in terms of size, composition, and source presents significant challenges for reliable detection and analysis. Recent advances in bioreceptor-based biosensor technologies provide promising solutions by offering high sensitivity and specificity in EV detection and characterization. These technologies address the limitations of conventional methods, such as ultracentrifugation and bulk analysis. Biosensors utilizing antibodies, aptamers, peptides, lectins, and molecularly imprinted polymers enable precise detection of EV subpopulations by targeting specific EV surface markers, including proteins, lipids, and glycans. Additionally, these biosensors support multiplexed and real-time analysis while preserving the structural integrity of EVs. This review highlights the transformative potential of combining modern biosensing tools with bioreceptor technologies to advance EV research and diagnostics, paving the way for innovations in disease diagnostics and therapeutic monitoring.
Collapse
Affiliation(s)
- Myoung Gyu Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Soo Min Ryu
- Life Science and Biotechnology, Underwood International College, Yonsei University, 50 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yong Shin
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Life Science and Biotechnology, Underwood International College, Yonsei University, 50 Yonsei Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
17
|
Yan R, Ke X, Cheng Y, Liu X, Wang Z, Meltzer SJ. Delivery of cancer cell-derived extracellular vesicles modulates the morphology and gene expression of Barrett esophagus and duodenal organoids (Article). Biochem Biophys Res Commun 2025; 769:151976. [PMID: 40349460 DOI: 10.1016/j.bbrc.2025.151976] [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: 01/24/2025] [Revised: 04/25/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Extracellular vesicles (EVs) facilitate intercellular communication, especially in the signaling mechanisms employed by tumor cells to influence both local and distant cells and tissues. This study investigated the impact of cancer cell-derived EVs (CEVs) on patient-derived organoids. Co-culture experiments examined the morphology, growth, proliferation, and cancer-related gene/miRNA expression in Barrett's esophagus (BE) and duodenal organoids. Our results indicate that CEVs promoted organoid proliferation, increased cancer-related mRNA/miRNA expression, and induced phenotypic changes. Artificial modulation of specific oncomiRNAs in CEVs-such as miR-21 and miR-210, influenced CEV-mediated effects on co-cultured organoid growth. These findings align with EV-mediated transformations in benign organoid models, providing a valuable tool to study EV-associated miRNAs/proteins in gastrointestinal preneoplastic/neoplastic conditions and potentially other organs. This lays a foundation for future research on cancer cell-microenvironment interactions and EV roles in tumorigenesis/metastasis.
Collapse
Affiliation(s)
- Rong Yan
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China; Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Xiquan Ke
- First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| | - Yulan Cheng
- Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Xi Liu
- First Affiliated Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi, China.
| | - Zhixiong Wang
- First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Stephen J Meltzer
- Johns Hopkins School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
18
|
Lim Falk V, Mueller-Wirth N, Karathanasis D, Evangelopoulos ME, Maleska Maceski A, Zadic A, Kuhle J, Schlup C, Marti S, Guse K, Chan A, Pernet V. Extracellular Vesicle Marker Changes Associated With Disease Activity in Relapsing-Remitting Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200404. [PMID: 40300121 PMCID: PMC12056760 DOI: 10.1212/nxi.0000000000200404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/19/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND AND OBJECTIVES Multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), and myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) are autoimmune disorders of the CNS causing severe neurologic impairment. Evidence suggests that extracellular vesicles (EVs) may play a disease-specific role in the orchestration of the immune cell response of MS, NMOSD, and MOGAD. In addition, EVs are considered as a potential source of biomarkers that may allow us to establish molecular signatures for these diseases and perhaps as well to follow treatment effects and disease progression. The aim of this study was to analyze the composition of EVs in patients with relapsing-remitting MS (RRMS) (n = 52), NMOSD (n = 19), and MOGAD (n = 10) and healthy controls ([HCs], n = 15). METHODS The concentrations of neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) were determined in plasma using single-molecule array (SIMOA). The size and concentration of tetraspanin-presenting EVs were evaluated in plasma samples with a single-particle interferometric resonance imaging sensor (SP-IRIS). Tetraspanin-independent analyses were performed by nanoparticle-tracking analysis (NTA) after EV isolation by size exclusion (SmartSEC) and cryo-electron microscopy observations. EV epitopes were analyzed by extended multiplex analysis using flow cytometry. RESULTS The plasma concentration of NfL and GFAP was significantly higher in patients with RRMS than in HCs. For patients with NMOSD, only GFAP increased. The density of EVs assessed by NTA was lower in plasma of patients with RRMS than in HC plasma. In addition, the 3 disease groups presented increased mean EV sizes in comparison with HCs. Tetraspanin-based EV analyses by SP-IRIS allowed us to observe a modest difference in the level of CD81 in RRMS EVs. In patients with RRMS, but not in those with NMOSD and MOGAD, multiplex/flow cytometry analyses revealed changes in the EV levels of CD29, CD31, and CD69 associated with the time elapsed since the last relapse. The negative correlations established between the vesicular levels of CD31, CD40, CD44, CD49c, CD69, and NfL or GFAP z-scores suggest a negative relationship specifically in RRMS. DISCUSSION We speculate that the higher release of EVs containing CD29, CD31, CD40, CD44, CD49c, and CD69 in plasma, at low levels of circulating NfL/GFAP, may be associated with reduced immune cell activity in RRMS. These EV markers may characterize patients with RRMS in disease stabilization.
Collapse
Affiliation(s)
- Victoria Lim Falk
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Switzerland
| | - Nicole Mueller-Wirth
- CSL Behring, CSL Biologics Research Center, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, Sitem-Insel, Bern, Switzerland
| | | | | | - Aleksandra Maleska Maceski
- Departments of Biomedicine and Clinical Research, Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Switzerland
| | - Amar Zadic
- Departments of Biomedicine and Clinical Research, Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Switzerland
| | - Jens Kuhle
- Departments of Biomedicine and Clinical Research, Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), University Hospital and University of Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Switzerland
| | - Cornelia Schlup
- CSL Behring, CSL Biologics Research Center, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, Sitem-Insel, Bern, Switzerland
| | - Stefanie Marti
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
| | - Kirsten Guse
- CSL Behring, CSL Biologics Research Center, Bern, Switzerland
- Swiss Institute for Translational and Entrepreneurial Medicine, Sitem-Insel, Bern, Switzerland
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
| | - Vincent Pernet
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Switzerland
- Regenerative Medicine Unit, University Hospital Center of Quebec, Laval University, Quebec City, Canada; and
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, Canada
| |
Collapse
|
19
|
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] [MESH Headings] [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
|
20
|
Wu Y, Zhao J, Lu X. Profiling the Ocular Landscape of sEVs miRNAs: Mechanisms and Applications. Exp Eye Res 2025; 256:110396. [PMID: 40280535 DOI: 10.1016/j.exer.2025.110396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/23/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
In this review, we comprehensively discuss the application of sEVs miRNAs in ophthalmic diseases by examining their basic characteristics and clinical application potential. Initially, we provide an overview of sEVs, including their definition, source, and functions, while particularly highlighting the importance of miRNAs contained in sEVs and its prospects in the treatment of ocular diseases. Subsequently, the structure and composition of sEVs as well as the biological functions of their encapsulated miRNAs, which expands the current knowledge about their roles in ophthalmic physiology and pathology. In addition, the functions of sEVs miRNAs in the growth of ocular tissues, ocular tissue homeostasis, and common eye diseases such as glaucoma, age-related macular degeneration, and diabetic retinopathy, are discussed. The application potential of sEVs miRNAs as diagnostic biomarkers and drug delivery systems for ophthalmic conditions and therapeutic value in diverse eye diseases are explored. Finally, the current challenges affecting research in this field are outlined to provide a basis that guide future utilization of sEVs miRNAs in ophthalmic disease research and clinical management of diverse ophthalmological conditions.
Collapse
Affiliation(s)
- Yifei Wu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Guangzhou, 510280, GuangDong, China
| | - Jiaqi Zhao
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Guangzhou, 510280, GuangDong, China
| | - Xiaohe Lu
- Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Guangzhou, 510280, GuangDong, China.
| |
Collapse
|
21
|
Oliveira Dias J, Sampaio Fagundes I, Bisio MDC, da Silva Barboza V, Jacinto AA, Altei WF. Extracellular vesicles as the common denominator among the 7 Rs of radiobiology: From the cellular level to clinical practice. Biochim Biophys Acta Rev Cancer 2025; 1880:189315. [PMID: 40216093 DOI: 10.1016/j.bbcan.2025.189315] [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: 11/22/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Extracellular vesicles (EVs) are lipid-bound particles released by tumor cells and widely explored in cancer development, progression, and treatment response, being considered as valuable components to be explored as biomarkers or cellular targets to modulate the effect of therapies. The mechanisms underlying the production and profile of EVs during radiotherapy (RT) require addressing radiobiological aspects to determine cellular responses to specific radiation doses and fractionation. In this review, we explore the role of EVs in the 7 Rs of radiobiology, known as the molecular basis of a biological tissue response to radiation, supporting EVs as a shared player in all the seven processes. We also highlight the relevance of EVs in the context of liquid biopsy and resistance to immunotherapy, aiming to establish the connection and utility of EVs as tools in contemporary and precision radiotherapy.
Collapse
Affiliation(s)
- Júlia Oliveira Dias
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | - Wanessa Fernanda Altei
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Radiation Oncology Department, Barretos Cancer Hospital, Barretos, Brazil.
| |
Collapse
|
22
|
Park KS, Lässer C, Lötvall J. Extracellular vesicles and the lung: from disease pathogenesis to biomarkers and treatments. Physiol Rev 2025; 105:1733-1821. [PMID: 40125970 DOI: 10.1152/physrev.00032.2024] [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: 08/14/2024] [Revised: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nanosized extracellular vesicles (EVs) are released by all cells to convey cell-to-cell communication. EVs, including exosomes and microvesicles, carry an array of bioactive molecules, such as proteins and RNAs, encapsulated by a membrane lipid bilayer. Epithelial cells, endothelial cells, and various immune cells in the lung contribute to the pool of EVs in the lung microenvironment and carry molecules reflecting their cellular origin. EVs can maintain lung health by regulating immune responses, inducing tissue repair, and maintaining lung homeostasis. They can be detected in lung tissues and biofluids such as bronchoalveolar lavage fluid and blood, offering information about disease processes, and can function as disease biomarkers. Here, we discuss the role of EVs in lung homeostasis and pulmonary diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis, and lung injury. The mechanistic involvement of EVs in pathogenesis and their potential as disease biomarkers are discussed. Finally, the pulmonary field benefits from EVs as clinical therapeutics in severe pulmonary inflammatory disease, as EVs from mesenchymal stem cells attenuate severe respiratory inflammation in multiple clinical trials. Further, EVs can be engineered to carry therapeutic molecules for enhanced and broadened therapeutic opportunities, such as the anti-inflammatory molecule CD24. Finally, we discuss the emerging opportunity of using different types of EVs for treating severe respiratory conditions.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
23
|
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
|
24
|
Wei YH, Lin F. Barcodes based on nucleic acid sequences: Applications and challenges (Review). Mol Med Rep 2025; 32:187. [PMID: 40314098 PMCID: PMC12076290 DOI: 10.3892/mmr.2025.13552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/04/2025] [Indexed: 05/03/2025] Open
Abstract
Cells are the fundamental structural and functional units of living organisms and the study of these entities has remained a central focus throughout the history of biological sciences. Traditional cell research techniques, including fluorescent protein tagging and microscopy, have provided preliminary insights into the lineage history and clonal relationships between progenitor and descendant cells. However, these techniques exhibit inherent limitations in tracking the full developmental trajectory of cells and elucidating their heterogeneity, including sensitivity, stability and barcode drift. In developmental biology, nucleic acid barcode technology has introduced an innovative approach to cell lineage tracing. By assigning unique barcodes to individual cells, researchers can accurately identify and trace the origin and differentiation pathways of cells at various developmental stages, thereby illuminating the dynamic processes underlying tissue development and organogenesis. In cancer research, nucleic acid barcoding has played a pivotal role in analyzing the clonal architecture of tumor cells, exploring their heterogeneity and resistance mechanisms and enhancing our understanding of cancer evolution and inter‑clonal interactions. Furthermore, nucleic acid barcodes play a crucial role in stem cell research, enabling the tracking of stem cells from diverse origins and their derived progeny. This has offered novel perspectives on the mechanisms of stem cell self‑renewal and differentiation. The present review presented a comprehensive examination of the principles, applications and challenges associated with nucleic acid barcode technology.
Collapse
Affiliation(s)
- Ying Hong Wei
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Faquan Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
25
|
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
|
26
|
Llach CD, Le GH, Shah H, Marcato LM, Brietzke E, Gill H, Tabassum A, Badulescu S, Rosenblat JD, McIntyre RS, Mansur RB. Peripheral and central inflammation in depression: How large is the gap and can we bridge it with PET neuroimaging and neural-derived extracellular vesicles? J Neuroimmunol 2025; 403:578587. [PMID: 40174479 DOI: 10.1016/j.jneuroim.2025.578587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 02/28/2025] [Accepted: 03/16/2025] [Indexed: 04/04/2025]
Abstract
Major depressive disorder (MDD) presents as a multifaceted syndrome with complex pathophysiology and variable treatment responses, posing significant challenges in clinical management. Neuroinflammation is known to play pivotal mechanism in depression, linking immune responses with central nervous system (CNS) dysfunction. This review explores the interplay between peripheral and central inflammatory processes in MDD, emphasizing discrepancies in biomarker validity and specificity. While peripheral markers like cytokines have historically been investigated as proxies for neuroinflammation, their reliability remains contentious due to inconsistent findings, lack of correlation with neuroinflammatory markers, the influence of confounding variables, and the role of regulatory mechanism within the CNS. Additionally, the human brain shows a pattern of regionalized inflammation. Current methodologies for investigating neuroinflammation in humans in vivo, including neural-derived extracellular vesicles (EVs) and positron emission tomography (PET) neuroimaging using translocator protein, offer promising avenues while facing substantial limitations. We propose that future research in MDD may benefit from combined microglia-derived EV-TSPO PET neuroimaging analyses to leverage the strengths and mitigate the limitations of both individual methods.
Collapse
Affiliation(s)
- Cristian-Daniel Llach
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| | - Gia Han Le
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Hiya Shah
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Liz M Marcato
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Aniqa Tabassum
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sebastian Badulescu
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Guendulain GG, Moyano AL, Mattera V, Monteleone MC. 2024 Argentine Group for Extracellular Vesicles (GAVE) Workshop: promoting science in challenging times. Biol Open 2025; 14:bio062002. [PMID: 40492302 DOI: 10.1242/bio.062002] [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] [Indexed: 06/11/2025] Open
Abstract
The Argentine Group for Extracellular Vesicles (GAVE) was established in 2022 with the objective of bringing together researchers working in Argentina dedicated to extracellular vesicle (EV) studies. Following its successful inaugural meeting in 2023, the II GAVE Workshop was held on 12-13 September 2024, at the University of Buenos Aires, Argentina. This event brought together over 140 participants from diverse disciplines, fostering collaboration and strengthening the national EV research field. Moreover, international speakers and renowned experts in their fields shared valuable insights and experiences with the audience. Despite the challenges posed by the national government's funding cuts, the 2024 GAVE workshop showcased the Argentine scientists' strong commitment to high-quality research and the growth of local science in the field of EVs. Supported by international organizations and local companies, the II GAVE Workshop prioritized inclusivity and provided valuable networking opportunities, particularly for students and early-career researchers. This financial support was fundamental to broadening the impact of the event by promoting the assistance of underrepresented groups. This Meeting Review highlights the outcomes of our workshop and shows the advances of the Argentinian scientific community involved in EV research.
Collapse
Affiliation(s)
- Gonzalo Germán Guendulain
- Centro de Investigación en Medicina Traslacional "Severo R. Amuchástegui" (CIMETSA), Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Naciones Unidas 420, Barrio Parque Vélez Sarsfield, X5016KEJ Córdoba, Argentina
| | - Ana Lis Moyano
- Centro de Investigación en Medicina Traslacional "Severo R. Amuchástegui" (CIMETSA), Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Naciones Unidas 420, Barrio Parque Vélez Sarsfield, X5016KEJ Córdoba, Argentina
| | - Vanesa Mattera
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB), Universidad de Buenos Aires, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires (CABA), C1113AAD Buenos Aires, Argentina
| | - Melisa Carolina Monteleone
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1650 San Martín, Argentina
- Escuela de Bio y Nanotecnologías (EByN), Universidad Nacional de San Martín, 1650 San Martín, Argentina
| |
Collapse
|
28
|
Kothandan VK, Ouyang Y, Sadovsky E, Komsky-Elbaz A, Powell JS, Xia J, Huang TJ, Sadovsky Y. A multi-platform assessment of extracellular vesicles from the plasma and urine of women with preeclampsia. Placenta 2025; 166:96-102. [PMID: 39746834 PMCID: PMC12146095 DOI: 10.1016/j.placenta.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION MicroRNAs (miRNAs), packaged within extracellular vesicles (EVs), have been used to interrogate the pathogenesis of preeclampsia and to identify its biomarkers. We have previously shown that miRNA species were differentially expressed in small plasma EVs from women with preeclampsia vs healthy controls. We sought to assess the use of rapid technologies for isolation of plasma and urine EVs from parturients with preeclampsia and determine differences in the expression of selected EV miRNA species. METHODS We collected blood and urine samples before delivery from parturients with severe preeclampsia vs healthy controls and used size exclusion chromatography (SEC) as an acceptable standard for rapid isolation of plasma EVs. We also isolated urine and plasma EVs using ExoDisc, a rapid nanofiltration technology for EV isolation. All samples were examined using a nanoparticle tracking analyzer, immunoblotting, and RT-qPCR for selected miRNA levels. RESULTS Whereas the concentration of EVs was higher in the urine from preeclampsia compared to controls, we observed the opposite change in plasma EVs, with no difference in EV size. Comparing the two patient groups for miRNA levels in EVs isolated by ExoDisc or SEC, we found that EV miR-93-5p was upregulated in the plasma and urine of parturients with preeclampsia vs healthy controls. Notably, miR-31-5p was upregulated in SEC- or ExoDisc-isolated plasma EVs, and miR-92-3p was upregulated in or ExoDisc-isolated plasma or urine EVs of parturients with preeclampsia. DISCUSSION Technologies for rapid analysis of plasma and urine EVs and their miRNA cargo provide complementary information that might be useful for deciphering pathways leading to preeclampsia and biomarkers for this disease.
Collapse
Affiliation(s)
- Vinoth K Kothandan
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alisa Komsky-Elbaz
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Department of Entomology, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Juliana S Powell
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianping Xia
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Tony J Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
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
|
30
|
Liu Y, Liu H, Zhu C, Yang Y, Shen Z, Shan G, Chen L, Zhou Y. Tumor Small Extracellular Vesicle-Transmitted LncRNA CATED Promotes Platinum-Resistance in High-Grade Serous Ovarian Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05963. [PMID: 40492382 DOI: 10.1002/advs.202505963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/15/2025] [Indexed: 06/12/2025]
Abstract
High-grade serous ovarian cancer (HGSOC) is the most lethal type of gynecological cancer, and platinum-resistance is a serious challenge in its treatment. Long non-coding RNAs (lncRNAs) play critical regulatory roles in the occurrence and development of cancers. Here, using RNA sequencing of tumor small extracellular vesicles (sEVs) from HGSOC patients, the lncRNA CATED is identified as significantly upregulated in both tumors and tumor-derived sEVs in platinum-resistant HGSOC, and low CATED levels correlate with good prognosis. Functionally, CATED enhances cisplatin resistance by promoting cell proliferation and inhibiting apoptosis in vitro and in vivo. These effects could be transferred via CATED-overexpressing sEVs from donor cells and HGSOC tumor sEVs. Mechanistically, CATED binds to and upregulates DHX36 via PIAS1-mediated SUMOylation at the K105 site, and elevated DHX36 levels increase downstream RAP1A protein levels by enhancing RAP1A mRNA translation, consequently activating the MAPK pathway to promote platinum-resistance in HGSOC. Antisense oligonucleotide mediated knockdown of CATED reverse platinum-resistance in sEV-transmitted mouse models via the DHX36-RAP1A-MAPK pathway. This study newly identifies a sEV-transmitted lncRNA CATED in driving HGSOC platinum-resistance and elucidates the mechanism it regulates the interacting protein through SUMOylation. These findings also provide a novel strategy for improving chemotherapy in HGSOC by targeting CATED.
Collapse
Affiliation(s)
- Yi Liu
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hanyuan Liu
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Chenchen Zhu
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yan Yang
- Hefei National Laboratory for Physical Sciences at Microscale, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhen Shen
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Ge Shan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Liang Chen
- Department of Cardiology, The First Affiliated Hospital of USTC, The RNA Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
31
|
Zhang Q, Huang J, Chen X, Li L, Chen L, Zhou X, Zhao X, Liu M, Zhao W, Yan J, Wang Y, Su Y, Liu Y, Xu S, Zeng W. Mesenchymal Stem Cell-Derived Mitochondrial Transfer Promotes Tip Cell Phenotype via Glutathione Metabolic Reprogramming in Stroke Mice. ACS NANO 2025; 19:20452-20471. [PMID: 40448640 DOI: 10.1021/acsnano.4c15759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Angiogenesis is crucial to improving neurovascular remodeling poststroke. Therein, the transformation of endothelial cells (ECs) to tip cells is essential in initiating angiogenesis. Mitochondrial damage in ECs poststroke and associated metabolic disorder are key factors repressing angiogenesis, but the mechanisms are unknown. Here, we designed an Arg-Gly-Asp peptide (RGD)-modified, mitochondria-enriched, and extracellular vesicle mimetics (mitoEVMs) platform for mitochondrial transfer. RGD mediated the mesenchymal stem cell-derived mitochondria transfer to ECs around the lesion targetedly. We found MSC-derived mitochondria promoted tip cell transition and further stimulated angiogenesis after stroke, alleviated brain atrophy, and improved functional rehabilitation. We noticed mitochondrial transfer rescued mitochondrial function in ECs and reprogrammed glutathione metabolism to activate the mTORC1 pathway, upregulated the expression of p4E-BP1 and VEGFR2, and ultimately facilitated tip cell transition. Our work elucidates the mechanism of MSC-derived mitochondrial transfer in poststroke treatment and proposes a potential approach for rehabilitation after stroke.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
- Department of Pain and Rehabilitation, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiaxin Huang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Xi Chen
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lang Li
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Lin Chen
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Xin Zhou
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Xingli Zhao
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Min Liu
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Wenyan Zhao
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Juan Yan
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Yueying Wang
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Yang Su
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
| | - Yong Liu
- Department of Pain and Rehabilitation, Xinqiao Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shangcheng Xu
- School of public health, Chongqing Medical University, Chongqing 400016, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400038, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
32
|
Lu X, Xu R, Dong X, Bai D, Ji W, Chen X, Chen H, Hou C, Gao J. Cell-derived exosome therapy for diabetic peripheral neuropathy: a preclinical animal studies systematic review and meta-analysis. Stem Cell Res Ther 2025; 16:297. [PMID: 40490808 DOI: 10.1186/s13287-025-04432-0] [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: 04/23/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUNDS Exosomes is a promising cell-free therapy for Diabetic peripheral neuropathy (DPN) that imposes long-term negative effects on patients' finances, mental health, and quality of life. We conducted a meta-analysis to assess the therapeutic effects of exosomes (such as SCs-derived, FCs-derived, BMSCs-derived, MSCs-derived, and Plasma-derived) on DPN. METHODS We searched nine databases from inception to February 2025, then two researchers independently screened studies, extracted data, and assessed the quality of included studies using SYRCLE's tool. The outcome indicators consisted of at least one of the three key DPN endpoints (electrophysiology, behavioural assessment, and nerve structure) based on the Neurodiab guidelines. R 4.4.2 software was used to conduct all statistical analyses. RESULTS 11 studies were identified, and the risk of bias in most studies was unclear generally. Pooled analyses demonstrated that exosome improved the nerve conduction velocity [MCV (SMD = 4.71 [2.18;7.25], P = 0.0003; I²= 91.8%), SCV (SMD = 1.07 [0.30;1.85], P = 0.0069; I²= 85.3%)], may restore IENFD [SMD = 1.46 [-0.85; 3.77], P = 0.2164; I²=88.7%], alleviated neuropathic pain [mechanical allodynia (SMD= -0.27 [-1.02;0.47], P = 0.4697; I2 = 85.0%), thermal hyperalgesia (SMD= -1.48 [-2.45;-0.50], P = 0.003; I2 = 88.4%)], ameliorated vascular function [blood flow perfusion in plantar (SMD = 2.84 [0.89; 4.80], P = 0.0043; I2 = 74.9%), blood flow perfusion in sciatic nerves (SMD = 2.62 [0.80; 4.43], P = 0.0047; I2 = 75.9%), vessel density (SMD = 2.69 [0.90; 4.49], P = 0.0032; I2 = 0%)], and restored the peripheral nerve structure [sciatic nerve fiber diameter (SMD = 3.29 [1.61; 4.96], P = 0.0066; I2 = 75.5%), axon diameter (SMD = 2.26 [1.64; 2.88], P < 0.0001; I2 = 54.3%), myelin sheath thickness (SMD = 2.56 [1.39; 3.72], P < 0.0001; I2 = 73.0%), g-ratio (SMD= -1.64 [-3.28; 0.00], P = 0.0502; I2 = 34.17)]. Furthermore, after exosome therapy, the expressions of NF-200 (SMD = 2.57 [0.39; 4.75], P = 0.0210; I2 = 33.0%), MBP (SMD = 2.27 [-1.49; 6.02], P = 0.1064; I2 = 59.0%), and S-100β (SMD = 1.90 [0.09; 3.72], P = 0.0399; I2 = 32.5%) evaluating axonal regeneration and remyelination increased significantly. Notably, high-glucose pretreatment of exosomes significantly attenuated these effects, while genetic overexpression modifications or novel dressings-mediated delivery partially counteracted this suppression. CONCLUSIONS Exosome therapy provides a novel therapeutic strategy for the benefit of neurovascular remodeling and functional recovery of DPN, especially when used in conjunction with exosome modification and novel dressings. To bridge the translational gap between preclinical and clinical studies, future research should conduct more large-scale, meticulously designed preclinical trials adhering to ARRIVE criteria before proceeding to clinical translation, to enhance translational rigor and mitigate risks associated with variability in study design.
Collapse
Affiliation(s)
- Xianying Lu
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ran Xu
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohui Dong
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dingxi Bai
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenting Ji
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Chen
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Chen
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaoming Hou
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jing Gao
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
33
|
Deng XH, Wu ZC, Sun Q, Huang LX, Xie YC, Lou DX, Li CG, Liu XQ, Zhou ZR, Tian T, Lian CL, Fu QL. The effects of Klotho delivering mesenchymal stem cell-derived small extracellular vesicles on acute kidney injury. J Nanobiotechnology 2025; 23:427. [PMID: 40481485 PMCID: PMC12144696 DOI: 10.1186/s12951-025-03499-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Acute kidney injury (AKI) is a life-threating syndrome characterized by sudden loss of kidney function, and its management is challenging and often suboptimal. Mesenchymal stem cells (MSCs) have shown promise in AKI therapy in pre-clinical and clinical trials; however, their clinical application still faces many challenges. MSC-derived small extracellular vesicles (sEV) may help overcome these challenges. In the current study, we overexpressed Klotho in MSCs and then isolated Klotho-loaded sEV (Klotho-sEV) using anion-exchange chromatography. Klotho-sEV displayed characteristics comparable to those of sEV in terms of size, morphology, conventional markers, and biosafety, as well as a higher abundance of Klotho protein. In rhabdomyolysis-induced AKI, sEV showed preferential tropism in injured kidneys. We found significantly and stably accelerated renal recovery, mitigated functional and histological abnormalities, stimulated tubular cell proliferation, reduced injury and inflammatory marker expression, and restored endogenous Klotho loss in mice after the administration of Klotho-sEV. In addition, Klotho-sEV treatment activated the mTOR and MEK1/2 signaling pathways. Proteomics and small RNA sequencing analyses of sEV and Klotho-sEV revealed abundant proteins and miRNAs involved in anti-inflammation and reno-protection, and Klotho-sEV showed characteristics that were different from those of sEV. In conclusion, Klotho-sEV may be a promising cell-free strategy for the treatment of AKI.
Collapse
Affiliation(s)
- Xiao-Hui Deng
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Zi-Cong Wu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Qi Sun
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Long-Xin Huang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Ying-Chun Xie
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Dong-Xiao Lou
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Chan-Gu Li
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Xiao-Qing Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Zhi-Rou Zhou
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Tian Tian
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China
| | - Chang-Lin Lian
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China
| | - Qing-Ling Fu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, PR China.
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, PR China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, PR China.
| |
Collapse
|
34
|
Liu JJJ, Liu D, To SKY, Wong AST. Exosomes in cancer nanomedicine: biotechnological advancements and innovations. Mol Cancer 2025; 24:166. [PMID: 40481526 PMCID: PMC12144782 DOI: 10.1186/s12943-025-02372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 05/28/2025] [Indexed: 06/11/2025] Open
Abstract
Exosomes, as natural intercellular messengers, are gaining prominence as delivery vehicles in nanomedicine, offering a superior alternative to conventional synthetic nanoparticles for cancer therapeutics. Unlike lipid, polymer, or metallic nanoparticles, which often face challenges related to immunogenicity, targeting precision, and off-tumor toxicity, exosomes can effectively encapsulate a diverse range of therapeutic agents while exhibiting low toxicity, favorable pharmacokinetics, and organotropic properties. This review examines recent advancements in exosome bioengineering over the past decade. Innovations such as microfluidics-based platforms, nanoporation, fusogenic hybrids, and genetic engineering have significantly improved loading efficiencies, production scalability, and pharmacokinetics of exosomes. These advancements facilitate tumor-specific cargo delivery, resulting in substantial improvements in retention and efficacy essential for clinical success. Moreover, enhanced biodistribution, targeting, and bioavailability-through strategies such as cell selection, surface modifications, membrane composition alterations, and biomaterial integration-suggests a promising future for exosomes as an ideal nanomedicine delivery platform. We also highlight the translational impact of these strategies through emerging clinical trials. Additionally, we outline a framework for clinical translation that focuses on: cargo selection, organotropic cell sourcing, precision loading methodologies, and route-specific delivery optimization. In summary, this review emphasizes the potential of exosomes to overcome the pharmacokinetic and safety challenges that have long impeded oncology drug development, thus enabling safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Jacky J J Liu
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Duanrui Liu
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Sally K Y To
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, 17 W, Hong Kong Science and Technology Parks, New Territories, Hong Kong.
| | - Alice S T Wong
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
| |
Collapse
|
35
|
Leung PYM, Graver AS, Katerelos M, Skene A, Whitlam JB, Power DA, Mount PF. Glycolytic control proteins in urinary extracellular vesicles are elevated during kidney transplant T cell-mediated rejection. BMC Nephrol 2025; 26:283. [PMID: 40481420 PMCID: PMC12144834 DOI: 10.1186/s12882-025-04196-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 05/19/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND A priority in kidney transplant management is the ability to monitor allograft health accurately, frequently and less-invasively. Metabolic reprogramming from fatty acid oxidation to glycolysis has been associated with kidney injury. Given the histological localisation of T cell-mediated rejection (TCMR) to the tubulointerstitium, we hypothesised that expression of glycolytic control proteins contained in urinary extracellular vesicles (UEV) may increase during TCMR. METHODS In this prospective observational study, urine samples were collected from kidney transplant recipients prior to indication biopsy. UEV were separated by differential ultracentrifugation. Vesicle markers, glycolytic control proteins and CD3 were assayed by immunoblotting. Differences in protein detection were compared across biopsy diagnoses (TCMR versus not) and Banff lesion scores. RESULTS 51 paired urine and biopsy samples from 43 subjects were included. The TCMR group comprised of 6 cases of TCMR and 1 borderline TCMR. The remaining 44 samples comprised a "No TCMR" group. There was significant increase in phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4) (p = 0.018) in TCMR compared to No TCMR, and similarly when tubulitis (p = 0.037) and interstitial inflammation (p = 0.047) were present. Total inflammation score ≥ 1 was associated with increases in PFKFB2 (p = 0.027), PFKFB3 (p = 0.090) and PFKFB4 (p = 0.0098). Interstitial fibrosis was associated with increased PFKFB2 (p = 0.0045) and PFKFB3 (p = 0.045). CD3 + UEV did not correlate with TCMR diagnosis. When combining the four glycolytic control proteins governing the phosphofructokinase-1 step of glycolysis (PFK-L, PFKFB2, PFKFB3 and PFKFB4), presence of ≥ 3 markers discriminated TCMR with ROC AUC of 0.73 (95% CI 0.50-0.96). CONCLUSION Increased rate-limiting enzymes of glycolysis in UEV were detected in association with tubulointerstitial inflammation and fibrosis. This suggests altered energy metabolism in the form of increased renal glycolysis occurring in the tubular epithelium, consistent with findings in native kidney injury. Further work is required to evaluate whether this could serve as a non-invasive strategy to study pathology in kidney transplantation.
Collapse
Affiliation(s)
- P Y M Leung
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Parkville, Australia.
- Department of Nephrology, Austin Health, 145 Studley Road, Heidelberg, Melbourne, VIC 3084, Australia.
| | - A S Graver
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Parkville, Australia
- Department of Nephrology, Austin Health, 145 Studley Road, Heidelberg, Melbourne, VIC 3084, Australia
- Australian Centre for Transplantation Excellence and Research, Austin Health, Melbourne, Australia
| | - M Katerelos
- Department of Nephrology, Austin Health, 145 Studley Road, Heidelberg, Melbourne, VIC 3084, Australia
| | - A Skene
- Department of Anatomical Pathology, Austin Health, Melbourne, Australia
| | - J B Whitlam
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Parkville, Australia
- Department of Nephrology, Austin Health, 145 Studley Road, Heidelberg, Melbourne, VIC 3084, Australia
- Australian Centre for Transplantation Excellence and Research, Austin Health, Melbourne, Australia
| | - D A Power
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Parkville, Australia
- Department of Nephrology, Austin Health, 145 Studley Road, Heidelberg, Melbourne, VIC 3084, Australia
| | - P F Mount
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Parkville, Australia
- Department of Nephrology, Austin Health, 145 Studley Road, Heidelberg, Melbourne, VIC 3084, Australia
| |
Collapse
|
36
|
Silva TF, Hutchins E, Zhao W, Ciani Y, Kim M, Ko E, Mariscal J, Qiu Z, Bedier F, Kittel A, Zhou B, Wang Y, Hall M, Galasso F, Reiman R, Freeman MR, Parker S, Van Eyk J, Yang W, Posadas E, Guarnerio J, Nolan J, Théry C, Zijlstra A, Stott S, You S, Demichelis F, Boutros PC, Van Keuren-Jensen K, Di Vizio D. Extracellular vesicle heterogeneity through the lens of multiomics. Cell Rep Med 2025:102161. [PMID: 40482644 DOI: 10.1016/j.xcrm.2025.102161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 11/19/2024] [Accepted: 05/08/2025] [Indexed: 06/11/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous in size, biogenesis, content, and function. Aggressive cancer cells release a distinct, poorly characterized, and particularly large EV subtype, namely large oncosomes (LOs). This study employs an optimized method to improve LO yields and integrates mass spectrometry and RNA sequencing (RNA-seq) to profile their molecular cargo. A consistent set of proteins enriched in LOs is identified across glioma, prostate, and breast cancer cell lines. These proteins are also present as mRNA in LOs from the prostate cancer model and are abundant in plasma LOs from 20 patients with metastasis. Single-LO RNA-seq confirms bulk LO cargo, demonstrating the utility of single-cell technologies for large vesicle analysis. Our patient study provides proof-of-principle evidence that we can use multiomics to delve into EV heterogeneity, biogenesis, and composition. It also suggests that plasma LOs help stratify patients, supporting their potential prognostic value for developing a multi-analyte approach for liquid biopsy.
Collapse
Affiliation(s)
- Taylon F Silva
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Wenyan Zhao
- Jonsson Comprehensive Cancer Center, University of California: Los Angeles, Los Angeles, CA, USA
| | - Yari Ciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Minhyung Kim
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Emily Ko
- Department of Radiation Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Javier Mariscal
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Zhuyu Qiu
- Jonsson Comprehensive Cancer Center, University of California: Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California: Los Angeles, Los Angeles, CA, USA
| | - Fatima Bedier
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Agnes Kittel
- HUN-REN Institute of Experimental Medicine, Budapest, Hungary
| | - Bo Zhou
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yang Wang
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Megan Hall
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Francesca Galasso
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Rebecca Reiman
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Michael R Freeman
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sarah Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Wei Yang
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Edwin Posadas
- Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Medicine, David Geffen School of Medicine, University of California: Los Angeles, Los Angeles, CA, USA
| | - Jlenia Guarnerio
- Department of Radiation Oncology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Hematology Oncology, David Geffen Medical School, University of California: Los Angeles, Los Angeles, CA, USA
| | - John Nolan
- Scintillon Institute for Biomedical and Bioenergy Research, San Diego, CA, USA
| | - Clotilde Théry
- Institut Curie, PSL Research University, INSERMU932, Paris, France
| | - Andries Zijlstra
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shannon Stott
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA; BioMEMS Resource Center, Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sungyong You
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Paul C Boutros
- Jonsson Comprehensive Cancer Center, University of California: Los Angeles, Los Angeles, CA, USA; Department of Human Genetics, University of California: Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California: Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California: Los Angeles, Los Angeles, CA, USA.
| | | | - Dolores Di Vizio
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Samuel Oschin Comprehensive Cancer Institute, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
37
|
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
|
38
|
Trautmann-Rodriguez M, Fromen CA. Nanoparticle-Based Pulmonary Immune Engineering. Annu Rev Chem Biomol Eng 2025; 16:249-270. [PMID: 40073112 DOI: 10.1146/annurev-chembioeng-082223-105117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Respiratory conditions represent a significant global healthcare burden impacting hundreds of millions worldwide and necessitating new treatment paradigms. Pulmonary immune engineering using synthetic nanoparticle (NP) platforms can reprogram immune responses for therapeutically beneficial or protective responses directly within the lung tissue. However, effectively localizing these game-changing approaches to the lung remains a significant challenge due to the lung's natural defense. We highlight the target pulmonary immune cells and address advances to localize NPs to the lung via both aerosol and vascular delivery. For each administration route, we discuss physiochemical design rules and recent immune-modulatory successes of synthetic, extracellular vesicle, and cell-mediated NP delivery. We aim to provide readers with an updated summary of this emerging field and offer a roadmap for future research aimed at enhancing the efficacy of pulmonary immunotherapies.
Collapse
Affiliation(s)
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware, USA;
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
39
|
Seth G, Singh S, Sharma G, Suvedi D, Kumar D, Nagraik R, Sharma A. Harnessing the power of stem cell-derived exosomes: a rejuvenating therapeutic for skin and regenerative medicine. 3 Biotech 2025; 15:184. [PMID: 40417660 PMCID: PMC12102458 DOI: 10.1007/s13205-025-04345-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/04/2025] [Indexed: 05/27/2025] Open
Abstract
Exosomes are small extracellular vesicles produced by most cell types and contain proteins, lipids, and nucleic acids (non-coding RNAs, mRNA, and DNA) that can be released by donor cells to influence the function of recipient cells. Skin photoaging is the premature aging of skin structures caused by prolonged exposure to ultraviolet (UV), as demonstrated by depigmentation, roughness, rhytides, elastosis, and precancerous alterations. Exosomes are associated with aging processes such as oxidative damage, inflammation, and senescence. Exosomes' anti-aging properties have been linked to various in vitro and preclinical investigations. There are still several unanswered questions about the use of MSC exosomes for skin rejuvenation, despite encouraging results. Uncertainty surrounds the precise processes by which exosomes stimulate the creation of collagen, skin tissue via a variety of mechanisms, including reduced matrix metalloproteinase (MMP) expression, increased collagen and elastin production, and modulation of intracellular signaling pathways and intercellular communication. These findings suggest the therapeutic potential of exosomes in skin aging. This review provides information on the molecular mechanisms and consequences of exosome anti-aging.
Collapse
Affiliation(s)
- Gracy Seth
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Siddharth Singh
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Geetansh Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Divyesh Suvedi
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Dinesh Kumar
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Rupak Nagraik
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, 248002 India
| | - Avinash Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, 248002 India
| |
Collapse
|
40
|
Fujii N, Urabe F, Yamamoto S, Inoue K, Kimura T, Shiraishi K. Extracellular vesicles in renal cell carcinoma: A review of the current landscape and future directions. Urol Oncol 2025; 43:370-379. [PMID: 40069067 DOI: 10.1016/j.urolonc.2025.02.022] [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] [Accepted: 02/23/2025] [Indexed: 05/19/2025]
Abstract
Liquid biopsy, a minimally invasive biopsy method that uses patient body fluids (e.g., blood, urine, or saliva), is considered a useful biomarker for early diagnosis, monitoring of tumor progression, and evaluating treatment efficacy. Extracellular vesicles (EVs), a diverse group of particles classified according to their size and biosynthetic method, are liquid bilayer structures released from various cells. EVs contain specific information, such as DNA, RNA, and proteins derived from released cells. Consequently, they have attracted attention for use in liquid biopsy. EV-derived microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are useful biomarkers for cancer diagnosis, tumor progression, and drug treatment resistance. Renal cell carcinoma (RCC), one of the most common type of urological cancer, accounts for 90% of all renal tumors. In contrast to prostate cancer, for which a tumor marker has been established, clinically applicable and useful biomarkers remain to be established for RCC. EV-derived miRNAs and lncRNAs have been identified as useful biomarkers in several types of carcinoma for determining the diagnosis and predicting tumor progression, and drug treatment resistance in patients with RCC. The development and identification of biomarkers to diagnose and predict tumor progression in RCC will improve the management and prognosis of patients with RCC. This review focuses on EV-derived miRNAs and lncRNAs and discusses the currently available EV-based biomarkers in RCC and their future prospects.
Collapse
Affiliation(s)
- Nakanori Fujii
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan.
| | | | - Keiji Inoue
- Department of Urology, Kochi Medical School, Nankoku, Kochi, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | - Koji Shiraishi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| |
Collapse
|
41
|
Hong Y, Feng Z, Ge Y, Xi Y, Zhang B, Wu J, Xia T, Tang B, Wang W, Chen J, Wang H, Xiao H. miR-145-enriched BMSCs-derived exosomes ameliorate neurogenic erectile dysfunction in aged rats via TGFBR2 inhibition. Regen Ther 2025; 29:455-465. [PMID: 40308644 PMCID: PMC12041780 DOI: 10.1016/j.reth.2025.04.004] [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: 02/14/2025] [Revised: 03/13/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025] Open
Abstract
Background Neurogenic erectile dysfunction (ED) is a prevalent complication following radical prostatectomy in elderly patients, primarily resulting from the apoptosis of corpus cavernosum smooth muscle cells (CCSMCs) and the subsequent excessive fibrosis of the corpus cavernosum. Aim This study aimed to compare the therapeutic effects of exosomes derived from lentivirus-transfected miR-145 bone marrow mesenchymal stem cells (Exo-145) and unmodified BMSCs-derived exosomes (Exo) in aged rats with bilateral cavernous nerve injury (BCNI) and investigate the underlying mechanisms. Methods Twenty-four-month-old male rats were assigned to four groups, namely Sham, BCNI, Exo, and Exo-145. Three weeks after treatment, erectile function was assessed by measuring the maximal intracavernosal pressure to mean arterial pressure (ICP/MAP) ratio. Apoptosis and fibrosis were semi-quantitatively analyzed using TUNEL and Masson's trichrome staining, respectively. In vitro, CCSMCs were subjected to H2O2-induced oxidative stress, and the protective effects of Exo-145 were evaluated through flow cytometry and Western blot. Lastly, the targets and mechanisms of miR-145 were further validated using dual-luciferase reporter assays and rescue experiments. Results Exo-145 significantly outperformed Exo in restoring erectile function in aged BCNI rats, as evidenced by the significantly higher maximal ICP/MAP ratio, a marked reduction in TUNEL-positive cell count, and marked suppression of fibrosis in cavernous tissue. Moreover, Masson's trichrome staining displayed a substantial decrease in collagen deposition. In vitro, Exo-145 alleviated H2O2-induced apoptosis in CCSMCs by downregulating Cleaved Caspase-3 expression and Bax while concurrently upregulating Bcl-2 expression. TGFBR2 was identified as a direct target of miR-145 through dual-luciferase reporter assays, with its overexpression partially reversing the protective effects of Exo-145. Conclusion Exo-145 demonstrates superior efficacy compared to Exo in treating aged neurogenic ED by targeting TGFBR2 to alleviate apoptosis and fibrosis. It may represent a promising cell-free therapeutic option for neurogenic erectile dysfunction in elderly patients and could offer new perspectives for improving their prognosis.
Collapse
Affiliation(s)
- Yude Hong
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Urology, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Zejia Feng
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunlong Ge
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuhang Xi
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bowen Zhang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianjie Wu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tian Xia
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bowen Tang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Wang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Chen
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hua Wang
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
42
|
Bavafa A, Izadpanahi M, Hosseini E, Hajinejad M, Abedi M, Forouzanfar F, Sahab-Negah S. Exosome: an overview on enhanced biogenesis by small molecules. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6473-6508. [PMID: 39862264 DOI: 10.1007/s00210-024-03762-9] [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: 10/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
Exosomes are extracellular vesicles that received attention for their potential use in the treatment of various injuries. They communicate intercellularly by transferring genetic and bioactive molecules from parent cells. Although exosomes hold immense promise for treating neurodegenerative and oncological diseases, their actual clinical use is very limited because of their biogenesis and secretion. Recent studies have shown that small molecules can significantly enhance exosome biogenesis, thereby remarkably improving yield, functionality, and therapeutic effects. These molecules modulate critical pathways toward optimum exosome production in a mode that is either ESCRT dependent or ESCRT independent. Improved exosome biogenesis may provide new avenues for targeted cancer therapy, neuroprotection in neurodegenerative diseases, and regenerative medicine in wound healing. This review explores the role of small molecules in enhancing exosome biogenesis and secretion, highlights their underlying mechanisms, and discusses emerging clinical applications. By addressing current challenges and focusing on translational opportunities, this study provides a foundation for advancing cell-free therapies in regenerative medicine and beyond.
Collapse
Affiliation(s)
- Amir Bavafa
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Izadpanahi
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Hosseini
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Hajinejad
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Qaen Faculty of Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahsa Abedi
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
43
|
Nabariya DK, Knüpfer LM, Hartwich P, Killian MS, Centler F, Krauß S. Transcriptomic analysis of intracellular RNA granules and small extracellular vesicles: Unmasking their overlap in a cell model of Huntington's disease. Mol Cell Probes 2025; 81:102026. [PMID: 40090627 DOI: 10.1016/j.mcp.2025.102026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Huntington's disease (HD) arises from the abnormal expansion of a CAG repeat in the HTT gene. The mutant CAG repeat triggers aberrant RNA-protein interactions and translates into toxic aggregate-prone polyglutamine protein. These aberrant RNA-protein ineractions also seed the formation of cytoplasmic liquid-like granules, such as stress granules. Emerging evidence demonstrates that granules formed via liquid-liquid phase separation can mature into gel-like inclusions that persist within the cell and may act as precursor to aggregates that occur in patients' tissue. Thus, deregulation of RNA granules is an important component of neurodegeneration. Interestingly, both the formation of intracellular membrane-less organelles like stress granules and the secretion of small extracellular vesicles (sEVs) increase upon stress and under disease conditions. sEVs are lipid membrane-bound particles that are secreted from all cell types and may participate in the spreading of misfolded proteins and aberrant RNA-protein complexes across the central nervous system in neurodegenerative diseases like HD. In this study, we performed a comparative transcriptomic analysis of sEVs and RNA granules in an HD model. RNA granules and sEVs were isolated from an inducible HD cell model. Both sEVs and RNA granules were isolated from induced (HD) and non-induced (control) cells and analyzed by RNA sequencing. Our comparative analysis between the transcriptomics data of HD RNA granules and sEVs showed that: (I) intracellular RNA granules and extracellular RNA vesicles share content, (II) several non-coding RNAs translocate to RNA granules, and (III) the composition of RNA granules and sEVs is affected in HD cells. Our data showing common transcripts in intracellular RNA granules and extracellular sEVs suggest that formation of RNA granules and sEV loading may be related. Moreover, we found a high abundance of lncRNAs in both control and HD samples, with several transcripts under REST regulation, highlighting their potential role in HD pathogenesis and selective incorporation into sEVs. The transcriptome cargo of RNA granules or sEVs may serve as a source for diagnostic strategies. For example, disease-specific RNA-signatures of sEVs can serve as biomarker of central nervous system diseases. Therefore, we compared our dataset to transcriptomic data from HD patient sEVs in blood. However, our data suggest that the cell-type specific signature of sEV-secreted RNAs as well as their high variability may make it difficult to detect these biomarkers in blood.
Collapse
Affiliation(s)
- Deepti Kailash Nabariya
- Human and Neurobiology, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Lisa Maria Knüpfer
- Human and Neurobiology, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Patrick Hartwich
- Chemistry and Structure of Novel Materials, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Manuela S Killian
- Chemistry and Structure of Novel Materials, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Florian Centler
- Bioinformatics, School of Life Sciences, University of Siegen, Siegen, Germany
| | - Sybille Krauß
- Human and Neurobiology, Department of Chemistry and Biology, University of Siegen, Siegen, Germany.
| |
Collapse
|
44
|
Chen H, Pang B, Liu Z, Li B, Wang Q, Fan B, Han M, Gong J, Zhou C, Chen Y, Li Y, Jiang J. The Diagnostic Value of Plasma Small Extracellular Vesicle-Derived CAIX Protein in Prostate Cancer and Clinically Significant Prostate Cancer: A Study on Predictive Models. Prostate 2025; 85:723-741. [PMID: 40013658 PMCID: PMC12038087 DOI: 10.1002/pros.24879] [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: 05/18/2024] [Revised: 01/25/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Current diagnostic tools are inaccurate and not specific to prostate cancer (PCa) diagnosis. Cancer-derived small extracellular vehicles (sEVs) play a key role in intercellular communication. In this study, we examined the diagnostic value of plasma sEV-derived carbonic anhydrase IX (CAIX) protein for PCa and clinically significant prostate cancer (csPCa) diagnosis and avoiding unnecessary biopsies. METHODS Plasma samples (n = 230) were collected from the patients who underwent prostate biopsy with elevated prostate-specific antigen (PSA) levels. sEVs were isolated and characterized, and sEV protein CAIX was measured using an enzyme-linked immunosorbent assay. Independent predictors of csPCa (Gleason score ≥ 7) were identified, and a predictive model was established. A Nomogram for predicting csPCa was developed using data from the training cohort. RESULTS The expression of sEV protein CAIX was significantly higher in both PCa and csPCa compared to benign patients and nonsignificant PCa (nsPCa) (Gleason score < 7, p < 0.001). sEV protein CAIX performed well in distinguishing PCa from benign patients. The predictive model defined by sEV protein CAIX and PSA density (PSAD) demonstrated the highest discriminative ability for csPCa (AUC = 0.895), with diagnostic sensitivity and specificity of 82.5% and 85.8%, respectively. Furthermore, sEV protein CAIX is an effective predictor of 2-year biochemical recurrence (BCR) in PCa patients (p = 0.013), and its high expression is significantly associated with poorer BCR-free survival (p < 0.05). CONCLUSIONS Our findings demonstrate the excellent performance of sEV protein CAIX in PCa and csPCa diagnosis. The Nomogram-based csPCa predictive model incorporating sEV protein CAIX and PSAD exhibits strong predictive value. Additionally, assessing plasma sEV protein CAIX expression levels can further aid in evaluating patient prognosis and provide a basis for making effective treatment decisions.
Collapse
Affiliation(s)
- Haotian Chen
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Bairen Pang
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Zhihan Liu
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Benjie Li
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Qi Wang
- Cancer Care Centre, St George HospitalKogarahNew South WalesAustralia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW SydneyKensingtonNew South WalesAustralia
| | - Baokun Fan
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Meng Han
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Jie Gong
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Cheng Zhou
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| | - Yingzhi Chen
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
| | - Yong Li
- Cancer Care Centre, St George HospitalKogarahNew South WalesAustralia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW SydneyKensingtonNew South WalesAustralia
| | - Junhui Jiang
- The First Affiliated Hospital of Ningbo University, Health Science CenterNingbo UniversityNingboZhejiangChina
- Ningbo Clinical Research Center for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Translational Research Laboratory for Urology, Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiangChina
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System DiseasesNingboZhejiangChina
| |
Collapse
|
45
|
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
|
46
|
Saylor LM, Cherukuri R, Kammala AK, Richardson L, Ferrer M, Antich C, Frebert S, Han A, Menon R. Exosomal Delivery of Interleukin-10 Reduces Infection-Associated Inflammation in a 3D-Printed Model of a Humanized Feto-Maternal Interface. FASEB J 2025; 39:e70634. [PMID: 40356417 PMCID: PMC12103302 DOI: 10.1096/fj.202500545r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/05/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
Spontaneous preterm birth (PTB) is associated with fetal inflammatory responses that are due to infections. Effective interventions to minimize these fetal responses are limited as drugs do not usually cross the feto-maternal interface (FMi) barrier, and reliable models to test drug efficacy and other pharmacologic parameters have not been available. We leveraged New Approach Methods (NAMs), including employing extracellular vesicles (exosomes of 30-200 nm) to deliver the anti-inflammatory cytokine interleukin (IL)-10 and using a high-throughput 3D-printed FMi model to test the efficacy of this delivery. IL-10 encapsulated exosomes were prepared by encapsulating recombinant IL-10 (rIL-10) using electroporation (eIL-10) or by transfecting RAW264.7 cells with an IL-10-expression plasmid (tIL-10) that enabled the expression of IL-10 in the cells during exosome biogenesis, which was then collected. Using a biocompatible polymer resin, we 3D printed a two-chambered FMi scaffold to mimic the amnion-decidual (feto-maternal) interface. Microchannels were integrated into the lower portions of the scaffold to facilitate intercellular communication. The device was composed of a mix of cells and gelatin methacrylate hydrogel material (lower part) and cell-specific culture medium (upper part). We showed that empty exosomes and IL-10-loaded exosomes delivered to the maternal side of the scaffold were able to cross to the fetal side of our FMi device. Furthermore, the effectiveness of eIL-10 and tIL-100 (500 ng) in reducing LPS-induced FMi inflammation on both the maternal and fetal sides was demonstrated by measuring pro-inflammatory IL-6 and IL-8 concentrations via multiplex assays at 6 h and 24 h timepoints. We determined that our 3D-printed two-chamber FMi model enabled the propagation of IL-10 encapsulated exosomes between the interconnected chambers. LPS treatment to the maternal decidua induced expression of pro-inflammatory IL-6 (p < 0.001) and IL-8 (p < 0.001) in both decidua and amnion compared with healthy (control) conditions. Co-treatment of LPS along with IL-10-loaded exosomes, regardless of its formulation, significantly reduced levels of the maternal and fetal inflammatory cytokines IL-6 and IL-8 at both 6 and 24 h after delivery. A high-throughput 3D-printed FMi model was used to show that IL-10 encapsulated exosomes can reduce infection-induced FMi inflammation. We describe two NAMs with the potential to significantly improve perinatal medicine: (1) an exosomal drug delivery method that protects the drug and can cross feto-maternal barriers and (2) a 3D-printed device that can be used for high-throughput drug screening.
Collapse
Affiliation(s)
- Leah M Saylor
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Rahul Cherukuri
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Ananth K Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Lauren Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory, National Center for Advancing Translational Sciences, National Institute of Sciences, Bethesda, MD, USA
| | - Cristina Antich
- 3D Tissue Bioprinting Laboratory, National Center for Advancing Translational Sciences, National Institute of Sciences, Bethesda, MD, USA
| | - Shayne Frebert
- 3D Tissue Bioprinting Laboratory, National Center for Advancing Translational Sciences, National Institute of Sciences, Bethesda, MD, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| |
Collapse
|
47
|
Soni N, Rameshwari R. "Silent messengers of chaos: unveiling the dual threat of immune infiltrates in Japanese encephalitis virus neuroinflammatory storm". Virol J 2025; 22:173. [PMID: 40450327 DOI: 10.1186/s12985-025-02805-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 05/22/2025] [Indexed: 06/03/2025] Open
Affiliation(s)
- Naina Soni
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India.
| | - Rashmi Rameshwari
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
| |
Collapse
|
48
|
Hutson HK, Qin G, Cai C, Nestorova GG. Comparative proteomic profiling of glioblastoma and healthy brain cell-derived extracellular vesicles reveals enrichment of cancer-associated proteins. J Proteomics 2025; 316:105418. [PMID: 40058457 DOI: 10.1016/j.jprot.2025.105418] [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: 09/25/2024] [Revised: 01/30/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
Extracellular vesicles (EVs)-mediated cellular communication plays a role in cancer development and progression. This study focuses on identifying glioblastoma-specific EV protein markers through a comparative mass spectrometry bottom-up proteomic analysis of the LN-229 cell line and human neurons, astrocytes, and endothelial brain cells (HEBCs) using timsTOF Pro 2 instrument. The statistically significant upregulated proteins with fold change greater than 2 in the glioblastoma-derived EVs were clustered based on physical and functional interactions using the STRING database and analyzed using Gene Ontology enrichment. LN229-derived EVs contained an average of 2635 proteins, while human astrocytes, neurons, and HEBC encapsulated 2647, 716, and 2285 proteins, respectively. NanoParticle Tracking Analysis indicated that glioblastoma-derived EVs exhibited greater size variability compared to EVs from healthy cells. Statistical analysis identified 25 statistically significant proteins with increased levels in LN229 EVs relative to at least two healthy cell lines suggesting their potential as glioblastoma markers. Functional clustering using the STRING database and GO analysis indicated involvement in epigenetic regulation, metastasis, angiogenesis, and protein folding. Post-translational modification analysis identified a subset of 17 proteins unique to the cancer-derived EVs involved in chromatin regulation, extracellular matrix remodeling, and basement membrane organization pathways, highlighting their role in tumor progression.
Collapse
Affiliation(s)
- Hope K Hutson
- Molecular Science and Nanotechnology, Louisiana Tech University, United States
| | - Guoting Qin
- College of Optometry, University of Houston, United States
| | - Chengzhi Cai
- Department of Chemistry, University of Houston, United States
| | | |
Collapse
|
49
|
Tavares NT, Lourenço C, Constâncio V, Fernandes-Pontes F, Fonseca D, Silva-Santos R, Braga I, Maurício J, Henrique R, Liu M, Weiss RS, Bagrodia A, Jerónimo C, Lobo J. MicroRNA-371-373 cluster extracellular vesicle-based communication in testicular germ cell tumors. Cell Commun Signal 2025; 23:252. [PMID: 40448114 DOI: 10.1186/s12964-025-02250-8] [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: 03/30/2025] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
Testicular germ cell tumors (TGCTs) represent the most common type of cancer in young adults. The cluster of microRNAs 371-373 is highly upregulated in TGCTs, and detection of miR-371a-3p specifically is currently being developed for clinical implementation as a sensitive and specific biomarker for TGCT, except for teratoma. Extracellular vesicles (EVs) are nano-sized particles used for cell communication, being increasingly regarded as potential sources of cancer biomarkers. Thus, the aim of this study was to characterize EVs from a wide range of TGCT samples, including cell lines, tissue explants and matched plasma samples from patients and healthy donors, and then use these samples to assess microRNA expression (miR-371-373 cluster and let-7e). TGCT-derived EVs were successfully isolated and characterized according to MISEV guidelines. TGCT cell lines showed different levels of EV-derived miR-371-373 cluster and let-7e. Upon differentiation of NT2 cells with ATRA, both cellular and EV-derived miR-371-373 cluster were downregulated, whereas let-7e was upregulated. TGCT patient samples presented high levels of EV-derived miR-371-373, except for the teratoma samples. We conclude that a significant portion of the circulating miR-371-373 cluster used as a TGCT biomarker in the clinic is secreted into EVs, and that this cluster and the let-7 family of microRNAs may be related with TGCT intercellular communication and differentiation.
Collapse
Affiliation(s)
- Nuno Tiago Tavares
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - Catarina Lourenço
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
- i3S - Instituto de Investigação e Inovação Em Saúde - University of Porto, Porto, 4200-135, Portugal
- Instituto Nacional de Engenharia Biomédica, Porto, 4200-135, Portugal
| | - Vera Constâncio
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Doctoral Programme in Biomedical Sciences, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - Fernanda Fernandes-Pontes
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
| | - Diana Fonseca
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
| | - Rui Silva-Santos
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
| | - Isaac Braga
- Department of Urology, Urology Clinic, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
| | - Joaquina Maurício
- Department of Medical Oncology, Urology Clinic, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - Michelle Liu
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Robert S Weiss
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / CI-IPOP@RISE Health Research Network - Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC Raquel Seruca), Porto, 4200-072, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, 4200-072, Portugal.
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, 4050-313, Portugal.
| |
Collapse
|
50
|
You Y, Zhang Z, Cortes S, Nguyen SN, Vadakattu P, Melvin BC, Jr Mann SD, Ray NB, Bregendahl M, McLean PJ, Gonzalez-Perez MP, Ikezu S, Ikezu T. Rapid and high-yield recovery of plasma-derived extracellular vesicles using modified chromatography with soluble protein depletion for biomarker discovery. Cell Commun Signal 2025; 23:253. [PMID: 40448170 DOI: 10.1186/s12964-025-02263-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
Extracellular vesicles (EVs) are critical mediators of intercellular communication by transferring proteins, lipid and nucleic acids between cells. EVs in biofluids, particularly blood, have gathered significant interest as potential biomarkers for disease diagnosis. However, isolating EVs from blood poses a challenge due to the high concentration of plasma proteins, which obscure the detection of low abundant EV-associated proteins. Here, we optimized a simplified and efficient method for isolating plasma-derived EVs by combining size exclusion chromatography (SEC) with flow-through chromatography using Capto Core 700 beads. A brief incubation of SEC-derived EV fractions with Capto Core beads (qEV + CC) enabled us to isolate intact, high-purity EVs with reduced soluble plasma protein contamination. As a comparison, MagReSyn-based method was not compatible with elution of intact EVs after the purification and showed significant contamination of soluble plasma proteins. Data-independent acquisition-based liquid chromatography-mass spectrometry of isolated plasma-EVs using the qEV + CC approach identified over 1,000 EV-associated proteins, including an increased presence of brain derived proteins and markers linked to neurodegenerative diseases, such as amyloid precursor protein and apolipoprotein E. These findings were further validated by super-resolution microscopy at a single EV resolution. Bioinformatic pathway and network analyses revealed enrichment of pathways involved in RNA processing, cell adhesion and synaptic function, highlighting the potential of EV molecules for broad disease biomarker discovery. Our findings present an optimized method for efficient purification of plasma-derived EVs, providing a valuable tool for advancing EV-based biomarker development.
Collapse
Affiliation(s)
- Yang You
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Zhengrong Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Samuel Cortes
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Biomedical Education and Anatomy, The Ohio State University, Columbus, OH, USA
| | - Son N Nguyen
- Mass Spectrometry Facility, University of Massachusetts Medical School, Shrewsbury, MA, USA
| | | | | | - Sean D Jr Mann
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Pam J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Seiko Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Regenerative Science Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA.
| |
Collapse
|