1
|
Cao LM, Qiu YZ, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Wu Q, Bu LL. Extracellular Vesicles: Hermes between cancers and lymph nodes. Cancer Lett 2025; 623:217735. [PMID: 40268131 DOI: 10.1016/j.canlet.2025.217735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Cancer is one of the main causes of death and a major obstacle to increasing life expectancy in all countries of the world. Lymph node metastasis (LNM) of in cancer patients indicates poor prognosis and it is an important indication to determine the therapeutic regime. Therefore, more attention should be given to the molecular mechanics of tumor lymphangiogenesis and LNM. Extracellular vesicles (EVs) are nanoscale cargo-bearing membrane vesicles that can serve as key mediators for the intercellular communication. Like Hermes, the messenger of the Greek gods, EVs can be secreted by tumor cells to regulate the LNM process. Many evidence has proved the clinical correlation between EVs and LNM in various cancer types. EVs plays an active role in the process of metastasis by expressing its connotative molecules, including proteins, nucleic acids, and metabolites. However, the clear role of EVs in the process of cancer LNM has not been thoroughly studied yet. In this review, we will summarize the clinical and mechanical findings of EVs regulating role on cancer LNM, and discuss the advanced modification of the research proposal. We propose the "PUMP" principle of EVs in LNM, including Preparation, Unleash, Migration, and Planting.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yu-Zhong Qiu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Han-Yue Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
2
|
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
|
3
|
Gu X, Fan Z, Lu L, Xu H, He L, Shen H, Huang R, Li Z. Machine learning-assisted washing-free detection of extracellular vesicles by target recycling amplification based fluorescent aptasensor for accurate diagnosis of gastric cancer. Talanta 2025; 287:127506. [PMID: 39837204 DOI: 10.1016/j.talanta.2024.127506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/23/2025]
Abstract
Extracellular vesicles (EVs) are promising non-invasive biomarkers for cancer diagnosis. EVs proteins play a critical role in tumor progress and metastasis. However, accurately and reliably diagnosing cancers is greatly limited by single protein marker on EVs. Here, we reported an accurate diagnosis model of gastric cancer by analyzing five types of EVs surface proteins using machine learning in a retrospective study design. A washing-free detection method based on aptasensor and exonuclease Ⅰ was used to profile EVs surface proteins. The aptamer was designed as hairpin structure. The presence of target protein positive EVs converted the conformation of hairpin probes, which subsequently degraded by exonuclease Ⅰ. The exposed target protein could bind with and then open new hairpin probes, thus forming an amplification cycle. The lengths of different detection probes were optimized for detection. With the combination of five target proteins, five machine learning algorithms were compared to achieve a higher diagnostic accuracy. The best model, XGBoost, validated with 20 % of detection results could reach an accuracy of 0.8421. Furthermore, the XGBoost-based surface protein analysis could precisely identify gastric cancer patients with the area under the curve value of 0.9347 (95 % confidential interval (CI) = 0.8590 to 1.000). Since our method utilized a simple and versatile design of detection probes, its diagnostic scope could potentially be expanded to include different protein markers and accurately diagnose other diseases in the future.
Collapse
Affiliation(s)
- Xinrui Gu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Zeyu Fan
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China
| | - Luying Lu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Hongpan Xu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Lei He
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China.
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, 211816, China.
| | - Zhiyang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu Province, 210008, China
| |
Collapse
|
4
|
Yoon H, Jo J, Hyun H, Lee G, Ma S, Sohn J, Sung DK, Han CY, Kim M, Hwang D, Lee H, Shin Y, Oh KT, Lim C. Extracellular vesicle as therapeutic agents in anti-aging: Mechanistic insights and future potential. J Control Release 2025:113796. [PMID: 40348131 DOI: 10.1016/j.jconrel.2025.113796] [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: 02/14/2025] [Revised: 04/14/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
Aging is a multifaceted biological process marked by a gradual decline in physiological functions, driven by cellular senescence, oxidative stress, chronic inflammation, and stem cell exhaustion. Extracellular vesicles (EVs), naturally occurring nanoscale vesicles secreted by various cell types, have gained attention as potential therapeutic agents due to their ability to mediate intercellular communication by delivering bioactive molecules, including proteins, lipids, and RNAs. This review provides a comprehensive overview of EV biogenesis, cargo composition, and their mechanistic roles in counteracting aging processes. EVs from diverse sources-such as mesenchymal stem cells, embryonic stem cells, dermal fibroblasts, and colostrum-exhibit regenerative properties by modulating immune responses, enhancing tissue repair, and promoting extracellular matrix homeostasis. Recent preclinical and clinical studies further highlight their potential in addressing age-related diseases and skin rejuvenation. However, significant challenges remain, including standardization of EV production, large-scale manufacturing, safety profiling, and regulatory approval. By leveraging advancements in EV engineering, targeted delivery systems, and combination strategies with existing anti-aging interventions, EV-based therapies hold promise as next-generation approaches in regenerative medicine and longevity enhancement.
Collapse
Affiliation(s)
- Hyejoo Yoon
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Junyeong Jo
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Hyesun Hyun
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gyuwon Lee
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Seoyoung Ma
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Jungho Sohn
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| | - Dong Kyung Sung
- CHA Advanced Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Chae Young Han
- CHA Advanced Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Minkyung Kim
- CHA Advanced Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea
| | - Duhyeong Hwang
- College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, Republic of Korea
| | - Hyunji Lee
- College of Pharmacy, Kyungsung University, Busan 48434, Republic of Korea
| | - Yuseon Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Kyung Taek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| | - Chaemin Lim
- College of Pharmacy, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; CHA Advanced Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
5
|
Su Y, He W, Zheng L, Fan X, Hu TY. Toward Clarity in Single Extracellular Vesicle Research: Defining the Field and Correcting Missteps. ACS NANO 2025; 19:16193-16203. [PMID: 40271998 PMCID: PMC12060644 DOI: 10.1021/acsnano.5c00705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/25/2025]
Abstract
Single extracellular vesicle (EV) research holds the potential to revolutionize our understanding of cellular communication and enable breakthroughs in diagnostics and therapeutics. However, the lack of a clear, consensus-driven definition of single EV research has led to methodological inconsistencies, overgeneralized interpretations, and, in some cases, misleading claims. In this perspective, we propose a framework for defining single EV research, critique current challenges and misconceptions in this field, and discuss its implications for biomedical applications. We argue that precise experimental design, rigorous validation, and interdisciplinary collaboration approaches are needed to establish single EV research as a cornerstone of precision medicine.
Collapse
Affiliation(s)
- Yun Su
- Department
of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and
Ocular Oncology, Shanghai Ninth People’s
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Wanzhuo He
- Department
of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and
Ocular Oncology, Shanghai Ninth People’s
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Lei Zheng
- Department
of Laboratory Medicine, Guangdong Provincial Key Laboratory of Precision
Medical Diagnostics, Guangdong Engineering and Technology Research
Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory
of Single Cell Technology and Application, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xianqun Fan
- Department
of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and
Ocular Oncology, Shanghai Ninth People’s
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P. R. China
| | - Tony Y. Hu
- Department
of Biochemistry and Molecular Biology, Center for Cellular and Molecular
Diagnostics, Tulane University School of
Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
6
|
Cai S, Wei X, Li Q, Jiang Z, Li L. Smart materials in pharmacological drug development: Neutrophils and its constituents for drug delivery and consequent antitumor effects. Mol Immunol 2025; 183:18-32. [PMID: 40318595 DOI: 10.1016/j.molimm.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/17/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Neutrophil-based drug delivery systems for targeted therapy of cancer have been studied widely in the recent past. Chemotactic cytokines including colony-stimulating factors (CSFs) recruit various immune cells including the neutrophils to the tumor microenvironment (TME) leading to enhanced metastasis. These cytokines can be targeted effectively by immunotherapeutic agents such as checkpoint inhibitors and mAbs that can lead to systemic toxicity. To minimize the systemic adverse effects, camouflaged nanoparticles can be used significantly as alternative therapeutic agents. The neutrophil-interacting NPs and neutrophil membrane coated NPs have been exploited recently for their antitumor properties in vitro and pose limited systemic adverse effects in vivo. Neutrophil-derived exosomes derived from immune cells can efficiently escape immune-surveillance and pass through the blood-brain barrier. They possess several intrinsic properties in drug delivery as they are nano-sized, extremely biocompatible, non-immunogenic, biodegradable, stable and can carry targeting agents with limited toxicity and display antitumor properties. Also, neutrophil-based nanotherapy is dependent on factors such as neutrophil kinetics and the physicochemical properties of NPs such as size, shape, and surface chemistry. Therefore, neutrophil-based drug delivery for cancer therapy via the use of polymer nanoparticles is widely studied as their clinical appliance in nanomedicine is still at its infancy.
Collapse
Affiliation(s)
- Shengjie Cai
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Xuehan Wei
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China
| | - Qian Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China; Department of Oncology, Ganyu Hospital of Traditional Chinese Medicine, Lianyungang, Jiangsu 222000, China
| | - Ziyu Jiang
- Department of Oncology, Lianyungang Integrated Traditional Chinese and Western Medicine Clinical College, Nanjing University of Chinese Medicine, Nanjing 222002, China; Department of Oncology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, China.
| | - Lingchang Li
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210028, China; Department of Oncology, Jiangsu Integrated Traditional Chinese and Western Medicine Hospital, Nanjing 210028, China.
| |
Collapse
|
7
|
Meng L, Pan Y, Yonezawa R, Yang K, Bailey-Kobayashi N, Hashimoto N, Maeyama K, Yoshitake K, Kinoshita S, Yoshida T, Nagai K, Watabe S, Asakawa S. Identification and comparison of exosomal and non-exosomal microRNAs in mantle tissue of Pinctada fucata (Akoya pearl oyster). Int J Biol Macromol 2025; 309:142991. [PMID: 40210052 DOI: 10.1016/j.ijbiomac.2025.142991] [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: 11/05/2024] [Revised: 03/25/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
MicroRNAs (miRNA) are a class of endogenous non-coding small RNA molecules that are widely found in tissues, biological fluids, and vesicles such as exosomes. Exosomes are extracellular vesicles released from multivesicular bodies of various cell types. They are involved in intercellular communication and transport and immune regulation and may serve as potential biomarkers for diagnosis and monitoring. The function of exosomal miRNAs and their potential applications as biomarkers are a topic of interest. However, identification and comparison of miRNA expression in different biological sample types have rarely been studied. Therefore, in this study, the miRNA profiles of tissue- and tissue-derived exosomes of Pinctada fucata were characterized and compared to screen for differentially expressed miRNAs. The miRNAs functioned within tissues and were also packaged into exosomes. Simultaneously, some miRNAs were preferentially exported to exosomes for their biological functions. Functional analyses suggested that the predicted genes targeted by these differentially expressed miRNAs were extensively involved in intracellular vesicle trafficking and vesicle-mediated substrate transport. Overall, our findings provide insights into the roles of tissue-derived miRNAs and circulating exosomal miRNAs in cell communication and gene regulation. Moreover, this study serves as an additional reference for sample type selection for P. fucata small RNA analysis.
Collapse
Affiliation(s)
- Lingxin Meng
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | - Yida Pan
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | - Ryo Yonezawa
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan; Signal Peptidome Research Laboratory, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | - Kaiqiao Yang
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | | | - Naoki Hashimoto
- Pearl Research Institute, MIKIMOTO & CO., LTD., Osaki Hazako 923, Hamajima, Shima, Mie 517-0403, Japan
| | - Kaoru Maeyama
- Mikimoto Pharmaceutical CO., LTD., Kurose 1425, Ise, Mie 516-8581, Japan
| | - Kazutoshi Yoshitake
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | - Shigeharu Kinoshita
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | - Tetsuhiko Yoshida
- Institute for Advanced Sciences, TOAGOSEI CO., LTD., Tsukuba, Ibaraki 300-2611, Japan
| | - Kiyohito Nagai
- Pearl Research Institute, MIKIMOTO & CO., LTD., Osaki Hazako 923, Hamajima, Shima, Mie 517-0403, Japan
| | - Shugo Watabe
- School of Marine Biosciences, Kitasato University, Minami-ku, Sagamihara, Kanagawa 252-0313, Japan
| | - Shuichi Asakawa
- Laboratory of Aquatic Molecular Biology and Biotechnology, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Science, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan; Signal Peptidome Research Laboratory, Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan.
| |
Collapse
|
8
|
Tiszbein K, Koss-Mikołajczyk I, Martysiak-Żurowska D. Unlocking the Secrets of Human Milk: Isolation and Characterization of Extracellular Vesicles. Adv Nutr 2025; 16:100430. [PMID: 40288493 DOI: 10.1016/j.advnut.2025.100430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Extracellular vesicles from human milk (HMEVs) are crucial for neonatal development, immune modulation, and protection against pathogens. However, the lack of standardized isolation and characterization protocols poses significant challenges. This review aims to evaluate and compare various methods for the isolation and characterization of HMEVs, highlighting their effectiveness and potential applications. Preliminary purification steps, including the removal of cells, fat globules, and casein micelles, enhance the purity of isolated HMEVs. We categorized isolation methods into density-based, size-based, and affinity-based techniques. Density-based methods include differential and density gradient ultracentrifugation. Size-based methods encompass polymer precipitation, membrane filtration, electrophoretic filtration, size exclusion chromatography, and microfluidics. Affinity-based methods involve immunoisolation using antibodies specific to HMEV surface proteins. Characterization techniques discussed include flow cytometry, dynamic light scattering, nanoparticle tracking analysis, tunable resistive pulse sensing, electron microscopy, atomic force microscopy, confocal microscopy, western blotting, ELISA, and lateral flow immunoassay systems. Differential ultracentrifugation, considered the "gold standard," provides high purity but is time-consuming. Density gradient ultracentrifugation offers precise separation. Size-based methods like polyethylene glycol precipitation and membrane filtration are simple and fast. Electrophoretic filtration and microfluidics provide precise control of sample flow. Affinity-based methods are highly specific but costly. Advanced characterization techniques provide comprehensive insights into HMEV properties and functions. Standardizing isolation protocols and employing advanced characterization techniques are essential for advancing HMEV research. Future studies should focus on understanding the molecular mechanisms of HMEVs, exploring the impact of maternal health, and developing targeted delivery technologies. These efforts will enhance the therapeutic potential of HMEVs in neonatal care and contribute to personalized nutritional interventions.
Collapse
|
9
|
Chen M, Feng X, Liu C, Huang Y, Su L, Li X, Zhu J. Diagnostic value of exosome-derived lncRNA PITPNA-AS1 in lung cancer. Front Immunol 2025; 16:1539557. [PMID: 40342419 PMCID: PMC12058797 DOI: 10.3389/fimmu.2025.1539557] [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: 12/04/2024] [Accepted: 03/25/2025] [Indexed: 05/11/2025] Open
Abstract
Background Lung cancer is one of the most lethal types of cancer, and effective diagnostic biomarkers are required. There is increasing evidences that exosome-secreted lncRNAs could play an important role in lung cancer diagnosis. However, the diagnostic value and molecular mechanism of the key lncRNA PITPNA-AS1 in lung cancer remain unclear. Methods qRT-PCR was conducted to determine the levels of exosomal lncRNA PITPNA-AS1 in pleural effusions from lung adenocarcinoma, squamous cell lung carcinoma, and small cell lung cancer patients. Receiver operating characteristic (ROC) curve analyses were used to evaluate the diagnostic accuracy of PITPNA-AS1. Its role in lung cancer development was determined by a series of experiments, including CCK-8, flow cytometry, and transwell assays. RNA pull-down and RNA immunoprecipitation assays were carried out to examine the interaction between PITPNA-AS1 and Fragile X messenger ribonucleoprotein 1 (FMR1). Results We discovered PITPNA-AS1 in exosomes from lung cancer patients. Its expression was significantly increased in lung cancer patients compared to non-cancer patients, and it was strongly associated with tumor stage, lymph node metastasis, and distant metastasis in all lung cancer subtypes assessed (all p<0.05). ROC curve analyses demonstrated that exosomal PITPNA-AS1 had a high accuracy for differentiating among lung cancer subtypes. Furthermore, PITPNA-AS1 boosted H1299 and A549 cell proliferation, migration, and invasion. Mechanistically, via direct interaction, PITPNA-AS1 increased FMR1 stability by preventing its ubiquitination. Conclusions These results reveal that exosome-derived lncRNA PITPNA-AS1 acts as an oncogene to promote malignant biological behaviors and is a promising diagnostic biomarker in lung cancer.
Collapse
Affiliation(s)
- Mujin Chen
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - XiaoHui Feng
- Department of Oncology, Loujiang New City Hospital of Taicang (Ruijin Hospital, Shanghai Jiao Tong University School of Medicine), Suzhou, China
| | - ChengChen Liu
- Department of Gastroenterology, WuWei City The Second People’s Hospital, Gansu, China
| | - Yan Huang
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - LiJuan Su
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - XiaoFeng Li
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| | - JinFeng Zhu
- Department of Oncology, First Hospital of Quanzhou Affiliated to Fujian Medical University, Fujian, Quanzhou, China
| |
Collapse
|
10
|
Lee H, Lee J, Ko M, Lee KN, Kim Y, Seo B, Lee J, Jeong S, Heo K, Lee YK, Jung I, Do YR. Advanced Exosome Isolation through Electrophoretic Oscillation-Assisted Tangent-Flow Ultrafiltration with a PVDF-Fiber-Coated SiN x Nanofilter. ACS APPLIED BIO MATERIALS 2025; 8:2965-2976. [PMID: 40063836 DOI: 10.1021/acsabm.4c01821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
This study introduces a comprehensive approach to enhancing SiNx nanofilters for exosome isolation from bovine milk using the electrophoretic oscillation-assisted tangent-flow ultrafiltration (EPOTF) process. Reinforcing the nanofilter with electro-spun poly(vinylidene fluoride) (PVDF) fibers significantly improved durability under high-pressure conditions, withstanding nearly 2.8 times greater pressures than nonreinforced nanofilters. The PVDF-fiber-coated nanofilters achieved a flow rate of over 70 mL min-1, compared to just 25 mL min-1 for nonreinforced nanofilters. A filter housing system with copper electrodes isolated from the solution flow path further enhanced the electrical stability of the entire system, widening the EPO voltage range while reducing the risk of corrosion and contamination. The PVDF-fiber-coated nanofilter with the electrode in a separated housing efficiently prevented clogging and bioparticle agglomeration, maintaining constant filtration performance across various voltages and duty cycles. Biochemical analyses confirmed the high concentration and structural integrity of exosomes isolated at high flow rates. Long-term tests verified the superior performance of PVDF-coated filters, successfully filtering 3400 mL of milk over 24 h. These results demonstrate the potential of these advances for highly efficient exosome isolation while maintaining the integrity and shape of exosomes, offering promise for the future of exosome isolation research.
Collapse
Affiliation(s)
- Hansol Lee
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Jaehyuk Lee
- Department of Mechanical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea
- Advanced Institutes of Convergence Technology 8F, R&D Center, Metapore Co., Ltd., Suwon 16229, Republic of Korea
| | - Minji Ko
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Keyong Nam Lee
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Yeonjae Kim
- R&D Center, Incospharm Corp., Daejeon 34000, Republic of Korea
| | - Bosung Seo
- Advanced Institutes of Convergence Technology 8F, R&D Center, Metapore Co., Ltd., Suwon 16229, Republic of Korea
| | - Jungwon Lee
- Advanced Institutes of Convergence Technology 8F, R&D Center, Metapore Co., Ltd., Suwon 16229, Republic of Korea
| | - Sekyoo Jeong
- R&D Center, Incospharm Corp., Daejeon 34000, Republic of Korea
| | - Kyun Heo
- Department of Biopharmaceutical Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| | - Young Kwang Lee
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California 92182, United States
| | - Inhwa Jung
- Department of Mechanical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Young Rag Do
- Department of Chemistry, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
11
|
Kranc W, Kaczmarek M, Kowalska K, Pieńkowski W, Ciesiółka S, Konwerska A, Mozdziak P, Brązert M, Jeseta M, Spaczyński RZ, Pawelczyk L, Kempisty B. Morphological characteristics, extracellular vesicle structure and stem-like specificity of human follicular fluid cell subpopulation during osteodifferentiation. Exp Mol Pathol 2025; 142:104965. [PMID: 40253818 DOI: 10.1016/j.yexmp.2025.104965] [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: 08/29/2024] [Revised: 03/28/2025] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
Extracellular vesicles can play an important role in the processes occurring after stem cell transplantation, preventing cell apoptosis, stimulating immunological processes, and promoting the synthesis of extracellular matrix. Human follicular fluid (FF) can be a source of a subpopulation of cells with mesenchymal stem cells (MSCs) properties. Moreover these subpopulations of FF cells can differentiate into osteoblasts. In presented studies flow cytometry of ovarian FF cells confirmed positive expression of MSCs markers such as: CD44, CD90, CD105, CD73 and negative expression of a hematopoietic marker: CD45. The CD90+, CD105+, CD45- cell subpopulation has been obtained during magnetic separation using appropriate antibodies conjugated with microbeads. The extracellular vesicles (EVs) secreted by the cells during osteodifferentiation process differed from those secreted by cells culture in the basal medium. Based on the previous and current electron microscopy research, changes in size, number, and shape would support the notion that released EVs could be crucial to the ovarian FF cell subpopulation differentiation process. Osteogenic differentiation has been confirmed via Alizarin red staining. Therefore, follicular fluid (FF) can be a new source of a cell subpopulation with MSC properties, with the cells capable of differentiating into the osteogenic lineage. EVs could play a key role as mediators in tissue regeneration, especially bone tissue regeneration.
Collapse
Affiliation(s)
- Wiesława Kranc
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland.
| | - Mariusz Kaczmarek
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, 15 Garbary St., 61-866 Poznań, Poland; Department of Cancer Immunology, Poznan University of Medical Sciences, 5 Garbary St., 61-866 Poznań, Poland.
| | - Katarzyna Kowalska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Swiecickiego St., 60-781 Poznan, Poland.
| | - Wojciech Pieńkowski
- Division of Perinatology and Women's Diseases, Poznan University of Medical Sciences, 33 Polna St. 60-535 Poznan, Poland.
| | - Sylwia Ciesiółka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Swiecickiego St., 60-781 Poznan, Poland.
| | - Aneta Konwerska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Swiecickiego St., 60-781 Poznan, Poland.
| | - Paul Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, NC 27695, USA; Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA.
| | - Maciej Brązert
- Department of Diagnostic and Treatment of Infertility, Department of Gynecological Endocrinology and Infertility Treatment Karol Marcinkowski University, Poznan University of Medical Sciences, 33 Polna St., 60-535 Poznan, Poland.
| | - Michal Jeseta
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 62500 Brno, Czechia.
| | - Robert Z Spaczyński
- Center for Gynecology, Obstetrics and Infertility Treatment Pastelova, Pastelowa 8, 60-198, Poznan, Poland..
| | - Leszek Pawelczyk
- Department of Diagnostic and Treatment of Infertility, Department of Gynecological Endocrinology and Infertility Treatment Karol Marcinkowski University, Poznan University of Medical Sciences, 33 Polna St., 60-535 Poznan, Poland.
| | - Bartosz Kempisty
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA; Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 62500 Brno, Czechia; Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Chalubinskiego 6a, 50-368 Wroclaw, Poland; Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 1 Lwowska St., 87-100 Torun, Poland.
| |
Collapse
|
12
|
Wang Y, Yuan S, Zhou L, Yang K, Jin Z, Lin A, Yang C, Tian W. Cutting-Edge Progress in the Acquisition, Modification and Therapeutic Applications of Exosomes for Drug Delivery. Int J Nanomedicine 2025; 20:5059-5080. [PMID: 40271148 PMCID: PMC12015628 DOI: 10.2147/ijn.s516840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
Exosomes are vesicles secreted by cells, typically ranging from 30 to 150 nm in diameter, and serve as crucial mediators of intercellular communication. Exosomes are capable of loading various therapeutic substances, such as small molecule compounds, proteins, and oligonucleotides, thereby making them an ideal vehicle for drug delivery. The distinctive biocompatibility, high stability, and targeting properties of exosomes render them highly valuable for future treatments of diseases like cancer and cardiovascular diseases. Despite the potential advantage of exosomes in delivering biologically active molecules, the techniques for the preparation, purification, preservation, and other aspects of stem cell exosomes are not yet mature enough. In this paper, we briefly introduce the composition, biogenesis, and benefits of exosomes, and primarily focus on summarizing the isolation and purification methods of exosomes, the preparation of engineered exosomes, and their clinical applications, to better provide new ideas for the development of exosome drug delivery systems.
Collapse
Affiliation(s)
- Yuhao Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Shengmeng Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Lihua Zhou
- National Institute of Measurement and Testing Technology, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Kexin Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhaorui Jin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - An Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Chao Yang
- Chengdu Shiliankangjian Biotechnology Co., Ltd., Chengdu, Sichuan, 610041, People’s Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| |
Collapse
|
13
|
Zhu C, Ke X, Gu Y, Wang C, Lin S, Qian Y, Cheng J, Chen Y, Xu L, Chen Z. Antimicrobial properties and preservation potential of Allium sativum L-derived extracellular vesicle-like particles for food applications. Food Chem 2025; 484:144419. [PMID: 40267679 DOI: 10.1016/j.foodchem.2025.144419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
This study proposes an innovative approach to food preservation by leveraging extracellular vesicle-like particles derived from Garlic (ASL-EVLPs) as a natural and effective preservation agent. To address the limitations of chemical preservatives and sensory drawbacks of garlic, we systematically investigated the antibacterial mechanisms, stability, and sensory impact of ASL-EVLPs. The isolated ASL-EVLPs exhibited notable stability and biocompatibility. Antibacterial evaluations demonstrated significant inhibition of Escherichia coli (ATCC 25922) and Staphylococcus aureus (CMCC(B) 26003) through membrane disruption mechanisms. ASL-EVLPs effectively delayed spoilage and preserved sensory attributes in carrot juice, with in vivo safety confirmed. These findings position ASL-EVLPs as a dual-functional alternative, overcoming both microbial and sensory challenges in food preservation.
Collapse
Affiliation(s)
- Chenqi Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China; Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Xiaoxiao Ke
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yi Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China; Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Chunmeng Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shangyang Lin
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China; Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Yijie Qian
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China; Gusu School, Nanjing Medical University, Suzhou 215002, China
| | - Jiale Cheng
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China; Gusu School, Nanjing Medical University, Suzhou 215002, China; Linzhou County People's Hospital, Lhasa 851600, China
| | - Yan Chen
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China; Gusu School, Nanjing Medical University, Suzhou 215002, China.
| | - Liu Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhipeng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
14
|
Araujo-Abad S, Berna JM, Lloret-Lopez E, López-Cortés A, Saceda M, de Juan Romero C. Exosomes: from basic research to clinical diagnostic and therapeutic applications in cancer. Cell Oncol (Dordr) 2025; 48:269-293. [PMID: 39298081 PMCID: PMC11997007 DOI: 10.1007/s13402-024-00990-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer continues to pose a global threat despite potent anticancer drugs, often accompanied by undesired side effects. To enhance patient outcomes, sophisticated multifunctional approaches are imperative. Small extracellular vesicles (EVs), a diverse family of naturally occurring vesicles derived from cells, offer advantages over synthetic carriers. Among the EVs, the exosomes are facilitating intercellular communication with minimal toxicity, high biocompatibility, and low immunogenicity. Their tissue-specific targeting ability, mediated by surface molecules, enables precise transport of biomolecules to cancer cells. Here, we explore the potential of exosomes as innovative therapeutic agents, including cancer vaccines, and their clinical relevance as biomarkers for clinical diagnosis. We highlight the cargo possibilities, including nucleic acids and drugs, which make them a good delivery system for targeted cancer treatment and contrast agents for disease monitoring. Other general aspects, sources, and the methodology associated with therapeutic cancer applications are also reviewed. Additionally, the challenges associated with translating exosome-based therapies into clinical practice are discussed, together with the future prospects for this innovative approach.
Collapse
Affiliation(s)
- Salomé Araujo-Abad
- Cancer Research Group, Faculty of Engineering and Applied Sciences, Universidad de Las Américas, Quito, 170124, Ecuador
| | - José Marcos Berna
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Elena Lloret-Lopez
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, 170124, Ecuador
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Camino de Juan Romero
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain.
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain.
| |
Collapse
|
15
|
Murphy CA, O'Reilly D, Weiss L, Madden S, Macleod H, Chevillier AL, Neary E, O'Loughlin J, EL‐Khuffash A, Kevane B, NíAinle F, Zivny J, McCallion N, Maguire PB. Unique Patterns of Circulating Extracellular Vesicles in Preterm Infants During Adaptation to Extra-Uterine Life. J Extracell Vesicles 2025; 14:e70064. [PMID: 40194994 PMCID: PMC11975508 DOI: 10.1002/jev2.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
There is growing interest in the role of extracellular vesicles (EVs) in neonatal pathology. This study aimed to characterise circulating EVs following preterm birth. This single-centre prospective observational study included cord and postnatal plasma from preterm (n = 101) and full-term infants (n = 66). EVs were analysed using nanoparticle tracking analysis, flow cytometry, proteomics and procoagulant activity assay. We found changes in the concentration, size, cellular origin and proteomic content of circulating EVs in preterm infants during perinatal adaptation. To understand if these changes were related to prematurity or normal adaptation to extrauterine life, they were also investigated in term infants. There was a dramatic increase in the concentration of small and large EVs on Day 3 in the preterm group; specific subsets of platelet (CD42b+ and CD62P+), endothelial (VEGFR2) and tissue factor EVs were elevated. Differentially expressed proteins relating to haemostasis, pulmonary physiology and immunity were identified between Day 1 and 3 in preterm infants. These changes have never previously been described in a large cohort of preterm infants and differ from healthy term infants. These findings have major implications for future neonatal EV studies, particularly the timing of sample collection. Further work is required to understand the clinical implications of this unique EV profile following preterm birth.
Collapse
Affiliation(s)
- Claire A. Murphy
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Daniel O'Reilly
- Department of NeonatologyRotunda HospitalDublinIreland
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Luisa Weiss
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Stephen Madden
- School of Population HealthRoyal College of Surgeons in IrelandDublinIreland
| | - Hayley Macleod
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Ana Le Chevillier
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Elaine Neary
- Department of NeonatologyLiverpool Women's HospitalLiverpoolUK
- Department of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - John O'Loughlin
- Department of Laboratory MedicineRotunda HospitalDublinIreland
| | - Afif EL‐Khuffash
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Barry Kevane
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
| | - Fionnuala NíAinle
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
- Department of HaematologyRotunda HospitalDublinIreland
| | - Jan Zivny
- Institute of Pathological PhysiologyFirst Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Naomi McCallion
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Patricia B. Maguire
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| |
Collapse
|
16
|
Zhang X, Ma L, Liu X, Zhou X, Wang A, Lai Y, Zhang J, Li Y, Chen S. Sustained release of miR-21 carried by mesenchymal stem cell-derived exosomes from GelMA microspheres inhibits ovarian granulosa cell apoptosis in premature ovarian insufficiency. Mater Today Bio 2025; 31:101469. [PMID: 39906205 PMCID: PMC11790500 DOI: 10.1016/j.mtbio.2025.101469] [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: 04/18/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/06/2025] Open
Abstract
Background Premature ovarian insufficiency (POI) refers to the severe decline or failure of ovarian function in women younger than 40 years of age. It is a serious hazard to women's physical and mental health, but current treatment options are limited. Mesenchymal stem cell-derived exosomes (MSC-Exo) exhibit promising potential as a therapeutic approach for POI. However, their clinical application is hindered by their instability and low long-term retention rate in vivo. Methods and results In this study, miR-21 was identified as the predominant miRNA with low-expression in follicular fluid exosomes of POI patients and was shown to possess antiapoptotic activity. Next, we loaded miR-21 agomir to MSC-Exo to form Agomir21-Exo, which significantly reversed the apoptosis of granulosa cells in vitro. Moreover, we successfully developed GelMA hydrogel microspheres for encapsulating Agomir21-Exo through microfluidic technology, named GelMA-Ag21Exo, which had good injectability and significantly enhanced the stability and long-term retention of Agomir21-Exo in mice through sustained release. The release of Agomir21-Exo from GelMA-Ag21Exo notably alleviated the apoptosis of ovarian granulosa cells and improved the ovarian reserve and fertility in POI mice. Conclusion Our findings illustrate that activating miR-21 through Agomir21-Exo could improve the function of ovarian granulosa cells. The GelMA-Ag21Exo enhanced the exosome-based therapeutic efficacy of the Agomir21-Exo in vivo. These findings provide a novel and promising treatment strategy for POI patients.
Collapse
Affiliation(s)
| | | | | | - Xingyu Zhou
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ao Wang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yunhui Lai
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Zhang
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ying Li
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shiling Chen
- Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
17
|
Mukerjee N, Bhattacharya A, Maitra S, Kaur M, Ganesan S, Mishra S, Ashraf A, Rizwan M, Kesari KK, Tabish TA, Thorat ND. Exosome isolation and characterization for advanced diagnostic and therapeutic applications. Mater Today Bio 2025; 31:101613. [PMID: 40161926 PMCID: PMC11950786 DOI: 10.1016/j.mtbio.2025.101613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Advancements in exosome isolation technologies are pivotal for transforming personalized medicine and enhancing clinical diagnostics. Exosomes, small extracellular vesicles with diameters ranging between 30 and 150 nm, are secreted into bodily fluids by a variety of cells and play essential roles in intercellular communication. These vesicles facilitate the transfer of nucleic acids, lipids, and proteins, affecting a wide range of biological and pathological processes. Given their importance in disease diagnostics, therapy, and as biomarkers, there has been a surge in developing methods to isolate them from fluids such as urine, saliva, blood, and cerebrospinal fluid. While traditional isolation techniques like ultracentrifugation and polymer-based precipitation have been foundational, recent technological advances have introduced more precise methods like microfluidics and immunoaffinity capture. These newer methods enable high-throughput and specific exosome isolation by targeting surface markers, thus enhancing purity. However, challenges such as balancing purity with yield and the lack of standardized protocols across different laboratories persist, impacting the consistency of findings. By integrating advanced isolation techniques and discussing their implications in diagnostics and therapy, this review aims to catalyze further research and adoption of exosome-based technologies in medicine, marking a significant stride towards tailored healthcare solutions.
Collapse
Affiliation(s)
- Nobendu Mukerjee
- Centre for Infectious Diseases & Microbiology, School of Public Health Sciences and Technology, Malla Reddy Vishwavidyapeeth, Hyderabad 500 055, Telangana, India
| | - Arghya Bhattacharya
- Department of Pharmacology, Bengal School of Technology, West Bengal, Kolkata, 712102, India
| | - Swastika Maitra
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shivang Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ayash Ashraf
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, 140307, Punjab, India
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Department of Ophthalmology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Tanveer A. Tabish
- Radcliffe Department of Medicine, University of Oxford, OX3 7BN, United Kingdom
| | - Nanasaheb D. Thorat
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| |
Collapse
|
18
|
Yang Y, Duan Y, Yue J, Yin Y, Ma Y, Wan X, Shao J. Exosomes: an innovative therapeutic target for cerebral ischemia-reperfusion injury. Front Pharmacol 2025; 16:1552500. [PMID: 40206077 PMCID: PMC11979243 DOI: 10.3389/fphar.2025.1552500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Ischemic stroke is caused by artery stenosis or occlusion, which reduces blood flow and may cause brain damage. Treatment includes restoring blood supply; however, ischemia-reperfusion can still aggravate tissue injury. Reperfusion injury can increase levels of reactive oxygen species, exacerbate mitochondrial dysfunction, create excessive autophagy and ferroptosis, and cause inflammation during microglial infiltration. Cerebral ischemia-reperfusion injury (CIRI) is a key challenge in the treatment of ischemic stroke. Currently, thrombolysis (e.g., rt-PA therapy) and mechanical thrombectomy are the primary treatments, but their application is restricted by narrow therapeutic windows (<4.5 h) and risks of hemorrhagic complications. Exosomes reduce CIRI by regulating oxidative stress, mitochondrial autophagy, inflammatory responses, and glial cell polarization. In addition, their noncellular characteristics provide a safer alternative to stem cell therapy. This article reviews the research progress of exosomes in CIRI in recent years.
Collapse
Affiliation(s)
- Yuan Yang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yushan Duan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jinxi Yue
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yue Yin
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiming Ma
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaohong Wan
- Department of Critical Care Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Jianlin Shao
- Department of Anesthesiology, The First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
19
|
Jangam TC, Desai SA, Patel VP, Pagare NB, Raut ND. Exosomes as Therapeutic and Diagnostic Tools: Advances, Challenges, and Future Directions. Cell Biochem Biophys 2025:10.1007/s12013-025-01730-5. [PMID: 40122928 DOI: 10.1007/s12013-025-01730-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Exosomes are tiny extracellular vesicles that are essential for intercellular communication and have shown great promise in the detection and treatment of disease. They are especially useful in the treatment of cancer, cardiovascular conditions, and neurological diseases because of their capacity to transport bioactive substances including proteins, lipids, and nucleic acids. Because of their low immunogenicity, ability to traverse biological barriers, and biocompatibility, exosome-based medicines have benefits over conventional treatments. Large-scale production, standardization of separation methods, possible immunological reactions, and worries about unforeseen biological effects are some of the obstacles that still need to be overcome. Furthermore, there are major barriers to the clinical use of exosomes due to their complex cargo sorting mechanisms and heterogeneity. Future studies should concentrate on enhancing separation and purification procedures, optimizing exosome engineering techniques, and creating plans to reduce immune system modifications. This review examines the most recent developments in exosome-based diagnostics and treatments, identifies current issues, and suggests ways to improve their clinical translation in the future.
Collapse
Affiliation(s)
- Tejas C Jangam
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nishant B Pagare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita D Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
20
|
Chi H, Shi L, Gan S, Fan G, Dong Y. Innovative Applications of Nanopore Technology in Tumor Screening: An Exosome-Centric Approach. BIOSENSORS 2025; 15:199. [PMID: 40277513 PMCID: PMC12024935 DOI: 10.3390/bios15040199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/26/2025]
Abstract
Cancer remains one of the leading causes of death worldwide. Its complex pathogenesis and metastasis pose significant challenges for early diagnosis, underscoring the urgent need for innovative and non-invasive tumor screening methods. Exosomes, small extracellular vesicles that reflect the physiological and pathological states of their parent cells, are uniquely suited for cancer liquid biopsy due to their molecular cargo, including RNA, DNA, and proteins. However, traditional methods for exosome isolation and detection are often limited by inadequate sensitivity, specificity, and efficiency. Nanopore technology, characterized by high sensitivity and single-molecule resolution, offers powerful tools for exosome analysis. This review highlights its diverse applications in tumor screening, such as magnetic nanopores for high-throughput sorting, electrochemical sensing for real-time detection, nanomaterial-based assemblies for efficient capture, and plasmon resonance for ultrasensitive analysis. These advancements have enabled precise exosome detection and demonstrated promising potential in the early diagnosis of breast, pancreatic, and prostate cancers, while also supporting personalized treatment strategies. Additionally, this review summarizes commercialized products for exosome-based cancer diagnostics and examines the technical and translational challenges in clinical applications. Finally, it discusses the future prospects of nanopore technology in advancing liquid biopsy toward clinical implementation. The continued progress of nanopore technology not only accelerates exosome-based precision medicine but also represents a significant step forward in next-generation liquid biopsy and tumor screening.
Collapse
Affiliation(s)
- Heng Chi
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
| | - Liuxin Shi
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
| | | | | | - Yuliang Dong
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
- BGI Research, Hangzhou 310030, China;
| |
Collapse
|
21
|
Xiao Q, Tan M, Yan G, Peng L. Revolutionizing lung cancer treatment: harnessing exosomes as early diagnostic biomarkers, therapeutics and nano-delivery platforms. J Nanobiotechnology 2025; 23:232. [PMID: 40119368 PMCID: PMC11929271 DOI: 10.1186/s12951-025-03306-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 03/08/2025] [Indexed: 03/24/2025] Open
Abstract
Lung cancer, known for its high morbidity and mortality rates, remains one of the most critical health challenges globally. Conventional treatment options, such as chemotherapy and surgery, are often limited by high costs, significant side effects, and often yield a poor prognosis. Notably, recent research has shed light on the potential therapeutic roles of exosomes, which essentially influence lung cancer's development, diagnosis, treatment, and prognosis. Exosomes have been revealed for their exceptional properties, including natural intercellular communication, excellent biocompatibility, minimal toxicity, prolonged blood circulation ability, and biodegradability. These unique characteristics position exosomes as highly effective drug delivery systems, nanotherapeutics, and potential diagnostic and prognostic biomarkers in lung cancer. This review provides a comprehensive review of the physiological and pathological roles of exosomes in lung cancer, emphasizing their potential as innovative diagnostic biomarkers, therapeutics, and delivery platforms. By harnessing their unique properties, exosomes are poised to revolutionize the diagnosis and treatment of lung cancer, offering a promising avenue for more personalized and effective therapies.
Collapse
Affiliation(s)
- Qiyao Xiao
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Minhong Tan
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Ge Yan
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China
| | - Lihua Peng
- College of Pharmaceutical Sciences, Zhejiang University, 866# Yuhangtang Road, Hangzhou, 310058, People's Republic of China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, People's Republic of China.
| |
Collapse
|
22
|
Zubair M, Abouelnazar FA, Iqbal MA, Pan J, Zheng X, Chen T, Shen W, Yin J, Yan Y, Liu P, Mao F, Chu Y. Mesenchymal stem cell-derived exosomes as a plausible immunomodulatory therapeutic tool for inflammatory diseases. Front Cell Dev Biol 2025; 13:1563427. [PMID: 40129569 PMCID: PMC11931156 DOI: 10.3389/fcell.2025.1563427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs), especially, exosomes are considered to have diverse therapeutic effects for various significant diseases. MSC-derived exosomes (MSCex) offer substantial advantages over MSCs due to their long-term preservation, stability, absence of nuclei and fewer adverse effects such as infusion toxicity, thereby paving the way towards regenerative medicine and cell-free therapeutics. These exosomes harbor several cellular contents such as DNA, RNA, lipids, metabolites, and proteins, facilitating drug delivery and intercellular communication. MSCex have the ability to immunomodulate and trigger the anti-inflammatory process hence, playing a key role in alleviating inflammation and enhancing tissue regeneration. In this review, we addressed the anti-inflammatory effects of MSCex and the underlying immunomodulatory pathways. Moreover, we discussed the recent updates on MSCex in treating specific inflammatory diseases, including arthritis, inflammatory bowel disease, inflammatory eye diseases, and respiratory diseases such as asthma and acute respiratory distress syndrome (ARDS), as well as neurodegenerative and cardiac diseases. Finally, we highlighted the challenges in using MSCex as the successful therapeutic tool and discussed future perspectives.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Fatma A. Abouelnazar
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, Egypt
| | | | - Jingyun Pan
- Department of Traditional Chinese Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Xuwen Zheng
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Tao Chen
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Wenming Shen
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Jinnan Yin
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Pengjun Liu
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Chu
- Wujin Clinical College, Xuzhou Medical University, Changzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
23
|
Ondracek AS, Afonyushkin T, Aszlan A, Taqi S, Koller T, Artner T, Porsch F, Resch U, Sharma S, Scherz T, Spittler A, Haertinger M, Hofbauer TM, Ozsvar-Kozma M, Seidl V, Beitzke D, Krueger M, Testori C, Lang IM, Binder CJ. Malondialdehyde-specific natural IgM inhibit NETosis triggered by culprit site-derived extracellular vesicles from myocardial infarction patients. Eur Heart J 2025; 46:926-939. [PMID: 39215577 PMCID: PMC11887544 DOI: 10.1093/eurheartj/ehae584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/08/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS Neutrophil extracellular traps (NETs) trigger atherothrombosis during acute myocardial infarction (AMI), but mechanisms of induction remain unclear. Levels of extracellular vesicles (EV) carrying oxidation-specific epitopes (OSE), which are targeted by specific natural immunoglobulin M (IgM), are increased at the culprit site in AMI. This study investigated EV as inducers of NETosis and assessed the inhibitory effect of natural anti-OSE-IgM in this process. METHODS Blood from the culprit and peripheral site of ST-segment elevation myocardial infarction (STEMI) patients (n = 28) was collected, and myocardial function assessed by cardiac magnetic resonance imaging (cMRI) 4 ± 2 days and 195 ± 15 days post-AMI. Extracellular vesicles were isolated from patient plasma and cell culture supernatants for neutrophil stimulation in vitro and in vivo, in the presence of a malondialdehyde (MDA)-specific IgM or an isotype control. NETosis and neutrophil functions were assessed via enzyme-linked immunosorbent assay and fluorescence microscopy. Pharmacological inhibitors were used to map signalling pathways. Neutrophil extracellular trap markers and anti-OSE-IgM were measured by ELISA. RESULTS CD45+ MDA+ EV and NET markers were elevated at the culprit site. Extracellular vesicles induced neutrophil activation and NET formation via TLR4 and PAD4, and mice injected with EV showed increased NETosis. Malondialdehyde-specific IgM levels were inversely associated with citH3 in STEMI patient blood. An MDA-specific IgM inhibited EV-induced NET release in vitro and in vivo. CD45+ MDA+ EV concentrations inversely correlated with left ventricular ejection fraction post-AMI. CONCLUSIONS Culprit site-derived EV induce NETosis, while MDA-specific natural IgM inhibit this effect, potentially impacting outcome after AMI.
Collapse
Affiliation(s)
- Anna S Ondracek
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Taras Afonyushkin
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 5H, 1090 Vienna, Austria
| | - Adrienne Aszlan
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Soreen Taqi
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 5H, 1090 Vienna, Austria
| | - Thomas Koller
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 5H, 1090 Vienna, Austria
| | - Tyler Artner
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Florentina Porsch
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 5H, 1090 Vienna, Austria
| | - Ulrike Resch
- Department of Vascular Biology and Thrombosis Research, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Institute for Genetics and Cologne Excellence Cluster for Aging and Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Smriti Sharma
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Thomas Scherz
- Department of Dermatology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
| | - Andreas Spittler
- Department of Surgery and Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Maximilian Haertinger
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Thomas M Hofbauer
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Maria Ozsvar-Kozma
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 5H, 1090 Vienna, Austria
| | - Veronika Seidl
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-guided therapy, Medical University of Vienna, Vienna, Austria
| | - Marcus Krueger
- Institute for Genetics and Cologne Excellence Cluster for Aging and Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Christoph Testori
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Irene M Lang
- Department of Internal Medicine II, Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 6L, 1090 Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 5H, 1090 Vienna, Austria
| |
Collapse
|
24
|
Perrone MG, Filieri S, Azzariti A, Armenise D, Baldelli OM, Liturri A, Sardanelli AM, Ferorelli S, Miciaccia M, Scilimati A. Exosomes in Ovarian Cancer: Towards Precision Oncology. Pharmaceuticals (Basel) 2025; 18:371. [PMID: 40143147 PMCID: PMC11946531 DOI: 10.3390/ph18030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Identification of targetable biomarkers to improve early disease detection and overall patient outcomes is becoming an urgent need in clinical oncology. Ovarian cancer (OC) has one of the highest mortality rates among gynecological cancers. It is asymptomatic and almost always diagnosed at an advanced stage (III or IV), leading to a 5-year survival rate of approximately 35%. Methods: Current therapeutic approaches for OC are very limited and mainly consist of cytoreductive surgery and cisplatin plus taxane-based chemotherapy. No gender and tumor specific biomarkers are known. Exosomes, lipid bilayer vesicles of endocytic origin secreted by most cell types, represent sources of information for their involvement in the onset and progression of many diseases. Hence, research on exosome contents as tools and targets in precise oncology therapy provides knowledge essential to improving diagnosis and prognosis of the disease. Results: This review attempts to give an overview of how exosomes are implicated in ovarian carcinoma pathogenesis to trigger further cancer exosome-based investigations aimed at developing ovarian cancer fine-tuning diagnostic methodologies. Conclusions: It is essential to investigate exosome-based cancer drugs to advance understanding, improve treatment plans, create personalized strategies, ensure safety, and speed up clinical translation to increase patients' overall survival and quality of life. Papers published in PubMed and Web of Science databases in the last five years (2020-2024) were used as a bibliographic source.
Collapse
Affiliation(s)
- Maria Grazia Perrone
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| | - Silvana Filieri
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy; (S.F.); (A.M.S.)
| | - Amalia Azzariti
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, V. O. Flacco, 65, 70124 Bari, Italy;
| | - Domenico Armenise
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| | - Olga Maria Baldelli
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| | - Anselma Liturri
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| | - Anna Maria Sardanelli
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, 70124 Bari, Italy; (S.F.); (A.M.S.)
| | - Savina Ferorelli
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| | - Morena Miciaccia
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| | - Antonio Scilimati
- Research Laboratory for Woman and Child Health, Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125 Bari, Italy; (M.G.P.); (D.A.); (O.M.B.); (A.L.); (S.F.)
| |
Collapse
|
25
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
26
|
Taori S, Habib A, Adida S, Gecici NN, Sharma N, Calcaterra M, Tang A, Pandya S, Mehra A, Deng H, Elidrissy H, Idrissi YA, Amjadzadeh M, Zinn PO. Circulating biomarkers in high-grade gliomas: current insights and future perspectives. J Neurooncol 2025; 172:41-49. [PMID: 39671020 DOI: 10.1007/s11060-024-04903-z] [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: 07/06/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
PURPOSE High-grade gliomas (HGG) represent a challenging subset of brain tumors characterized by aggressive nature and poor prognosis. Histopathology remains to be the standard for diagnosis, however, it is invasive, prone to sampling errors, and may not capture the full tumor heterogeneity and evolution over time. In recent years, there has been a growing interest in the potential utility of circulating biomarkers, obtained through minimally-invasive liquid biopsies, providing an opportunity for diagnosis, prognostication, monitoring treatment response and developing targeted therapies. METHODS We have reviewed the literature on circulating biomarkers for HGG, including circulating tumor cells (CTCs), circulating tumor-derived exosomes/extracellular vesicles (ctEVs), circulating tumor-derived DNA (ctDNA), circulating tumor-derived miRNA (ctmiRNA), and circulating tumor-derived proteins. RESULTS CTCs provide real-time information about tumor characteristics for molecular profiling and monitoring treatment response, yet their low numbers in circulation makes detection challenging. ctEVs carry a range of biomolecules and are easily detectable. However, they are not exclusively released from tumor cells and heterogeneity in their content requires standardized isolation and analysis methods. ctDNA is another promising biomarker with its levels correlating with the disease stage. However, its low concentration in blood requires highly sensitive techniques for identification and differentiation from normal cell-free DNA. ctmiRNA and tumor-derived proteins show promise but are limited by their susceptibility to dilution and lack of specificity in current technology. CONCLUSION This review highlights the transformative potential of circulating biomarkers in the management of HGG, with implications for improving patient outcomes, optimizing treatment strategies, and advancing precision oncology in neuro-oncology practice.
Collapse
Affiliation(s)
- Suchet Taori
- School of Medicine, University of Pittsburgh, Pennsylvania, PA, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Ahmed Habib
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Samuel Adida
- School of Medicine, University of Pittsburgh, Pennsylvania, PA, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Neslihan Nisa Gecici
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Nikhil Sharma
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | | | - Anthony Tang
- School of Medicine, University of Pittsburgh, Pennsylvania, PA, USA
| | - Sumaarg Pandya
- School of Medicine, University of Pittsburgh, Pennsylvania, PA, USA
| | - Arnav Mehra
- School of Medicine, University of Pittsburgh, Pennsylvania, PA, USA
| | - Hansen Deng
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Hayat Elidrissy
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Yassine Alami Idrissi
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Mohammadreza Amjadzadeh
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA
| | - Pascal O Zinn
- School of Medicine, University of Pittsburgh, Pennsylvania, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, PA, USA.
| |
Collapse
|
27
|
Shakouri-Motlagh A, O'Connor AJ, Brennecke SP, Heath DE, Kalionis B. Extracellular vesicles support increased expansion of mesenchymal stromal cells on fetal membrane-derived decellularized extracellular matrix. Cell Tissue Res 2025; 399:323-336. [PMID: 39715869 DOI: 10.1007/s00441-024-03946-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
Decidual mesenchymal stromal cells (DMSC) were the source of extracellular vesicles (DMSC_EV). The xCELLigence real-time cell growth assay revealed increasing concentrations of EVs decreased DMSC attachment in the early growth phase but stimulated DMSC proliferation at day 7 when grown on tissue culture plastic (TCP). DMSC attachment and proliferation varied depending on the growth surface and DMSC_EV supplementation. DMSC attachment increased on decellularized and solubilized amniotic (s-dAM) whether or not EVs were added. Only Matrigel substrate increased DMSC attachment with added EVs. The addition of EVs increased DMSC proliferation only on the s-dAM substrate. DMSCs were more motile on s-dAM and decellularized and solubilized chorionic (s-dCM) membranes following EV addition. The osteogenic potential of DMSCs was improved on s-dAM substrates when supplanted with EVs. Finally, the levels of reactive oxygen species in DMSCs varied depending on the substrate but not on added EVs. We show that the addition of in vitro EVs isolated from the source being expanded (i.e., DMSCs) and the presence of ECM improve DMSC behaviours during ex vivo expansion. The inclusion of two key components of the MSC niche, EVs and ECM, benefitted the ex vivo expansion of MSCs. Added in vitro EVs increased the proliferation of DMSCs when grown on s-dAM but not on s-dCM, whereas they improved DMSC mobility on both surfaces. Testing different ECMs could be used to promote specific desired characteristics of DMSCs, and different combinations of EVs and ECM may enhance desirable MSC characteristics for specific therapeutic settings.
Collapse
Affiliation(s)
- Aida Shakouri-Motlagh
- Department of Biomedical Engineering, School of Engineering, The University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Royal Women's Hospital Campus, Parkville, VIC, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, School of Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Shaun P Brennecke
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Royal Women's Hospital Campus, Parkville, VIC, Australia
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia
| | - Daniel E Heath
- Department of Biomedical Engineering, School of Engineering, The University of Melbourne, Parkville, VIC, Australia.
| | - Bill Kalionis
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Royal Women's Hospital Campus, Parkville, VIC, Australia.
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia.
| |
Collapse
|
28
|
Nakamura K, Ida N, Hirasawa A, Okamoto K, Vu TH, Hai Ly DT, Masuyama H. CD63 as a potential biomarker for patients with ovarian cancer. Eur J Obstet Gynecol Reprod Biol 2025; 306:87-93. [PMID: 39799740 DOI: 10.1016/j.ejogrb.2025.01.011] [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: 12/12/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Exosomes play an important role in regulating physiological processes and mediating the systemic dissemination of various types of cancer. We investigated the association of exosomal tetraspanins CD9, CD63, and CD81 in patients with ovarian cancer (OC). MATERIAL AND METHODS We measured the plasma tetraspanins CD9, CD63, and CD81 by enzyme-linked immunosorbent assay in 91 patients who underwent treatment for OC between April 2018 and March 2024. Additionally, we analyzed clinical pathologic factors, chemotherapy response, and prognosis. RESULTS In terms of stages, CD63 expression was significantly higher in patients with stage IV compared to those with stage I OC (p = 0.003). In terms of histological type, CD63 expression was significantly higher in high-grade serous carcinoma (HGSC) than in clear cell carcinoma (CCC) with OC (p = 0.009). Furthermore, CD63 levels were significantly higher in advanced-stage, HGSC than in patients with early-stage, non-HGSC and early-stage, HGSC OC (p = 0.045 and p = 0.002, respectively). In the Neoadjuvant chemotherapy (NAC) of 12 patients with OC assessed as having either a partial response (PR) or complete response (CR), CD63 was significantly decreased (p = 0.043), whereas perforin was significantly increased (p = 0.001). In the NAC of 16 patients with OC, CD63 of the response rate to chemotherapy tended to differ between the progressive disease (PD) and PR/CR groups (p = 0.056). A moderate inverse correlation was observed between CD63 and perforin levels (R = 0.638, R2 = 0.428, p = 0.008). CONCLUSIONS CD63 could be a potential biomarker for all types of OC patients.
Collapse
Affiliation(s)
- Keiichiro Nakamura
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| | - Naoyuki Ida
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan
| | - Akira Hirasawa
- Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| | - Kazuhiro Okamoto
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan
| | - Thuy Ha Vu
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan
| | - Dao Thi Hai Ly
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| | - Hisashi Masuyama
- Department of Obstetrics and Gynecology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho kitaku, Okayama 700-8558, Japan.
| |
Collapse
|
29
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Patel AA, Shafie A, Mohamed AH, Ali SAJ, Tayeb FJ, Waggiallah HA, Ahmad I, Sheweita SA, Muzammil K, AlShahrani AM, Al Abdulmonem W. The promise of mesenchymal stromal/stem cells in erectile dysfunction treatment: a review of current insights and future directions. Stem Cell Res Ther 2025; 16:98. [PMID: 40012076 PMCID: PMC11866689 DOI: 10.1186/s13287-025-04221-9] [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: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
Erectile dysfunction is a common and multifactorial condition that significantly impacts men's quality of life. Traditional treatments, such as phosphodiesterase type 5 inhibitors (PDE5i), often fail to provide lasting benefits, particularly in patients with underlying health conditions. In recent years, regenerative medicine, particularly stem cell therapies, has emerged as a promising alternative for managing erectile dysfunction. This review explores the potential of mesenchymal stromal/stem cells (MSCs) and their paracrine effects, including extracellular vesicles (EVs), in the treatment of erectile dysfunction. MSCs have shown remarkable potential in promoting tissue repair, reducing inflammation, and regenerating smooth muscle cells, offering therapeutic benefits in models of erectile dysfunction. Clinical trials have demonstrated positive outcomes in improving erectile function and other clinical parameters. This review highlights the promise of MSC therapy for erectile dysfunction, discusses existing challenges, and emphasizes the need for continued research to refine these therapies and improve long-term patient outcomes.
Collapse
Affiliation(s)
- Ayyub Ali Patel
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | | | - Faris J Tayeb
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkarj, Saudi Arabia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Salah Ahmed Sheweita
- Department of Clinical Biochemistry, Faculty of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait, King Khalid University, 62561, Abha, Saudi Arabia
| | - Abdullah M AlShahrani
- Department of Basic Medical Science, College of Applied Medical Sciences, Khamis Mushait, King Khalid University (KKU), 62561, Abha, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| |
Collapse
|
31
|
Zhang X, Yang M, Chen X, Zhang M, Peng Y, Lu M. Melatonin-pretreated mesenchymal stem cell-derived exosomes alleviate cavernous fibrosis in a rat model of nerve injury-induced erectile dysfunction via miR-145-5p/TGF-β/Smad axis. Stem Cell Res Ther 2025; 16:96. [PMID: 40001250 PMCID: PMC11863846 DOI: 10.1186/s13287-025-04173-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cavernous nerve injury-induced erectile dysfunction (CNI-ED) is a common complication after radical prostatectomy. Conventional treatment approaches have had little success in treating the severe cavernous fibrosis which is a consequence of CNI-ED. METHODS Pre-treatment of adipose-derived stem cells with melatonin allows for the extraction of active exosomes (MT-hASC-EVs) from the conditioned medium. The therapeutic effects of MT-hASC-EVs were assessed in a rat model of CNI-ED, and the anti-fibrotic properties were evaluated. MicroRNA sequencing was used to identify specific microRNAs highly expressed in MT-hASC-EVs, and differential microRNAs were screened for regulatory pathways through target gene enrichment analysis. Finally, the conclusions from bioinformatics analysis were validated through in vitro experiments. RESULTS Intracavernous injection of MT-hASC-EVs significantly restored erectile function and reduced the extent of corpus cavernosum fibrosis in the CNI-ED rat model. MT-hASC-EVs promoted the proliferation and anti-apoptotic effects of corpus cavernosum smooth muscle cells (CCSMCs) in vitro. Mechanistically, MT-hASC-EVs inhibit fibrosis by delivering miR-145-5p, which targets TGF-β2/Smad3 axis. CONCLUSIONS MT-hASCs-EVs can inhibit cavernous fibrosis and improve erectile function in a rat model of CNI-ED by targeting the miR-145-5p/TGF-β/Smad axis.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Mengbo Yang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Xinda Chen
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Ming Zhang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Yiliang Peng
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Mujun Lu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China.
| |
Collapse
|
32
|
Dong X, Lin Y, Li K, Liang G, Huang X, Pan J, Wang L, Zhang D, Liu T, Wang T, Yan X, Zhang L, Li X, Qu X, Jia D, Li Y, Zhang H. Consensus statement on extracellular vesicles in liquid biopsy for advancing laboratory medicine. Clin Chem Lab Med 2025; 63:465-482. [PMID: 38896030 DOI: 10.1515/cclm-2024-0188] [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: 02/07/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024]
Abstract
Extracellular vesicles (EVs) represent a diverse class of nanoscale membrane vesicles actively released by cells. These EVs can be further subdivided into categories like exosomes and microvesicles, based on their origins, sizes, and physical attributes. Significantly, disease-derived EVs have been detected in virtually all types of body fluids, providing a comprehensive molecular profile of their cellular origins. As a result, EVs are emerging as a valuable addition to liquid biopsy techniques. In this collective statement, the authors share their current perspectives on EV-related research and product development, with a shared commitment to translating this newfound knowledge into clinical applications for cancer and other diseases, particularly as disease biomarkers. The consensus within this document revolves around the overarching recognition of the merits, unresolved questions, and existing challenges surrounding EVs. This consensus manuscript is a collaborative effort led by the Committee of Exosomes, Society of Tumor Markers, Chinese anti-Cancer Association, aimed at expediting the cultivation of robust scientific and clinically applicable breakthroughs and propelling the field forward with greater swiftness and efficacy.
Collapse
Affiliation(s)
- Xingli Dong
- 558113 Central Laboratory, Department of Hematology and Oncology, Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Shenzhen Clinical Research Center for hematologic disease, Shenzhen University General Hospital , Shenzhen, Guangdong, China
| | - Yusheng Lin
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Thoracic Surgery, 47885 The First Affiliated Hospital of Jinan University , Guangzhou, China
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Kai Li
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Gaofeng Liang
- 74623 School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology , Luoyang, China
| | - Xiaoyi Huang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Heilongjiang Province, Harbin, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Jingxuan Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, School of Medicine
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, and College of Pharmacy, State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
| | - Tingjiao Liu
- Department of Oral Pathology, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Tong Wang
- 47885 MOE Key Laboratory of Tumor Molecular Biology, College of Life Science and Technology, Jinan University , Guangzhou, China
| | - Xiaomei Yan
- Department of Chemical Biology, 534787 MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University , Xiamen, China
| | - Long Zhang
- 12377 MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University , Hangzhou, China
| | - Xiaowu Li
- Department of Hepatobiliary Surgery, 558113 Shenzhen Key Laboratory, Shenzhen University General Hospital , Shenzhen, Guangdong, China
| | - Xiujuan Qu
- Department of Medical Oncology, 159407 The First Hospital of China Medical University , Shenyang, China
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, Australia
| | - Hao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China
- Institute of Precision Cancer Medicine and Pathology, and Department of Pathology, School of Medicine, Jinan University, Guangzhou, P.R. China
| |
Collapse
|
33
|
Jin W, Li Y, Yu M, Ren D, Han C, Guo S. Advances of exosomes in diabetic wound healing. BURNS & TRAUMA 2025; 13:tkae078. [PMID: 39980588 PMCID: PMC11836438 DOI: 10.1093/burnst/tkae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/11/2024] [Accepted: 11/09/2024] [Indexed: 02/22/2025]
Abstract
Poor wound healing is a refractory process that places an enormous medical and financial burden on diabetic patients. Exosomes have recently been recognized as crucial players in the healing of diabetic lesions. They have excellent stability, homing effects, biocompatibility, and reduced immunogenicity as novel cell-free therapies. In addition to transporting cargos to target cells to enhance intercellular communication, exosomes are beneficial in nearly every phase of diabetic wound healing. They participate in modulating the inflammatory response, accelerating proliferation and reepithelization, increasing angiogenesis, and regulating extracellular matrix remodeling. Accumulating evidence indicates that hydrogels or dressings in conjunction with exosomes can prolong the duration of exosome residency in diabetic wounds. This review provides an overview of the mechanisms, delivery, clinical application, engineering, and existing challenges of the use of exosomes in diabetic wound repair. We also propose future directions for biomaterials incorporating exosomes: 2D or 3D scaffolds, biomaterials loaded with wound healing-promoting gases, intelligent biomaterials, and the prospect of systematic application of exosomes. These findings may might shed light on future treatments and enlighten some studies to improve quality of life among diabetes patients.
Collapse
Affiliation(s)
- Weixue Jin
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 1511 Jiang Hong Road, Binjiang District, Hangzhou 310009, Zhejiang, China
| | - Yi Li
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 1511 Jiang Hong Road, Binjiang District, Hangzhou 310009, Zhejiang, China
| | - Meirong Yu
- Center for Basic and Translational Research, Second Affiliated Hospital Zhejiang University School of Medicine, 88 Jie Fang Road, Shangcheng District, Hangzhou 310009, Zhejiang, China
| | - Danyang Ren
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 1511 Jiang Hong Road, Binjiang District, Hangzhou 310009, Zhejiang, China
| | - Chunmao Han
- Department of Burns and Wound Repair, Second Affiliated Hospital Zhejiang University School of Medicine, 88 Jie Fang Road, Shangcheng District, Hangzhou 310009, Zhejiang, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, 88 Jie Fang Road, Shangcheng District, Hangzhou 310009, Zhejiang, China
| | - Songxue Guo
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 1511 Jiang Hong Road, Binjiang District, Hangzhou 310009, Zhejiang, China
- Zhejiang Key Laboratory of Trauma, Burn, and Medical Rescue, 88 Jie Fang Road, Shangcheng District, Hangzhou 310009, Zhejiang, China
| |
Collapse
|
34
|
Mohamed AH, Abaza T, Youssef YA, Rady M, Fahmy SA, Kamel R, Hamdi N, Efthimiado E, Braoudaki M, Youness RA. Extracellular vesicles: from intracellular trafficking molecules to fully fortified delivery vehicles for cancer therapeutics. NANOSCALE ADVANCES 2025; 7:934-962. [PMID: 39823046 PMCID: PMC11733735 DOI: 10.1039/d4na00393d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) are emerging as viable tools in cancer treatment due to their ability to carry a wide range of theranostic activities. This review summarizes different forms of EVs such as exosomes, microvesicles, apoptotic bodies, and oncosomes. It also sheds the light onto isolation methodologies, characterization techniques and therapeutic applications of all discussed EVs. Evidence indicates that EVs are particularly effective in delivering chemotherapeutic medications, and immunomodulatory agents. However, the advancement of EV-based therapies into clinical practice is hindered by challenges including EVs heterogeneity, cargo loading efficiency, and in vivo stability. Overall, EVs have the potential to change cancer therapeutic paradigms. Continued research and development activities are critical for improving EV-based medications and increasing their therapeutic impact.
Collapse
Affiliation(s)
- Adham H Mohamed
- Department of Chemistry, Faculty of Science, Cairo University 12613 Giza Egypt
| | - Tasneem Abaza
- Biotechnology and Biomolecular Chemistry Program, Faculty of Science, Cairo University 12613 Giza Egypt
- Université Paris-Saclay, Université d'Evry Val D'Essonne 91000 Évry-Courcouronnes Île-de-France France
| | - Yomna A Youssef
- Department of Physiology, Faculty of Physical Therapy, German International University (GIU) 11835 Cairo Egypt
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
- Faculty of Biotechnology, German International University New Administrative Capital 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre 12622 Cairo Egypt
| | - Nabila Hamdi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
| | - Eleni Efthimiado
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens Athens Greece
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical, and Biological Science, School of Life and Medical Sciences, University of Hertfordshire Hatfield AL10 9AB UK
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| |
Collapse
|
35
|
Shibuta T, Takada Y, Nishinosono S, Yasuda S, Ono Y, Hirooka Y, Irikura D, Saito K, Umemura T. Disease-specific signatures of circulating extracellular vesicles detected by the surface plasmon resonance imaging: a pilot study. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:36-53. [PMID: 40206804 PMCID: PMC11977349 DOI: 10.20517/evcna.2024.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/24/2025] [Accepted: 02/06/2025] [Indexed: 04/11/2025]
Abstract
Aim: Cells in the human body release extracellular vesicles (EVs) into fluids, such as plasma, urine, and cerebrospinal fluid. EVs express tetraspanin family proteins (e.g., CD63, CD9, and CD81) and cell-specific antigens on their surface as common and specific markers, respectively. In this study, we hypothesized that the profile of blood cell-derived circulating EVs could reveal both common and specific pathophysiology in atherogenic diseases. Methods: Using surface plasmon resonance imaging (SPRi), we analyzed EVs surface molecules and identified circulating EVs in healthy controls (n = 18), patients with type 2 diabetes mellitus (T2DM; n = 71), and those with hypertension (HT; n = 47). Results: Patients with T2DM and HT exhibited distinct EV profiles: (i) CD9, CD110, CD20, activin receptor type-2A (AcvRIIA), Duffy antigen receptor for chemokine, and CD44 positive EVs were upregulated in T2DM; (ii) CD9, Maackia amurensis agglutinin lectin binding molecules (MBM), CD20, AcvRIIA, and CD44 positive EVs were upregulated in HT. By analyzing an appropriate set of three antigens or using dimensional reduction clustering, we were able to clearly differentiate between T2DM, HT, and control groups. In some patients, disease severity correlated with CD44 and CD20 in T2DM and MBM and AcvRIIA in HT. Conclusion: Our findings demonstrate that profiling of circulating EVs via the SPRi method offers a novel approach for diagnosing and monitoring human diseases.
Collapse
Affiliation(s)
- Tatsuki Shibuta
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka 831-8501, Japan
| | - Yukichi Takada
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka 831-8501, Japan
| | - Shiori Nishinosono
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka 831-8501, Japan
| | - Seiko Yasuda
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka 831-8501, Japan
| | - Yasuhiro Ono
- Department of Diabetes and Metabolism, Kouhoukai Takagi Hospital, Fukuoka 831-0016, Japan
| | - Yoshitaka Hirooka
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka 831-8501, Japan
- Hypertension and Heart Failure Center, Kouhoukai Takagi Hospital, Fukuoka 831-0016, Japan
- Graduate School, International University of Health and Welfare, Fukuoka 831-8501, Japan
| | | | | | - Tsukuru Umemura
- Department of Medical Technology and Sciences, International University of Health and Welfare, Fukuoka 831-8501, Japan
- Graduate School, International University of Health and Welfare, Fukuoka 831-8501, Japan
| |
Collapse
|
36
|
Krishnan I, Ng CY, Kee LT, Ng MH, Law JX, Thangarajah T, Zainuddin AA, Mahmood Z, Rajamanickam S, Subramani B, Lokanathan Y. Quality Control of Fetal Wharton's Jelly Mesenchymal Stem Cells-Derived Small Extracellular Vesicles. Int J Nanomedicine 2025; 20:1807-1820. [PMID: 39963415 PMCID: PMC11830757 DOI: 10.2147/ijn.s497586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Background Quality control (QC) is an important element in ensuring drug substances' safety, efficacy, and quality. The dosing regimen for sEVs can be in the form of protein concentration or the number of particles based on the results of a series of quality controls applied as in-process control. Methods Wharton's Jelly Mesenchymal Stem Cells (WJMSCs) were isolated from four independent umbilical cord samples and were characterized following the International Society for Cellular Therapy (ISCT) guidelines. Small extracellular vesicles (sEVs) were isolated separately from these four WJMSCs samples using the Tangential Flow Filtration (TFF) method and were characterized per Minimal Information for Studies of Extracellular Vesicles (MISEV2018) guidelines. Each isolated and concentrated sEV preparation was standardized and its purity was determined by the ratio of the number of particles to protein concentration. Results All the WJMSCs samples passed the Mesenchymal Stem Cells (MSCs) characterization QC tests. Qualitatively, EVs-positive markers (CD63 and TSG101) and intact bilipid membrane vesicles were detected in all the sEV preparations. Quantitatively, the protein and particle concentrations revealed that all the sEV preparations were "impure" with < 1.5 × 109 particles/µg protein. Albumin was co-isolated in all the sEV preparations. Conclusion In short, all characterized and standardized individual and pooled sEV preparations were deemed "impure" due to albumin co-isolation using the TFF method. For therapeutic development, it is essential to report protein and particle concentrations in EV preparations based on these QC results.
Collapse
Affiliation(s)
- Illayaraja Krishnan
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
| | - Chiew Yong Ng
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
| | - Li Ting Kee
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
| | - Min Hwei Ng
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
| | - Jia Xian Law
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
| | - Thavachelvi Thangarajah
- Department of Obstetrics and Gynaecology, Hospital Angkatan Tentera (HAT) Tuanku Mizan, Kuala Lumpur, Malaysia
| | - Ani Amelia Zainuddin
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
| | - Zalina Mahmood
- Production and Blood Supply Management Division, National Blood Centre, Kuala Lumpur, Malaysia
| | | | | | - Yogeswaran Lokanathan
- Department of Tissue Engineering and Regenerative Medicine (DTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia (UKM), Cheras, Kuala Lumpur, Malaysia
- Advance Bioactive Materials-Cells UKM Research Group, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| |
Collapse
|
37
|
Puspadewi R, Milanda T, Muhaimin M, Chaerunisaa AY. Nanoparticle-Encapsulated Plant Polyphenols and Flavonoids as an Enhanced Delivery System for Anti-Acne Therapy. Pharmaceuticals (Basel) 2025; 18:209. [PMID: 40006023 PMCID: PMC11858878 DOI: 10.3390/ph18020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
This study conducted a literature review by searching for articles related to the treatment of skin infections/wrinkles using nano-delivery systems containing natural compounds. The search was conducted in various databases for articles published in the last 10 years, with strict inclusion and exclusion criteria. Of the 490 articles found, 40 were considered relevant. Acne vulgaris is a common dermatological disorder characterised by inflammation of the sebaceous glands, often resulting in the development of pimples, cysts, and scarring. Conventional treatments, including antibiotics and topical retinoids, frequently demonstrate limitations such as side effects, resistance, and insufficient skin absorption. Recent advancements in nanotechnology have enabled the creation of innovative drug-delivery systems that enhance the effectiveness and reduce the adverse effects of anti-acne medications. Polyphenols and flavonoids, natural bioactive compounds with notable anti-inflammatory, antioxidant, and antibacterial properties, are recognised for their therapeutic effectiveness in acne treatment. However, their practical application is hindered by insufficient solubility, stability, and bioavailability. The incorporation of these compounds into nanoparticle-based delivery systems has shown promise in resolving these challenges. Various nanoparticle platforms, including lipid-based nanoparticles, polymeric nanoparticles, and solid lipid nanoparticles, are evaluated for their ability to improve the stability, controlled release, and targeted delivery of polyphenols and flavonoids to the skin. The advent of polyphenol and flavonoid-loaded nanoparticles marks a new acne therapy era.
Collapse
Affiliation(s)
- Ririn Puspadewi
- Doctoral Program of Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia;
- Faculty of Pharmacy, Jenderal Achmad Yani University, Cimahi 40531, Indonesia
| | - Tiana Milanda
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| | - Muhaimin Muhaimin
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
- Center of Herbal Studies, Padjadjaran University, Sumedang 45363, Indonesia
| | - Anis Yohana Chaerunisaa
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| |
Collapse
|
38
|
Xu Y, Ye Z, Wang Y, Ma Y, Chen X, Wang S, Zhang B, Xia C. Alleviating osteoarthritis-induced damage through extracellular vesicles derived from inflammatory chondrocytes. Int Immunopharmacol 2025; 146:113829. [PMID: 39675196 DOI: 10.1016/j.intimp.2024.113829] [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: 05/22/2024] [Revised: 11/19/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
The role of extracellular vesicles (EVs) derived from inflammatory chondrocytes in EV-based therapy for osteoarthritis (OA) has received little attention. We examined the effects of EVs derived from both normal rat chondrocytes (nEVs) and IL-1β-treated rat chondrocytes (iEVs) on IL-1β-treated rat chondrocytes, macrophages, and osteoblasts, alongside mRNA-seq and miRNA-seq analyses of both them. Additionally, nEVs and iEVs were administered intra-articularly in the joints of rat models subjected to anterior cruciate ligament transection (ACLT), and the morphological alterations across the joints were assessed. These findings indicated that iEVs, compared with nEVs, significantly enhanced collagen II synthesis in IL-1β-treated chondrocytes, accompanied by marked increases in ER stress and autophagy. In comparison to nEVs, iEVs exhibited a greater effect on facilitating M2-type macrophage polarization while simultaneously diminishing M1-type polarization, a process likely mediated by the downregulation of chemotactic cytokines such as Cxcl10, Ccl5, Cxcl9, Cxcl1, and Cxcl11. iEVs exerted a more pronounced influence on the phenotypic characteristics of IL-1β-treated osteoblasts than nEVs. In the ACLT-rat model, iEVs, akin to nEVs, effectively mitigated articular cartilage degradation. However, there was no significant difference in OARSI Scores between the two groups, despite iEVs exerting a greater effect on increasing hyaline cartilage thickness and proteoglycan content. iEVs were superior to nEVs in attenuating synovium inflammation and promoting trabecula formation in the femur subchondral bone. Consequently, iEVs, akin to nEVs, significantly alleviated OA-induced damage. Moreover, iEVs outperformed nEVs in certain aspects, notably in augmenting hyaline cartilage, reducing synovium inflammation, and promoting trabecular formation in the subchondral bone during the early stage of OA.
Collapse
Affiliation(s)
- Yang Xu
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Zesen Ye
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Yue Wang
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Yongkang Ma
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Xiaolei Chen
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China
| | - Shaojie Wang
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China.
| | - Bing Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Chun Xia
- Department of Joint Surgery & Sports Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361004, China.
| |
Collapse
|
39
|
Chen G, Ouyang X, Mu Y, Chen Y. Human breast milk-derived exosomes and their positive role on neonatal intestinal health. Pediatr Res 2025:10.1038/s41390-025-03813-8. [PMID: 39865171 DOI: 10.1038/s41390-025-03813-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 01/28/2025]
Abstract
Although the role of breast milk in promoting neonatal growth and maintaining intestinal homeostasis is well established, underlying mechanisms by which it protects the intestine from damage remain to be elucidated. Human breast milk-derived exosomes (HMDEs) are newly discovered active signaling vesicles with a diameter of 30-150 nm, which are key carriers of biological information exchange between mother and child. In addition, due to their ability to cross the gastrointestinal barrier, low immunogenicity, good biocompatibility and stability, HMDEs play an important role in regulating intestinal barrier integrity in newborns. In addition, HMDEs possess specific properties that are reformable and modifiable, offering promising strategies for the prevention and treatment of neonatal intestinal diseases. However, challenges such as purification, complex content, and quality control hinder their clinical application. This paper provides a comprehensive review of the biogenesis and properties of HMDEs, their isolation and purification, composition, and effects on neonatal intestinal barrier function, and further explores their potential biomedical applications. IMPACT: Breast milk helps maintain intestinal homeostasis in newborns and can prevent diseases, especially necrotizing enterocolitis (NEC). Breast milk contains abundant exosomes, which are important carriers of maternal and infant biological information exchange. Breast milk have the advantages of low immunogenicity, good biocompatibility and good stability, which helps to maintain the integrity of the intestinal barrier. Exosomes can be modified, which is expected to provide a more effective strategy for the prevention and treatment of intestinal diseases.
Collapse
Affiliation(s)
- Gen Chen
- Department of Pediatrics, The First People's Hospital of Chenzhou, Chenzhou, Hunan, 423000, China
| | - Xiangdong Ouyang
- Department of Pediatrics, The First People's Hospital of Chenzhou, Chenzhou, Hunan, 423000, China
| | - Yide Mu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China
| | - Yuqiong Chen
- Department of Pediatrics, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
40
|
Chu CP, Nabity MB. Technical considerations and review of urinary microRNAs as biomarkers for chronic kidney disease in dogs and cats. Vet Clin Pathol 2025. [PMID: 39865558 DOI: 10.1111/vcp.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/11/2024] [Accepted: 12/28/2024] [Indexed: 01/28/2025]
Abstract
MicroRNAs (miRNAs or miRs) are small, non-coding RNAs that play a crucial role in gene regulation, making them potential biomarkers for various diseases. In the field of veterinary medicine, there is a growing interest in exploring the diagnostic and therapeutic potential of miRNAs in kidney diseases affecting dogs and cats. This review focuses on the use of urinary miRNAs as biomarkers for chronic kidney disease (CKD) in these companion animals. We introduce miRNAs, their biogenesis, and their presence in biofluids, particularly within exosomes, and discuss studies investigating miRNAs in kidney tissue and urine. We acknowledge the challenges associated with miRNA studies, including preanalytical factors such as biological variation, sample collection/processing, storage conditions, and experimental design. We highlight the importance of technical considerations, such as sample pooling, sequencing depth, multiplexing, and the various steps of the miRNA experimental workflow. Furthermore, we discuss RNA isolation methods, small RNA sequencing data analysis, and the use of quantitative reverse transcription PCR (qRT-PCR) and droplet digital PCR for verification. We emphasize the importance of internal controls, spike-ins, and normalization methods to minimize technical variation and ensure reliable results in qRT-PCR analysis. This review concludes that while urinary miRNAs hold promise as non-invasive biomarkers for CKD in dogs and cats, addressing the challenges and standardization of protocols is vital for the successful translation of this research into clinical practice.
Collapse
Affiliation(s)
- Candice P Chu
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Mary B Nabity
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
41
|
Chen J, Zheng Y, Wang Z, Gao Q, Hao K, Chen X, Ke N, Lv X, Weng J, Zhong Y, Huang Z, Fu M, Zhao L, Lin F, Mi H, Tang H, Yu C, Huang Y. Development a glycosylated extracellular vesicle-derived miRNA Signature for early detection of esophageal squamous cell carcinoma. BMC Med 2025; 23:39. [PMID: 39849483 PMCID: PMC11755925 DOI: 10.1186/s12916-025-03871-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is often diagnosed at an advanced stage due to the lack of non-invasive early detection tools, which significantly impacts patient prognosis. Given that glycosylation alterations especially high sialylation and fucosylation, frequently occur during cellular malignant transformation, but their roles are not elucidated. We examined alterations in disease-specific glycosylated extracellular vesicles (EVs)-derived miRNAs in the serum of ESCC patients, evaluating their utility as diagnostic biomarkers. METHODS A total of 371 ESCC and 303 healthy controls (HCs) were recruited in this multi-stage, multicentre case-control study. Fucosylated (Fuc-) and sialylated (Sia-) EVs were isolated utilizing Lentil lectin (LCA) and wheat germ lectin (WGA)-coated magnetic beads, respectively. The glycosylated EVs-derived miRNAs-based signature (RiskscoreFuc-&Sia-) was established through logistic regression in a training cohort and subsequently validated in an internal and an external multicentre cohort. RESULTS The RiskscoreFuc-&Sia- effectively identified ESCC across all stages, demonstrating high AUC values in training (0.980), internal validation (0.957), and external multicentre validation (0.973) cohorts, markedly higher than carcinoembryonic antigen (CEA) (AUC = 0.769, training cohort; AUC = 0.749, internal validation cohort; AUC = 0.765, external validation cohort). Notably, this score exhibited robust accuracy in detecting CEA (-) ESCC cases (CEA < 5 ng/ml) (AUC = 0.974, training & internal cohort; AUC = 0.973, external multicentre validation cohort). Additionally, it displayed strong efficacy in differentiating early-stage ESCC patients (AUC = 0.982, training cohort; AUC = 0.977, external multicentre validation cohort). CONCLUSIONS Our study illustrates the effectiveness of glycosylated EVs capture strategy for isolating tumour-specific EVs. The unique glycosylated EVs-derived miRNAs-based signature shows the optimal potential as a biomarker for early detection of ESCC.
Collapse
Affiliation(s)
- Jianlin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yue Zheng
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Zhen Wang
- Department of Clinical Laboratory, Shishi Hospital, Fujian, 362700, Shishi, China
| | - Qi Gao
- Research and development center, Beijing Youngen Technology Co. Ltd, Beijing, 102600, People's Republic of China
| | - Kun Hao
- Research and development center, Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, People's Republic of China
| | - Xiongfeng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Scientific Research, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Nantian Ke
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Xiang Lv
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jiamiao Weng
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yuhong Zhong
- Department of Clinical Laboratory, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, 310009, China
| | - Zhixin Huang
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
- Integrated Chinese and Western Medicine College, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou, 350108, China
| | - Miao Fu
- Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Zhejiang, 321000, Jinhua, China
| | - Lilan Zhao
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Fan Lin
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
- Fujian Provincial Centre for Geriatrics, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Hui Mi
- Departments of Clinical Laboratory, Changzhi People's Hospital, Shanxi, Changzhi, 046000, China
| | - Haijun Tang
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
| | - Chundong Yu
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Centre for Cell Signalling Network, School of Life Sciences, Xiamen University, Fujian, Xiamen, 361102, China.
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- Central Laboratory, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian, Fuzhou, 350001, China.
| |
Collapse
|
42
|
Kong F, Upadya M, Wong ASW, Dalan R, Dao M. Isolating Small Extracellular Vesicles from Small Volumes of Blood Plasma using size exclusion chromatography and density gradient ultracentrifugation: A Comparative Study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.30.564707. [PMID: 37961562 PMCID: PMC10634961 DOI: 10.1101/2023.10.30.564707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Small extracellular vesicles (sEVs) are heterogeneous biological vesicles released by cells under both physiological and pathological conditions. Due to their potential as valuable diagnostic and prognostic biomarkers in human blood, there is a pressing need to develop effective methods for isolating high-purity sEVs from the complex milieu of blood plasma, which contains abundant plasma proteins and lipoproteins. Size exclusion chromatography (SEC) and density gradient ultracentrifugation (DGUC) are two commonly employed isolation techniques that have shown promise in addressing this challenge. In this study, we aimed to determine the optimal combination and sequence of SEC and DGUC for isolating sEVs from small plasma volumes, in order to enhance both the efficiency and purity of the resulting isolates. To achieve this, we compared sEV isolation using two combinations: SEC-DGUC and DGUC-SEC, from unit volumes of 500 μl plasma. Both protocols successfully isolated high-purity sEVs; however, the SEC-DGUC combination yielded higher sEV protein and RNA content. We further characterized the isolated sEVs obtained from the SEC-DGUC protocol using flow cytometry and mass spectrometry to assess their quality and purity. In conclusion, the optimized SEC-DGUC protocol is efficient, highly reproducible, and well-suited for isolating high-purity sEVs from small blood volumes.
Collapse
Affiliation(s)
- Fang Kong
- School of Biological Sciences, Nanyang Technological University, SINGAPORE
| | - Megha Upadya
- School of Biological Sciences, Nanyang Technological University, SINGAPORE
| | - Andrew See Weng Wong
- Facility for Analysis, Characterisation, Testing and Simulation, Nanyang Technological University, SINGAPORE
| | - Rinkoo Dalan
- Lee Kong Chian School of Medicine, Nanyang Technological University, SINGAPORE
| | - Ming Dao
- School of Biological Sciences, Nanyang Technological University, SINGAPORE
- Department of Material Science and Engineering, Massachusetts Institute of Technology, USA
| |
Collapse
|
43
|
Khristov V, Weber SR, Caton-Darby M, Campbell G, Sundstrom JM. Diagnostic and Therapeutic Utility of Extracellular Vesicles in Ocular Disease. Int J Mol Sci 2025; 26:836. [PMID: 39859553 PMCID: PMC11765869 DOI: 10.3390/ijms26020836] [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: 12/01/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer particles released by virtually all cells, with prominent roles in both physiological and pathological processes. The size, number, and molecular composition of released EVs correlate to the cells of origin, modulated by the cell's environment and pathologic state. The proteins, DNA, RNA, and protein cargo carried by EVs are protected by degradation, with a prominent role in targeted intercellular signaling. These properties make EVs salient targets as both carriers of biomarkers and potential therapeutic delivery vehicles. The majority of EV research has focused on blood, urine, saliva, and cerebrospinal fluid due to easy accessibility. EVs have also been identified and studied in all ocular biofluids, including the vitreous humor, the aqueous humor, and the tear film, and the study of EVs in ocular disease is a new, promising, and underexplored direction with unique challenges and considerations. This review covers recent advances in the diagnostic and therapeutic use of ocular EVs, with a focus on human applications and key preceding in vitro and in vivo animal studies. We also discuss future directions based on the study of EVs in other organ systems and disease sates.
Collapse
Affiliation(s)
- Vladimir Khristov
- Penn State Hershey College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (V.K.); (G.C.)
| | - Sarah R. Weber
- Department of Ophthalmology, Penn State University, Hershey, PA 17033, USA; (S.R.W.); (M.C.-D.)
| | - Mireille Caton-Darby
- Department of Ophthalmology, Penn State University, Hershey, PA 17033, USA; (S.R.W.); (M.C.-D.)
| | - Gregory Campbell
- Penn State Hershey College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA; (V.K.); (G.C.)
| | - Jeffrey M. Sundstrom
- Department of Ophthalmology, Penn State University, Hershey, PA 17033, USA; (S.R.W.); (M.C.-D.)
| |
Collapse
|
44
|
Yin X, Yi J, Mao F, Tang Q, Zhang X, Yang X, Xie H, Wang L, Sun S, Yu X, Liu J, Jiang L. Identification of key miRNAs and target genes in extracellular vesicles derived from low-intensity pulsed ultrasound-treated stem cells. Front Genet 2025; 15:1407671. [PMID: 39882071 PMCID: PMC11774887 DOI: 10.3389/fgene.2024.1407671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/18/2024] [Indexed: 01/31/2025] Open
Abstract
Objectives This study aimed to investigate the impact of low-intensity pulsed ultrasound (LIPUS) treatment on the miRNA and mRNA profiles of stem cell-derived extracellular vesicles (EVs). Specifically, it sought to identify key miRNAs and their target mRNAs associated with enhanced therapeutic efficacy in LIPUS-treated stem cell-derived EVs. Methods Utilizing miRNA deep-sequencing data from the Gene Expression Omnibus database, differential gene analysis was performed. MiRNA-mRNA target analysis, functional and pathway enrichment analysis, protein-protein interaction network construction, and hub gene identification were conducted. Validation of differentially expressed miRNAs was performed via RT-qPCR in human umbilical cord mesenchymal stem cells (hUC-MSCs) treated with LIPUS. Results Ten differentially expressed miRNAs were identified, with six upregulated and four downregulated miRNAs in LIPUS-treated stem cell-derived EVs. Functional enrichment analysis revealed involvement in biological processes such as regulation of metabolic processes, cellular component organization, and response to stress, as well as signaling pathways like cell cycle, MAPK signaling, and Hippo signaling. Protein-protein interaction network analysis identified key hub genes including MYC, GAPDH, HSP90AA1, EP300, JUN, PTEN, DAC1, STAT3, HSPA8, and HIF1A associated with LIPUS treatment. RT-qPCR validation confirmed differential expression of selected miRNAs (hsa-miR-933, hsa-miR-3943, hsa-miR-4633-5p, hsa-miR-592, hsa-miR-659-5p, hsa-miR-4766-3p) in LIPUS-treated hUC-MSCs. Conclusion This study sheds light on the potential therapeutic mechanisms underlying LIPUS-treated stem cell-derived EVs. The identified differentially expressed miRNAs and their potential target mRNAs offer valuable insights into the biological processes influenced by LIPUS treatment. While further investigation is necessary to validate their roles as therapeutic targets, this study lays the groundwork for future research on optimizing SC-EV therapy with LIPUS preconditioning.
Collapse
Affiliation(s)
- Xin Yin
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jialian Yi
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Fugang Mao
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Qisheng Tang
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xinyu Zhang
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xiaoyu Yang
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Hongqing Xie
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Linping Wang
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Shuifen Sun
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Xin Yu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Jie Liu
- The First People’s Hospital of Yunnan Province, Kunming, China
| | - Lihong Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
45
|
Vasu S, Johnson V, M A, Reddy KA, Sukumar UK. Circulating Extracellular Vesicles as Promising Biomarkers for Precession Diagnostics: A Perspective on Lung Cancer. ACS Biomater Sci Eng 2025; 11:95-134. [PMID: 39636879 DOI: 10.1021/acsbiomaterials.4c01323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Extracellular vesicles (EVs) have emerged as promising biomarkers in liquid biopsy, owing to their ubiquitous presence in bodily fluids and their ability to carry disease-related cargo. Recognizing their significance in disease diagnosis and treatment, substantial efforts have been dedicated to developing efficient methods for EV isolation, detection, and analysis. EVs, heterogeneous membrane-encapsulated vesicles secreted by all cells, contain bioactive substances capable of modulating recipient cell biology upon internalization, including proteins, lipids, DNA, and various RNAs. Their prevalence across bodily fluids has positioned them as pivotal mediators in physiological and pathological processes, notably in cancer, where they hold potential as straightforward tumor biomarkers. This review offers a comprehensive examination of advanced nanotechnology-based techniques for detecting lung cancer through EV analysis. It begins by providing a brief overview of exosomes and their role in lung cancer progression. Furthermore, this review explores the evolving landscape of EV isolation and cargo analysis, highlighting the importance of characterizing specific biomolecular signatures within EVs for improved diagnostic accuracy in lung cancer patients. Innovative strategies for enhancing the sensitivity and specificity of EV isolation and detection, including the integration of microfluidic platforms and multiplexed biosensing technologies are summarized. The discussion then extends to key challenges associated with EV-based liquid biopsies, such as the standardization of isolation and detection protocols and the establishment of robust analytical platforms for clinical translation. This review highlights the transformative impact of EV-based liquid biopsy in lung cancer diagnosis, heralding a new era of personalized medicine and improved patient care.
Collapse
Affiliation(s)
- Sunil Vasu
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Vinith Johnson
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Archana M
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - K Anki Reddy
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| | - Uday Kumar Sukumar
- Department of Chemical Engineering, Indian Institute of Technology Tirupati, Tirupati, Andhra Pradesh, India-517 619
| |
Collapse
|
46
|
Černoša A, Gostinčar C, Holcar M, Kostanjšek R, Lenassi M, Gunde-Cimerman N. The impact of Aureobasidium melanogenum cells and extracellular vesicles on human cell lines. Sci Rep 2025; 15:1413. [PMID: 39789015 PMCID: PMC11718310 DOI: 10.1038/s41598-024-84189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
Aureobasidium melanogenum is a black yeast-like fungus that occurs frequently both in nature and in domestic environments. It is becoming increasingly important as an opportunistic pathogen. Nevertheless, its effect on human cells has not yet been studied. In this study, we investigated the effect of A. melanogenum cells and extracellular vesicles (EVs) on human cell lines A549 (human lung cells), HDFa (human dermal fibroblasts), and SH-SY5Y (human neuroblastoma cells). Scanning electron microscopy (SEM) showed no direct interaction between A. melanogenum cells and human cell lines, but there were some changes in HDFa cells. As a possible cause for this change, we tested the cytotoxic effect of EVs from A. melanogenum on the same cell lines. We isolated EVs from the fungus and prepared three different pools: a non-melanin pool (containing mainly EVs), a melanin pool (containing mainly melanin nanoparticles), and a total pool (containing both EVs and melanin nanoparticles). All three pools were characterized and then added to human cell lines to test their cytotoxicity. Unlike in some other fungal opportunistic pathogens, no effects of fungal EVs on human cell viability were observed. Therefore, the opportunistic potential of A. melanogenum remains only partially understood.
Collapse
Affiliation(s)
- Anja Černoša
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia.
- InnoRenew CoE, Livade 6a, Izola, 6310, Slovenia.
| | - Cene Gostinčar
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Marija Holcar
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Rok Kostanjšek
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Metka Lenassi
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Nina Gunde-Cimerman
- Department of Biology, Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
47
|
Muzaffer U, Nisar N, Ali SI, Kareem O, Paul V. Immunotoxicogenomics: Moving from observation to prediction. IMMUNOTOXICOGENOMICS 2025:181-206. [DOI: 10.1016/b978-0-443-18502-1.00007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
48
|
Kumar A, Yang E, Du Y. Trabecular Meshwork Regeneration for Glaucoma Treatment Using Stem Cell-Derived Trophic Factors. Methods Mol Biol 2025; 2848:59-71. [PMID: 39240516 PMCID: PMC11971979 DOI: 10.1007/978-1-0716-4087-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Glaucoma is one of the leading causes of irreversible blindness. Stem cell therapy has shown promise in the treatment of primary open-angle glaucoma in animal models. Stem cell-free therapy using stem cell-derived trophic factors might be in demand in patients with high-risk conditions or religious restrictions. In this chapter, we describe methods for trabecular meshwork stem cell (TMSC) cultivation, secretome harvesting, and protein isolation, as well as assays to ensure the health of TMSC post-secretome harvesting and for secretome periocular injection into mice for therapeutic purposes.
Collapse
Affiliation(s)
- Ajay Kumar
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Enzhi Yang
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Yiqin Du
- Department of Ophthalmology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
49
|
Namdari M, McDonnell FS. Extracellular vesicles as emerging players in glaucoma: Mechanisms, biomarkers, and therapeutic targets. Vision Res 2025; 226:108522. [PMID: 39581065 PMCID: PMC11640964 DOI: 10.1016/j.visres.2024.108522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
In recent years, extracellular vesicles (EVs) have attracted significant scientific interest due to their widespread distribution, their potential as disease biomarkers, and their promising applications in therapy. Encapsulated by lipid bilayers these nanovesicles include small extracellular vesicles (sEV) (30-150 nm), microvesicles (100-1000 nm), and apoptotic bodies (100-5000 nm) and are essential for cellular communication, immune responses, biomolecular transport, and physiological regulation. As they reflect the condition and functionality of their originating cells, EVs play critical roles in numerous physiological processes and diseases. Therefore, EVs offer valuable opportunities for uncovering disease mechanisms, enhancing drug delivery systems, and identifying novel biomarkers. In the context of glaucoma, a leading cause of irreversible blindness, the specific roles of EVs are still largely unexplored. This review examines the emerging role of EVs in the pathogenesis of glaucoma, with a focus on their potential as diagnostic biomarkers and therapeutic agents. Through a thorough analysis of current literature, we summarize key advancements in EV research and identify areas where further investigation is needed to fully understand their function in glaucoma.
Collapse
Affiliation(s)
- Maral Namdari
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Pharmacology and Toxicology, University of Utah Salt Lake City, UT, USA.
| |
Collapse
|
50
|
Park M, Lee CH, Noh H, Kang G, Lee J, Bae JH, Moon H, Park J, Kong S, Baek MC, Park H. High-precision extracellular-vesicle isolation-analysis integrated platform for rapid cancer diagnosis directly from blood plasma. Biosens Bioelectron 2025; 267:116863. [PMID: 39442437 DOI: 10.1016/j.bios.2024.116863] [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: 07/17/2024] [Revised: 09/29/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Cancer-derived small extracellular vesicles (sEVs) in body fluids hold promise as biomarkers for cancer diagnosis. For sEV-based liquid biopsy, isolation of sEVs with a high-purity and cancer-sEV detection with an extremely high sensitivity are essential because body fluids include much higher density of normal-cell-derived sEVs and other biomolecules and bioparticles. Here, we propose an isolation-analysis-integrated cancer-diagnosis platform based on dielectrophoresis(DEP)-ELISA technique which enables a three orders of magnitude higher sensitivity over conventional ELISA method and direct cancer diagnosis from blood plasma with high accuracy. The limit of detection (LOD) for sEVs in human plasma was as low as 104 sEVs/mL without a time-consuming and low-yield sEV isolation and purification process. The capability of this platform was validated by monitoring mice with cancer cell inoculation and assessing the effect of cancer-sEV-inhibiting drug. Using the developed sEV-based liquid biopsy, we diagnosed clinical samples from healthy donors (N = 39) and cancer patients (N = 90). The diagnostic accuracy was 94.2%, 98.6%, and 91.3% for breast, colon, and lung cancers, respectively. This integrated sEV isolation and analysis platform could be applied for high-sensitivity biomarker profiling and sEV-based liquid biopsy.
Collapse
Affiliation(s)
- Minsu Park
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea
| | - Chan-Hyeong Lee
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, 41944, Daegu, South Korea
| | - Hyowoong Noh
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea; Digital Biomedical Research Division, Electronics and Telecommunications Research Institute (ETRI) , 34129, Daejeon, South Korea
| | - Geeyoon Kang
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea
| | - Junyeong Lee
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea; Digital Biomedical Research Division, Electronics and Telecommunications Research Institute (ETRI) , 34129, Daejeon, South Korea
| | - Ju-Hyun Bae
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, 41944, Daegu, South Korea
| | - Hyeri Moon
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea
| | - Jonghoo Park
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea
| | - Seongho Kong
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Exosome Convergence Research Center (ECRC), School of Medicine, Kyungpook National University, 41944, Daegu, South Korea.
| | - Hongsik Park
- School of Electronic and Electrical Engineering, Kyungpook National University, 41566, Daegu, South Korea.
| |
Collapse
|