1
|
Ge W, Mu Z, Yang S, Zeng Y, Deng Y, Lin Y, Xie P, Li G. Biosensor-based methods for exosome detection with applications to disease diagnosis. Biosens Bioelectron 2025; 279:117362. [PMID: 40157151 DOI: 10.1016/j.bios.2025.117362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/09/2025] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Exosomes are nanoscale extracellular vesicles (EVs) secreted by most eukaryotic cells and can be found in nearly all human body fluids. Increasing evidence has revealed their pivotal roles in intercellular communication, and their active participation in myriad physiological and pathological activities. Exosomes' functions rely on their contents that are closely correlated with the biological characteristics of parental cells, which may provide a rich resource of molecular information for accurate and detailed diagnosis of a diverse array of diseases, such as differential diagnosis of Alzheimer's disease, early detection and subtyping of various tumors. As a category of sensitive detection devices, biosensors can fully reveal the molecular information and convert them into actionable clinical information. In this review, recent advances in biosensor-based methods for the detection of exosomes are summarized. We have described the fabrication of various biosensors based on the analysis of exosomal proteins, RNAs or glycans for accurate diagnosis, with respect to their elaborate recognition designs, signal amplification strategies, sensing properties, as well as their application potential. The challenges along with corresponding technologies in the future development and clinical translation of these biosensors are also discussed.
Collapse
Affiliation(s)
- Weikang Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zheying Mu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Shiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yujing Zeng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yifan Lin
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
2
|
Saint-Pol J, Culot M. Minimum information for studies of extracellular vesicles (MISEV) as toolbox for rigorous, reproducible and homogeneous studies on extracellular vesicles. Toxicol In Vitro 2025; 106:106049. [PMID: 40074066 DOI: 10.1016/j.tiv.2025.106049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Studies based on extracellular vesicles (EVs) have been multiplying exponentially for almost two decades, since they were first identified as vectors of cell-cell communication. However, several of these studies display a lack of rigor in EVs characterization and isolation, without discriminating between the different EV populations, thus generating conflicting and unreproducible results. There is therefore a strong need for standardization and guidelines to conduct studies that are rigorous, transparent, reproducible and comply with certain nomenclatures concerning the type of EVs used. The International Society for Extracellular Vesicles (ISEV) published the Minimum Information for Studies of Extracellular Vesicles (MISEV) in 2014, updating it in 2018 and 2023 to reflect different study contexts and technical advancements. The primary objective of this review is to inform future authors about EVs, including their history, nomenclature, and technical recommendations for the for isolation and functionality analysis for conducing EV-based studies according to current standards. Additionally, it aims to inform reviewers about the key parameters required for characterizing EV preparations.
Collapse
Affiliation(s)
- Julien Saint-Pol
- Univ. Artois, UR 2465, Blood-Brain Barrier laboratory (LBHE), F-62300 Lens, France.
| | - Maxime Culot
- Univ. Artois, UR 2465, Blood-Brain Barrier laboratory (LBHE), F-62300 Lens, France
| |
Collapse
|
3
|
Atabay M, Inci F, Saylan Y. Computational studies for the development of extracellular vesicle-based biosensors. Biosens Bioelectron 2025; 277:117275. [PMID: 39999607 DOI: 10.1016/j.bios.2025.117275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/25/2024] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
Cancer affects millions of people, and early detection and efficient treatment are two strong levers to hurdle this disease. Recent studies on exosomes, a subset of extracellular vesicles, have deliberately shown the potential to function as a biomarker or treatment tool, thereby attracting the attention of researchers who work on developing biosensors. Due to the ability of computational methods to predict of the behavior of biomolecules, the combination of experimental and computational methods would enhance the analytical performance of the biosensor, including sensitivity, accuracy, and specificity, even detecting such vesicles from bodily fluids. In this regard, the role of computational methods such as molecular docking, molecular dynamics simulation, and density functional theory is overviewed in the development of biosensors. This review highlights the investigations and studies that have been reported using these methods to design exosome-based biosensors. This review concludes with the role of the quantum mechanics/molecular mechanics method in the investigation of chemical processes of biomolecular systems and the deficiencies in using this approach to develop exosome-based biosensors. In addition, the artificial intelligence theory is explained briefly to show its importance in the study of these biosensors.
Collapse
Affiliation(s)
- Maryam Atabay
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, Turkey; Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Yeşeren Saylan
- Department of Chemistry, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
4
|
Niu Z, Zhou H, Zheng W, Hayes OG, Hou VWQ, Görgens A, Roudi S, Zhou G, Wiklander RJ, Sych T, Sezgin E, Nordin JZ, Zhao Y, Liang X, Andaloussi SEL. Screening scaffold proteins for improved functional delivery of luminal proteins using engineered extracellular vesicles. J Control Release 2025; 384:113882. [PMID: 40425092 DOI: 10.1016/j.jconrel.2025.113882] [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: 11/19/2024] [Revised: 05/16/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
The potential for engineered extracellular vesicles (EVs) to efficiently deliver biotherapeutics is still largely untapped. One of the key structures in determining cargo loading and subsequent functional delivery efficiency of engineered EVs is the sorting protein (scaffold). To determine the role of scaffold protein identity, a functional screen of scaffold proteins for efficient cargo delivery is required. Here, we applied the VEDIC (VSV-G plus EV-sorting Domain-Intein-Cargo) system, previously developed by our group, for the functional screen of 55 different scaffold proteins. Three tetraspanins (TSPAN2, TSPAN4 and TSPAN9) were identified that demonstrate enhanced intracellular delivery of cargo when compared to traditionally used CD63. We further explored the in vivo and ex vivo protein delivery performance of the best performing engineered EVs (TSPAN2) using melanoma xenografts and isolated primary cells from Cre-LoxP R26-LSL-tdTomato reporter mice, respectively. Finally, we report successful treatment of LPS-induced systemic inflammation by delivering a super-repressor inhibitor of NF-ĸB using TSPAN2 engineered EVs. This work highlights the importance of screening critical EV engineering elements, such as the scaffold protein, to modulate EV properties.
Collapse
Affiliation(s)
- Zheyu Niu
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Houze Zhou
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Wenyi Zheng
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Oliver G Hayes
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Vicky W Q Hou
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - André Görgens
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Samantha Roudi
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden
| | - Guannan Zhou
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rim Jawad Wiklander
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Taras Sych
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Joel Z Nordin
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Immunology and Transfusion Medicine (KITM), Karolinska University Hospital, Stockholm, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden; Cancer Research Laboratory, Shandong University-Karolinska Institutet collaborative Laboratory, School of Basic Medical Science, Shandong University, Jinan, Shandong, PR China.
| | - Samir E L Andaloussi
- Biomolecular Medicine, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Karolinska ATMP Center, ANA Futura, Karolinska Institutet, Stockholm, Sweden; Department of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University, Stockholm, Sweden; Evox Therapeutics Limited, Oxford, United Kingdom
| |
Collapse
|
5
|
Liu C, Cheng C, Cheng K, Gao AS, Li Q, Atala A, Zhang Y. Precision exosome engineering for enhanced wound healing and scar revision. J Transl Med 2025; 23:578. [PMID: 40410904 PMCID: PMC12103044 DOI: 10.1186/s12967-025-06578-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
The dysfunction of wound-healing processes can result in chronic non-healing wounds and pathological scar formation. Current treatment options often fall short, necessitating innovative approaches. Exosomes, extracellular vesicles secreted by various cells, have emerged as promising therapeutic agents serving as an intercellular communication system. By engineering exosomes, their cargo and surface properties can be tailored to enhance therapeutic efficacy and specificity. Engineered exosomes (eExo) are emerging as a favorable tool for treating non-healing wounds and pathological scars. In this review, we delve into the underlying mechanisms of non-healing wounds and pathological scars, outline the current state of engineering strategies, and explore the clinical potential of eExo based on preclinical and clinical studies. In addition, we address the current challenges and future research directions, including standardization, safety and efficacy assessments, and potential immune responses. In conclusion, eExo hold great promise as a novel therapeutic approach for non-healing wounds and non-healing wounds and pathological scars. Further research and clinical trials are warranted to translate preclinical findings into effective clinical treatments.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chen Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108-2718, USA
| | - Allen S Gao
- Department of Urologic Surgery, School of Medicine, University of California, Davis Sacramento, CA, 95817, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27151, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27151, USA.
| |
Collapse
|
6
|
Kang H, Qiu L, Li Y, Xu X, Pei R, Yang T, Yang L, Xu X, Sun N. Si Microanemones Integrated Microfluidic Chip for Highly Efficient Isolation of Extracellular Vesicles. Adv Healthc Mater 2025:e2500439. [PMID: 40395100 DOI: 10.1002/adhm.202500439] [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: 03/14/2025] [Revised: 04/29/2025] [Indexed: 05/22/2025]
Abstract
Liquid biopsy has emerged as a transformative approach for early cancer detection and treatment monitoring, offering significant potential to improve patient outcomes. However, isolating tumor-derived extracellular vesicles (EVs) from body fluids is often impeded by background noise, making subsequent analysis challenging. Herein, a bio-inspired 3D silicon microanemone (SMA) microfluidic chip is reported. This innovative structure is prepared by a two-step lithographic method combined with nanosphere lithography, achieving an impressive isolation efficiency of 89.4%. Simulation results reveal that the hierarchical structure not only provides more antibody binding sites but also synergizes with an integrated chaotic mixer to amplify fluid perturbations, while inducing a flow around circular cylinder phenomenon to enhance EV-antibody interactions. Finally, the SMA chip's performance is assessed with clinical samples and combined with RT-qPCR-based β-actin (ACTB) mRNA quantification in purified EVs. The results demonstrate its high sensitivity and specificity in isolating cancer-related EV subgroups, enabling non-invasive and precise detection of cancer biomarkers in blood samples.
Collapse
Affiliation(s)
- Hanyue Kang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Key Laboratory of D&A for Metal-Functional Materials, School of Materials Science & Engineering, Tongji University, Shanghai, 201804, China
| | - Lei Qiu
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yecheng Li
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Xiaocheng Xu
- Department of Thyroid and Breast Surgery, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, 215000, China
| | - Renjun Pei
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Tongqing Yang
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Key Laboratory of D&A for Metal-Functional Materials, School of Materials Science & Engineering, Tongji University, Shanghai, 201804, China
| | - Lizhi Yang
- Zhejiang Dongfang Polytechnic School of Health Medicine, Wenzhou, Zhejiang, 325000, China
| | - Xiaobin Xu
- Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Key Laboratory of D&A for Metal-Functional Materials, School of Materials Science & Engineering, Tongji University, Shanghai, 201804, China
| | - Na Sun
- Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
7
|
Müller GA. The Transformation Experiment of Frederick Griffith II: Inclusion of Cellular Heredity for the Creation of Novel Microorganisms. Bioengineering (Basel) 2025; 12:532. [PMID: 40428151 DOI: 10.3390/bioengineering12050532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
So far, synthetic biology approaches for the construction of artificial microorganisms have fostered the transformation of acceptor cells with genomes from donor cells. However, this strategy seems to be limited to closely related bacterial species only, due to the need for a "fit" between donor and acceptor proteomes and structures. "Fitting" of cellular regulation of metabolite fluxes and turnover between donor and acceptor cells, i.e. cybernetic heredity, may be even more difficult to achieve. The bacterial transformation experiment design 1.0, as introduced by Frederick Griffith almost one century ago, may support integration of DNA, macromolecular, topological, cybernetic and cellular heredity: (i) attenuation of donor Pneumococci of (S) serotype fosters release of DNA, and hypothetically of non-DNA structures compatible with subsequent transfer to and transformation of acceptor Pneumococci from (R) to (S) serotype; (ii) use of intact donor cells rather than of subcellular or purified fractions may guarantee maximal diversity of the structural and cybernetic matter and information transferred; (iii) "Blending" or mixing and fusion of donor and acceptor Pneumococci may occur under accompanying transfer of metabolites and regulatory circuits. A Griffith transformation experiment design 2.0 is suggested, which may enable efficient exchange of DNA as well as non-DNA structural and cybernetic matter and information, leading to unicellular hybrid microorganisms with large morphological/metabolic phenotypic differences and major features compared to predeceding cells. The prerequisites of horizontal gene and somatic cell nuclear transfer, the molecular mechanism of transformation, the machineries for the biogenesis of bacterial cytoskeleton, micelle-like complexes and membrane landscapes are briefly reviewed on the basis of underlying conceptions, ranging from Darwin's "gemmules" to "stirps", cytoplasmic and "plasmon" inheritance, "rhizene agency", "communicology", "transdisciplinary membranology" to up to Kirschner's "facilitated variation".
Collapse
Affiliation(s)
- Günter A Müller
- Biology and Technology Studies Institute Munich (BITSIM), 80939 Munich, Germany
- Institute of Media Sociology, Department of Cultural Sciences, University of Paderborn, 33104 Paderborn, Germany
| |
Collapse
|
8
|
Park J, Feng M, Yang J, Shen H, Qin Z, Guo W, Issadore DA. Agarose Microgel-Based In Situ Cleavable Immuno-Rolling Circle Amplification for Multiplexed Single-Molecule Quantitation on Single Extracellular Vesicles. ACS NANO 2025; 19:17884-17899. [PMID: 40320637 DOI: 10.1021/acsnano.5c04207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
We have developed a platform for the multiplexed and ultrasensitive profiling of individual extracellular vesicles (EVs) directly in plasma, which we call GDEVA─Agarose microGel-based Digital single-molecule-single EV Assay. GDEVA achieves single-molecule sensitivity and moderate multiplexing (demonstrated 3-plex), and can achieve a throughput of ∼104 EVs per minute necessary to resolve EVs directly in human plasma when read out using flow cytometry. Our platform integrates a rolling circle amplification (RCA) immunoassay of EV surface proteins, which are cleaved from single EVs, and amplified within agarose microgels, followed by flow cytometry-based readout or imaging after fluorescence-activated cell sorting (FACS). It overcomes steric hindrance of RCA products, nonspecific binding of RCA templates, and the lack of quantitation of multiple proteins on EVs that have plagued earlier approaches. We evaluated the analytical capabilities of GDEVA through head-to-head comparison with conventional technology and demonstrated a ∼100× improvement in the limit of detection (LOD) of EV subpopulations. We evaluate GDEVA's potential in cancer immunology, by analyzing single EVs in plasma samples from patients with melanoma, where EV heterogeneity plays a critical role in disease progression and response to therapy. We demonstrate profiling of individual EVs for key immune markers PD-L1, CD155, and the melanoma marker TYRP-1, and showed that GDEVA can precisely quantify EVs, offering the resolution to detect rare EV subpopulations in complex clinical specimens.
Collapse
Affiliation(s)
- Juhwan Park
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| | - Michelle Feng
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Jingbo Yang
- Department of Biology, School of Arts and Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Hanfei Shen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Zhiyuan Qin
- Department of Biology, School of Arts and Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Wei Guo
- Department of Biology, School of Arts and Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - David A Issadore
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
9
|
Xu Z, Yang X, Lu X, Su D, Wang Y, Wu H, Zhang Z, Long C, Su L, Wang Y, Chen H, Xiang S, Zhou B. PD-L1 antibody-modified plant-derived nanovesicles carrying a STING agonist for the combinational immunotherapy of melanoma. Biomaterials 2025; 322:123396. [PMID: 40367814 DOI: 10.1016/j.biomaterials.2025.123396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025]
Abstract
Combination therapies for melanoma face challenges due to asynchronous drug delivery and associated toxicity, underscoring the need for advanced delivery systems. While immune checkpoint inhibitors (ICIs) enhance T cell activity, optimal cytotoxic responses require efficient antigen presentation by mature dendritic cells (DCs), which are often functionally impaired in the tumor microenvironment. Thus, effective treatment requires coordinated T cell activation, DC-mediated priming, and direct tumor suppression. Herein, wild Glycyrrhiza uralensis Fisch roots-derived nanovesicles (GC NV) are demonstrated to be effective inhibitors of melanoma proliferation. The vesicles exert this activity through the intracellular delivery of encapsulated miRNA (miR2916) and bioactive molecules (isoliquiritigenin), with this capacity for intracellular delivery extending to the STING agonist DMXAA. We also demonstrate how chemical modification can be used to install PD-L1 antibodies on the membrane surface of these GC NV, imbuing these vesicles with selectivity for tumor cells. Combining DMXAA encapsulation with surface-displayed PD-L1 antibodies creates vesicles (GP@DMX NV) that both promote DCs maturation and elicit CD8+ T cell response. Our multifunctional GP@DMX NV reverse the immunosuppressive microenvironment of melanoma and significantly enhance the immunotherapeutic potential of immune checkpoints.
Collapse
Affiliation(s)
- Zhanxue Xu
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xinrui Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xingyu Lu
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Department of Orthopedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yidan Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Huixing Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhenhua Zhang
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Changrui Long
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Liqian Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yanyu Wang
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Shijian Xiang
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Benjie Zhou
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
10
|
Llach CD, Le GH, Badulescu S, Anmella G, Hasan HA, Giménez-Palomo A, Pacchiarotti I, Vieta E, McIntyre RS, Rosenblat JD, Mansur RB. Extracellular vesicles in mood disorders: A systematic review of human studies. Eur Neuropsychopharmacol 2025; 94:59-75. [PMID: 40057988 DOI: 10.1016/j.euroneuro.2025.02.009] [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: 12/18/2024] [Revised: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 05/02/2025]
Abstract
Extracellular vesicles (EVs) are small, membrane-bound particles that are naturally released by nearly all cell types in the body. They serve as molecular biosignatures, reflecting the state of their cells of origin and providing a non-invasive peripheral marker of central nervous system (CNS) activity under physiological and pathological conditions. We conducted a systematic review (ID: CRD42024528824) of studies investigating the use of EVs in mood disorders within clinical populations. We screened articles indexed in PubMed, EMBASE, Scopus, ISI Web of Science, and APA PsycInfo from January 2010 to October 2024. Available research has focused on four key areas: (1) EV cargo as mechanistic and diagnostic biomarkers; (2) EV cargo as predictive or tracking biomarkers for antidepressant response; (3) EV cargo and neuroimaging correlates; and (4) EV physical properties. Most studies examined major depressive disorder (MDD), with others addressing bipolar disorder (BD), adolescent depression, postpartum depression, and late-life depression. Notably, only 35,55 % of the studies utilized brain-derived EVs. Through analyses of EV-derived miRNA, proteins, mtDNA, and metabolites, these studies have explored neural mitochondrial function, brain insulin resistance, neurogenesis, neuroinflammation, and blood-brain barrier permeability in the context of mood disorders. Some EV-derived markers demonstrated diagnostic and predictive potential. This review discusses key findings, limitations of current research, and future directions for leveraging EVs in the study of mood disorders.
Collapse
Affiliation(s)
- Cristian-Daniel Llach
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada.
| | - Gia Han Le
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Sebastian Badulescu
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Gerard Anmella
- Bipolar and Depressive Disorders Unit, Hospital Clínic de Barcelona, Barcelona, Spain; Institute of Neurosciences (UBNeuro); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Biomedical Research Networking Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III (ISCIII);; Department of Medicine, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Hayder Ali Hasan
- Department of Neurosciences, Psychiatry and Pediatric Psychiatry, Faculty of Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj, Napoca, Romania
| | - Anna Giménez-Palomo
- Bipolar and Depressive Disorders Unit, Hospital Clínic de Barcelona, Barcelona, Spain; Institute of Neurosciences (UBNeuro); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Isabella Pacchiarotti
- Bipolar and Depressive Disorders Unit, Hospital Clínic de Barcelona, Barcelona, Spain; Institute of Neurosciences (UBNeuro); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Biomedical Research Networking Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III (ISCIII);; Department of Medicine, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Hospital Clínic de Barcelona, Barcelona, Spain; Institute of Neurosciences (UBNeuro); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Biomedical Research Networking Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III (ISCIII);; Department of Medicine, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Roger S McIntyre
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Rodrigo B Mansur
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| |
Collapse
|
11
|
Théry C, Louvard D. The roles and applications of extracellular vesicles in cancer. Mol Oncol 2025; 19:1287-1290. [PMID: 39989268 PMCID: PMC12077283 DOI: 10.1002/1878-0261.70007] [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: 01/22/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025] Open
Abstract
Extracellular vesicles (EVs) have been studied for several decades and are attracting growing interest among life scientists and oncologists. Understanding the extent of diversity of their cellular origins, structure, molecular composition, and consequently functions is still under progress. EVs offer numerous diagnostic and therapeutic possibilities, but many fundamental questions about their functions need to be resolved in order to effectively and safely implement their applications in the treatment of human diseases.
Collapse
Affiliation(s)
- Clotilde Théry
- Institut Curie Research CenterPSL Research University, INSERM U932, Immunity and Cancer, and CurieCoreTech Extracellular VesiclesParisFrance
| | - Daniel Louvard
- Institut Curie Research CenterPSL Research University, CNRS UMR 144, Cell Biology and CancerParisFrance
| |
Collapse
|
12
|
Hoang VT, Nguyen QT, Phan TTK, Pham TH, Dinh NTH, Anh LPH, Dao LTM, Bui VD, Dao H, Le DS, Ngo ATL, Le Q, Nguyen Thanh L. Tissue Engineering and Regenerative Medicine: Perspectives and Challenges. MedComm (Beijing) 2025; 6:e70192. [PMID: 40290901 PMCID: PMC12022429 DOI: 10.1002/mco2.70192] [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: 07/06/2024] [Revised: 12/30/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
From the pioneering days of cell therapy to the achievement of bioprinting organs, tissue engineering, and regenerative medicine have seen tremendous technological advancements, offering solutions for restoring damaged tissues and organs. However, only a few products and technologies have received United States Food and Drug Administration approval. This review highlights significant progress in cell therapy, extracellular vesicle-based therapy, and tissue engineering. Hematopoietic stem cell transplantation is a powerful tool for treating many diseases, especially hematological malignancies. Mesenchymal stem cells have been extensively studied. The discovery of induced pluripotent stem cells has revolutionized disease modeling and regenerative applications, paving the way for personalized medicine. Gene therapy represents an innovative approach to the treatment of genetic disorders. Additionally, extracellular vesicle-based therapies have emerged as rising stars, offering promising solutions in diagnostics, cell-free therapeutics, drug delivery, and targeted therapy. Advances in tissue engineering enable complex tissue constructs, further transforming the field. Despite these advancements, many technical, ethical, and regulatory challenges remain. This review addresses the current bottlenecks, emphasizing novel technologies and interdisciplinary research to overcome these hurdles. Standardizing practices and conducting clinical trials will balance innovation and regulation, improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Van T. Hoang
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Quyen Thi Nguyen
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Trang Thi Kieu Phan
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Trang H. Pham
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Nhung Thi Hong Dinh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Le Phuong Hoang Anh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Lan Thi Mai Dao
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Van Dat Bui
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- School of Chemical EngineeringCollege of EngineeringSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Hong‐Nhung Dao
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Duc Son Le
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Anh Thi Lan Ngo
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Quang‐Duong Le
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| |
Collapse
|
13
|
Assunção RRS, Santos NL, Andrade LNDS. Extracellular vesicles as cancer biomarkers and drug delivery strategies in clinical settings: Advances, perspectives, and challenges. Clinics (Sao Paulo) 2025; 80:100635. [PMID: 40315797 PMCID: PMC12090321 DOI: 10.1016/j.clinsp.2025.100635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/26/2025] [Indexed: 05/04/2025] Open
Abstract
Cancer is a leading cause of death worldwide, and despite the introduction of new therapeutic approaches for advanced cases aimed at improving patient survival, only a subset of patients benefits from a complete response. In this context, there is a growing need for new cancer biomarkers and therapeutic strategies, and the use of Extracellular Vesicles (EVs) has been widely explored in various approaches. As circulating lipid-bilayer particles carrying a variety of biological information from tumor cells, EVs can be employed as good biomarkers of diagnosis, prognosis, therapy evaluation, and as adjuvants in cancer treatment. In this review, we provide a brief overview of the different types of EVs and their biogenesis and discuss how tumor-derived EV cargo can serve as a potential biomarker in clinical settings through liquid biopsy. We also highlight recent advances in EV nanoengineering and their potential as adjuvants in cancer treatment. Finally, we discuss the key unknowns, gaps, and bottlenecks that must be addressed to fully integrate EVs into precision oncology.
Collapse
Affiliation(s)
- Raphaela Rebeca Silveira Assunção
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Nathalia Leal Santos
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Luciana Nogueira de Sousa Andrade
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
14
|
Li H, Liu H, Zhou Y, Cheng L, Wang B, Ma J. The multifaceted roles of extracellular vesicles in osteonecrosis of the femoral head. J Orthop Translat 2025; 52:70-84. [PMID: 40256260 PMCID: PMC12008682 DOI: 10.1016/j.jot.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a severe disease characterized by bone tissue necrosis due to vascular impairment, often leading to joint collapse and requiring surgical intervention. Extracellular vesicles (EVs) serve as crucial mediators of intercellular communication, influencing osteogenesis, angiogenesis, and immune regulation. This review summarizes the dual role of EVs in both the pathogenesis of ONFH and post-necrosis bone repair, highlighting the impact of various EV-mediated signaling pathways on bone regeneration and the potential crosstalk among these pathways. Additionally, EVs hold promise as diagnostic biomarkers or contrast agents to complement conventional imaging techniques for ONFH detection. By elucidating the role of EVs in osteonecrosis and addressing the current challenges, we aspire to establish a foundation for the timely identification and treatment of ONFH. The translational potential of this article: This review comprehensively discusses the role of EVs in ONFH, providing innovative and promising insights for its diagnosis and treatment, which also establishes a theoretical foundation for the future clinical application of EVs in ONFH.
Collapse
Affiliation(s)
- Hongxu Li
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Haoyang Liu
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Yu Zhou
- Department of Orthopaedic Surgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Liming Cheng
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Bailiang Wang
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jinhui Ma
- Department of Orthopaedic Surgery, Center for Osteonecrosis and Joint Preserving & Reconstruction, China-Japan Friendship Hospital, Beijing, 100029, China
| |
Collapse
|
15
|
Tandoh KZ, Avalos-Padilla Y, Ameyaw P, Laryea-Akrong EK, Awandare GA, Wilson MD, Quashie NB, Fernàndez-Busquets X, Duah-Quashie NO. Extracellular Vesicle Abundance, but Not a High Aggregation-Prone Peptide Cargo, Is Associated with Dihydroartemisinin Exposure in Plasmodium falciparum. Int J Mol Sci 2025; 26:3962. [PMID: 40362203 PMCID: PMC12072043 DOI: 10.3390/ijms26093962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 05/15/2025] Open
Abstract
Our understanding of the molecular mechanisms undergirding artemisinin (ART) resistance in Plasmodium falciparum is currently based on two organizing principles: reduced hemoglobin trafficking into the digestive food vacuole, resulting in lower levels of activated ART, and increased tolerance to ART-induced oxidative stress in the infected erythrocyte. We had previously proposed an extracellular vesicle (EV) export model of ART resistance in P. falciparum. This model predicts that EV abundance will be altered by ART exposure and that the peptide cargo of EVs from the ART-exposed condition will be enriched with aggregation-prone peptides. We tested the predictions of the EV export hypothesis in this study using in vitro culture assays of an ART-resistant transgenic line engineered on a 3D7 background (R561H) and a 3D7 knock-out line (PfVps60KO) with deficient EV production phenotype. EV enrichment was obtained from in vitro parasite culture supernatants via a series of ultracentrifugation and filtration steps, followed by size exclusion chromatography. A quality check on EVs was performed using dynamic light scattering. Liquid chromatography with tandem mass spectrometry was used to determine the proteome cargo from extracted EVs, and parasite peptides were queried for aggregation-prone tendency using open-access software. We report that dihydroartemisinin (DHA) exposure was positively correlated with EV abundance (coefficient estimate = 1038.58, confidence interval of 194.86-1882.30, and p-value = 0.018) and suggests that EV biogenesis is part of the parasite's response to DHA/ART. Furthermore, our findings suggest the expression of a non-constitutive DHA-induced alternate EV biogenesis pathway as the PfVps60KO was observed to produce the highest number of EVs under DHA exposure. Finally, we show that EVs from both ART-susceptible and resistant parasites under DHA exposure carry a cargo of Chorein N-terminal domain-containing protein (PF3D7_1021700) with a high aggregation-prone index (prion-like domain [PrLD] score = 26.5) out of nine identified parasite peptides. The former of these findings is in concordance with the EV export hypothesis, which posits that the removal of DHA/ART-induced aggregated and/or misfolded peptides is critical to the parasite's survival under DHA/ART exposure. This observation further implicates EVs in the development of the ART-resistant phenotype. However, the finding of one aggregation-prone peptide out of the nine parasite proteins in the EV cargo does not sufficiently support the EV export hypothesis. Future replicates of this study and further interrogations of the EV export hypothesis are needed.
Collapse
Affiliation(s)
- Kwesi Z. Tandoh
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (E.K.L.-A.); (G.A.A.); (N.B.Q.); (N.O.D.-Q.)
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana;
| | - Yunuen Avalos-Padilla
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10-12, 08028 Barcelona, Spain;
- Barcelona Institute for Global Health (ISGlobal, Hospital Clınic-Universitat de Barcelona), Rosselló 149-153, 08036 Barcelona, Spain
| | - Prince Ameyaw
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana;
| | - Elisabeth K. Laryea-Akrong
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (E.K.L.-A.); (G.A.A.); (N.B.Q.); (N.O.D.-Q.)
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana;
| | - Gordon A. Awandare
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (E.K.L.-A.); (G.A.A.); (N.B.Q.); (N.O.D.-Q.)
| | - Michael David Wilson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana
| | - Neils B. Quashie
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (E.K.L.-A.); (G.A.A.); (N.B.Q.); (N.O.D.-Q.)
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana;
- Centre for Tropical Clinical Pharmacology and Therapeutics, School of Medicine and Dentistry, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana
| | - Xavier Fernàndez-Busquets
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10-12, 08028 Barcelona, Spain;
- Barcelona Institute for Global Health (ISGlobal, Hospital Clınic-Universitat de Barcelona), Rosselló 149-153, 08036 Barcelona, Spain
| | - Nancy O. Duah-Quashie
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (E.K.L.-A.); (G.A.A.); (N.B.Q.); (N.O.D.-Q.)
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana;
| |
Collapse
|
16
|
Li X, Lu X, Liu M, Chen J, Lu X. Extracellular vesicles: messengers of cross-talk between gastric cancer cells and the tumor microenvironment. Front Cell Dev Biol 2025; 13:1561856. [PMID: 40309240 PMCID: PMC12040901 DOI: 10.3389/fcell.2025.1561856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Gastric cancer is a common malignancy characterized by an insidious onset and high mortality rate. Exosomes, a special type of extracellular vesicle, contain various bioactive molecules and have been found to play crucial roles in maintaining normal physiological functions and homeostasis in the body. Recent research has shown that the contents of exosome play a significant role in the progression and metastasis of gastric cancer through communication and regulatory functions. These mechanisms involve promoting gastric cancer cell proliferation and drug resistance. Additionally, other cells in the gastric cancer microenvironment can regulate the progression of gastric cancer through exosomes. These include exosomes derived from fibroblasts and immune cells, which modulate gastric cancer cells. Therefore, in this review, we provide a brief overview of recent advances in the contents and occurrence mechanisms of exosome. This review specifically focused on the regulatory mechanisms of exosomes derived from gastric cancer and other cellular subtypes in the tumor microenvironment. Subsequently, we summarize the latest research progress on the use of exosomes in liquid biopsy, discussing the potential of gastric cancer exosomes in clinical applications.
Collapse
Affiliation(s)
- Xiwen Li
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Xian Lu
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| | - Mi Liu
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Junjie Chen
- Department of Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Xirong Lu
- Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Kunshan, China
| |
Collapse
|
17
|
Du G, He J, Zhan Y, Chen L, Hu Y, Qian J, Huang H, Meng F, Shan L, Chen Z, Hu D, Zhu C, Yue M, Qi Y, Tan W. Changes and application prospects of biomolecular materials in small extracellular vesicles (sEVs) after flavivirus infection. Eur J Med Res 2025; 30:275. [PMID: 40229861 PMCID: PMC11998145 DOI: 10.1186/s40001-025-02539-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Small extracellular vesicles (sEVs), also known as exosomes, are membranous vesicles filled with various proteins and nucleic acids, serving as a communication vector between cells. Recent research has highlighted their role in viral diseases. This review synthesizes current understanding of viral sEVs and includes recent findings on sEVs infected with flaviviruses. It discusses the implications of viral sEVs research for advancing arbovirus sEVs research and anticipates the potential applications of sEVs in flavivirus infections.
Collapse
Affiliation(s)
- Gengting Du
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Junhua He
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Yan Zhan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Leru Chen
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Yue Hu
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Jiaojiao Qian
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Huan Huang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Fanjin Meng
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Laiyou Shan
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | - Zhiyu Chen
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China
| | | | - Changqiang Zhu
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Ming Yue
- Department of Infectious Diseases, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yong Qi
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China
| | - Weilong Tan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, People's Republic of China.
- Nanjing Bioengineering (Gene) Technology Center for Medicines, Nanjing, People's Republic of China.
- Nanjing Jinling Hospital, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Rey-Cadilhac F, Rachenne F, Marquant A, Kee Him JL, Ancelin A, Foisor V, Morille M, Lyonnais S, Cazevieille C, Missé D, Pompon J. Characterization of size distribution and markers for mosquito extracellular vesicles. Front Cell Dev Biol 2025; 13:1497795. [PMID: 40292329 PMCID: PMC12021844 DOI: 10.3389/fcell.2025.1497795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
Extracellular vesicles (EVs) are non-replicative, cell-derived membranous structures secreted by potentially all eukaryotic cells, playing a crucial role in intercellular communication. The study of EVs requires approaches and tools, which have predominantly been developed for mammalian models. Here, we undertook a multimodal characterization of mosquito EVs to provide a technical and knowledge foundation for their study. First, using a cell line model from Aedes aegypti and applying multiple analytical technologies (i.e., NTA, TEM, cryo-EM, and AFM), we observed that mosquito EVs range from 20 to 500 nm in diameter and that a majority are smaller than 100 nm. Second, we showed that smaller EVs are secreted in mosquito saliva. Third, we evaluated the capacity of differential centrifugation and size exclusion chromatography to separate mosquito EVs, revealing the strengths and weaknesses of each technology. Finally, we identified a mosquito homolog of CD63 as an extravesicular marker and the mosquito syntenin as a putative luminal marker. Overall, our results promote the development of tools and approaches for the study of mosquito EVs.
Collapse
Affiliation(s)
| | | | | | - Josephine Lai Kee Him
- CBS (Centre de Biologie Structurale), Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | - Aurélie Ancelin
- CBS (Centre de Biologie Structurale), Univ. Montpellier, CNRS, Inserm, Montpellier, France
| | | | - Marie Morille
- ICGM, Univ. Montpellier, CNRS, ENSCM, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| | | | - Chantal Cazevieille
- INM (Institut de Neuroscience de Montpellier), Electronic Microscopy Plateform, Saint Eloi Hospital, Montpellier, France
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| | - Julien Pompon
- MIVEGEC, Univ. Montpellier, IRD, CNRS, Montpellier, France
| |
Collapse
|
19
|
Hnath B, Dokholyan NV. Novel extracellular vesicle release pathway facilitated by toxic superoxide dismutase 1 oligomers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647611. [PMID: 40291716 PMCID: PMC12026985 DOI: 10.1101/2025.04.07.647611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease resulting in paralysis and death within three to five years. Mutations in over forty different proteins have been linked to ALS, leading to controversy whether ALS is one disease or many diseases with a similar phenotype. Mutations in Cu,Zn superoxide dismutase 1 (SOD1) are only found in 2-3% of ALS cases, yet misfolded SOD1 is found in both sporadic (sALS) and familial (fALS) patients. Yet, mutations in TDP-43 or FUS increase the level of misfolded SOD1 on extracellular vesicles (EVs). Additionally, small EVs isolated from ALS patient samples caused cell death of wild type motor neurons and myotubules. The toxicity and protein alterations of ALS EVs have led to the theory that EVs are responsible for the spread of ALS. We hypothesize that previously-identified toxic trimeric SOD1 is spreading on EVs in ALS and altering the spread of other ALS-related proteins, linking them to a common mechanism. To test our hypothesis, we isolate EVs from motor neuron-like cells expressing trimer stabilizing mutations and perform a sandwich enzyme-linked immunoassay (ELISA) (CD9 capture antibody) to quantify whether misfolded SOD1 and 17 other ALS-related proteins increase or decrease on EVs with trimer stabilization. We identify which EV release pathway is being affected by trimeric SOD1 utilizing endocytosis and exocytosis inhibitors, and determine if any specific EV-related proteins are altered with trimer stabilization. We establish that VAPB, VCP, and Stathmin-2 increase on EVs with trimer stabilization. The common pathway between SOD1 and three other ALS-associated proteins is affected by multiple pathways, including the Caveolae endocytosis pathway, suggesting a novel hybrid pathway of EV release present in ALS.
Collapse
|
20
|
Yadav P, Debnath N, Pradhan D, Mehta PK, Kumar A, Yadav ML, Yadav AK. Probiotic Lactobacillus-Derived Extracellular Vesicles: Insights Into Disease Prevention and Management. Mol Nutr Food Res 2025:e70013. [PMID: 40200671 DOI: 10.1002/mnfr.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 04/10/2025]
Abstract
Bacterial extracellular vesicles (BEVs) have emerged as versatile and promising tools for therapeutic interventions across a spectrum of medical applications. Among these, Lactobacillus-derived extracellular vesicles (LDEVs) have garnered significant attention due to their diverse physiological functions and applications in health advancement. These LDEVs modulate host cell signaling pathways through the delivery of bioactive molecules, including nucleic acids and proteins. The immunomodulatory properties of LDEVs are important, as they have been shown to regulate the balance between pro-inflammatory and anti-inflammatory responses in various diseases. These LDEVs play a crucial role in maintaining gut homeostasis by modulating the composition and function of the gut microbiota, which has implications for health conditions, including inflammatory bowel diseases, metabolic disorders, and neurological disorders. Furthermore, LDEVs hold potential to deliver therapeutic payloads to specific tissues or organs. Engineered LDEVs can be loaded with therapeutic agents such as antimicrobial peptides or nucleic acid-based therapies to treat various diseases. By leveraging the unique properties of LDEVs, researchers can develop innovative strategies for disease prevention, treatment, and overall well-being. Thus, this review aims to provide a comprehensive overview of the therapeutic benefits of LDEVs and their implications for promoting overall well-being.
Collapse
Affiliation(s)
- Pooja Yadav
- Centre for Molecular Biology, Central University of Jammu, Jammu, Jammu & Kashmir, India
| | - Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Jammu, Jammu & Kashmir, India
| | - Diwas Pradhan
- Dairy Microbiology Division, National Dairy Research Institute, Karnal, Haryana, India
| | - Praveen Kumar Mehta
- Centre for Molecular Biology, Central University of Jammu, Jammu, Jammu & Kashmir, India
| | - Ashwani Kumar
- Department of Nutrition Biology, Central University of Haryana, Mahendergarh, Haryana, India
| | - Munna Lal Yadav
- Discovery Research Division, Indian Council of Medical Research (ICMR), New Delhi, India
| | - Ashok Kumar Yadav
- Centre for Molecular Biology, Central University of Jammu, Jammu, Jammu & Kashmir, India
- Department of Zoology, Central University of Jammu, Jammu, Jammu & Kashmir, India
| |
Collapse
|
21
|
Li W, Fang W, Zhang Y, Chen Q, Shentu W, Lai Q, Cheng L, Yan S, Kong Q, Qiao S. Research progress on resistance exercise therapy for improving cognitive function in patients with AD and muscle atrophy. Front Aging Neurosci 2025; 17:1552905. [PMID: 40271180 PMCID: PMC12016217 DOI: 10.3389/fnagi.2025.1552905] [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: 12/29/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Alzheimer's disease (AD) significantly reduces the quality of life of patients and exacerbates the burden on their families and society. Resistance exercise significantly enhances the overall cognitive function of the elderly and patients with AD while positively improving memory, executive function, and muscle strength, reducing fall risks, and alleviating psychological symptoms. As AD is a neurodegenerative disorder, some nerve factors are readily activated and released during exercise. Therefore, several prior studies have concentrated on exploring the molecular mechanisms of resistance exercise and their impact on brain function and neural plasticity. Recent investigations have identified an intrinsic relationship between individuals with AD and the pathological mechanisms of skeletal muscle atrophy, establishing a correlation between patients with AD cognitive level and skeletal muscle content. Resistance exercise primarily targets the skeletal muscle, which improves cognitive impairment in patients with AD by reducing vascular and neuroinflammatory factors and further enhances cognitive function in patients with AD by restoring the structural function of skeletal muscle. Furthermore, the effects of resistance training vary among distinct subgroups of cognitive impairment. Individuals exhibiting lower cognitive function demonstrate more pronounced adaptive responses in physical performance over time. Consequently, further investigation is warranted to determine whether tailored guidelines-such as variations in the frequency and duration of resistance exercise-should be established for patients with varying levels of dementia, in order to optimize the benefits for those experiencing cognitive impairment. This study aimed to review the relationship between AD and skeletal muscle atrophy, the impact of skeletal muscle atrophy on AD cognition, the mechanism by which resistance exercise improves cognition through skeletal muscle improvement, and the optimal resistance exercise mode to elucidate the additional advantages of resistance exercise in treating cognitive function in patients with AD and skeletal muscle atrophy.
Collapse
Affiliation(s)
- Wenyao Li
- Department of Special Inspection, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Wei Fang
- Department of Neurology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yier Zhang
- Zhejiang Chinese Medical University Hangzhou, Hangzhou, Zhejiang, China
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Qiulu Chen
- Department of Neurology, Zhejiang Medical and Health Group Hangzhou Hospital, Hangzhou, Zhejiang, China
| | - Wuyue Shentu
- Zhejiang Chinese Medical University Hangzhou, Hangzhou, Zhejiang, China
| | - Qilun Lai
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Lin Cheng
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Sicheng Yan
- Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Qi Kong
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Song Qiao
- Department of Neurology, Zhejiang Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Ragab SS. Signature of click chemistry in advanced techniques for cancer therapeutics. RSC Adv 2025; 15:10583-10601. [PMID: 40190630 PMCID: PMC11970365 DOI: 10.1039/d5ra01196e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Click chemistry has made a revolution in the field of chemical biology owing to its high efficiency, specificity, and mild reaction conditions. The copper(i)-catalyzed azide-alkyne cycloaddition (CuAAC) and strain-promoted [3 + 2] azide-alkyne cycloaddition (SPAAC) stand out as the most popular click reactions that construct a stable triazole ring by reacting an azide with an alkyne. These two reactions represent an ideal choice for biological applications due to its specificity, reliability, and biocompatibility. As a powerful modular synthetic approach for creating new molecular entities, it has seen increasing use in anticancer drug discovery. The present "state of the art" focuses mainly on the signature of click chemistry (CuAAC and SPAAC) in advanced techniques for cancer therapeutics, which includes cancer immunotherapy, antibody-drug conjugates, development of proteolysis-targeting chimeras, targeted dual-agent combination therapy for cancer, exosome modification for cancer therapy, and photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Sherif Shaban Ragab
- Photochemistry Department, Chemical Industries Research Institute, National Research Centre El-Buhouth St, P.O. 12622, Dokki Giza Egypt
| |
Collapse
|
23
|
Araujo-Abad S, Berna JM, Lloret-Lopez E, López-Cortés A, Saceda M, de Juan Romero C. Exosomes: from basic research to clinical diagnostic and therapeutic applications in cancer. Cell Oncol (Dordr) 2025; 48:269-293. [PMID: 39298081 PMCID: PMC11997007 DOI: 10.1007/s13402-024-00990-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer continues to pose a global threat despite potent anticancer drugs, often accompanied by undesired side effects. To enhance patient outcomes, sophisticated multifunctional approaches are imperative. Small extracellular vesicles (EVs), a diverse family of naturally occurring vesicles derived from cells, offer advantages over synthetic carriers. Among the EVs, the exosomes are facilitating intercellular communication with minimal toxicity, high biocompatibility, and low immunogenicity. Their tissue-specific targeting ability, mediated by surface molecules, enables precise transport of biomolecules to cancer cells. Here, we explore the potential of exosomes as innovative therapeutic agents, including cancer vaccines, and their clinical relevance as biomarkers for clinical diagnosis. We highlight the cargo possibilities, including nucleic acids and drugs, which make them a good delivery system for targeted cancer treatment and contrast agents for disease monitoring. Other general aspects, sources, and the methodology associated with therapeutic cancer applications are also reviewed. Additionally, the challenges associated with translating exosome-based therapies into clinical practice are discussed, together with the future prospects for this innovative approach.
Collapse
Affiliation(s)
- Salomé Araujo-Abad
- Cancer Research Group, Faculty of Engineering and Applied Sciences, Universidad de Las Américas, Quito, 170124, Ecuador
| | - José Marcos Berna
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Elena Lloret-Lopez
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, 170124, Ecuador
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Camino de Juan Romero
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain.
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain.
| |
Collapse
|
24
|
Yadav K, Sahu KK, Sucheta, Minz S, Pradhan M. Unlocking exosome therapeutics: The critical role of pharmacokinetics in clinical applications. Tissue Cell 2025; 93:102749. [PMID: 39904192 DOI: 10.1016/j.tice.2025.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Exosomes are microscopic vesicles released by cells that transport various biological materials and play a vital role in intercellular communication. When they are engineered, they serve as efficient delivery systems for therapeutic agents, making it possible to precisely deliver active pharmaceutical ingredients to organs, tissues, and cells. Exosomes' pharmacokinetics, or how they are transported and metabolized inside the body, is affected by several factors, including their source of origination and the proteins in their cell membranes. The pharmacokinetics and mobility of both native and modified exosomes are being observed in living organisms using advanced imaging modalities such as in vitro-in vivo simulation, magnetic resonance imaging, and positron emission tomography. Establishing comprehensive criteria for the investigation of exosomal pharmacokinetic is essential, given its increasing significance in both therapy and diagnostics. To obtain a thorough understanding of exosome intake, distribution, metabolism, and excretion, molecular imaging methods are crucial. The development of industrial processes and therapeutic applications depends on the precise measurement of exosome concentration in biological samples. To ensure a seamless incorporation of exosomes into clinical practice, as their role in therapeutics grows, it is imperative to conduct a complete assessment of their pharmacokinetics. This review provides a brief on how exosome-based research is evolving and the need for pharmacokinetic consideration to realize the full potential of these promising new therapeutic approaches.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 491024, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 11 122103, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | |
Collapse
|
25
|
Semeradtova A, Liegertova M, Herma R, Capkova M, Brignole C, Del Zotto G. Extracellular vesicles in cancer´s communication: messages we can read and how to answer. Mol Cancer 2025; 24:86. [PMID: 40108630 PMCID: PMC11921637 DOI: 10.1186/s12943-025-02282-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Extracellular vesicles (EVs) are emerging as critical mediators of intercellular communication in the tumor microenvironment (TME), profoundly influencing cancer progression. These nano-sized vesicles, released by both tumor and stromal cells, carry a diverse cargo of proteins, nucleic acids, and lipids, reflecting the dynamic cellular landscape and mediating intricate interactions between cells. This review provides a comprehensive overview of the biogenesis, composition, and functional roles of EVs in cancer, highlighting their significance in both basic research and clinical applications. We discuss how cancer cells manipulate EV biogenesis pathways to produce vesicles enriched with pro-tumorigenic molecules, explore the specific contributions of EVs to key hallmarks of cancer, such as angiogenesis, metastasis, and immune evasion, emphasizing their role in shaping TME and driving therapeutic resistance. Concurrently, we submit recent knowledge on how the cargo of EVs can serve as a valuable source of biomarkers for minimally invasive liquid biopsies, and its therapeutic potential, particularly as targeted drug delivery vehicles and immunomodulatory agents, showcasing their promise for enhancing the efficacy and safety of cancer treatments. By deciphering the intricate messages carried by EVs, we can gain a deeper understanding of cancer biology and develop more effective strategies for early detection, targeted therapy, and immunotherapy, paving the way for a new era of personalized and precise cancer medicine with the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Alena Semeradtova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic.
| | - Michaela Liegertova
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Regina Herma
- Centre for Nanomaterials and Biotechnology, Faculty of Science, Jan Evangelista Purkyně University in Ústí Nad Labem, Pasteurova 3632/15, Ústí Nad Labem, 40096, Czech Republic
| | - Magdalena Capkova
- Institute of Photonics and Electronics of the CAS, Chaberská 1014/57, Prague, 182 51, Czech Republic
| | - Chiara Brignole
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147, Genoa, Italy.
| | - Genny Del Zotto
- Core Facilities, Department of Research and Diagnostics, IRCCS Istituto Giannina Gaslini, 16147, Genoa, Italy.
| |
Collapse
|
26
|
Erasha AM, EL-Gendy H, Aly AS, Fernández-Ortiz M, Sayed RKA. The Role of the Tumor Microenvironment (TME) in Advancing Cancer Therapies: Immune System Interactions, Tumor-Infiltrating Lymphocytes (TILs), and the Role of Exosomes and Inflammasomes. Int J Mol Sci 2025; 26:2716. [PMID: 40141358 PMCID: PMC11942452 DOI: 10.3390/ijms26062716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Understanding how different contributors within the tumor microenvironment (TME) function and communicate is essential for effective cancer detection and treatment. The TME encompasses all the surroundings of a tumor such as blood vessels, fibroblasts, immune cells, signaling molecules, exosomes, and the extracellular matrix (ECM). Subsequently, effective cancer therapy relies on addressing TME alterations, known drivers of tumor progression, immune evasion, and metastasis. Immune cells and other cell types act differently under cancerous conditions, either driving or hindering cancer progression. For instance, tumor-infiltrating lymphocytes (TILs) include lymphocytes of B and T cell types that can invade malignancies, bringing in and enhancing the ability of immune system to recognize and destroy cancer cells. Therefore, TILs display a promising approach to tackling the TME alterations and have the capability to significantly hinder cancer progression. Similarly, exosomes and inflammasomes exhibit a dual effect, resulting in either tumor progression or inhibition depending on the origin of exosomes, type of inflammasome and tumor. This review will explore how cells function in the presence of a tumor, the communication between cancer cells and immune cells, and the role of TILs, exosomes and inflammasomes within the TME. The efforts in this review are aimed at garnering interest in safer and durable therapies for cancer, in addition to providing a promising avenue for advancing cancer therapy and consequently improving survival rates.
Collapse
Affiliation(s)
- Atef M. Erasha
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Hanem EL-Gendy
- Department of Pharmacology, Faculty of Veterinary Medicine, Sadat City University, Sadat City 32897, Egypt;
| | - Ahmed S. Aly
- Department of Animal Production, Faculty of Agriculture, Ain Shams University, Cairo 11241, Egypt;
| | - Marisol Fernández-Ortiz
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Ramy K. A. Sayed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Sohag University, Sohag 82524, Egypt;
| |
Collapse
|
27
|
An Y, Sun JX, Ma SY, Xu MY, Xu JZ, Liu CQ, Wang SG, Xia QD. From Plant Based Therapy to Plant-Derived Vesicle-Like Nanoparticles for Cancer Treatment: Past, Present and Future. Int J Nanomedicine 2025; 20:3471-3491. [PMID: 40125436 PMCID: PMC11927496 DOI: 10.2147/ijn.s499893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
Cancer stands as a formidable malady profoundly impacting human health. Throughout history, plant-based therapies have remained pivotal in the arsenal against cancer, evolving alongside the epochs. Presently, challenges such as the arduous extraction of active components and potential safety concerns impede the progression of plant-based anticancer therapies. The isolation of plant-derived vesicle-like nanoparticles (PDVLNs), a kind of lipid bilayer capsules isolated from plants, has brought plant-based anticancer therapy into a novel realm and has led to decades of research on PDVLNs. Accumulating evidence indicates that PDVLNs can deliver plant-derived active substances to human cells and regulate cellular functions. Regulating immunity, inducing cell cycle arrest, and promoting apoptosis in cancer cells are the most commonly reported mechanisms of PDVLNs in tumor suppression. Low immunogenicity and lack of tumorigenicity make PDVLNs a good platform for drug delivery. The molecules within the PDVLNs are all from source plants, so the selection of source plants is crucial. In recent years, there has been a clear trend that the source plants have changed from vegetables or fruits to medicinal plants. This review highlights the mechanisms of medicinal plant-based cancer therapies to identify candidate source plants. More importantly, the current research on PDVLN-based cancer therapy and the applications of PDVLNs for drug delivery are systematically discussed.
Collapse
Affiliation(s)
- Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
28
|
Hao Z, Guan W, Wei W, Li M, Xiao Z, Sun Q, Pan Y, Xin W. Unlocking the therapeutic potential of tumor-derived EVs in ischemia-reperfusion: a breakthrough perspective from glioma and stroke. J Neuroinflammation 2025; 22:84. [PMID: 40089793 PMCID: PMC11909855 DOI: 10.1186/s12974-025-03405-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
Clinical studies have revealed a bidirectional relationship between glioma and ischemic stroke, with evidence of spatial overlap between the two conditions. This connection arises from significant similarities in their pathological processes, including the regulation of cellular metabolism, inflammation, coagulation, hypoxia, angiogenesis, and neural repair, all of which involve common biological factors. A significant shared feature of both diseases is the crucial role of extracellular vesicles (EVs) in mediating intercellular communication. Extracellular vesicles, with their characteristic bilayer structure, encapsulate proteins, lipids, and nucleic acids, shielding them from enzymatic degradation by ribonucleases, deoxyribonucleases, and proteases. This structural protection facilitates long-distance intercellular communication in multicellular organisms. In gliomas, EVs are pivotal in intracranial signaling and shaping the tumor microenvironment. Importantly, the cargos carried by glioma-derived EVs closely align with the biological factors involved in ischemic stroke, underscoring the substantial impact of glioma on stroke pathology, particularly through the crucial roles of EVs as key mediators in this interaction. This review explores the pathological interplay between glioma and ischemic stroke, addressing clinical manifestations and pathophysiological processes across the stages of hypoxia, stroke onset, progression, and recovery, with a particular focus on the crucial role of EVs and their cargos in these interactions.
Collapse
Affiliation(s)
- Zhongnan Hao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Neurology, The Affiliated Hospital of Qingdao University, Medical School of Qingdao University, Qingdao, 266100, Shandong Province, China
| | - Wenxin Guan
- Queen Mary School, Nanchang University, Xuefu Avenue, Nanchang, Jiangxi, China
| | - Wei Wei
- Department of Neurology, the Affiliated Hospital of Southwest Jiaotong University & The Third People's Hospital of Chengdu, Chengdu, 610031, Sichuan, PR China
| | - Meihua Li
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhipeng Xiao
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qinjian Sun
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China
| | - Yongli Pan
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P. R. China.
| | - Wenqiang Xin
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
29
|
Ribas-Maynou J, Parra A, Martínez-Díaz P, Rubio CP, Lucas X, Yeste M, Roca J, Barranco I. Protective role of extracellular vesicles against oxidative DNA damage. Biol Res 2025; 58:14. [PMID: 40075425 PMCID: PMC11905505 DOI: 10.1186/s40659-025-00595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Oxidative stress, a source of genotoxic damage, is often the underlying mechanism in many functional cell disorders. Extracellular vesicles (EVs) have been shown to be key regulators of cellular processes and may be involved in maintaining cellular redox balance. Herein, we aimed to develop a method to assess the effects of EVs on DNA oxidation using porcine seminal plasma extracellular vesicles (sEVs). RESULTS The technique was set using a cell-free plasmid DNA to avoid the bias generated by the uptake of sEVs by sperm cells, employing increasing concentrations of hydrogen peroxide (H2O2) that generate DNA single-strand breaks (SSBs). Because SSBs contain a free 3'-OH end that allow the extension through quantitative PCR, such extension -and therefore the SYBR intensity- showed to be proportional to the amount of SSB. In the next stage, H2O2 was co-incubated with two size-differentiated subpopulations (small and large) of permeabilized and non-permeabilized sEVs to assess whether the intravesicular content (IC) or the surface of sEVs protects the DNA from oxidative damage. Results obtained showed that the surface of small sEVs reduced the incidence of DNA SSBs (P < 0.05), whereas that of large sEVs had no impact on the generation of SSBs (P > 0.05). The IC showed no protective effect against DNA oxidation (P > 0.05). CONCLUSIONS Our results suggest that the surface of small sEVs, including the peripheral corona layer, may exert a protective function against alterations that are originated by oxidative mechanisms. In addition, our in vitro study opens path to detect, localize and quantify the protective effects against oxidation of extracellular vesicles derived from different fluids, elucidating their function in physiopathological states.
Collapse
Affiliation(s)
- Jordi Ribas-Maynou
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
- Unit of Cell Biology and Medical Genetics; Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Ana Parra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Pablo Martínez-Díaz
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Camila Peres Rubio
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
| | - Xiomara Lucas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (Technosperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Jordi Roca
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain.
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain.
| | - Isabel Barranco
- Department of Medicine and Animal Surgery, Faculty of Veterinary Science, University of Murcia, Murcia, Spain
- International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| |
Collapse
|
30
|
Liu Y, Wang C, Fu X, Ren M. The Progress and Evolving Trends in Nucleic-Acid-Based Therapies. Biomolecules 2025; 15:376. [PMID: 40149911 PMCID: PMC11940734 DOI: 10.3390/biom15030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Nucleic-acid-based therapies have emerged as a pivotal domain within contemporary biomedical science, marked by significant advancements in recent years. These innovative treatments primarily operate through the precise binding of DNA or RNA molecules to discrete target genes, subsequently suppressing the expression of the target proteins. The spectrum of nucleic-acid-based therapies encompasses antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), etc. Compared to more traditional medicinal approaches, nucleic-acid-based therapies stand out for their highly targeted action on specific genes, as well as their potential for chemical modification to improve resistance to nucleases, ensuring sustained therapeutic activity and mitigating immunogenicity concerns. Nevertheless, these molecules' limited cellular permeability necessitates the deployment of delivery vectors to enhance their intracellular uptake and stability. As nucleic-acid-based therapies progressively display promising pharmacodynamic profiles, there has been a burgeoning interest in these treatments for applications in clinical research. This review aims to summarize the variety of nucleic acid drugs and their mechanisms, evaluate the present status in research and application, discourse on prospective trends, and potential challenges ahead. These innovative therapeutics are anticipated to assume a pivotal role in the management of a wide array of diseases.
Collapse
Affiliation(s)
| | | | - Xiuping Fu
- School of Chemistry and School of Life Sciences, Tiangong University, Tianjin 300387, China; (Y.L.); (C.W.)
| | - Mengtian Ren
- School of Chemistry and School of Life Sciences, Tiangong University, Tianjin 300387, China; (Y.L.); (C.W.)
| |
Collapse
|
31
|
Wang X, Xu L, Wu Z, Lou L, Xia C, Miao H, Dai J, Fei W, Wang J. Exosomes of stem cells: a potential frontier in the treatment of osteoarthritis. PRECISION CLINICAL MEDICINE 2025; 8:pbae032. [PMID: 39781279 PMCID: PMC11705996 DOI: 10.1093/pcmedi/pbae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
The aging population has led to a global issue of osteoarthritis (OA), which not only impacts the quality of life for patients but also poses a significant economic burden on society. While biotherapy offers hope for OA treatment, currently available treatments are unable to delay or prevent the onset or progression of OA. Recent studies have shown that as nanoscale bioactive substances that mediate cell communication, exosomes from stem cell sources have led to some breakthroughs in the treatment of OA and have important clinical significance. This paper summarizes the mechanism and function of stem cell exosomes in delaying OA and looks forward to the development prospects and challenges of exosomes.
Collapse
Affiliation(s)
- Xiaofei Wang
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Lei Xu
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Zhimin Wu
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Linbing Lou
- The Graduate School, Dalian Medical University, Dalian 116044, China
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Cunyi Xia
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jihang Dai
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Wenyong Fei
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| | - Jingcheng Wang
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225001, China
| |
Collapse
|
32
|
Espinoza P, Cheng M, Ng C, Cruz DDL, Wasson ED, McCarthy DM, Bhide PG, Maguire CA, Santoscoy MC. Metabolic engineering improves transduction efficiency and downstream vector isolation by altering the lipid composition of extracellular vesicle-enclosed AAV. Metab Eng 2025; 88:40-49. [PMID: 39653070 PMCID: PMC11850184 DOI: 10.1016/j.ymben.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Adeno-associated viruses (AAV) are promising vectors for gene therapy due to their efficacy in vivo. However, there is room for improvement to address key limitations such as the pre-existing immunity to AAV in patients, high-dose toxicity, and relatively low efficiency for some cell types. This study introduces a metabolic engineering approach, using knockout of the enzyme phosphatidylserine synthase 1 (PTDSS1) to increase the abundance of extracellular vesicle-enclosed AAV (EV-AAV) relative to free AAV in the supernatant of producer cells, simplifying downstream purification processes. The lipid-engineered HEK293T-ΔPTDSS1 cell line achieved a 42.7-fold enrichment of EV-AAV9 compared to free AAV9 in the supernatant. The rational genetic strategy also led to a 300-fold decrease of free AAV in supernatant compared to wild-type HEK293T. The membrane-engineered EV-AAV9 (mEV-AAV9) showed unique envelope composition alterations, including cholesterol enrichment and improved transduction efficiency in human AC16 cardiomyocytes by 1.5-fold compared to conventional EV-AAV9 and by 11-fold compared to non-enveloped AAV9. Robust in-vivo transduction four weeks after intraparenchymal administration of mEV-AAV9 was observed in the murine brain. This study shows promise in the potential of lipid metabolic engineering strategies to improve the efficiency and process development of enveloped gene delivery vectors.
Collapse
Affiliation(s)
- Paula Espinoza
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Ming Cheng
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Carrie Ng
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Demitri de la Cruz
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA
| | - Elizabeth D Wasson
- FSU Institute for Pediatric Rare Diseases, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Deirdre M McCarthy
- FSU Institute for Pediatric Rare Diseases, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Pradeep G Bhide
- FSU Institute for Pediatric Rare Diseases, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Casey A Maguire
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Miguel C Santoscoy
- Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
33
|
Li P, Xu X, Zhang C, Chang Q, Wang J, Wang W, Ren H. Glycosylation on extracellular vesicles and its detection strategy: Paving the way for clinical use. Int J Biol Macromol 2025; 295:139714. [PMID: 39798737 DOI: 10.1016/j.ijbiomac.2025.139714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Extracellular vesicles (EVs) contain various glycans during their life cycle, from biogenesis to cellular recognition and uptake by recipient cells. EV glycosylation has substantial diagnostic significance in multiple health conditions, highlighting the necessity of determining an accurate glycosylation pattern for EVs from diverse biological fluids. Reliable and accessible glycan detection techniques help to elaborate the glycosylation-related functional alterations of specific proteins or lipids. However, multiple obstacles exist, including the inconsistency in glycosylation patterns between an entire batch of EVs and a specific EV protein, and difficulty in distinguishing glycosylation types after tedious separation and purification procedures. This review outlines recent advances in EV glycan detection, either at the glycomic level for a collection of intact EVs or at the molecular level for a specific protein on EVs. Particular emphasis has been placed on the abundance of EVs in body fluids and their unique characteristics for drug delivery of EVs, indicating an opportunity for diagnostic and therapeutic purposes via EV glycans.
Collapse
Affiliation(s)
- Ping Li
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266024, China
| | - Xiao Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China
| | - Cong Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210042, China
| | - Qi Chang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| | - Weijie Wang
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao 266024, China.
| | - He Ren
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
34
|
Mittal A, Jakhmola VR, Baweja S. Bioengineered extracellular vesicles: The path to precision medicine in liver diseases. LIVER RESEARCH (BEIJING, CHINA) 2025; 9:17-28. [PMID: 40206438 PMCID: PMC11977285 DOI: 10.1016/j.livres.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 04/11/2025]
Abstract
Extracellular vesicles (EVs) are membrane-bound entities secreted by each cell, categorized as, exosomes, microvesicles or apoptotic bodies based on their size and biogenesis. They serve as promising vectors for drug delivery due to their capacity to carry diverse molecular signatures reflective of their cell of origin. EV research has significantly advanced since their serendipitous discovery, with recent studies focusing on their roles in various diseases and their potential for targeted therapy. In liver diseases, EVs are particularly promising for precision medicine, providing diagnostic and therapeutic potential in conditions such as metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis, hepatocellular carcinoma, alcoholic liver disease, liver fibrosis, and acute liver failure. Despite challenges in isolation and characterization, engineered EVs have shown efficacy in delivering therapeutic agents with improved targeting and reduced side effects. As research progresses, EVs hold great promise to revolutionize precision medicine in liver diseases, offering targeted, efficient, and versatile therapeutic options. In this review, we summarize various techniques for loading EVs with therapeutic cargo including both passive and active methods, and the potential of bioengineered EVs loaded with various molecules, such as miRNAs, proteins, and anti-inflammatory drugs in ameliorating clinical pathologies of liver diseases.
Collapse
Affiliation(s)
| | | | - Sukriti Baweja
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| |
Collapse
|
35
|
Palma C, Masud MK, Guanzon D, Lai A, Razo M, Nakahara A, Nair S, Salas-Burgos A, Hossain MSA, Carrion F, Duncombe G, McIntyre HD, Handberg A, Longo S, Yamauchi Y, Salomon C. Rapid and high-sensitivity screening of pregnancy complications by profiling circulating placental extracellular vesicles. SCIENCE ADVANCES 2025; 11:eadr4074. [PMID: 40009685 PMCID: PMC11864196 DOI: 10.1126/sciadv.adr4074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Herein, we developed a specific, rapid sensor to quantify placental extracellular vesicle (EV) protein biomarkers of early pregnancy complications. A distinct tetraspanin CD9 and placental alkaline phosphatase (PLAP) expression pattern was observed via targeted multiple reaction monitoring of EVs from maternal plasma collected before 18 weeks of gestation. A classification model was developed using training and validation patient sets, distinguishing between individuals at high risk of developing complications from those with normal pregnancies, achieving 80% sensitivity, 90% specificity, 89% positive predictive value (PPV), and 82% negative predictive value (NPV). Superparamagnetic nanoflowers that captured target EVs (CD9+/PLAP+) were used to construct a 4-flex glass strip nanozymatic readout system. The sensor analyzes plasma for EVs, identifying gestational diabetes mellitus risk with a 95% combined sensitivity, 100% specificity, 100% PPV, and 96% NPV. This nanoplatform identifies individuals at risk of developing pregnancy complications with a >90% classification accuracy, exhibiting potential for clinical applications.
Collapse
Affiliation(s)
- Carlos Palma
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Mostafa Kamal Masud
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dominic Guanzon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Melissa Razo
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - Angela Nakahara
- Department of Obstetrics and Gynecology, Maternal-Fetal Medicine, Ochsner Clinic Foundation, New Orleans, LA 70115, USA
| | - Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
| | - Alexis Salas-Burgos
- Department of Pharmacology, Faculty of Biological Sciences, University of Concepción, Concepción 160C, Chile
| | - Md Shahriar A. Hossain
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
- School of Mechanical and Mining Engineering, Faculty of Engineering, Architecture, and Information Technology (EAIT), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago 8320000, Chile
| | - Gregory Duncombe
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| | - H. David McIntyre
- Mater Research, Faculty of Medicine, University of Queensland, Mater Health, South Brisbane 4101, Australia
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg 9220, Denmark
| | - Sherri Longo
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yusuke Yamauchi
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, Brisbane, QLD 4072, Australia
- Department of Materials Process Engineering Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, University of Queensland Centre for Clinical Research, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland, Herston, QLD 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4006, Australia
| |
Collapse
|
36
|
García-Barberán V, Gómez Del Pulgar ME, Guamán HM, Benito-Martin A. The times they are AI-changing: AI-powered advances in the application of extracellular vesicles to liquid biopsy in breast cancer. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:128-140. [PMID: 40206803 PMCID: PMC11977355 DOI: 10.20517/evcna.2024.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 01/03/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
Artificial intelligence (AI) is revolutionizing scientific research by facilitating a paradigm shift in data analysis and discovery. This transformation is characterized by a fundamental change in scientific methods and concepts due to AI's ability to process vast datasets with unprecedented speed and accuracy. In breast cancer research, AI aids in early detection, prognosis, and personalized treatment strategies. Liquid biopsy, a noninvasive tool for detecting circulating tumor traits, could ideally benefit from AI's analytical capabilities, enhancing the detection of minimal residual disease and improving treatment monitoring. Extracellular vesicles (EVs), which are key elements in cell communication and cancer progression, could be analyzed with AI to identify disease-specific biomarkers. AI combined with EV analysis promises an enhancement in diagnosis precision, aiding in early detection and treatment monitoring. Studies show that AI can differentiate cancer types and predict drug efficacy, exemplifying its potential in personalized medicine. Overall, the integration of AI in biomedical research and clinical practice promises significant changes and advancements in diagnostics, personalized medicine-based approaches, and our understanding of complex diseases like cancer.
Collapse
Affiliation(s)
- Vanesa García-Barberán
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - María Elena Gómez Del Pulgar
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Heidy M. Guamán
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Alberto Benito-Martin
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio, Madrid 28691, Spain
| |
Collapse
|
37
|
Zhang X, Yang M, Chen X, Zhang M, Peng Y, Lu M. Melatonin-pretreated mesenchymal stem cell-derived exosomes alleviate cavernous fibrosis in a rat model of nerve injury-induced erectile dysfunction via miR-145-5p/TGF-β/Smad axis. Stem Cell Res Ther 2025; 16:96. [PMID: 40001250 PMCID: PMC11863846 DOI: 10.1186/s13287-025-04173-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cavernous nerve injury-induced erectile dysfunction (CNI-ED) is a common complication after radical prostatectomy. Conventional treatment approaches have had little success in treating the severe cavernous fibrosis which is a consequence of CNI-ED. METHODS Pre-treatment of adipose-derived stem cells with melatonin allows for the extraction of active exosomes (MT-hASC-EVs) from the conditioned medium. The therapeutic effects of MT-hASC-EVs were assessed in a rat model of CNI-ED, and the anti-fibrotic properties were evaluated. MicroRNA sequencing was used to identify specific microRNAs highly expressed in MT-hASC-EVs, and differential microRNAs were screened for regulatory pathways through target gene enrichment analysis. Finally, the conclusions from bioinformatics analysis were validated through in vitro experiments. RESULTS Intracavernous injection of MT-hASC-EVs significantly restored erectile function and reduced the extent of corpus cavernosum fibrosis in the CNI-ED rat model. MT-hASC-EVs promoted the proliferation and anti-apoptotic effects of corpus cavernosum smooth muscle cells (CCSMCs) in vitro. Mechanistically, MT-hASC-EVs inhibit fibrosis by delivering miR-145-5p, which targets TGF-β2/Smad3 axis. CONCLUSIONS MT-hASCs-EVs can inhibit cavernous fibrosis and improve erectile function in a rat model of CNI-ED by targeting the miR-145-5p/TGF-β/Smad axis.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Mengbo Yang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Xinda Chen
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Ming Zhang
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Yiliang Peng
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China
| | - Mujun Lu
- Department of Urology and Andrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, NO. 145 Middle Shandong Road, Shanghai, 200001, China.
| |
Collapse
|
38
|
Li H, Zhang P, Lin M, Li K, Zhang C, He X, Gao K. Pyroptosis: candidate key targets for mesenchymal stem cell-derived exosomes for the treatment of bone-related diseases. Stem Cell Res Ther 2025; 16:68. [PMID: 39940049 PMCID: PMC11816542 DOI: 10.1186/s13287-025-04167-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Bone-related diseases impact a large portion of the global population and, due to their high disability rates and limited treatment options, pose significant medical and economic challenges. Mesenchymal stem cells (MSCs) can differentiate into multiple cell types and offer strong regenerative potential, making them promising for treating various diseases. However, issues with the immune response and cell survival limit the effectiveness of cell transplantation. This has led to increased interest in cell-free stem cell therapy, particularly the use of exosomes, which is the most studied form of this approach. Exosomes are extracellular vesicles that contain proteins, lipids, and nucleic acids and play a key role in cell communication and material exchange. Pyroptosis, a form of cell death involved in innate immunity, is also associated with many diseases. Studies have shown that MSC-derived exosomes have therapeutic potential for treating a range of conditions by regulating inflammation and pyroptosis. This study explored the role of MSC-derived exosomes in modulating pyroptosis to improve the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Haiming Li
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Peng Zhang
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Minghui Lin
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China
| | - Kang Li
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China
| | - Cunxin Zhang
- Department of Spine Surgery, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Xiao He
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| | - Kai Gao
- Shandong University of Traditional Chinese Medicine, Jinan, CN, China.
- Department of Orthopaedics, Jining No. 1 People's Hospital, Jining, 272011, People's Republic of China.
| |
Collapse
|
39
|
Liu P, Guo H, Huang X, Liu A, Zhu T, Zheng C, Fu F, Zhang K, Li S, Luo X, Tian J, Jin Y, Xuan K, Sui B. Golgi-restored vesicular replenishment retards bone aging and empowers aging bone regeneration. Bone Res 2025; 13:21. [PMID: 39922812 PMCID: PMC11807224 DOI: 10.1038/s41413-024-00386-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/15/2024] [Accepted: 10/22/2024] [Indexed: 02/10/2025] Open
Abstract
Healthy aging is a common goal for humanity and society, and one key to achieving it is the rejuvenation of senescent resident stem cells and empowerment of aging organ regeneration. However, the mechanistic understandings of stem cell senescence and the potential strategies to counteract it remain elusive. Here, we reveal that the aging bone microenvironment impairs the Golgi apparatus thus diminishing mesenchymal stem cell (MSC) function and regeneration. Interestingly, replenishment of cell aggregates-derived extracellular vesicles (CA-EVs) rescues Golgi dysfunction and empowers senescent MSCs through the Golgi regulatory protein Syntaxin 5. Importantly, in vivo administration of CA-EVs significantly enhanced the bone defect repair rate and improved bone mass in aging mice, suggesting their therapeutic value for treating age-related osteoporosis and promoting bone regeneration. Collectively, our findings provide insights into Golgi regulation in stem cell senescence and bone aging, which further highlight CA-EVs as a potential rejuvenative approach for aging bone regeneration.
Collapse
Affiliation(s)
- Peisheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Hao Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiaoyao Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Anqi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Ting Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Chenxi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Fei Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Kaichao Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Shijie Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xinyan Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiongyi Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yan Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, Shaanxi, China.
| | - Kun Xuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Disease, Department of Preventive Dentistry, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Bingdong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Disease, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
40
|
Gustafson D, Nieuwland R, Lucien F. MIBLood-EV: An Online Reporting Tool to Facilitate the Standardized Reporting of Preanalytical Variables and Quality Control of Plasma and Serum to Enhance Rigor and Reproducibility in Liquid Biopsy Research. Biopreserv Biobank 2025; 23:62-64. [PMID: 39247973 DOI: 10.1089/bio.2024.0083] [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: 09/10/2024] Open
Abstract
Pre-analytical variability significantly impacts the reproducibility of liquid biopsy research, which is critical for precision medicine and biomedical research. This report highlights the challenges and variability in the pre-analytical processes of liquid biopsies, especially regarding extracellular vesicles (EVs), which are crucial for diagnostics in oncology. The MIBlood-EV initiative aims to standardize the reporting of pre-analytical variables and the quality control of plasma and serum samples to enhance reproducibility in EV research. By providing a comprehensive and flexible reporting framework, MIBlood-EV seeks to improve the reliability of EV studies and facilitate the development of evidence-based protocols, ultimately advancing the field of liquid biopsy research.
Collapse
Affiliation(s)
- Dakota Gustafson
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, Toronto, Canada
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle Center, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
41
|
Rohm TV, Cunha E Rocha K, Olefsky JM. Metabolic Messengers: small extracellular vesicles. Nat Metab 2025; 7:253-262. [PMID: 39920357 DOI: 10.1038/s42255-024-01214-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/19/2024] [Indexed: 02/09/2025]
Abstract
Small extracellular vesicles (sEVs) are signalling molecules and biomarkers of cell status that govern a complex intraorgan and interorgan communication system through their cargo. Initially recognized as a waste disposal mechanism, they have emerged as important metabolic regulators. They transfer biological signals to recipient cells through their cargo content, and microRNAs (miRNAs) often mediate their metabolic effects. This review provides a concise overview of sEVs, specifically in the context of obesity-associated chronic inflammation and related metabolic disorders, describing their role as metabolic messengers, identifying their key sites of action and elucidating their mechanisms. We highlight studies that have shaped our understanding of sEV metabolism, address critical questions for future exploration, discuss the use of miRNAs as disease biomarkers and provide insights into the therapeutic potential of sEVs or specific miRNAs for treating metabolic diseases and related disorders in the future.
Collapse
Affiliation(s)
- Theresa V Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Karina Cunha E Rocha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jerrold M Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
42
|
Saneh H, Wanczyk H, Walker J, Finck C. Stem cell-derived extracellular vesicles: a potential intervention for Bronchopulmonary Dysplasia. Pediatr Res 2025; 97:497-509. [PMID: 39251881 PMCID: PMC12014501 DOI: 10.1038/s41390-024-03471-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 09/11/2024]
Abstract
Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among extreme preterm infants. The pathogenesis of BPD is multifactorial, with inflammation playing a central role. There is strong evidence that stem cell therapy reduces inflammatory changes and restores normal lung morphology in animal models of hyperoxia-induced lung injury. These therapeutic effects occur without significant engraftment of the stem cells in the host lung, suggesting more of a paracrine mechanism mediated by their secretome. In addition, there are multiple concerns with stem cell therapy which may be alleviated by administering only the effective vesicles instead of the cells themselves. Extracellular vesicles (EVs) are cell-derived components secreted by most eukaryotic cells. They can deliver their bioactive cargo (mRNAs, microRNAs, proteins, growth factors) to recipient cells, which makes them a potential therapeutic vehicle in many diseases, including BPD. The following review will highlight recent studies that investigate the effectiveness of EVs derived from stem cells in preventing or repairing injury in the preterm lung, and the potential mechanisms of action that have been proposed. Current limitations will also be discussed as well as suggestions for advancing the field and easing the transition towards clinical translation in evolving or established BPD. IMPACT: Extracellular vesicles (EVs) derived from stem cells are a potential intervention for neonatal lung diseases. Their use might alleviate the safety concerns associated with stem cell therapy. This review highlights recent studies that investigate the effectiveness of stem cell-derived EVs in preclinical models of bronchopulmonary dysplasia. It adds to the existing literature by elaborating on the challenges associated with EV research. It also provides suggestions to advance the field and ease the transition towards clinical applications. Optimizing EV research could ultimately improve the quality of life of extreme preterm infants born at vulnerable stages of lung development.
Collapse
Affiliation(s)
- Hala Saneh
- Department of Neonatal Medicine, Connecticut Children's Medical Center, Hartford, CT, USA.
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Heather Wanczyk
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Joanne Walker
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Christine Finck
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
- Department of Pediatric Surgery, Connecticut Children's Medical Center, Hartford, CT, USA
| |
Collapse
|
43
|
Tunset ME, Haslene-Hox H, Larsen JB, Kondziella D, Nygård M, Pedersen SA, Vaaler A, Llorente A. Clinical studies of blood-borne Extracellular vesicles in psychiatry: A systematic review. J Psychiatr Res 2025; 182:373-390. [PMID: 39862765 DOI: 10.1016/j.jpsychires.2025.01.037] [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: 10/14/2024] [Revised: 12/02/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Biomarkers for the diagnosis and clinical management of psychiatric disorders are currently lacking. Extracellular vesicles (EVs), lipid membrane-encapsulated vesicles released by cells, hold promise as a source of biomarkers due to their ability to carry molecules that reflect the status of their donor cells and their ubiquitous presence in biofluids. This review examines the literature on EVs in biofluids from psychiatric disorder patients, and discuss how the published studies contribute to our understanding of the pathophysiology of these conditions and to the discovery of potential biomarkers. We analyzed 46 studies on blood-borne EVs; no investigations on cerebrospinal fluid-derived EVs were found. A significant number of studies lacked optimal description of the methodology and/or characterization of the isolated EVs. Moreover, many studies aimed to capture brain-derived EVs, but often capture-proteins with low brain specificity were used. Considering biomarkers, miRNAs were the most investigated molecular type, but based on the studies analyzed it was not possible to identify robust biomarker candidates for the investigated disorders. Additionally, we describe the contribution of EV studies in illuminating the pathophysiology of psychiatric disorders, including research on insulin resistance, inflammation, mitochondrial dysfunction, and the microbiota. We conclude that there is a shortage of studies with detailed methodology description and EV sample characterization in psychiatric research. To exploit the potential of EVs to investigate psychiatric disorders and identify biomarkers more studies and validated protocols using capture proteins with high specificity to brain cells are needed. The review protocol was pre-registered in the PROSPERO database under the registration number CRD42021277534.
Collapse
Affiliation(s)
- Mette Elise Tunset
- Department of Psychosis and Rehabilitation, Division of Mental Healthcare, St. Olavs University Hospital, Trondheim, Norway; Department of Mental Health- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Hanne Haslene-Hox
- Department of Biotechnology and Nanomedicine, SINTEF, Trondheim, Norway
| | - Jeanette Brun Larsen
- Department of Psychosis and Rehabilitation, Division of Mental Healthcare, St. Olavs University Hospital, Trondheim, Norway
| | - Daniel Kondziella
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mona Nygård
- Department of Psychosis and Rehabilitation, Division of Mental Healthcare, St. Olavs University Hospital, Trondheim, Norway; Department of Mental Health- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | | | - Arne Vaaler
- Department of Mental Health- Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway; Department of Acute Psychiatry, Division of Mental Healthcare, St. Olavs University Hospital, Trondheim, Norway
| | - Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, 0379, Oslo, Norway; Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway; Department for Mechanical, Electronics and Chemical Engineering, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
44
|
Orefice NS, Petrillo G, Pignataro C, Mascolo M, De Luca G, Verde S, Pentimalli F, Condorelli G, Quintavalle C. Extracellular vesicles and microRNAs in cancer progression. Adv Clin Chem 2025; 125:23-54. [PMID: 39988407 DOI: 10.1016/bs.acc.2024.11.004] [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] [Indexed: 02/25/2025]
Abstract
Extracellular vesicles (EVs) have emerged as critical mediators of intercellular communication in cancer. These membranous structures, secreted by normal and cancerous cells, carry a cargo of bioactive molecules including microRNAs (miRNAs) that modulate various cellular processes. miRNAs are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation and have been implicated in cancer initiation, progression, and metastasis. In cancer, tumor-derived EVs transport specific miRNAs to recipient cells, modulating tumorigenesis, growth, angiogenesis, and metastasis. Dysregulation of miRNA expression profiles within EVs contributes to the acquisition of cancer hallmarks that include increased proliferation, survival, and migration. EV miRNAs influence the tumor microenvironment, promoting immune evasion, remodeling the extracellular matrix, and establishing pre-metastatic niches. Understanding the complex interplay between EVs, miRNAs, and cancer holds significant promise for developing novel diagnostic and therapeutic strategies. This chapter provides insights into the role of EV-mediated miRNA signaling in cancer pathogenesis, highlighting its potential as a biomarker for cancer detection, prognosis, and treatment response assessment.
Collapse
Affiliation(s)
- Nicola S Orefice
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Gianluca Petrillo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Martina Mascolo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Giada De Luca
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| | - Sara Verde
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; Aka biotech S.r.l., Napoli, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe DeGennaro", Bari, Italy
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy; Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy.
| | - Cristina Quintavalle
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| |
Collapse
|
45
|
Jimenez-Trinidad FR, Calvo-Gomez S, Sabaté M, Brugaletta S, Campuzano V, Egea G, Dantas AP. Extracellular Vesicles as Mediators of Endothelial Dysfunction in Cardiovascular Diseases. Int J Mol Sci 2025; 26:1008. [PMID: 39940780 PMCID: PMC11816526 DOI: 10.3390/ijms26031008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
This comprehensive review aims to provide a thorough overview of the vital role that extracellular vesicles (EVs) play in endothelial dysfunction, particularly emphasizing how physiological factors-such as sex and aging-along with significant cardiovascular risk factors, influence this process. The review covers studies ranging from the first description of EVs in 1945 to contemporary insights into their biological roles in intercellular signaling and endothelial dysfunction. A comprehensive analysis of peer-reviewed articles and reviews indexed in the PubMed database was conducted to compile the information. Initially, Medical Subject Headings (MeSH) terms included keywords aimed at providing general knowledge about the role of EVs in the regulation of endothelial signaling, such as "extracellular vesicles", "endothelium", and "intercellular signaling". Subsequently, terms related to the pathophysiological implications of EV interactions with endothelial dysfunction and cardiovascular disease were added, including "cardiovascular disease", "sex", "aging", "atherosclerosis", "obesity", and "diabetes". Additionally, the potential applications of EVs in cardiovascular disease were explored using the MeSH terms "extracellular vesicles", "cardiovascular disease", "biomarker", and "therapeutic strategy". The results of this bibliographical review reveal that EVs have the capacity to induce various cellular responses within the cardiovascular system and play a significant role in the complex landscape of endothelial dysfunction and cardiovascular disease. The composition of the EV cargo is subject to modification by pathophysiological conditions such as sex, aging, and cardiovascular risk factors, which result in a complex regulatory influence on endothelial function and neighboring cells when released from a dysfunctional endothelium. Moreover, the data suggest that this field still requires further exploration, as EV biology is continuously evolving, presenting a dynamic and engaging area for research. A deeper understanding of the molecular cargo involved in EV-endothelium interactions could yield valuable biomarkers for monitoring cardiovascular disease progression and facilitate the development of innovative bioengineered therapeutic strategies to enhance patient outcomes.
Collapse
Affiliation(s)
- Francisco Rafael Jimenez-Trinidad
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Sergi Calvo-Gomez
- Department of Biomedical Sciences, School of Medicine, Universitat Internacional de Catalunya (UIC), 08195 Barcelona, Spain;
| | - Manel Sabaté
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Salvatore Brugaletta
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Rare Diseases Biomedical Research Network Center (CIBERER), Instituto de Salud Carlos III, 28222 Madrid, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Center of Medical Genetics, University of Antwerpen, 2659 Edegem, Belgium
| | - Ana Paula Dantas
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain; (F.R.J.-T.); (V.C.); (G.E.)
- Institut Clínic Cardiovascular (ICCV), Hospital Clínic, 08036 Barcelona, Spain; (M.S.); (S.B.)
- Division of Respiratory, Cardiovascular and Renal Pathobiology and Bioengineering, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| |
Collapse
|
46
|
Osorio-Méndez JJ, Gómez-Grosso LA, Montoya-Ortiz G, Novoa-Herrán S, Domínguez-Romero Y. Small Extracellular Vesicles from Breast Cancer Cells Induce Cardiotoxicity. Int J Mol Sci 2025; 26:945. [PMID: 39940718 PMCID: PMC11816698 DOI: 10.3390/ijms26030945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Cardiovascular diseases and cancer are leading global causes of morbidity and mortality, necessitating advances in diagnosis and treatment. Doxorubicin (Doxo), a potent chemotherapy drug, causes long-term heart damage due to cardiotoxicity. Small extracellular vesicles (sEVs) carry bioactive molecules-such as proteins, lipids, and nucleic acids-that can modulate gene expression and signaling pathways in recipient cells, including cardiomyocytes. Through the delivery of cytokines, microRNAs, and growth factors, sEVs can influence cell survival, which plays a critical role in the development of cardiotoxicity. This study investigates the role of sEVs derived from breast cancer cells treated or not with Doxo and their potential to induce cardiomyocyte damage, thereby contributing to cardiotoxicity. We isolated sEVs from MCF-7 cells treated or not to Doxo using ultracentrifugation and characterized them through Nanoparticle Tracking Analysis (NTA), Scanning Electron Microscopy (SEM), and Western Blotting (WB) for the markers CD63, CD81, and TSG101. We analyzed cytokine profiles using a Multiplex Assay and Cytokine Membrane Array. We exposed Guinea pig cardiomyocytes to different concentrations of sEVs. We assessed their viability (MTT assay), shortening, reactive oxygen species (ROS-DHE dye) production, mitochondrial membrane potential (JC-1 dye), and calcium dynamics (FLUO-4 dye). We performed statistical analyses, including t-tests, ANOVA, Cohen's d, and η2 to validate the robustness of the results. Treatment of MCF-7 cells with 0.01 μM Doxorubicin resulted in increased sEVs production, particularly after 48 h of exposure (~1.79 × 108 ± 2.77 × 107 vs. ~5.1 × 107 ± 1.28 × 107 particles/mL, n = 3, p = 0.0019). These sEVs exhibited protein profiles in the 130-25 kDa range and 93-123 nm sizes. They carried cytokines including TNF-α, IL-1β, IL-4, IFN-γ, and IL-10. Exposure of cardiomyocytes to sEVs (0.025 μg/mL to 2.5 μg/mL) from both Doxo-treated and untreated cells significantly reduced cardiomyocyte viability, shortened cell length by up to 20%, increased ROS production, and disrupted calcium homeostasis and mitochondrial membrane potential, indicating severe cellular stress and cardiotoxicity. These findings suggest that Doxo enhances sEVs production from breast cancer cells, which plays a key role in cardiotoxicity through their cytokine cargo. The study highlights the potential of these sEVs as biomarkers for early cardiotoxicity detection and as therapeutic targets to mitigate cardiovascular risks in chemotherapy patients. Future research should focus on understanding the mechanisms by which Doxorubicin-induced sEVs contribute to cardiotoxicity and exploring their diagnostic and therapeutic potential to improve patient safety and outcomes in cancer therapy.
Collapse
Affiliation(s)
- Jhon Jairo Osorio-Méndez
- Master in Biochemistry Program, Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia (Y.D.-R.)
| | - Luis Alberto Gómez-Grosso
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia (Y.D.-R.)
- Department of Physiological Sciences, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| | - Gladis Montoya-Ortiz
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia (Y.D.-R.)
| | - Susana Novoa-Herrán
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia (Y.D.-R.)
| | - Yohana Domínguez-Romero
- Molecular Physiology Group, Sub-Direction of Scientific and Technological Research, Direction of Public, Health Research, National Institute of Health, Bogotá 111321, Colombia (Y.D.-R.)
- Doctorate in Biotechnology Program, Faculty of Sciences, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| |
Collapse
|
47
|
Zhang L, Zhou Y, Yang Z, Jiang L, Yan X, Zhu W, Shen Y, Wang B, Li J, Song J. Lipid droplets in central nervous system and functional profiles of brain cells containing lipid droplets in various diseases. J Neuroinflammation 2025; 22:7. [PMID: 39806503 PMCID: PMC11730833 DOI: 10.1186/s12974-025-03334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Lipid droplets (LDs), serving as the convergence point of energy metabolism and multiple signaling pathways, have garnered increasing attention in recent years. Different cell types within the central nervous system (CNS) can regulate energy metabolism to generate or degrade LDs in response to diverse pathological stimuli. This article provides a comprehensive review on the composition of LDs in CNS, their generation and degradation processes, their interaction mechanisms with mitochondria, the distribution among different cell types, and the roles played by these cells-particularly microglia and astrocytes-in various prevalent neurological disorders. Additionally, we also emphasize the paradoxical role of LDs in post-cerebral ischemia inflammation and explore potential underlying mechanisms, aiming to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Longxiao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yunfei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Zhongbo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liangchao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyang Yan
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wenkai Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yi Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bolong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiaxi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Jinning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
48
|
Emami A, Arabpour Z, Izadi E. Extracellular vesicles: essential agents in critical bone defect repair and therapeutic enhancement. Mol Biol Rep 2025; 52:113. [PMID: 39798011 DOI: 10.1007/s11033-024-10209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025]
Abstract
Bone serves as a fundamental structural component in the body, playing pivotal roles in support, protection, mineral supply, and hormonal regulation. However, critical-sized bone injuries have become increasingly prevalent, necessitating extensive medical interventions due to limitations in the body's capacity for self-repair. Traditional approaches, such as autografts, allografts, and xenografts, have yielded unsatisfactory results. Stem cell therapy emerges as a promising avenue, but challenges like immune rejection and low cell survival rates hinder its widespread clinical implementation. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have garnered attention for their regenerative capabilities, which surpass those of MSCs themselves. EVs offer advantages such as reduced immunogenicity, enhanced stability, and simplified storage, positioning them as a promising tool in stem cell-based therapies. This review explores the potential of EV-based therapy in bone tissue regeneration, delving into their biological characteristics, communication mechanisms, and preclinical applications across various physiological and pathological conditions. The mechanisms underlying EV-mediated bone regeneration, including angiogenesis, osteoblast proliferation, mineralization, and immunomodulation, are elucidated. Preclinical studies demonstrate the efficacy of EVs in promoting bone repair and neovascularization, even in pathological conditions like osteoporosis. EVs hold promise as a potential alternative for regenerating bone tissue, particularly in the context of critical-sized bone defects, offering new avenues for effective bone defect repair and management.
Collapse
Affiliation(s)
- Asrin Emami
- Iranian Tissue Bank and Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Zohreh Arabpour
- Department of Ophthalmology and Visual Science and University of Illinois, Chicago, IL, 60612, USA
| | - Elaheh Izadi
- Pediatric Cell, and Gene Therapy Research Center Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Zhang Y, Liu Z, Chopp M, Millman M, Li Y, Cepparulo P, Kemper A, Li C, Zhang L, Zhang ZG. Small extracellular vesicles derived from cerebral endothelial cells with elevated microRNA 27a promote ischemic stroke recovery. Neural Regen Res 2025; 20:224-233. [PMID: 38767487 PMCID: PMC11246145 DOI: 10.4103/nrr.nrr-d-22-01292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/14/2023] [Accepted: 01/22/2024] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00030/figure1/v/2024-05-14T021156Z/r/image-tiff Axonal remodeling is a critical aspect of ischemic brain repair processes and contributes to spontaneous functional recovery. Our previous in vitro study demonstrated that exosomes/small extracellular vesicles (sEVs) isolated from cerebral endothelial cells (CEC-sEVs) of ischemic brain promote axonal growth of embryonic cortical neurons and that microRNA 27a (miR-27a) is an elevated miRNA in ischemic CEC-sEVs. In the present study, we investigated whether normal CEC-sEVs engineered to enrich their levels of miR-27a (27a-sEVs) further enhance axonal growth and improve neurological outcomes after ischemic stroke when compared with treatment with non-engineered CEC-sEVs. 27a-sEVs were isolated from the conditioned medium of healthy mouse CECs transfected with a lentiviral miR-27a expression vector. Small EVs isolated from CECs transfected with a scramble vector (Scra-sEVs) were used as a control. Adult male mice were subjected to permanent middle cerebral artery occlusion and then were randomly treated with 27a-sEVs or Scra-sEVs. An array of behavior assays was used to measure neurological function. Compared with treatment of ischemic stroke with Scra-sEVs, treatment with 27a-sEVs significantly augmented axons and spines in the peri-infarct zone and in the corticospinal tract of the spinal grey matter of the denervated side, and significantly improved neurological outcomes. In vitro studies demonstrated that CEC-sEVs carrying reduced miR-27a abolished 27a-sEV-augmented axonal growth. Ultrastructural analysis revealed that 27a-sEVs systemically administered preferentially localized to the pre-synaptic active zone, while quantitative reverse transcription-polymerase chain reaction and Western Blot analysis showed elevated miR-27a, and reduced axonal inhibitory proteins Semaphorin 6A and Ras Homolog Family Member A in the peri-infarct zone. Blockage of the Clathrin-dependent endocytosis pathway substantially reduced neuronal internalization of 27a-sEVs. Our data provide evidence that 27a-sEVs have a therapeutic effect on stroke recovery by promoting axonal remodeling and improving neurological outcomes. Our findings also suggest that suppression of axonal inhibitory proteins such as Semaphorin 6A may contribute to the beneficial effect of 27a-sEVs on axonal remodeling.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Michael Millman
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Yanfeng Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | - Amy Kemper
- Department of Pathology, Henry Ford Hospital, Detroit, MI, USA
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | |
Collapse
|
50
|
Nakatsutsumi K, Choi W, Johnston W, Pool K, Park DJ, Weaver JL, Coimbra R, Eliceiri B, Costantini TW. Lung contusion complicated by pneumonia worsens lung injury via the inflammatory effect of alveolar small extracellular vesicles on macrophages and epithelial cells. J Trauma Acute Care Surg 2025; 98:55-63. [PMID: 39621452 DOI: 10.1097/ta.0000000000004499] [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: 12/19/2024]
Abstract
BACKGROUND Lung contusion (LC) complicated by pneumonia is associated with a higher risk of acute lung injury (ALI) mediated by activation of immune cells and injury to the lung epithelium. Small extracellular vesicles (sEVs) are essential mediators of cellular crosstalk; however, their role in the development of postinjury ALI remains unclear. We hypothesized that LC complicated by pneumonia increases the pro-inflammatory effect of alveolar sEVs on macrophages and the cytotoxicity of alveolar sEVs to pulmonary epithelial cells, worsening the severity of ALI. METHODS Studies in C57BL/6 mice were designed with four groups: sham, LC, Pneumonia (Pneu), and LC + Pneu. Lung contusion was induced by a cortical controlled impactor, while pneumonia was conducted by intratracheal injection of 10 5 cfu Pseudomonas aeruginosa . Bronchoalveolar lavage fluid (BAL) was harvested 24 hours postinfection, and sEVs were purified by centrifugation and size exclusion chromatography. To evaluate the effect of alveolar sEV on cells, sEVs from each group were cocultured with macrophages (RAW 264.7) to assess cytokine release and lung epithelial cells (MLE 12) to assess epithelial cytotoxicity. RESULTS The LC + Pneu group severely injured lungs histologically and increased the susceptibility to the bacteria. The LC + Pneu group showed higher concentrations of proteins, macrophage inflammatory protein 1-alpha (MIP1α), and intercellular adhesion molecule 1 (ICAM-1) in BAL. MIP1α and ICAM-1 expression in the macrophages increased after incubation with sEVs from the LC + Pneu group. Moreover, the sEVs demonstrated higher cytotoxicity to epithelial cells and increased apoptosis in epithelial cells after incubation with sEVs from the LC + Pneu group. CONCLUSION Lung contusion complicated by pneumonia increased the pro-inflammatory effect of alveolar sEVs on macrophages and the cytotoxicity of alveolar sEVs to pulmonary epithelial cells, worsening the severity of ALI. These results demonstrate the potential importance of alveolar sEVs in lung inflammation following a bacterial infection after trauma.
Collapse
Affiliation(s)
- Keita Nakatsutsumi
- From the Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery (K.N., W.C., W.J., K.P., D.P., J.W., B.E., T.C.), UC San Diego School of Medicine, San Diego; Comparative Effectiveness and Clinical Outcomes Research Center (R.C.), Riverside University Health System, Loma Linda University School of Medicine, Riverside, California; and Trauma and Acute Critical Care Center (K.N.), Tokyo Medical and Dental University Hospital, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|