51
|
Acevedo-Sánchez V, Rodríguez-Hernández RM, Aguilar-Ruíz SR, Torres-Aguilar H, Pina-Canseco S, Chávez-Olmos P, Garrido E, Baltiérrez-Hoyos R, Romero-Tlalolini MA. Keratinocyte-derived extracellular vesicles induce macrophage polarization toward an M1-like phenotype. Biochem Biophys Res Commun 2025; 758:151659. [PMID: 40121968 DOI: 10.1016/j.bbrc.2025.151659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Multiple reports have shown an effect of keratinocyte-derived extracellular vesicles (EVs) on keratinocytes and other cell types. However, the contribution of keratinocyte-derived EVs under physiological and pathological conditions is not fully elucidated. Therefore, whether there is an effect of EVs on macrophages in cervical cancer (CC) is also unknown. Here, we evaluated the effect of tumor and non-tumor keratinocyte-derived EVs on the polarization of peripheral blood mononuclear cells (PBMCs)-derived macrophages and THP-1 cell line. Flow cytometric evaluation of macrophages cultured in the presence of keratinocyte-derived EVs mainly indicated an increase in classical activation markers CD80 and CD86 (M1 phenotype) and little or no modification of alternative activation markers (M2 phenotype). ELISA evaluation of macrophage supernatants revealed an increase in the secretion of proinflammatory cytokines such as IL-1β and IL-6. On the other hand, TGF-β was not significantly modified and only EVs derived from non-cancerous keratinocytes induced a significant increase in IL-10. The expression levels of transcripts associated with the M1 phenotype were also evaluated by qRT-PCR with similar results to ELISA for TGF-β and IL-10; but also an increase in the expression of HLA-DRα and TNF-α was observed, and no statistically significant changes in ARG1. The ROS production was also evaluated and this increase mainly in macrophages treated with CC keratinocytes-derived EVs. So, our results suggest that the uptake of EVs derived from released by non-tumor and cervical cancer keratinocytes promotes in macrophages their polarization to an M1-like phenotype.
Collapse
Affiliation(s)
- V Acevedo-Sánchez
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - R M Rodríguez-Hernández
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - S R Aguilar-Ruíz
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - H Torres-Aguilar
- Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez de Oaxaca, Av. Universidad S/N, Cinco Señores, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - S Pina-Canseco
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - P Chávez-Olmos
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación, Gustavo A. Madero, 07360, Mexico City, Mexico.
| | - E Garrido
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Delegación, Gustavo A. Madero, 07360, Mexico City, Mexico.
| | - R Baltiérrez-Hoyos
- Facultad de Medicina y Cirugía, CONAHCYT-Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| | - M A Romero-Tlalolini
- Facultad de Medicina y Cirugía, CONAHCYT-Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda de Aguilera S/N, Calz. San Felipe del Agua, Oaxaca de Juárez, 68120, Oaxaca, Mexico.
| |
Collapse
|
52
|
Sabatke B, Rossi IV, Bonato L, Fucio S, Cortés A, Marcilla A, Ramirez MI. Host-Pathogen Cellular Communication: The Role of Dynamin, Clathrin, and Macropinocytosis in the Uptake of Giardia-Derived Extracellular Vesicles. ACS Infect Dis 2025; 11:954-962. [PMID: 40155351 PMCID: PMC11997983 DOI: 10.1021/acsinfecdis.4c00996] [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: 12/12/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Giardia intestinalis, a protozoan causing giardiasis, disrupts gastrointestinal health through complex host-parasite interactions. This study explores the differential uptake mechanisms of extracellular vesicles (EVs) derived from Giardia (gEVs), host cells (hEVs), and the host-parasite interaction (intEVs) in intestinal Caco-2 cells. Results show that intEVs are internalized more rapidly than gEVs and hEVs, underscoring their pivotal role in pathogenesis. To delineate uptake pathways, various endocytosis inhibitors were applied, and clathrin-mediated endocytosis inhibition using monodansylcadaverine (MDC) significantly reduced intEV and gEV uptake, confirming the role of clathrin-mediated endocytosis (CME). The use of dynasore, a dynamin inhibitor, strongly reduced the internalization of all EV types, demonstrating that uptake is dynamin-dependent. In contrast, methyl-β-cyclodextrin (MβCD), which disrupts lipid rafts and caveolae-mediated pathways, had no effect on EV uptake, indicating that caveolae are not involved in this process. Furthermore, inhibition of Na+/H+ exchange and phosphoinositide 3-kinase activity, both essential for macropinocytosis, also led to a significant reduction in intEV internalization. These findings strongly support that gEVs are internalized primarily through a dynamin- and clathrin-dependent pathway, independent of caveolae and lipid rafts, but modulated by tyrosine kinase signaling and macropinocytosis. These insights into selective and comprehensive inhibition pathways offer promising therapeutic targets to mitigate giardiasis.
Collapse
Affiliation(s)
- Bruna Sabatke
- Graduate
Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
- EVAHPI-Extracellular
Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| | - Izadora V Rossi
- Graduate
Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
- EVAHPI-Extracellular
Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| | - Leticia Bonato
- Graduate
Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
- EVAHPI-Extracellular
Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| | - Sarah Fucio
- EVAHPI-Extracellular
Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
- Graduate
Program in Cell and Molecular Biology, Federal
University of Paraná, Curitiba, PR 81350-010, Brazil
| | - Alba Cortés
- Department of Parasitology and Cellular Biology,
Faculty of Pharmacy, University of Valencia, Valencia 46010, Spain
| | - Antonio Marcilla
- Department of Parasitology and Cellular Biology,
Faculty of Pharmacy, University of Valencia, Valencia 46010, Spain
| | - Marcel I. Ramirez
- EVAHPI-Extracellular
Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| |
Collapse
|
53
|
Xu S, Zhang Z, Zhou X, Liao Y, Peng Z, Meng Z, Nüssler AK, Ma L, Xia H, Liu L, Yang W. Gouqi-derived Nanovesicles (GqDNVs) promoted MC3T3-E1 cells proliferation and improve fracture healing. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156755. [PMID: 40252435 DOI: 10.1016/j.phymed.2025.156755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/25/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Lycium barbarum L., also known as Gouqi, a traditional Chinese herbal medicine, is widely utilized in health care products and clinical therapies. Its muscle and bone strengthening efficacy has been recorded in medical classics for a long time. In addition, plant exosome-like nanovesicles (PELNVs) have attracted more and more attention owing to their biological traits. Therefore, we intended to explore the functions, regulatory role, and underlying mechanism of Gouqi-derived Nanovesicles (GqDNVs) on fracture healing. METHODS In this study, we employed the sucrose density gradient differential ultracentrifugation to isolate GqDNVs. The effects of GqDNVs on the proliferation and differentiation of MC3T3-E1 cells were evaluated using the CCK-8 assay, ALP activity measurement, and cell scratch assay. Additionally, leveraging a fracture mouse model, we utilized Micro-CT, immunological staining, and histologic analyses to comprehensively assess the impact of GqDNVs on fracture healing in mice. RESULTS GqDNVs stimulated cell viability, increased ALP activity, and promoted cellular osteogenic protein expression (OPN, ALP, and RUNX2). Subsequently, in the mouse fracture model, trabecular thickness, and bone marrow density were increased in the GqDNVs treatment group after 28 days of injection. Meanwhile, the expressions of OPN and BGP were significantly elevated after both 14 and 28 days. Additionally, the expressions of p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR, p-4EBP1/4EBP1 and p-p70S6K/ p70S6K were also increased after14 days of treatment. CONCLUSIONS GqDNVs effectively promoted the proliferation and differentiation of MC3T3-E1 cells. Furthermore, GqDNVs could improve fracture healing, which is associated with PI3K/Akt/mTOR/p70S6K/4EBP1 signaling pathway.
Collapse
Affiliation(s)
- Shiyin Xu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zixuan Zhang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Xiaolei Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Yuxiao Liao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zhao Peng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zitong Meng
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Andreas K Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, Tübingen 72076, Germany
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hui Xia
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, China
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Wei Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; Department of Nutrition and Food Hygiene and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China; NHC Specialty Laboratory of Food Safety Risk Assessment and Standard Development, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China.
| |
Collapse
|
54
|
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
|
55
|
Hasan R, Zhao Z, Li Y, Liu Y, Zhang Y, Cheng K. Small extracellular vesicles (sEVs) in pancreatic cancer progression and diagnosis. J Control Release 2025; 380:269-282. [PMID: 39889882 PMCID: PMC11908897 DOI: 10.1016/j.jconrel.2025.01.072] [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: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Pancreatic cancer is one of the most aggressive malignancies with poor prognostic outcomes, necessitating the exploration of novel biomarkers and therapeutic targets for early detection and effective treatment. Small extracellular vesicles (sEVs) secreted by cells, have gained considerable attention in cancer research due to their role in intercellular communication and their potential as non-invasive biomarkers. This review focuses on the role of sEVs in the progression of pancreatic cancer and their application as biomarkers. We delve into the biogenesis, composition, and functional implications of sEVs in pancreatic tumor biology, emphasizing their involvement in processes such as tumor growth, metastasis, immune modulation, and chemotherapy resistance. In addition, we discuss the challenges in isolating and characterizing sEVs. The review also highlights recent advances in the utilization of sEV-derived biomarkers for the early diagnosis, prognosis, and monitoring of pancreatic cancer. By synthesizing the latest findings, we aim to underscore the significance of sEVs in pancreatic cancer and their potential to revolutionize patient management through improved diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Reaid Hasan
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Zhen Zhao
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yuanke Li
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yanli Liu
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA.
| |
Collapse
|
56
|
Praveena G, Jayachandran A, Manda Venkata S, Asthana A. From bench to bedside: The evolution of extracellular vesicle diagnostics through microfluidic and paper-based technologies. Colloids Surf B Biointerfaces 2025; 252:114675. [PMID: 40222114 DOI: 10.1016/j.colsurfb.2025.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 04/15/2025]
Abstract
"Extracellular vesicles (EVs) have emerged as key mediators of intercellular communication and valuable biomarkers for various diseases. However, traditional EV isolation and detection methods often struggle with efficiency, scalability, and purity, limiting their clinical utility. Recent advances in microfluidic and paper-based technologies offer innovative solutions that enhance EV isolation and detection by reducing sample volume, accelerating processing times, and integrating multiple analytical steps into compact platforms. These technologies hold significant promise for advancing point-of-care diagnostics, enabling rapid disease detection, personalized treatment monitoring, and better patient outcomes. For example, early detection of cancer biomarkers through EVs can facilitate timely intervention, potentially improving survival rates, while rapid infectious disease diagnostics can support prompt treatment. Despite their potential, challenges such as standardization, scalability, and regulatory hurdles remain. This review discusses recent advancements in microfluidic and paper-based EV diagnostic technologies, their comparative advantages over traditional methods, and their transformative potential in clinical practice."
Collapse
Affiliation(s)
- Ganji Praveena
- Urvogelbio Private Limited, AHERF, Film Nagar, Hyderabad, Telangana 500033, India
| | - Arjun Jayachandran
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad (NIPER - Hyderabad), Balanagar, Hyderabad, Telangana 500037, India
| | - Sasidhar Manda Venkata
- Urvogelbio Private Limited, AHERF, Film Nagar, Hyderabad, Telangana 500033, India; Apollo Hospitals Educational and Research Foundation (AHERF), Cell and Molecular Biology Research Lab, Hyderabad, India.
| | - Amit Asthana
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Hyderabad (NIPER - Hyderabad), Balanagar, Hyderabad, Telangana 500037, India.
| |
Collapse
|
57
|
Wylie C, Rowan R, Malinova D, Crawford L. Extracellular vesicles in multiple myeloma: pathogenesis and therapeutic application. FEBS J 2025. [PMID: 40205752 DOI: 10.1111/febs.70093] [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: 10/02/2024] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Multiple myeloma (MM), characterised by the clonal proliferation of plasma cells in the bone marrow, is the second most common haematological malignancy worldwide. Although there is now an impressive artillery of therapeutics to tackle this condition, resistance remains a prevalent issue. The bone marrow microenvironment performs a crucial role in supporting MM pathogenesis and promoting the development of therapeutic resistance. Extracellular vesicles (EVs), small vesicles that carry bioactive molecules, are a key component of cell-to-cell communication within the bone marrow microenvironment. In this review, we summarise the contribution of EVs to disease progression and anticancer treatment resistance and discuss the potential therapeutic applications of EVs in MM.
Collapse
Affiliation(s)
- Chloe Wylie
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Rebecca Rowan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, UK
| | - Dessi Malinova
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, UK
| | - Lisa Crawford
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, UK
| |
Collapse
|
58
|
Wang L, Liu R, Wang Y. The roles of extracellular vesicles in mental disorders: information carriers, biomarkers, therapeutic agents. Front Pharmacol 2025; 16:1591469. [PMID: 40271072 PMCID: PMC12014780 DOI: 10.3389/fphar.2025.1591469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Mental disorders are complex conditions that encompass various symptoms and types, affecting approximately 1 in 8 people globally. They place a significant burden on both families and society as a whole. So far, the etiology of mental disorders remains poorly understood, making diagnosis and treatment particularly challenging. Extracellular vesicles (EVs) are nanoscale particles produced by cells and released into the extracellular space. They contain bioactive molecules including nucleotides, proteins, lipids, and metabolites, which can mediate intercellular communication and are involved in various physiological and pathological processes. Recent studies have shown that EVs are closely linked to mental disorders like schizophrenia, major depressive disorder, and bipolar disorder, playing a key role in their development, diagnosis, prognosis, and treatment. Therefore, based on recent research findings, this paper aims to describe the roles of EVs in mental disorders and summarize their potential applications in diagnosis and treatment, providing new ideas for the future clinical transformation and application of EVs.
Collapse
Affiliation(s)
| | | | - Ying Wang
- Department of Pharmacy, Tianjin Anding Hospital, Tianjin, China
| |
Collapse
|
59
|
Xiang W, Zhu Z, Shang Q, Yasin P, Wu Y, Song X. Multi-Omics Analysis of the Immune Effect of the Engineered Exosome Drug Delivery System in Inducing Macrophage Apoptosis. Pharmaceutics 2025; 17:494. [PMID: 40284489 PMCID: PMC12030349 DOI: 10.3390/pharmaceutics17040494] [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: 02/21/2025] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
Background: In this study, exosomes were engineered with anti-CD47 antibody and loaded with rifapentine to improve their ability to target macrophages for drug delivery. Methods: Exosomes from RAW264.7 cell supernatant were extracted by differential centrifugation, antibody-modified, and drug-loaded ultrasonically. After co-culturing with macrophages, transcriptomics and proteomics screened differentially expressed genes and proteins. Western Blot identified macrophage polarization, ELISA detected inflammatory indicators, and an apoptosis kit was used for fluorescence staining. Results: Transcriptome sequencing showed that 406 genes in the macrophages changed significantly, with pathways like TNF and NF-κB. Proteomics identified 7478 proteins, 433 with significant differences. Western Blot indicated M1 polarization. Fluorescence staining showed apoptosis in the antiMExo-RIF group. Conclusions: The study provides multi-omics evidence of the immune mechanism of the engineered exosome drug delivery system in inducing macrophage apoptosis, revealing potential molecular mechanisms and the great potential use of engineered exosomes in treating macrophage-related diseases.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Zhoujun Zhu
- Department of Joint Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China;
| | - Qisong Shang
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Parhat Yasin
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Yuanyuan Wu
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| | - Xinghua Song
- Department of Spine Surgery, The Sixth Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, China; (W.X.); (Q.S.); (P.Y.); (Y.W.)
| |
Collapse
|
60
|
Pan X, Huang C, Bai X, Li F. Causal relationship between breast cancer and acute myeloid leukemia based on two-sample bidirectional Mendelian randomization and transcriptome overlap analysis. Discov Oncol 2025; 16:492. [PMID: 40198525 PMCID: PMC11979033 DOI: 10.1007/s12672-025-02288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer is the most prevalent malignancy and the leading cause of cancer-related deaths among women worldwide. Several case reports have shown that some breast cancer patients subsequently develop acute myeloid leukemia (AML) within a short period. However, the causal relationship and pathogenic mechanisms between breast cancer and AML remain incompletely understood. METHODS Mendelian randomization (MR) analyses were conducted to explore the bidirectional causal relationships between breast cancer and AML. Additionally, we applied the Bayesian Weighted Mendelian Randomization (BWMR) approach to validate the results of the MR analysis. Subsequently, we utilized RNA-seq data from various sources to explore the potential molecular signaling pathways between breast cancer and AML. RESULTS Both IVW method and BWMR approach demonstrated that data from three distinct sources consistently indicated breast cancer as a risk factor for AML, with all sources showing statistically significant results (all P < 0.05, Odds Ratios [ORs] > 1). Bioinformatic analyses suggested that extracellular vesicle functions and p53 signaling pathway may mediate molecular links between breast cancer and AML. Using machine learning, we identified 8 genes with high diagnostic efficacy for predicting the occurrence of AML in breast cancer patients. CONCLUSIONS MR analyses indicated a causal relationship between breast cancer and AML. Additionally, transcriptome analysis offered a theoretical basis for understanding the potential mechanisms and therapeutic targets of AML in breast cancer patients.
Collapse
Affiliation(s)
- Xin'an Pan
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Zheng Street, East Lake District, Nanchang City, 330006, Jiangxi Province, China
| | - Cuihan Huang
- The First Clinical Medical College of Nanchang University, Xuefu Road, Nanchang, 330006, Jiangxi, China
| | - Xinyi Bai
- School of Public, Health of Nanchang University, Xuefu Road, Nanchang, 330006, Jiangxi, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Zheng Street, East Lake District, Nanchang City, 330006, Jiangxi Province, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China.
| |
Collapse
|
61
|
Zhang J, Weng S, Fan Z, Hu D, Le J, Sheng K. Migrasomes: Critical players in intercellular nanovesicle communication. Cell Signal 2025; 132:111796. [PMID: 40209968 DOI: 10.1016/j.cellsig.2025.111796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/24/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
Migrasomes are vesicular structures that form on elongated tethers originating from the tips or junctions of cellular tails during migration. These organelles, named for their vesicle rich lumen and release during cell movement, have gained attention for their role in intercellular communication and signal transduction. Migrasome formation is closely associated with the dynamic and active movement of cells, as well as with the intrinsic properties of cells and the extracellular microenvironment under various pathophysiological conditions. This review provides a comprehensive overview of migrasome dynamics, examining the mechanisms and distinct features of nanoscale vesicle-mediated intercellular signaling. It also highlights the influence of microscopic secretory factors on migrasome generation and formation. By comparing migrasomes with other active extracellular vesicles, this review highlights the advantages of migrasomes and addresses future challenges.
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of ECG, Sir Run Run Shaw Hospital, Sir Run Run Shaw Institute of Clinical Medicine of Zhejiang University, Hangzhou, China
| | - Shoutao Weng
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zaiwei Fan
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical College Nanchang University, Nanchang, Jiangxi 330006, China
| | - Dongyang Hu
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jiadi Le
- Department of Orthopaedics, Key Laboratory of Orthopaedics of Zhejiang Province, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kongsheng Sheng
- Department of Pharmacy, Taizhou Municipal Hospital, Taizhou 318000, China.
| |
Collapse
|
62
|
Mateos H, Mallardi A, Serrano-Pertierra E, Blanco-López MC, Liguori M, Antonacci Y, Casiello M, Palazzo G. Hetero Sandwich Immunoassay as Tool to Probe the Composition of the Extracellular Vesicles Membranes: The Case Study of L1CAM Localization. ACS OMEGA 2025; 10:12983-12992. [PMID: 40224478 PMCID: PMC11983164 DOI: 10.1021/acsomega.4c09363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Lateral flow immunoassays (LFIAs) are widely used for point-of-care diagnostic devices due to their simplicity, low cost, and rapid results. In this work, we demonstrate that a heterosandwich design LFIA can be an effective tool for verifying the presence of different proteins on the same particles. As a case study, we address a recent controversy regarding the presence of the protein L1CAM on the extracellular vesicles (EVs). EVs are crucial for cell communication and may serve as valuable disease biomarkers, including for neurodegenerative disorders. EVs from neuronal cells can cross the blood-brain barrier and be selectively isolated from plasma. Although L1CAM has been suggested as a marker for neuron-derived EVs, recent studies report that L1CAM exists as a cleaved soluble protein in plasma, not associated with EVs. We propose a heterosandwich LFIA to detect and quantify L1CAM and a confirmed EV marker, tetraspanin CD63 or CD9, on the same EV. This assay, together with several control experiments on EVs isolated from plasma by size exclusion chromatography (SEC), demonstrates that although most L1CAM in plasma is present as soluble cleaved proteins, 13% of the EVs are strongly associated with this protein. This evidence is confirmed by dynamic light scattering measurements, showing a significant size increase of gold nanoparticles conjugated with L1CAM antibodies when exposed to EVs but not to cleaved soluble L1CAM. Our results validate the selective immune-isolation of L1CAM-EVs, resolving the controversy by confirming that L1CAM is indeed associated with a significant fraction of EVs despite the presence of its soluble form in plasma.
Collapse
Affiliation(s)
- Helena Mateos
- Dipartimento
di Chimica and CSGI (Center for Colloid and Surface Science), Università degli Studi di Bari “Aldo
Moro”, Via Orabona n. 4 70125 Bari, Italy
| | - Antonia Mallardi
- CNR-IPCF,
Institute for Physical and Chemical Processes—Bari Division,
National Research Council (CNR), 00185 Rome, Italy
| | - Esther Serrano-Pertierra
- Departamento
de Bioquímica y Biología Molecular & Instituto Universitario
de Biotecnología de Asturias, Universidad
de Oviedo, 33006 Oviedo, Spain
| | - Maria Carmen Blanco-López
- Departamento
de Química Física y Analítica & Instituto
Universitario de Biotecnología de Asturias, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Maria Liguori
- CNR-ITB,
Institute of Biomedical Technologies—Bari Unit, National Research
Council (CNR), Via Amendola
n. 122, 70125 Bari, Italy
| | - Ylenia Antonacci
- CNR-ITB,
Institute of Biomedical Technologies—Bari Unit, National Research
Council (CNR), Via Amendola
n. 122, 70125 Bari, Italy
| | - Michele Casiello
- Dipartimento
di Chimica and CSGI (Center for Colloid and Surface Science), Università degli Studi di Bari “Aldo
Moro”, Via Orabona n. 4 70125 Bari, Italy
| | - Gerardo Palazzo
- Dipartimento
di Chimica and CSGI (Center for Colloid and Surface Science), Università degli Studi di Bari “Aldo
Moro”, Via Orabona n. 4 70125 Bari, Italy
| |
Collapse
|
63
|
Tarin M, Oryani MA, Javid H, Karimi-Shahri M. Exosomal PD-L1 in non-small cell lung Cancer: Implications for immune evasion and resistance to immunotherapy. Int Immunopharmacol 2025; 155:114519. [PMID: 40199140 DOI: 10.1016/j.intimp.2025.114519] [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: 11/05/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
Exosomes, characterized by their bilayer lipid structure, are crucial in mediating intercellular signaling and contributing to various physiological processes. Tumor cells produce distinct exosomes facilitating cancer progression, angiogenesis, and metastasis by conveying signaling molecules. A notable feature of these tumor-derived exosomes is the presence of programmed death-ligand 1 (PD-L1) on their surface. The PD-L1/programmed cell death receptor-1 (PD-1) signaling axis serves as a critical immune checkpoint, enabling tumors to evade immune detection and antitumor activity. The advancement of immunotherapy targeting the PD-1/PD-L1 pathway has significantly impacted the treatment landscape for non-small cell lung cancer (NSCLC). Despite its promise, evidence indicates that many patients experience limited responses or develop resistance to PD-1/PD-L1 inhibitors. Recent studies suggest that exosomal PD-L1 contributes to this resistance by modulating immune responses and tumor adaptability. This study reviews the PD-1/PD-L1 pathway's characteristics, current clinical findings on PD-L1 inhibitors in NSCLC, and exosome-specific attributes, with a particular focus on exosomal PD-L1. Furthermore, it examines the growing body of research investigating the role of exosomal PD-L1 in cancer progression and response to immunotherapy, underscoring its potential as a target for overcoming resistance in NSCLC treatment.
Collapse
Affiliation(s)
- Mojtaba Tarin
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
64
|
Kozela E, Petrovich-Kopitman E, Berger Y, Camacho AC, Shoham Y, Morandi MI, Rosenhek-Goldian I, Rotkopf R, Regev-Rudzki N. Spectral flow cytometry for detecting DNA cargo in malaria parasite-derived extracellular vesicles. J Biol Chem 2025:108481. [PMID: 40199399 DOI: 10.1016/j.jbc.2025.108481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025] Open
Abstract
Cells across biological kingdoms release extracellular vesicles (EVs) as a means of communication with other cells, be their friends or foes. This is indeed true for the intracellular malaria parasite Plasmodium falciparum (Pf), which utilizes EVs to transport bioactive molecules to various human host systems. Yet, the study of this mode of communication in malaria research is currently constrained due to limitations in high-resolution tools and the absence of commercial antibodies. Here, we demonstrate the power of an advanced spectral flow cytometry approach to robustly detect secreted EVs, isolated from Pf-infected red blood cells. By labeling both EV membrane lipids and the DNA cargo within (non-antibody staining approach), we were able to detect a subpopulation of parasitic-derived EVs enriched in DNA. Furthermore, we could quantitatively measure the DNA-carrying EVs isolated from two distinct blood stages of the parasite: rings and trophozoites. Our findings showcase the potential of spectral flow cytometry to monitor dynamic changes in nucleic acid cargo within pathogenic EVs.
Collapse
Affiliation(s)
- Ewa Kozela
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | | | - Yuval Berger
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Abel Cruz Camacho
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Yaara Shoham
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Mattia I Morandi
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Science, Prague, Czech Republic; The International Institute of Molecular Mechanisms and Machines, Polish Academy of Sciences, Warsaw, Poland
| | - Irit Rosenhek-Goldian
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
65
|
Zhao M, Wang J, Zhu S, Zhang S, Han C, Tan C, Huang Y, Sun Z, Wang L, Liu J. Human neural stem cell-derived exosomes activate PINK1/Parkin pathway to protect against oxidative stress-induced neuronal injury in ischemic stroke. J Transl Med 2025; 23:402. [PMID: 40188077 PMCID: PMC11971779 DOI: 10.1186/s12967-025-06283-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: 11/25/2024] [Accepted: 02/20/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Mitochondria play a critical role in oxidative stress (OS)-induced neuronal injury during ischemic stroke (IS), making them promising therapeutic targets. Mounting evidence underscores the extraordinary therapeutic promise of exosomes derived from human neural stem cells (hNSCs) in the management of central nervous system (CNS) diseases. Nonetheless, the precise mechanisms by which these exosomes target mitochondria to ameliorate the effects of IS remain only partially elucidated. This study investigates the protective effects of hNSC derived exosomes (hNSC-Exos) on neuronal damage. METHODS Using a rat model of middle cerebral artery occlusion (MCAO) in vivo and OS-induced HT22 cells in vitro. Firstly, our research group independently isolated human neural stem cells (hNSCs) and subsequently prepared hNSC-Exos. In vivo, MCAO rats were restored to blood flow perfusion to simulate ischemia-reperfusion injury, and hNSC-Exos were injected through stereotaxic injection into the brain. Subsequently, the protective effects of hNSC-Exos on MCAO rats were evaluated, including histological studies, behavioral assessments. In vivo, H2O2 was used in HT22 cells to simulate the OS environment in MCAO, and then its protective effects on HT22 were evaluated by co-culturing with hNSC-Exos, including immunofluorescence staining, western blotting (WB), quantitative real time PCR (qRT-PCR). In the process of exploring specific mechanisms, we utilized RNA sequencing (RNA-seq) to detect the potential induction of mitophagy in OS-induced HT22 cells. Afterwards, we employed a series of mitochondrial function assessments and autophagy related detection techniques, including measuring mitochondrial membrane potential, reactive oxygen species (ROS) levels, transmission electron microscopy (TEM) imaging, monodansylcadaverine (MDC) staining, and mCherry-GFP-LC3B staining. In addition, we further investigated the regulatory pathway of hNSC-Exos by using autophagy inhibitor mdivi-1 and knocking out PTEN induced kinase 1 (PINK1) in HT22 cells. RESULTS Administration of hNSC-Exos significantly ameliorated brain tissue damage and enhanced behavioral outcomes in MCAO rats. This treatment led to a reduction in brain tissue apoptosis and facilitated the normalization of impaired neurogenesis and neuroplasticity. Notably, the application of hNSC-Exos in vitro resulted in an upregulation of mitophagy in HT22 cells, thereby remedying mitochondrial dysfunction. We demonstrate that hNSC-Exos activate mitophagy via the PINK1/Parkin pathway, improving mitochondrial function and reducing neuronal apoptosis. CONCLUSIONS These findings suggest that hNSC-Exos alleviate OS-induced neuronal damage by regulating the PINK1/Parkin pathway. These reveals a novel role of stem cell-derived mitochondrial therapy in promoting neuroprotection and suggest their potential as a therapeutic approach for OS-associated CNS diseases, including IS.
Collapse
Affiliation(s)
- Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Shuaiyu Zhu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Shensen Zhang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Chao Han
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Chengcheng Tan
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Yubing Huang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Zhaokai Sun
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China.
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Local Joint Engineering Laboratory, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- National Genetic Test Center, The First Affiliated Hospital of Dalian Medical University, No. 193, Lianhe Road, Shahekou District, Dalian, 116011, People's Republic of China.
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Innovation Institute of Stem Cell and Precision Medicine, No. 57, Xinda Street, High-Tech Park, Dalian, 116023, People's Republic of China.
| |
Collapse
|
66
|
Wang J, Li Y, Wang Y, Wang G, Zhao C, Zhang Y, Lu H. Comparison of Protein Solubilization and Normalization Methods for Proteomics Analysis of Extracellular Vesicles from Urine. J Proteome Res 2025. [PMID: 40184522 DOI: 10.1021/acs.jproteome.4c01085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
Extracellular vesicles (EVs) play a vital role in numerous biological processes. Proteomic research of EVs is crucial for understanding their functions and potential therapeutic implications. Despite many sample preparation protocols for mass spectrometry-based proteomics of EVs being described, the variability in protein extraction across different protocols has not been extensively investigated. Moreover, given the inherent heterogeneity of EVs, it is vital to conduct a thorough evaluation of normalization methods. Here, we present a comprehensive comparison of three widely used lysis agents─sodium dodecyl sulfate (SDS), urea, and sodium deoxycholate (SDC)─for protein extraction from EVs. We also assess the impact of different normalization strategies on protein quantification, which is crucial for ensuring reliable results. Our results show that method-dependent differences in protein recovery were observed, particularly for membrane-associated proteins. We also find that common normalization strategies, such as urine creatinine and EV markers, did not significantly stabilize protein quantification, indicating that these methods are not universally applicable as normalization standards. Our work thereby provides a reference for the selection of MS sample preparation and normalization strategies for a given EV proteomics project.
Collapse
Affiliation(s)
- Jun Wang
- Liver Cancer Institute, Zhongshan Hospital and Department of Chemistry, Fudan University, Shanghai 200032, P. R. China
| | - Yang Li
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, P. R. China
| | - Yisheng Wang
- Liver Cancer Institute, Zhongshan Hospital and Department of Chemistry, Fudan University, Shanghai 200032, P. R. China
| | - Guoli Wang
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, P. R. China
| | - Chenyang Zhao
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, P. R. China
| | - Ying Zhang
- Liver Cancer Institute, Zhongshan Hospital and Department of Chemistry, Fudan University, Shanghai 200032, P. R. China
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, P. R. China
| | - Haojie Lu
- Liver Cancer Institute, Zhongshan Hospital and Department of Chemistry, Fudan University, Shanghai 200032, P. R. China
- Institutes of Biomedical Sciences and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
67
|
Rydland A, Heinicke F, Nyman TA, Trøseid AMS, Flåm ST, Stensland M, Gehin J, Eikeland J, Øvstebø R, Mjaavatten MD, Lie BA. Newly-diagnosed rheumatoid arthritis patients have elevated levels of plasma extracellular vesicles with protein cargo altered towards inflammatory processes. Sci Rep 2025; 15:11632. [PMID: 40185859 PMCID: PMC11971360 DOI: 10.1038/s41598-025-96325-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Extracellular vesicles (EVs) are implicated in rheumatoid arthritis (RA) but have mainly been assessed in RA patients taking disease modifying anti-rheumatic drugs. EVs are nanoparticles important in cell-cell communication and their molecular cargo are biomarker candidates. We characterized the protein profiles of EVs from blood plasma from newly diagnosed, treatment naïve RA patients (N = 32) and compared them to healthy controls (N = 20), by size exclusion chromatography-based EV enrichment coupled with high-resolution quantitative proteomics. The RA patients had higher EV concentration and larger EVs than controls. A total of 682 EV proteins were reliably quantified, and the overall profiles were distinctly different between patients and controls. Specifically, 26 proteins were significantly upregulated and 31 downregulated in RA patients, with several proteins acting in inflammatory networks and with immunologically important upstream regulators. The RA associated EVs appear, based on the tissue expression of their cargo proteins, to originate mainly from hepatocytes or immune cells, like neutrophils. Interestingly, the strongest RA associated EV proteins were inflammatory molecules, like SAA1 and S100A9, already suggested as biomarkers in RA. Furthermore, the RA associated EV proteins were generally not correlated with total serum protein levels, stressing the importance of EV transport of inflammatory proteins in RA pathogenesis.
Collapse
Affiliation(s)
- Anne Rydland
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.
| | - Fatima Heinicke
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Department of Biostatistics, Institute of Basic Medical Sciences, Oslo Centre for Biostatistics and Epidemiology, University of Oslo, Oslo, Norway
| | - Tuula A Nyman
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Anne-Marie Siebke Trøseid
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Siri T Flåm
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Maria Stensland
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Johanna Gehin
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Joakim Eikeland
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Reidun Øvstebø
- The Blood Cell Research Group, Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Maria Dahl Mjaavatten
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Division of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Benedicte A Lie
- Department of Medical Genetics, University of Oslo and Oslo University Hospital, Postboks 4956 Nydalen, OUS HF Ullevål sykehus, 0424, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Center for Treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway.
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
68
|
Eerdekens H, Pirlet E, Willems S, Bronckaers A, Pincela Lins PM. Extracellular vesicles: innovative cell-free solutions for wound repair. Front Bioeng Biotechnol 2025; 13:1571461. [PMID: 40248643 PMCID: PMC12003306 DOI: 10.3389/fbioe.2025.1571461] [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: 02/05/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
Chronic non-healing wounds are often associated with conditions such as diabetes and peripheral vascular disease, pose significant medical and socioeconomic challenges. Cell-based therapies have shown promise in promoting wound healing but have major drawbacks such as immunogenicity and tumor formation. As a result, recent research has shifted to the potential of extracellular vesicles (EVs) derived from these cells. EVs are nanosized lipid bilayer vesicles, naturally produced by all cell types, which facilitate intercellular communication and carry bioactive molecules, offering advantages such as low immunogenicity, negligible toxicity and the potential to be re-engineered. Recent evidence recognizes that during wound healing EVs are released from a wide range of cells including immune cells, skin cells, epithelial cells and platelets and they actively participate in wound repair. This review comprehensively summarizes the latest research on the function of EVs from endogenous cell types during the different phases of wound healing, thereby presenting interesting therapeutic targets. Additionally, it gives a critical overview of the current status of mesenchymal stem cell-derived EVs in wound treatment highlighting their tremendous therapeutic potential as a non-cellular of-the-shelf alternative in wound care.
Collapse
Affiliation(s)
- Hanne Eerdekens
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Elke Pirlet
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Sarah Willems
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Annelies Bronckaers
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
| | - Paula M. Pincela Lins
- Hasselt University, Faculty of Medicine and Life Sciences, Biomedical Research Institute (BIOMED), Diepenbeek, Belgium
- Flemish Institute for Technological Research (VITO), Environmental Intelligence Unit, Mol, Belgium
| |
Collapse
|
69
|
Manolopoulos A, Yao PJ, Kapogiannis D. Extracellular vesicles: translational research and applications in neurology. Nat Rev Neurol 2025:10.1038/s41582-025-01080-z. [PMID: 40181198 DOI: 10.1038/s41582-025-01080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2025] [Indexed: 04/05/2025]
Abstract
Over the past few decades, extensive basic, translational and clinical research has been devoted to deciphering the physiological and pathogenic roles of extracellular vesicles (EVs) in the nervous system. The presence of brain cell-derived EVs in the blood, carrying diverse cargoes, has enabled the development of predictive, diagnostic, prognostic, disease-monitoring and treatment-response biomarkers for various neurological disorders. In this Review, we consider how EV biomarkers can bring us closer to understanding the complex pathogenesis of neurological disorders such as Alzheimer disease, Parkinson disease, stroke, traumatic brain injury, amyotrophic lateral sclerosis and multiple sclerosis. We describe how translational research on EVs might unfold bidirectionally, proceeding from basic to clinical studies but also in the opposite direction, with biomarker findings in the clinic leading to novel hypotheses that can be tested in the laboratory. We demonstrate the potential value of EVs across all stages of the therapeutic development pipeline, from identifying therapeutic targets to the use of EVs as reporters in model systems and biomarkers in clinical research. Finally, we discuss how the cargo and physicochemical properties of naturally occurring and custom-engineered EVs can be leveraged as novel treatments and vehicles for drug delivery, potentially revolutionizing neurotherapeutics.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Pamela J Yao
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
70
|
Tiwari A, Soni N, Dongre S, Chaudhary M, Bissa B. The role of plant-derived extracellular vesicles in ameliorating chronic diseases. Mol Biol Rep 2025; 52:360. [PMID: 40180626 DOI: 10.1007/s11033-025-10466-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Plant-derived extracellular vesicles (PDEVs) have been shown to have a promising role in treating chronic illnesses. Plants secrete these vesicles containing bioactive chemicals such as proteins, lipids, nucleic acids, and small metabolites. Because of their unique structure, PDEVs affect many biological processes, which makes them an ideal candidate for treating the complex pathophysiology of chronic diseases. Recent studies have shown that PDEVs have anti-inflammatory and antioxidant properties. Extracellular vesicles (EVs) possess diverse therapeutic potential, including anti-inflammatory, antioxidant, and regenerative properties. By regulating immune responses, scavenging free radicals, and promoting tissue repair, EVs can address various chronic diseases such as cardiovascular disorders, neurological conditions, skin diseases, and inflammatory ailments. In preclinical models, PDEVs have been demonstrated to improve heart function and minimize the size of myocardial infarctions. In neurodegenerative illnesses, they can pass through the blood-brain barrier and deliver neuroprotective medicines to the brain. Furthermore, PDEVs have shown promise in enhancing insulin sensitivity and lowering hyperglycemia in diabetic animals. In this review article, we attempt to explain the diverse therapeutic potential of PDEVs in ameliorating chronic diseases.
Collapse
Affiliation(s)
- Ashwani Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Naveen Soni
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Shweta Dongre
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Megha Chaudhary
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India
| | - Bhawana Bissa
- Department of Biochemistry, Central University of Rajasthan, Ajmer, India.
| |
Collapse
|
71
|
Cho H, Ju H, Ahn Y, Jang J, Cho J, Park E, Kang SM, Lee J, Seo D, Baek MC, Yea K. Engineered extracellular vesicles with surface FGF21 and enclosed miR-223 for treating metabolic dysfunction-associated steatohepatitis. Biomaterials 2025; 321:123321. [PMID: 40209593 DOI: 10.1016/j.biomaterials.2025.123321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disorder with a complex pathogenesis that requires combination therapies rather than monotherapies. Extracellular vesicles (EVs) exhibit inherently efficient delivery to the liver and can be engineered to carry various therapeutic substances, making them promising agents. In this study, EVs were engineered to display fibroblast growth factor 21 (FGF21) on their surface and encapsulate miR-223 (223/F-EVs), aiming to improve steatosis and alleviate inflammation and fibrosis, respectively. Introducing the 223/F-EVs into human liver cell lines significantly reduced both basal and induced levels of lipid storage, inflammation, and fibrosis markers. Furthermore, using an FGF21-blocking antibody or miR-223 inhibitor effectively diminished the efficacy of the 223/F-EVs, confirming the essential roles of FGF21 and miR-223 in these processes. In a Choline-Deficient, l-Amino acid-defined, High-Fat Diet (CDAHFD)-fed mouse model, intravenously administered 223/F-EVs demonstrated liver-preferential delivery and a marked reduction in the MASH phenotype without compromising bone density, unlike conventional FGF21 treatment. Collectively, 223/F-EVs convey FGF21 and miR-223 exclusively to the liver, offering strategic advantages by mitigating MASH progression via multiple pathways. This study lays a solid foundation for further investigation of engineered EVs as a transformative therapeutic approach for treating MASH.
Collapse
Affiliation(s)
- Hanchae Cho
- Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Hyunji Ju
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Yongdeok Ahn
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhyeong Cho
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Eunju Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Sung-Min Kang
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea.
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea; New Biology Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 43024, Republic of Korea.
| |
Collapse
|
72
|
Shen H, Chen J, Liu M, Zhao M, Hu D, Xie F, Jin Q, Xiao D, Peng Z, Qin T, Rao D, Huang D. Research progress of extracellular vesicles derived from mesenchymal stem cells in the treatment of neurodegenerative diseases. Front Immunol 2025; 16:1496304. [PMID: 40242755 PMCID: PMC12000061 DOI: 10.3389/fimmu.2025.1496304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
As the world's population ages, neurodegenerative diseases are becoming more widely acknowledged as serious global health and socioeconomic issues. Although many resources have been devoted to the research of these illnesses, little progress has been made in the creation of novel diagnostic and therapeutic approaches. Extracellular vesicles (EVs) are released by all cell types and contain proteins, microRNAs, mRNAs, and other biologically active molecules. EVs play an important role in intercellular communication as well as in the regulation of neuroinflammation. Determining the mechanisms by which EVs contribute to the pathogenesis of neurodegenerative diseases will aid in the development of new therapeutic approaches and diagnostic tools. Mesenchymal stem cells (MSCs) have been shown in studies to control immunological responses, promote the growth of new brain connections, promote the production of blood vessels, and heal damaged tissues. There is growing evidence that MSCs' ability to treat patients is mostly due to the neurotrophic compounds they secrete through EVs. Since their tiny size allows them to pass through biological barriers and reach injured parts of the central nervous system, MSC-derived extracellular vesicles (MSC-EVs) retain many of the therapeutic qualities of their parent MSCs. This review discusses the role of EVs in neurodegenerative diseases and highlights the potential of MSC-EVs in the treatment of neurodegenerative diseases. The paper also examines the challenges that still need to be overcome and the prospects for using MSC-EVs to treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Haibin Shen
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jie Chen
- Department of Laboratory Medicine, Yongchuan Hospital of Chongqing Medical University, Chongqing, Yongchuan, China
| | - Meijin Liu
- Laboratory Medicine, People’s Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Minghong Zhao
- Laboratory Medicine, Guizhou Aerospace Hospital, Zunyi, China
| | - Die Hu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fangfang Xie
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qing Jin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Dewang Xiao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Zongbo Peng
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Tao Qin
- Laboratory Medicine, Guizhou Aerospace Hospital, Zunyi, China
| | - Dingyu Rao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
73
|
Kim J, Park H, Park NY, Hwang SI, Kim YE, Sung SI, Chang YS, Koh A. Functional maturation of preterm intestinal epithelium through CFTR activation. Commun Biol 2025; 8:540. [PMID: 40169914 PMCID: PMC11961738 DOI: 10.1038/s42003-025-07944-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
Preterm birth disrupts intestinal epithelial maturation, impairing digestive and absorptive functions. This study integrates analysis of single-cell RNA sequencing datasets, spanning fetal to adult stages, with human preterm intestinal models derived from the ileal tissue of preterm infants. We investigate the potential of extracellular vesicles (EVs) derived from human Wharton's jelly mesenchymal stem cells to promote intestinal maturation. Distinct enterocyte differentiation trajectories are identified during the transition from immature to mature stages of human intestinal development. EV treatment, particularly with the EV39 line, significantly upregulates maturation-specific gene expression related to enterocyte function. Gene set enrichment analysis reveals an enrichment of TGFβ1 signaling pathways, and proteomic analysis identifies TGFβ1 and FGF2 as key mediators of EV39's effects. These treatments enhance cell proliferation, epithelial barrier integrity, and fatty acid uptake, primarily through CFTR-dependent mechanisms-unique to human preterm models, not observed in mouse intestinal organoids. This highlights the translational potential of EV39 and CFTR activation in promoting the functional maturation of the premature human intestine.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Hyunji Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Na-Young Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Se In Hwang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, 06351, South Korea
| | - Young Eun Kim
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, 06351, South Korea
| | - Se In Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea.
| | - Yun Sil Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, 06351, South Korea.
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, 06351, South Korea.
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, South Korea.
| | - Ara Koh
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea.
| |
Collapse
|
74
|
Yu Z, Swift KA, Hedges MA, Theiss AL, Andres SF. Microscopic messengers: Extracellular vesicles shaping gastrointestinal health and disease. Physiol Rep 2025; 13:e70292. [PMID: 40165585 PMCID: PMC11959161 DOI: 10.14814/phy2.70292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The field of extracellular vesicles (EVs) is advancing rapidly, and this review aims to synthesize the latest research connected to EVs and the gastrointestinal tract. We will address new and emerging roles for EVs derived from internal sources such as the pancreas and immune system and how these miniature messengers alter organismal health or the inflammatory response within the GI tract. We will examine what is known about external EVs from dietary and bacterial sources and the immense anti-inflammatory, immune-modulatory, and proliferative potential within these nano-sized information carriers. EV interactions with the intestinal and colonic epithelium and associated immune cells at homeostatic and disease states, such as necrotizing enterocolitis (NEC) and inflammatory bowel disease (IBD) will also be covered. We will discuss how EVs are being leveraged as therapeutics or for drug delivery and conclude with a series of unanswered questions in the field.
Collapse
Affiliation(s)
- Zhantao Yu
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Kevin A. Swift
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Madeline A. Hedges
- Department of Neonatology, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| | - Arianne L. Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine and the Mucosal Inflammation ProgramUniversity of Colorado School of MedicineAuroraColoradoUSA
- Rocky Mountain Regional Veterans Affairs Medical CenterAuroraColoradoUSA
| | - Sarah F. Andres
- Department of Pediatrics, Pediatric GI Division, School of MedicineOregon Health and Science UniversityPortlandOregonUSA
| |
Collapse
|
75
|
Tao K, Tao K, Wang J. The potential mechanisms of extracellular vesicles in transfusion-related adverse reactions: Recent advances. Transfus Clin Biol 2025:S1246-7820(25)00049-7. [PMID: 40180029 DOI: 10.1016/j.tracli.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Blood transfusion is an irreplaceable clinical treatment. Blood components are differentiated and stored according to specific guidelines. Storage temperatures and times vary depending on the blood component, but they all release extracellular vesicles (EVs) during storage. Although blood transfusions can be life-saving, they can also cause many adverse transfusion reactions, among which the effects of EVs are of increasing interest to researchers. EVs are submicron particles that vary in size, composition, and surface biomarkers, are encapsulated by a lipid bilayer, and are not capable of self-replication. EVs released by blood cells are important contributors to pathophysiologic states through proinflammatory, coagulant, and immunosuppressive effects, which in turn promote or inhibit the associated disease phenotype. Therefore, this review explores the potential mechanisms of hematopoietic-derived EVs in transfusion-associated adverse reactions and discusses the potential of the latest proteomics tools to be applied to the analysis of EVs in the field of transfusion medicine with a view to reducing the risk of blood transfusion.
Collapse
Affiliation(s)
- Keyi Tao
- Panzhihua University, Panzhihua 617000 Sichuan, China
| | - Keran Tao
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000 Hubei, China
| | - Jing Wang
- Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000 China.
| |
Collapse
|
76
|
Khorrami-Nejad M, Hashemian H, Majdi A, Jadidi K, Aghamollaei H, Hadi A. Application of stem cell-derived exosomes in anterior segment eye diseases: A comprehensive update review. Ocul Surf 2025; 36:209-219. [PMID: 39884389 DOI: 10.1016/j.jtos.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Mesenchymal stem cell (MSC) therapy has emerged as a promising approach for addressing various eye-related conditions. Yet, its clinical application faces challenges due to issues such as limited biocompatibility and difficulties in effectively delivering treatment to specific ocular tissues. Recent studies have shifted attention towards MSC-derived exosomes, which share similar regenerative, reparative, and immunomodulatory capabilities with their origin cells. This review delves into the latest research on the use of MSC-derived exosomes for treating anterior segment diseases of the eye. It explores the exosomes' composition, biological functions, and the methods used for their isolation, as well as their roles in disease progression, diagnosis, and therapy. The review critically assesses the therapeutic advantages and mechanisms of action of MSC-derived exosomes in treating conditions like dry eye disease, Sjogren's syndrome, keratoconus, corneal lesions, and corneal allograft rejection. Additionally, it discusses the obstacles and future prospects of employing MSC-derived exosomes as innovative therapies for anterior segment eye diseases. This comprehensive overview underscores the significant potential of MSC-derived exosomes in transforming the treatment paradigm for anterior segment eye disorders, while also highlighting the necessity for further research to enhance their clinical application.
Collapse
Affiliation(s)
- Masoud Khorrami-Nejad
- Optometry Department, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran; Translational Ophthalmology Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hesam Hashemian
- Translational Ophthalmology Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Majdi
- Optical Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Aghamollaei
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Hadi
- Optometry Department, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
77
|
Chen L, Shao Y, Geng J, Liu H, Liu Q, Cheng Y, Sun T. Dual Role of Exosomes in Parkinson's Disease: Adenine Exerts a Beneficial Effect. CNS Neurosci Ther 2025; 31:e70331. [PMID: 40237545 PMCID: PMC12001426 DOI: 10.1111/cns.70331] [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: 01/09/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 04/18/2025] Open
Abstract
AIMS Developing validated treatments for Parkinson's disease (PD) remains a priority for clinicians and researchers. The lack of viable therapies may stem from an incomplete understanding of PD pathogenesis and inadequate therapeutic candidates. The production and transmission of exosomes are gaining recognition in the pathogenesis of neurodegenerative diseases. However, how exosomes affect the pathophysiology of PD has not been well elucidated. METHODS Here, we investigated the effect of exosomes secreted by rats that were treated with saline or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP) in treating healthy or PD model mice, and we evaluated the efficacy of peripheral and intracranial administration of adenine, which is an exosomal metabolite identified through widely targeted metabolomics. RESULTS We found that exosomes derived from the blood of healthy rats alleviated motor dysfunction, dopaminergic neuron loss in the substantia nigra pars compacta and striatum, oxidative injury, and neuroinflammation. Conversely, exosomes from the blood of PD model rats reproduced the behavioral phenotype and pathology of PD in healthy mice. Additionally, peripheral and intracranial administration of adenine ameliorated the motor coordination disorder and dopaminergic neuron loss, and maintained the homeostasis of oxidative stress and neuroinflammation by activating cAMP/PKA signaling in PD. CONCLUSION Together, these findings shed light on the mechanism by which exosomes participate in the pathophysiology of PD by transmitting the metabolite adenine and providing potential therapeutic strategies.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
| | - Yi‐Ting Shao
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
| | - Ji Geng
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
| | - Hua Liu
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
| | - Qing‐Shan Liu
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
| | - Yong Cheng
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
- Institute of National SecurityMinzu University of ChinaBeijingP. R. China
| | - Ting Sun
- Key Laboratory for Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, School of PharmacyMinzu University of ChinaBeijingChina
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingP. R. China
| |
Collapse
|
78
|
Zhao Y, Wen C, Wang Q, Qing Y, Tondi S, Reina C, Šabanović B, Chang CY, Lai C, Wang H, Agerbaek MØ, Clausen TM, Gustavsson T, Theander TG, Salanti A, Meny CC, Shen B, Aicher A, Tang J, Heeschen C. Use of the Malaria Protein VAR2CSA for the Detection of Small Extracellular Vesicles to Diagnose Adenocarcinoma. J Extracell Vesicles 2025; 14:e70067. [PMID: 40241173 PMCID: PMC12003099 DOI: 10.1002/jev2.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge for early diagnosis due to the lack of sensitive and specific biomarkers. This encouraged us to explore the diagnostic value of cancer-derived small extracellular vesicles (sEVs) as early detection biomarkers. We previously showed that the recombinant malaria protein VAR2CSA (rVAR2) selectively binds to oncofetal chondroitin sulfate (ofCS) on the surfaces of cancer cells, which might be useful for identifying cancer-derived sEVs. Indeed, flow cytometry revealed strong ofCS expression in PDAC cell-derived sEVs, as evidenced by the presence of mutant KRAS, a common genetic alteration in PDAC. Plasma from PDAC patients showed significantly higher ofCS+ sEV levels compared to healthy donors and patients with benign gastrointestinal diseases. ROC analysis for ofCS+ sEVs revealed an AUC of 0.9049 for the detection of all-stage and 0.9222 for early-stage PDAC. Notably, mutant KRAS was also detected in these patient-derived sEVs. Most intriguingly, combining ofCS+ sEVs and CA19-9 resulted in an AUC of 0.9707 for the detection of early PDAC. Our study demonstrates that rVAR2 is suitable for detecting ofCS+ cancer-derived sEVs in plasma, thereby providing high efficiency for identifying PDAC patients among a diverse population. These findings suggest that rVAR2-based sEV detection could serve as a powerful diagnostic tool to improve patient survival through early detection.
Collapse
Affiliation(s)
- Yaru Zhao
- Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chenlei Wen
- Research Institute of Pancreatic Disease, Ruijin Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Pancreatic Disease Center, Ruijin Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Qi Wang
- Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yue Qing
- Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Serena Tondi
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute FPO‐IRCCS, CandioloTurinItaly
| | - Chiara Reina
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute FPO‐IRCCS, CandioloTurinItaly
| | - Berina Šabanović
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute FPO‐IRCCS, CandioloTurinItaly
| | - Cherry Yin‐Yi Chang
- Department of Medicine, School of MedicineChina Medical UniversityTaichungTaiwan
- Department of Obstetrics and GynecologyChina Medical University HospitalTaichungTaiwan
| | - Chu‐Hu Lai
- Precision Immunotherapy, Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
| | - Huimin Wang
- Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mette Ø. Agerbaek
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
- VarCT DiagnosticsCopenhagenDenmark
| | - Thomas M. Clausen
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Tobias Gustavsson
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
- VAR2 PharmaceuticalsCopenhagenDenmark
| | - Thor G. Theander
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology (CMP) at Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
| | - Clara Csilla Meny
- 2 Institute for Pathology and Experimental Oncology ResearchSemmelweis UniversityBudapestHungary
| | - Baiyong Shen
- Research Institute of Pancreatic Disease, Ruijin Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Pancreatic Disease Center, Ruijin Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical SciencesChina Medical UniversityTaichungTaiwan
| | - Jiajia Tang
- Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Christopher Heeschen
- Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Systems Medicine for CancerShanghai Jiao Tong University School of MedicineShanghaiChina
- Pancreatic Cancer HeterogeneityCandiolo Cancer Institute FPO‐IRCCS, CandioloTurinItaly
| |
Collapse
|
79
|
Brugière O, Dreyfuss D, Guilet R, Rong S, Hirschi S, Renaud-Picard B, Reynaud-Gaubert M, Coiffard B, Bunel V, Messika J, Demant X, Le Pavec J, Dauriat G, Saint Raymond C, Falque L, Mornex JF, Tissot A, Lair D, Le Borgne Krams A, Bousseau V, Magnan A, Picard C, Roux A, Glorion M, Carmagnat M, Gazeau F, Aubertin K, Carosella E, Vallée A, Landais C, Rouas-Freiss N, LeMaoult J. Circulating Vesicular-bound HLA-G as Noninvasive Predictive Biomarker of CLAD After Lung Transplantation. Transplantation 2025; 109:736-745. [PMID: 39294868 DOI: 10.1097/tp.0000000000005175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
BACKGROUND Circulating extracellular vesicles (EVs) have shown promising results as noninvasive biomarkers for predicting disease outcomes in solid organ transplantation. Because in situ graft cell expression of the tolerogenic molecule HLA-G is associated with acceptance after lung transplantation (LTx), we hypothesized that plasma EV-bound HLA-G (HLA-G EV ) levels could predict chronic lung allograft dysfunction (CLAD) development. METHODS We analyzed 78 LTx recipients from the Cohort-for-Lung-Transplantation cohort, all in a stable (STA) state within the first year post-LTx. At 3 y, 41 patients remained STA, and 37 had CLAD (bronchiolitis obliterans syndrome, BOS, [n = 32] or restrictive allograft syndrome [n = 5]). HLA-G EV plasma levels were measured at month 6 (M6) and M12 in 78 patients. CLAD occurrence and graft failure at 3 y post-LTx were assessed according to early HLA-G EV plasma levels. RESULTS In patients with subsequent BOS, (1) HLA-G EV levels at M12 were significantly lower than those in STA patients ( P = 0.013) and (2) also significantly lower than their previous levels at M6 ( P = 0.04).A lower incidence of CLAD and BOS and higher graft survival at 3 y were observed in patients with high HLA-G EV plasma levels at M12 (high versus low HLA-G EVs patients [cutoff 21.3 ng/mL]: freedom from CLAD, P = 0.002; freedom from BOS, P < 0.001; and graft survival, P = 0.04, [log-rank]). Furthermore, in multivariate analyses, low HLA-G EV levels at M12 were independently associated with a subsequent risk of CLAD, BOS, and graft failure at 3 y ( P = 0.015, P = 0.036, and P = 0.026, respectively [Cox models]). CONCLUSIONS This exploratory study suggests the potential of EV-bound HLA-G plasma levels as a liquid biopsy in predicting CLAD/BOS onset and subsequent graft failure.
Collapse
Affiliation(s)
- Olivier Brugière
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital Foch, Suresnes, France
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| | - Dora Dreyfuss
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| | - Ronan Guilet
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| | - Sophie Rong
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| | - Sandrine Hirschi
- Department of Pneumology, Strasbourg Lung Transplant Program, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | - Benjamin Renaud-Picard
- Department of Pneumology, Strasbourg Lung Transplant Program, Strasbourg University Hospital, Strasbourg, France
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | | | - Benjamin Coiffard
- Service de Pneumologie et Transplantation Pulmonaire, CHU de Marseille, Marseille, France
| | - Vincent Bunel
- APHP.Nord-Université de Paris, Hôpital Bichat-Claude Bernard, Service de Pneumologie B et Transplantation Pulmonaire, Paris, France
| | - Jonathan Messika
- APHP.Nord-Université de Paris, Hôpital Bichat-Claude Bernard, Service de Pneumologie B et Transplantation Pulmonaire, Paris, France
| | - Xavier Demant
- Service de Pneumologie et Transplantation Pulmonaire, CHU de Bordeaux, Bordeaux, France
| | - Jérôme Le Pavec
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - Gaelle Dauriat
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - Christel Saint Raymond
- Service Hospitalier Universitaire de Pneumologie et Physiologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Échirolles, France
| | - Loic Falque
- Service Hospitalier Universitaire de Pneumologie et Physiologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Échirolles, France
| | - Jean-Francois Mornex
- Université Claude Bernard Lyon1, INRAE, IVPC, Lyon, France
- Hospices Civils de Lyon, Inserm CIC1407, Bron, France
| | - Adrien Tissot
- Nantes Université, CHU Nantes, INSERM, Service de Pneumologie, l'institut du thorax, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - David Lair
- Nantes Université, CHU Nantes, INSERM, Service de Pneumologie, l'institut du thorax, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | | | - Veronique Bousseau
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital HEGP, Paris, France
| | - Antoine Magnan
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital Foch, Suresnes, France
| | - Clément Picard
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital Foch, Suresnes, France
| | - Antoine Roux
- Service de Pneumologie et Transplantation Pulmonaire, Hôpital Foch, Suresnes, France
| | | | | | - Florence Gazeau
- Université Paris Cité, MSC et intégrateur IVETh, CNRS UMR7057, Paris, France
| | - Kelly Aubertin
- Université Paris Cité, MSC et intégrateur IVETh, CNRS UMR7057, Paris, France
| | - Edgardo Carosella
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| | - Alexandre Vallée
- Service d'épidémiologie et santé publique, Hôpital Foch, Suresnes, France
| | - Cecile Landais
- Departement de biostatistiques, DRCI Hôpital Foch, Suresnes, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| | - Joel LeMaoult
- CEA, DRF-Institut de Biologie Francois Jacob, Service de Recherches en Hémato-Immunologie, Hôpital Saint-Louis, Paris, France
- U976 HIPI, IRSL, Université Paris Cité, Paris, France
| |
Collapse
|
80
|
Bare Y, Defourny K, Bretou M, Van Niel G, Nolte-'t Hoen E, Gaudin R. The endoplasmic reticulum as a cradle for virus and extracellular vesicle secretion. Trends Cell Biol 2025; 35:282-293. [PMID: 39730274 DOI: 10.1016/j.tcb.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024]
Abstract
Extracellular vesicles (EVs) are small membranous carriers of protein, lipid, and nucleic acid cargoes and play a key role in intercellular communication. Recent work has revealed the previously under-recognized participation of endoplasmic reticulum (ER)-associated proteins (ERAPs) during EV secretion, using pathways reminiscent of viral replication and secretion. Here, we present highlights of the literature involving ER/ERAPs in EV biogenesis and propose mechanistic parallels with ERAPs exploited during viral infections. We propose that ERAPs play an active role in the release of EVs and viral particles, and we present views on whether viruses hijack or enhance pre-existing ERAP-dependent secretory machineries or whether they repurpose ERAPs to create new secretory pathways.
Collapse
Affiliation(s)
- Yonis Bare
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS UMR9004, Université Montpellier, Montpellier, France.
| | - Kyra Defourny
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marine Bretou
- Université Paris Cité, Institut de Psychiatrie et Neurosciences de Paris (IPNP), INSERM U1266, Paris, France
| | - Guillaume Van Niel
- CRCI2NA, Nantes Université, INSERM UMR1307, CNRS UMR6075, Université d'Angers, Nantes, France; GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| | - Esther Nolte-'t Hoen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Raphael Gaudin
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS UMR9004, Université Montpellier, Montpellier, France.
| |
Collapse
|
81
|
Peng J, He J, Lin L, Li Y, Xia Y. Neural Stem Cell Extracellular Vesicles Carrying YBX1 Inhibited Neuronal Pyroptosis Through Increasing m6A-modified GPR30 Stability and Expression in Ischemic Stroke. Transl Stroke Res 2025; 16:262-279. [PMID: 37966628 DOI: 10.1007/s12975-023-01210-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
Neural stem cell-derived extracellular vesicles (NSC-derived EVs) alleviated ischemic stroke (IS) by suppressing the activation of nucleotide-binding domain leucine-rich repeats family protein 3 (NLRP3) inflammasome and neuronal pyroptosis. However, the specific mechanism needs further investigation. qRT-qPCR, Western blotting, and immunofluorescence detected related gene expression. Immunofluorescent analyzed the expression of Ki-67, βIII-Tubulin (Tuj1), and GFAP. Lactate dehydrogenase (LDH) release and IL-1β and IL-18 levels were analyzed by LDH and ELISA kits. TTC staining evaluated the infarction of brain tissues. Flow cytometric analysis measured caspase-1 activity. M6A methylated RNA immunoprecipitation PCR (MeRIP-PCR) measured methylation levels of G protein-coupled receptor 30 (GPR30). RIP and Co-IP analyzed the interactions of Y box binding protein (YBX1)/GPR30, YBX1/IGF2BP1 and NLRP3/speckle-type POZ protein (SPOP), as well as the ubiquitination levels of NLRP3. NSC-derived EVs inhibited the ischemia-reperfusion (I/R) injury of rats and the neuronal pyroptosis induced by oxygen-glucose deprivation/reoxygenation (OGD/R). Knockdown of EVs carrying YBX1 or GPR30 silencing abolished these inhibiting effects. GPR30 mRNA and IGF2BP1 protein were enriched by YBX1 antibody. YBX1 enhanced the stability of m6A-modified GPR30 by interacting with IGF2BP1 and thus promoting GPR30 expression. Knockdown of IGF2BP1 suppressed the binding between YBX1 and GPR30 mRNA. GPR30 promoted NLRP3 ubiquitination by interacting with SPOP. EVs carrying YBX1 could reduce the infarction of brain tissues and inhibit neuronal pyroptosis in rats with I/R injury. NSC-derived EVs carrying YBX1 increased the stability of m6A-modified GPR30 by interacting with IGF2BP1; the upregulation of GPR30 inhibited the activation of NLRP3 inflammasome through promoting NLRP3 ubiquitination by SPOP, ultimately suppressing the neuronal pyroptosis in IS.
Collapse
Affiliation(s)
- Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - Jun He
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - Long Lin
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - You Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China.
| |
Collapse
|
82
|
Zhang J, Liu J, Zhang H, Liu B, Li L, Li Y, Pei J, Lin Q, Chen Q, Lin J. Lymph node-targeted delivery of Lonicera japonica thunb. polysaccharides for enhancing antitumor immunotherapy. Mater Today Bio 2025; 31:101559. [PMID: 40026631 PMCID: PMC11871467 DOI: 10.1016/j.mtbio.2025.101559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/15/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Dendritic cells (DCs) are crucial for the initiation and regulation of innate and adaptive immunity. Their maturity and infiltration in the tumor largely determine the efficiency of antigen presentation, the CTL responses, and the prognosis of tumors. However, the application of common immunoregulatory plant polysaccharides to DCs in vivo still represents major challenges due to the off-target effect and short biological lifespan. Lonicera japonica Thunb. polysaccharides (LJP) were found to exert benign immunoregulatory ability, but the effectiveness of utilizing LJP alone is unsatisfactory. As a result, we innovatively encapsulated LJP in into the exosomes derived from mouse bone mesenchymal stem cells (BMSCs) to form a DC-activated inducer (LJP-exosome). LJP-exosomes possessed a profound ability to target lymph nodes and the co-stimulatory capability of DCs compared with the application of LJP alone. Adequate results have shown that DCs primed by LJP-exosomes enhanced the tumor-reactive CD8+ T cell responses, leading to prophylactic tumor inhibition in an immunologically ignorant tumor model. The study proposed offers a promising strategy for enhancing the immune activation efficacy of extracted polysaccharides of traditional Chinese medicine by building the patients' immunity, thus consolidating the overall prognosis.
Collapse
Affiliation(s)
- Jiatong Zhang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Jintong Liu
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Hong Zhang
- The Eighth Medical Center of the Chinese People's Liberation Army General Hospital, Beijing, China
| | - Biao Liu
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Lujie Li
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Yifan Li
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Jingrou Pei
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Qiao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
| | - Qi Chen
- Molecular and Nanoscale Physics Group, School of Physics and Astronomy, University of Leeds, Leeds LS2 9JT, UK
| | - Jiahao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing 100193, China
- Center of Research and Innovation of Chinese Traditional Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
83
|
Wakale S, Chen Y, Sun AR, Liyanage C, Gunter J, Batra J, Crawford R, Sang H, Prasadam I. Comparative Analysis of the Therapeutic Potential of Extracellular Vesicles Secreted by Aged and Young Bone Marrow-Derived Mesenchymal Stem Cells in Osteoarthritis Pathogenesis. Cell Prolif 2025; 58:e13776. [PMID: 39703140 PMCID: PMC11969246 DOI: 10.1111/cpr.13776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Osteoarthritis (OA), a joint disease, burdens global healthcare due to aging and obesity. Recent studies show that extracellular vesicles (EVs) from bone marrow-derived mesenchymal stem cells (BMSCs) contribute to joint homeostasis and OA management. However, the impact of donor age on BMSC-derived EV efficacy remains underexplored. In this study, we investigated EV efficacy from young BMSCs (2-month-old) in mitigating OA, contrasting them with EVs from aged BMSCs (27-month-old). The study used destabilisation of the medial meniscus (DMM) surgery on mouse knee joints to induce accelerated OA. Cartilage degeneration markers and senescence markers' expression levels were investigated in response to EV treatment. The therapeutic impact of EVs on chondrocytes under inflammatory responses was also evaluated. Despite having similar morphologies, EVs from young BMSCs markedly decreased senescence and improved chondroprotection by activating the PTEN pathway while simultaneously suppressing the upregulation of the PI3K/AKT pathways, proving to be more effective than those from older BMSCs in vitro. Furthermore, intraperitoneal injections of EVs from young donors significantly mitigated OA progression by preserving cartilage and reducing synovitis in a surgical OA model using DMM in mice. These findings highlight that donor age as a critical determinant in the therapeutic potential of BMSC-derived EVs for clinical use in OA treatment.
Collapse
Affiliation(s)
- Shital Wakale
- Centre for Biomedical TechnologiesQueensland University of TechnologyBrisbaneAustralia
- School of Mechanical, Medical & Process EngineeringQueensland University of TechnologyBrisbaneAustralia
| | - Yang Chen
- Department of OrthopaedicsShenzhen Hospital of Southern Medical UniversityShenzhenChina
| | - Antonia Rujia Sun
- Centre for Biomedical TechnologiesQueensland University of TechnologyBrisbaneAustralia
- School of Mechanical, Medical & Process EngineeringQueensland University of TechnologyBrisbaneAustralia
| | - Chamikara Liyanage
- Cancer Single Cell Genomics LaboratoryTranslational Breast Cancer Program, Olivia Newton‐John Cancer Research InstituteHeidelbergVictoriaAustralia
| | - Jennifer Gunter
- Australian Prostate Cancer Research Centre‐Queensland, Centre for Genomics and Personalised Health, School of Biomedical ScienceQueensland University of Technology, Translational Research InstituteBrisbaneQueenslandAustralia
| | - Jyotsna Batra
- Australian Prostate Cancer Research Centre‐Queensland, Centre for Genomics and Personalised Health, School of Biomedical ScienceQueensland University of Technology, Translational Research InstituteBrisbaneQueenslandAustralia
| | - Ross Crawford
- Centre for Biomedical TechnologiesQueensland University of TechnologyBrisbaneAustralia
- Orthopaedic DepartmentThe Prince Charles HospitalBrisbaneAustralia
| | - Hongxun Sang
- Department of OrthopaedicsShenzhen Hospital of Southern Medical UniversityShenzhenChina
| | - Indira Prasadam
- Centre for Biomedical TechnologiesQueensland University of TechnologyBrisbaneAustralia
- School of Mechanical, Medical & Process EngineeringQueensland University of TechnologyBrisbaneAustralia
| |
Collapse
|
84
|
Abraham MT, Wilson J. Exosomes in Facial Plastic Surgery. Facial Plast Surg 2025; 41:274-276. [PMID: 39832778 DOI: 10.1055/a-2510-6807] [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: 01/22/2025] Open
Abstract
Exosomes have emerged as a promising therapeutic frontier in facial plastic surgery. Preclinical studies have demonstrated their ability to modulate wound healing, skin rejuvenation, hair growth, and nerve regeneration. Early clinical evidence suggests potential benefits in enhancing recovery after laser resurfacing, treating acne scars, and promoting hair growth. Despite their potential, there are currently no exosome products that are FDA-approved for medical use, and they should be considered experimental until receiving regulatory approval and robust clinical validation. As research advances, exosomes may offer valuable tools for facial plastic surgeons to improve patient outcomes and expand regenerative medicine applications in facial aesthetics and reconstruction.
Collapse
Affiliation(s)
- Manoj T Abraham
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John Wilson
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
85
|
Sama S, Cho S, Rehni AK, Jy W, Dave KR. Red cell microparticles produced using high-pressure extrusion enhance both primary and secondary hemostasis. Pharmacol Rep 2025; 77:508-516. [PMID: 39775702 PMCID: PMC11911262 DOI: 10.1007/s43440-024-00688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Current therapies to treat excessive bleeding are associated with significant complications, which may outweigh their benefits. Red blood cell-derived microparticles (RMPs) are a promising hemostatic agent. Previous studies demonstrated that they reduce bleeding in animal models, correct coagulation defects in patient blood, and have an excellent safety profile. However, their exact mechanism of action is not known. We investigated the potential role of RMPs on primary and secondary hemostasis. METHODS To evaluate the effects of RMPs, prepared using high-pressure extrusion, on primary hemostasis, we employed platelet aggregometry with platelet inhibitors, eptifibatide, and ticagrelor, with and without RMPs. To evaluate their effects on secondary hemostasis, we employed thromboelastography with plasma deficient in factors VII, VIII, IX, XI, and XII with and without RMPs. RESULTS We found that RMPs significantly increased collagen-induced platelet aggregation. However, there were no significant differences with and without RMP in the presence of the platelet inhibitors, indicating that RMPs may work through these receptors, either directly or indirectly. For secondary hemostasis, RMPs significantly decreased clotting times for plasma deficient in factors VII, VIII, IX, and XI but not in XII. CONCLUSIONS Our results indicate that RMPs enhance primary hemostasis and both pathways of secondary hemostasis.
Collapse
Affiliation(s)
- Snigdha Sama
- Department of Neurology, Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB #7046, Miami, FL, 33136, USA
| | - Sunjoo Cho
- Department of Neurology, Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB #7046, Miami, FL, 33136, USA
| | - Ashish K Rehni
- Department of Neurology, Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB #7046, Miami, FL, 33136, USA
| | - Wenche Jy
- The Wallace H. Coulter Platelet Laboratory, Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kunjan R Dave
- Department of Neurology, Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, 1600 NW 10th Ave RMSB #7046, Miami, FL, 33136, USA.
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
86
|
Belényesi SK, Patmore S, O'Driscoll L. Extracellular vesicles and the tumour microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189275. [PMID: 39900204 DOI: 10.1016/j.bbcan.2025.189275] [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: 06/11/2024] [Revised: 12/18/2024] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Extracellular vesicles (EVs), tiny packages of information released by cells, are well established as being involved in unwanted cell-to-cell communication in cancer. EVs from cancer cells have been associated with the spread of drug resistance, immune suppression, and metastasis. Additional to cancer cells, the tumour microenvironment (TME) involves many cell types -including immune cells, fibroblasts, and endothelial cells, each of which has a potential role in how tumours grow, spread, and respond (or otherwise) to therapy. This review collates and distils research developments regarding the role of EVs in multi-way communication between cells in the TME. Further research including tailored clinical studies are now warranted to determine how best to prevent this extensive adverse communication occurring and/or how best to exploit it for biomarker discovery and as a therapeutic approach, in the interest of patients and also for economic benefit.
Collapse
Affiliation(s)
- Szilárd-Krisztián Belényesi
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| | - Sean Patmore
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland.
| |
Collapse
|
87
|
Khereldin RM, Abouelela YS, Yasin NAE, Youssef FS, Abdelhameed MI, Tohamy AF, Rizk H, Daghash SM. Comparing the therapeutic influence of bone marrow Mesenchymal stem cells versus its derived exosomes against diabetic hepatopathy in rats. Exp Cell Res 2025; 447:114436. [PMID: 40057260 DOI: 10.1016/j.yexcr.2025.114436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 03/15/2025]
Abstract
Diabetes mellitus (DM) is a chronic widespread metabolic disorder, involving a high blood glucose level which causes multiple serious complications including liver, kidney, brain and peripheral nerves damage. Due to the undesirable side effects of the anti-diabetic drugs, the current studies directed to use stem cells and exosomes to overcome the limitations of traditional therapy. We aimed to compare the antidiabetic effect of Bone marrow mesenchymal stem cells (BMMSCs) and its derived exosomes against diabetic hepatopathy induced by streptozotocin (STZ) in albino rats. Our study was conducted on 28 male albino rats divided into 4 groups {control negative non diabetic group, control positive diabetic group, exosomes treated group received (5 × 109 particle/rat) through tail vein twice per week for one month} and Stem cell treated group received (107) BMMSCs through tail vein twice per week for one month. Hepatic structure together with blood glucose level, liver function enzymes were assayed in addition to a lipid profile tests, oxidative stress, and gene expression. Both treated groups by exosomes and stem cells expressed significantly low levels of fasting blood glucose, liver function parameters (ALT, AST, ALP), lipid profile tests (cholesterol and triglycerides), lipid peroxidation index (MDA), with substantial reduction in IL-1β expression compared to diabetic group. Significantly downregulating the VEGF and elevation of eNOS genes and GSH which suggest the effective role provided by BMMSCs and its derived exosomes for treatment of diabetic hepatopathy. Although, the results of both groups showed near average outcomes, the exosome treated group significantly enhanced liver function enzymes and triglyceride, cholesterol level compared to stem cells treated group. These findings were reinforced by the histopathological and immunohistochemistry examination. The latter showed slight but non-significant improvements in VEGF, eNOS, and IL-1β expression. These minor differences together with practical advantages of exosomes make it preferable over BMMSCs in treatment of diabetic hepatopathy.
Collapse
Affiliation(s)
- Rehab Mahmoud Khereldin
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Yara Sayed Abouelela
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Noha Ali Elsayed Yasin
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Fady Sayed Youssef
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Marwa Ibrahim Abdelhameed
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Adel Fathy Tohamy
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Hamdy Rizk
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| | - Samer Mohamed Daghash
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza square, 12211, Giza, Egypt.
| |
Collapse
|
88
|
Chen S, Bao Q, Xu W, Zhai X. Extracellular particles: emerging insights into central nervous system diseases. J Nanobiotechnology 2025; 23:263. [PMID: 40170148 PMCID: PMC11960037 DOI: 10.1186/s12951-025-03354-6] [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: 01/13/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Extracellular particles (EPs), including extracellular vesicles (EVs) and non-vesicular extracellular particles (NVEPs), are multimolecular biomaterials released by cells that play a crucial role in intercellular communication. Recently, new subtypes of EPs associated with central nervous system (CNS), such as exophers and supermeres have been identified. These EPs provide new perspectives for understanding the pathological progression of CNS disorders and confer potential diagnostic value for liquid biopsies in neurodegenerative diseases (NDs). Moreover, EPs have emerged as promising drug delivery vehicles and targeted platforms for CNS-specific therapies. In this review, we delineate the landscape of EP subtypes and their roles in the pathophysiology of CNS diseases. We also review the recent advances of EP-based diagnosis in NDs and highlight the importance of analytical platforms with single-particle resolution in the exploitation of potential biomarkers. Furthermore, we summarize the application of engineered EVs in the treatment of CNS diseases and outline the underexplored potential of NVEPs as novel therapeutic agents.
Collapse
Affiliation(s)
- Shenyuan Chen
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou, Jiangsu, 215600, China
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Qinghua Bao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou, Jiangsu, 215600, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Suzhou, Jiangsu, 215600, China.
- Zhenjiang Key Laboratory of High Technology Research on sEVs Foundation and Transformation Application, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| | - Xiao Zhai
- Department of Orthopedics, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
89
|
Mao X, Xu H, Liu X, Guan J, Shi J, Yang S. Proteomics of urinary exosomes for discovering novel non-invasive biomarkers of acute myocardial infarction patients. Int J Biol Macromol 2025; 302:140427. [PMID: 39890005 DOI: 10.1016/j.ijbiomac.2025.140427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/08/2025] [Accepted: 01/26/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Patients with acute myocardial infarction (AMI) can be identified by myocardial enzymes in peripheral blood, but no protein markers have been found in urinary exosomes derived from AMI patients. METHODS In the present study, a comprehensive proteomics analysis of urinary exosomes derived from patients with AMI was performed. Firstly, we employed the outstanding separation method known as EXODUS to isolate urinary exosomes from AMI patients and healthy controls. Then, we characterized urinary exosomes by nanoparticle tracking analysis (NTA), western blotting and transmission electron microscopy (TEM). After that, we identified the protein components of exosomes through label-free proteomics and conducted bioinformatics analysis. RESULT High-quality exosomes were obtained through separation using EXODUS, which could be demonstrated by NTA, Western blotting and TEM. NTA analysis showed that partilce amount in AMI patients was significantly higher than healthy controls. The equal-volume Western blotting experiment indicated that the expression level of classic exosomal markers Alix, heat shock protein90 (HSP90), CD63 and TSG101 (Tumor susceptibility gene101) in AMI patents was obviously stronger than healthy subjects. We first described the protein profiles of urinary exosomes in AMI patients through proteomics. In this study, We have identified 3194 proteins, among which a total of 30 differential proteins were detected between the urinary exosomes of AMI patients and healthy controls. We investigated F2 and OLR1 among identified exosomal proteins significantly elevated in AMI group, whereas F3 and APCS dysregulated in AMI development. CONCLUSIONS F2, F3, OLR1 and APCS are able to distinguish individuals between the AMI group and the healthy controls, and the protein panel represent a novel prospective non-invasive biomarkers for the diagnostic process of acute myocardial infarction.
Collapse
Affiliation(s)
- Xulong Mao
- The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, China.
| | - Hao Xu
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaoling Liu
- Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai 519087, China
| | - Jiale Guan
- Basic Medical College of Wenzhou Medical University, Zhejiang 325035, China
| | - Jiachong Shi
- Department of Cardiovascular Medicine, Qianjiang Central Hospital of Hubei Province, Qianjiang City 434000, Hubei, China
| | - Shaning Yang
- The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, China
| |
Collapse
|
90
|
Obi PO, Souza TFG, Özerkliğ B, Seif S, Bydak B, Klassen N, Duhamel TA, West AR, Gordon JW, Saleem A. Extracellular Vesicles Released From Skeletal Muscle Post-Chronic Contractile Activity Increase Mitochondrial Biogenesis in Recipient Myoblasts. J Extracell Vesicles 2025; 14:e70045. [PMID: 40205946 PMCID: PMC11982704 DOI: 10.1002/jev2.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/22/2025] [Indexed: 04/11/2025] Open
Abstract
The effect of chronic contractile activity (CCA) on the biophysical properties and functional activity of skeletal muscle extracellular vesicles (Skm-EVs) is poorly understood due to challenges in distinguishing Skm-EVs originating from exercising muscle in vivo. To address this, myoblasts were differentiated into myotubes, and electrically paced (3 h/day, 4 days @ 14 V). CCA evoked an increase in mitochondrial biogenesis in stimulated versus non-stimulated (CON) myotubes as expected. EVs were isolated from conditioned media (CM) from control and stimulated myotubes using differential ultracentrifugation (dUC) and characterised biophysically using tunable resistive pulse sensing (TRPS, Exoid), TEM and western blotting. TEM images confirmed isolated round-shaped vesicles of about 30-150 nm with an intact lipid bilayer. EVs ranged from 98 to 138 nm in diameter, and the mean size was not altered by CCA. Zeta potential and total EV protein yield remained unchanged between groups, and total EV secretion increased after 4 days of CCA. Concomitant analysis of EVs after each day of CCA also demonstrated a progressive increase in CCA-EV concentration, whilst size and zeta potential remained unaltered, and EV protein yield increased in both CON-EVs and CCA groups. CCA-EVs were enriched with small-EVs versus CON-EVs, concomitant with higher expression of small-EV markers CD81, Tsg101 and HSP70. In whole cell lysates, CD63 and ApoA1 were reduced with CCA in myotubes, whereas CD81, Tsg101, Flotillin-1 and HSP70 levels remained unchanged. To evaluate the functional effect of EVs secreted post-CCA, we treated C2C12 myoblasts with all EVs isolated from CON or CCA myotubes after each day of stimulation, and measured cell count, cell viability, protein yield and mitochondrial biogenesis in recipient cells. There was no effect on cell count, viability and protein yield. Myoblasts treated with CCA-EVs exhibited increased mitochondrial biogenesis as indicated by enhanced MitoTracker Red staining, cytochrome c oxidase (COX) activity and protein expression of electron transport chain subunit, CIV-MTCO1. Further, CCA-EV treatment enhanced maximal oxygen consumption rates (OCR) in a dose-dependent manner, and ATP production in treated myoblasts. This increase in maximal OCR was abrogated when CCA-EVs pre-treated with proteinase K were co-cultured with myoblasts, indicating the pro-metabolic effect was likely mediated by transmembrane or peripheral membrane proteins in CCA-EVs. Our data highlight the novel effect of Skm-EVs isolated post-CCA in mediating pro-metabolic effects in recipient cells and thereby transmitting the effects associated with traditional exercise. Further investigation to interrogate the underlying mechanisms involved in downstream cellular metabolic adaptations is warranted.
Collapse
Affiliation(s)
- Patience O. Obi
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Applied Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
| | - Tamiris F. G. Souza
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
| | - Berkay Özerkliğ
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
- Department of Exercise and Sport Sciences, Faculty of Sport SciencesHacettepe UniversityAnkaraTurkey
| | - Samira Seif
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
| | - Benjamin Bydak
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
| | - Nicholas Klassen
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
| | - Todd A. Duhamel
- Applied Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
- Institute of Cardiovascular SciencesSt. Boniface General Hospital Albrechtsen Research CentreWinnipegManitobaCanada
| | - Adrian R. West
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Department of Physiology and Pathophysiology, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Joseph W. Gordon
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Department of Pediatrics and Child Health, Rady Faculty of Health SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Ayesha Saleem
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research ThemeWinnipegManitobaCanada
- Biology of Breathing Research ThemeWinnipegManitobaCanada
- Applied Health SciencesUniversity of ManitobaWinnipegManitobaCanada
- Children's Hospital Research Institute of Manitoba (CHRIM)WinnipegManitobaCanada
- Faculty of Kinesiology and Recreation ManagementUniversity of ManitobaWinnipegManitobaCanada
| |
Collapse
|
91
|
Bhavsar V, Sahu A, Taware R. Stress-induced extracellular vesicles: insight into their altered proteomic composition and probable physiological role in cancer. Mol Cell Biochem 2025; 480:2025-2041. [PMID: 39302488 DOI: 10.1007/s11010-024-05121-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: 04/17/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
EVs (extracellular vesicles) are phospholipid bilayer vesicles that can be released by both prokaryotic and eukaryotic cells in normal as well as altered physiological conditions. These vesicles also termed as signalosomes, possess a distinctive cargo comprising nucleic acids, proteins, lipids, and metabolites, enabling them to play a pivotal role in both local and long-distance intercellular communication. The composition, origin, and release of EVs can be influenced by different physiological conditions and a variety of stress factors, consequently affecting the contents carried within these vesicles. Therefore, identifying the modified contents of EVs can provide valuable insights into their functional role in stress-triggered communication. Particularly, this is important when EVs released from tumor microenvironment are investigated for their role in the development and dissemination of cancer. This review article emphasizes the importance of differential EV shedding and altered proteomic content in response to reduced oxygen concentration, altered levels of glucose and glutamine, pH variations, oxidative stress and Ca2+ ion concertation and it is subsequent effects on the behavior of recipient cells.
Collapse
Affiliation(s)
- Vaidehi Bhavsar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Ashish Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Ravindra Taware
- Department of Natural Products, National Institute of Pharmaceutical Education and Research-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
92
|
Boselli D, Clemente F, Di Terlizzi S, Pagiatakis C, Papa L, Del Zotto G, Villa C, Ramirez GA, Maugeri N, Manfredi AA, Anselmo A. Unravelling Plasma Extracellular Vesicle Diversity With Optimised Spectral Flow Cytometry. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70045. [PMID: 40292386 PMCID: PMC12025886 DOI: 10.1002/jex2.70045] [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: 09/27/2024] [Revised: 02/27/2025] [Accepted: 03/13/2025] [Indexed: 04/30/2025]
Abstract
Extracellular vesicles (EVs) are crucial for intercellular communication and are found in various biological fluids. The identification and immunophenotyping of such small particles continue to pose significant challenges. Here, we have developed a workflow for the optimisation of a next-generation panel for in-depth immunophenotyping of circulating plasma EVs using spectral flow cytometry. Our data collection followed a multistep optimisation phase for both instrument setup and 21-colour panel design, thus maximising fluorescent signal recovery. This spectral approach enabled the identification of novel EV subpopulations. Indeed, besides common EVs released by erythrocytes, platelets, leukocytes and endothelial cells, we observed rare and poorly known EV subsets carrying antigens related to cell activation or exhaustion. Notably, the unsupervised data analysis of major EV subsets revealed subpopulations expressing up to five surface antigens simultaneously. However, the majority of EVs expressed only a single surface antigen, suggesting they may not fully represent the phenotype of their parent cells. This is likely due to the small surface area or the biogenesis of EVs rather than antibody steric hindrance. Finally, we tested our workflow by analysing the plasma EV landscape in a cohort of systemic lupus erythematosus (SLE) patients. Interestingly, we observed a significant increase in CD54+ EVs, supporting the notion of elevated circulating ICAM under SLE conditions. To our knowledge, these are the first data highlighting the importance of a spectral flow cytometry approach in deciphering the heterogeneity of plasma EVs paving the way for the routine use of a high-dimensional immunophenotyping in EV research.
Collapse
Affiliation(s)
- Daniela Boselli
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
| | - Francesca Clemente
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
| | - Simona Di Terlizzi
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
| | - Christina Pagiatakis
- Department of Cardiovascular MedicineIRCCS Humanitas Research HospitalRozzanoMilanItaly
- Department of Biotechnology and Life SciencesUniversity of InsubriaVareseItaly
| | - Laura Papa
- Department of Cardiovascular MedicineIRCCS Humanitas Research HospitalRozzanoMilanItaly
| | - Genny Del Zotto
- Department of Research and DiagnosticsIRCCS Istituto Giannina GasliniGenoaItaly
| | - Chiara Villa
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
- Università Vita‐Salute San RaffaeleMilanItaly
| | - Giuseppe Alvise Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare DiseasesIRCCS Ospedale San RaffaeleMilanItaly
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS Ospedale San RaffaeleMilanItaly
| | - Norma Maugeri
- Università Vita‐Salute San RaffaeleMilanItaly
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS Ospedale San RaffaeleMilanItaly
| | - Angelo A. Manfredi
- Università Vita‐Salute San RaffaeleMilanItaly
- Unit of Immunology, Rheumatology, Allergy and Rare DiseasesIRCCS Ospedale San RaffaeleMilanItaly
- Division of Immunology, Transplantation and Infectious DiseasesIRCCS Ospedale San RaffaeleMilanItaly
| | - Achille Anselmo
- Experimental Imaging Center, FRACTAL, Flow cytometry Resource, Advanced Cytometry Technical Applications LaboratoryIRCCS Ospedale San RaffaeleMilanItaly
- Università Vita‐Salute San RaffaeleMilanItaly
| |
Collapse
|
93
|
Gao K, Xi W, Ni J, Jiang J, Lei Y, Li L, Chu J, Li R, An Y, Ouyang Y, Su R, Zhang R, Wu G. Genetically modified extracellular vesicles loaded with activated gasdermin D potentially inhibit prostate-specific membrane antigen-positive prostate carcinoma growth and enhance immunotherapy. Biomaterials 2025; 315:122894. [PMID: 39461061 DOI: 10.1016/j.biomaterials.2024.122894] [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: 07/07/2024] [Revised: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
Prostate cancer (PCa) is associated with poor immunogenicity and lymphocytic infiltration, and immunotherapy effective against PCa remains unavailable. Pyroptosis, a novel immunotherapeutic modality for cancer, promotes systemic immune responses leading to immunogenic cell death in solid tumors. This paper describes the preparation and analysis of PSMAscFv-EVN-GSDMD; this genetically engineered recombinant extracellular vesicle (EV) expresses a single-chain variable antibody fragment (scFv) with high affinity for prostate-specific membrane antigen (PSMA) on their surfaces and is loaded with the N-terminal domain of gasdermin D (GSDMD). Both in vitro and in vivo, PSMAscFv-EVN-GSDMD effectively targeted PSMA-positive PCa cells and induced pyroptosis through the carrier properties of EVs and the specificity of PSMAscFv. In the 22RV1 and PSMA-transfected RM-1-inoculated PCa mouse models, PSMAscFv-EVN-GSDMD efficiently inhibited tumor growth and promoted tumor immune responses. In conclusion, PSMAscFv-EVN-GSDMD can convert the immunosuppressive "cold" tumor microenvironment of PCa into an immunogenic "hot" tumor microenvironment.
Collapse
Affiliation(s)
- Ke Gao
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China; State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Wenjin Xi
- State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Jianxin Ni
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Jun Jiang
- Department of Health Service, Base of Health Service, Air Force Medical University, Xi'an, China
| | - Yonghua Lei
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Lin Li
- Department of Immunology, School of Basic Medicine, Yan'an University. Yan'an, 716099, China
| | - Jie Chu
- State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Ruixiao Li
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Yongpan An
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Yanan Ouyang
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Ruiping Su
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.
| | - Guojun Wu
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China.
| |
Collapse
|
94
|
Sandira MI, Lim K, Yoshida T, Sajidah ES, Narimatsu S, Imakawa R, Yoshimura K, Nishide G, Qiu Y, Taoka A, Hazawa M, Ando T, Hanayama R, Wong RW. Nanoscopic Profiling of Small Extracellular Vesicles via High-Speed Atomic Force Microscopy (HS-AFM) Videography. J Extracell Vesicles 2025; 14:e270050. [PMID: 40139685 PMCID: PMC11943829 DOI: 10.1002/jev2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 01/21/2025] [Accepted: 01/31/2025] [Indexed: 03/29/2025] Open
Abstract
Small extracellular vesicles (sEVs), which carry lipids, proteins and RNAs from their parent cells, serve as biomarkers for specific cell types and biological states. These vesicles, including exosomes and microvesicles, facilitate intercellular communication by transferring cellular components between cells. Current methods, such as ultracentrifugation and Tim-4 affinity method, yield high-purity sEVs. However, despite their small size, purified sEVs remain heterogeneous due to their varied intracellular origins. In this technical note, we used high-speed atomic force microscopy (HS-AFM) in conjunction with exosome markers (IgGCD63 and IgGCD81) to explore the intracellular origins of sEVs at single-sEV resolution. Our results first revealed the nanotopology of HEK293T-derived sEVs under physiological conditions. Larger sEVs (diameter > 100 nm) exhibited greater height fluctuations compared to smaller sEVs (diameter ≤ 100 nm). Next, we found that mouse-origin IgGCD63, and rabbit-origin IgGcontrol and IgGCD81, exhibited the iconic 'Y' conformation, and similar structural dynamics properties. Last, exosome marker antibodies predominantly co-localised with sEVd ≤ 100 nm but not with sEVd > 100 nm, demonstrating the CD63-CD81-enriched sEV and CD63-CD81-depleted sEV subpopulations. In summary, we demonstrate that nanoscopic profiling of surface exosome markers on sEVs using HS-AFM is feasible for characterising distinct sEV subpopulations in a heterogeneous sEV mixture.
Collapse
Affiliation(s)
- Muhammad Isman Sandira
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Keesiang Lim
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Takeshi Yoshida
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Department of ImmunologyGraduate School of Medical SciencesKanazawa UniversityKanazawaIshikawaJapan
| | | | - Shinnosuke Narimatsu
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Reon Imakawa
- The School of Biological Science and TechnologyCollege of Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Kota Yoshimura
- The School of Biological Science and TechnologyCollege of Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Goro Nishide
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeWISE Program for Nano‐Precision Medicine, Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Yujia Qiu
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Azuma Taoka
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Masaharu Hazawa
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Toshio Ando
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Rikinari Hanayama
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Department of ImmunologyGraduate School of Medical SciencesKanazawa UniversityKanazawaIshikawaJapan
| | - Richard W. Wong
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeWISE Program for Nano‐Precision Medicine, Science and TechnologyKanazawa UniversityKanazawaIshikawaJapan
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
95
|
Alves Â, Ferreira M, Eiras M, Lima L, Medeiros R, Teixeira AL, Dias F. Exosome-derived hsa-miR-200c-3p, hsa-miR-25-3p and hsa-miR-301a-3p as potential biomarkers and therapeutic targets for restoration of PTEN expression in clear cell renal cell carcinoma. Int J Biol Macromol 2025; 302:140607. [PMID: 39900161 DOI: 10.1016/j.ijbiomac.2025.140607] [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: 01/07/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is an aggressive kidney cancer subtype with limited biomarkers and therapeutic options. Thus, the present study aimed to evaluate the biomarker and therapeutic potential of Phosphatase and Tensin Homologue (PTEN)-regulating microRNAs (miRNAs) in 2D and 3D ccRCC models. Extracellular vesicles (EVs) from four renal cell lines were characterized, and selected miRNAs (hsa-miR-200c-3p, hsa-miR-25-3p, and hsa-miR-301a-3p) were quantified in cells and EVs. PTEN mRNA levels were measured intracellularly. 786-O cells were transfected with miRNA inhibitors in both models and effects on miRNA and PTEN expression were assessed alongside phenotypic alterations. The expression of target miRNAs increased with ccRCC cell aggressiveness, both intracellularly and in EVs, while PTEN mRNA expression decreased. Combined inhibition of these miRNAs significantly increased PTEN expression, reducing tumor cell proliferation and migration in 2D models and decreasing spheroid size and metabolic capacity in 3D models. These miRNAs show potential as biomarkers for monitoring disease aggressiveness and as therapeutic targets in ccRCC, potentially leading to more effective and personalized treatment approaches for ccRCC patients.
Collapse
Affiliation(s)
- Ângela Alves
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Mariana Ferreira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Mariana Eiras
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Experimental Pathology and Therapeutics Group, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Luís Lima
- Experimental Pathology and Therapeutics Group, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Faculty of Medicine (FMUP), University of Porto, 4200-319 Porto, Portugal; Laboratory Medicine, Clinical Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; Biomedicine Research Center (CEBIMED), Research Innovation and Development Institute (FP-I3ID), Faculty of Health Sciences, Fernando Pessoa University (UFP), 4249-004 Porto, Portugal; Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| |
Collapse
|
96
|
Lyu X, Yamano T, Nagamori K, Imai S, Van Le T, Bolidong D, Ueda M, Warashina S, Mukai H, Hayashi S, Matoba K, Nishino T, Hanayama R. Direct delivery of immune modulators to tumour-infiltrating lymphocytes using engineered extracellular vesicles. J Extracell Vesicles 2025; 14:e70035. [PMID: 40154979 PMCID: PMC11952836 DOI: 10.1002/jev2.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/18/2024] [Accepted: 12/17/2024] [Indexed: 04/01/2025] Open
Abstract
Extracellular vesicles (EVs) are important mediators of cell-cell communication, including immune regulation. Despite the recent development of several EV-based cancer immunotherapies, their clinical efficacy remains limited. Here, we created antigen-presenting EVs to express peptide-major histocompatibility complex (pMHC) class I, costimulatory molecule and IL-2. This enabled the selective delivery of multiple immune modulators to antigen-specific CD8+ T cells, promoting their expansion in vivo without severe adverse effects. Notably, antigen-presenting EVs accumulated in the tumour microenvironment, increasing IFN-γ+ CD8+ T cell and decreasing exhausted CD8+ T cell numbers, suggesting that antigen-presenting EVs transformed the 'cold' tumour microenvironment into a 'hot' one. Combination therapy with antigen-presenting EVs and anti-PD-1 demonstrated enhanced anticancer immunity against established tumours. We successfully engineered humanized antigen-presenting EVs, which selectively stimulated tumour antigen-specific CD8+ T cells. In conclusion, engineering EVs to co-express multiple immunomodulators represents a promising method for cancer immunotherapy.
Collapse
Affiliation(s)
- Xiabing Lyu
- WPI Nano Life Science Institute (NanoLSI)Kanazawa UniversityKanazawaJapan
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| | - Tomoyoshi Yamano
- WPI Nano Life Science Institute (NanoLSI)Kanazawa UniversityKanazawaJapan
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| | - Kanto Nagamori
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| | - Shota Imai
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| | - Toan Van Le
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| | - Dilireba Bolidong
- WPI Nano Life Science Institute (NanoLSI)Kanazawa UniversityKanazawaJapan
| | - Makie Ueda
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| | - Shota Warashina
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
- Department of Pharmaceutical Informatics, Graduate School of Biomedical SciencesNagasaki UniversityNagasakiJapan
| | - Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging TechnologyRIKEN Center for Biosystems Dynamics ResearchKobeJapan
- Department of Pharmaceutical Informatics, Graduate School of Biomedical SciencesNagasaki UniversityNagasakiJapan
| | - Seigo Hayashi
- Biological Research LaboratoriesNissan Chemical CorporationSaitamaJapan
| | - Kazutaka Matoba
- Biological Research LaboratoriesNissan Chemical CorporationSaitamaJapan
| | - Taito Nishino
- Biological Research LaboratoriesNissan Chemical CorporationSaitamaJapan
| | - Rikinari Hanayama
- WPI Nano Life Science Institute (NanoLSI)Kanazawa UniversityKanazawaJapan
- Department of Immunology, Graduate School of MedicineKanazawa UniversityKanazawaJapan
| |
Collapse
|
97
|
Feng X, Shen A, Zhang W, Jia S, Iliuk A, Wang Y, Zhang W, Zhang Y, Tao WA, Hu L. High-throughput capture and in situ protein analysis of extracellular vesicles by chemical probe-based array. Nat Protoc 2025; 20:1057-1081. [PMID: 39438698 DOI: 10.1038/s41596-024-01082-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are small particles with phospholipid bilayers that carry a diverse range of cargoes including nucleic acids, proteins and metabolites. EVs have important roles in various cellular processes and are increasingly recognized for their ubiquitous role in cell-cell communications and potential applications in therapeutics and diagnostics. Although many methods have been developed for the characterization and measurement of EVs, analyzing them from biofluids remains a challenge with regard to throughput and sensitivity. Recently, we introduced an approach to facilitate high-throughput analysis of EVs from trace amounts of sample. In this method, an amphiphile-dendrimer supramolecular probe (ADSP) is coated onto a nitrocellulose membrane for array-based capture and to enable an in situ immunoblotting assay. Here, we describe the protocol for our array-based method of EV profiling. We describe an enhanced version of the method that incorporates an automated printing workstation, ensuring high throughput and reproducibility. We further demonstrate the use of our array to profile specific glycosylations on the EV surface using click chemistry of an azide group introduced by metabolic labeling. In this protocol, the synthesis of ADSP and the fabrication of ADSP nitrocellulose membrane array can be completed on the same day. EVs are efficiently captured from biological or clinical samples through a 30-min incubation, followed by an immunoblotting assay within a 3-h window, thus providing a high-throughput platform for EV isolation and in situ targeted analysis of EV proteins and their modifications.
Collapse
Affiliation(s)
- Xin Feng
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, China
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Ao Shen
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, China
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Shengnan Jia
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, China
| | - Anton Iliuk
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Yuling Wang
- School of Natural Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales, Australia
| | - Wenke Zhang
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, China
| | - Ying Zhang
- Department of Chemistry and NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai, China.
| | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, China.
| |
Collapse
|
98
|
Huang Z, Li J, Wo J, Li C, Wu Z, Deng X, Liang Y, Li F, Chen B, Jia B, Wang L, Wang Y, Sun G, Li Z, Zhu H, Guest JD, So K, Fu Q, Zhou L. Intranasal Delivery of Brain-Derived Neurotrophic Factor (BDNF)-Loaded Small Extracellular Vesicles for Treating Acute Spinal Cord Injury in Rats and Monkeys. J Extracell Vesicles 2025; 14:e70066. [PMID: 40194993 PMCID: PMC11975507 DOI: 10.1002/jev2.70066] [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: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Besides surgical decompression, neuroprotection and neuroinflammation reduction are critical for acute spinal cord injury (SCI). In this study, we prepared small extracellular vesicles (sEVs) from immortalised mesenchymal stem cells overexpressing brain-derived neurotrophic factor (BDNF) and evaluated whether intranasal administration of BDNF-sEVs is a therapeutic option for acute SCI. In cultured neurons, BDNF loading enhanced neurite outgrowth promoted by sEVs. After intranasal administration, mCherry-labelled sEVs were transported to the injured spinal cords of rats and monkeys and mainly taken up by neurons. In acute SCI rats, intranasal administration of sEVs and BDNF-sEVs reduced glial responses and proinflammatory cytokine production, enhanced neuronal survival and angiogenesis in the lesion, promoted injured axon rewiring, delayed lumbar spinal motoneuron atrophy below the lesion, and improved functional performance. The rats receiving BDNF-sEV treatment showed improved neural repair and functional recovery compared to those with sEV treatment. Intranasal administration of BDNF-sEVs, but not of sEVs, increased BDNF levels and phosphorylation of downstream signals in the rat-injured spinal cord samples, indicating activation of the BDNF/TrkB signalling pathway. In acute SCI monkeys, intranasal administration of BDNF-sEVs was further confirmed to inhibit glial reactivities and proinflammatory cytokine release, increasing BDNF levels in the cerebrospinal fluid, enhancing neural network rewiring of injured spinal cords and neuronal activities of the brain, and improving functional performances in behavioural tests and electrophysiological recordings. In conclusion, BDNF-sEVs play a combinatory therapeutic role of sEVs and BDNF, and intranasal administration of BDNF-sEVs is a potential option for the clinical treatment of acute SCI.
Collapse
Affiliation(s)
- Zhonghai Huang
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Jing Li
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of Human Anatomy, The College of Basic Medical SciencesJinan UniversityGuangzhouChina
| | - Jin Wo
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Cheng‐Lin Li
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zi‐Cong Wu
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xiao‐Hui Deng
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yaying Liang
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Fuxiang Li
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Boli Chen
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Bin Jia
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Ying Wang
- Medical Imaging CenterThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Zhizhong Li
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
| | - Hui Zhu
- Kunming International Spine and Spinal Cord Injury Treatment CenterKunming Tongren HospitalKunmingChina
| | - James D Guest
- Neurological Surgery and the Miami Project to Cure ParalysisMiller School of MedicineMiamiFloridaUSA
| | - Kwok‐Fai So
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of Human Anatomy, The College of Basic Medical SciencesJinan UniversityGuangzhouChina
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdaoShandongChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
- Center for Exercise and Brain ScienceSchool of PsychologyShanghai University of SportShanghaiChina
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital of The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Libing Zhou
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord ReconstructionThe Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital)Jinan UniversityHeyuanChina
- Guangdong‐Hongkong‐Macau Institute of CNS RegenerationKey Laboratory of CNS Regeneration (Jinan University)‐Ministry of EducationGuangdong Key Laboratory of Non‐Human Primate ResearchJinan UniversityGuangzhouChina
- Department of OrthopedicsThe First Affiliated HospitalJinan UniversityGuangzhouChina
- Neuroscience and Neurorehabilitation InstituteUniversity of Health and Rehabilitation SciencesQingdaoShandongChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityJiangsuChina
- Center for Exercise and Brain ScienceSchool of PsychologyShanghai University of SportShanghaiChina
| |
Collapse
|
99
|
Murphy CA, O'Reilly D, Weiss L, Madden S, Macleod H, Chevillier AL, Neary E, O'Loughlin J, EL‐Khuffash A, Kevane B, NíAinle F, Zivny J, McCallion N, Maguire PB. Unique Patterns of Circulating Extracellular Vesicles in Preterm Infants During Adaptation to Extra-Uterine Life. J Extracell Vesicles 2025; 14:e70064. [PMID: 40194994 PMCID: PMC11975508 DOI: 10.1002/jev2.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
There is growing interest in the role of extracellular vesicles (EVs) in neonatal pathology. This study aimed to characterise circulating EVs following preterm birth. This single-centre prospective observational study included cord and postnatal plasma from preterm (n = 101) and full-term infants (n = 66). EVs were analysed using nanoparticle tracking analysis, flow cytometry, proteomics and procoagulant activity assay. We found changes in the concentration, size, cellular origin and proteomic content of circulating EVs in preterm infants during perinatal adaptation. To understand if these changes were related to prematurity or normal adaptation to extrauterine life, they were also investigated in term infants. There was a dramatic increase in the concentration of small and large EVs on Day 3 in the preterm group; specific subsets of platelet (CD42b+ and CD62P+), endothelial (VEGFR2) and tissue factor EVs were elevated. Differentially expressed proteins relating to haemostasis, pulmonary physiology and immunity were identified between Day 1 and 3 in preterm infants. These changes have never previously been described in a large cohort of preterm infants and differ from healthy term infants. These findings have major implications for future neonatal EV studies, particularly the timing of sample collection. Further work is required to understand the clinical implications of this unique EV profile following preterm birth.
Collapse
Affiliation(s)
- Claire A. Murphy
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Daniel O'Reilly
- Department of NeonatologyRotunda HospitalDublinIreland
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Luisa Weiss
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Stephen Madden
- School of Population HealthRoyal College of Surgeons in IrelandDublinIreland
| | - Hayley Macleod
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Ana Le Chevillier
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Elaine Neary
- Department of NeonatologyLiverpool Women's HospitalLiverpoolUK
- Department of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - John O'Loughlin
- Department of Laboratory MedicineRotunda HospitalDublinIreland
| | - Afif EL‐Khuffash
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Barry Kevane
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
| | - Fionnuala NíAinle
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
- Department of HaematologyRotunda HospitalDublinIreland
| | - Jan Zivny
- Institute of Pathological PhysiologyFirst Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Naomi McCallion
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Patricia B. Maguire
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| |
Collapse
|
100
|
Zhang Y, Song J, Wang B, Wen Y, Jiang W, Zhang YL, Li ZL, Yu H, Qin SF, Lv LL, Tang TT, Liu BC. Comprehensive Comparison of Extracellular Vesicles Derived from Mesenchymal Stem Cells Cultured with Fetal Bovine Serum and Human Platelet Lysate. ACS NANO 2025; 19:12366-12381. [PMID: 40110859 DOI: 10.1021/acsnano.5c02532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as a promising approach in regenerative therapy. However, the clinical application of MSC-EVs is hindered by the presence of xenogenic components, such as fetal bovine serum (FBS), which is the most used culture supplement for MSCs. Human platelet lysate (HPL) has been proposed as an alternative to FBS, but whether MSC-EVs derived from HPL-cultured MSCs are suitable for clinical translation remains unclear. In this study, we comprehensively compared the characterization of EVs derived from MSCs cultured in the medium with FBS (F-EVs) and HPL (H-EVs). Our study showed that HPL promoted MSC-EV production without compromising EVs critical quality attributes. Multiomics sequencing revealed the stability of H-EVs from different umbilical cord donors and global functional alterations for MSC-EVs under different culture conditions. In comparison to F-EVs, H-EVs enriched more angiogenesis-related molecules and exhibited enhanced angiogenesis, which were further confirmed by in vivo and in vitro studies. H-EVs significantly reduced renal microvascular rarefaction and promoted the regeneration of umbilical vein endothelial cells to hypoxia stimulation compared to that of F-EVs. In conclusion, our findings demonstrated that HPL as culture supplements did not alter the critical quality attributes of MSC-EVs, specifically holding a higher yield and quality of MSC-EVs with enhanced angiogenic potential.
Collapse
Affiliation(s)
- Yue Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Jing Song
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Yi Wen
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Wei Jiang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Yi-Lin Zhang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Zuo-Lin Li
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Hong Yu
- Department of Obstetrics and Gynecology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Suo-Fu Qin
- Shenzhen Kexing Pharmaceutical Co., Ltd., Shenzhen 518057, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing 210009, China
| |
Collapse
|