1
|
Wang J, Zhao M, Fu D, Wang M, Han C, Lv Z, Wang L, Liu J. Human neural stem cell-derived extracellular vesicles protect against ischemic stroke by activating the PI3K/AKT/mTOR pathway. Neural Regen Res 2025; 20:3245-3258. [PMID: 39248158 PMCID: PMC11881723 DOI: 10.4103/nrr.nrr-d-23-01144] [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: 12/06/2023] [Revised: 05/11/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00028/figure1/v/2024-12-20T164640Z/r/image-tiff Human neural stem cell-derived extracellular vesicles exhibit analogous functions to their parental cells, and can thus be used as substitutes for stem cells in stem cell therapy, thereby mitigating the risks of stem cell therapy and advancing the frontiers of stem cell-derived treatments. This lays a foundation for the development of potentially potent new treatment modalities for ischemic stroke. However, the precise mechanisms underlying the efficacy and safety of human neural stem cell-derived extracellular vesicles remain unclear, presenting challenges for clinical translation. To promote the translation of therapy based on human neural stem cell-derived extracellular vesicles from the bench to the bedside, we conducted a comprehensive preclinical study to evaluate the efficacy and safety of human neural stem cell-derived extracellular vesicles in the treatment of ischemic stroke. We found that administration of human neural stem cell-derived extracellular vesicles to an ischemic stroke rat model reduced the volume of cerebral infarction and promoted functional recovery by alleviating neuronal apoptosis. The human neural stem cell-derived extracellular vesicles reduced neuronal apoptosis by enhancing phosphorylation of phosphoinositide 3-kinase, mammalian target of rapamycin, and protein kinase B, and these effects were reversed by treatment with a phosphoinositide 3-kinase inhibitor. These findings suggest that human neural stem cell-derived extracellular vesicles play a neuroprotective role in ischemic stroke through activation of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway. Finally, we showed that human neural stem cell-derived extracellular vesicles have a good in vivo safety profile. Therefore, human neural stem cell-derived extracellular vesicles are a promising potential agent for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Dong Fu
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Meina Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Chao Han
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Zhongyue Lv
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| |
Collapse
|
2
|
Sanie-Jahromi F, Sadeghi N, Moayedfard Z, Gharegezloo Z, Nejabat M, Nowroozzadeh MH. Effects of exosomes derived from activated corneal stromal keratocytes on the inflammation, proliferation, neuroprotection and epithelial-mesenchymal transition in retinal pigment epithelium cells. Life Sci 2025; 371:123592. [PMID: 40174671 DOI: 10.1016/j.lfs.2025.123592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/04/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
AIMS This study investigated the effects of activated keratocyte-derived exosomes (aKExo) on retinal pigment epithelial (RPE) cells in-vitro, focusing on cell viability, inflammatory cytokine expression, and neuroprotective properties. MATERIALS AND METHODS Keratocytes were cultured, and exosomes were extracted and characterized using transmission electron microscopy (TEM), scanning electron microscopy (SEM), flow cytometry, and dynamic light scattering (DLS). RPE cells, isolated from a human donor, were confirmed via RPE65 expression. aKExo effects on RPE cells were assessed using MTT assay at concentrations from 10-1 (35 μg/mL) to 10-5 (3.5 × 10-3 μg/mL). The optimal aKExo concentration (10-5) enhanced cell viability and exhibited the highest proliferative potential compared to the control group, making it the optimal dose for subsequent experiments including gene expression analysis, and ELISA. KEY FINDINGS aKExo downregulated IL-6 mRNA (0.70 ± 0.06, p = 0.0009) and marginally reduced TGF-β mRNA (0.75 ± 0.16, p = 0.0575). ELISA confirmed a reduction in IL-6 (31.33 ± 5.77 pg/mL vs. 50.22 ± 13.47 pg/mL, p = 0.0894) and TGF-β (8.91 ± 0.16 pg/mL vs. 11.39 ± 1.49 pg/mL, p = 0.0460). No significant changes were observed for IL-1β expression or other epithelial-mesenchymal transition (EMT)-related genes (α-SMA, ZEB-1, β-catenin). Neuroprotective genes NGF (4.34 ± 1.05, p = 0.0053) and CD90 (1.55 ± 0.25, p = 0.0184) were significantly upregulated, while VEGF-A was elevated (1.65 ± 0.15, p = 0.0018). SIGNIFICANCE These findings highlight aKExo's immunomodulatory, neuroprotective, and anti-EMT effects, suggesting potential therapeutic applications for retinal disorders, while noting that VEGF-A upregulation requires further investigation.
Collapse
Affiliation(s)
- Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Niloofar Sadeghi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Moayedfard
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Gharegezloo
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Nejabat
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - M Hossein Nowroozzadeh
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Wu P, Wang Z, Sun Y, Cheng Z, Wang M, Wang B. Extracellular vesicles: a new frontier in diagnosing and treating graft-versus-host disease after allogeneic hematopoietic cell transplantation. J Nanobiotechnology 2025; 23:251. [PMID: 40133949 PMCID: PMC11938667 DOI: 10.1186/s12951-025-03297-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Graft-versus-host disease (GvHD) is a prevalent complication following allogeneic hematopoietic stem cell transplantation (HSCT) and is characterized by relatively high morbidity and mortality rates. GvHD can result in extensive systemic damage in patients following allogeneic HSCT (allo-HSCT), with the skin, gastrointestinal tract, and liver frequently being the primary target organs affected. The severe manifestations of acute intestinal GvHD often indicate a poor prognosis for patients after allo-HSCT. Endoscopy and histopathological evaluation remain employed to diagnose GvHD, and auxiliary examinations exclude differential diagnoses. Currently, reliable serum biomarkers for the diagnosis and differential diagnosis of GvHD are scarce. As an essential part of standard transplant protocols, early application of immunosuppressive drugs effectively prevents GvHD. Among them, steroids represent first-line therapeutic agents, and the JAK2 inhibitor ruxolitinib represents the second-line therapeutic agent. Currently, no efficacious treatment modality exists for steroid-resistant aGvHD. Therefore, the diagnosis and treatment of GvHD still face significant medical demands. Extracellular vesicles (EVs) are nanometer to micrometer-scale biomembrane vesicles containing various bioactive components, such as proteins, nucleotides, and metabolites. Distinctive changes in serum-derived EV components occur in patients after allo-HSCT; Hence, EVs are expected to be potential biomarkers for diagnosing and treating GvHD. Furthermore, cell-free therapeutics characterized by EVs derived from mesenchymal stem cells (MSCs) have manifested remarkable therapeutic efficacy in preclinical models and preclinical trials of GvHD. Customized engineered EVs with fewer toxic and side effects for the combined treatment of GvHD hold broad prospects for clinical translation. This review article examines the potential value of translating EVs into clinical applications for the diagnosis and treatment of GvHD. It summarizes the latest advancements and prospects of engineered EVs applying GvHD.
Collapse
Affiliation(s)
- Peipei Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Zhangfei Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yongping Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhixiang Cheng
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Anhui Public Health Clinical Center, Hefei, China.
| | - Min Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, China.
| | - Baolong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
4
|
Yoo D, Jung SY, Go D, Park JY, You DG, Jung WK, Li Y, Ding J, Park JH, Um W. Functionalized extracellular vesicles of mesenchymal stem cells for regenerative medicine. J Nanobiotechnology 2025; 23:219. [PMID: 40102934 PMCID: PMC11921732 DOI: 10.1186/s12951-025-03300-6] [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: 06/09/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Stem cell-derived extracellular vesicles (EVs) have emerged as a safe and potent alternative to regenerative medicine in recent decades. Furthermore, the adjustment of EV functions has been recently enabled by certain stem cell preconditioning methods, providing an exceptional opportunity to enhance the therapeutic potential or confer additional functions of stem cell-derived EVs. In this review, we discuss the recent progress of functionalized EVs, based on stem cell preconditioning, for treating various organ systems, such as the musculoskeletal system, nervous system, integumentary system, cardiovascular system, renal system, and respiratory system. Additionally, we summarize the expected outcomes of preconditioning methods for stem cells and their EVs. With recent progress, we suggest considerations and future directions for developing personalized medicine based on preconditioned stem cell-derived EVs.
Collapse
Affiliation(s)
- Donghyeon Yoo
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Se Young Jung
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dabin Go
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Ji Yeong Park
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dong Gil You
- Department of Chemical Engineering & Biotechnology, Tech University of Korea, Siheung, 15073, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| | - Yuce Li
- College of Life Science and Health, Wuhan University of Science and Technology (WUST), Wuhan, 430065, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
5
|
van Maanen JC, Bach FC, Snuggs JW, Ito K, Wauben MHM, Le Maitre CL, Tryfonidou MA. Explorative Study of Modulatory Effects of Notochordal Cell-Derived Extracellular Vesicles on the IL-1β-Induced Catabolic Cascade in Nucleus Pulposus Cell Pellets and Explants. JOR Spine 2025; 8:e70043. [PMID: 39881783 PMCID: PMC11775941 DOI: 10.1002/jsp2.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/27/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025] Open
Abstract
Background Cell-free regenerative strategies, such as notochordal cell (NC)-derived extracellular vesicles (EVs), are an attractive alternative in developing new therapies for intervertebral disc (IVD) degeneration. NC-EVs have been reported to elicit matrix anabolic effects on nucleus pulposus cells from degenerated IVDs cultured under basal conditions. However, the degenerative process is exacerbated by pro-inflammatory cytokines contributing to the vicious degenerative cycle. Therefore, this study explores whether NC-EVs modulate interleukin (IL)-1β-mediated pro-inflammatory responses in the degenerating disc. Methods This study utilized two IL-1β induced pro-catabolic culture models; a dog 3D nucleus pulposus (NP) cell pellet culture and a human patient-derived, ex vivo NP tissue culture system. Porcine NC-EVs were generated from NC-conditioned medium by differential centrifugation followed by size exclusion chromatography. Donor matched EV-depleted media were generated by overnight ultracentrifugation, whereafter the EV-depleted NCCM supernatant was subjected to size exclusion chromatography. To investigate whether observed effects were EV-associated, NC-EVs conditions were compared to EV-depleted controls in the absence and presence of IL-1β. Results The size and concentration of NC-EVs were quantified by nanoparticle tracking analysis, which showed minimal donor variation and confirmed depletion of EVs in the EV-depleted media. In the IL-1β-induced catabolic cascade, the NC-EVs did not elicit anabolic effects at the matrix level nor did they rescue the pro-catabolic phenotype within dog pellets. Modification of the CCL2 secretion seemed to be context dependent in the human explants: where EVs treatment stimulated CCL2 secretion but in the presence of IL-1β this effect was counteracted. Secretion of IL-6 and C-X-C motif chemokine ligand 1 was significantly decreased in NC-EV + IL-1β vs. control+IL-1β but not compared to EV-depleted human explant controls. Altogether, this data provides evidence for a protective modulatory role of NC-EVs. Considering the homeostatic function EVs exert, inherently encompassing subtle biologic modifications, the current study may have lacked sufficient power to demonstrate statistical significance in a sample set with evident donor variation. Conclusions NC-EVs may modulate the production of specific cytokines and chemokines in human degenerate explants when the key pro-inflammatory cytokine IL-1β is present. Implementation of the technical EV-depleted controls in further studies is essential to robustly demonstrate that these effects are EV-mediated and not associated with other secreted factors co-isolated during EV-isolation.
Collapse
Affiliation(s)
- J. C. van Maanen
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtNetherlands
| | - F. C. Bach
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtNetherlands
| | - J. W. Snuggs
- Division of Clinical Medicine, Faculty of HealthUniversity of SheffieldSheffieldUK
| | - K. Ito
- Orthopedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenNetherlands
- Department of OrthopedicsUniversity Medical Centre UtrechtUtrechtNetherlands
| | - M. H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtNetherlands
| | - C. L. Le Maitre
- Division of Clinical Medicine, Faculty of HealthUniversity of SheffieldSheffieldUK
| | - M. A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtNetherlands
| |
Collapse
|
6
|
Wang B, Zhao Y, Zhao M, Han Y, Zhang H, Kong W, Qu F. Al-Based Metal-Organic Nanoslice-Coupled Phospholipid Layer Enables Highly Enriched Biological External Vesicles. Anal Chem 2025; 97:3319-3327. [PMID: 39899402 DOI: 10.1021/acs.analchem.4c05089] [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: 02/05/2025]
Abstract
Systematic analysis of the information content of biological external vesicles (BEVs) is essential for understanding the complex relationships between metabolic processes and cells at a systemic level. Although considerable efforts have been made to enrich BEVs, their comprehensive analysis in a way that maintains and stabilizes information maintenance at high doses remains a challenge. To address this issue, we developed a metal-organic nanoslice-coupled exocytosis phospholipid layer method that utilized subtle interactions between the metal-organic nanoslice and the phospholipid molecular layer of the membrane to achieve the convenient and efficient enrichment of BEVs. The method bypassed the conventional ultracentrifugation and size exclusion separation and helped to highly preserve the internal information on BEVs. By integrating and analyzing the enriched BEVs for elements such as membrane proteins and endotoxins, the simplicity and robustness of the metal-organic nanoslice-coupled vesicle surface phospholipid layer technology were verified, which provided a reliable platform for studying the cellular events associated with outer vesicles and expanded the biological applications of the metal-organic nanoslice.
Collapse
Affiliation(s)
- Bangming Wang
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yan Zhao
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Mingzhu Zhao
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuke Han
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Hui Zhang
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| | - Weiheng Kong
- Key Laboratory of Life-Organic Analysis of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Fengli Qu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310022, P. R. China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, P. R. China
| |
Collapse
|
7
|
Kim JE, Lee JW, Cha GD, Yoon JK. The Potential of Mesenchymal Stem Cell-Derived Exosomes to Treat Diabetes Mellitus. Biomimetics (Basel) 2025; 10:49. [PMID: 39851765 PMCID: PMC11760843 DOI: 10.3390/biomimetics10010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Diabetes mellitus (DM) is a fatal metabolic disease characterized by persistent hyperglycemia. In recent studies, mesenchymal stem cell (MSC)-derived exosomes, which are being investigated clinically as a cell-free therapy for various diseases, have gained attention due to their biomimetic properties that closely resemble natural cellular communication systems. These MSC-derived exosomes inherit the regenerative and protective effects from MSCs, inducing pancreatic β-cell proliferation and inhibiting apoptosis, as well as ameliorating insulin resistance by suppressing the release of various inflammatory cytokines. Consequently, MSC-derived exosomes have attracted attention as a novel treatment for DM as an alternative to stem cell therapy. In this review, we will introduce the potential of MSC-derived exosomes for the treatment of DM by discussing the studies that have used MSC-derived exosomes to treat DM, which have shown therapeutic effects in both type 1 and type 2 DM.
Collapse
Affiliation(s)
| | | | | | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea (G.D.C.)
| |
Collapse
|
8
|
Zhu Q, Mao X, Zhu X, Xiao Y, Xu H, Su L, Liu X, Huang X, Wang L. Hypoxia-Induced and Glucuronic Acid-Modified Extracellular Vesicles from Mesenchymal Stromal Cells Treat Pulmonary Arterial Hypertension by Improving Vascular Remodeling. NANO LETTERS 2024; 24:16342-16350. [PMID: 39660764 DOI: 10.1021/acs.nanolett.4c04638] [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: 12/12/2024]
Abstract
Achieving precise delivery of extracellular vesicles (EVs) to treat pulmonary arterial hypertension (PAH) remains challenging. Here, we propose a strategy using hypoxia-induced and glucuronic acid (GA)-modified mesenchymal stromal-cell-derived EVs (MSC-EVs) to enhance their functionalities and therapeutic targeting. The hypoxia-induced EVs (Hypo-EVs) exhibit enriched exosomal signatures and display heightened inhibition of the proliferation of pulmonary arterial smooth muscle cells (PASMCs) compared to normoxic EVs (Norm-EV). We then modify Hypo-EVs by incorporating GA into their outer membrane, targeting glucose transporter-1 overexpressed on PASMCs. Our studies show that GA-EVs significantly enhance the therapeutic efficacy, both in vitro and in vivo, through improved targeted delivery to diseased PASMCs for improving vascular remodeling. Additionally, we identify miR-5119 involved in the PAH-associated calcium signaling pathway as a key contributor to GA-EVs' superior effects. This work provides a promising strategy for PAH treatment and advances the clinical potential of MSC-EV-based therapies.
Collapse
Affiliation(s)
- Qingfu Zhu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University,Xueyuan Road 270, Wenzhou 325027, China
| | - Xulong Mao
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xinxi Zhu
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yijia Xiao
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Hao Xu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University,Xueyuan Road 270, Wenzhou 325027, China
| | - Lihuang Su
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaohu Liu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University,Xueyuan Road 270, Wenzhou 325027, China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Liangxing Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
9
|
Kim J, Jo W. Engineering of buried interfaces in perovskites: advancing sustainable photovoltaics. NANO CONVERGENCE 2024; 11:57. [PMID: 39681713 DOI: 10.1186/s40580-024-00464-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024]
Abstract
Perovskite solar cells (PSCs) have garnered significant attention for their high power conversion efficiency (PCE) and potential for cost-effective, large-scale manufacturing. This comprehensive review focuses on the role of buried interface engineering in enhancing the performance and stability of PSCs with both n-type electron transport layer/perovskite/p-type hole transport layer (n-i-p) and p-type hole transport layer/perovskite/n-type electron transport layer (p-i-n) structures. This study highlights key challenges associated with interface engineering, such as charge extraction, recombination loss, and energy level alignment. Various interface engineering techniques, such as surface passivation, self-assembled monolayers, and additive engineering, are explored in terms of their effectiveness in mitigating recombination loss and improving long-term device stability. This review also provides an in-depth analysis of material selection for the electron and hole transport layers, defect management techniques, and the influence of these on perovskite film quality and device stability. Advanced characterization methods for buried interfaces are discussed, providing insights into the structural, morphological, and electronic properties that govern device performance. Furthermore, we explore emerging approaches that target homogenous cation distribution and phase stability at buried interfaces, both of which are crucial for improving PCEs beyond current benchmarks. By synthesizing the latest research findings and identifying key challenges, this review aims to guide future directions in interface engineering for PSCs and ensure their successful use in next-generation sustainable energy technologies.
Collapse
Affiliation(s)
- Jihyun Kim
- New and Renewable Energy Research Center, Ewha Womans University, Seoul, 03760, Korea
| | - William Jo
- New and Renewable Energy Research Center, Ewha Womans University, Seoul, 03760, Korea.
- Department of Physics, Ewha Womans University, Seoul, 03760, Korea.
| |
Collapse
|
10
|
Ma YN, Hu X, Karako K, Song P, Tang W, Xia Y. Exploring the multiple therapeutic mechanisms and challenges of mesenchymal stem cell-derived exosomes in Alzheimer's disease. Biosci Trends 2024; 18:413-430. [PMID: 39401895 DOI: 10.5582/bst.2024.01306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder, and the current treatment options are limited. Mesenchymal stem cell-derived exosomes (MSC-Exos) have garnered significant attention due to their unique biological properties, showcasing tremendous potential as an acellular alternative therapy for AD. MSC-Exos exhibit excellent biocompatibility and low immunogenicity, enabling them to effectively cross the blood-brain barrier (BBB) and deliver therapeutic molecules directly to target cells. They are highly efficacious in delivering nucleic acid-based drugs. Moreover, the production process of MSC-Exos benefits from a high proliferation capacity and multilineage differentiation potential, allowing for production while maintaining a stable composition. Despite the significant theoretical advantages of MSC-Exos, their clinical use still faces multiple challenges, including cross-contamination during isolation and purification processes, the complexity of their components, and the presence of potential adverse paracrine factors. Future research needs to focus on optimizing separation and purification techniques, enhancing delivery methods to improve therapeutic efficacy, and performing detailed analyses of the components of MSC-Exos. In summary, MSC-Exos hold promise as an effective option for the treatment of AD and other neurodegenerative diseases, driving their clinical research and use in related fields.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Xiqi Hu
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Kenji Karako
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Peipei Song
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| |
Collapse
|
11
|
Ahmed W, Mushtaq A, Ali S, Khan N, Liang Y, Duan L. Engineering Approaches for Exosome Cargo Loading and Targeted Delivery: Biological versus Chemical Perspectives. ACS Biomater Sci Eng 2024; 10:5960-5976. [PMID: 38940421 DOI: 10.1021/acsbiomaterials.4c00856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Exosomes are nanoscale membrane bound vesicles secreted by almost all types of cells. Their unique attributes, such as minimal immunogenicity and compatibility with biological systems, make them novel carriers for drug delivery. These native exosomes harbor proteins, nucleic acids, small molecule compounds, and fluorogenic agents. Moreover, through a combination of chemical and bioengineering methodologies, exosomes are tailored to transport precise therapeutic payloads to designated cells or tissues. In this review, we summarize the strategies for exosome modification and drug loading modalities in engineered exosomes. In addition, we provide an overview of the advances in the use of engineered exosomes for targeted drug delivery. Lastly, we discuss the merits and limitations of chemically engineered versus bioengineered exosome-mediated target therapies. These insights offer additional options for refining engineered exosomes in pharmaceutical development and hold promise for expediting the successful translation of engineered exosomes from the bench to the bedside.
Collapse
Affiliation(s)
- Waqas Ahmed
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
- Medical School, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Asim Mushtaq
- Centre for Future Materials, University of Southern Queensland, Springfield, Queensland 4300, Australia
| | - Shahzad Ali
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
- Medical School, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Nawaz Khan
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
- Medical School, Shenzhen University, Shenzhen 518060, Guangdong, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Institute of Mental Health, Shenzhen Mental Health Center, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen 518020, Guangdong, China
| | - Li Duan
- Department of Orthopedics, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, Guangdong, China
| |
Collapse
|
12
|
Lee S, Jung SY, Yoo D, Go D, Park JY, Lee JM, Um W. Alternatives of mesenchymal stem cell-derived exosomes as potential therapeutic platforms. Front Bioeng Biotechnol 2024; 12:1478517. [PMID: 39315312 PMCID: PMC11417005 DOI: 10.3389/fbioe.2024.1478517] [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: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
With outstanding therapeutic potential in the tissue regeneration and anti-inflammation, mesenchymal stem cell-derived exosomes (MSC-EXOs) have emerged as a prominent therapeutic in recent. However, poor production yield and reproducibility have remained as significant challenges of their practical applications. To surmount these challenges, various alternative materials with stem cell-like functions, have been recently investigated, however, there has been no comprehensive analysis in these alternatives so far. Here, we discuss the recent progress of alternatives of MSC-EXOs, including exosomes and exosome-like nanovesicles from various biological sources such as plants, milk, microbes, and body fluids. Moreover, we extensively compare each alternative by summarizing their unique functions and mode of actions to suggest the expected therapeutic target and future directions for developing alternatives for MSC-EXOs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jong Min Lee
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
13
|
Yang Y, Liu H, Guo K, Yu Q, Zhao Y, Wang J, Huang Y, Li W. Extracellular Vesicles from Compression-Loaded Cementoblasts Promote the Tissue Repair Function of Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402529. [PMID: 39101239 PMCID: PMC11423227 DOI: 10.1002/advs.202402529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/19/2024] [Indexed: 08/06/2024]
Abstract
Treatment strategies for hard tissue defects aim to establish a mineralized microenvironment that facilitates tissue remodeling. As a mineralized tissue, cementum shares a similar structure with bone and exhibits an excellent capacity to resist resorption under compression. Macrophages are crucial for mineralized remodeling; however, their functional alterations in the microenvironment of cementum remain poorly understood. Therefore, this study explores the mechanisms by which cementum resists resorption under compression and the regulatory roles of cementoblasts in macrophage functions. As a result, extracellular vesicles from compression-loaded cementoblasts (Comp-EVs) promote macrophage M2 polarization and enhance the clearance of apoptotic cells (efferocytosis) by 2- to 3-fold. Local injection of Comp-EVs relieves cementum destruction in mouse root resorption model by activating the tissue repair function of macrophages. Moreover, Comp-EV-loaded hydrogels achieve significant bone healing in calvarial bone defect. Unexpectedly, under compression, EV secretion in cementoblasts is reduced by half. RNA-Seq analysis and verification reveal that Rab35 expression decreases by 60% under compression, thereby hampering the release of EVs. Rab35 overexpression is proposed as a modification of cementoblasts to boost the yield of Comp-EVs. Collectively, Comp-EVs activate the repair function of macrophages, which will be a potential therapeutic strategy for hard tissue repair and regeneration.
Collapse
Affiliation(s)
- Yuhui Yang
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Hao Liu
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Kunyao Guo
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Qianyao Yu
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Yi Zhao
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Jiayi Wang
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Yiping Huang
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| | - Weiran Li
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory for Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| |
Collapse
|
14
|
Niu Q, Lin C, Yang S, Rong S, Wei J, Zhao T, Peng Y, Cheng Z, Xie Y, Wang Y. FoxO1-Overexpressed Small Extracellular Vesicles Derived from hPDLSCs Promote Periodontal Tissue Regeneration by Reducing Mitochondrial Dysfunction to Regulate Osteogenesis and Inflammation. Int J Nanomedicine 2024; 19:8751-8768. [PMID: 39220194 PMCID: PMC11365494 DOI: 10.2147/ijn.s470419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Periodontitis is a chronic infectious disease characterized by progressive inflammation and alveolar bone loss. Forkhead box O1 (FoxO1), an important regulator, plays a crucial role in maintaining bone homeostasis and regulating macrophage energy metabolism and osteogenic differentiation of mesenchymal stem cells (MSCs). In this study, FoxO1 was overexpressed into small extracellular vesicles (sEV) using engineering technology, and effects of FoxO1-overexpressed sEV on periodontal tissue regeneration as well as the underlying mechanisms were investigated. Methods Human periodontal ligament stem cell (hPDLSCs)-derived sEV (hPDLSCs-sEV) were isolated using ultracentrifugation. They were then characterized using transmission electron microscopy, Nanosight, and Western blotting analyses. hPDLSCs were treated with hPDLSCs-sEV in vitro after stimulation with lipopolysaccharide, and osteogenesis was evaluated. The effect of hPDLSCs-sEV on the polarization phenotype of THP-1 macrophages was also evaluated. In addition, we measured the reactive oxygen species (ROS) levels, adenosine triphosphate (ATP) production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells. Experimental periodontitis was established in vivo in mice. HPDLSCs-sEV or phosphate-buffered saline (PBS) were injected into periodontal tissues for four weeks, and the maxillae were collected and assessed by micro-computed tomography, histological staining, and small animal in vivo imaging. Results In vitro, FoxO1-overexpressed sEV promoted osteogenic differentiation of hPDLSCs in the inflammatory environment and polarized THP-1 cells from the M1 phenotype to the M2 phenotype. Furthermore, FoxO1-overexpressed sEV regulated the ROS level, ATP production, mitochondrial characteristics, and metabolism of hPDLSCs and THP-1 cells in the inflammatory environment. In the in vivo analyses, FoxO1-overexpressed sEV effectively promoted bone formation and inhibited inflammation. Conclusion FoxO1-overexpressed sEV can regulate osteogenesis and immunomodulation. The ability of FoxO1-overexpressed sEV to regulate inflammation and osteogenesis can pave the way for the establishment of a therapeutic approach for periodontitis.
Collapse
Affiliation(s)
- Qingru Niu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Chuanmiao Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Shuqing Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Shuxuan Rong
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Junbin Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Tingting Zhao
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yingying Peng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Zhilan Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yunyi Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangzhou, People’s Republic of China
- Sun Yat-Sen University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, People’s Republic of China
| |
Collapse
|
15
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-w] [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: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
16
|
Li XL, Fan W, Fan B. Dental pulp regeneration strategies: A review of status quo and recent advances. Bioact Mater 2024; 38:258-275. [PMID: 38745589 PMCID: PMC11090883 DOI: 10.1016/j.bioactmat.2024.04.031] [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: 11/27/2023] [Revised: 02/18/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024] Open
Abstract
Microorganisms, physical factors such as temperature or mechanical injury, and chemical factors such as free monomers from composite resin are the main causes of dental pulp diseases. Current clinical treatment methods for pulp diseases include the root canal therapy, vital pulp therapy and regenerative endodontic therapy. Regenerative endodontic therapy serves the purpose of inducing the regeneration of new functional pulp tissues through autologous revascularization or pulp tissue engineering. This article first discusses the current clinical methods and reviews strategies as well as the research outcomes regarding the pulp regeneration. Then the in vivo models, the prospects and challenges for regenerative endodontic therapy were further discussed.
Collapse
Affiliation(s)
- Xin-Lu Li
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, 430079, Wuhan, China
| | - Wei Fan
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, 430079, Wuhan, China
| | - Bing Fan
- The State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, 430079, Wuhan, China
| |
Collapse
|
17
|
Li G, Zhang S, Zou Y, Ai H, Zheng X, Qian K, Lei C, Fu W. The therapeutic potential of exosomes in immunotherapy. Front Immunol 2024; 15:1424081. [PMID: 39040108 PMCID: PMC11260647 DOI: 10.3389/fimmu.2024.1424081] [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: 04/27/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Exosomes are found in various tissues of the body and carry abundant contents including nucleic acids, proteins, and metabolites, which continuously flow between cells of various tissues and mediate important intercellular communication. In addition, exosomes from different cellular sources possess different physiopathological immunomodulatory effects, which are closely related to the immune regeneration of normal or abnormal organs and tissues. Here, we focus on the mechanistic interactions between exosomes and the human immune system, introduce the immuno-regenerative therapeutic potential of exosomes in common clinical immune-related diseases, such as infectious diseases, autoimmune diseases, and tumors, and reveal the safety and efficacy of exosomes as a novel cell-free immune regenerative therapy.
Collapse
Affiliation(s)
- Guangyao Li
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Shuyi Zhang
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yitan Zou
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hongru Ai
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xinya Zheng
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Kewen Qian
- Department of Biomedical Engineering, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Changhai Lei
- Department of Biophysics, College of Basic Medical Sciences, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenyan Fu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Shimizu Y, Ntege EH, Inoue Y, Matsuura N, Sunami H, Sowa Y. Optimizing mesenchymal stem cell extracellular vesicles for chronic wound healing: Bioengineering, standardization, and safety. Regen Ther 2024; 26:260-274. [PMID: 38978963 PMCID: PMC11228664 DOI: 10.1016/j.reth.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/12/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Chronic wounds represent a significant global burden, afflicting millions with debilitating complications. Despite standard care, impaired healing persists due to factors like persistent inflammation and impaired tissue regeneration. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) offer an innovative regenerative medicine approach, delivering stem cell-derived therapeutic cargo in engineered nanoscale delivery systems. This review examines pioneering bioengineering strategies to engineer MSC-EVs into precision nanotherapeutics for chronic wounds. Emerging technologies like CRISPR gene editing, microfluidic manufacturing, and biomimetic delivery systems are highlighted for their potential to enhance MSC-EV targeting, optimize therapeutic cargo enrichment, and ensure consistent clinical-grade production. However, key hurdles remain, including batch variability, rigorous safety assessment for potential tumorigenicity, immunogenicity, and biodistribution profiling. Crucially, collaborative frameworks harmonizing regulatory science with bioengineering and patient advocacy hold the key to expediting global clinical translation. By overcoming these challenges, engineered MSC-EVs could catalyze a new era of off-the-shelf regenerative therapies, restoring hope and healing for millions afflicted by non-healing wounds.
Collapse
Affiliation(s)
- Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Edward Hosea Ntege
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshikazu Inoue
- Department of Plastic and Reconstructive Surgery, School of Medicine, Fujita Health University, 1-98, Dengakugakubo, Kutsukake, Toyoake, Aichi, 470-1192, Japan
| | - Naoki Matsuura
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Hiroshi Sunami
- Center for Advanced Medical Research, School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke, 329-0498, Tochigi, Japan
| |
Collapse
|
19
|
Bhat A, Malik A, Yadav P, Ware WJ, Kakalij P, Chand S. Mesenchymal stem cell‐derived extracellular vesicles: Recent therapeutics and targeted drug delivery advances. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3. [DOI: 10.1002/jex2.156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/25/2024] [Indexed: 01/03/2025]
Abstract
AbstractThe targeted drug delivery field is rapidly advancing, focusing on developing biocompatible nanoparticles that meet rigorous criteria of non‐toxicity, biocompatibility, and efficient release of encapsulated molecules. Conventional synthetic nanoparticles (SNPs) face complications such as elevated immune responses, complex synthesis methods, and toxicity, which restrict their utility in therapeutics and drug delivery. Extracellular vesicles (EVs) have emerged as promising substitutes for SNPs, leveraging their ability to cross biological barriers, biocompatibility, reduced toxicity, and natural origin. Notably, mesenchymal stem cell‐derived EVs (MSC‐EVs) have garnered much curiosity due to their potential in therapeutics and drug delivery. Studies suggest that MSC‐EVs, the central paracrine contributors of MSCs, replicate the therapeutic effects of MSCs. This review explores the characteristics of MSC‐EVs, emphasizing their potential in therapeutics and drug delivery for various diseases, including CRISPR/Cas9 delivery for gene editing. It also delves into the obstacles and challenges of MSC‐EVs in clinical applications and provides insights into strategies to overcome the limitations of biodistribution and target delivery.
Collapse
Affiliation(s)
- Anjali Bhat
- Department of Anesthesiology University of Nebraska Medical Center Omaha Nebraska USA
| | - Anshu Malik
- Institute for Quantitative Health Science and Engineering (IQ) Michigan State University East Lansing Michigan USA
- Department of Biomedical Engineering Michigan State University East Lansing Michigan USA
| | - Poonam Yadav
- Medical Science Interdepartmental Area University of Nebraska Medical Center Omaha Omaha Nebraska USA
| | | | - Pratiksha Kakalij
- Department of Pharmaceutical Sciences University of Nebraska Medical Center Omaha Omaha Nebraska USA
| | - Subhash Chand
- Department of Anesthesiology University of Nebraska Medical Center Omaha Nebraska USA
| |
Collapse
|
20
|
Xue K, Mi B. Engineered Extracellular Vesicles in Chronic Kidney Diseases: A Comprehensive Review. Int J Nanomedicine 2024; 19:2377-2393. [PMID: 38469058 PMCID: PMC10926925 DOI: 10.2147/ijn.s452393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/19/2024] [Indexed: 03/13/2024] Open
Abstract
Chronic kidney diseases (CKD) present a formidable global health challenge, characterized by a deficiency of effective treatment options. Extracellular vesicles (EVs), recognized as multifunctional drug delivery systems in biomedicine, have gained accumulative interest. Specifically, engineered EVs have emerged as a promising therapeutic approach for targeted drug delivery, potentially addressing the complexities of CKD management. In this review, we systematically dissect EVs, elucidating their classification, biogenesis, composition, and cargo molecules. Furthermore, we explore techniques for EV engineering and strategies for their precise renal delivery, focusing on cargo loading and transportation, providing a comprehensive perspective. Moreover, this review also discusses and summarizes the diverse therapeutic applications of engineered EVs in CKD, emphasizing their anti-inflammatory, immunomodulatory, renoprotective, and tissue-regenerating effects. It critically evaluates the challenges and limitations in translating EV therapies from laboratory settings to clinical applications, while outlining future prospects and emerging trends.
Collapse
Affiliation(s)
- Kaming Xue
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
21
|
Yang Q, Li S, Ou H, Zhang Y, Zhu G, Li S, Lei L. Exosome-based delivery strategies for tumor therapy: an update on modification, loading, and clinical application. J Nanobiotechnology 2024; 22:41. [PMID: 38281957 PMCID: PMC10823703 DOI: 10.1186/s12951-024-02298-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Malignancy is a major public health problem and among the leading lethal diseases worldwide. Although the current tumor treatment methods have therapeutic effect to a certain extent, they still have some shortcomings such as poor water solubility, short half-life, local and systemic toxicity. Therefore, how to deliver therapeutic agent so as to realize safe and effective anti-tumor therapy become a problem urgently to be solved in this field. As a medium of information exchange and material transport between cells, exosomes are considered to be a promising drug delivery carrier due to their nano-size, good biocompatibility, natural targeting, and easy modification. In this review, we summarize recent advances in the isolation, identification, drug loading, and modification of exosomes as drug carriers for tumor therapy alongside their application in tumor therapy. Basic knowledge of exosomes, such as their biogenesis, sources, and characterization methods, is also introduced herein. In addition, challenges related to the use of exosomes as drug delivery vehicles are discussed, along with future trends. This review provides a scientific basis for the application of exosome delivery systems in oncological therapy.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Haibo Ou
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shaohong Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Lanjie Lei
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
22
|
Han KH, Kim CH, Kim SH, Lee CH, Park M, Bui VD, Duong VH, Kwon S, Ha M, Kang H, Park JH. Immunogenic Extracellular Vesicles Derived from Endoplasmic Reticulum-Stressed Tumor Cells: Implications as the Therapeutic Cancer Vaccine. ACS NANO 2024; 18:199-209. [PMID: 38109681 DOI: 10.1021/acsnano.3c05645] [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: 12/20/2023]
Abstract
Tumor-derived extracellular vesicles (TDEs) have potential for therapeutic cancer vaccine applications since they innately possess tumor-associated antigens, mediate antigen presentation, and can incorporate immune adjuvants for enhanced vaccine efficacy. However, the original TDEs also contain immune-suppressive proteins. To address this, we proposed a simple yet powerful preconditioning method to improve the overall immunogenicity of the TDEs. This approach involved inducing endoplasmic reticulum (ER) stress on parental tumor cells via N-glycosylation inhibition with tunicamycin. The generated immunogenic TDEs (iTDEs) contained down-regulated immunosuppressive proteins and up-regulated immune adjuvants, effectively activating dendritic cells (DCs) in vitro. Furthermore, in vivo evidence from a tumor-bearing mouse model showed that iTDEs activated DCs, enabling cytotoxic T lymphocytes (CTLs) to target tumors, and eventually established a systemic antitumor immune response. Additionally, iTDEs significantly delayed tumor recurrence in a postsurgery model compared with control groups. These findings highlight the immense potential of our strategy for utilizing TDEs to develop effective cancer vaccines.
Collapse
Affiliation(s)
- Kyung Hee Han
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - So Hee Kim
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Chang Hyun Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Minsung Park
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351 Republic of Korea
| | - Van Dat Bui
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Van Hieu Duong
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Seunglee Kwon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Minji Ha
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Heegun Kang
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351 Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| |
Collapse
|
23
|
Wang YY, Cheng J, Liu YD, Wang YP, Yang QW, Zhou N. Exosome-based regenerative rehabilitation: A novel ice breaker for neurological disorders. Biomed Pharmacother 2023; 169:115920. [PMID: 37995565 DOI: 10.1016/j.biopha.2023.115920] [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: 09/14/2023] [Revised: 11/11/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Neurological disorders affect a large population, often leading to different levels of disability and resulting in decreased quality of life. Due to the limited recovery obtained from surgical procedures and other medical approaches, a large number of patients with prolonged dysfunction receive neurorehabilitation protocols to improve their neural plasticity and regeneration. However, the poor neural regeneration ability cannot effectively rebuild the tissue integrity and neural functional networks; consequently, the prognoses of neurorehabilitation remain undetermined. To increase the chances of neural regeneration and functional recovery for patients with neurological disorders, regenerative rehabilitation was introduced with combined regenerative medicine and neurorehabilitation protocols to repair neural tissue damage and create an optimized biophysical microenvironment for neural regeneration potential. With the deepening of exosome research, an increasing number of studies have found that the systemic therapeutic effects of neurorehabilitation approaches are mediated by exosomes released by physically stimulated cells, which provides new insight into rehabilitative mechanisms. Meanwhile, exosome therapy also serves as an alternative cell-free therapy of regenerative medicine that is applied in partnership with neurorehabilitation approaches and formulates exosome-based neurological regenerative rehabilitation. In this study, we review the current state of exosome-associated neurorehabilitation. On the one hand, we focus on presenting the varied mediating effects of exosomes in neurorehabilitation protocols of specific neurological pathologies; on the other hand, we discuss the diverse combinations of exosome therapies and neurorehabilitation approaches in the field of neurological regenerative rehabilitation, aiming to increase the awareness of exosome research and applications in the rehabilitation of neurological disorders.
Collapse
Affiliation(s)
- Yuan-Yi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jin Cheng
- Department of Sport Medicine, Peking University Third Hospital, Beijing, China
| | - Ya-Dong Liu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yi-Peng Wang
- Department of Orthopedics, Peking Union Medical College Hospital, Beijing, China.
| | - Qi-Wei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Nan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Henan Province, China.
| |
Collapse
|
24
|
Ye H, Wang F, Xu G, Shu F, Fan K, Wang D. Advancements in engineered exosomes for wound repair: current research and future perspectives. Front Bioeng Biotechnol 2023; 11:1301362. [PMID: 38033824 PMCID: PMC10682480 DOI: 10.3389/fbioe.2023.1301362] [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: 09/24/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
Wound healing is a complex and prolonged process that remains a significant challenge in clinical practice. Exosomes, a type of nanoscale extracellular vesicles naturally secreted by cells, are endowed with numerous advantageous attributes, including superior biocompatibility, minimal toxicity, and non-specific immunogenicity. These properties render them an exceptionally promising candidate for bioengineering applications. Recent advances have illustrated the potential of exosome therapy in promoting tissue repair. To further augment their therapeutic efficacy, the concept of engineered exosomes has been proposed. These are designed and functionally modifiable exosomes that have been tailored on the attributes of natural exosomes. This comprehensive review delineates various strategies for exosome engineering, placing specific emphasis on studies exploring the application of engineered exosomes for precision therapy in wound healing. Furthermore, this review sheds light on strategies for integrating exosomes with biomaterials to enhance delivery effectiveness. The insights presented herein provide novel perspectives and lay a robust foundation for forthcoming research in the realm of cutaneous wound repair therapies.
Collapse
Affiliation(s)
- Hailian Ye
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Wang
- Department of Burn and Plastic Surgery, Department of Wound Repair, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Guangchao Xu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Feihong Shu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| | - Kunwu Fan
- Department of Burn and Plastic Surgery, Department of Wound Repair, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Dali Wang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
25
|
Kmiotek-Wasylewska K, Bobis-Wozowicz S, Karnas E, Orpel M, Woźnicka O, Madeja Z, Dawn B, Zuba-Surma EK. Anti-inflammatory, Anti-fibrotic and Pro-cardiomyogenic Effects of Genetically Engineered Extracellular Vesicles Enriched in miR-1 and miR-199a on Human Cardiac Fibroblasts. Stem Cell Rev Rep 2023; 19:2756-2773. [PMID: 37700183 PMCID: PMC10661813 DOI: 10.1007/s12015-023-10621-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
RATIONALE Emerging evidence indicates that stem cell (SC)- derived extracellular vesicles (EVs) carrying bioactive miRNAs are able to repair damaged or infarcted myocardium and ameliorate adverse remodeling. Fibroblasts represent a major cell population responsible for scar formation in the damaged heart. However, the effects of EVs on cardiac fibroblast (CFs) biology and function has not been investigated. OBJECTIVE To analyze the biological impact of stem cell-derived EVs (SC-EVs) enriched in miR-1 and miR-199a on CFs and to elucidate the underlying molecular mechanisms. METHODS AND RESULTS Genetically engineered human induced pluripotent stem cells (hiPS) and umbilical cord-derived mesenchymal stem cells (UC-MSCs) expressing miR-1 or miR-199a were used to produce miR-EVs. Cells and EVs were thoughtfully analyzed for miRNA expression using RT-qPCR method. Both hiPS-miRs-EVs and UC-MSC-miRs-EVs effectively transferred miRNAs to recipient CFs, however, hiPS-miRs-EVs triggered cardiomyogenic gene expression in CFs more efficiently than UC-MSC-miRs-EVs. Importantly, hiPS-miR-1-EVs exhibited cytoprotective effects on CFs by reducing apoptosis, decreasing levels of pro-inflammatory cytokines (CCL2, IL-1β, IL-8) and downregulating the expression of a pro-fibrotic gene - α-smooth muscle actin (α-SMA). Notably, we identified a novel role of miR-199a-3p delivered by hiPS-EVs to CFs, in triggering the expression of cardiomyogenic genes (NKX2.5, TNTC, MEF2C) and ion channels involved in cardiomyocyte contractility (HCN2, SCN5A, KCNJ2, KCND3). By targeting SERPINE2, miR-199a-3p may reduce pro-fibrotic properties of CFs, whereas miR-199a-5p targeted BCAM and TSPAN6, which may be implicated in downregulation of inflammation. CONCLUSIONS hiPS-EVs carrying miR-1 and miR-199a attenuate apoptosis and pro-fibrotic and pro-inflammatory activities of CFs, and increase cardiomyogenic gene expression. These finding serve as rationale for targeting fibroblasts with novel EV-based miRNA therapies to improve heart repair after myocardial injury.
Collapse
Affiliation(s)
- Katarzyna Kmiotek-Wasylewska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Sylwia Bobis-Wozowicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Monika Orpel
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Olga Woźnicka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, 1701 W Charleston Blvd., Las Vegas, NV, 89102, USA
| | - Ewa K Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland.
| |
Collapse
|
26
|
Xue C, Ma X, Guan X, Feng H, Zheng M, Yang X. Small extracellular vesicles derived from umbilical cord mesenchymal stem cells repair blood-spinal cord barrier disruption after spinal cord injury through down-regulation of Endothelin-1 in rats. PeerJ 2023; 11:e16311. [PMID: 37927780 PMCID: PMC10624166 DOI: 10.7717/peerj.16311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023] Open
Abstract
Spinal cord injury could cause irreversible neurological dysfunction by destroying the blood-spinal cord barrier (BSCB) and allowing blood cells like neutrophils and macrophages to infiltrate the spinal cord. Small extracellular vesicles (sEVs) derived from mesenchymal stem cells (MSCs) found in the human umbilical cord have emerged as a potential therapeutic alternative to cell-based treatments. This study aimed to investigate the mechanism underlying the alterations in the BSCB permeability by human umbilical cord MSC-derived sEVs (hUC-MSCs-sEVs) after SCI. First, we used hUC-MSCs-sEVs to treat SCI rat models, demonstrating their ability to inhibit BSCB permeability damage, improve neurological repair, and reduce SCI-induced upregulation of prepro-endothelin-1 (prepro-ET-1) mRNA and endothelin-1 (ET-1) peptide expression. Subsequently, we confirmed that hUC-MSCs-sEVs could alleviate cell junction destruction and downregulate MMP-2 and MMP-9 expression after SCI, contributing to BSCB repair through ET-1 inhibition. Finally, we established an in vitro model of BSCB using human brain microvascular endothelial cells and verified that hUC-MSCs-sEVs could increase the expression of junction proteins in endothelial cells after oxygen-glucose deprivation by ET-1 downregulation. This study indicates that hUC-MSCs-sEVs could help maintain BSCB's structural integrity and promote functional recovery by suppressing ET-1 expression.
Collapse
Affiliation(s)
- Chenhui Xue
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xun Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoming Guan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haoyu Feng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingkui Zheng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xihua Yang
- Laboratory Animal Center, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
27
|
Zhang X, Wang J, Zhang J, Tan Y, Li Y, Peng Z. Exosomes Highlight Future Directions in the Treatment of Acute Kidney Injury. Int J Mol Sci 2023; 24:15568. [PMID: 37958550 PMCID: PMC10650293 DOI: 10.3390/ijms242115568] [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: 09/20/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Acute kidney injury (AKI) is a severe health problem associated with high morbidity and mortality rates. It currently lacks specific therapeutic strategies. This review focuses on the mechanisms underlying the actions of exosomes derived from different cell sources, including red blood cells, macrophages, monocytes, mesenchymal stem cells, and renal tubular cells, in AKI. We also investigate the effects of various exosome contents (such as miRNA, lncRNA, circRNA, mRNA, and proteins) in promoting renal tubular cell regeneration and angiogenesis, regulating autophagy, suppressing inflammatory responses and oxidative stress, and preventing fibrosis to facilitate AKI repair. Moreover, we highlight the interactions between macrophages and renal tubular cells through exosomes, which contribute to the progression of AKI. Additionally, exosomes and their contents show promise as potential biomarkers for diagnosing AKI. The engineering of exosomes has improved their clinical potential by enhancing isolation and enrichment, target delivery to injured renal tissues, and incorporating small molecular modifications for clinical use. However, further research is needed to better understand the specific mechanisms underlying exosome actions, their delivery pathways to renal tubular cells, and the application of multi-omics research in studying AKI.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (X.Z.); (J.W.); (J.Z.); (Y.T.)
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Jing Wang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (X.Z.); (J.W.); (J.Z.); (Y.T.)
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Jing Zhang
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (X.Z.); (J.W.); (J.Z.); (Y.T.)
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Yuwei Tan
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (X.Z.); (J.W.); (J.Z.); (Y.T.)
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (X.Z.); (J.W.); (J.Z.); (Y.T.)
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, Wuhan 430071, China; (X.Z.); (J.W.); (J.Z.); (Y.T.)
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan 430071, China
- Department of Critical Care Medicine, Center of Critical Care Nephrology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
28
|
van de Wakker SI, Meijers FM, Sluijter JPG, Vader P. Extracellular Vesicle Heterogeneity and Its Impact for Regenerative Medicine Applications. Pharmacol Rev 2023; 75:1043-1061. [PMID: 37280097 DOI: 10.1124/pharmrev.123.000841] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/20/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-enclosed particles that are involved in physiologic and pathologic processes. EVs are increasingly being studied for therapeutic applications in the field of regenerative medicine. Therapeutic application of stem cell-derived EVs has shown great potential to stimulate tissue repair. However, the exact mechanisms through which they induce this effect have not been fully clarified. This may to a large extent be attributed to a lack of knowledge on EV heterogeneity. Recent studies suggest that EVs represent a heterogeneous population of vesicles with distinct functions. The heterogeneity of EVs can be attributed to differences in their biogenesis, and as such, they can be classified into distinct populations that can then be further subcategorized into various subpopulations. A better understanding of EV heterogeneity is crucial for elucidating their mechanisms of action in tissue regeneration. This review provides an overview of the latest insights on EV heterogeneity related to tissue repair, including the different characteristics that contribute to such heterogeneity and the functional differences among EV subtypes. It also sheds light on the challenges that hinder clinical translation of EVs. Additionally, innovative EV isolation techniques for studying EV heterogeneity are discussed. Improved knowledge of active EV subtypes would promote the development of tailored EV therapies and aid researchers in the translation of EV-based therapeutics to the clinic. SIGNIFICANCE STATEMENT: Within this review we discuss the differences in regenerative properties of extracellular vesicle (EV) subpopulations and implications of EV heterogeneity for development of EV-based therapeutics. We aim to provide new insights into which aspects are leading to heterogeneity in EV preparations and stress the importance of EV heterogeneity studies for clinical applications.
Collapse
Affiliation(s)
- Simonides Immanuel van de Wakker
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| | - Fleur Michelle Meijers
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| | - Joost Petrus Gerardus Sluijter
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| | - Pieter Vader
- Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (S.I.V.D.W., F.M.M., J.P.G.S., P.V.) and CDL Research, University Medical Center Utrecht, The Netherlands (P.V.)
| |
Collapse
|
29
|
Chen WY, Xiao X, Pan C, Huang FH, Xu HY, Wei QJ, Jiang H. Scientific publications on orthopedic surgery from three major East Asian countries (2012-2021). World J Orthop 2023; 14:641-650. [PMID: 37662667 PMCID: PMC10473913 DOI: 10.5312/wjo.v14.i8.641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/08/2023] [Accepted: 07/07/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND East Asia is the most dynamic region in the world and includes three major countries: Japan, South Korea and China. Due to rapid economic growth, orthopedics research in East Asia has achieved great advances during the past 10 years. However, the current status of orthopedic research in Japan, South Korea and China is still unclear. AIM To understand the current status of orthopedic research in Japan, South Korea, and China. METHODS Journals listed in the ''Orthopedics'' category of Science Citation Index Expanded subject categories were included. The PubMed and Web of Knowledge electronic databases were searched to identify scientific publications from the selected journals written by researchers from Japan, South Korea and China. A systematic analysis was conducted to analyze orthopedic research articles published in the three countries based on the number of articles, study design, impact factors (IFs) and citations. Furthermore, we also ranked the top 10 countries worldwide with the highest publications in the past 10 years. Additionally, we ranked the top 10 countries with the highest number of publications in the world in the past 10 years. Statistical analyses were performed using SPSS 20.0 software (SPSS Inc., Chicago, IL, United States), and statistical results are given in Tables and Figures. The Kruskal-Wallis test and the Mann-Whitney test were used to detect differences between countries. The tendency regarding the number of articles was analyzed by curvilinear regression. A two-tailed P < 0.05 was considered significant. RESULTS From 2012-2021, a total of 144518 articles were published in the 86 selected orthopedic journals. During this period, the number of worldwide published orthopedic articles has shown an annual increasing trend. A total of 27164 orthopedic research articles were published by Japan, South Korea and China during the past 10 years; 44.32% were from China, 32.98% were from Japan, and 22.70% were from South Korea. From 2012 to 2021, the annual number of articles markedly increased in each of the three countries. Over time, the worldwide share of articles increased substantially in South Korea (3.37% to 6.53%, P < 0.001) and China (5.29% to 9.61%, P < 0.001). However, the worldwide share of articles significantly decreased in Japan (5.22% to 3.80%, P < 0.001). The annual total IFs of articles from China were well above those of articles from Japan and South Korea (36597.69 vs 27244.48 vs 20657.83, P < 0.05). There was no significant difference among the articles in the top 10 high-IF orthopedics journals published from those three countries [South Korea (800) > China (787) > Japan (646), P > 0.05]. CONCLUSION Over the past 10 years, China's scientific publications in orthopedic journals have shown an increasing trend. Considering the relative scale of the populations, Japan and South Korea have outpaced China with respect to quality.
Collapse
Affiliation(s)
- Wei-You Chen
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Xin Xiao
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Cheng Pan
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Fei-Hong Huang
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hong-Yuan Xu
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Qing-Jun Wei
- Department of Orthopedic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Hua Jiang
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
30
|
Kandeel M, Morsy MA, Alkhodair KM, Alhojaily S. Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities. Biomolecules 2023; 13:1250. [PMID: 37627315 PMCID: PMC10452295 DOI: 10.3390/biom13081250] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a type of versatile adult stem cells present in various organs. These cells give rise to extracellular vesicles (EVs) containing a diverse array of biologically active elements, making them a promising approach for therapeutics and diagnostics. This article examines the potential therapeutic applications of MSC-derived EVs in addressing neurodegenerative disorders such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Furthermore, the present state-of-the-art for MSC-EV-based therapy in AD, HD, PD, ALS, and MS is discussed. Significant progress has been made in understanding the etiology and potential treatments for a range of neurodegenerative diseases (NDs) over the last few decades. The contents of EVs are carried across cells for intercellular contact, which often results in the control of the recipient cell's homeostasis. Since EVs represent the therapeutically beneficial cargo of parent cells and are devoid of many ethical problems connected with cell-based treatments, they offer a viable cell-free therapy alternative for tissue regeneration and repair. Developing innovative EV-dependent medicines has proven difficult due to the lack of standardized procedures in EV extraction processes as well as their pharmacological characteristics and mechanisms of action. However, recent biotechnology and engineering research has greatly enhanced the content and applicability of MSC-EVs.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Mohamed A. Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
| | - Khalid M. Alkhodair
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Sameer Alhojaily
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| |
Collapse
|
31
|
Sun Y, Sun F, Xu W, Qian H. Engineered Extracellular Vesicles as a Targeted Delivery Platform for Precision Therapy. Tissue Eng Regen Med 2023; 20:157-175. [PMID: 36637750 PMCID: PMC10070595 DOI: 10.1007/s13770-022-00503-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 01/14/2023] Open
Abstract
Extracellular vesicles (EVs)-based cell-free strategy has shown therapeutic potential in tissue regeneration. Due to their important roles in intercellular communications and their natural ability to shield cargos from degradation, EVs are also emerged as novel delivery vehicles for various bioactive molecules and drugs. Accumulating studies have revealed that EVs can be modified to enhance their efficacy and specificity for the treatment of many diseases. Engineered EVs are poised as the next generation of targeted delivery platform in the field of precision therapy. In this review, the unique properties of EVs are overviewed in terms of their biogenesis, contents, surface features and biological functions, and the recent advances in the strategies of engineered EVs construction are summarized. Additionally, we also discuss the potential applications of engineered EVs in targeted therapy of cancer and damaged tissues, and evaluate the opportunities and challenges for translating them into clinical practice.
Collapse
Affiliation(s)
- Yuntong Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Fengtian Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
32
|
Tu J, Zeng Y, An R, Sun J, Wen H. Engineered nanovesicles from stromal vascular fraction promote angiogenesis and adipogenesis inside decellularized adipose tissue through encapsulating growth factors. Sci Rep 2023; 13:750. [PMID: 36639385 PMCID: PMC9839776 DOI: 10.1038/s41598-022-27176-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Acellular matrix is a commonly used biomaterial in the field of biomedical engineering and revascularization is the key process to affect the effect of acellular matrix on tissue regeneration. The application of bioactive factors related to angiogenesis has been popular in the regulation of revascularization, but the immune system clearance, uncontrollable systemic reactions, and other factors make this method face challenges. Recent reports showed that engineered cells into nanovesicles can reorganize cell membranes and encapsulate cellular active factors, extending the in vitro preservation of cytokines. However, the problems of exogenous biological contamination and tumorigenicity restricted the clinical transformation and wide application of this method. Here, we for the first time engineer stromal vascular fraction (SVF) which is extracted from fat into nanovesicles (SVF-EVs) for angiogenesis in the acellular matrix. SVF-EVs not only promote the migration of vascular endothelial cells in vitro, but also facilitate the lipogenic differentiation of mesenchymal stem cells. In vivo, SVF-EVs enhanced the retention of decellularized adipose tissue after transplanting to the subcutaneous area of nude mice. Immunofluorescence staining further showed that SVF-EVs promoted the formation of vascular networks with large lumen diameter in the grafted acellular matrix, accompanied by adipocyte regeneration peripherally. These findings reveal that SVF-EVs can be a viable method for accelerating revascularization in acellular matrix, and this process of squeezing tissue into nanovesicles shows the potential for rapid clinical transformation.
Collapse
Affiliation(s)
- Jun Tu
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yuyang Zeng
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran An
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huicai Wen
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, China.
| |
Collapse
|
33
|
Chen X, Li H, Song H, Wang J, Zhang X, Han P, Wang X. Meet changes with constancy: Defence, antagonism, recovery, and immunity roles of extracellular vesicles in confronting SARS-CoV-2. J Extracell Vesicles 2022; 11:e12288. [PMID: 36450704 PMCID: PMC9712136 DOI: 10.1002/jev2.12288] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has wrought havoc on the world economy and people's daily lives. The inability to comprehensively control COVID-19 is due to the difficulty of early and timely diagnosis, the lack of effective therapeutic drugs, and the limited effectiveness of vaccines. The body contains billions of extracellular vesicles (EVs), which have shown remarkable potential in disease diagnosis, drug development, and vaccine carriers. Recently, increasing evidence has indicated that EVs may participate or assist the body in defence, antagonism, recovery and acquired immunity against SARS-CoV-2. On the one hand, intercepting and decrypting the general intelligence carried in circulating EVs from COVID-19 patients will provide an important hint for diagnosis and treatment; on the other hand, engineered EVs modified by gene editing in the laboratory will amplify the effectiveness of inhibiting infection, replication and destruction of ever-mutating SARS-CoV-2, facilitating tissue repair and making a better vaccine. To comprehensively understand the interaction between EVs and SARS-CoV-2, providing new insights to overcome some difficulties in the diagnosis, prevention and treatment of COVID-19, we conducted a rounded review in this area. We also explain numerous critical challenges that these tactics face before they enter the clinic, and this work will provide previous 'meet change with constancy' lessons for responding to future similar public health disasters. Extracellular vesicles (EVs) provide a 'meet changes with constancy' strategy to combat SARS-CoV-2 that spans defence, antagonism, recovery, and acquired immunity. Targets for COVID-19 diagnosis, therapy, and prevention of progression may be found by capture of the message decoding in circulating EVs. Engineered and biomimetic EVs can boost effects of the natural EVs, especially anti-SARS-CoV-2, targeted repair of damaged tissue, and improvement of vaccine efficacy.
Collapse
Affiliation(s)
- Xiaohang Chen
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
- Fujian Key Laboratory of Oral Diseases, School and Hospital of StomatologyFujian Medical UniversityFuzhouChina
| | - Huifei Li
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Haoyue Song
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Jie Wang
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Xiaoxuan Zhang
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Pengcheng Han
- CAS Key Laboratory of Pathogen Microbiology and ImmunologyInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
- School of MedicineZhongda Hospital, Southeast UniversityNanjingChina
| | - Xing Wang
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| |
Collapse
|
34
|
Birjandi AA, Sharpe P. Potential of extracellular space for tissue regeneration in dentistry. Front Physiol 2022; 13:1034603. [DOI: 10.3389/fphys.2022.1034603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/24/2022] [Indexed: 11/19/2022] Open
Abstract
With the proven relationship between oral and general health and the growing aging population, it is pivotal to provide accessible therapeutic approaches to regenerate oral tissues and restore clinical function. However, despite sharing many core concepts with medicine, dentistry has fallen behind the progress in precision medicine and regenerative treatments. Stem cell therapies are a promising avenue for tissue regeneration, however, ethical, safety and cost issues may limit their clinical use. With the significance of paracrine signalling in stem cell and tissue regeneration, extracellular space comprising of the cell secretome, and the extracellular matrix can serve as a potent source for tissue regeneration. Extravesicles are secreted and naturally occurring vesicles with biologically active cargo that can be harvested from the extracellular space. These vesicles have shown great potential as disease biomarkers and can be used in regenerative medicine. As a cell free therapy, secretome and extracellular vesicles can be stored and transferred easily and pose less ethical and safety risks in clinical application. Since there are currently many reviews on the secretome and the biogenesis, characterization and function of extracellular vesicles, here we look at the therapeutic potential of extracellular space to drive oral tissue regeneration and the current state of the field in comparison to regenerative medicine.
Collapse
|
35
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Potential Therapy for Diabetes Mellitus and Diabetic Complications. Pharmaceutics 2022; 14:pharmaceutics14102208. [PMID: 36297643 PMCID: PMC9607185 DOI: 10.3390/pharmaceutics14102208] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 12/02/2022] Open
Abstract
As a novel cell-free strategy, mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) inherit the therapeutic potential of donor cells, and are widely used for the treatment of many diseases. Increasing studies have shown that MSC-EVs transfer various bioactive molecules to create a beneficial microenvironment, thus exerting protective roles in diabetic mellitus (DM) and diabetic complications. To overcome the limitations of natural MSC-EVs such as heterogeneity and insufficient function, several modification methods have been established for constructing engineered MSC-EVs with elevated repairing efficiency. In this review, the PubMed library was searched from inception to August 2022, using a combination of Medical Subject Headings (MeSH) and keywords related to MSC-EVs, DM, and diabetic complications. We provide an overview of the major characteristics of MSC-EVs and summarize the recent advances of MSC-EV-based therapy for hyperglycemia-induced tissue damage with an emphasis on MSC-EV-mediated delivery of functional components. Moreover, the potential applications of engineered MSC-EVs in DM-related diseases therapy are discussed by presenting examples, and the opportunities and challenges for the clinical translation of MSC-EVs, especially engineered MSC-EVs, are evaluated.
Collapse
|
36
|
Cheng J, Sun Y, Ma Y, Ao Y, Hu X, Meng Q. Engineering of MSC-Derived Exosomes: A Promising Cell-Free Therapy for Osteoarthritis. MEMBRANES 2022; 12:membranes12080739. [PMID: 36005656 PMCID: PMC9413347 DOI: 10.3390/membranes12080739] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is characterized by progressive cartilage degeneration with increasing prevalence and unsatisfactory treatment efficacy. Exosomes derived from mesenchymal stem cells play an important role in alleviating OA by promoting cartilage regeneration, inhibiting synovial inflammation and mediating subchondral bone remodeling without the risk of immune rejection and tumorigenesis. However, low yield, weak activity, inefficient targeting ability and unpredictable side effects of natural exosomes have limited their clinical application. At present, various approaches have been applied in exosome engineering to regulate their production and function, such as pretreatment of parental cells, drug loading, genetic engineering and surface modification. Biomaterials have also been proved to facilitate efficient delivery of exosomes and enhance treatment effectiveness. Here, we summarize the current understanding of the biogenesis, isolation and characterization of natural exosomes, and focus on the large-scale production and preparation of engineered exosomes, as well as their therapeutic potential in OA, thus providing novel insights into exploring advanced MSC-derived exosome-based cell-free therapy for the treatment of OA.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Yixin Sun
- Peking Unversity First Hospital, Peking University Health Science Center, Beijing 100034, China;
| | - Yong Ma
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
- Correspondence: (X.H.); (Q.M.); Tel.: +86-010-8226-5680 (Q.M.)
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
- Correspondence: (X.H.); (Q.M.); Tel.: +86-010-8226-5680 (Q.M.)
| |
Collapse
|
37
|
Therapeutic Strategy of Mesenchymal-Stem-Cell-Derived Extracellular Vesicles as Regenerative Medicine. Int J Mol Sci 2022; 23:ijms23126480. [PMID: 35742923 PMCID: PMC9224400 DOI: 10.3390/ijms23126480] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer membrane particles that play critical roles in intracellular communication through EV-encapsulated informative content, including proteins, lipids, and nucleic acids. Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal ability derived from bone marrow, fat, umbilical cord, menstruation blood, pulp, etc., which they use to induce tissue regeneration by their direct recruitment into injured tissues, including the heart, liver, lung, kidney, etc., or secreting factors, such as vascular endothelial growth factor or insulin-like growth factor. Recently, MSC-derived EVs have been shown to have regenerative effects against various diseases, partially due to the post-transcriptional regulation of target genes by miRNAs. Furthermore, EVs have garnered attention as novel drug delivery systems, because they can specially encapsulate various target molecules. In this review, we summarize the regenerative effects and molecular mechanisms of MSC-derived EVs.
Collapse
|