1
|
Xie Y, Mi X, Xing Y, Dai Z, Pu Q. Past, present, and future of exosomes research in cancer: A bibliometric and visualization analysis. Hum Vaccin Immunother 2025; 21:2488551. [PMID: 40207548 PMCID: PMC11988232 DOI: 10.1080/21645515.2025.2488551] [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: 01/21/2025] [Revised: 03/12/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer seriously threatens the lives and health of people worldwide, and exosomes seem to play an important role in managing cancer effectively, which has attracted extensive attention from researchers in recent years. This study aimed to scientifically visualize exosomes research in cancer (ERC) through bibliometric analysis, reviewing the past, summarizing the present, and predicting the future, with a view to providing valuable insights for scholars and policy makers. Researches search and data collection from Web of Science Core Collection and clinical trial.gov. Calculations and visualizations were performed using Microsoft Excel, VOSviewer, Bibliometrix R-package, and CiteSpace. As of December 1, 2024, and March 8, 2025, we identified 8,001 ERC-related publications and 107 ERC-related clinical trials, with an increasing trend in annual publications. Our findings supported that China, Nanjing Medical University, and International Journal of Molecular Sciences were the most productive countries, institutions, and journals, respectively. Whiteside, Theresa L. had the most publications, while Théry, C was the most co-cited scholar. In addition, Cancer Research was the most co-cited journal. Spatial and temporal distribution of clinical trials was the same as for publications. High-frequency keywords were "extracellular vesicle," "microRNA" and "biomarker." Additional, "surface functionalization," "plant," "machine learning," "nanomaterials," "promotes metastasis," "engineered exosomes," and "macrophage-derived exosomes" were promising research topics. Our study comprehensively and visually summarized the structure, hotspots, and evolutionary trends of ERC. It would inspire subsequent studies from a macroscopic perspective and provide a basis for rational allocation of resources and identification of collaborations among researchers.
Collapse
Affiliation(s)
- Yafei Xie
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xingqi Mi
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yikai Xing
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhangyi Dai
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Pu
- Department of Thoracic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
2
|
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
|
3
|
Yang S, Zhu H, Jin H, Wang K, Song J, Sun N, Liu Y, Yin X, Wang R, Wu X, Liu H, Zhang C, Zhao W, Yu F. Bio-orthogonal-labeled exosomes reveals specific distribution in vivo and provides potential application in ARDS therapy. Biomaterials 2025; 319:123208. [PMID: 40023928 DOI: 10.1016/j.biomaterials.2025.123208] [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: 11/07/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Exosomes derived from specific cells may be useful for targeted drug delivery, but tracking them in vivo is essential for their clinical application. However, their small size and complex structure challenge the development of exosome-tracking techniques, and traditional labeling methods are limited by weak affinity and potential toxicity. To address these issues, here we developed a novel bio-orthogonal labeling strategy based on phosphatidylinositol derivatives to fluorescently label exosomes from various human and mouse cell types. The different cell-derived exosomes revealed organ-specific distribution patterns and a favorable safety profile. Notably, 4T1 cell-derived exosomes specifically targeted the lungs. When used as drug carriers loaded with anti-inflammatory resveratrol, these exosomes showed significant therapeutic efficacy in mice with acute respiratory distress syndrome (ARDS), effectively reducing inflammatory responses, mitigating pulmonary fibrosis, and restoring lung tissue morphology and function. Our findings provide a novel exosome labeling strategy and an invaluable tool for their in vivo tracking and targeting screening, while exosomes that specifically target the lungs offer a potential therapeutic strategy for organ-specific diseases such as ARDS.
Collapse
Affiliation(s)
- Song Yang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Haomiao Zhu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China; Department of Pharmacy, Qilu Hospital, Shandong University, No.107 Cultural West Road, Jinan, 250012, China
| | - Hongzhen Jin
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Kun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Junna Song
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Na Sun
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Yonghui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China; School of Chemistry, Tiangong University, No.399 BinShuiXi Road, Tianjin, 300387, China
| | - Xiaona Yin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Rui Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China
| | - Xiao Wu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Huadong Liu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China
| | - Chunling Zhang
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China.
| | - Wei Zhao
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China.
| | - Fan Yu
- Qingdao Central Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, No. 369, Qingdao National High-Tech Industrial Development Zone, Qingdao, 266113, China; State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Molecular Drug Research and KLMDASR of Tianjin, Nankai University, No.38 Tongyan Road, Haihe Education Park, Tianjin, 300350, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Liu YJ, Miao HB, Lin S, Chen Z. Exosomes derived let-7f-5p is a potential biomarker of SLE with anti-inflammatory function. Noncoding RNA Res 2025; 12:116-131. [PMID: 40144341 PMCID: PMC11938083 DOI: 10.1016/j.ncrna.2025.02.004] [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: 10/09/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
This study found that in patients with SLE (n = 5), lethal (let)-7f-5p expression was significantly downregulated in peripheral blood mononuclear cells. Further, high-throughput RNA sequencing was used to mine the differential transcriptome expression in renal tissue exosomes of systemic lupus erythematosus (SLE)-prone mice, and bioinformatics was utilized to analyze non-coding RNAs and coding RNAs in exosomes for their possible roles in SLE. In renal tissues of MRL/lpr SLE-prone mice with exosomes and Pristane-induced SLE mice, we also demonstrated aberrant expression levels of microRNA (miRNA) let-7f-5p. Meanwhile, in the macrophage inflammation model, the expression levels of let-7f-5p were downregulated, that of guanylate binding protein (Gbp2 and Gbp7) were upregulated, and the inflammatory state of macrophages was alleviated following transfection with the let-7f-5p mimic. Co-culturing mesenchymal stem cells with a macrophage model of inflammation resulted in increased let-7f-5p expression and downregulated inflammatory factors, Gbp2 and Gbp7 expression in macrophages. Dual luciferase reporter gene assays confirmed that let-7f-5p directly binds to the 3' UTR of Gbp7 to regulate its expression. Let-7f-5p regulation of the Gbp family is involved in SLE pathogenesis and is a biomarker associated with the inflammatory response with potential clinical applications.
Collapse
Affiliation(s)
- Yi-jing Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hai-bing Miao
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia
| | - Zhen Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
7
|
Bertolio M, Li Q, Mowry FE, Reynolds KE, Alananzeh R, Wei H, Keum K, Jarvis R, Wu J, Yang Y. Glutamatergic Regulation of miRNA-Containing Intraluminal Vesicle Trafficking and Extracellular Vesicle Secretion From Cortical Neurons. J Extracell Vesicles 2025; 14:e70100. [PMID: 40439163 PMCID: PMC12120566 DOI: 10.1002/jev2.70100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 05/08/2025] [Indexed: 06/02/2025] Open
Abstract
Neuronal extracellular vesicles (microvesicles and exosomes) are emerging secreted vesicular signals that play important roles in the CNS. Currently, little is known about how glutamatergic signalling affects the subcellular localisation of exosome precursor intraluminal vesicles (ILVs), microRNA (miR) packaging into ILVs and in vivo spreading of neuronal EVs. By selectively labelling ILVs and exosomes (but not plasma membrane-derived MVs) with GFP-tagged human CD63 (hCD63-GFP) in cortical neurons, we found that glutamate stimulation significantly redistributes subcellular localisation of hCD63-GFP+ ILVs, especially decreasing its co-localisation with multi-vesicular body (MVB) marker Rab7 while substantially promoting EV secretion. Interestingly, glutamate stimulation only modestly alters EV miR profiles based on small RNA sequencing. Subsequent in vivo cortical neuronal DREADD activation leads to significantly more widespread hCD63-GFP+ area in hCD63-GFPf/+ mice, consistently supporting the stimulatory effect of glutamatergic activation on neuronal EV secretion and spreading. Moreover, in situ localisation of hCD63-GFP+ ILVs and hCD63-GFP+ secreted exosomes from specialised HB9+ and DAT+ neurons were also illustrated in the CNS. Taken together, our results demonstrated that glutamate activity stimulates neuronal exosome secretion and spreading in vitro and in vivo, but only modestly affects miR cargo packaging in neuronal exosomes.
Collapse
Affiliation(s)
- Marcela Bertolio
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Qiyi Li
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Francesca E. Mowry
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Kathryn E. Reynolds
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Rashed Alananzeh
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Haichao Wei
- Department of Neurosurgery, McGovern Medical SchoolThe University of Texas Health Science Center at Houston (UTHealth)HoustonTexasUSA
| | - Kyoeun Keum
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Rachel Jarvis
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Jiaqian Wu
- Department of Neurosurgery, McGovern Medical SchoolThe University of Texas Health Science Center at Houston (UTHealth)HoustonTexasUSA
| | - Yongjie Yang
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
- Graduate School of Biomedical SciencesTufts UniversityBostonMassachusettsUSA
| |
Collapse
|
8
|
Samanta A, Yoo MJ, Koh J, Lufkin SC, Lufkin T, Kraus P. Proteomic profiling of small extracellular vesicles from bovine nucleus pulposus cells. PLoS One 2025; 20:e0324179. [PMID: 40440285 PMCID: PMC12121814 DOI: 10.1371/journal.pone.0324179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/21/2025] [Indexed: 06/02/2025] Open
Abstract
Small extracellular vesicles (small EV) are a conserved means of communication across the domains of life and lately gained more interest in mammalian non-cancerous work as non-cellular, biological therapeutic with encouraging results in recent studies of chronic degenerative diseases. The nucleus pulposus (NP) is the avascular and aneural center of an intervertebral disc (IVD), home to unique niche conditions and affected in IVD degeneration. We investigated autologous and mesenchymal stem cell (MSC) small EVs for their potential to contribute to cell and tissue homeostasis in the NP niche via mass spectrometric proteome and functional enrichment analysis using adult and fetal donors. We compared these findings to published small EV databases and MSC small EV data. We propose several mechanisms associated with NP small EVs: Membrane receptor trafficking to modify signal responses promoting niche homeostasis; Redox and energy homeostasis via metabolic enzymes delivery; Cell homeostasis via proteasome delivery and immunomodulation beyond an association with a serum protein corona. The proteome signature of small EVs generated by NP parent cells is similar to previously published small EV data, yet with a focus on supplementing anaerobic metabolism and redox balance while contributing to the maintenance of an aneural and avascular microniche.
Collapse
Affiliation(s)
- Ankita Samanta
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Mi-Jeong Yoo
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Jin Koh
- The Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States of America
| | - Sina Charlotte Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| |
Collapse
|
9
|
Gan L, Guo X, Dong S, Sun C. The biology of exosomes and exosomal non-coding RNAs in cardiovascular diseases. Front Pharmacol 2025; 16:1529375. [PMID: 40492132 PMCID: PMC12147041 DOI: 10.3389/fphar.2025.1529375] [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: 11/16/2024] [Accepted: 04/07/2025] [Indexed: 06/11/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide, both in developed and developing countries. Despite the implementation of various measures in clinical practice that have shown certain curative effects, poor prognosis and irreversible pathological cardiac remodeling continue to limit the therapeutic effect of CVDs. There are still many new mechanisms worth exploring for the regulation of CVDs. Previous studies have highlighted the potential applicability of exosomes in CVDs, and significant research has been conducted in this area. In this review, we summarize the physiological mechanisms of exosomes and the basic research achievements in regulating CVDs via exosomal non-coding RNAs. We also discuss the limitations and prospects of exosome application in CVD treatment.
Collapse
Affiliation(s)
- Lu Gan
- Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofei Guo
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shichao Dong
- Department of Pharmacy, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chuan Sun
- Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
10
|
Stricker PEF, de Oliveira NB, Mogharbel BF, Irioda AC, da Rosa NN, Lührs L, Saçaki CS, Munhoz da Rocha I, Alves LR, Poubel SB, Cardoso da Silva J, Carvalho PC, Fischer JSDG, de Carvalho KAT. Proteomic Characterization of Extracellular Vesicles from Human Neural Precursor Cells: A Promising Advanced Therapy for Neurodegenerative Diseases. Int J Nanomedicine 2025; 20:6675-6699. [PMID: 40444011 PMCID: PMC12121667 DOI: 10.2147/ijn.s502031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 05/01/2025] [Indexed: 06/02/2025] Open
Abstract
Background The therapeutic effect of stem cells is attributed to their direct maturation into somatic cells and their paracrine effects, which influence the extracellular environment. One such component released is extracellular vesicles containing proteins and genetic materials with immunomodulatory functions and facilitating cell-to-cell communication. Purpose The study's main objective was to characterize extracellular vesicles (EVs) from Human Neural Precursor Cells (hNPCs). Methods Wharton's Jelly mesenchymal stem cells (WJ-MSCs) were isolated by explant technique and characterized by flow cytometry and trilineage differentiation. The hNPCs obtained from neurospheres were produced by seeding WJ-MSCs on a natural functional biopolymer matrix. EVs derived from WJ-MSCs and hNPCs were isolated by precipitation methodology and characterized by flow cytometry, nanoparticle tracking analysis (NTA), scanning electron microscopy (TEM), and proteomic. Results hNPCs expressed proteins and genes characteristic of neural precursor cells. The EVs were characterized by flow cytometry and showed varied expression for the markers CD63, CD9, and CD81, indicating different subpopulations based on their origin of formation. NTA and TEM of the EVs exhibited characteristic size, shape, and structural integrity consistent with the criteria established by the International Society for Extracellular Vesicles (ISEV). EV-hNPCs function enrichment analysis of the proteomic results showed that these vesicles presented abundant proteins directly involved in neuronal biological processes such as plasticity, transduction, postsynaptic density, and overall brain development. Discussion The results indicate that EVs derived from hNPCs maintain key neural precursor characteristics and exhibit marker variability, suggesting distinct subpopulations. Their structural integrity aligns with ISEV standards, supporting their potential as reliable biological entities. The proteomic analysis highlights their role in neuronal functions, reinforcing their applicability in neurodegenerative research and therapeutic strategies. Conclusion The EVs were successfully isolated from hNPCs with abundant proteins involved in neuronal processes, making them attractive for acellular therapies to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Priscila Elias Ferreira Stricker
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Nathalia Barth de Oliveira
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Bassam Felipe Mogharbel
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Ana Carolina Irioda
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Nádia Nascimento da Rosa
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Larissa Lührs
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Claudia Sayuri Saçaki
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Isadora Munhoz da Rocha
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, FIOCRUZ, Curitiba, PR, Brazil
| | - Lysangela Ronalte Alves
- Gene Expression Regulation Laboratory, Carlos Chagas Institute, FIOCRUZ, Curitiba, PR, Brazil
| | - Saloe Bispo Poubel
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| | - Julia Cardoso da Silva
- Computational Mass Spectrometry Group, Carlos Chagas Institute, FIOCRUZ, Curitiba, PR, Brazil
| | - Paulo Costa Carvalho
- Computational Mass Spectrometry Group, Carlos Chagas Institute, FIOCRUZ, Curitiba, PR, Brazil
| | | | - Katherine Athayde Teixeira de Carvalho
- Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, Curitiba, PR, Brazil
| |
Collapse
|
11
|
Esmaeili A, Esmaeili V, Shahverdi A, Eslaminejad MB. Engineered extracellular vesicles: a breakthrough approach to overcoming sperm cryopreservation challenges. Reprod Biol Endocrinol 2025; 23:75. [PMID: 40399922 PMCID: PMC12093887 DOI: 10.1186/s12958-025-01407-x] [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: 01/07/2025] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
Freezing sperm for artificial insemination (AI) has been common for decades, but this method causes damage to sperm, which affects its viability and fertility. Various strategies have been used to treat sperm cryopreservation complications, but their results are still not satisfactory. The latest approach in this field is using extracellular vesicles (EVs). The role of EVs in reproduction, such as spermatogenesis, sperm capacitation, and fertility has been proven. EVs can deliver proteins, lipids, nucleic acids, and other molecules to the sperm for repair. The EVs carry proteins, lipids, nucleic acids, and other molecules, which could be involved in sperm quality, functionality or fertility. The application of EV derived from animal and human cell sources for cryoinjury treatment indicates the improvement of sperm quality after freeze-thawing. In addition, different EV engineering methods regarding various EV cargos could be more influential for cryopreserved sperm treatment because they could provide EV customized content for delivering to cryoinjured sperm, according to their unique needs to enhance viability and fertility. In this review, first, we reminded the sperm cryopreservation complications, and next explained the conventional and modern strategies for overcoming them. Then, we have pointed out the role of EV in sperm development and the following mentioned the study results of using EV from different cell sources in sperm cryoinjuries repair. Also, we suggested several predisposing molecules (including microRNAs and proteins) for EV engineering to treat sperm cryopreservation complications by indirect engineering procedure, including genetic manipulation and incubation with therapeutic molecules, and direct engineering procedure, including electroporation, sonication, incubation, saponin permeabilization, extrusion, CaCl2-heat shock, and freeze/thawing. Finally, we discussed the limitations of EV application and ethical considerations in this context. In the meantime, despite these limitations, we pointed out the promising potential of the EV engineering strategies to reduce infertility rates by helping to overcome sperm cryopreservation challenges.
Collapse
Affiliation(s)
- Abazar Esmaeili
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Vahid Esmaeili
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
12
|
Zhang Y, Wang C, Shao H. Nanoplasmonic Sensing of Heterogeneous Extracellular Vesicles: From Bulk to Single Vesicles. SMALL METHODS 2025:e2500097. [PMID: 40391615 DOI: 10.1002/smtd.202500097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/16/2025] [Indexed: 05/22/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoscale membrane vesicles released by almost all cell types into the circulation. Depending on their biogenesis and cells of origin, EVs show considerable heterogeneity in their biophysical and biomolecular composition and can serve as reflective and dynamic blood biomarkers for personalized medicine. Conventional analytical technologies, however, often lack the compatibility to reveal nanoscale EV features and resolve vesicle heterogeneity. The past decade has since witnessed the development of various nanoplasmonic technologies to empower EV analysis, through bulk and single-vesicle characterization, at an unprecedented scale and resolution. These platforms achieve versatile measurements that are not only size-matched to EV dimensions but can also probe multiplexed biomolecular contents, thereby providing new insights into EV heterogeneity and enabling transformative clinical opportunities. In this review, key characteristics of EVs and their remarkable heterogeneity are introduced. The sensing principles of plasmonic platforms are also discussed, with recent technology developments highlighted to resolve EV heterogeneity, through bulk analyses of EV subpopulations as well as high-resolution single-EV measurements. An outlook is further provided on emerging opportunities, at the interface of biomarker discovery and technology innovation, to develop empowering nanoplasmonic EV platforms for personalized medicine. biosensing; bulk analysis; extracellular vesicles; nanoplasmonics; single-vesicle analysis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Chao Wang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Materials Science and Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 138673, Singapore
| |
Collapse
|
13
|
Longo A, Manganelli V, Misasi R, Riitano G, Caglar TR, Fasciolo E, Recalchi S, Sorice M, Garofalo T. Extracellular Vesicles in the Crosstalk of Autophagy and Apoptosis: A Role for Lipid Rafts. Cells 2025; 14:749. [PMID: 40422252 DOI: 10.3390/cells14100749] [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: 04/14/2025] [Revised: 05/13/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Autophagy and apoptosis are two essential mechanisms regulating cell fate. Although distinct, their signaling pathways are closely interconnected through various crosstalk mechanisms. Lipid rafts are described to act as both physical and functional platforms during the early stages of autophagic and apoptotic processes. Only recently has a role for lipid raft-associated molecules in regulating EV biogenesis and release begun to emerge. In particular, lipids of EV membranes are essential components in conferring stability to these vesicles in different extracellular environments and/or to facilitate binding or uptake into recipient cells. In this review we highlight these aspects, focusing on the role of lipid molecules during apoptosis and secretory autophagy pathways. We describe the molecular machinery that connects autophagy and apoptosis with vesicular trafficking and lipid metabolism during the release of EVs, and how their alterations contribute to the development of various diseases, including autoimmune disorders and cancer. Overall, these findings emphasize the complexity of autophagy/apoptosis crosstalk and its key role in cellular dynamics, supporting the role of lipid rafts as new therapeutic targets.
Collapse
Affiliation(s)
- Agostina Longo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tuba Rana Caglar
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Elena Fasciolo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Serena Recalchi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
14
|
Smith SM, Kumari A, Marvar JP, Onukwugha NE, Kang YT, Nagrath S. Stellate silicon microneedles for rapid point-of-care melanoma exosome isolation and detection via a lateral flow assay. Biosens Bioelectron 2025; 285:117560. [PMID: 40403613 DOI: 10.1016/j.bios.2025.117560] [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: 02/26/2025] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025]
Abstract
Melanoma is the most aggressive type of skin cancer with high mortality rates. Early diagnosis is crucial because it significantly improves treatment outcomes, but conventional methods relying on dermoscopy and lesion biopsy have limitations in accuracy during early stages and are invasive. Liquid biopsies offer a minimally invasive alternative, particularly for routine screening. The abundance of cancer cell-driven extracellular vesicles in interstitial fluid can be utilized for point-of-care cancer diagnostics. Here, we developed a stellate silicon microneedle patch, the ExoPatch, coated with Annexin V functionalized hydrogel to isolate melanoma-specific exosomes. The ExoPatch captures exosomes directly from the skin, followed by dissolution of the hydrogel to release the exosomes, which are then detected using a lateral flow immunoassay specific to melanoma markers (MCAM and MCSP). After validating with cell line derived extracellular vesicles and testing with mouse tissue, the ExoPatch isolated 11.5 times more protein from melanoma tissue compared to healthy tissue. Additionally, the ExoPatch effectively distinguished between melanoma and healthy tissues, with its specificity confirmed through Western Blot and electron microscopy analysis. The ExoPatch with melanoma mouse samples produced a 3.5-fold higher signal in the lateral flow immunoassay compared to that of healthy controls. The ExoPatch presents a promising point-of-care diagnostic tool for melanoma, offering significant advantages in terms of rapidness, minimal invasiveness, and ease of use. It has the potential to enhance early detection and routine monitoring in melanoma patients, ultimately improving patient outcomes by reducing the reliance on traditional, invasive biopsies.
Collapse
Affiliation(s)
- Scott M Smith
- Department of Chemical Engineering and Biomedical Engineering, Biointerfaces Institute, Translational Oncology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Abha Kumari
- Department of Chemical Engineering and Biomedical Engineering, Biointerfaces Institute, Translational Oncology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joseph P Marvar
- Department of Chemical Engineering and Biomedical Engineering, Biointerfaces Institute, Translational Oncology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nna-Emeka Onukwugha
- Department of Chemical Engineering and Biomedical Engineering, Biointerfaces Institute, Translational Oncology Program, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yoon-Tae Kang
- Department of Chemical Engineering and Biomedical Engineering, Biointerfaces Institute, Translational Oncology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Sunitha Nagrath
- Department of Chemical Engineering and Biomedical Engineering, Biointerfaces Institute, Translational Oncology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
15
|
Sonar S, Das A, Yeong Zher L, Narayanan Ravi R, Zheng Kong EQ, Dhar R, Narayanan K, Gorai S, Subramaniyan V. Exosome-Based Sensor: A Landmark of the Precision Cancer Diagnostic Era. ACS APPLIED BIO MATERIALS 2025. [PMID: 40366154 DOI: 10.1021/acsabm.5c00288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Extracellular vesicles are nanoscale vesicles released by a diversity of cells that mediate intercellular communication by transporting an array of biomolecules. They are gaining increasing attention in cancer research due to their ability to carry specific biomarkers. This characteristic makes them potentially useful for highly sensitive, noninvasive diagnostic procedures and more precise prognostic assessments. Consequently, EVs are emerging as a transformative tool in cancer treatment, facilitating early detection and personalized medicine. Despite significant progress, clinical implementation is hindered by challenges in EV isolation, purification, and characterization. However, developing advanced biosensor technologies offers promising solutions to these obstacles. This review highlights recent progress in biosensors for EV detection and analysis, focusing on various sensing modalities including optical, electrochemical, microfluidic, nanomechanical, and biological sensors. We also explore techniques for EV isolation, characterization, and analysis, such as electron microscopy, atomic force microscopy, nanoparticle tracking analysis, and single-particle analysis. Furthermore, the review critically assesses the challenges associated with EV detection and put forward future directions, aiming to usher in a cutting-edge era of precision medicine through advanced, sensor-based, noninvasive early cancer diagnosis by detecting EV-carried biomarkers.
Collapse
Affiliation(s)
- Swarup Sonar
- Department of Oncology, Neuron Institute of Applied Research, Amravati, Maharashtra 444605, India
| | - Asmit Das
- Department of Oncology, Neuron Institute of Applied Research, Amravati, Maharashtra 444605, India
| | - Lee Yeong Zher
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Ram Narayanan Ravi
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Eason Qi Zheng Kong
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Rajib Dhar
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Selangor (Darul Ehsan), Malaysia
| | - Kumaran Narayanan
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center, 1620 W Harrison Street, Chicago, Illinois 60612, United States
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Selangor (Darul Ehsan), Malaysia
| |
Collapse
|
16
|
Ajwad N, Mustapha M, Idris Z, Lee SY. The Recent Applications of Stem Cell-Derived Exosomes and Hydrogels in Neurological Disorders. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40323680 DOI: 10.1089/ten.teb.2024.0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Neurological disorders such as Alzheimer's disease, Parkinson's disease, and stroke pose significant challenges for conventional therapy due to the complexities of the blood-brain barrier (BBB) and the restricted delivery of drugs to the central nervous system. Exosomes, a type of small extracellular vesicle secreted by nearly all cell types, hold substantial promise as delivery vehicles for therapeutic agents in treating these conditions. Notably, stem cell-secreted exosomes have emerged as particularly effective due to their regenerative potential and natural ability to cross the BBB. Similarly, hydrogels have gained recognition as versatile biomaterials capable of supporting sustained release and targeted delivery of therapeutics. The combination of the regenerative properties of stem cell-derived exosomes (SC-Exos) with the structural and functional benefits of hydrogels offers a promising approach for enhancing neurogenesis, modulating neuroinflammation, and facilitating tissue repair. This review explores the origin, structure, and modifications of exosomes as well as the synthesis and incorporation methods of hydrogels in the therapeutic context for debilitating neurological disorders. It highlights recent advancements in using SC-Exos and hydrogels for therapeutic delivery, addressing both current challenges and future applications. Improving our understanding of hydrogels loaded with SC-Exos for cargo transportation and neural tissue regeneration may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Nabil Ajwad
- Regenerative Medicine Research Group, Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Zamzuri Idris
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Si-Yuen Lee
- Regenerative Medicine Research Group, Department of Hematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
17
|
Peng X, Gao Y, Liu J, Shi X, Li W, Ma Y, Li X, Li H. Mitochondria-derived vesicles: A promising and potential target for tumour therapy. Clin Transl Med 2025; 15:e70320. [PMID: 40356246 PMCID: PMC12069804 DOI: 10.1002/ctm2.70320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondria-derived vesicles (MDVs) participate in early cellular defence mechanisms initiated in response to mitochondrial damage. They maintain mitochondrial quality control (MQC) by clearing damaged mitochondrial components, thereby ensuring the normal functioning of cellular processes. This process is crucial for cell survival and health, as mitochondria are the energy factories of cells, and their damage can cause cellular dysfunction and even death. Recent studies have shown that MDVs not only maintain mitochondrial health but also have a significant impact on tumour progression. MDVs selectively encapsulate and transport damaged mitochondrial proteins under oxidative stress and reduce the adverse effects of mitochondrial damage on cells, which may promote the survival and proliferation of tumour cells. Furthermore, it has been indicated that after cells experience mild stress, the number of MDVs significantly increases within 2-6 h, whereas mitophagy, a process of clearing damaged mitochondria, occurs 12-24 h later. This suggests that MDVs play a critical role in the early stress response of cells. Moreover, MDVs also have a significant role in intercellular communication, specifically in the tumour microenvironment. They can carry and transmit various bioactive molecules, such as proteins, nucleic acids, and lipids, which regulate tumour cell's growth, invasion, and metastasis. This intercellular communication may facilitate tumour spread and metastasis, making MDVs a potential therapeutic target. Advances in MDV research have identified novel biomarkers, clarified regulatory mechanisms, and provided evidence for clinical use. These breakthroughs pave the way for novel MDV-targeted therapies, offering improved treatment alternatives for cancer patients. Further research can identify MDVs' role in tumour development and elucidate future cancer treatment horizons.
Collapse
Affiliation(s)
- Xueqiang Peng
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Group of Chronic Disease and Environmental GenomicsSchool of Public HealthChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
| | - Yu Gao
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
| | - Jiaxing Liu
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
| | - Xinxin Shi
- Department of General SurgeryThe First Hospital of Anhui University of Science & TechnologyHuainanChina
| | - Wei Li
- Department of General SurgeryThe First Hospital of Anhui University of Science & TechnologyHuainanChina
| | - Yingbo Ma
- Depatment of Hepatobiliary SurgeryAir Force Medical CenterBeijingChina
| | - Xuexin Li
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
- Division of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Hangyu Li
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
- Department of General SurgeryThe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouLiaoningChina
| |
Collapse
|
18
|
Kozela E, Petrovich-Kopitman E, Berger Y, Camacho AC, Shoham Y, Morandi MI, Rosenhek-Goldian I, Rotkopf R, Regev-Rudzki N. Spectral flow cytometry for detecting DNA cargo in malaria parasite-derived extracellular vesicles. J Biol Chem 2025; 301:108481. [PMID: 40199399 PMCID: PMC12136778 DOI: 10.1016/j.jbc.2025.108481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
Cells across biological kingdoms release extracellular vesicles (EVs) as a means of communication with other cells, be their friends or foes. This is indeed true for the intracellular malaria parasite Plasmodium falciparum (Pf), which utilizes EVs to transport bioactive molecules to various human host systems. Yet, the study of this mode of communication in malaria research is currently constrained due to limitations in high-resolution tools and the absence of commercial antibodies. Here, we demonstrate the power of an advanced spectral flow cytometry approach to robustly detect secreted EVs, isolated from Pf-infected red blood cells. By labeling both EV membrane lipids and the DNA cargo within (non-antibody staining approach), we were able to detect a subpopulation of parasitic-derived EVs enriched in DNA. Furthermore, we could quantitatively measure the DNA-carrying EVs isolated from two distinct blood stages of the parasite: rings and trophozoites. Our findings showcase the potential of spectral flow cytometry to monitor dynamic changes in nucleic acid cargo within pathogenic EVs.
Collapse
Affiliation(s)
- Ewa Kozela
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | | | - Yuval Berger
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Abel Cruz Camacho
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Yaara Shoham
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Mattia I Morandi
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Science, Prague, Czech Republic; The International Institute of Molecular Mechanisms and Machines, Polish Academy of Sciences, Warsaw, Poland
| | - Irit Rosenhek-Goldian
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
19
|
Manolopoulos A, Yao PJ, Kapogiannis D. Extracellular vesicles: translational research and applications in neurology. Nat Rev Neurol 2025; 21:265-282. [PMID: 40181198 DOI: 10.1038/s41582-025-01080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2025] [Indexed: 04/05/2025]
Abstract
Over the past few decades, extensive basic, translational and clinical research has been devoted to deciphering the physiological and pathogenic roles of extracellular vesicles (EVs) in the nervous system. The presence of brain cell-derived EVs in the blood, carrying diverse cargoes, has enabled the development of predictive, diagnostic, prognostic, disease-monitoring and treatment-response biomarkers for various neurological disorders. In this Review, we consider how EV biomarkers can bring us closer to understanding the complex pathogenesis of neurological disorders such as Alzheimer disease, Parkinson disease, stroke, traumatic brain injury, amyotrophic lateral sclerosis and multiple sclerosis. We describe how translational research on EVs might unfold bidirectionally, proceeding from basic to clinical studies but also in the opposite direction, with biomarker findings in the clinic leading to novel hypotheses that can be tested in the laboratory. We demonstrate the potential value of EVs across all stages of the therapeutic development pipeline, from identifying therapeutic targets to the use of EVs as reporters in model systems and biomarkers in clinical research. Finally, we discuss how the cargo and physicochemical properties of naturally occurring and custom-engineered EVs can be leveraged as novel treatments and vehicles for drug delivery, potentially revolutionizing neurotherapeutics.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Pamela J Yao
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
20
|
Kashkoulinejad Kouhi T. Exosome-mediated communication between T cells and dendritic cells: Implications for therapeutic strategies. Cytokine 2025; 189:156914. [PMID: 40073808 DOI: 10.1016/j.cyto.2025.156914] [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: 10/05/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Cell communication is crucial for coordinating physiological functions in multicellular organisms, with exosomes playing a significant role. Exosomes mediate intercellular communication by transporting proteins, lipids, and nucleic acids between cells. These small, membrane-bound vesicles, derived from the endosomal pathway, are integral to various biological processes, including signal transmission and cellular behavior modulation. Recent advances highlight the potential of exosomes, especially dendritic cell-derived exosomes (DEXs), for diagnostic and therapeutic applications, particularly in cancer immunotherapy. DEXs are distinguished by their ability to present antigens and stimulate immune responses more effectively than exosomes from other cell types. They carry a cargo rich in immunostimulatory molecules and MHC-peptide complexes, which facilitate robust T-cell activation and enhance tumor-specific immune responses. The unique properties of DEXs, such as their ability to cross biological barriers and resist tumor-induced immunosuppression, position them as promising candidates for therapeutic applications. Here, I review the reports on the bidirectional interaction between dendritic cells and T cells through exosomes and their role in medicine.
Collapse
Affiliation(s)
- Tahereh Kashkoulinejad Kouhi
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada; CTOAM | Cancer Treatment Options & Management, Vancouver, British Columbia, Canada.
| |
Collapse
|
21
|
Dauphin T, de Beaurepaire L, Salama A, Pruvost Q, Claire C, Haurogné K, Sourice S, Dupont A, Bach JM, Hervé J, Olmos E, Bosch S, Lieubeau B, Mosser M. Scalability of spheroid-derived small extracellular vesicles production in stirred systems. Front Bioeng Biotechnol 2025; 13:1516482. [PMID: 40365014 PMCID: PMC12069995 DOI: 10.3389/fbioe.2025.1516482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Small extracellular vesicle (sEV)-based therapies have gained widespread interest, but challenges persist to ensure standardization and high-scale production. Implementing upstream processes in a chemically defined media in stirred-tank bioreactors (STBr) is mandatory to closely control the cell environment, and to scale-up production, but it remains a significant challenge for anchorage-dependent cells. Methods We used a human β cell line, grown as monolayer or in suspension as spheroid in stirred systems. We assessed the consequences of culturing these cells in 3D with, or without fetal bovine serum in a chemically defined medium, for cell growth, viability and metabolism. We next explored how different scale-up strategies might influence cell and spheroid formation in spinner flask, with the aim to transfer the process in instrumented Ambr®250 STBr. Lastly, we analyzed and characterized sEV production in monolayer, spinner flask and STBr. Results and discussion Generation of spheroids in a chemically defined medium allowed the culture of highly viable cells in suspension in stirred systems. Spheroid size depended on the system's volumetric power input (P/V), and maintaining this parameter constant during scale-up proved to be the optimal strategy for standardizing the process. However, transferring the spinner flask (SpF) process to the Ambr®250 STBr at constant P/V modified spheroid size, due to important geometric differences and impeller design. Compared to a monolayer reference process, sEV yield decreased two-fold in SpF, but increased two-fold in STBr. Additionally, a lower expression of the CD63 tetraspanin was observed in sEV produced in both stirred systems, suggesting a reduced release of exosomes compared to ectosomes. This study addresses the main issues encountered in spheroid culture scale-up in stirred systems, rather conducive for the production of ectosomes.
Collapse
Affiliation(s)
| | | | | | | | - Clémentine Claire
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
- Oniris VetAgroBio, B-FHIT, Nantes, France
| | | | | | - Aurélien Dupont
- CNRS, INSERM, BIOSIT_UAR 3480, Univ Rennes, Inserm 018, Rennes, France
| | - Jean-Marie Bach
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
- Oniris VetAgroBio, B-FHIT, Nantes, France
| | - Julie Hervé
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
| | - Eric Olmos
- University of Lorraine, CNRS, LRGP, Nancy, France
| | | | | | - Mathilde Mosser
- Oniris VetAgroBio, INRAE, IECM, Nantes, France
- Oniris VetAgroBio, B-FHIT, Nantes, France
| |
Collapse
|
22
|
Lorico A, Santos MF, Karbanová J, Corbeil D. Extracellular membrane particles en route to the nucleus - exploring the VOR complex. Biochem Soc Trans 2025:BST20253005. [PMID: 40366329 DOI: 10.1042/bst20253005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025]
Abstract
Intercellular communication is an essential hallmark of multicellular organisms for their development and adult tissue homeostasis. Over the past two decades, attention has been focused on communication mechanisms based on various membrane structures, as illustrated by the burst of scientific literature in the field of extracellular vesicles (EVs). These lipid bilayer-bound nano- or microparticles, as vehicle-like devices, act as regulators in various biological and physiological processes. When EVs are internalized by recipient cells, their membrane and cytoplasmic cargoes can interfere with cellular activities, affecting pathways that regulate cell proliferation, differentiation, and migration. In cancer, EVs can transfer oncogenic factors, stimulate neo-angiogenesis and immunosuppression, reprogram stromal cells, and confer drug resistance traits, thereby remodeling the surrounding microenvironment. Although the mechanisms underlying EV biogenesis and uptake are now better understood, little is known about the spatiotemporal mechanism(s) of their actions after internalization. In this respect, we have shown that a fraction of endocytosed EVs reaches the nuclear compartment via the VOR (VAP-A-ORP3-Rab7) complex-mediated docking of late endosomes to the outer nuclear membrane in the nucleoplasmic reticulum, positioning and facilitating the transfer of EV cargoes into the nucleoplasm via nuclear pores. Here, we highlight the EV heterogeneity, the cellular pathways governing EV release and uptake by donor and recipient cells, respectively, and focus on a novel intracellular pathway leading to the nuclear transfer of EV cargoes. We will discuss how to intercept it, which could open up new avenues for clinical applications in which EVs and other small extracellular particles (e.g., retroviruses) are implicated.
Collapse
Affiliation(s)
- Aurelio Lorico
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV 89014, U.S.A
| | - Mark F Santos
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV 89014, U.S.A
| | - Jana Karbanová
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Saxony, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Dresden, Saxony, Germany
| | - Denis Corbeil
- Biotechnology Center (BIOTEC), Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Saxony, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Dresden, Saxony, Germany
| |
Collapse
|
23
|
Zhdanova DY, Bobkova NV, Chaplygina AV, Svirshchevskaya EV, Poltavtseva RA, Vodennikova AA, Chernyshev VS, Sukhikh GT. Effect of Small Extracellular Vesicles Produced by Mesenchymal Stem Cells on 5xFAD Mice Hippocampal Cultures. Int J Mol Sci 2025; 26:4026. [PMID: 40362265 PMCID: PMC12071690 DOI: 10.3390/ijms26094026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is one of the most common progressive neurodegenerative diseases leading to impairments in memory, orientation, and behavior. However, significant work is still needed to fully understand the progression of such disease and develop novel therapeutic agents for AD prevention and treatment. Small extracellular vesicles (sEVs) have received attention in recent years due to their potential therapeutic effects on AD. The aim of this study was to determine the potential effect of sEVs in an in vitro model of AD. sEVs were isolated from human Wharton's jelly mesenchymal stem cells (MSCs) by asymmetric depth filtration, a method developed recently by us. AD was modeled in vitro using cells obtained from the hippocampi of newborn 5xFAD transgenic mice carrying mutations involved in familial AD. After isolation, sEVs underwent detailed characterization that included scanning electron microscopy, nanoparticle tracking analysis, confocal microscopy, Western blotting, and Luminex assay. When added to 5xFAD hippocampal cells, sEVs were nontoxic, colocalized with neurons and astrocytes, decreased the level of Aβ peptide, and increased the synaptic density. These results support the possibility that sEVs can improve brain cell function during aging, decrease the risk of AD, and potentially be used for AD therapeutics.
Collapse
Affiliation(s)
- Daria Y. Zhdanova
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Natalia V. Bobkova
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Alina V. Chaplygina
- Institute of Cell Biophysics, Federal Research Center Pushchino Research Center for Biological Studies, Russian Academy of Sciences, Institutskaya 3, Pushchino, 142290 Moscow, Russia (A.V.C.)
| | - Elena V. Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ulitsa M0iklukho-Maklaya 16/10, 117997 Moscow, Russia;
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| | - Rimma A. Poltavtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| | - Anastasia A. Vodennikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ulitsa M0iklukho-Maklaya 16/10, 117997 Moscow, Russia;
- Institute of Bioorganic Chemistry, National Research Nuclear University “MEPhI”, Kashirskoe Shosse 31, 115409 Moscow, Russia
| | - Vasiliy S. Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Bld. 1, 121205 Moscow, Russia
| | - Gennadiy T. Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V. I. Kulakov, Ministry of Healthcare of the Russian Federation, Oparina St. 4, 117997 Moscow, Russia; (R.A.P.); (V.S.C.); (G.T.S.)
| |
Collapse
|
24
|
Huang Y, Li W, Liu T, Lin X, Xia Y, Zhu W, Jin H, Cai Q. Rice extracellular vesicles send defense proteins into fungus Rhizoctonia solani to reduce disease. Dev Cell 2025; 60:1168-1181.e6. [PMID: 39755117 DOI: 10.1016/j.devcel.2024.12.020] [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: 05/12/2024] [Revised: 09/18/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025]
Abstract
The exchange of molecular information across kingdoms is crucial for the survival of both plants and their pathogens. Recent research has identified that plants transfer their small RNAs and microRNAs into fungal pathogens to suppress infection. However, whether and how plants send defense proteins into pathogens remains unknown. Here, we report that rice (Oryza sativa) plants package defense proteins into extracellular vesicles (EVs) and deliver them to the fungal pathogen Rhizoctonia solani. These EVs, enriched with host defense proteins, are internalized by the fungal cells. Reducing the transfer of host defense proteins via EVs results in increased disease susceptibility. Furthermore, the overexpression of host defense proteins in either rice plants or the fungal cells reduced the infection. Therefore, plants use EVs to send defense proteins into fungal pathogens, thereby combating infection. This mechanism represents a form of protein exchange between plants and pathogens, which contributes to reducing crop diseases.
Collapse
Affiliation(s)
- Yifan Huang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China
| | - Wei Li
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China
| | - Tiangu Liu
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China
| | - Xiaoli Lin
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China
| | - Yanhui Xia
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China
| | - Wenjing Zhu
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China
| | - Hailing Jin
- Department of Microbiology and Plant Pathology and Center for Plant Cell Biology, Institute for Integrative Genome Biology, University of California, Riverside, CA 92507, USA
| | - Qiang Cai
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China; Hubei Hongshan Laboratory, Wuhan 430072, China.
| |
Collapse
|
25
|
Fernández-Pérez AG, Herrera-González A, López-Naranjo EJ, Martínez-Álvarez IA, Uribe-Rodríguez D, Ramírez-Arreola DE, Sánchez-Peña MJ, Navarro-Partida J. Extracellular Vesicles from Different Mesenchymal Stem Cell Types Exhibit Distinctive Surface Protein Profiling and Molecular Characteristics: A Comparative Analysis. Int J Mol Sci 2025; 26:3393. [PMID: 40244251 PMCID: PMC11989379 DOI: 10.3390/ijms26073393] [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: 02/19/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
The current medical need to respond to different diseases has sparked great interest in extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) due to their great regenerative potential and as drug carriers by playing a critical role in cell-cell communication. However, due to their heterogeneity, there is no standardized universal method for their identification and characterization, which limits their clinical application. This study, following the recommendations and methodologies proposed by MISEV2023 for the characterization of EVs, shows for the first time a detailed morphological, protein, and biochemical comparison between EVs derived from three different MSCs sources (placenta, endometrium, and dental pulp). The information obtained from the different applied assays suggests that there are substantial differences between one EVs source and another. It also offers valuable insights that provide the guidelines to ease their profiling and therefore improve their selection, in order to speed up their use and clinical application; additionally, the knowledge obtained from each characterization test could facilitate new researchers in the field to choose a specific cell source to obtain EVs and select the appropriate methods that provide the necessary information according to their requirements.
Collapse
Affiliation(s)
- Atziri G. Fernández-Pérez
- Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), University of Guadalajara, Guadalajara 44430, Jalisco, Mexico; (A.G.F.-P.); (A.H.-G.); (E.J.L.-N.); (M.J.S.-P.)
| | - Azucena Herrera-González
- Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), University of Guadalajara, Guadalajara 44430, Jalisco, Mexico; (A.G.F.-P.); (A.H.-G.); (E.J.L.-N.); (M.J.S.-P.)
| | - Edgar J. López-Naranjo
- Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), University of Guadalajara, Guadalajara 44430, Jalisco, Mexico; (A.G.F.-P.); (A.H.-G.); (E.J.L.-N.); (M.J.S.-P.)
| | | | - David Uribe-Rodríguez
- Centro de Biotecnología Santer S.C., Guadalajara 45040, Jalisco, Mexico; (I.A.M.-Á.); (D.U.-R.)
| | - Daniel E. Ramírez-Arreola
- Centro Universitario de la Costa Sur (CUCSUR), University of Guadalajara, Autlan 48900, Jalisco, Mexico;
| | - María Judith Sánchez-Peña
- Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), University of Guadalajara, Guadalajara 44430, Jalisco, Mexico; (A.G.F.-P.); (A.H.-G.); (E.J.L.-N.); (M.J.S.-P.)
| | - Jose Navarro-Partida
- School of Medicine and Health Sciences, Monterrey Institute of Technology, Zapopan 45201, Jalisco, Mexico
| |
Collapse
|
26
|
Sandira MI, Lim K, Yoshida T, Sajidah ES, Narimatsu S, Imakawa R, Yoshimura K, Nishide G, Qiu Y, Taoka A, Hazawa M, Ando T, Hanayama R, Wong RW. Nanoscopic Profiling of Small Extracellular Vesicles via High-Speed Atomic Force Microscopy (HS-AFM) Videography. J Extracell Vesicles 2025; 14:e270050. [PMID: 40139685 PMCID: PMC11943829 DOI: 10.1002/jev2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 03/29/2025] Open
Abstract
Small extracellular vesicles (sEVs), which carry lipids, proteins and RNAs from their parent cells, serve as biomarkers for specific cell types and biological states. These vesicles, including exosomes and microvesicles, facilitate intercellular communication by transferring cellular components between cells. Current methods, such as ultracentrifugation and Tim-4 affinity method, yield high-purity sEVs. However, despite their small size, purified sEVs remain heterogeneous due to their varied intracellular origins. In this technical note, we used high-speed atomic force microscopy (HS-AFM) in conjunction with exosome markers (IgGCD63 and IgGCD81) to explore the intracellular origins of sEVs at single-sEV resolution. Our results first revealed the nanotopology of HEK293T-derived sEVs under physiological conditions. Larger sEVs (diameter > 100 nm) exhibited greater height fluctuations compared to smaller sEVs (diameter ≤ 100 nm). Next, we found that mouse-origin IgGCD63, and rabbit-origin IgGcontrol and IgGCD81, exhibited the iconic 'Y' conformation, and similar structural dynamics properties. Last, exosome marker antibodies predominantly co-localised with sEVd ≤ 100 nm but not with sEVd > 100 nm, demonstrating the CD63-CD81-enriched sEV and CD63-CD81-depleted sEV subpopulations. In summary, we demonstrate that nanoscopic profiling of surface exosome markers on sEVs using HS-AFM is feasible for characterising distinct sEV subpopulations in a heterogeneous sEV mixture.
Collapse
Affiliation(s)
- Muhammad Isman Sandira
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Keesiang Lim
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Takeshi Yoshida
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Department of ImmunologyGraduate School of Medical SciencesKanazawa UniversityKanazawaIshikawaJapan
| | | | - Shinnosuke Narimatsu
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Reon Imakawa
- The School of Biological Science and TechnologyCollege of Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Kota Yoshimura
- The School of Biological Science and TechnologyCollege of Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Goro Nishide
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeWISE Program for Nano‐Precision Medicine, Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Yujia Qiu
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Azuma Taoka
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Masaharu Hazawa
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Toshio Ando
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Rikinari Hanayama
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Department of ImmunologyGraduate School of Medical SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Richard W. Wong
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeWISE Program for Nano‐Precision Medicine, Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
27
|
Feng G, Lan X, Qin S, Shi Y, Zhao Q, Li Q, Zhong L. Advances in Research on Exosomal miRNAs in Central Nervous System Diseases. ASN Neuro 2025; 17:2465546. [PMID: 40165664 DOI: 10.1080/17590914.2025.2465546] [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: 10/29/2024] [Revised: 01/03/2025] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Neurological diseases present a wide range of conditions, intricate diagnosis and treatment processes, and complex prognostic considerations. Therefore, research focusing on the diagnosis and treatment of these diseases is crucial. Exosomal miRNAs are small RNA molecules enclosed in membrane vesicles, released by cells and known to play roles in the development of various neurological disorders. They also serve as specific biomarkers for these conditions. Drawing on extensive research on exosomal miRNAs in diseases like stroke, Alzheimer's, epilepsy, Parkinson's, and neuroregeneration, this paper provides a comprehensive review of the relationship between exosomal miRNAs and neurological diseases. We strive to offer current and detailed theoretical understandings to help with the diagnosis and treatment of these disorders.
Collapse
Affiliation(s)
- Guangli Feng
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaoqian Lan
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shiyi Qin
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuting Shi
- Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qinxi Zhao
- The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Kunming Medical University, Kunming, Yunnan, China
| | - Lianmei Zhong
- Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Farm YR, Chuah BH, Law JX, Leong XF, Razali M, Ng SL. Therapeutic Potential of Extracellular Vesicles in Oral Inflammation. Int J Mol Sci 2025; 26:3031. [PMID: 40243684 PMCID: PMC11988662 DOI: 10.3390/ijms26073031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/18/2025] [Accepted: 03/22/2025] [Indexed: 04/18/2025] Open
Abstract
The therapeutic potential of extracellular vesicles (EVs) in reducing oral inflammation is thoroughly examined in this review, with an emphasis on gingivitis, periodontitis, and oral mucositis. It explains the complex relationship between microbial dysbiosis and host immune responses in the aetiology of oral inflammation. Pathophysiological mechanisms of periodontitis are examined, emphasising the roles played by periodontal pathogens and inflammatory mediators in the disease's chronic course and systemic effects. Preclinical research is providing new evidence that EVs originating from various cellular sources control immune cell dynamics towards a pro-healing phenotype, promote tissue regeneration, and have immunomodulatory qualities. EV-based therapies appear to be a promising new therapeutic technique with potential benefits over traditional methods for the treatment of oral inflammatory illnesses by specifically altering inflammatory signalling pathways. This review highlights the potential of EVs to improve patient outcomes in oral health and emphasises the need for additional clinical research to clarify the therapeutic efficacy and underlying mechanisms of EVs in periodontal therapy.
Collapse
Affiliation(s)
- Yan Rou Farm
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Bing Huan Chuah
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Xin Fang Leong
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| | - Masfueh Razali
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Sook Luan Ng
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (Y.R.F.); (B.H.C.); (X.F.L.)
| |
Collapse
|
29
|
Yeat NY, Liu LH, Chang YH, Lai CPK, Chen RH. Bro1 proteins determine tumor immune evasion and metastasis by controlling secretion or degradation of multivesicular bodies. Dev Cell 2025:S1534-5807(25)00155-8. [PMID: 40185104 DOI: 10.1016/j.devcel.2025.03.008] [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: 06/12/2024] [Revised: 11/25/2024] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Exosomes play pleiotropic tumor-promoting functions and are secreted by fusion of multivesicular bodies (MVBs) with the plasma membrane. However, MVBs are also directed to lysosomes for degradation, and the mechanism controlling different fates of MVBs remains elusive. Here, we show that the pro-tumor protein WDR4 enhances exosome secretion from mouse and human cancer cells through degrading the endosomal sorting complex required for transport (ESCRT)-associated Bro1-family protein PTPN23. Mechanistically, PTPN23 and ALIX compete for binding to syntenin, thereby directing MVBs toward degradation and secretion, respectively. ALIX, but not PTPN23, recruits actin-capping proteins CAPZA1/CAPZB to prevent branched filamentous actin (F-actin) accumulation around MVBs, thus enabling MVBs trafficking to the cell periphery for secretion. Functionally, WDR4/ALIX-dependent exosomes load a set of pro-tumor proteins through LAMP2A, thereby potentiating metastasis and immune evasion in mice. Our study highlights a previously unappreciated coupling between the biogenesis mechanism and the fate decision of MVBs and its importance in determining exosomal cargos, which have a profound impact on tumor progression.
Collapse
Affiliation(s)
- Nai Yang Yeat
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Li-Heng Liu
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yu-Hsuan Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | | | - Ruey-Hwa Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan; Chemical Biology and Molecular Biophysics Program, Taiwan International Graduate Program, Academia Sinica, Taipei 115, Taiwan; Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
30
|
Cortés-Hernández LE, Eslami-S Z, Attina A, Batista S, Cayrefourcq L, Vialeret J, Di Vizio D, Hirtz C, Costa-Silva B, Alix-Panabières C. Proteomic profiling and functional analysis of extracellular vesicles from metastasis-competent circulating tumor cells in colon cancer. J Exp Clin Cancer Res 2025; 44:102. [PMID: 40119417 PMCID: PMC11929255 DOI: 10.1186/s13046-025-03360-4] [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: 01/23/2025] [Accepted: 03/09/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are pivotal in cancer progression, and in vitro CTC models are crucial for understanding their biological mechanisms. This study focused on the characterization of extracellular vesicles (EVs) from CTC lines derived from a patient with metastatic colorectal cancer (mCRC) at different stages of progression who progressed despite therapy (thus mirroring the clonal evolution of cancer). METHODS AND RESULTS Morphological and size analyses revealed variations among EVs derived from different CTC lines. Compared with the Vesiclepedia database, proteomic profiling of these EVs revealed enrichment of proteins related to stemness, endosomal biogenesis, and mCRC prognosis. Integrin family proteins were significantly enriched in EVs from CTC lines derived after therapy failure. The role of these EVs in cancer progression was analyzed by assessing their in vivo distribution, particularly in the liver, lungs, kidneys, and bones. EVs accumulate significantly in the liver, followed by the lungs, kidneys and femurs. CONCLUSIONS This study is a pioneering effort in highlighting therapy progression-associated changes in EVs from mCRC patients via an in vitro CTC model. The results offer insights into the role of metastasis initiator CTC-derived EVs in cancer spread, suggesting their utility for studying cancer tissue distribution mechanisms. However, these findings must be confirmed and extended to patients with mCRC. This work underscores the potential of CTC-derived EVs as tools for understanding cancer dissemination.
Collapse
Affiliation(s)
- Luis Enrique Cortés-Hernández
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France.
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Aurore Attina
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Silvia Batista
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Jérôme Vialeret
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Dolores Di Vizio
- Department of Urology, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christophe Hirtz
- IRMB-PPC, INM, Univ Montpellier, CHU Montpellier, INSERM CNRS, Montpellier, France
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC, MIVEGEC, University of Montpellier, CNRS, IRD, Montpellier, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
| |
Collapse
|
31
|
Wu YY, Yu LSY, Zhou HY, Xue JC. Effect of HepG2-Derived Exosome with PDGF-D Knockdown on Transformation of Normal Fibroblasts into Tumor-Associated Fibroblasts in Liver Cancer. FRONT BIOSCI-LANDMRK 2025; 30:26045. [PMID: 40152369 DOI: 10.31083/fbl26045] [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: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND It is known that the transformation of liver cancer-mediated fibroblasts into cancer-related fibroblasts (CAFs) is beneficial to the development of liver cancer. However, the specific mechanism is still unclear. METHODS Human hepatocarcinoma (HepG2) cells were treated with short hairpin RNA (shRNA) of platelet-derived growth factor-D (shPDGF-D) vector, and the exosomes secreted by the cells were separated using ultracentrifugation and identified by using nanoparticle tracking analysis, transmission electron microscope, and western blot analysis. Exosomes were co-cultured with mouse primary fibroblasts, and then the activity, proliferation, cell cycle, migration, epithelial-mesenchymal transition- (EMT-) and CAF marker-related protein expression levels of fibroblasts were determined by cell counting kit-8 (CCK-8), immunofluorescence, flow cytometry, wound healing, real-time reverse transcription-PCR, and western blotting assays, respectively. Co-cultured fibroblasts were mixed with HepG2 cells and injected subcutaneously into mice to construct animal models. The size and weight of xenograft tumor and the expression of epithelial-mesenchymal transition- (EMT-), angiogenesis- and CAFs marker-related proteins were detected. RESULTS The exosomes inhibited the proliferation, migration, EMT, and induced cell cycle arrest, as well as decreased the expression of α-SMA, FAP, MMP-9, and VEGF in fibroblasts. In vivo, sh-PDGF-D inhibited tumor growth, reduced the expressions of CD31, vimentin, α-SMA, FAP, MMP9, and VEGF, and promoted the expression of E-cadherin. CONCLUSIONS Exosomes derived from HepG2 cells transfected with shPDGF-D prevent normal fibroblasts from transforming into CAFs, thus inhibiting angiogenesis and EMT of liver cancer.
Collapse
Affiliation(s)
- Yan-Yan Wu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Liu-Shen-Yan Yu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Han-Yu Zhou
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| | - Jun-Chao Xue
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Karbowniczek M, Kalvala A, Silwal A, Patel B, Kasetti A, Shetty K, Cho JH, Lara G, Daugherity B, Diesler R, Pooladanda V, Rueda B, Henske E, Yu J, Markiewski M. Extracellular vesicles modulate integrin signaling and subcellular energetics to promote pulmonary lymphangioleiomyomatosis metastasis. RESEARCH SQUARE 2025:rs.3.rs-5390547. [PMID: 40166013 PMCID: PMC11957204 DOI: 10.21203/rs.3.rs-5390547/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pulmonary lymphangioleiomyomatosis (LAM) is metastatic sarcoma but mechanisms of LAM metastasis are unknown. Extracellular vesicles (EV) regulate cancer metastasis but their roles in LAM have not yet been thoroughly investigated. Here, we report the discovery of distinct LAM-EV subtypes derived from primary tumor or metastasizing LAM cells that promote LAM metastasis through ITGα6/β1-c-Src-FAK signaling, triggered by shuttling ATP synthesis to cell pseudopodia or the activation of integrin adhesion complex, respectively. This signaling leads to increased LAM cell migration, invasiveness, and stemness and regulates metastable (hybrid) phenotypes that are all pivotal for metastasis. Mouse models corroborate in vitro data by demonstrating a significant increase in metastatic burden upon the exposure to EV through distinct mechanisms involving either lung resident fibroblasts or metalloproteinases' activation that are EV subtype dependent. The clinical relevance of these findings is underscored by increased EV biogenies in LAM patients and the enrichment of these EV cargo with lung tropic integrins and metalloproteinases. These findings establish EV as novel therapeutic target in LAM, warranting the future clinical studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gerard Lara
- Texas Tech University Health Sciences Center
| | | | - Remi Diesler
- Brigham and Women's Hospital and Harvard Medical School
| | | | | | | | - Jane Yu
- University of Cincinnati College of Medicine
| | | |
Collapse
|
33
|
Vafadar A, Younesi M, Babadi S, Alizadeh M, Movahedpour A, Savardashtaki A. Exosome biosensors for detection of liver cancer. Clin Chim Acta 2025; 570:120199. [PMID: 39961411 DOI: 10.1016/j.cca.2025.120199] [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: 11/09/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Liver cancer is a significant global health concern due to its poor prognosis, often resulting from late-stage diagnosis and limited treatment options. While non-invasive methods such as ultrasound, blood tests (like AFP and PIVKA-II), CT scans, and MRIs are commonly employed in liver cancer diagnosis, they can occasionally be limited in sensitivity or associated with high costs. This has heightened the demand for innovative, non-invasive biomarkers that enable early and accurate diagnosis, leading to increased interest in the potential of exosomes. Exosomes are small vesicles released by cells and have the potential to serve as biomarkers for liver cancer. They contain a variety of biomolecules, including nucleic acids, proteins, and lipids, which can offer important information about cell health and disease progression. Developing fast, accurate, sensitive, and reliable techniques for detecting exosomes is essential. Biosensors, analytical tools for biological samples, have emerged as powerful instruments for analyzing exosomes. This review focuses on recent advancements in biosensor technology for exosome detection and explores future perspectives. The goal is to promote the development of innovative biosensor-based methods for detecting exosomes to enable earlier diagnosis and better clinical management of liver cancer.
Collapse
Affiliation(s)
- Asma Vafadar
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Younesi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sepideh Babadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Alizadeh
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Movahedpour
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
34
|
Ribas-Maynou J, Parra A, Martínez-Díaz P, Rubio CP, Lucas X, Yeste M, Roca J, Barranco I. Protective role of extracellular vesicles against oxidative DNA damage. Biol Res 2025; 58:14. [PMID: 40075425 PMCID: PMC11905505 DOI: 10.1186/s40659-025-00595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Oxidative stress, a source of genotoxic damage, is often the underlying mechanism in many functional cell disorders. Extracellular vesicles (EVs) have been shown to be key regulators of cellular processes and may be involved in maintaining cellular redox balance. Herein, we aimed to develop a method to assess the effects of EVs on DNA oxidation using porcine seminal plasma extracellular vesicles (sEVs). RESULTS The technique was set using a cell-free plasmid DNA to avoid the bias generated by the uptake of sEVs by sperm cells, employing increasing concentrations of hydrogen peroxide (H2O2) that generate DNA single-strand breaks (SSBs). Because SSBs contain a free 3'-OH end that allow the extension through quantitative PCR, such extension -and therefore the SYBR intensity- showed to be proportional to the amount of SSB. In the next stage, H2O2 was co-incubated with two size-differentiated subpopulations (small and large) of permeabilized and non-permeabilized sEVs to assess whether the intravesicular content (IC) or the surface of sEVs protects the DNA from oxidative damage. Results obtained showed that the surface of small sEVs reduced the incidence of DNA SSBs (P < 0.05), whereas that of large sEVs had no impact on the generation of SSBs (P > 0.05). The IC showed no protective effect against DNA oxidation (P > 0.05). CONCLUSIONS Our results suggest that the surface of small sEVs, including the peripheral corona layer, may exert a protective function against alterations that are originated by oxidative mechanisms. In addition, our in vitro study opens path to detect, localize and quantify the protective effects against oxidation of extracellular vesicles derived from different fluids, elucidating their function in physiopathological states.
Collapse
Affiliation(s)
- Jordi Ribas-Maynou
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
- Unit of Cell Biology and Medical Genetics; Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Ana Parra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Pablo Martínez-Díaz
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Camila Peres Rubio
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
| | - Xiomara Lucas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (Technosperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain.
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain.
| | - Isabel Barranco
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| |
Collapse
|
35
|
Hirosawa KM, Sato Y, Kasai RS, Yamaguchi E, Komura N, Ando H, Hoshino A, Yokota Y, Suzuki KGN. Uptake of small extracellular vesicles by recipient cells is facilitated by paracrine adhesion signaling. Nat Commun 2025; 16:2419. [PMID: 40075063 PMCID: PMC11903687 DOI: 10.1038/s41467-025-57617-9] [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: 04/16/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Small extracellular vesicles (sEVs) play crucial roles in intercellular communication. However, the internalization of individual sEVs by recipient cells has not been directly observed. Here, we examined these mechanisms using state-of-the-art imaging techniques. Single-molecule imaging shows that tumor-derived sEVs can be classified into several subtypes. Simultaneous single-sEV particle tracking and observation of super-resolution movies of membrane invaginations in living cells reveal that all sEV subtypes are internalized via clathrin-independent endocytosis mediated by galectin-3 and lysosome-associated membrane protein-2C, while some subtypes that recruited raft markers are internalized through caveolae. Integrin β1 and talin-1 accumulate in recipient cell plasma membranes beneath all sEV subtypes. Paracrine, but not autocrine, sEV binding triggers Ca2+ mobilization induced by the activation of Src family kinases and phospholipase Cγ. Subsequent Ca2+-induced activation of calcineurin-dynamin promotes sEV internalization, leading to the recycling pathway. Thus, we clarified the detailed mechanisms of sEV internalization driven by paracrine adhesion signaling.
Collapse
Affiliation(s)
- Koichiro M Hirosawa
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Yusuke Sato
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, 980-8578, Japan
| | - Rinshi S Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, 104-0045, Japan
| | - Eriko Yamaguchi
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Hiromune Ando
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan
- Innovation Research Center for Quantum Medicine. Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan
| | - Ayuko Hoshino
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, 153-8904, Japan
- Inamori Research Institute for Science, Inamori Foundation, Kyoto, 600-8411, Japan
| | - Yasunari Yokota
- Department of Electrical, Electronics and Computer Engineering, Faculty of Engineering, Gifu University, Gifu, 501-1193, Japan
| | - Kenichi G N Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan.
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, 104-0045, Japan.
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan.
- Innovation Research Center for Quantum Medicine. Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
36
|
Tricoli L, Sase S, Hacker JL, Pham V, Chappell M, Breda L, Hurwitz SN, Tanaka N, Castruccio Castracani C, Guerra A, Hou Z, Schlotawa L, Radhakrishnan K, Hogenauer M, Roche A, Everett J, Bushman F, Kurre P, Ahrens-Nicklas R, Adang LA, Vanderver AL, Rivella S. Effective gene therapy for metachromatic leukodystrophy achieved with minimal lentiviral genomic integrations. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102464. [PMID: 40171445 PMCID: PMC11960508 DOI: 10.1016/j.omtn.2025.102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025]
Abstract
Metachromatic leukodystrophy (MLD) is a fatal lysosomal storage disease characterized by the deficient enzymatic activity of arylsulfatase A (ARSA). Combined autologous hematopoietic stem cell transplantion (HSCT) with lentiviral (LV)-based gene therapy has great potential to treat MLD. Achieving the optimal balance between high enzyme production for therapeutic efficacy and maintaining a low vector copy number (VCN) is crucial. Insufficient enzyme levels can lead to the progression of motor symptoms, undermining treatment goals. Conversely, elevated VCN increases the risk of genotoxicity, which poses safety concerns, and contributes to higher production costs, making the therapy less accessible. Striking this balance is essential to maximize clinical benefit while minimizing risks and costs. To address this need, we increased the expression of ARSA cDNA at single integration by generating novel LVs, optimizing ARSA expression and enhancing safety. In addition, our vectors achieved optimal transduction in mouse and human hematopoietic stem cells (HSCs) with minimal multiplicity of infection (MOI). Our top-performing vector (EA1) showed at least 4× more ARSA activity than the currently US and European Union (EU)-approved vector and a superior ability to secrete vesicle-associated ARSA, a critical modality to transfer functional enzymes from microglia to oligodendrocytes. Three-month-old Arsa-knockout (KO) MLD mice transplanted with Arsa-KO bone marrow (BM) cells transduced with 0.6 VCN of EA1 demonstrated behavior and CNS histology matching wild-type (WT) mice. Our novel vector boosts efficacy while improving safety as a robust approach for treating MLD patients.
Collapse
Affiliation(s)
- Lucas Tricoli
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sunetra Sase
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julia L. Hacker
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Vi Pham
- Department of Pediatrics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology affinity group (CAMB), University of Pennsylvania, Philadelphia, PA, USA
| | - Maxwell Chappell
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laura Breda
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephanie N. Hurwitz
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Comprehensive Bone Marrow Failure Center, CHOP, Philadelphia, PA, USA
| | - Naoto Tanaka
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Carlo Castruccio Castracani
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amaliris Guerra
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zhongqi Hou
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lars Schlotawa
- University Medical Centre Göttingen, Göttingen, Germany
- Fraunhofer Institute for Translational Medicine – Translational Neuroinflammation and Automated Microscopy, Göttingen, Germany
| | | | - Matthew Hogenauer
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aoife Roche
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Kurre
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Comprehensive Bone Marrow Failure Center, CHOP, Philadelphia, PA, USA
| | - Rebecca Ahrens-Nicklas
- Department of Pediatrics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A. Adang
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adeline L. Vanderver
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- RNA Institute, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics-CHOP, Philadelphia, PA, USA
| |
Collapse
|
37
|
Wu Q, Wu Q, Lin H, Zhang C, You Z, Kang S, Xu Y, Chen X, Yang C, Song Y, Zhu L. Microfluidic Replication and Phenotypic Profiling of Extracellular Vesicles from the Tumor Microenvironment Using Dual-Switch Aptamer Logic Gates. Anal Chem 2025; 97:5313-5323. [PMID: 40012368 DOI: 10.1021/acs.analchem.5c00234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The phenotypic profiling of extracellular vesicles (EVs) within the tumor microenvironment (TME) provides critical insights into the intercellular communication mechanisms of EVs underlying tumor physiology. However, conventional methods typically isolate EVs from the extracellular space through tissue fragmentation, which compromises tissue viability, and neglects the spatial organization of the tissue and the dynamic nature of EV secretion. Herein, we introduce an innovative microfluidic platform to cultivate intact tumor tissues while preserving their spatial architecture and facilitating natural EV secretion. This system enables the direct replication of EVs onto the chip for high-fidelity phenotypic analysis. Utilizing a combinatorial-aptamer-induced dual-switch logic gate methodology, this approach allows for the precise subtyping of EVs derived from both tumor cells and immune cells within the TME. Specifically, aptamers targeting EpCAM and PD-L1, along with the connector probe, were employed to induce a dual-switch signal to identify distinct EV populations. This strategy enables noninvasive, real-time capture and phenotypic profiling of EVs directly within the microfluidic environment. Furthermore, our findings indicate that immunotherapy with PD-1 antibodies significantly enhances the secretion of EVs by immune cells within the TME, underscoring the potential role of EVs as mediators of therapeutic responses. Overall, we have developed a robust, noninvasive method for the phenotypic profiling of EVs in the TME, offering a powerful tool for investigating the biological functions and implications of EVs in tumor pathophysiology.
Collapse
Affiliation(s)
- Qiaoyi Wu
- Department of Trauma Center and Emergency Surgery, The First Affiliated Hospital of Fujian Medical University, Department of Trauma Center & Emergency Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P. R. China
| | - Qiuyue Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Haoting Lin
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Chi Zhang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Zhenlong You
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Siyin Kang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Yuanfeng Xu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Xiaofeng Chen
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, P. R. China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P. R. China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
| | - Lin Zhu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, P. R. China
- School of Environmental Science and Engineering, Hainan University, Haikou 570228, P. R. China
| |
Collapse
|
38
|
Zhang G, Ma C, Ma L, Wei D, Wu Y, Li Y, Xu Z, Liu Y, Cai Y, Yu EY, Zhu Y, Zhang H. High-Efficiency Capture and Proteomic Analysis of Plasma-Derived Extracellular Vesicles through Affinity Purification. Anal Chem 2025; 97:4889-4897. [PMID: 39908429 DOI: 10.1021/acs.analchem.4c04269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
Abstract
Plasma-derived extracellular vesicles (EVs) are promising sources of biomarkers. It is still a challenge to isolate EVs from a small amount of human plasma for downstream proteomic analysis. The isolation process is hindered by contamination with high-abundance blood proteins and lipoprotein particles, which adversely impact proteomic analyses. Moreover, although EV immune-isolation via magnetic beads often integrates with flow sorting and Western blotting (WB), it lacks compatibility with nanoparticle tracking analysis (NTA) and proteomic analysis. To address these issues, we have developed a functional affinity magnetic bead, EVlent (Extracellular Vesicles isoLated Efficiently, Naturally, and Totally), enabling the rapid and efficient isolation of EVs from plasma. By optimizing the quantities of magnetic beads and plasma used, we characterized the isolated EVs through WB, NTA, and transmission electron microscopy (TEM), showing the successful isolation of EVs from plasma. Proteomic analysis of these EVs identified over 2000 proteins and 15,000 peptides from 100 μL of plasma and nearly 1000 proteins from trace samples as small as 5 μL. Additionally, this isolation method significantly reduced contaminants, including plasma proteins and lipoproteins, compared to ultracentrifugation. Finally, we applied this strategy to plasma samples of healthy individuals and those with Parkinson's disease, identifying four potential biomarkers that provide promising guidance for clinical diagnosis.
Collapse
Affiliation(s)
- Guiyuan Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- EVLiXiR Biotech, Nanjing 210032, China
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Chengxiao Ma
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Le Ma
- Shanghai JINCE Clinical Laboratories, Shanghai 201101, China
| | - Dong Wei
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Yanan Wu
- Laboratory of Medical Genetics, Harbin Medical University, Harbin 150081, China
| | - Ying Li
- Center of Clinical Laboratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhehui Xu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Yufeng Liu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Yuhan Cai
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Evan Yiwen Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Epidemiology & Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China
- Department of Epidemiology, CAPHRI Care and Public Health Research Institute, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6229ER, The Netherlands
| | - Yefei Zhu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Laboratory Medicine Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- Department of Laboratory Medicine, Jianhu People's Hospital, Yanchen 224700, China
| | - Hao Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- EVLiXiR Biotech, Nanjing 210032, China
| |
Collapse
|
39
|
Singh M, Tiwari PK, Kashyap V, Kumar S. Proteomics of Extracellular Vesicles: Recent Updates, Challenges and Limitations. Proteomes 2025; 13:12. [PMID: 40137841 PMCID: PMC11944546 DOI: 10.3390/proteomes13010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid-bound vesicles secreted by cells, including exosomes, microvesicles, and apoptotic bodies. Proteomic analyses of EVs, particularly in relation to cancer, reveal specific biomarkers crucial for diagnosis and therapy. However, isolation techniques such as ultracentrifugation, size-exclusion chromatography, and ultrafiltration face challenges regarding purity, contamination, and yield. Contamination from other proteins complicates downstream processing, leading to difficulties in identifying biomarkers and interpreting results. Future research will focus on refining EV characterization for diagnostic and therapeutic applications, improving proteomics tools for greater accuracy, and exploring the use of EVs in drug delivery and regenerative medicine. In this review, we provide a bird's eye view of various challenges, starting with EV isolation methods, yield, purity, and limitations in the proteome analysis of EVs for identifying protein targets.
Collapse
Affiliation(s)
- Mohini Singh
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida UP-201310, India
| | - Prashant Kumar Tiwari
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida UP-201310, India
| | - Vivek Kashyap
- Division of Cancer Immunology and Microbiology, Medicine and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Sanjay Kumar
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida UP-201310, India
- Division of Nephrology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
40
|
Borger A, Haertinger M, Millesi F, Semmler L, Supper P, Stadlmayr S, Rad A, Radtke C. Conditioning period impacts the morphology and proliferative effect of extracellular vesicles derived from rat adipose tissue derived stromal cell. J Nanobiotechnology 2025; 23:164. [PMID: 40033315 PMCID: PMC11877948 DOI: 10.1186/s12951-025-03273-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025] Open
Abstract
A serum-free conditioning period is a crucial step during small extracellular vesicle (sEV) preparation ranging from 12 to 72h. There is a paucity of knowledge about downstream effects of serum-free conditioning on sEVs and the optimal duration of the conditioning period. The aim of this study was to investigate the influence of the serum-free conditioning period on the sEVs derived from primary adipose stromal cells (AdSCs) and their regenerative potential. Primary AdSCs were conditioned in serum-free medium for 72h. Conditioned medium was collected and refreshed every 24h obtaining three fractions, namely sEVs released after 24h (early), 24h to 48h (intermediate) and 48h to 72h (late). After sEV enrichment with ultracentrifugation, the sEV fractions were analyzed by their size, phenotypic expression, and morphology. Proliferation assays of primary Schwann cells after treatment with sEVs were performed. Particles meeting criteria to be classified as sEVs were detected in all fractions. However, sEVs differed by their size and phenotypic expression. A long conditioning period led to a heterogenous population of larger sEVs and increased protein per particle ratio. Moreover, the expression of tetraspanines was affected. Lastly, the proliferative effect of sEVs on Schwann cells decreased with increasing conditioning period. In conclusion, particles meeting the criteria of EVs are released by primary AdSCs over 72h under serum free conditioning. Nonetheless, they significantly differ in their proliferative effect on Schwann cells cultures.
Collapse
Affiliation(s)
- Anton Borger
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Maximilian Haertinger
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Flavia Millesi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Lorenz Semmler
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Paul Supper
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sarah Stadlmayr
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anda Rad
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
41
|
Zhan Y, Zhou Z, Zhu Z, Zhang L, Yu S, Liu Y, Zhang X. Exosome-transmitted LUCAT1 promotes stemness transformation and chemoresistance in bladder cancer by binding to IGF2BP2. J Exp Clin Cancer Res 2025; 44:80. [PMID: 40025525 PMCID: PMC11874664 DOI: 10.1186/s13046-025-03330-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
The chemotherapy resistance is an awkward challenge in management of bladder cancer (BC). Cancer organoid model is an effective preclinical tumor model that could faithfully represent clinical manifestations and simulate the biological processes of chemoresistance. Recent studies have revealed that cancer stem cells (CSCs) play a significant role in the development of chemoresistance in cancer. Exosomes act as essential intercellular messengers and participate in controlling the conversion of distinct cell characteristics, including chemoresistance. However, the role of exosome-transmitted lncRNAs in bladder cancer chemoresistance has rarely been reported. In this study, cancer organoid models were developed from urothelial carcinomas to explore the pathophysiology mechanism of BC chemoresistance, and RNA-seq was performed to screen for lncRNAs involved in chemoresistance of BC. We found chemotherapy enriches stem-like cells in BC, and significant upregulation of Lung Cancer Associated Transcript 1 (LUCAT1) occurs in chemotherapy-resistant organoids and correlated with chemotherapy response. Further experimental results demonstrated that LUCAT1 promotes chemoresistance in bladder cancer by enhancing the stemness phenotype of BC cells in vivo and in vitro. Moreover, exosomes derived from bladder cancer stem cells can enhance the stemness phenotype and chemoresistance of BC cells by delivering LUCAT1. Mechanistically, LUCAT1 could significantly enhance the mRNA stability of HMGA1 via binding to IGF2BP2 in an m6A-dependent manner. The study demonstrates an important role for exosome-transmitted LUCAT1 in chemoresistance and LUCAT1 has the potential to function as both a diagnostic biomarker and therapeutic target for BC.
Collapse
Affiliation(s)
- Yonghao Zhan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| | - Zhenzhen Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Zhaowei Zhu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Lianghao Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Shuanbao Yu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Yuchen Liu
- Shenzhen Institute of Translational Medicine, Health Science Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China.
| | - Xuepei Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China.
| |
Collapse
|
42
|
Njoku GC, Forkan CP, Soltysik FM, Nejsum PL, Pociot F, Yarani R. Unleashing the potential of extracellular vesicles for ulcerative colitis and Crohn's disease therapy. Bioact Mater 2025; 45:41-57. [PMID: 39610953 PMCID: PMC11602541 DOI: 10.1016/j.bioactmat.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Image 1.
Collapse
Affiliation(s)
- George Chigozie Njoku
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, USA
| | - Cathal Patrick Forkan
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Pharmacy, Université Grenoble Alpes, France
| | - Fumie Mitani Soltysik
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Peter Lindberg Nejsum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| |
Collapse
|
43
|
Colao IL, Corteling RL, Bracewell DG, Wall IB. Neural stem cell-derived extracellular vesicles purified by monolith chromatography retain stimulatory effect in in vitro scratch assay. Cytotherapy 2025; 27:365-377. [PMID: 39755977 DOI: 10.1016/j.jcyt.2024.11.007] [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: 03/11/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND AIMS Extracellular vesicles (EVs) have gained traction as potential cell-free therapeutic candidates. Development of purification methods that are scalable and robust is a major focus of EV research. Yet there is still little in the literature that evaluates purification methods against potency of the EV product. In the present study, we examined two monolith chromatography methods with a focus on assessing the ability of purified EVs to retain stimulatory effects on fibroblasts to connect scalable purification methods with product outputs. METHODS We characterized EVs recovered from CTX0E03 (CTX) neural stem cell-conditioned medium in terms of biomarker distribution, functional capacity and purity. We evaluated the ability of EVs to promote wound closure in an in vitro scratch assay prior to and following two monolith chromatography steps (anion exchange and hydrophobic interaction) to determine whether these options may better serve EV bioprocessing. RESULTS EVs from CTX cells were successful in initiating wound repair in a fibroblast scratch assay over 72 h with a single 20-μg dose. EV preparations presented the markers CD9, CD81 and CD63 but also contained culture albumin and DNA as process impurities. EVs recovered by tangential flow filtration could be successfully purified further by both monolith chromatography steps. Post-monolith EV stimulation was conserved. CONCLUSIONS The results indicate that monolith chromatography is a viable purification method for EVs derived from cell culture that does not detract from the product's ability to stimulate fibroblasts, suggesting that product functionality is conserved. Further work is needed in developing suitable downstream processes and analytics to achieve clinically relevant purities for injectable biologics.
Collapse
Affiliation(s)
- Ivano Luigi Colao
- Department of Biochemical Engineering, University College London, London, UK
| | | | - Daniel G Bracewell
- Department of Biochemical Engineering, University College London, London, UK.
| | - Ivan B Wall
- Institute of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
44
|
Xu L, Xiong J, Li X, Wang J, Wang P, Wu X, Wang J, Liu Y, Guo R, Fan X, Zhu X, Guan Y. Role of Lactobacillus plantarum-Derived Extracellular Vesicles in Regulating Alcohol Consumption. Mol Neurobiol 2025; 62:2889-2902. [PMID: 39180695 DOI: 10.1007/s12035-024-04447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Alcohol Use Disorder (AUD), characterized by repeated alcohol consumption and withdrawal symptoms, poses a significant public health issue. Alcohol-induced impairment of the intestinal barrier results in alterations in intestinal permeability and the composition of the intestinal microbiota. Such alterations lead to a reduced relative abundance of intestinal lactic acid bacteria. However, the role of gut microbiota in alcohol consumption is not yet fully understood. In this study, we explore the mechanism by which gut microbiota regulates alcohol consumption, specifically using extracellular vesicles derived from Lactobacillus plantarum (L-EVs). L-EVs were administered to Sprague-Dawley rats either through intraperitoneal injection or microinjection into the ventral tegmental area (VTA), resulting in a significant reduction in alcohol consumption 72 hours after withdrawal. The observed reduction was akin to the effect of an intra-VTA microinjection of Brain-Derived Neurotrophic Factor (BDNF). Intriguingly, the microinjection of K252a (a Trk B antagonist) into the VTA blocked the reducing effect of L-EVs on alcohol consumption. The intraperitoneal injection of L-EVs restored the diminished BDNF expression in the VTA of alcohol-dependent rats. Furthermore, L-EVs rescued the low BDNF expression in alcohol-incubated PC12 cells. In conclusion, our study demonstrates that L-EVs attenuated alcohol consumption by enhancing BDNF expression in alcohol-dependent rats, thus suggesting the significant therapeutic potential of L-EVs in preventing excessive alcohol consumption.
Collapse
Affiliation(s)
- Lulu Xu
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Junwei Xiong
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xinxin Li
- Heilongjiang Province Key Laboratory of Mechanism and Prevention of Substance Dependence Disease, Mudanjiang, 157011, China
| | - Jiajia Wang
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Pengyu Wang
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xiaobin Wu
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jiaxi Wang
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yong Liu
- Heilongjiang Province Key Laboratory of Mechanism and Prevention of Substance Dependence Disease, Mudanjiang, 157011, China
| | - Ran Guo
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xiaohe Fan
- Heilongjiang Province Key Laboratory of Mechanism and Prevention of Substance Dependence Disease, Mudanjiang, 157011, China
| | - Xiaofeng Zhu
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China.
- Heilongjiang Province Key Laboratory of Mechanism and Prevention of Substance Dependence Disease, Mudanjiang, 157011, China.
- Development and Application of North Traditional Chinese Medicine Collaborative Innovation Center in Mudanjiang, Mudanjiang, 157011, China.
| | - Yanzhong Guan
- Department of Physiology & Neurobiology, Mudanjiang Medical University, Mudanjiang, 157011, China.
- Heilongjiang Province Key Laboratory of Mechanism and Prevention of Substance Dependence Disease, Mudanjiang, 157011, China.
- Development and Application of North Traditional Chinese Medicine Collaborative Innovation Center in Mudanjiang, Mudanjiang, 157011, China.
| |
Collapse
|
45
|
Kostyusheva A, Romano E, Yan N, Lopus M, Zamyatnin AA, Parodi A. Breaking barriers in targeted Therapy: Advancing exosome Isolation, Engineering, and imaging. Adv Drug Deliv Rev 2025; 218:115522. [PMID: 39855273 DOI: 10.1016/j.addr.2025.115522] [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: 07/30/2024] [Revised: 12/23/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
Exosomes have emerged as promising tools for targeted drug delivery in biomedical applications and medicine. This review delves into the scientific advancements, challenges, and future prospects specifically associated with these technologies. In this work, we trace the research milestones that led to the discovery and characterization of exosomes and extracellular vesicles, and discuss strategies for optimizing the synthetic yield and the loading of these particles with various therapeutics. In addition, we report the current major issues affecting the field and hampering the clinical translation of these technologies. Highlighting the pivotal role of imaging techniques, we explore how they drive exosome therapy and development by offering insights into biodistribution and cellular trafficking dynamics. Methodologies for vesicle isolation, characterization, loading, and delivery mechanisms are thoroughly examined, alongside strategies aimed at enhancing their therapeutic efficacy. Special emphasis was dedicated to their therapeutic properties, particularly to their ability to deliver biologics into the cytoplasm. Furthermore, we delve into the intricate balance between surface modifications and targeting properties including also transgenic methods aimed at their functionalization and visualization within biological systems. This review underscores the transformative potential of these carriers in targeted drug delivery and identifies crucial areas for further research and clinical translation.
Collapse
Affiliation(s)
- Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia
| | | | - Neng Yan
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai Kalina Campus, Vidyanagari, Mumbai 400098, India
| | - Andrey A Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Department of Biological Chemistry, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia
| | - Alessandro Parodi
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia.
| |
Collapse
|
46
|
Liu X, To KK, Zeng Q, Fu L. Effect of Extracellular Vesicles Derived From Tumor Cells on Immune Evasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417357. [PMID: 39899680 PMCID: PMC11948033 DOI: 10.1002/advs.202417357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Indexed: 02/05/2025]
Abstract
The crosstalk between immunity and cancer in the regulation of tumor growth is considered a hallmark of cancer. Antitumor immunity refers to the innate and adaptive immune responses that regulate cancer development and proliferation. Tumor immune evasion represents a major hindrance to effective anticancer treatment. Extracellular vesicles (EVs) are nano-sized and lipid-bilayer-enclosed particles that are secreted to the extracellular space by all cell types. They are critically involved in numerous biological functions including intercellular communication. Tumor-derived extracellular vesicles (TEVs) can transport a variety of cargo to modulate immune cells in the tumor microenvironment (TME). This review provides the latest update about how tumor cells evade immune surveillance by exploiting TEVs. First, the biogenesis of EVs and the cargo-sorting machinery are discussed. Second, how tumor cells modulate immune cell differentiation, activation, and function via TEVs to evade immune surveillance is illustrated. Last but not least, the novel antitumor strategies that can reverse immune escape are summarized.
Collapse
Affiliation(s)
- Xuanfan Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
| | - Kenneth K.W. To
- School of PharmacyThe Chinese University of Hong KongHong Kong999077P. R. China
| | - Qinsong Zeng
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080P. R. China
- Guangxi Hospital Division of The First Affiliated HospitalSun Yat‐sen UniversityNanning530025P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerCollaborative Innovation Center for Cancer MedicineGuangdong Esophageal Cancer InstituteSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
47
|
Norman M, Shami‐shah A, D'Amaddio SC, Travis BG, Ter‐Ovanesyan D, Dougan TJ, Walt DR. Toward Identification of Markers for Brain-Derived Extracellular Vesicles in Cerebrospinal Fluid: A Large-Scale, Unbiased Analysis Using Proximity Extension Assays. J Extracell Vesicles 2025; 14:e70052. [PMID: 40098346 PMCID: PMC11913887 DOI: 10.1002/jev2.70052] [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: 04/13/2024] [Revised: 12/27/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Extracellular vesicles (EVs) captured in biofluids have opened a new frontier for liquid biopsies. To enrich for vesicles coming from a particular cell type or tumour, scientists utilize antibodies to transmembrane proteins that are relatively unique to the cell type of interest. However, recent evidence has called into question the basic assumption that all transmembrane proteins measured in biofluids are, in fact, EV-associated. To identify both candidate markers for brain-derived EV immunocapture and cargo proteins to validate the EVs' cell of origin, we conducted an unbiased Olink screen, measuring 5416 unique proteins in cerebrospinal fluid after size exclusion chromatography. We identified proteins that demonstrated a clear EV fractionation pattern and created a searchable dataset of candidate EV-associated markers-both proteins that are cell type-specific within the brain, and proteins found across multiple cell types for use as general EV markers. We further implemented the DeepTMHMM deep learning model to differentiate predicted cytosolic, transmembrane, and external proteins and found that intriguingly, only 10% of the predicted transmembrane proteins have a clear EV fractionation pattern based on our stringent criteria. This dataset further bolsters the critical importance of verifying EV association of candidate proteins using methods such as size exclusion chromatography before downstream use of the targets for EV analysis.
Collapse
Affiliation(s)
- Maia Norman
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
- Department of Pathology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of PsychiatryMassachusetts General HospitalBostonMassachusettsUSA
| | - Adnan Shami‐shah
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
- Department of Pathology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Sydney C. D'Amaddio
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
- Department of Pathology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Benjamin G. Travis
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
- Department of Pathology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Tyler J. Dougan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
- Department of Pathology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Harvard‐MIT Program in Health Sciences and TechnologyCambridgeMassachusettsUSA
| | - David R. Walt
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
- Department of Pathology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
48
|
Luther K, Navaei A, Gens L, Semple C, Moharil P, Passalacqua I, Vyas K, Wang Q, Liu SL, Sun L, Ramaswamy S, Zocco D, Nabhan JF. Scalable production and purification of engineered ARRDC1-mediated microvesicles in a HEK293 suspension cell system. Sci Rep 2025; 15:7299. [PMID: 40025043 PMCID: PMC11873033 DOI: 10.1038/s41598-025-87674-5] [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: 08/11/2024] [Accepted: 01/21/2025] [Indexed: 03/04/2025] Open
Abstract
Engineering of human ARRDC1-mediated microvesicles (ARMMs) as non-viral vehicles for delivery of gene therapies bears the potential to enable novel therapeutic paradigms. We evaluated two scalable strategies to generate ARMMs loaded with protein cargo, by transient transfection or stable cell line-based production. The upstream ARMMs production processes utilized a suspension-adapted HEK293-derived line, termed 5B8. 5B8 cells yielded robust production of ARMMs after transient transfection with the ARMMs loading construct or using a stable cell line containing a transgene that encodes the ARMMs loading cassette, in shake flasks or a stirred tank bioreactor, respectively. ARMMs were purified by ultracentrifugation (small scale) or a combination of TFF and AEX (scalable production). Both purification methods produced comparable ARMMs, in terms of size and payload incorporation. Single particle analysis showed approximately 50% were payload-containing ARMMs. Additionally, an in vivo study was conducted in mice to investigate the half-life and biodistribution of ARMMs administered intravenously. ARMMs showed rapid biodistribution predominantly to the spleen and liver and, to a lesser extent, kidneys, and lungs. The half-life of ARMMs in plasma was 6 ± 0.4 min. Altogether, this work advances knowledge on scale-up of engineered cell-derived vesicles for future in vivo delivery of therapeutic molecules.
Collapse
Affiliation(s)
- Kristin Luther
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA.
| | - Ali Navaei
- Lonza Cell & Gene Technologies, Lonza Walkersville Inc., Walkersville, MD, 21793, USA
| | - Leah Gens
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | - Carson Semple
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | - Pearl Moharil
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | | | - Komal Vyas
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | - Qiyu Wang
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | - Shu-Lin Liu
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | - Lucy Sun
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA
| | - Senthil Ramaswamy
- Lonza Cell & Gene Technologies, Lonza Walkersville Inc., Walkersville, MD, 21793, USA
| | - Davide Zocco
- Lonza Siena, Strada del Petriccio e Belriguardo 35, 53100, Siena, Italy
| | - Joseph F Nabhan
- Vesigen Therapeutics, 790 Memorial Drive, Cambridge, MA, 02139, USA.
| |
Collapse
|
49
|
Duke LC, Cone AS, Sun L, Dittmer DP, Meckes DG, Tomko RJ. Tetraspanin CD9 alters cellular trafficking and endocytosis of tetraspanin CD63, affecting CD63 packaging into small extracellular vesicles. J Biol Chem 2025; 301:108255. [PMID: 39909378 PMCID: PMC11919600 DOI: 10.1016/j.jbc.2025.108255] [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: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Small extracellular vesicles (sEVs) are particles secreted from cells that play vital roles both in normal physiology and in human disease. sEVs are highly enriched in tetraspanin proteins, such as CD9 and CD63, and contain tetraspanin-enriched membrane microdomains involved in loading of sEVs with macromolecule cargoes and in sEV biogenesis. However, the precise roles of individual tetraspanins in sEV biogenesis and cargo loading remain poorly understood. Here, we report that CD9 negatively regulated CD63 trafficking to tetraspanin-enriched microdomains and its subsequent packaging into sEVs, whereas CD63 had no discernable effect on CD9 localization or packaging. Using super resolution microscopy of individual vesicles, we showed that CD9 governs the fraction of sEVs that are loaded with CD63. Interestingly, CD9-dependent suppression of CD63 packaging was rescued by pharmacological blockade of endocytosis. Together, our data support a model where CD9 contributes to the regulation and secretion of CD63 in an endocytosis-dependent manner to reprogram the contents of sEVs and tetraspanin-enriched microdomains.
Collapse
Affiliation(s)
- Leanne C Duke
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA.
| | - Allaura S Cone
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Robert J Tomko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
50
|
Kostas JC, Brainard CS, Cristea IM. A Primer on Proteomic Characterization of Intercellular Communication in a Virus Microenvironment. Mol Cell Proteomics 2025; 24:100913. [PMID: 39862905 PMCID: PMC11889360 DOI: 10.1016/j.mcpro.2025.100913] [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: 11/17/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Intercellular communication is fundamental to multicellular life and a core determinant of outcomes during viral infection, where the common goals of virus and host for persistence and replication are generally at odds. Hosts rely on encoded innate and adaptive immune responses to detect and clear viral pathogens, while viruses can exploit or disrupt these pathways and other intercellular communication processes to enhance their spread and promote pathogenesis. While virus-induced signaling can result in systemic changes to the host, striking alterations are observed within the cellular microenvironment directly surrounding a site of infection, termed the virus microenvironment (VME). Mechanisms employed by viruses to condition their VMEs are emerging and are critical for understanding the biology and pathologies of viral infections. Recent advances in experimental approaches, including proteomic methods, have enabled study of the VME in unprecedented detail. In this review article, we provide a primer on proteomic approaches used to study how viral infections alter intercellular communication, highlighting the ways in which these approaches have been implemented and the exciting biology they have uncovered. First, we consider the different molecules secreted by an infected cell, including proteins, either soluble or contained within extracellular vesicles, and metabolites. We further discuss the modalities of interactions facilitated by alteration at the cell surface of infected cells, including immunopeptide presentation and interactions with the extracellular matrix. Finally, we review spatial profiling approaches that have allowed distinguishing how specific subpopulations of cells within a VME respond to infection and alter their protein composition, discussing valuable insights these methods have offered.
Collapse
Affiliation(s)
- James C Kostas
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Colter S Brainard
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA.
| |
Collapse
|