1
|
Lou C, Cai X. The emerging roles of platelet-derived extracellular vesicles in disease. Ann Med 2025; 57:2499029. [PMID: 40317251 PMCID: PMC12054590 DOI: 10.1080/07853890.2025.2499029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Platelet-derived extracellular vesicles (pEVs) are nanoscale, membrane-bound vesicles released by platelets during activation or apoptosis. They contain various bioactive and non-bioactive molecules and play significant roles in numerous physiological and pathological processes through intercellular communication, thus attracting growing attention in biomedical research. METHODS This review comprehensively overviews the biogenesis, clearance, and molecular characteristics of pEVs. It also covers current methodologies for their isolation and characterization. The therapeutic implications of pEVs in key clinical settings like tissue regeneration, hemostasis, immune modulation, and vascular repair, with a focus on cancer progression, wound healing, and hemorrhagic shock management, are explored. Their role in cellular signal transduction is examined, and their functional properties are compared with other platelet-derived products such as platelet-rich plasma. RESULTS pEVs show potential as both therapeutic agents and diagnostic biomarkers. They are involved in modulating inflammatory responses, promoting angiogenesis, and enhancing cellular repair mechanisms. CONCLUSION Future research should concentrate on optimizing their therapeutic efficacy, refining biomarker applications, and exploring targeted delivery strategies to fully utilize their potential in regenerative medicine, oncology, and hemostasis management.
Collapse
Affiliation(s)
- Can Lou
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Blood Transfusion, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Cai
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Blood Transfusion, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Chen J, Li Y, Quan X, Chen J, Han Y, Yang L, Zhou M, Mok GSP, Wang R, Zhao Y. Utilizing engineered extracellular vesicles as delivery vectors in the management of ischemic stroke: a special outlook on mitochondrial delivery. Neural Regen Res 2025; 20:2181-2198. [PMID: 39101653 PMCID: PMC11759020 DOI: 10.4103/nrr.nrr-d-24-00243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 08/06/2024] Open
Abstract
Ischemic stroke is a secondary cause of mortality worldwide, imposing considerable medical and economic burdens on society. Extracellular vesicles, serving as natural nano-carriers for drug delivery, exhibit excellent biocompatibility in vivo and have significant advantages in the management of ischemic stroke. However, the uncertain distribution and rapid clearance of extracellular vesicles impede their delivery efficiency. By utilizing membrane decoration or by encapsulating therapeutic cargo within extracellular vesicles, their delivery efficacy may be greatly improved. Furthermore, previous studies have indicated that microvesicles, a subset of large-sized extracellular vesicles, can transport mitochondria to neighboring cells, thereby aiding in the restoration of mitochondrial function post-ischemic stroke. Small extracellular vesicles have also demonstrated the capability to transfer mitochondrial components, such as proteins or deoxyribonucleic acid, or their sub-components, for extracellular vesicle-based ischemic stroke therapy. In this review, we undertake a comparative analysis of the isolation techniques employed for extracellular vesicles and present an overview of the current dominant extracellular vesicle modification methodologies. Given the complex facets of treating ischemic stroke, we also delineate various extracellular vesicle modification approaches which are suited to different facets of the treatment process. Moreover, given the burgeoning interest in mitochondrial delivery, we delved into the feasibility and existing research findings on the transportation of mitochondrial fractions or intact mitochondria through small extracellular vesicles and microvesicles to offer a fresh perspective on ischemic stroke therapy.
Collapse
Affiliation(s)
- Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Li Yang
- Department of Pharmacy, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Manfei Zhou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Greta Seng Peng Mok
- Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| |
Collapse
|
3
|
Yu L, Shi H, Gao T, Xu W, Qian H, Jiang J, Yang X, Zhang X. Exomeres and supermeres: Current advances and perspectives. Bioact Mater 2025; 50:322-343. [PMID: 40276541 PMCID: PMC12020890 DOI: 10.1016/j.bioactmat.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Recent studies have revealed a great diversity and complexity in extracellular vesicles and particles (EVPs). The developments in techniques and the growing awareness of the particle heterogeneity have spurred active research on new particle subsets. Latest discoveries highlighted unique features and roles of non-vesicular extracellular nanoparticles (NVEPs) as promising biomarkers and targets for diseases. These nanoparticles are distinct from extracellular vesicles (EVs) in terms of their smaller particle sizes and lack of a bilayer membrane structure and they are enriched with diverse bioactive molecules particularly proteins and RNAs, which are widely reported to be delivered and packaged in exosomes. This review is focused on the two recently identified membraneless NVEPs, exomeres and supermeres, to provide an overview of their biogenesis and contents, particularly those bioactive substances linked to their bio-properties. This review also explains the concepts and characteristics of these nanoparticles, to compare them with other EVPs, especially EVs, as well as to discuss their isolation and identification methods, research interests, potential clinical applications and open questions.
Collapse
Affiliation(s)
- Li Yu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Tingxin Gao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
4
|
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
|
5
|
Zununi Vahed S, Hejazian SM, Bakari WN, Landon R, Gueguen V, Meddahi-Pellé A, Anagnostou F, Barzegari A, Pavon-Djavid G. Milking mesenchymal stem cells: Updated protocols for cell lysate, secretome, and exosome extraction, and comparative analysis of their therapeutic potential. Methods 2025; 238:40-60. [PMID: 40058715 DOI: 10.1016/j.ymeth.2025.03.004] [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: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/21/2025] Open
Abstract
The potential of the cell lysate, secretome, and extracellular vesicles (EVs) of mesenchymal stem cells (MSCs) to modulate the immune response and promote tissue regeneration has positioned them as a promising option for cell-free therapy. Currently, many clinical trials in stem cells-derived EVs and secretome are in progress various diseases and sometimes the results are failing. The major challenge on this roadmap is the lack of a standard extraction method for exosome, secretome, and lysate. The most optimal method for obtaining the secretome of MSCs for clinical utilization involves a comprehensive approach that includes non-destructive collection methods, time optimization, multiple collection rounds, optimization of culture conditions, and quality control measures. Further research and clinical studies are warranted to validate and refine these methods for safe and effective utilization of the MSC exosome, secretome, and lysate in various clinical applications. To address these challenges, it is imperative to establish a standardized and unified methodology to ensure reliable evaluation of these extractions in clinical trials. This review seeks to outline the pros and cons of methods for the preparation of MSCs-derived exosome, and secretome/lysate, and comparative analysis of their therapeutic potential.
Collapse
Affiliation(s)
| | | | - William Ndjidda Bakari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France; Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Rebecca Landon
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Fani Anagnostou
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France.
| |
Collapse
|
6
|
Ye Y, Zhang J, Liu Y, Sun X. Migrasome: a novel insight into unraveling physiological and pathological function. Mol Biol Rep 2025; 52:509. [PMID: 40434508 DOI: 10.1007/s11033-025-10615-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: 02/17/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025]
Abstract
The migrasome, a recently discovered organelle formed during cell migration, is a membrane-bound vesicular structure containing numerous smaller intracavitary vesicles and cellular contents. It is generated at the tips and intersections of retraction fibers (RFs). Upon the rupture of RFs, migrasomes can either be engulfed by surrounding cells or undergo lysis to release their contents into the extracellular microenvironment. The process through which cells release their contents via migrasomes is termed migracytosis. Migrasomes play crucial roles in intercellular signaling, cellular homeostasis maintenance, and intercellular material transport. This article provides a comprehensive review of the discovery, biogenesis, isolation and characterization, biological functions of migrasomes, as well as their roles in the occurrence, progression, diagnosis, and treatment of clinical diseases. Furthermore, this paper proposes novel hypotheses and future directions regarding the current research challenges of migrasomes and their potential clinical applications, which may facilitate future clinical diagnosis and treatment involving migrasomes.
Collapse
Affiliation(s)
- Yu Ye
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jun Zhang
- Department of Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Yue Liu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
7
|
Zhang X, Wang Y, E Q, Naveed M, Wang X, Liu Y, Li M. The biological activity and potential of probiotics-derived extracellular vesicles as postbiotics in modulating microbiota-host communication. J Nanobiotechnology 2025; 23:349. [PMID: 40380331 PMCID: PMC12082936 DOI: 10.1186/s12951-025-03435-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Probiotics such as Lactobacillus and Bifidobacterium spp. have been shown to be critical for maintaining host homeostasis. In recent years, key compounds of postbiotics derived from probiotic metabolism and cellular secretion have been identified for their role in maintaining organ immunity and regulating intestinal inflammation. In particular, probiotic-derived extracellular vesicles (PEVs) can act as postbiotics, maintaining almost the same functional activity as probiotics. They also have strong biocompatibility and loading capacity to carry exogenous or parental active molecules to reach distal organs to play their roles. This provides a new direction for understanding the intrinsic microbiota-host communication mechanism. However, most current studies on PEVs are limited to their functional effects/benefits, and their specific physicochemical properties, composition, intrinsic mechanisms for maintaining host homeostasis, and possible threats remain to be explored. Here, we review and summarize the unique physicochemical properties of PEVs and their bioactivities and mechanisms in mediating microbiota-host communication, and elucidate the limitations of the current research on PEVs and their potential application as postbiotics.
Collapse
Affiliation(s)
- Xiaoming Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ye Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiyu E
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Naveed
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
8
|
Yeh H, Gupta K, Lu YH, Srinivasan A, Delila L, Yen NTH, Nyam-Erdene A, Burnouf T. Platelet Extracellular Vesicles as Natural Delivery Vehicles for Mitochondrial Dysfunction Therapy? ACS Biomater Sci Eng 2025; 11:2601-2621. [PMID: 40280866 PMCID: PMC12076291 DOI: 10.1021/acsbiomaterials.5c00473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Mitochondria are vital for energy production, metabolic regulation, and cellular signaling. Their dysfunction is strongly implicated in neurological, cardiovascular, and muscular degenerative diseases, where energy deficits and oxidative stress accelerate disease progression. Platelet extracellular vesicles (PEVs), once called "platelet dust", have emerged as promising agents for mitigating mitochondrial dysfunction. Like other extracellular vesicles (EVs), PEVs carry diverse molecular cargo and surface markers implicated in disease processes and therapeutic efficacy. Notably, they may possibly contain intact or partially functional mitochondrial components, making them tentatively attractive for targeting mitochondrial damage. Although direct research on PEVs-mediated mitochondrial rescue remains limited, current evidence suggests that PEVs can modulate diseases associated with mitochondrial dysfunction and potentially enhance mitochondrial health. This review explores the therapeutic potential of PEVs in neurodegenerative and cardiovascular disorders, highlighting their role in restoring mitochondrial health. By examining recent advancements in PEVs research, we aim to shed light on novel strategies for utilizing PEVs as therapeutic agents. Our goal is to underscore the importance of further fundamental and applied research into PEVs-based interventions, as innovative tools for combating a wide range of diseases linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hsien
Chang Yeh
- School
of Medicine, College of Medicine, Taipei
Medical University, Xin-Yi
Campus, Taipei City 110, Taiwan
| | - Kirti Gupta
- International
Graduate Program in Medicine, College of Medicine, Taipei Medical University, Xin-Yi Campus, Taipei 110, Taiwan
| | - Ya-Hsuan Lu
- School
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
| | - Abinaya Srinivasan
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei
City 110, Taiwan
| | - Liling Delila
- Graduate
Institute of Biomedical Materials and Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
| | - Nguyen Tran Hai Yen
- Graduate
Institute of Biomedical Materials and Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
| | - Ariunjargal Nyam-Erdene
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei
City 110, Taiwan
| | - Thierry Burnouf
- International
PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Shuang-Ho Campus, New Taipei
City 110, Taiwan
- Graduate
Institute of Biomedical Materials and Tissue Engineering, College
of Biomedical Engineering, Taipei Medical
University, Shuang-Ho
Campus, New Taipei City 110, Taiwan
- International
PhD Program in Cell Therapy and Regeneration Medicine, College of
Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
9
|
Suresh PS, Zhang Q. Comprehensive Comparison of Methods for Isolation of Extracellular Vesicles from Human Plasma. J Proteome Res 2025. [PMID: 40356199 DOI: 10.1021/acs.jproteome.5c00149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Extracellular vesicles (EVs) are a vital component in cell-cell communication and hold significant potential as biomarkers and therapeutic carriers. Having a reproducible and simple EV isolation method for small volumes of human plasma is essential for biomarker discovery. Although combining multiple methods has been a recent trend in its ability to minimize contamination, it is not ideal for clinical specimens due to the large sample number and small sample volume. This study compared EVs isolated from 100 μL of plasma by nine commonly used methods based on different principles, including centrifugation, polymer precipitation, size exclusion, electrostatic interaction, and affinity enrichment. The isolated EVs were characterized by particle size and number using nanoparticle tracking analysis, purity, and contaminants using Simple Western and overall proteomic profiles using bottom-up proteomics. Despite the same EV enrichment principle, individual methods isolated EVs exhibited distinct characteristics, likely due to variations in the physicochemical properties of materials used and specific protocols. Overall, all of the methods evaluated are reproducible. MagNet and MagCap methods result in purer EVs with the narrowest size distribution and the highest proteome coverage but modest yield. This is the first report on isolating EVs from 100 μL of plasma using nine different methods with detailed characterization.
Collapse
Affiliation(s)
- Patil Shivprasad Suresh
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
| | - Qibin Zhang
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
- Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27402, United States
| |
Collapse
|
10
|
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
|
11
|
Sun J, Li Z, Chen Y, Chang Y, Yang M, Zhong W. Enhancing Analysis of Extracellular Vesicles by Microfluidics. Anal Chem 2025; 97:6922-6937. [PMID: 40133233 DOI: 10.1021/acs.analchem.4c07016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Extracellular vesicles (EVs) play crucial roles in intercellular communication and hold great promise as biomarkers for noninvasive disease diagnosis. Intensive research efforts have been devoted to discovering the EV subpopulations responsible for specific functions or with enhanced effectiveness as disease markers, through extensive EV purification and content analysis. However, their high heterogeneity in size and cargo composition poses significant challenges for reaching such goals. Isolation methods like ultracentrifugation and size-exclusion chromatography, as well as content analysis approaches like polymerase chain reaction and enzyme-linked immunosorbent assay, have made significant contributions to improving our understanding of EV biology. Nonetheless, these methods face limitations in isolation efficiency, EV purity, and detection sensitivity and specificity due to issues like large sample consumption, unsatisfactory purity, and insufficient resolution in EV subtyping. Microfluidic technology presents promising solutions to these challenges, leveraging their intrinsic capabilities in precise flow and external energy field manipulation, sample compartmentalization, and signal enhancement at the micro- and nanoscale. Hence, this review summarizes the recent developments in microfluidics-enabled EV analysis, paying special attention to the unique microfluidic features exploited. Strategies such as viscoelastic and inertial flow, fluid mixing, and external-field-assisted approaches in improving EV purification, as well as compartmentalization and micro/nanostructures for enhancing EV detection, are examined. Furthermore, the current limitations and potential future directions are discussed to inspire advancements in this rapidly developing field.
Collapse
Affiliation(s)
- Jiayu Sun
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518057, P. R. China
| | | | | | | | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen 518057, P. R. China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen 518057, P. R. China
| | | |
Collapse
|
12
|
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: 8] [Impact Index Per Article: 8.0] [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
|
13
|
Zheng B, Wang X, Guo M, Tzeng CM. Current Development of Mesenchymal Stem Cell-Derived Extracellular Vesicles. Cell Transplant 2025; 34:9636897241297623. [PMID: 39874070 PMCID: PMC11775985 DOI: 10.1177/09636897241297623] [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: 06/22/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 01/30/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal. They play a critical role in cell therapy due to their powerful immunomodulatory and regenerative effects. Recent studies suggest that one of the key therapeutic mechanisms of MSCs seems to derive from their paracrine product, called extracellular vesicles (EVs). The EVs contain much DNA, messenger RNA (mRNA), microRNA, and protein components, which can exert intracellular communication to target cells. In clinical applications, the MSC-EVs have been widely used in tissue repair and immune disorder diseases. However, there are serval issues need to be considered such as how to accomplish the large-scale production of EVs and how to verify the exact mechanism of EVs. In this review, we summarize the current progress of MSC-EVs and discuss the challenges and future of MSC-EVs.
Collapse
Affiliation(s)
- Bingyi Zheng
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xueting Wang
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Meizhai Guo
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chi-Meng Tzeng
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
14
|
Qiu M, Ye C, Zhao X, Zou C, Tang R, Xie J, Liu Y, Hu Y, Hu X, Zhang N, Fu Y, Wang J, Zhao C. Succinate exacerbates mastitis in mice via extracellular vesicles derived from the gut microbiota: a potential new mechanism for mastitis. J Nanobiotechnology 2024; 22:712. [PMID: 39543623 PMCID: PMC11566393 DOI: 10.1186/s12951-024-02997-1] [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: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND A high grain diet causes an ecological imbalance in the gut microbiota and serves as an important endogenous trigger of mastitis in dairy cows, but the underlying mechanisms are unclear. Our previous study revealed that subacute rumen acidosis (SARA)-associated mastitis has distinct metabolic profiles in the rumen, especially a significant increase in succinate, but the role of succinate in the pathogenesis of mastitis remains unclear. RESULTS Succinate treatment exacerbates low-grade endotoxemia-induced mastitis in mice. Specifically, succinate increased the production of gut microbiota-extracellular vehicles (mEVs) containing lipopolysaccharides, which can diffuse across the damaged intestinal barrier into the mammary glands. Administration of mEVs promotes mammary inflammation via activation of the TLR4/NF-κB pathway. CONCLUSIONS Our findings suggest that succinate promotes mastitis through the proliferation of enteric pathogens and mEVs production, suggesting a potential strategy for mastitis intervention on the basis of intestinal metabolic regulation and pathogen inhibition. The role of mEVs in interspecific communication has also been elucidated.
Collapse
Affiliation(s)
- Min Qiu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Cong Ye
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Xiaotong Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Chenyu Zou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Ruibo Tang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Jiaxin Xie
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yiheng Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yubo Hu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| | - Jun Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, 130118, China.
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
15
|
Santos P, Rezende CP, Piraine R, Oliveira B, Ferreira FB, Carvalho VS, Calado RT, Pellegrini M, Almeida F. Extracellular vesicles from human breast cancer-resistant cells promote acquired drug resistance and pro-inflammatory macrophage response. Front Immunol 2024; 15:1468229. [PMID: 39474419 PMCID: PMC11518763 DOI: 10.3389/fimmu.2024.1468229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/30/2024] [Indexed: 05/01/2025] Open
Abstract
Introduction Breast cancer is a significant public health problem around the world, ranking first in deaths due to cancer in females. The therapy to fight breast cancer involves different methods, including conventional chemotherapy. However, the acquired resistance that tumors develop during the treatment is still a central cause of cancer-associated deaths. One mechanism that induces drug resistance is cell communication via extracellular vesicles (EVs), which can carry efflux transporters and miRNA that increase sensitive cells' survivability to chemotherapy. Methods Our study investigates the transcription changes modulated by EVs from tamoxifen- and doxorubicin-resistant breast cancer cells in sensitive cells and how these changes may induce acquired drug resistance, inhibit apoptosis, and increase survivability in the sensitive cells. Additionally, we exposed human macrophages to resistant EVs to understand the influence of EVs on immune responses. Results Our results suggest that the acquired drug resistance is associated with the ability of resistant EVs to upregulate several transporter classes, which are directly related to the increase of cell viability and survival of sensitive cells exposed to EVs before a low-dose drug treatment. In addition, we show evidence that resistant EVs may downregulate immune system factors to evade detection and block cell death by apoptosis in sensitive breast cancer cells. Our data also reveals that human macrophages in contact with resistant EVs trigger a pro-inflammatory cytokine secretion profile, an effect that may be helpful for future immunotherapy studies. Discussion These findings are the first transcriptome-wide analysis of cells exposed to resistant EVs, supporting that resistant EVs are associated with the acquired drug resistance process during chemotherapy by modulating different aspects of sensitive cancer cells that coffer the chemoresistance.
Collapse
Affiliation(s)
- Patrick Santos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Caroline P. Rezende
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Renan Piraine
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Bianca Oliveira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francielle B. Ferreira
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Vinicius S. Carvalho
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rodrigo T. Calado
- Department of Medical Imaging, Hematology, and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, CA, United States
| | - Fausto Almeida
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
16
|
Kapoor KS, Harris K, Arian KA, Ma L, Schueng Zancanela B, Church KA, McAndrews KM, Kalluri R. High throughput and rapid isolation of extracellular vesicles and exosomes with purity using size exclusion liquid chromatography. Bioact Mater 2024; 40:683-695. [PMID: 39290685 PMCID: PMC11407901 DOI: 10.1016/j.bioactmat.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/19/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as potential biomarkers for diagnosing a range of diseases without invasive procedures. Extracellular vesicles also offer advantages compared to synthetic vesicles for delivery of various drugs; however, limitations in segregating EVs from other particles and soluble proteins have led to inconsistent EV retrieval rates with low levels of purity. Here, we report a new high-yield (88.47 %) and rapid (<20 min) EV isolation method termed size exclusion - fast protein liquid chromatography (SE-FPLC). We show SE-FPLC can effectively isolate EVs from multiple sources including EVs derived from human and mouse cells and serum samples. The results indicate that SE-FPLC can successfully remove highly abundant protein contaminants such as albumin and lipoprotein complexes, which can represent a major hurdle in large scale isolation of EVs. The high-yield nature of SE-FPLC allows for easy industrial scaling up of EV production for various clinical utilities. SE-FPLC also enables analysis of small volumes of blood for use in point-of-care diagnostics in the clinic. Collectively, SE-FPLC offers many advantages over current EV isolation methods and offers rapid clinical translation.
Collapse
Affiliation(s)
- Kshipra S Kapoor
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, USA
| | - Kristen Harris
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kent A Arian
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lihua Ma
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Beatriz Schueng Zancanela
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kaira A Church
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kathleen M McAndrews
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
17
|
Humaira, Ahmad I, Shakir HA, Khan M, Franco M, Irfan M. Bacterial Extracellular Vesicles: Potential Therapeutic Applications, Challenges, and Future Prospects. J Basic Microbiol 2024; 64:e2400221. [PMID: 39148315 DOI: 10.1002/jobm.202400221] [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: 04/16/2024] [Revised: 07/14/2024] [Accepted: 07/28/2024] [Indexed: 08/17/2024]
Abstract
Almost all cell types naturally secret extracellular vesicles (EVs) in the extracellular space with variable metabolic cargo facilitating intracellular communication, posing immune-modulation capacity. Thus, "bacterial extracellular vesicles" (BEVs), with their great immunoregulatory, immune response stimulation and disease condition-altering potential, have gained importance in the medical and therapeutic industry. Various subtypes of BEVs were observed and reported in the literature, such as exosomes (30-150 nm), microvesicles (100-1000 nm), apoptotic bodies (1000-5000 nm), and oncosomes (1000-10,000 nm). As biological systems are complex entities, inserting BEVs requires extra high purity. Various techniques for BEV isolation have been employed alone or with other strategies, such as ultracentrifugation, precipitation, size-exclusion chromatography, affinity-based separation, ultrafiltration, and field-flow fractionation. But to date, no BEV isolation method is considered perfect as the lack of standard protocols limits their scale-up. Medical research has focused on BEVs to explore their diverse therapeutic potential. This review particularly focused on the recent advancements in the potential medical application of BEVs, current challenges, and prospects associated with their scale-up.
Collapse
Affiliation(s)
- Humaira
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Hafiz Abdullah Shakir
- Institute of Zoology, Faculty of Life Science, University of the Punjab New Campus, Lahore, Pakistan
| | - Muhammad Khan
- Institute of Zoology, Faculty of Life Science, University of the Punjab New Campus, Lahore, Pakistan
| | - Marcelo Franco
- Department of Exact Science, State University of Santa Cruz, Ilheus, Brazil
| | - Muhammad Irfan
- Department of Biotechnology, University of Sargodha, Sargodha, Pakistan
| |
Collapse
|
18
|
Nanru P. Immunomodulatory effects of immune cell-derived extracellular vesicles in melanoma. Front Immunol 2024; 15:1442573. [PMID: 39391320 PMCID: PMC11464304 DOI: 10.3389/fimmu.2024.1442573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
Melanoma, recognized as one of the most immunogenic malignancies in humans, holds paramount significance in the realm of immunotherapy. However, the emergence of drug resistance and the occurrence of adverse drug reactions underscore the pressing need to explore increasingly personalized immunotherapeutic modalities. Extracellular Vesicles (EVs), pivotal derivatives of immune cells, assume pivotal roles by encapsulating proteins, lipids, and nucleic acids within bilayer lipid structures, thereby facilitating targeted delivery to other immune cells. This orchestrated process orchestrates critical functions including antigen presentation, immune modulation, and the induction of apoptosis in tumor cells. A burgeoning body of evidence underscores the vast therapeutic potential of EVs in melanoma treatment. This comprehensive review aims to delineate the roles of EVs derived from immune cells such as dendritic cells, natural killer cells, macrophages, and T cells in the context of melanoma patients, thereby furnishing invaluable insights for the future direction of melanoma immunotherapy.
Collapse
Affiliation(s)
- Peng Nanru
- Department of Plastic Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou
University, Zhengzhou, China
| |
Collapse
|
19
|
Di Bella MA, Taverna S. Extracellular Vesicles: Diagnostic and Therapeutic Applications in Cancer. BIOLOGY 2024; 13:716. [PMID: 39336143 PMCID: PMC11446462 DOI: 10.3390/biology13090716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024]
Abstract
In recent years, knowledge of cell-released extracellular vesicle (EV) functions has undergone rapid growth. EVs are membrane vesicles loaded with proteins, nucleic acids, lipids, and bioactive molecules. Once released into the extracellular space, EVs are delivered to target cells that may go through modifications in physiological or pathological conditions. EVs are nano shuttles with a crucial role in promoting short- and long-distance cell-cell communication. Comprehension of the mechanism that regulates this process is a benefit for both medicine and basic science. Currently, EVs attract immense interest in precision and nanomedicine for their potential use in diagnosis, prognosis, and therapies. This review reports the latest advances in EV studies, focusing on the nature and features of EVs and on conventional and emerging methodologies used for their separation, characterization, and visualization. By searching an extended portion of the relevant literature, this work aims to give a summary of advances in nanomedical applications of EVs. Moreover, concerns that require further studies before translation to clinical applications are discussed.
Collapse
Affiliation(s)
- Maria Antonietta Di Bella
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, 90133 Palermo, Italy
| | - Simona Taverna
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 90146 Palermo, Italy
| |
Collapse
|
20
|
Zhu T, Fan B, He Z, Fan Z, He B, Miao W, Huang R. Self-Assembled DNA Microflowers for Platelet-Derived Extracellular Vesicle Isolation and Infected Wound Healing. Biomacromolecules 2024; 25:4956-4964. [PMID: 38985329 DOI: 10.1021/acs.biomac.4c00398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Platelet-derived extracellular vesicles (PEVs) showing great potential in wound healing have attracted increasing attention recently. Nondestructive isolation and effective utilization strategies are highly conducive for PEVs developing into recognized therapeutic entities. Here, we present an efficient strategy for PEV isolation and bacterial infected wound healing based on self-assembled DNA microflowers. First, DNA microflowers are prepared using rolling circle amplification. Then, the hydrophobic interaction between cholesteryl modified on DNA microflowers and the phospholipid bilayer membrane of PEVs leads to the formation of a network structure with improved mechanical strength and the separation of PEVs from biological samples. Finally, controlled release of PEVs is achieved through bacterial-induced hydrogel degradation. In vitro experiments demonstrate the obtained DNA hydrogel with good cytocompatibility and therapeutic potential. Taken together, the DNA microflower-based hydrogels with bioadhesive, self-healing, tunable mechanical properties and bacteria-responsive behavior offer substantial potential for EV isolation and wound healing.
Collapse
Affiliation(s)
- Tong Zhu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Boyue Fan
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, P. R. China
| | - Ziju He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Zeyu Fan
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, P. R. China
| | - Wenjun Miao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, P. R. China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, P. R. China
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, P. R. China
| |
Collapse
|
21
|
Xu F, Luo S, Lu P, Cai C, Li W, Li C. Composition, functions, and applications of exosomal membrane proteins. Front Immunol 2024; 15:1408415. [PMID: 39148736 PMCID: PMC11324478 DOI: 10.3389/fimmu.2024.1408415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Exosomes play a crucial role in various biological processes, such as human development, immune responses, and disease occurrence. The membrane proteins on exosomes are pivotal factors for their biological functionality. Currently, numerous membrane proteins have been identified on exosome membranes, participating in intercellular communication, mediating target cell recognition, and regulating immune processes. Furthermore, membrane proteins from exosomes derived from cancer cells can serve as relevant biomarkers for early cancer diagnosis. This article provides a comprehensive review of the composition of exosome membrane proteins and their diverse functions in the organism's biological processes. Through in-depth exploration of exosome membrane proteins, it is expected to offer essential foundations for the future development of novel biomedical diagnostics and therapies.
Collapse
Affiliation(s)
- Fang Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Shumin Luo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Pengpeng Lu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chao Cai
- Integrated Chinese and Western Medicine Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Weihua Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Integrated Chinese and Western Medicine Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chuanyun Li
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Kowalczyk A, Kordan W. Evaluation of the effectiveness of the use of exosomes in the regulation of the mitochondrial membrane potential of frozen/thawed spermatozoa. PLoS One 2024; 19:e0303479. [PMID: 38959270 PMCID: PMC11221688 DOI: 10.1371/journal.pone.0303479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/25/2024] [Indexed: 07/05/2024] Open
Abstract
Numerous studies confirm the involvement of extracellular vesicles (EVs) in the regulation of physiological processes of mammalian sperm cells. It has been proven that they take part in the processes of capacitation, acrosonmal reaction, and anti-oxidation. Despite growing interest in the biomedical potential (including the search for new reproductive biomarkers) of EVs, the role of extracellular seminal vesicles in maintaining semen quality during cryopreservation has not yet been established. Therefore, the objective of this experiment was to evaluate the effectiveness of the use in the regulation of the mitochondrial membrane potential of bovine sperm and to explain the mechanisms of EV action during cell cryopreservation. Exosomes were isolated from bull semen plasma, measured, and used for extender supplementation. Semen samples were collected from Simmental bulls, diluted, and pre-evaluated. Then they were divided into equal fractions that did not contain EVs or were supplemented with 0.75; 1.5 and 2.25 mg/ml of EVs. The test samples were frozen/thawed and the mitochondrial membrane potential, DNA integrity, and viability were evaluated. EVs have been established to have a positive effect on cryopreserved sperm structures. The most favourable level of EVs was 1.5 mg / ml, which can be successfully to improve cell cryostability during freezing/thawing. In this study, exosomes isolated from the sperm plasma and supplemented with a concentrated dose in the extender for sperm freezing were shown to significantly improve cryostability of cells by supporting the potentials of the mitochondrial membrane and protecting the cytoplasmic membrane of spermatozoa.
Collapse
Affiliation(s)
- Alicja Kowalczyk
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Władysław Kordan
- Department of Animal Biochemistry and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
23
|
Deng S, Wu Y, Huang S, Yang X. Novel insights into the roles of migrasome in cancer. Discov Oncol 2024; 15:166. [PMID: 38748047 PMCID: PMC11096295 DOI: 10.1007/s12672-024-00942-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 03/18/2024] [Indexed: 05/18/2024] Open
Abstract
Cell migration, a hallmark of cancer malignancy, plays a critical role in cancers. Improperly initiated or misdirected cell migration can lead to invasive metastatic cancer. Migrasomes are newly discovered vesicular cellular organelles produced by migrating cells and depending on cell migration. Four marker proteins [NDST1 (bifunctionalheparan sulfate N-deacetylase/N-sulfotransferase 1), EOGT (Epidermal growth factor domains pecific O-linked N-acetylglucosaminetransferase), CPQ (carboxypeptidase Q), and PIGK (phosphatidylinositol glycan anchor biosynthesis, class K)] of migrasomes were successfully identified. There are three marker proteins (NDST1, PIGK, and EOGT) of migrasome expressed in cancer. In this review, we will discuss the process of migrasome discovery, the formation of migrasome, the possible functions of migrasome, and the differences between migrasomes and exosomes, especially, the biological functions of migrasome marker proteins in cancer, and discuss some possible roles of migrasomes in cancer. We speculate that migrasomes and migracytosis can play key roles in regulating the development of cancer.
Collapse
Affiliation(s)
- Sijun Deng
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang , 421001, Hunan, People's Republic of China
| | - Yiwen Wu
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang , 421001, Hunan, People's Republic of China
| | - Sheng Huang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang , 421001, Hunan, People's Republic of China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang , 421001, Hunan, People's Republic of China.
| |
Collapse
|
24
|
Jiang Y, Lv H, Shen F, Fan L, Zhang H, Huang Y, Liu J, Wang D, Pan H, Yang J. Strategies in product engineering of mesenchymal stem cell-derived exosomes: unveiling the mechanisms underpinning the promotive effects of mesenchymal stem cell-derived exosomes. Front Bioeng Biotechnol 2024; 12:1363780. [PMID: 38756412 PMCID: PMC11096451 DOI: 10.3389/fbioe.2024.1363780] [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: 12/31/2023] [Accepted: 04/08/2024] [Indexed: 05/18/2024] Open
Abstract
Articular cartilage injuries present a significant global challenge, particularly in the aging population. These injuries not only restrict movement due to primary damage but also exacerbate elderly degenerative lesions, leading to secondary cartilage injury and osteoarthritis. Addressing osteoarthritis and cartilage damage involves overcoming several technical challenges in biological treatment. The use of induced mesenchymal stem cells (iMSCs) with functional gene modifications emerges as a solution, providing a more stable and controllable source of Mesenchymal Stem Cells (MSCs) with reduced heterogeneity. Furthermore, In addition, this review encompasses strategies aimed at enhancing exosome efficacy, comprising the cultivation of MSCs in three-dimensional matrices, augmentation of functional constituents within MSC-derived exosomes, and modification of their surface characteristics. Finally, we delve into the mechanisms through which MSC-exosomes, sourced from diverse tissues, thwart osteoarthritis (OA) progression and facilitate cartilage repair. This review lays a foundational framework for engineering iMSC-exosomes treatment of patients suffering from osteoarthritis and articular cartilage injuries, highlighting cutting-edge research and potential therapeutic pathways.
Collapse
Affiliation(s)
- Yudong Jiang
- Orthopedics Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanning Lv
- Orthopedics Department, Longgang District People’s Hospital of Shenzhen and the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuguo Shen
- Orthopedics Department, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Lei Fan
- Orthopedics Department, Longgang District People’s Hospital of Shenzhen and the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, China
| | - Hongjun Zhang
- Orthopedics Department, Longgang District People’s Hospital of Shenzhen and the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, China
| | - Yong Huang
- Orthopedics Department, Longgang District People’s Hospital of Shenzhen and the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, China
| | - Jia Liu
- Central Laboratory, Longgang District People’s Hospital of Shenzhen and the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, China
| | - Dong Wang
- The Biomechanics Group, Department of Mechanical Engineering, Imperial College London, London, United Kingdom
- Department of Engineering, Faculty of Environment, Science and Economy, University of Exeter, Exeter, United Kingdom
| | - Haile Pan
- Orthopedics Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianhua Yang
- Orthopedics Department, Longgang District People’s Hospital of Shenzhen and the Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
25
|
Chen S, Pan J, Gong Z, Wu M, Zhang X, Chen H, Yang D, Qi S, Peng Y, Shen J. Hypochlorous acid derived from microglial myeloperoxidase could mediate high-mobility group box 1 release from neurons to amplify brain damage in cerebral ischemia-reperfusion injury. J Neuroinflammation 2024; 21:70. [PMID: 38515139 PMCID: PMC10958922 DOI: 10.1186/s12974-023-02991-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 12/11/2023] [Indexed: 03/23/2024] Open
Abstract
Myeloperoxidase (MPO) plays critical role in the pathology of cerebral ischemia-reperfusion (I/R) injury via producing hypochlorous acid (HOCl) and inducing oxidative modification of proteins. High-mobility group box 1 (HMGB1) oxidation, particularly disulfide HMGB1 formation, facilitates the secretion and release of HMGB1 and activates neuroinflammation, aggravating cerebral I/R injury. However, the cellular sources of MPO/HOCl in ischemic brain injury are unclear yet. Whether HOCl could promote HMGB1 secretion and release remains unknown. In the present study, we investigated the roles of microglia-derived MPO/HOCl in mediating HMGB1 translocation and secretion, and aggravating the brain damage and blood-brain barrier (BBB) disruption in cerebral I/R injury. In vitro, under the co-culture conditions with microglia BV cells but not the single culture conditions, oxygen-glucose deprivation/reoxygenation (OGD/R) significantly increased MPO/HOCl expression in PC12 cells. After the cells were exposed to OGD/R, MPO-containing exosomes derived from BV2 cells were released and transferred to PC12 cells, increasing MPO/HOCl in the PC12 cells. The HOCl promoted disulfide HMGB1 translocation and secretion and aggravated OGD/R-induced apoptosis. In vivo, SD rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) plus different periods of reperfusion. Increased MPO/HOCl production was observed at the reperfusion stage, accomplished with enlarged infarct volume, aggravated BBB disruption and neurological dysfunctions. Treatment of MPO inhibitor 4-aminobenzoic acid hydrazide (4-ABAH) and HOCl scavenger taurine reversed those changes. HOCl was colocalized with cytoplasm transferred HMGB1, which was blocked by taurine in rat I/R-injured brain. We finally performed a clinical investigation and found that plasma HOCl concentration was positively correlated with infarct volume and neurological deficit scores in ischemic stroke patients. Taken together, we conclude that ischemia/hypoxia could activate microglia to release MPO-containing exosomes that transfer MPO to adjacent cells for HOCl production; Subsequently, the production of HOCl could mediate the translocation and secretion of disulfide HMGB1 that aggravates cerebral I/R injury. Furthermore, plasma HOCl level could be a novel biomarker for indexing brain damage in ischemic stroke patients.
Collapse
Affiliation(s)
- Shuang Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jingrui Pan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhe Gong
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiaoni Zhang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Hansen Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Dan Yang
- Department of Chemistry, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Suhua Qi
- Medical and Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China.
| | - Ying Peng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Jiangang Shen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Medical and Technology School, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
26
|
Wang S, Xia K, Zhu X, Liu Y, Sun L, Zhu Q. Separation of high-purity plasma extracellular vesicles for investigating proteomic signatures in diabetic retinopathy. J Chromatogr A 2024; 1718:464700. [PMID: 38354507 DOI: 10.1016/j.chroma.2024.464700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/16/2024]
Abstract
Extracellular vesicles (EVs) play a multifaceted role in intercellular communication and hold significant promise as bio-functional indicators for clinical diagnosis. Although plasma samples represent one of the most critical sources of circulating EVs, the existing technical challenges associated with plasma-EV isolation have restricted their application in disease diagnosis and biomarker discovery. In this study, we introduce a two-step purification method utilizing ultracentrifugation (UC) to isolate crude extracellular vesicle (EV) samples, followed by a phospholipid affinity-based technique for the selective isolation of small EVs, ensuring a high level of purity for downstream proteomic analysis. Our research demonstrates that the UC & TiO2-coated magnetic bead (TiMB) purification system significantly improves the purity of EVs when compared to conventional UC or TiMB along. We further revealed that proteomic alterations in plasma EVs effectively reflect key gene ontology components associated with diabetic retinopathy (DR) pathogenesis, including the VEGF-activated neuropilin pathway, positive regulation of angiogenesis, angiogenesis, cellular response to vascular endothelial growth factor stimulus, and immune response. By employing a comprehensive analytical approach, which incorporates both time-series analysis (cluster analysis) and differential analysis, we have identified three potential protein signatures including LGALS3, MYH10, and CPB2 that closely associated with the retinopathy process. These proteins exhibit promising diagnostic and severity-classification capabilities for DR disease. This adaptable EV isolation system can be regarded as an effective analytical tool for enhancing plasma-based liquid biopsies toward clinical applications.
Collapse
Affiliation(s)
- Siyao Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Kangfu Xia
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei China
| | - Xinxi Zhu
- Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuhan Liu
- Department of Laboratory Medicine, The Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lei Sun
- Department of Clinical Laboratory, Lu'an Hospital of Anhui Medical University, Lu'an People's Hospital of Anhui Province, Lu'an, 237005, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Qingfu Zhu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
27
|
Naquin TD, Canning AJ, Gu Y, Chen J, Naquin CM, Xia J, Lu B, Yang S, Koroza A, Lin K, Wang HN, Jeck WR, Lee LP, Vo-Dinh T, Huang TJ. Acoustic separation and concentration of exosomes for nucleotide detection: ASCENDx. SCIENCE ADVANCES 2024; 10:eadm8597. [PMID: 38457504 PMCID: PMC10923504 DOI: 10.1126/sciadv.adm8597] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/05/2024] [Indexed: 03/10/2024]
Abstract
Efficient isolation and analysis of exosomal biomarkers hold transformative potential in biomedical applications. However, current methods are prone to contamination and require costly consumables, expensive equipment, and skilled personnel. Here, we introduce an innovative spaceship-like disc that allows Acoustic Separation and Concentration of Exosomes and Nucleotide Detection: ASCENDx. We created ASCENDx to use acoustically driven disc rotation on a spinning droplet to generate swift separation and concentration of exosomes from patient plasma samples. Integrated plasmonic nanostars on the ASCENDx disc enable label-free detection of enriched exosomes via surface-enhanced Raman scattering. Direct detection of circulating exosomal microRNA biomarkers from patient plasma samples by the ASCENDx platform facilitated a diagnostic assay for colorectal cancer with 95.8% sensitivity and 100% specificity. ASCENDx overcomes existing limitations in exosome-based molecular diagnostics and holds a powerful position for future biomedical research, precision medicine, and point-of-care medical diagnostics.
Collapse
Affiliation(s)
- Ty D. Naquin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Aidan J. Canning
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Yuyang Gu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Jianing Chen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Chloe M. Naquin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Jianping Xia
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Brandon Lu
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Shujie Yang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Aleksandra Koroza
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Katherine Lin
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| | - Hsin-Neng Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - William R. Jeck
- Department of Pathology, Duke University Medical Center, Durham, NC 27708, USA
| | - Luke P. Lee
- Harvard Medical School, Harvard University; Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Bioengineering and Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Tony Jun Huang
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27708, USA
| |
Collapse
|
28
|
Wang W, Sun H, Duan H, Sheng G, Tian N, Liu D, Sun Z. Isolation and usage of exosomes in central nervous system diseases. CNS Neurosci Ther 2024; 30:e14677. [PMID: 38497529 PMCID: PMC10945885 DOI: 10.1111/cns.14677] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/10/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND Exosomes are vesicles secreted by all types of mammalian cells. They are characterized by a double-layered lipid membrane structure. They serve as carriers for a plethora of signal molecules, including DNA, RNA, proteins, and lipids. Their unique capability of effortlessly crossing the blood-brain barrier underscores their critical role in the progression of various neurological disorders. This includes, but is not limited to, diseases such as Alzheimer's, Parkinson's, and ischemic stroke. Establishing stable and mature methods for isolating exosomes is a prerequisite for the study of exosomes and their biomedical significance. The extraction technologies of exosomes include differential centrifugation, density gradient centrifugation, size exclusion chromatography, ultrafiltration, polymer coprecipitation, immunoaffinity capture, microfluidic, and so forth. Each extraction technology has its own advantages and disadvantages, and the extraction standards of exosomes have not been unified internationally. AIMS This review aimed to showcase the recent advancements in exosome isolation techniques and thoroughly compare the advantages and disadvantages of different methods. Furthermore, the significant research progress made in using exosomes for diagnosing and treating central nervous system (CNS) diseases has been emphasized. CONCLUSION The varying isolation methods result in differences in the concentration, purity, and size of exosomes. The efficient separation of exosomes facilitates their widespread application, particularly in the diagnosis and treatment of CNS diseases.
Collapse
Affiliation(s)
- Wenjing Wang
- Beijing Chest Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Hong Sun
- Beijing Chest Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Huijuan Duan
- Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| | - Gang Sheng
- Beijing Chest Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Na Tian
- Beijing Chest Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Dingyi Liu
- Beijing Chest Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Zhaogang Sun
- Beijing Chest Hospital Affiliated to Capital Medical UniversityBeijingChina
- Beijing Tuberculosis and Thoracic Tumor Research InstituteBeijingChina
| |
Collapse
|
29
|
Khoushab S, Aghmiuni MH, Esfandiari N, Sarvandani MRR, Rashidi M, Taheriazam A, Entezari M, Hashemi M. Unlocking the potential of exosomes in cancer research: A paradigm shift in diagnosis, treatment, and prevention. Pathol Res Pract 2024; 255:155214. [PMID: 38430814 DOI: 10.1016/j.prp.2024.155214] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
Exosomes, which are tiny particles released by cells, have the ability to transport various molecules, including proteins, lipids, and genetic material containing non-coding RNAs (ncRNAs). They are associated with processes like cancer metastasis, immunity, and tissue repair. Clinical trials have shown exosomes to be effective in treating cancer, inflammation, and chronic diseases. Mesenchymal stem cells (MSCs) and dendritic cells (DCs) are common sources of exosome production. Exosomes have therapeutic potential due to their ability to deliver cargo, modulate the immune system, and promote tissue regeneration. Bioengineered exosomes could revolutionize disease treatment. However, more research is needed to understand exosomes in tumor growth and develop new therapies. This paper provides an overview of exosome research, focusing on cancer and exosome-based therapies including chemotherapy, radiotherapy, and vaccines. It explores exosomes as a drug delivery system for cancer therapy, highlighting their advantages. The article discusses using exosomes for various therapeutic agents, including drugs, antigens, and RNAs. It also examines challenges with engineered exosomes. Analyzing exosomes for clinical purposes faces limitations in sensitivity, specificity, and purification. On the other hand, Nanotechnology offers solutions to overcome these challenges and unlock exosome potential in healthcare. Overall, the article emphasizes the potential of exosomes for personalized and targeted cancer therapy, while acknowledging the need for further research.
Collapse
Affiliation(s)
- Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Hobabi Aghmiuni
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esfandiari
- Department of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
30
|
Zheng X, Ai H, Qian K, Li G, Zhang S, Zou Y, Lei C, Fu W, Hu S. Small extracellular vesicles purification and scale-up. Front Immunol 2024; 15:1344681. [PMID: 38469310 PMCID: PMC10925713 DOI: 10.3389/fimmu.2024.1344681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/06/2024] [Indexed: 03/13/2024] Open
Abstract
Exosomes are small extracellular vesicles (sEVs) secreted by cells. With advances in the study of sEVs, they have shown great potential in the diagnosis and treatment of disease. However, sEV therapy usually requires a certain dose and purity of sEVs to achieve the therapeutic effect, but the existing sEV purification technology exists in the form of low yield, low purity, time-consuming, complex operation and many other problems, which greatly limits the application of sEVs. Therefore, how to obtain high-purity and high-quality sEVs quickly and efficiently, and make them realize large-scale production is a major problem in current sEV research. This paper discusses how to improve the purity and yield of sEVs from the whole production process of sEVs, including the upstream cell line selection and cell culture process, to the downstream isolation and purification, quality testing and the final storage technology.
Collapse
Affiliation(s)
- Xinya Zheng
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Hongru Ai
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| | - Kewen Qian
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Guangyao Li
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Shuyi Zhang
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Yitan Zou
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Wenyan Fu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Fahe Life Science and Technology Inc., Shanghai, China
| | - Shi Hu
- Department of Biomedical Engineering, College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
- School of Gongli Hospital Medical Technology, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
31
|
Berezin AE, Berezin AA. Extracellular vesicles in heart failure. Adv Clin Chem 2024; 119:1-32. [PMID: 38514208 DOI: 10.1016/bs.acc.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Physiologically, extracellular vesicles (EVs) have been implicated as crucial mediators of immune response, cell homeostasis, angiogenesis, cell differentiation and growth, and tissue repair. In heart failure (HF) they may act as regulators of cardiac remodeling, microvascular inflammation, micro environmental changes, tissue fibrosis, atherosclerosis, neovascularization of plaques, endothelial dysfunction, thrombosis, and reciprocal heart-remote organ interaction. The chapter summaries the nomenclature, isolation, detection of EVs, their biologic role and function physiologically as well as in the pathogenesis of HF. Current challenges to the utilization of EVs as diagnostic and predictive biomarkers in HF are also discussed.
Collapse
Affiliation(s)
- Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, Salzburg, Austria.
| | | |
Collapse
|
32
|
Chak PT, Kam NW, Choi TH, Dai W, Kwong DLW. Unfolding the Complexity of Exosome-Cellular Interactions on Tumour Immunity and Their Clinical Prospects in Nasopharyngeal Carcinoma. Cancers (Basel) 2024; 16:919. [PMID: 38473281 DOI: 10.3390/cancers16050919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy situated in the posterolateral nasopharynx. NPC poses grave concerns in Southeast Asia due to its late diagnosis. Together with resistance to standard treatment combining chemo- and radiotherapy, NPC presents high metastatic rates and common recurrence. Despite advancements in immune-checkpoint inhibitors (ICIs) and cytotoxic-T-lymphocytes (CTLs)-based cellular therapy, the exhaustive T cell profile and other signs of immunosuppression within the NPC tumour microenvironment (TME) remain as concerns to immunotherapy response. Exosomes, extracellular vesicles of 30-150 nm in diameter, are increasingly studied and linked to tumourigenesis in oncology. These bilipid-membrane-bound vesicles are packaged with a variety of signalling molecules, mediating cell-cell communications. Within the TME, exosomes can originate from tumour, immune, or stromal cells. Although there are studies on tumour-derived exosomes (TEX) in NPC and their effects on tumour processes like angiogenesis, metastasis, therapeutic resistance, there is a lack of research on their involvement in immune evasion. In this review, we aim to enhance the comprehension of how NPC TEX contribute to cellular immunosuppression. Furthermore, considering the detectability of TEX in bodily fluids, we will also discuss the potential development of TEX-related biomarkers for liquid biopsy in NPC as this could facilitate early diagnosis and prognostication of the disease.
Collapse
Affiliation(s)
- Paak-Ting Chak
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, New Territories, Hong Kong 999077, China
| | - Tsz-Ho Choi
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Wei Dai
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
33
|
Rizzuto A, Faggiano A, Macchi C, Carugo S, Perrino C, Ruscica M. Extracellular vesicles in cardiomyopathies: A narrative review. Heliyon 2024; 10:e23765. [PMID: 38192847 PMCID: PMC10772622 DOI: 10.1016/j.heliyon.2023.e23765] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by all cells under physiological and pathological conditions. EVs constitute a potential tool to unravel cell-specific pathophysiological mechanisms at the root of disease states and retain the potential to act as biomarkers for cardiac diseases. By being able to carry bioactive cargo (such as proteins and miRNAs), EVs harness great potential as accessible "liquid biopsies", given their ability to reflect the state of their cell of origin. Cardiomyopathies encompass a variety of myocardial disorders associated with mechanical, functional and/or electric dysfunction. These diseases exhibit different phenotypes, including inappropriate ventricular hypertrophy, dilatation, scarring, fibro-fatty replacement, dysfunction, and may stem from multiple aetiologies, most often genetic. Thus, the aims of this narrative review are to summarize the current knowledge on EVs and cardiomyopathies (e.g., hypertrophic, dilated and arrhythmogenic), to elucidate the potential role of EVs in the paracrine cell-to-cell communication among cardiac tissue compartments, in aiding the diagnosis of the diverse subtypes of cardiomyopathies in a minimally invasive manner, and finally to address whether certain molecular and phenotypical characteristics of EVs may correlate with cardiomyopathy disease phenotype and severity.
Collapse
Affiliation(s)
- A.S. Rizzuto
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - A. Faggiano
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Italy
| | - C. Macchi
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - S. Carugo
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Italy
| | - C. Perrino
- Department of Advanced Biomedical Sciences, Federico II University, 80131, Naples, Italy
| | - M. Ruscica
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Italy
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
34
|
Kuklin V, Sovershaev M, Bjerner J, Keith P, Scott LK, Thomas OMT, Szpirt W, Rock G, Stegmayr B. Influence of therapeutic plasma exchange treatment on short-term mortality of critically ill adult patients with sepsis-induced organ dysfunction: a systematic review and meta-analysis. Crit Care 2024; 28:12. [PMID: 38178170 PMCID: PMC10768220 DOI: 10.1186/s13054-023-04795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024] Open
Abstract
INTRODUCTION The impact of therapeutic plasma exchange (TPE) on short-term mortality in adult patients with sepsis-induced organ dysfunction remains uncertain. The objective of the study is to assess the effect of adjunct TPE in this setting through a comprehensive literature review. METHODS The National Library of Medicine's Medline, Ovid (Embase), the Cochrane Library database and clinicaltrial.gov from January 01, 1966, until October 01, 2022, were searched for terms: therapeutic plasma exchange, plasmapheresis, sepsis, and septic shock. We reviewed, selected and extracted data from relevant randomized clinical trials (RCTs) and matched cohort studies (MCSs) comparing short-term mortality in critically ill adult septic patients treated with standard therapy versus those receiving adjunct TPE. Risk of bias was assessed in the RCTs using Cochrane Collaboration tool and in MCSs using ROBINS-I tool. Summary statistics, risk ratios (RRs), and confidence intervals (CIs) were calculated using random effects model. RESULTS This systematic review included 937 adult critically ill septic patients from five RCTs (n = 367) and fifteen MCSs (n = 570). Of these total, 543 received treatment with TPE in addition to standard care. The meta-analysis includes all five RCTs and only six MCSs (n = 627). The adjunct TPE treatment (n = 300) showed a significant reduction in short-term mortality (RR 0.59, 95% CI 0.47-0.74, I2 3%) compared to standard therapy alone (n = 327). The systematic review of all 20 trials revealed that adding TPE to the standard therapy of critically ill septic patients resulted in faster clinical and/or laboratory recovery. CONCLUSIONS Our comprehensive and up-to-date review demonstrates that adjunct TPE may provide potential survival benefits when compared to standard care for critically ill adult patients with sepsis-induced organ dysfunction. While results of this meta-analysis are encouraging, large well-designed randomized trials are required to identify the optimal patient population and TPE procedure characteristics prior to widespread adoption into practice.
Collapse
Affiliation(s)
- Vladimir Kuklin
- Department of Anaesthesiology and Intensive Care Medicine, Ahus University Hospital, Sykehusveien, 25, 1478, Lorenskog, Norway.
| | | | | | - Philip Keith
- Critical Care Medicine, Lexington Medical Center, West Columbia, SC, USA
| | - L Keith Scott
- Division of Trauma and Surgical Critical Care, Louisiana State University Health Sciences Center, Shreveport, USA
| | | | - Wladimir Szpirt
- Department of Nephrology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Gail Rock
- University of Ottawa, Ottawa, ON, Canada
| | - Bernd Stegmayr
- Department of Public Health and Clinical Medicine, Umea University, Umea, Sweden
| |
Collapse
|
35
|
Santiago VF, Rosa-Fernandes L, Macedo-da-Silva J, Angeli CB, Mule SN, Marinho CRF, Torrecilhas AC, Marie SNK, Palmisano G. Isolation of Extracellular Vesicles Using Titanium Dioxide Microspheres. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:1-22. [PMID: 38409413 DOI: 10.1007/978-3-031-50624-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Extracellular vesicles (EVs) are bilayer membrane particles released from several cell types to the extracellular environment. EVs have a crucial role in cell-cell communication, involving different biological processes in health and diseases. Due to the potential of biomarkers for several diseases as diagnostic and therapeutic tools, it is relevant to understand the biology of the EVs and their content. One of the current challenges involving EVs is regarding the purification method, which is a critical step for EV's functional and characterization studies. Ultracentrifugation is the most used method for EV isolation, where the nanoparticles are separated in sequential centrifugation to isolate the EVs based on their size. However, for viscous biofluids such as plasma, there is a co-isolation of the most abundant proteins, which can impair the EV's protein identification due to the low abundance of these proteins and signal suppression by the most abundant plasma proteins. Emerging techniques have gained attention in recent years. Titanium dioxide (TiO2) is one of the most promising techniques due to its property for selective isolation based on the interaction with phospholipids in the EV membrane. Using a small amount of TiO2 beads and a low volume of plasma, it is possible to isolate EVs with reduced plasma protein co-isolation. This study describes a comprehensive workflow for the isolation and characterization of plasma extracellular vesicles (EVs) using mass spectrometry-based proteomics techniques. The aim of this chapter is describe the EV isolation using TiO2 beads enrichment and high-throughput mass spectrometry techniques to efficiently identify the protein composition of EVs in a fast and straightforward manner.
Collapse
Affiliation(s)
- Veronica Feijoli Santiago
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Livia Rosa-Fernandes
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Janaina Macedo-da-Silva
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Claudia B Angeli
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Simon Ngao Mule
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Claudio R F Marinho
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Torrecilhas
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas; Departamento de Ciências Farmacêuticas; Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários. Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Suely N K Marie
- Laboratory of Molecular and Cellular Biology (LIM15), Department of Neurology, Fac-uldade de Medicina FMUSP, University of São Paulo, São Paulo, Brazil
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, Australia.
| |
Collapse
|
36
|
Ghorbani R, Hosseinzadeh S, Azari A, Taghipour N, Soleimani M, Rahimpour A, Abbaszadeh HA. The Current Status and Future Direction of Extracellular Nano-vesicles in the Alleviation of Skin Disorders. Curr Stem Cell Res Ther 2024; 19:351-366. [PMID: 37073662 DOI: 10.2174/1574888x18666230418121053] [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: 11/19/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 04/20/2023]
Abstract
Exosomes are extracellular vesicles (EVs) that originate from endocytic membranes. The transfer of biomolecules and biological compounds such as enzymes, proteins, RNA, lipids, and cellular waste disposal through exosomes plays an essential function in cell-cell communication and regulation of pathological and physiological processes in skin disease. The skin is one of the vital organs that makes up about 8% of the total body mass. This organ consists of three layers, epidermis, dermis, and hypodermis that cover the outer surface of the body. Heterogeneity and endogeneity of exosomes is an advantage that distinguishes them from nanoparticles and liposomes and leads to their widespread usage in the remedy of dermal diseases. The biocompatible nature of these extracellular vesicles has attracted the attention of many health researchers. In this review article, we will first discuss the biogenesis of exosomes, their contents, separation methods, and the advantages and disadvantages of exosomes. Then we will highlight recent developments related to the therapeutic applications of exosomes in the treatment of common skin disorders like atopic dermatitis, alopecia, epidermolysis bullosa, keloid, melanoma, psoriasis, and systemic sclerosis.
Collapse
Affiliation(s)
- Raziyeh Ghorbani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezo Azari
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloofar Taghipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Rahimpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Lai JJ, Hill JJ, Huang CY, Lee GC, Mai KW, Shen MY, Wang SK. Unveiling the Complex World of Extracellular Vesicles: Novel Characterization Techniques and Manufacturing Considerations. Chonnam Med J 2024; 60:1-12. [PMID: 38304124 PMCID: PMC10828078 DOI: 10.4068/cmj.2024.60.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
Extracellular vesicles (EVs) function as potent mediators of intercellular communication for many in vivo processes, contributing to both health and disease related conditions. Given their biological origins and diverse functionality from correspondingly unique "cargo" compositions, both endogenous and modified EVs are garnering attention as promising therapeutic modalities and vehicles for targeted therapeutic delivery applications. Their diversity in composition, however, has revealed a significant need for more comprehensive analytical-based characterization methods, and manufacturing processes that are consistent and scalable. In this review, we explore the dynamic landscape of EV research and development efforts, ranging from novel isolation approaches, to their analytical assessment through novel characterization techniques, and to their production by industrial-scale manufacturing process considerations. Expanding the horizon of these topics to EVs for in-human applications, we underscore the need for stringent development and adherence to Good Manufacturing Practice (GMP) guidelines. Wherein, the intricate interplay of raw materials, production in bioreactors, and isolation practices, along with analytical assessments compliant with the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines, in conjunction with reference standard materials, collectively pave the way for standardized and consistent GMP production processes.
Collapse
Affiliation(s)
- James J. Lai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - John J. Hill
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
- BioProcess Technology Group, BDO, Boston, MA, USA
| | - Casey Y. Huang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Gino C. Lee
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Karol W. Mai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Maggie Y. Shen
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Simon K. Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
38
|
Wang H, Liu Y, Zhang L, Li X, Zhao G, Song Z, Jia Y, Qiao X. High Throughput and Noninvasive Exosomal PD-L1 Detection for Accurate Immunotherapy Response Prediction via Tim4-Functionalized Magnetic Core-Shell Metal-Organic Frameworks. Anal Chem 2023; 95:18268-18277. [PMID: 38011622 DOI: 10.1021/acs.analchem.3c04117] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Exosomal PD-L1 has been increasingly considered a noninvasive and accurate predictive marker for immunotherapy treatment response. However, the clinical monitoring of exosomal PD-L1 expression is still limited by its complex biological environment as well as the lack of a robust isolation strategy. Here, a Tim4-functionalized magnetic core-shell metal-organic framework (denoted as Fe3O4@SiO2-ILI-01@Tim4) was facilely constructed via layer-by-layer assembly. Owing to the strongly hydrophilic organic ligand of 1,3-bis(4-carboxybutyl)imidazolium bromide (ILI), magnetic Fe3O4@SiO2-ILI-01@Tim4 was endowed with the merits of low nonspecific adsorption and quick, easy, and convenient isolation of exosomes. The capture efficiency of Fe3O4@SiO2-ILI-01@Tim4 reached as high as 90.3 ± 0.5% and the recovery rate for exosomes was up to 93.0 ± 6.1%. The purity of the isolated exosomes was 7.5 times higher than that via the ultracentrifugation (UC) method. By further combination with immunofluorescence assay, high throughput and noninvasive exosomal PD-L1 detection for accurate immunotherapy response prediction was achieved. The prognosis accuracy of the developed Fe3O4@SiO2-ILI-01@Tim4-based strategy reached 85.7%, whereas the prognosis accuracy of the clinical gold standard, the PD-L1 combined positive score (CPS) test, was only 57.1%. Most interestingly, the developed method is especially suitable for those patients receiving false negative results in the CPS test. The proposed Fe3O4@SiO2-ILI-01@Tim4 is a highly efficient and robust technique showing great potential in high throughput and noninvasive exosomal PD-L1 detection for accurately predicting immunotherapy efficacy.
Collapse
Affiliation(s)
- Haiyan Wang
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding 071002, China
| | - Yanli Liu
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding 071002, China
| | - Liyuan Zhang
- Institute of Advanced Science Facilities, Shenzhen 518107, China
- College of Basic Medical Science, Key Laboratory for Proteomics of Liaoning Province, Dalian Medical University, Dalian 116044, China
| | - Xinran Li
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding 071002, China
| | - Guofa Zhao
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Zizheng Song
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Youchao Jia
- Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Xiaoqiang Qiao
- College of Pharmaceutical Sciences, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis, Ministry of Education, Hebei University, Baoding 071002, China
| |
Collapse
|
39
|
Hyung S, Ko J, Heo YJ, Blum SM, Kim ST, Park SH, Park JO, Kang WK, Lim HY, Klempner SJ, Lee J. Patient-derived exosomes facilitate therapeutic targeting of oncogenic MET in advanced gastric cancer. SCIENCE ADVANCES 2023; 9:eadk1098. [PMID: 38000030 PMCID: PMC10672184 DOI: 10.1126/sciadv.adk1098] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023]
Abstract
Gastric cancer (GC) with peritoneal metastases and malignant ascites continues to have poor prognosis. Exosomes mediate intercellular communication during cancer progression and promote therapeutic resistance. Here, we report the significance of exosomes derived from malignant ascites (EXOAscites) in cancer progression and use modified exosomes as resources for cancer therapy. EXOAscites from patients with GC stimulated invasiveness and angiogenesis in an ex vivo three-dimensional autologous tumor spheroid microfluidic system. EXOAscites concentration increased invasiveness, and blockade of their secretion suppressed tumor progression. In MET-amplified GC, EXOAscites contain abundant MET; their selective delivery to tumor cells enhanced angiogenesis and invasiveness. Exosomal MET depletion substantially reduced invasiveness; an additive therapeutic effect was induced when combined with MET and/or VEGFR2 inhibition in a patient-derived MET-amplified GC model. Allogeneic MET-harboring exosome delivery induced invasion and angiogenesis in a MET non-amplified GC model. MET-amplified patient tissues showed higher exosome concentration than their adjacent normal tissues. Manipulating exosome content and production may be a promising complementary strategy against GC.
Collapse
Affiliation(s)
- Sujin Hyung
- Precision Medicine Research Institute, Samsung Medical Center, Seoul, Republic of Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jihoon Ko
- Department of BioNano Technology, Gachon University, Gyeonggi 13120, Republic of Korea
| | | | - Steven M. Blum
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Hoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ho Yeong Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Samuel J. Klempner
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
40
|
Cai CS, He GJ, Xu FW. Advances in the Applications of Extracellular Vesicle for the Treatment of Skin Photoaging: A Comprehensive Review. Int J Nanomedicine 2023; 18:6411-6423. [PMID: 37954453 PMCID: PMC10638935 DOI: 10.2147/ijn.s433611] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023] Open
Abstract
Skin photoaging is a complex biological process characterized by the accumulation of oxidative damage and structural changes in the skin, resulting from chronic exposure to ultraviolet (UV) radiation. Despite the growing demand for effective treatments, current therapeutic options for skin photoaging remain limited. However, emerging research has highlighted the potential of extracellular vesicles (EVs), including exosomes, micro-vesicles, apoptotic bodies and liposomes, as promising therapeutic agents in skin rejuvenation. EVs are involved in intercellular communication and can deliver bioactive molecules, including proteins, nucleic acids, and lipids, to recipient cells, thereby influencing various cellular processes. This comprehensive review aims to summarize the current research progress in the application of EVs for the treatment of skin photoaging, including their isolation and characterization methods, roles in skin homeostasis, therapeutic potential and clinical applications for skin photoaging. Additionally, challenges and future directions in EVs-based therapies for skin rejuvenation are discussed.
Collapse
Affiliation(s)
- Chan-Sheng Cai
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Gui-Juan He
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, People’s Republic of China
| | - Fa-Wei Xu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, People’s Republic of China
| |
Collapse
|
41
|
Yu L, Wen H, Liu C, Wang C, Yu H, Zhang K, Han Q, Liu Y, Han Z, Li Z, Liu N. Embryonic stem cell-derived extracellular vesicles rejuvenate senescent cells and antagonize aging in mice. Bioact Mater 2023; 29:85-97. [PMID: 37449253 PMCID: PMC10336196 DOI: 10.1016/j.bioactmat.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/11/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Aging is a degenerative process that leads to tissue dysfunction and death. Embryonic stem cells (ESCs) have great therapeutic potential for age-related diseases due to their capacity for self-renewal and plasticity. However, the use of ESCs in clinical treatment is limited by immune rejection, tumourigenicity and ethical issues. ESC-derived extracellular vesicles (EVs) may provide therapeutic effects that are comparable to those of ESCs while avoiding unwanted effects. Here, we fully evaluate the role of ESC-EVs in rejuvenation in vitro and in vivo. Using RNA sequencing (RNA-Seq) and microRNA sequencing (miRNA-Seq) screening, we found that miR-15b-5p and miR-290a-5p were highly enriched in ESC-EVs, and induced rejuvenation by silencing the Ccn2-mediated AKT/mTOR pathway. These results demonstrate that miR-15b-5p and miR-290a-5p function as potent activators of rejuvenation mediated by ESC-EVs. The rejuvenating effect of ESC-EVs was further investigated in vivo by injection into aged mice. The results showed that ESC-EVs successfully ameliorated the pathological age-related phenotypes and rescued the transcriptome profile of aged mice. Our findings demonstrate that ESC-EVs treatment can rejuvenate senescence both in vitro and in vivo and suggest the therapeutic potential of ESC-EVs as a novel cell-free alternative to ESCs for age-related diseases.
Collapse
Affiliation(s)
- Lu Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Hang Wen
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Huaxin Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Kaiyue Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qingsheng Han
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yue Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhongchao Han
- Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Na Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University, Tianjin, 300071, China
| |
Collapse
|
42
|
Kapoor KS, Harris K, Arian K, Ma L, Church KA, Kalluri R. Rapid and high yield isolation of extracellular vesicles with purity by application of size exclusion fast performance liquid chromatography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.12.561425. [PMID: 37904947 PMCID: PMC10614745 DOI: 10.1101/2023.10.12.561425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Extracellular Vesicles (EVs) have emerged as potential biomarkers for diagnosing a range of diseases without invasive procedures. Extracellular vesicles also offer an advantage compared to synthetic vesicles, for delivery of various drugs. However, limitations in segregating EVs from soluble proteins have led to inconsistent EV retrieval rates with low levels of purity. Here, we report a new high-yield (>95%) and rapid (<20 min) EV isolation method called S ize E xclusion - F ast P erformance L iquid C hromatography (SE-FPLC). We show SE-FPLC can effectively isolate EVs from multiple sources including EVs derived from human and mouse cells and serum. The results indicate that SE-FPLC can successfully remove highly abundant protein contaminants such as albumin and lipoprotein complexes, which can represent a major hurdle in large scale isolation of EVs for clinical translation. Additionally, the high-yield nature of SE- FPLC allows for easy industrial upscaling of extracellular vesicles production for various clinical utilities. Moreover, SE-FPLC enables analysis of very small volumes of blood for use in point-of-care diagnostics in the clinic. Collectively, SE-FPLC offers many advantages over current EV isolation methods and offers rapid clinical utility potential.
Collapse
|
43
|
Fu Y, Xiong S. Differential traits between microvesicles and exosomes in enterovirus infection. MedComm (Beijing) 2023; 4:e384. [PMID: 37752943 PMCID: PMC10518433 DOI: 10.1002/mco2.384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/05/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Extracellular vesicles (EVs), including exosomes and microvesicles (MVs), are released by most cell types into the extracellular space and represent the pathophysiological condition of their source cells. Recent studies demonstrate that EVs derived from infected cells and tumors contribute to disease pathogenesis. However, very few studies have rigorously characterized exosomes and microvesicles in infectious diseases. In this study, we focused on subpopulations of EVs during the human enterovirus infection and explored the distinct traits and functions of EVs. We construct an effective immunomagnetic method to isolate exosomes and MVs from enterovirus-infected cells excluding virion. The morphology and sizes of exosomes and MVs have no significant alteration after enterovirus infection. Meanwhile, our study observed that the enterovirus infection could induce exosome secretion but not MVs. In vivo study showed that there was differential biodistribution between exosomes and MVs. Using deep RNA sequencing, we found that the cargo information in MVs rather than in exosomes could accurately reflect pathological condition of original cells. Our study demonstrated that it should be considered to use MVs as clinical diagnostics during in enterovirus infection because their composition is reflective of pathological changes.
Collapse
Affiliation(s)
- Yuxuan Fu
- Jiangsu Key Laboratory of Infection and ImmunityInstitutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and ImmunityInstitutes of Biology and Medical SciencesSoochow UniversitySuzhouChina
| |
Collapse
|
44
|
Yu L, Qin J, Xing J, Dai Z, Zhang T, Wang F, Zhou J, Zhang X, Chen X, Gu Y. The mechanisms of exosomes in diabetic foot ulcers healing: a detailed review. J Mol Med (Berl) 2023; 101:1209-1228. [PMID: 37691076 DOI: 10.1007/s00109-023-02357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 08/03/2023] [Accepted: 08/06/2023] [Indexed: 09/12/2023]
Abstract
As time goes by, the morbidity of diabetes mellitus continues to rise, and the economic burden of diabetic foot ulcers as a common and serious complication of diabetes is increasing. However, currently there is no unified clinical treatment strategy for this complication, and the therapeutic efficacy is unsatisfactory. Recent studies have revealed that biological effects of exosomes involved in multiple stages of the process of wound closure are similar to source cells. Compared with source cells, exosomes possess lowly immunogenicity, highly stability and easily stored, etc. Accumulating evidence confirmed that exosomes promote diabetic wound healing through various pathways such as promoting angiogenesis, collagen fiber deposition, and inhibiting inflammation. The superior therapeutic efficacy of exosomes in accelerating diabetic cutaneous wound healing has attracted an increasing attention. Notably, the molecular mechanisms of exosomes vary among different sources in the chronic wound closure of diabetes. This review focuses on the specific roles and mechanisms of different cell- or tissue-derived exosomes relevant to wound healing. Additionally, the paper provides an overview of the current pre-clinical and clinical applications of exosomes, illustrates their special advantages in wound repair. Furthermore, we discuss the potential obstacles and various solutions for future research on exosomes in the management of diabetic foot ulcer. The aim is to offer novel insights and approaches for the treatment of diabetic foot ulcer.
Collapse
Affiliation(s)
- Lei Yu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Jianxin Qin
- Department of Histology and Embryology, Medical School, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jiajun Xing
- Department of Histology and Embryology, Medical School, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Zihao Dai
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Tingting Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China
| | - Feng Wang
- Nantong Xingzhong Cell Engineering Co. LTD, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jin Zhou
- Nantong Xingzhong Cell Engineering Co. LTD, Nantong, Jiangsu, 226001, People's Republic of China
| | - Xiaobai Zhang
- Department of Respiratory Medicine, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Xia Chen
- Department of Histology and Embryology, Medical School, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China.
| | - Yunjuan Gu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, People's Republic of China.
| |
Collapse
|
45
|
Li C, Hu Y. Extracellular Vesicles Derived from Mesenchymal Stem Cells as Cell-Free Therapy for Intrauterine Adhesion. Int J Stem Cells 2023; 16:260-268. [PMID: 37385632 PMCID: PMC10465336 DOI: 10.15283/ijsc21177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/17/2021] [Indexed: 07/01/2023] Open
Abstract
Intrauterine adhesion (IUA) can occur after trauma to the basal layer of the endometrium, contributing to severe complications in females, such as infertility and amenorrhea. To date, the proposed therapeutic strategies are targeted to relieve IUA, such as hysteroscopic adhesiolysis, Foley catheter balloon, and hyaluronic acid injection have been applied in the clinic. However, these approaches showed limited effects in alleviating endometrial fibrosis and thin endometrium. Mesenchymal stem cells (MSCs) can offer the potential for endometrium regeneration owing to reduce inflammation and release growth factors. On this basis, MSCs have been proposed as promising methods to treat intrauterine adhesion. However, due to the drawbacks of cell therapy, the possible therapeutic use of extracellular vesicles released by stem cells is raising increasing interest. The paracrine effect, mediated by MSCs derived extracellular vehicles (MSC-EVs), has recently been suggested as a mechanism for their therapeutic properties. Here, we summarizes the main pathological mechanisms involved in intrauterine adhesion, the biogenesis and characteristics of extracellular vesicles, explaining how these vesicles could provide new opportunities for MSCs.
Collapse
Affiliation(s)
- Chao Li
- Department of Gynecology, Tianjin Medical University, Tianjin, China
- Department of Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| | - Yuanjing Hu
- Department of Gynecology, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, China
| |
Collapse
|
46
|
Zhang Z, Yu K, You Y, Jiang P, Wu Z, DeTure MA, Dickson DW, Ikezu S, Peng J, Ikezu T. Comprehensive characterization of human brain-derived extracellular vesicles using multiple isolation methods: Implications for diagnostic and therapeutic applications. J Extracell Vesicles 2023; 12:e12358. [PMID: 37563857 PMCID: PMC10415636 DOI: 10.1002/jev2.12358] [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: 05/03/2023] [Revised: 07/14/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Extracellular vesicles (EVs) have emerged as critical mediators of intercellular communication and promising biomarkers and therapeutics in the central nervous system (CNS). Human brain-derived EVs (BDEVs) provide a comprehensive snapshot of physiological changes in the brain's environment, however, the isolation of BDEVs and the comparison of different methods for this purpose have not been fully investigated. In this study, we compared the yield, morphology, subtypes and protein cargo composition of EVs isolated from the temporal cortex of aged human brains using three established separation methods: size-exclusion chromatography (SEC), phosphatidylserine affinity capture (MagE) and sucrose gradient ultracentrifugation (SG-UC). Our results showed that SG-UC method provided the highest yield and collected larger EVs compared to SEC and MagE methods as assessed by transmission electron microscopy and nanoparticle tracking analysis (NTA). Quantitative tandem mass-tag (TMT) mass spectrometry analysis of EV samples from three different isolation methods identified a total of 1158 proteins, with SG-UC showing the best enrichment of common EV proteins with less contamination of non-EV proteins. In addition, SG-UC samples were enriched in proteins associated with ATP activity and CNS maintenance, and were abundant in neuronal and oligodendrocytic molecules. In contrast, MagE samples were more enriched in molecules related to lipoproteins, cell-substrate junction and microglia, whereas SEC samples were highly enriched in molecules related to extracellular matrix, Alzheimer's disease and astrocytes. Finally, we validated the proteomic results by performing single-particle analysis using the super-resolution microscopy and flow cytometry. Overall, our findings demonstrate the differences in yield, size, enrichment of EV cargo molecules and single EV assay by different isolation methods, suggesting that the choice of isolation method will have significant impact on the downstream analysis and protein discovery.
Collapse
Affiliation(s)
- Zhengrong Zhang
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Kaiwen Yu
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and MetabolomicsSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Yang You
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Peizhou Jiang
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Zhiping Wu
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and MetabolomicsSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | | | | | - Seiko Ikezu
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and MetabolomicsSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Tsuneya Ikezu
- Department of NeuroscienceMayo Clinic FloridaJacksonvilleFloridaUSA
- Department of Pharmacology and Experimental TherapeuticsBoston University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
47
|
Shami-shah A, Travis BG, Walt DR. Advances in extracellular vesicle isolation methods: a path towards cell-type specific EV isolation. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:447-460. [PMID: 39698024 PMCID: PMC11648483 DOI: 10.20517/evcna.2023.14] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 07/10/2023] [Accepted: 07/19/2023] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles are small, heterogenous, phospholipid-rich vesicles that are secreted by all cells into the extracellular space. They play a key role in intercellular communication because they can transport a variety of biomolecules such as proteins, lipids, and nucleic acids between cells. As categorized by the International Society of Extracellular Vesicles (ISEV), the term EV encompasses different sub-types, including exosomes, microvesicles, and apoptotic bodies, which differ in their size, origin, and cargo. EVs can be isolated from biological fluids such as blood, urine, and cerebrospinal fluid, and their biomolecular content can be analyzed to monitor the progression of certain diseases. Therefore, EVs can be used as a new source of liquid biomarkers for advancing novel diagnostic and therapeutic tools. Isolating and analyzing EVs can be challenging due to their nanoscopic size and low abundance. Several techniques have been developed for the isolation and characterization of EVs, including ultracentrifugation, density gradient separation, size-exclusion chromatography, microfluidics, and magnetic bead-based/affinity methods. This review highlights advances in EV isolation techniques in the last decade and provides a perspective on their advantages, limitations, and potential application to cell-type specific EV isolation in the future.
Collapse
Affiliation(s)
- Adnan Shami-shah
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02120, USA
- Department of Pathology, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02120, USA
| | - Benjamin G Travis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02120, USA
- Department of Pathology, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02120, USA
| | - David R Walt
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02120, USA
- Department of Pathology, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
48
|
Zheng J, Hu X, Zeng Y, Zhang B, Sun Z, Liu X, Zheng W, Chai Y. Review of the advances in lipid anchors-based biosensors for the isolation and detection of exosomes. Anal Chim Acta 2023; 1263:341319. [PMID: 37225343 DOI: 10.1016/j.aca.2023.341319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/26/2023]
Abstract
Exosomes are nanoparticles with a bilayer lipid structure that carry cargo from their cells of origin. These vesicles are vital to disease diagnosis and therapeutics; however, conventional isolation and detection techniques are generally complicated, time-consuming, and costly, thus hampering the clinical applications of exosomes. Meanwhile, sandwich-structured immunoassays for exosome isolation and detection rely on the specific binding of membrane surface biomarkers, which may be limited by the type and amount of target protein present. Recently, lipid anchors inserted into the membranes of vesicles through hydrophobic interactions have been adopted as a new strategy for extracellular vesicle manipulation. By combining nonspecific and specific binding, the performance of biosensors can be improved variously. This review presents the reaction mechanisms and properties of lipid anchors/probes, as well as advances in the development of biosensors. The combination of signal amplification methods with lipid anchors is discussed in detail to provide insights into the design of convenient and sensitive detection techniques. Finally, the advantages, challenges, and future directions of lipid anchor-based exosome isolation and detection methods are highlighted from the perspectives of research, clinical use, and commercialization.
Collapse
Affiliation(s)
- Junyuan Zheng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Xiaoxiang Hu
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Yuping Zeng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Binmao Zhang
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Zhonghao Sun
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| | - Xiaowei Liu
- Department of Management, Shenzhen University, Shenzhen, 518055, China.
| | - Weidong Zheng
- Department of Laboratory Medicine, Shenzhen University General Hospital, Shenzhen, 518055, China.
| | - Yujuan Chai
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
49
|
Jiang Y, Liu X, Ye J, Ma Y, Mao J, Feng D, Wang X. Migrasomes, a new mode of intercellular communication. Cell Commun Signal 2023; 21:105. [PMID: 37158915 PMCID: PMC10165304 DOI: 10.1186/s12964-023-01121-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 03/30/2023] [Indexed: 05/10/2023] Open
Abstract
Migrasomes are newly discovered extracellular vesicles (EVs) that are formed in migrating cells and mediate intercellular communication. However, their size, biological generation, cargo packaging, transport, and effects on recipient cells by migrasomes are different from those of other EVs. In addition to mediating organ morphogenesis during zebrafish gastrulation, discarding damaged mitochondria, and lateral transport of mRNA and proteins, growing evidence has demonstrated that migrasomes mediate a variety of pathological processes. In this review, we summarize the discovery, mechanisms of formation, isolation, identification, and mediation of cellular communication in migrasomes. We discuss migrasome-mediated disease processes, such as osteoclast differentiation, proliferative vitreoretinopathy, tumor cell metastasis by PD-L1 transport, immune cell chemotaxis to the site of infection by chemokines, angiogenesis promotion via angiogenic factors by immune cells, and leukemic cells chemotaxis to the site of mesenchymal stromal cells. Moreover, as new EVs, we propose the potential of migrasomes for disease diagnosis and treatment. Video Abstract.
Collapse
Affiliation(s)
- Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yongbin Ma
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Department of Central Laboratory, Jintan Hospital, Jiangsu University, 500 Avenue Jintan, Jintan, 213200, People's Republic of China.
| | - Jiahui Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Dingqi Feng
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Department of Nuclear Medicine and Institute of Digestive Diseases, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
50
|
van de Looij SM, de Jong OG, Vermonden T, Lorenowicz MJ. Injectable hydrogels for sustained delivery of extracellular vesicles in cartilage regeneration. J Control Release 2023; 355:685-708. [PMID: 36739906 DOI: 10.1016/j.jconrel.2023.01.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/15/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are a population of small vesicles secreted by essentially all cell types, containing a wide variety of biological macromolecules. Due to their intrinsic capabilities for efficient intercellular communication, they are involved in various aspects of cellular functioning. In the past decade, EVs derived from stem cells attracted interest in the field of regenerative medicine. Owing to their regenerative properties, they have great potential for use in tissue repair, in particular for tissues with limited regenerative capabilities such as cartilage. The maintenance of articular cartilage is dependent on a precarious balance of many different components that can be disrupted by the onset of prevalent rheumatic diseases. However, while cartilage is a tissue with strong mechanical properties that can withstand movement and heavy loads for years, it is virtually incapable of repairing itself after damage has occurred. Stem cell-derived EVs (SC-EVs) transport regenerative components such as proteins and nucleic acids from their parental cells to recipient cells, thereby promoting cartilage healing. Many possible pathways through which SC-EVs execute their regenerative function have been reported, but likely there are still numerous other pathways that are still unknown. This review discusses various preclinical studies investigating intra-articular injections of free SC-EVs, which, while often promoting chondrogenesis and cartilage repair in vivo, showed a recurring limitation of the need for multiple administrations to achieve sufficient tissue regeneration. Potentially, this drawback can be overcome by making use of an EV delivery platform that is capable of sustainably releasing EVs over time. With their remarkable versatility and favourable chemical, biological and mechanical properties, hydrogels can facilitate this release profile by encapsulating EVs in their porous structure. Ideally, the optimal delivery platform can be formed in-situ, by means of an injectable hydrogel that can be administered directly into the affected joint. Relevant research fulfilling these criteria is discussed in detail, including the steps that still need to be taken before injectable hydrogels for sustained delivery of EVs can be applied in the context of cartilage regeneration in the clinic.
Collapse
Affiliation(s)
- Sanne M van de Looij
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Olivier G de Jong
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands
| | - Magdalena J Lorenowicz
- Regenerative Medicine Centre, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands; Centre for Molecular Medicine, University Medical Centre Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands; Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands.
| |
Collapse
|