1
|
Castagnino PA, Haas DA, Musante L, Tancler NA, Tran BV, Kean R, Steck AR, Martinez LA, Mostaghel EA, Hooper DC, Kim FJ. Sigma1 inhibitor suppression of adaptive immune resistance mechanisms mediated by cancer cell derived extracellular vesicles. Cancer Biol Ther 2025; 26:2455722. [PMID: 39863992 PMCID: PMC11776462 DOI: 10.1080/15384047.2025.2455722] [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: 08/07/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Adaptive immune resistance in cancer describes the various mechanisms by which tumors adapt to evade anti-tumor immune responses. IFN-γ induction of programmed death-ligand 1 (PD-L1) was the first defined and validated adaptive immune resistance mechanism. The endoplasmic reticulum (ER) is central to adaptive immune resistance as immune modulatory secreted and integral membrane proteins are dependent on ER. Sigma1 is a unique ligand-regulated integral membrane scaffolding protein enriched in the ER of cancer cells. PD-L1 is an integral membrane glycoprotein that is translated into the ER and processed through the cellular secretory pathway. At the cell surface, PD-L1 is an immune checkpoint molecule that binds PD-1 on activated T-cells and blocks anti-tumor immunity. PD-L1 can also be incorporated into cancer cell-derived extracellular vesicles (EVs), and EV-associated PD-L1 can inactivate T-cells within the tumor microenvironment. Here, we demonstrate that a selective small molecule inhibitor of Sigma1 can block IFN-γ mediated adaptive immune resistance in part by altering the incorporation of PD-L1 into cancer cell-derived EVs. Sigma1 inhibition blocked post-translational maturation of PD-L1 downstream of IFN-γ/STAT1 signaling. Subsequently, EVs released in response to IFN-γ stimulation were significantly less potent suppressors of T-cell activation. These results suggest that by reducing tumor derived immune suppressive EVs, Sigma1 inhibition may promote antitumor immunity. Sigma1 modulation presents a novel approach to regulating the tumor immune microenvironment by altering the content and production of EVs. Altogether, these data support the notion that Sigma1 may play a role in adaptive immune resistance in the tumor microenvironment.
Collapse
Affiliation(s)
- Paola A. Castagnino
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Derick A. Haas
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Luca Musante
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Nathalia A. Tancler
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Bach V. Tran
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Alexandra R. Steck
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Luis A. Martinez
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Elahe A. Mostaghel
- Geriatric Research, Education and Clinical Center, U.S. Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - D. Craig Hooper
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Felix J. Kim
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| |
Collapse
|
2
|
Powell JS, Larregina AT, Shufesky WJ, Sullivan ML, Stolz DB, Gould SJ, Camirand G, Catz SD, Watkins SC, Sadovsky Y, Morelli AE. Fetoplacental extracellular vesicles deliver conceptus-derived antigens to maternal secondary lymphoid tissues for immune recognition. JCI Insight 2025; 10:e186335. [PMID: 40401522 DOI: 10.1172/jci.insight.186335] [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: 09/06/2024] [Accepted: 04/09/2025] [Indexed: 05/23/2025] Open
Abstract
Pregnancy is an immunological paradox where despite a competent maternal immune system, regulatory mechanisms at the fetoplacental interface and maternal secondary lymphoid tissues (SLTs) circumvent rejection of semi-allogeneic concepti. Small extracellular vesicles (sEVs) are a vehicle for intercellular communication; nevertheless, the role of fetoplacental sEVs in transport of antigens to maternal SLTs has not been conclusively demonstrated. Using mice in which the conceptus generates fluoroprobe-tagged sEVs shed by the plasma membrane or released from the endocytic compartment, we show that fetoplacental sEVs are delivered to immune cells in the maternal spleen. Injection of sEVs from placentas of females impregnated with Act-mOVA B6 males elicited suboptimal activation of OVA-specific CD8+ OT-I T cells in virgin females as occurs during pregnancy. Furthermore, when OVA+ concepti were deficient in Rab27a, a protein required for sEV secretion, OT-I cell proliferation in the maternal spleen was decreased. Proteomics analysis revealed that mouse trophoblast sEVs were enriched in antiinflammatory and immunosuppressive mediators. Translational relevance was tested in humanized mice injected using sEVs from cultures of human trophoblasts. Our findings show that sEVs deliver fetoplacental antigens to the mother's SLTs that are recognized by maternal T cells. Alterations of such a mechanism may lead to pregnancy disorders.
Collapse
Affiliation(s)
- Juliana S Powell
- T.E. Starzl Transplantation Institute, Department of Surgery
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute
| | - Adriana T Larregina
- Department of Dermatology
- Department of Immunology
- McGowan Institute for Regenerative Medicine; and
| | | | - Mara Lg Sullivan
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donna Beer Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephen J Gould
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Geoffrey Camirand
- T.E. Starzl Transplantation Institute, Department of Surgery
- Department of Immunology
| | - Sergio D Catz
- The Scripps Research Institute, La Jolla, California, USA
| | - Simon C Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee-Womens Research Institute
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adrian E Morelli
- T.E. Starzl Transplantation Institute, Department of Surgery
- Department of Immunology
| |
Collapse
|
3
|
Czpakowska J, Głąbiński A, Szpakowski P. The Potential Role of Exosomes in Communication Between Astrocytes and Endothelial Cells. Int J Mol Sci 2025; 26:4676. [PMID: 40429819 PMCID: PMC12111803 DOI: 10.3390/ijms26104676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Exosomes are extracellular vesicles secreted by almost all types of cells. Their release allows for the transport of specific regulatory cargo into the recipient cells and the modulation of their activity. Vesicular communication has also been identified as an important mechanism for the regulation of numerous cellular activities in the brain tissue, contributing to proper neuronal functions and brain homeostasis. In this work, we focus on the role of exosomes and extracellular vesicles in the communication between astrocytes and brain endothelial cells, two major components of the blood-brain barrier. We perform a comprehensive review of the latest studies highlighting the role of exosomes in astrocyte-endothelial cell crosstalk within the blood-brain barrier. We have also described the role of particular exosomal miRNAs in the regulation of astrocytes and brain endothelial cell functions, and discuss some future implications.
Collapse
Affiliation(s)
| | - Andrzej Głąbiński
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland;
| | - Piotr Szpakowski
- Department of Neurology and Stroke, Medical University of Lodz, Zeromskiego 113 Street, 90-549 Lodz, Poland;
| |
Collapse
|
4
|
Liu J, Wang Z, Liang W, Zhang Z, Deng Y, Chen X, Hou Z, Xie Y, Wang Q, Li Y, Bai C, Li D, Mo F, Wang H, Wang D, Yuan J, Wang Y, Teng ZQ, Hu B. Microglial TMEM119 binds to amyloid-β to promote its clearance in an Aβ-depositing mouse model of Alzheimer's disease. Immunity 2025:S1074-7613(25)00181-5. [PMID: 40373772 DOI: 10.1016/j.immuni.2025.04.018] [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: 07/11/2024] [Revised: 01/19/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
The progression of Alzheimer's disease (AD) involves temporal dynamics of microglial activation. Restoring or maintaining microglial homeostasis has emerged as a promising therapeutic strategy to combat AD. Transmembrane protein 119 (TMEM119) is a homeostatic marker of microglia but has not been fully studied under AD pathological conditions. Here, we observed that amyloid-beta (Aβ) induced a decrease in TMEM119 expression in microglia, and TMEM119 deficiency increased AD progression in the 5×FAD mouse model. TMEM119 bound to Aβ oligomers and recruited low-density lipoprotein receptor 1, which in turn degraded TMEM119 itself. Overexpression of TMEM119 in microglia enhanced their phagocytic activity and alleviated cognitive deficits in 5xFAD mice. Administration of the small molecules Kartogenin and SRI-011381, which we found enhanced TMEM119 expression, substantially promoted Aβ clearance and improved cognitive function in AD mice, even during the mid-stage of the disease. These findings identify TMEM119 as a promising therapeutic target for AD.
Collapse
Affiliation(s)
- Jing Liu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhimeng Wang
- School of Pharmaceutical Sciences, Tsinghua University, Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Wenwen Liang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Zhenhao Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yusen Deng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaowei Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zongren Hou
- Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanzhi Xie
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Qi Wang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Yuan Li
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Chaobo Bai
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China
| | - Da Li
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Fan Mo
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huinan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Dongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Junliang Yuan
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing 100191, China.
| | - Yukai Wang
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Zhao-Qian Teng
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Baoyang Hu
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Medical School, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
5
|
Jin Y, Zhang J, Wu X, Qu C, Fang X, Yang Y, Yuan Y, Liu H, Han Z. Microfluidics-based label-free SERS profiling of exosomes with machine learning for osteosarcoma diagnosis. Talanta 2025; 294:128276. [PMID: 40344844 DOI: 10.1016/j.talanta.2025.128276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025]
Abstract
Osteosarcoma (OS) calls for early diagnosis to significantly improve patient survival rates. Exosomes hold significant potential as noninvasive biomarkers for the early diagnosis of cancer. Here, we design a microfluidic device to purify and analyze plasma-derived exosomes by label-free surface-enhanced Raman spectroscopy (SERS) profiling for OS diagnosis. Exosomes were isolated, purified, and enriched using a size-dependent microfluidic chip with tangential flow filtration, achieving a high recovery rate of 82 %. The isolated exosomes were then analyzed by label-free SERS using a nanoarray chip with self-assembly monolayers of gold nanoparticles (GNPs). Exosomes originating from different OS cell types were differentiated based on the intrinsic SERS signals. Our approach was further employed to analyze the plasma-derived exosomes from healthy donors and OS patients without the need for specific biomarker labeling. A machine learning-based diagnostic model for OS was constructed, achieving an accuracy of 93 %. The findings indicate that our method is valuable for noninvasive and precise diagnosis of OS and could be generalized to other diseases in the future.
Collapse
Affiliation(s)
- Ying Jin
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Junjie Zhang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Xinyi Wu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Cheng Qu
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, PR China
| | - Xingru Fang
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, PR China
| | - Yi Yang
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, PR China
| | - Yue Yuan
- Department of Pediatric Orthopedics, Anhui Children's Hospital (Children's Hospital of Anhui Medical University), Hefei, 230051, Anhui, PR China.
| | - Honglin Liu
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, School of Food and Biological Engineering, Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, 230009, PR China.
| | - Zhenzhen Han
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, PR China.
| |
Collapse
|
6
|
Hnath B, Dokholyan NV. Novel extracellular vesicle release pathway facilitated by toxic superoxide dismutase 1 oligomers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.07.647611. [PMID: 40291716 PMCID: PMC12026985 DOI: 10.1101/2025.04.07.647611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease resulting in paralysis and death within three to five years. Mutations in over forty different proteins have been linked to ALS, leading to controversy whether ALS is one disease or many diseases with a similar phenotype. Mutations in Cu,Zn superoxide dismutase 1 (SOD1) are only found in 2-3% of ALS cases, yet misfolded SOD1 is found in both sporadic (sALS) and familial (fALS) patients. Yet, mutations in TDP-43 or FUS increase the level of misfolded SOD1 on extracellular vesicles (EVs). Additionally, small EVs isolated from ALS patient samples caused cell death of wild type motor neurons and myotubules. The toxicity and protein alterations of ALS EVs have led to the theory that EVs are responsible for the spread of ALS. We hypothesize that previously-identified toxic trimeric SOD1 is spreading on EVs in ALS and altering the spread of other ALS-related proteins, linking them to a common mechanism. To test our hypothesis, we isolate EVs from motor neuron-like cells expressing trimer stabilizing mutations and perform a sandwich enzyme-linked immunoassay (ELISA) (CD9 capture antibody) to quantify whether misfolded SOD1 and 17 other ALS-related proteins increase or decrease on EVs with trimer stabilization. We identify which EV release pathway is being affected by trimeric SOD1 utilizing endocytosis and exocytosis inhibitors, and determine if any specific EV-related proteins are altered with trimer stabilization. We establish that VAPB, VCP, and Stathmin-2 increase on EVs with trimer stabilization. The common pathway between SOD1 and three other ALS-associated proteins is affected by multiple pathways, including the Caveolae endocytosis pathway, suggesting a novel hybrid pathway of EV release present in ALS.
Collapse
|
7
|
Duke LC, Cone AS, Sun L, Dittmer DP, Meckes DG, Tomko RJ. Tetraspanin CD9 alters cellular trafficking and endocytosis of tetraspanin CD63, affecting CD63 packaging into small extracellular vesicles. J Biol Chem 2025; 301:108255. [PMID: 39909378 PMCID: PMC11919600 DOI: 10.1016/j.jbc.2025.108255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/13/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Small extracellular vesicles (sEVs) are particles secreted from cells that play vital roles both in normal physiology and in human disease. sEVs are highly enriched in tetraspanin proteins, such as CD9 and CD63, and contain tetraspanin-enriched membrane microdomains involved in loading of sEVs with macromolecule cargoes and in sEV biogenesis. However, the precise roles of individual tetraspanins in sEV biogenesis and cargo loading remain poorly understood. Here, we report that CD9 negatively regulated CD63 trafficking to tetraspanin-enriched microdomains and its subsequent packaging into sEVs, whereas CD63 had no discernable effect on CD9 localization or packaging. Using super resolution microscopy of individual vesicles, we showed that CD9 governs the fraction of sEVs that are loaded with CD63. Interestingly, CD9-dependent suppression of CD63 packaging was rescued by pharmacological blockade of endocytosis. Together, our data support a model where CD9 contributes to the regulation and secretion of CD63 in an endocytosis-dependent manner to reprogram the contents of sEVs and tetraspanin-enriched microdomains.
Collapse
Affiliation(s)
- Leanne C Duke
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA.
| | - Allaura S Cone
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Robert J Tomko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
8
|
Chen Y, Douanne N, Wu T, Kaur I, Tsering T, Erzingatzian A, Nadeau A, Juncker D, Nerguizian V, Burnier JV. Leveraging nature's nanocarriers: Translating insights from extracellular vesicles to biomimetic synthetic vesicles for biomedical applications. SCIENCE ADVANCES 2025; 11:eads5249. [PMID: 40009680 PMCID: PMC11864201 DOI: 10.1126/sciadv.ads5249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Naturally occurring extracellular vesicles (EVs) and synthetic nanoparticles like liposomes have revolutionized precision diagnostics and medicine. EVs excel in biocompatibility and cell targeting, while liposomes offer enhanced drug loading capacity and scalability. The clinical translation of EVs is hindered by challenges including low yield and heterogeneity, whereas liposomes face rapid immune clearance and limited targeting efficiency. To bridge these gaps, biomimetic synthetic vesicles (SVs) have emerged as innovative platforms, combining the advantageous properties of EVs and liposomes. This review emphasizes critical aspects of EV biology, such as mechanisms of EV-cell interaction and source-dependent functionalities in targeting, immune modulation, and tissue regeneration, informing biomimetic SV engineering. We reviewed a broad array of biomimetic SVs, with a focus on lipid bilayered vesicles functionalized with proteins. These include cell-derived nanovesicles, protein-functionalized liposomes, and hybrid vesicles. By addressing current challenges and highlighting opportunities, this review aims to advance biomimetic SVs for transformative biomedical applications.
Collapse
Affiliation(s)
- Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Tad Wu
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Ishman Kaur
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- École de technologie supérieure ÉTS, Montreal, QC, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - David Juncker
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | | | - Julia V. Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Wang C, Feng Y, Rong X, Yan J, Lv B, Jiang H, Duan L, Jiang J. Mesenchymal stromal cell exosomes for drug delivery of prostate cancer treatments: a review. Stem Cell Res Ther 2025; 16:18. [PMID: 39849570 PMCID: PMC11755940 DOI: 10.1186/s13287-025-04133-8] [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: 10/09/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025] Open
Abstract
Interest in prostate cancer as a research topic has gradually increased. As a result, a series of innovative treatment strategies have emerged with an in-depth understanding of the disease. Owing to their unique biological characteristics, mesenchymal stromal cell exosomes (MSC-Exos) have garnered significant attention for their potential to deliver targeted drugs and enable precise prostate cancer treatment. Herein, prostate cancer treatment with MSC-Exos drug-delivery systems is reviewed. This review provides a comprehensive introduction to the advantages of these systems, current research trends and progress, as well as an analysis of current challenges and future research directions. Moreover, this review lays a solid foundation for the continued development and application of MSC-Exos.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Yanshuo Feng
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Xinao Rong
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Jingguo Yan
- Department of Intensive Care Unit, Dunhua Hospital, The First Hospital of Jilin University, Dunhua, Jilin Province, People's Republic of China
| | - Baisong Lv
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Hongyu Jiang
- Life Spring AKY Pharmaceuticals, Changchun, Jilin Province, People's Republic of China
| | - Lian Duan
- Department of Spine Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| | - Jinlan Jiang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
10
|
Bodart-Santos V, Ruan Z, Melvin BC, Pandey I, Ikezu S, Ikezu T. Selenoprotein P is a target for regulating extracellular vesicle biogenesis and secretion from activated microglia in vivo. Cell Rep 2024; 43:115025. [PMID: 39616613 PMCID: PMC11834494 DOI: 10.1016/j.celrep.2024.115025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 09/22/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
Microglia, brain innate immune cells, participate in the spread of inflammatory signals and aggregated proteins through secretion of extracellular vesicles (EVs). Selenoprotein P (Sepp1) is a potential regulator of microglial EV secretion. Here, we investigate the effect of Sepp1 silencing on microglial transcriptomics to elucidate the Sepp1 regulatory mechanism of EV secretion and validate this effect in APPNL-G-F knockin mice. Silencing of Sepp1 significantly reduces EV secretion and CD63 loading to EVs from BV-2 microglia, as determined by single-vesicle flow cytometry and super-resolution microscopy. Sepp1 deficiency downregulates EV biogenesis machinery, accompanied by increased lysosomal activity and lipid metabolism. Silencing of Sepp1 in astrocytes but not neurons suppresses EV secretion in vitro. Finally, Sepp1 silencing reduces EV secretion from activated neurodegenerative microglia associated with amyloid plaques in APPNL-G-F mouse brains in vivo. Sepp1 is thus an emerging therapeutic target for ameliorating microglia-mediated disease spread through EV secretion in neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Zhi Ruan
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Bridgette C Melvin
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Ikshu Pandey
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Seiko Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA; Regenerative Science Graduate Program, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA; Robert and Alene Kogod Center on Aging, Mayo Clinic, Jacksonville, FL 32224, USA; Alzheimer's Disease Research Center, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
11
|
Qiao Z, Wang X, Zhao H, Deng Y, Zeng W, Wu J, Chen Y. Research on the TSPAN6 regulating the secretion of ADSCs-Exos through syntenin-1 and promoting wound healing. Stem Cell Res Ther 2024; 15:430. [PMID: 39548518 PMCID: PMC11566053 DOI: 10.1186/s13287-024-04004-8] [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: 08/10/2024] [Accepted: 10/17/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Exosomes (Exos) from adipose-derived stem cells (ADSCs) have a high inclusion content and low immunogenicity, which helps to control inflammation and accelerate the healing of wounds. Unfortunately, the yield of exosomes is poor, which raises the expense and lengthens the treatment period in addition to impairing exosomes' therapeutic impact. Thus, one of the key problems that needs to be resolved in the current exosome study is increasing the exosome yield. METHODS Tetraspanin-6 (TSPAN6) overexpression and knockdown models of ADSCs were constructed to determine the number of exosomes secreted by each group of cells as well as the number of multivesicular bodies (MVBs) and intraluminal vesicles (ILVs) within the cells. Subsequently, the binding region of the interaction between TSPAN6 and syntenin-1 was identified using the yeast two-hybrid assay, and the interaction itself was identified by immunoprecipitation. Finally, cellular and animal studies were conducted to investigate the role of each class of exosomes. RESULTS When compared to the control group, the number of intracellular MVBs and ILVs was significantly larger, and the number of ADSCsTSPAN6+-Exos was more than three times higher. However, TSPAN6's ability to stimulate exosome secretion was reduced as a result of syntenin-1 knockdown. Additional yeast two-hybrid assay demonstrated that the critical structures for their interaction were the N-terminal, Postsynaptic density protein 95/Discs large protein/Zonula occludens 1 (PDZ1), and PDZ2 domains of syntenin-1, and the C-terminal of TSPAN6. In animal trials, the wound healing rate was best in the ADSCsTSPAN6+-Exos group, while cellular experiments demonstrated that ADSCsTSPAN6+-Exos better enhanced the proliferation and migration of human skin fibroblasts (HSFs) and human umbilical vein endothelial cells (HUVECs). CONCLUSION TSPAN6 stimulates exosome secretion and formation, as well as the creation of MVBs and ILVs in ADSCs. Syntenin-1 is essential for TSPAN6's stimulation of ADSCs-Exos secretion. Furthermore, ADSCsTSPAN6+-Exos has a greater ability to support wound healing, angiogenesis, and the proliferation and migration of a variety of cells.
Collapse
Affiliation(s)
- Zhihua Qiao
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiancheng Wang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hongli Zhao
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiwen Deng
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiliang Zeng
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingjing Wu
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yunzhu Chen
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Sun Y, Zhang S, Shen Y, Lu H, Zhao X, Wang X, Wang Y, Wang T, Liu B, Yao L, Wen J. Therapeutic application of mesenchymal stem cell-derived exosomes in skin wound healing. Front Bioeng Biotechnol 2024; 12:1428793. [PMID: 39161350 PMCID: PMC11330766 DOI: 10.3389/fbioe.2024.1428793] [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: 05/08/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Wound healing is a complicated obstacle, especially for chronic wounds. Mesenchymal stem cell-derived exosomes may be a promising cell-free approach for treating skin wound healing. Exosomes can accelerate wound healing by attenuating inflammation, promoting angiogenesis, cell proliferation, extracellular matrix production and remodeling. However, many issues, such as off-target effects and high degradation of exosomes in wound sites need to be addressed before applying into clinical therapy. Therefore, the bioengineering technology has been introduced to modify exosomes with greater stability and specific therapeutic property. To prolong the function time and the local concentration of exosomes in the wound bed, the use of biomaterials to load exosomes emerges as a promising strategy. In this review, we summarize the biogenesis and characteristics of exosomes, the role of exosomes in wound healing, and the therapeutic applications of modified-exosomes in wound healing. The challenges and prospects of exosomes in wound healing are also discussed.
Collapse
Affiliation(s)
- Yunhan Sun
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shun Zhang
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yukai Shen
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Haoyang Lu
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xincan Zhao
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xin Wang
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yongkai Wang
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Taiping Wang
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bing Liu
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lan Yao
- Eye Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Jie Wen
- School of Stomatology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
13
|
Xu F, Luo S, Lu P, Cai C, Li W, Li C. Composition, functions, and applications of exosomal membrane proteins. Front Immunol 2024; 15:1408415. [PMID: 39148736 PMCID: PMC11324478 DOI: 10.3389/fimmu.2024.1408415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
Exosomes play a crucial role in various biological processes, such as human development, immune responses, and disease occurrence. The membrane proteins on exosomes are pivotal factors for their biological functionality. Currently, numerous membrane proteins have been identified on exosome membranes, participating in intercellular communication, mediating target cell recognition, and regulating immune processes. Furthermore, membrane proteins from exosomes derived from cancer cells can serve as relevant biomarkers for early cancer diagnosis. This article provides a comprehensive review of the composition of exosome membrane proteins and their diverse functions in the organism's biological processes. Through in-depth exploration of exosome membrane proteins, it is expected to offer essential foundations for the future development of novel biomedical diagnostics and therapies.
Collapse
Affiliation(s)
- Fang Xu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Shumin Luo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Pengpeng Lu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chao Cai
- Integrated Chinese and Western Medicine Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Weihua Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Integrated Chinese and Western Medicine Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chuanyun Li
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|