1
|
Feng K, Liu J, Gong L, Ye T, Chen Z, Wang Y, Li Q, Xie X. Engineered MSC-sEVs as a Versatile Nanoplatform for Enhanced Osteoarthritis Treatment via Targeted Elimination of Senescent Chondrocytes and Maintenance of Cartilage Matrix Metabolic Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2413759. [PMID: 39755936 DOI: 10.1002/advs.202413759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/17/2024] [Indexed: 01/06/2025]
Abstract
Chondrocyte senescence is an important pathogenic factor causing osteoarthritis (OA) progression through persistently producing pro-inflammatory factors. Mesenchymal stem cells-derived small extracellular vesicles (MSC-sEVs) have shown anti-inflammatory effects in OA models, while persistent existence of senescent chondrocytes still promotes cartilage destruction. Therefore, improving the targeted elimination ability on senescent chondrocytes is required to facilitate the translation of MSC-sEVs in OA treatment. In this study, versatile engineered MSC-sEVs are developed to targetedly clear senescent chondrocytes and maintain cartilage metabolic homeostasis. Specifically, MSC-sEVs are loaded with siRNA mouse double minute 2 homologue (siMDM2) and modified with cartilage-targeting peptide WYRGRL-PEG2K-DSPE (WPD), named WPD-sEVssiMDM2. The results demonstrate versatile modification improves the cellular uptake of MSC-sEVs in chondrocytes, and thus improves the antiaging effects. Importantly, multifunctional modification enhances cartilage penetration ability and extends joint retention time of MSC-sEVs. In both post-traumatic OA mice and naturally aged mice, WPD-sEVssiMDM2 more effectively eliminates senescent chondrocytes and maintained matrix metabolic homeostasis. By using the P53 phosphorylation inhibitor, the essential role MDM2-P53 pathway in the antiaging function of WPD-sEVssiMDM2 on chondrocytes is verified. In ex vivo cultured human OA cartilage explants, it is confirmed that WPD-sEVssiMDM2 alleviates senescent phenotype. Altogether, the findings suggest that WPD-sEVssiMDM2 have promising translational potential for OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiashuo Liu
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liangzhi Gong
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Teng Ye
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhengsheng Chen
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qing Li
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xuetao Xie
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
2
|
Chen M, Wang J, Liu Y, Cui X, Liang J, Lei N, Xiao Y, Wang Q, Fan Y, Zhang X. Injectable biomimetic microcarriers harness synergistic effects of paracrine factors and cellular membranes to alleviate osteoarthritis. CHEMICAL ENGINEERING JOURNAL 2025; 503:158451. [DOI: 10.1016/j.cej.2024.158451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2024; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [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: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
4
|
Li B, Chen H, Hang R. Osseointegration-Related Exosomes for Surface Functionalization of Titanium Implants. Biomater Res 2024; 28:0124. [PMID: 39711824 PMCID: PMC11661649 DOI: 10.34133/bmr.0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Despite that the clinical application of titanium-based implants has achieved great success, patients' own diseases and/or unhealthy lifestyle habits often lead to implant failure. Many studies have been carried out to modify titanium implants to promote osseointegration and implant success. Recent studies showed that exosomes, proactively secreted extracellular vesicles by mammalian cells, could selectively target and modulate the functions of recipient cells such as macrophages, nerve cells, endothelial cells, and bone marrow mesenchymal stem cells that are closely involved in implant osseointegration. Accordingly, using exosomes to functionalize titanium implants has been deemed as a novel and effective way to improve their osseointegration ability. Herein, recent advances pertaining to surface functionalization of titanium implants with exosomes are analyzed and discussed, with focus on the role of exosomes in regulating the functions of osseointegration-related cells, and their immobilization strategies as well as resultant impact on osseointegration ability.
Collapse
Affiliation(s)
- Boqiong Li
- Department of Materials Science and Engineering,
Jinzhong University, Jinzhong 030619, China
| | - Huanming Chen
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering,
Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
5
|
Mohd Noor NA, Jun Quan N, Mazlan NAAA, Nurul AA, Ahmad Mohd Zain MR, Azlan M. Synovial Fluid-Derived Exosomes from Osteoarthritis Patients Modulate Cell Surface Phenotypes of Monocytes and Cytokine Secretions. Immunol Invest 2024:1-16. [PMID: 39702926 DOI: 10.1080/08820139.2024.2443244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
BACKGROUND Exosomes can be found in the synovial fluid of inflamed knee joints, which play a significant role in osteoarthritis (OA) progression. However, their role - in modulating the cellular environment within the body, particularly monocytes remain unexplored. This study aimed to evaluate the immunomodulatory effect of exosomes on monocytes. METHODS Exosomes were isolated by ultracentrifugation and characterized using nanoparticle tracking analysis (NTA), scanning electron microscopy (SEM), and Western blot. The effect of exosomes in modulating monocyte phenotypes as well as cytokine secretion were further assessed in a co-culture condition using flow cytometry and ELISA accordingly. RESULTS Exosomes were identified as spherical particles with a size distribution ranging from 30 nm to 150 nm. These nanoparticles intensely expressed exosome protein markers including CD9, CD63, CD81, and HSP70. The expression of HLA-DR, CD14, and CD11b on monocytes decreased in the presence of exosomes after 24 h of incubation, regardless of the dose. Exosomes significantly induced the release of anti-inflammatory cytokines IL-1Ra in a time- and dose-dependent manner, while TNF-α secretion remains unchanged regardless of the presence or absence of exosomes. CONCLUSION This study highlights the immunoregulatory role of exosomes on monocytes, emphasizing the need for further studies into the underlying mechanism.
Collapse
Affiliation(s)
- Nur Azira Mohd Noor
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Ng Jun Quan
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Asma Abdullah Nurul
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Maryam Azlan
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
6
|
Kong Y, Wang Y, Yang Y, Hou Y, Yu J, Liu M, Xie S, Song Y. Intra-articular injection of exosomes derived from different stem cells in animal models of osteoarthritis: a systematic review and meta- analysis. J Orthop Surg Res 2024; 19:834. [PMID: 39696589 DOI: 10.1186/s13018-024-05227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/01/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND In recent years, the increasing incidence of osteoarthritis (OA) has attracted widespread public attention; however, the available effective treatments are limited. As a result, new therapeutic approaches, including stem cell and exosome therapies, have been proposed and are gradually gaining popularity. Because exosomes are immunocompatible, there is thought to be more potential for their use in clinical settings. This study summarizes the efficacy of exosomes in the treatment of OA. METHODS In total, we conducted a comprehensive search of the PubMed, Web of Science, and Embase databases using medical subject headings terms to identify studies published from their inception until November 2023 that investigated the use of stem cell-derived exosomes in treating OA. We focused on specific outcomes including osteophyte score, chondrocyte count, pain level, qPCR and histological assessments such as the OARSI (Osteoarthritis research society international) score to measure cartilage degeneration. For data extraction, we used GetData Graph Digitizer to retrieve values from graphs, and the meta-analysis was conducted using RevMan 5.3 software. We chose mean difference (MD) as the primary effect measure since all included studies reported the same outcomes. Ultimately, 20 articles met the inclusion criteria and were included in the meta-analysis. RESULTS We evaluated 20 studies comprising a total of 400 subjects. Compared with control groups, the exosome-treated groups showed significantly improved histological outcomes, as measured by the OARSI score (n = 400; MD = -3.54; 95% CI = [-4.30, -2.79]; P < 0.00001; I2 = 98%). This indicates a marked reduction in cartilage degeneration and OA severity in the exosome-treated groups. Notably, exosome therapy was more effective when administered during the early stages of OA. Additionally, a once-weekly dosing schedule yielded better results compared to more frequent administrations. Of the three exosome isolation methods assessed, kit-based extraction demonstrated a trend toward superior therapeutic efficacy. CONCLUSIONS Exosome treatment improved OA compared to placebo treatment.
Collapse
Affiliation(s)
- Yajie Kong
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
| | - Yuzhong Wang
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
- Hebei Key Laboratory of Rare Disease, Shijiazhuang, 050000, Hebei Province, People's Republic of China
| | - Yujia Yang
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
| | - Yu Hou
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
| | - Jingjing Yu
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
| | - Meiling Liu
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
| | - Siyi Xie
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China
| | - Yongzhou Song
- Department of Orthopedics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People's Republic of China.
- Hebei Key Laboratory of Rare Disease, Shijiazhuang, 050000, Hebei Province, People's Republic of China.
- Hebei Medical University National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China.
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei Medical University, Shijiazhuang, 050017, Hebei Province, People's Republic of China.
| |
Collapse
|
7
|
Lai Q, Li B, Chen L, Zhou Y, Bao H, Li H. Substrate stiffness regulates the proliferation and inflammation of chondrocytes and macrophages through exosomes. Acta Biomater 2024:S1742-7061(24)00731-1. [PMID: 39662715 DOI: 10.1016/j.actbio.2024.12.021] [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: 08/13/2024] [Revised: 11/12/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024]
Abstract
Osteoarthritis (OA) progression is characterized by decreased cartilage stiffness and degradation of the extracellular matrix (ECM), which significantly influence cartilage behavior and fate. In contrast, processes such as chondrocyte calcification and aging often result in increased stiffness. Despite extensive studies on how ECM stiffness regulates cellular functions, the impact of substrate stiffness on the cartilage microenvironment and intercellular communications remains not well understood. Using tunable stiffness Gelatin methacryloyl (GelMA) hydrogel, we demonstrated that a potential optimal substrate stiffness can promote maximal chondrocyte proliferation and exosome secretion. The exogenous addition of stiffness-tuned exosomes induced significant changes in chondrocyte morphology, proliferation, migration, and inflammation. Notably, blocking Yes-associated protein (YAP) synthesis negated the proliferation enhancement induced by exosomes from chondrocytes cultured on medium stiffness substrates (ExoMedium), confirming that substrate stiffness regulates cell proliferation through exosomes by modulating YAP expression and its nuclear localization. Moreover, our study revealed that exosomes from medium stiffness substrates-mimicking normal cartilage stiffness-not only reduce inflammation in chondrocytes but also shift macrophage polarization from M1 to M2. Conversely, exosomes from soft stiffness substrates, akin to osteoarthritic tissue, exacerbate chondrocytes inflammation and M1 macrophage polarization. These findings highlight the crucial role of stiffness-tuned exosomes in OA progressing, affecting chondrocyte proliferation, migration, inflammation and macrophage polarization, and provide new insights into the potential novel treatment strategies using engineered scaffolds and exosomes. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by decreased cartilage stiffness and degradation of the extracellular matrix (ECM). While some studies suggest that increased substrate stiffness enhances cell proliferation, others have reported the opposite effect. Whether there exists an optimal matrix stiffness that promotes chondrocytes anabolism and how matrix stiffness regulates the cartilage microenvironment and intercellular communications remain unclear. Utilizing tunable stiffness Gelatin methacryloyl (GelMA) hydrogel, this study demonstrated that a potential optimal substrate stiffness can maximize chondrocyte proliferation and exosome secretion. The introduction of stiffness-tuned exosomes induced significant changes in chondrocyte and macrophage proliferation, migration, and inflammation, offering new insights into OA progression and highlighting their potential as a promising therapeutic strategy for osteoarthritis treatment and tissue regeneration.
Collapse
Affiliation(s)
- Qiling Lai
- Joint Research Centre on Medicine, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, PR China; NO.7 People's Hospital, Dongguan, Guangdong 523000, PR China
| | - Bo Li
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China.
| | - Linjie Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Yafen Zhou
- Joint Research Centre on Medicine, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, PR China
| | - Hongdan Bao
- Joint Research Centre on Medicine, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, PR China.
| | - Huaqiong Li
- Joint Research Centre on Medicine, Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang 315700, PR China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, PR China.
| |
Collapse
|
8
|
He D, Liu X, Yang W, Guan T, Wang G. The role of mechanosensitive ion channel Piezo1 in knee osteoarthritis inflammation. Channels (Austin) 2024; 18:2393088. [PMID: 39169878 PMCID: PMC11346567 DOI: 10.1080/19336950.2024.2393088] [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: 06/01/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Mechanosensitive ion channel Piezo1 is known to mediate a variety of inflammatory pathways and is also involved in the occurrence and development of many orthopedic diseases. Although its role in the inflammatory mechanism of knee osteoarthritis (KOA) has been reported, a systematic explanation is yet to be seen. This article aims to summarize the role of inflammatory responses in the pathogenesis of KOA and elucidate the mechanism by which the Piezo1-mediated inflammatory response contributes to the pathogenesis of KOA, providing a theoretical basis for the prevention and treatment of knee osteoarthritis. The results indicate that in the mechanism leading to knee osteoarthritis, Piezo1 can mediate the inflammatory response through chondrocytes and synovial cells, participating in the pathological progression of KOA. Piezo1 has the potential to become a new target for the prevention and treatment of this disease. Additionally, as pain is one of the most severe manifestations in KOA patients, the inflammatory response mediated by Piezo1, which causes the release of inflammatory mediators and pro-inflammatory factors leading to pain, can be further explored.
Collapse
Affiliation(s)
- Dingchang He
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Xin Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Wenhao Yang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Taiyuan Guan
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Guoyou Wang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Orthopedic Disorders, Southwest Medical University, Luzhou, China
| |
Collapse
|
9
|
Yang Q, Liu G, Chen G, Chen G, Chen K, Fan L, Tu Y, Chen J, Shi Z, Chen C, Liu S, Deng G, Deng X, Sun C, Li X, Yang S, Zheng S, Chen B. Novel injectable adhesive hydrogel loaded with exosomes for holistic repair of hemophilic articular cartilage defect. Bioact Mater 2024; 42:85-111. [PMID: 39280582 PMCID: PMC11399810 DOI: 10.1016/j.bioactmat.2024.08.018] [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: 06/07/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Hemophilic articular cartilage damage presents a significant challenge for surgeons, characterized by recurrent intraarticular bleeding, a severe inflammatory microenvironment, and limited self-repair capability of cartilage tissue. Currently, there is a lack of tissue engineering-based integrated therapies that address both early hemostasis, anti-inflammation, and long-lasting chondrogenesis for hemophilic articular cartilage defects. Herein, we developed an adhesive hydrogel using oxidized chondroitin sulfate and gelatin, loaded with exosomes derived from bone marrow stem cells (BMSCs) (Hydrogel-Exos). This hydrogel demonstrated favorable injectability, self-healing, biocompatibility, biodegradability, swelling, frictional and mechanical properties, providing a comprehensive approach to treating hemophilic articular cartilage defects. The adhesive hydrogel, featuring dynamic Schiff base bonds and hydrogen bonds, exhibited excellent wet tissue adhesiveness and hemostatic properties. In a pig model, the hydrogel could be smoothly injected into the knee joint cartilage defect site and gelled in situ under fluid-irrigated arthroscopic conditions. Our in vitro and in vivo experiments confirmed that the sustained release of exosomes yielded anti-inflammatory effects by modulating macrophage M2 polarization through the NF-κB pathway. This immunoregulatory effect, coupled with the extracellular matrix components provided by the adhesive hydrogel, enhanced chondrogenesis, promoted the cartilage repair and joint function restoration after hemophilic articular cartilage defects. In conclusion, our results highlight the significant application potential of Hydrogel-Exos for early hemostasis, immunoregulation, and long-term chondrogenesis in hemophilic patients with cartilage injuries. This innovative approach is well-suited for application during arthroscopic procedures, offering a promising solution for addressing the complex challenges associated with hemophilic articular cartilage damage.
Collapse
Affiliation(s)
- Qinfeng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Guihua Liu
- Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou, Guangdong, 516008, China
| | - Guanghao Chen
- Department of Orthopaedics, The Second Hospital of Jiaxing, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, 314000, China
| | - Guo Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Keyu Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lei Fan
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yuesheng Tu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jialan Chen
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhanjun Shi
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuan Chen
- Department of Obstetrics and Gynecology, Core Facility Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shubo Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Geyang Deng
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaoqian Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong, 510060, China
| | - Chunhan Sun
- Institute of Orthopaedics, Huizhou Central People's Hospital, Huizhou, Guangdong, 516008, China
| | - Xiaoyang Li
- Department of Vascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Shaowei Zheng
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Quality Research in Chinese Medicines, Laboratory of Drug Discovery from Natural Resources and Industrialization, School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| |
Collapse
|
10
|
Kitahara K, Ebata T, Liyile C, Nishida Y, Ogawa Y, Tokuhiro T, Shiota J, Nagano T, Takasuka TE, Endo T, Shimizu T, Alhasan H, Asano T, Takahashi D, Homan K, Onodera T, Kadoya K, Terkawi MA, Iwasaki N. Chondroprotective functions of neutrophil-derived extracellular vesicles by promoting the production of secreted frizzled-related protein 5 in cartilage. Cell Commun Signal 2024; 22:569. [PMID: 39604981 PMCID: PMC11603793 DOI: 10.1186/s12964-024-01953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common degenerative joint disease characterized by cartilage degradation and various degrees of inflammation in the synovium. Growing evidence highlights that neutrophil extracellular vesicles (EVs) play a protective role in arthritic joints by promoting the resolution of inflammation and the synthesis of proteoglycans in cartilage. However, this homeostatic function is dependent on the activation state of neutrophils and the surrounding environment/tissues. Hence, we explored the chondroprotective functions of neutrophil-derived EVs under different stimulation conditions and the underlying molecular mechanism. METHODS Human blood-derived neutrophils, murine bone marrow-derived neutrophils, C-28I2 cells and primary chondrocytes were used. Neutrophils were stimulated with different cytokines, and their EVs were isolated for chondrocyte stimulation and further subjected to RNA-sequencing analysis. Two experimental murine OA models were used, and the treatment was performed by intraarticular injections. RESULTS Conditioned medium from neutrophils stimulated with TGF-β (N-β) had the greatest inhibitory effect on the expression of catabolic factors in stimulated chondrocytes. These protective effects were not impaired when conditioned medium of N-β from AnxA1-deficient mice was used. Consistent with these results, EVs isolated from N-β significantly reduced the expression of catabolic factors in stimulated chondrocytes. Bulk RNA-seq analysis revealed that secreted frizzled-related protein 5 (SFRP5) is upregulated in N-β-EV-stimulated chondrocytes. Furthermore, recombinant SFRP5 treatment significantly reduced the expression of catabolic factors in vitro and catabolic process in experimental murine OA models. CONCLUSIONS The current study emphasizes the potential therapeutic application of neutrophils in OA and provides new knowledge on the molecular mechanisms underlying their function.
Collapse
Affiliation(s)
- Keita Kitahara
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Taku Ebata
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Chen Liyile
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yoshio Nishida
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yuki Ogawa
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Taiki Tokuhiro
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Junki Shiota
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tatsuya Nagano
- Research Faculty of Agriculture, Hokkaido University, Kita-9, Nishi-9, Kitaku, Sapporo, 060-8589, Japan
| | - Taichi E Takasuka
- Research Faculty of Agriculture, Hokkaido University, Kita-9, Nishi-9, Kitaku, Sapporo, 060-8589, Japan
| | - Tsutomu Endo
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tomohiro Shimizu
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hend Alhasan
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tsuyoshi Asano
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Kentaro Homan
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Ken Kadoya
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - M Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
11
|
Zhang X, Ma S, Naz SI, Soderblom EJ, Jain V, Aliferis C, Kraus VB. Immune System-Related Plasma Pathogenic Extracellular Vesicle Subpopulations Predict Osteoarthritis Progression. Int J Mol Sci 2024; 25:12504. [PMID: 39684216 DOI: 10.3390/ijms252312504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Certain molecules found on the surface or within the cargo of extracellular vesicles (EVs) are linked to osteoarthritis (OA) severity and progression. We aimed to identify plasma pathogenic EV subpopulations that can predict knee radiographic OA (rOA) progression. We analyzed the mass spectrometry-based proteomic data of plasma EVs and synovial fluid (SF) EVs from knee OA patients (n = 16, 50% female). The identified surface markers of interest were further evaluated in plasma EVs from an independent cohort of knee OA patients (n = 30, 47% female) using flow cytometry. A total of 199 peptides with significant correlation between plasma and SF EVs were identified. Of these, 41.7% were linked to immune system processes, 15.5% to inflammatory responses, and 16.7% to the complement system. Crucially, five previously identified knee rOA severity-indicating surface markers-FGA, FGB, FGG, TLN1, and AMBP-were confirmed on plasma EV subpopulations in an independent cohort. These markers' baseline frequencies on large plasma EVs predicted rOA progression with an AUC of 0.655-0.711. Notably, TLN1 was expressed in OA joint tissue, whereas FGA, FGB, FGG, and AMBP were predominantly liver derived. These surface markers define specific pathogenic EV subpopulations, offering potential OA prognostic biomarkers and novel therapeutic targets for disease modification.
Collapse
Affiliation(s)
- Xin Zhang
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27701, USA
| | - Sisi Ma
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Syeda Iffat Naz
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Erik J Soderblom
- Duke Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC 27701, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
| | - Constantin Aliferis
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27701, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC 27701, USA
| |
Collapse
|
12
|
Zhang S, Chen J, Cao Y, Cui Y, Zhang M, Yue C, Yang B. Divergent Proteomic Profiles and Uptake Mechanisms of Exosomes Derived from Human Dental Pulp Stem Cells, Endothelial Cells, and Fibroblasts. Mol Pharm 2024. [PMID: 39535266 DOI: 10.1021/acs.molpharmaceut.4c00911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Effective intercellular communication is crucial for tissue repair and regeneration, with exosomes playing a key role in mediating these processes by transferring proteins, lipids, and nucleic acids between cells. This study explored the mechanisms underlying the uptake of exosomes derived from human dental pulp stem cells (hDPSCs), human umbilical vein endothelial cells (HUVECs), and human fibroblasts (HFBs). Our findings revealed that hDPSCs exhibited the greatest capacity for exosome uptake across all three cell types. Moreover, exosomes originating from hDPSCs were also taken up in the highest amounts by all three cell types. Proteomic analysis uncovered significant differences in protein expression among exosomes from these different cell types, particularly in proteins related to endocytosis. Clathrin-dependent endocytosis emerged as the primary pathway for exosome uptake in hDPSCs and HUVECs, while HFBs appeared to use a different mechanism. Additionally, proteins such as fibronectin and tetraspanins were found to be highly expressed in hDPSC-derived exosomes, suggesting their potential involvement in exosome-cell interactions. This study offers new insights into exosome uptake mechanisms and highlights the potential of exosomes in advancing tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Siqi Zhang
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| | - Jun Chen
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| | - Yipu Cao
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| | - Yifan Cui
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| | - Mei Zhang
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| | - Chongxia Yue
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial and Institute of Regulatory Science for Medical Devices and NMPA Research Base of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| | - Bangcheng Yang
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, People's Republic of China
- National Engineering Research Center for Biomaterials, Chengdu 610064, People's Republic of China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, People's Republic of China
| |
Collapse
|
13
|
Jiang F, Zhao H, Zhang P, Bi Y, Zhang H, Sun S, Yao Y, Zhu X, Yang F, Liu Y, Xu S, Yu T, Xiao X. Challenges in tendon-bone healing: emphasizing inflammatory modulation mechanisms and treatment. Front Endocrinol (Lausanne) 2024; 15:1485876. [PMID: 39568806 PMCID: PMC11576169 DOI: 10.3389/fendo.2024.1485876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
Tendons are fibrous connective tissues that transmit force from muscles to bones. Despite their ability to withstand various loads, tendons are susceptible to significant damage. The healing process of tendons and ligaments connected to bone surfaces after injury presents a clinical challenge due to the intricate structure, composition, cellular populations, and mechanics of the interface. Inflammation plays a pivotal role in tendon healing, creating an inflammatory microenvironment through cytokines and immune cells that aid in debris clearance, tendon cell proliferation, and collagen fiber formation. However, uncontrolled inflammation can lead to tissue damage, and adhesions, and impede proper tendon healing, culminating in scar tissue formation. Therefore, precise regulation of inflammation is crucial. This review offers insights into the impact of inflammation on tendon-bone healing and its underlying mechanisms. Understanding the inflammatory microenvironment, cellular interactions, and extracellular matrix dynamics is essential for promoting optimal healing of tendon-bone injuries. The roles of fibroblasts, inflammatory cytokines, chemokines, and growth factors in promoting healing, inhibiting scar formation, and facilitating tissue regeneration are discussed, highlighting the necessity of balancing the suppression of detrimental inflammatory responses with the promotion of beneficial aspects to enhance tendon healing outcomes. Additionally, the review explores the significant implications and translational potential of targeted inflammatory modulation therapies in refining strategies for tendon-bone healing treatments.
Collapse
Affiliation(s)
- Fan Jiang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Haibo Zhao
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Po Zhang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yanchi Bi
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Haoyun Zhang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Shenjie Sun
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yizhi Yao
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xuesai Zhu
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Fenghua Yang
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yang Liu
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Sicong Xu
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Tengbo Yu
- Department of Orthopedic Surgery, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Xiao Xiao
- Central Laboratories, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
14
|
Feng K, Ye T, Xie X, Liu J, Gong L, Chen Z, Zhang J, Li H, Li Q, Wang Y. ESC-sEVs alleviate non-early-stage osteoarthritis progression by rejuvenating senescent chondrocytes via FOXO1A-autophagy axis but not inducing apoptosis. Pharmacol Res 2024; 209:107474. [PMID: 39433168 DOI: 10.1016/j.phrs.2024.107474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
Osteoarthritis (OA) is a common joint degenerative disease which currently lacks satisfactory disease-modifying treatments. Oxidative stress-mediated senescent chondrocytes accumulation is closely associated with OA progression, which abrogates cartilage metabolism homeostasis by secreting senescence-associated secretory phenotype (SASP) factors. Numerous studies suggested mesenchymal stem cells-derived small extracellular vesicles (MSC-sEVs) have been regarded as promising candidates for OA therapy. However, MSC-sEVs were applied before the occurrence of cartilage degeneration or at early-stage OA, while in clinical practice, most OA patients who present with pain are already in non-early-stage. Recently, embryonic stem cells-derived sEVs (ESC-sEVs) have been reported to possess powerful anti-aging effects. However, whether ESC-sEVs could attenuate non-early-stage OA progression remains unknown. In this study, we demonstrated ESC-sEVs ameliorated senescent phenotype and cartilage destruction in both mechanical stress-induced non-early-stage posttraumatic OA and naturally aged mice. More importantly, we found ESC-sEVs alleviated senescent phenotype by rejuvenating aged chondrocytes but not inducing apoptosis. We also provided evidence that the FOXO1A-autophagy axis played an important role in the anti-aging effects of ESC-sEVs. To promote clinical translation, we confirmed ESC-sEVs reversed senescent phenotype in ex-vivo cultured human end-stage OA cartilage explants. Collectively, our findings reveal that ESC-sEVs-based therapy is of high translational value in non-early-stage OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Teng Ye
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Xuetao Xie
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jiashuo Liu
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Liangzhi Gong
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zhengsheng Chen
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Juntao Zhang
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haiyan Li
- Chemical and Environmental Engineering, School of Engineering, STEM College, RMIT University, 124 La Trobe St, Melbourne, VIC 3000, Australia
| | - Qing Li
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Yang Wang
- Institute of Microsurgery on Extremities, Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
15
|
Abbas A, Almaghrbi H, Giordo R, Zayed H, Pintus G. Pathogenic mechanisms, diagnostic, and therapeutic potential of microvesicles in diabetes and its complications. Arch Biochem Biophys 2024; 761:110168. [PMID: 39349130 DOI: 10.1016/j.abb.2024.110168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
Extracellular vesicles (EVs), particularly microvesicles (MVs), have gained significant attention for their role as mediators of intercellular communication in both physiological and pathological contexts, including diabetes mellitus (DM) and its complications. This review provides a comprehensive analysis of the emerging roles of MVs in the pathogenesis of diabetes and associated complications such as nephropathy, retinopathy, cardiomyopathy, and neuropathy. MVs, through their cargo of proteins, lipids, mRNAs, and miRNAs, regulate critical processes like inflammation, oxidative stress, immune responses, and tissue remodeling, all of which contribute to the progression of diabetes and its complications. We examine the molecular mechanisms underlying MVs' involvement in these pathological processes and discuss their potential as biomarkers and therapeutic tools, particularly for drug delivery. Despite promising evidence, challenges remain in isolating and characterizing MVs, understanding their molecular mechanisms, and validating them for clinical use. Advanced techniques such as single-cell RNA sequencing and proteomics are required to gain deeper insights. Improved isolation and purification methods are essential for translating MVs into clinical applications, with potential to develop novel diagnostic and therapeutic strategies to improve patient outcomes in diabetes.
Collapse
Affiliation(s)
- Alaa Abbas
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Heba Almaghrbi
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, 505055, Dubai, United Arab Emirates; Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy
| | - Hatem Zayed
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, P.O. Box 2713, Doha, Qatar.
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100, Sassari, Italy; Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, University City Rd, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
16
|
Bolandnazar NS, Raeissadat SA, Haghighatkhah H, Rayegani SM, Oshnari RS, Keshel SH, Zahraei M, Aalipour K, Babaee M, Zamani A, Rad ZB, Soleimani M, Sefat F. Safety and efficacy of placental mesenchymal stromal cells-derived extracellular vesicles in knee osteoarthritis: a randomized, triple-blind, placebo-controlled clinical trial. BMC Musculoskelet Disord 2024; 25:856. [PMID: 39465400 PMCID: PMC11514941 DOI: 10.1186/s12891-024-07979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Knee osteoarthritis causes pain and disability in many people worldwide, for which no definitive treatment has yet been proposed. In this study, we investigated the safety and efficacy of placental mesenchymal stromal cells-derived extracellular vesicles in patients with knee osteoarthritis. METHODS This triple-blind, randomized clinical trial included patients suffering from bilateral knee osteoarthritis with grade 2 or 3. The knees of each patient were randomized to intervention and control. For the intervention knee, 5 cc of placental mesenchymal stromal cells-derived extracellular vesicles were injected, and for the control knee, 5 cc of normal saline was injected. The patients' symptoms were evaluated before the intervention and 2 and 6 months after the intervention using VAS, WOMAC questionnaire, and Lequesne index. MRI was performed before the intervention and 6 months after the intervention to evaluate retropatellar and tibiofemoral cartilage volume, medial and lateral meniscal disintegrity, ACL injury, and effusion-synovitis. RESULTS 62 knees (31 patients) were enrolled in this study. There were 31 knees as intervention and 31 knees as control. Finally, the data of 58 knees (29 patients) were analyzed, including 28 women and 1 man. The mean age of the patients was 55.38 ± 6.07 years. No statistically significant difference was detected between the two groups in clinical outcomes (including VAS, WOMAC, and Lequesne scores) before treatment and 2 and 6 months after treatment. Also, no statistically significant difference was detected between the two groups in MRI findings before treatment and 6 months after treatment. No systemic complications or severe local reactions occurred in the patients. CONCLUSION A single intra-articular injection of placental mesenchymal stromal cells-derived extracellular vesicles (5 cc, 7 × 109 particles/cc) is safe, but does not improve clinical symptoms or MRI findings in knee osteoarthritis beyond placebo. The protocol of this study was approved on 11 May 2022 with registration number IRCT20210423051054N1.
Collapse
Affiliation(s)
- Najmeh Sadat Bolandnazar
- Physical Medicine and Rehabilitation Research Center, Medical and Educational Center of Taleghani Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ahmad Raeissadat
- Physical Medicine and Rehabilitation Department, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Haghighatkhah
- Radiology Department, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mansoor Rayegani
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasa Salmani Oshnari
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Zahraei
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kianmehr Aalipour
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marzieh Babaee
- Physical Medicine and Rehabilitation Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Zamani
- Radiology Department, Shohada-e-Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK
- Interdisciplinary Research Center in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford, UK
| |
Collapse
|
17
|
Chu CH, Lee RP, Wu WT, Chen IH, Yeh KT, Wang CC. Advancing Osteoarthritis Treatment: The Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes and Biomaterial Integration. Biomedicines 2024; 12:2478. [PMID: 39595044 PMCID: PMC11591758 DOI: 10.3390/biomedicines12112478] [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: 09/30/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Osteoarthritis (OA) is a prevalent and debilitating joint disorder characterized by progressive cartilage degradation and inflammation, for which traditional treatments offer only symptomatic relief without halting disease progression. Exosomes, cell-free vesicles derived from mesenchymal stem cells, have emerged as a promising alternative therapy owing to their regenerative and anti-inflammatory properties. METHODS This review synthesizes findings from recent studies (2017-2023) on the therapeutic potential of exosomes in OA treatment, highlighting their ability to modulate the joint microenvironment, reduce inflammation, and promote cartilage repair by delivering bioactive molecules such as cytokines, growth factors, and regulatory ribonucleic acids. RESULTS We explore the integration of exosomes with biomaterials, such as hydrogels and scaffolds, to enhance their delivery and therapeutic efficacy, and we address the critical challenges associated with their clinical application, including standardization of isolation and characterization methods, scalability of production, mechanistic understanding, and long-term safety. Despite these challenges, exosome-based therapies offer several advantages over traditional and cell-based treatments, including lower immunogenicity, ease of handling, and targeted delivery of therapeutic agents to damaged tissues. CONCLUSIONS We provide an analytical perspective on the current state of exosome research in OA, emphasizing the need for standardized production methods, deeper mechanistic insights, and rigorous long-term safety assessments. Future directions should focus on optimizing delivery systems, exploring personalized medicine approaches, and conducting comparative effectiveness studies to fully realize the potential of exosome therapies for OA treatment. Addressing these gaps will be crucial for translating exosome therapies from bench to bedside and achieving a transformative impact on OA management.
Collapse
Affiliation(s)
- Chung-Hua Chu
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
- Department of Biomedical Engineering, National Taiwan University, Taipei 106216, Taiwan
| | - Ru-Ping Lee
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan; (R.-P.L.); (W.-T.W.)
| | - Wen-Tien Wu
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan; (R.-P.L.); (W.-T.W.)
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Ing-Ho Chen
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Kuang-Ting Yeh
- Institute of Medical Sciences, Tzu Chi University, Hualien 970374, Taiwan; (R.-P.L.); (W.-T.W.)
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
- Graduate Institute of Clinical Pharmacy, Tzu Chi University, Hualien 970374, Taiwan
| | - Chen-Chie Wang
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| |
Collapse
|
18
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
19
|
Deng Z, Zeng X, Lin B, Chen L, Wu J, Zheng J, Ma Y, Lyu FJ, Zheng Q. Human umbilical cord mesenchymal stem cells on treating osteoarthritis in a rabbit model: Injection strategies. Heliyon 2024; 10:e38384. [PMID: 39430502 PMCID: PMC11489144 DOI: 10.1016/j.heliyon.2024.e38384] [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: 05/19/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Human umbilical cord mesenchymal stem cells (UCMSCs) are a novel stem-cell source to treat osteoarthritis (OA). Here we investigated the therapeutic effects of UCMSCs injection strategies on knee OA in a rabbit model. Thirty OA rabbits randomly received normal saline, a single dose of 1 × 106 UCMSCs, or three injections of 1 × 106 UCMSCs at 2, 4, 6 weeks. Articular cartilages were collected after 8 weeks. Macroscopic and histological assessments indicated that intra-articular injection of UCMSCs, both single and multiple injection, significantly reduced the formation of periarticular osteophytes and articular cartilage degeneration when compared with the control. Furthermore, both UCMSCs injections increased the expression of chondrogenic markers in the articular cartilage, and reduced the levels of TNF-α and IL-6 in synovium. Micro-CT showed significant reduction of sub-chondral bone degeneration and osteophytes in the multiple-injection group compared to the control and single-injection group. Taken together, intra-articular injection of UCMSCs for OA treatment is safe and effective. Single and multiple injection of UCMSCs had comparable reparative effect on cartilage lesions, while multiple injection of UCMSCs further exerted effect on enhancing subchondral bone volume.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaoli Zeng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Bofu Lin
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lixuan Chen
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Jiwei Wu
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Jie Zheng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Zhu Y, Zhao J, Ding H, Qiu M, Xue L, Ge D, Wen G, Ren H, Li P, Wang J. Applications of plant-derived extracellular vesicles in medicine. MedComm (Beijing) 2024; 5:e741. [PMID: 39309692 PMCID: PMC11413507 DOI: 10.1002/mco2.741] [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: 05/28/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Plant-derived extracellular vesicles (EVs) are promising therapeutic agents owing to their natural abundance, accessibility, and unique biological properties. This review provides a comprehensive exploration of the therapeutic potential of plant-derived EVs and emphasizes their anti-inflammatory, antimicrobial, and tumor-inhibitory effects. Here, we discussed the advancements in isolation and purification techniques, such as ultracentrifugation and size-exclusion chromatography, which are critical for maintaining the functional integrity of these nanovesicles. Next, we investigated the diverse administration routes of EVs and carefully weighed their respective advantages and challenges related to bioavailability and patient compliance. Moreover, we elucidated the multifaceted mechanisms of action of plant-derived EVs, including their roles in anti-inflammation, antioxidation, antitumor activity, and modulation of gut microbiota. We also discussed the impact of EVs on specific diseases such as cancer and inflammatory bowel disease, highlighting the importance of addressing current challenges related to production scalability, regulatory compliance, and immunogenicity. Finally, we proposed future research directions for optimizing EV extraction and developing targeted delivery systems. Through these efforts, we envision the seamless integration of plant-derived EVs into mainstream medicine, offering safe and potent therapeutic alternatives across various medical disciplines.
Collapse
Affiliation(s)
- Yawen Zhu
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Junqi Zhao
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Haoran Ding
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Mengdi Qiu
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Lingling Xue
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Dongxue Ge
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Gaolin Wen
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Haozhen Ren
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| | - Peng Li
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalClinical College of Nanjing University of Chinese MedicineNanjingChina
| |
Collapse
|
21
|
Zhang Y, He X, Ge Z, Wang B, Ni M, Cai G. Investigating the differential therapeutic efficacy and mechanisms of human umbilical cord mesenchymal stem cells at various passages in osteoarthritis treatment. Tissue Cell 2024; 90:102499. [PMID: 39126832 DOI: 10.1016/j.tice.2024.102499] [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: 06/08/2023] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024]
Abstract
This study aimed to assess the clinical efficacy of umbilical cord mesenchymal stem cells (hUC-MSCs) from different passages (P3, P8, and P13) in the treatment of knee osteoarthritis (OA) and explore the underlying mechanisms. The hUC-MSCs from each passage were characterized and evaluated for their stemness, migration, proliferation, and marker expression. Rats with OA were treated with hUC-MSCs from each passage, and the therapeutic effects were assessed based on knee swelling, discomfort, and pathological examination of the knee joint. Co-culture experiments were conducted to examine the ability of hUC-MSCs to stimulate type II collagen synthesis and inhibit MMP13 expression in chondrocytes. Telomere length and telomerase activity of hUC-MSCs from each passage were measured to investigate the reasons for the observed differences in clinical efficacy. The results revealed that P3 and P8 hUC-MSCs exhibited superior osteogenic and chondrogenic differentiation potential compared to P13, while P13 demonstrated stronger adipogenic differentiation. The wound healing rate was significantly higher in the P3 and P8 groups compared to P13. All hUC-MSC groups expressed high levels of CD90 and CD105, indicating their mesenchymal stem cell characteristics, while CD31 and CD45 were not expressed. CD105 expression was significantly reduced in the P13 group. In the treatment of rat osteoarthritis, there were no significant differences in knee swelling, discomfort, Mankin scores, and pathological findings between P3 and P8 hUC-MSC treatments. However, there was a significant difference between the 8th and 13th passages. Co-culture experiments showed that hUC-MSCs from P3 and P8 enhanced type II collagen synthesis and reduced MMP13 expression in chondrocytes. Although no significant difference was observed between the P3 and P8 groups, a significant difference was found between the P13 and P8 groups. Telomere length analysis revealed that P13 samples had significantly shorter telomeres compared to both P3 and P8. The telomerase activity was positive in P3 and P8 hUC-MSCs, indicating no significant difference between these passages, while it was negative in P13 hUC-MSCs. In conclusion, P3 and P8 hUC-MSCs exhibited superior therapeutic potential for knee osteoarthritis compared to P13, possibly due to their enhanced differentiation capacity and telomerase activity.
Collapse
Affiliation(s)
- Yingkai Zhang
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China; Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University, Shanghai City 200032, PR China
| | - Xianwei He
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Zhe Ge
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Bingnan Wang
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Miaozhong Ni
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China
| | - Guoping Cai
- Department of Orthopaedic Surgery, Jinshan Hospital of Fudan University, Shanghai City 201508, PR China.
| |
Collapse
|
22
|
Cheung C, Tu S, Feng Y, Wan C, Ai H, Chen Z. Mitochondrial quality control dysfunction in osteoarthritis: Mechanisms, therapeutic strategies & future prospects. Arch Gerontol Geriatr 2024; 125:105522. [PMID: 38861889 DOI: 10.1016/j.archger.2024.105522] [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: 04/19/2024] [Revised: 05/25/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Osteoarthritis (OA) is a prevalent chronic joint disease characterized by articular cartilage degeneration, pain, and disability. Emerging evidence indicates that mitochondrial quality control dysfunction contributes to OA pathogenesis. Mitochondria are essential organelles to generate cellular energy via oxidative phosphorylation and regulate vital processes. Impaired mitochondria can negatively impact cellular metabolism and result in the generation of harmful reactive oxygen species (ROS). Dysfunction in mitochondrial quality control mechanisms has been increasingly linked to OA onset and progression. This review summarizes current knowledge on the role of mitochondrial quality control disruption in OA, highlighting disturbed mitochondrial dynamics, impaired mitochondrial biogenesis, antioxidant defenses and mitophagy. The review also discusses potential therapeutic strategies targeting mitochondrial Quality Control in OA, offering future perspectives on advancing OA therapeutic strategies.
Collapse
Affiliation(s)
- Chiyuen Cheung
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Shaoqin Tu
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Yi Feng
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Chuiming Wan
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Hong Ai
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Zheng Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
23
|
Wu J, Wu J, Xiang W, Gong Y, Feng D, Fang S, Wu Y, Liu Z, Li Y, Chen R, Zhang X, Li B, Chen L, Jin R, Li S, Zhang B, Zhang T, Yin L, Zhou Y, Huang S, Liu N, Xu H, Lian J, Wang Y, Zhou S, Ni Z. Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis. J Nanobiotechnology 2024; 22:555. [PMID: 39261846 PMCID: PMC11391629 DOI: 10.1186/s12951-024-02795-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. METHODS The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF-α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. RESULTS The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF-α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF-α, thereby exerting chondroprotective effects. CONCLUSION TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.
Collapse
Affiliation(s)
- Jiangyi Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Jinhui Wu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Wei Xiang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yunquan Gong
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
- Department of Orthopedics, Shanghai Hospital, Wanzhou District, Chongqing, 40400, China
| | - Daibo Feng
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Shunzheng Fang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Zheng Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yang Li
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China
| | - Xiaoqi Zhang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Bingfei Li
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Lifeng Chen
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Runze Jin
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Song Li
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400000, China
| | - Bin Zhang
- Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400000, China
- Rehabilitation Center, Strategic Support Force Xingcheng Special Duty Sanatorium, Xingcheng, 125105, China
| | - Tongyi Zhang
- Department of General Practice, Chinese PLA General Hospital of the Central Theater Command, Wuhan, 430012, China
| | - Lin Yin
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Yizhao Zhou
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Shu Huang
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Ningning Liu
- Department of Laboratory Medicine, The Fifth Clinical Medical College of, Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450003, China
| | - Hao Xu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450003, China
| | - Jiqin Lian
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Yongqian Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China.
| | - Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, People's Republic of China.
| | - Zhenhong Ni
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China.
| |
Collapse
|
24
|
Wu J, Wu J, Liu Z, Gong Y, Feng D, Xiang W, Fang S, Chen R, Wu Y, Huang S, Zhou Y, Liu N, Xu H, Zhou S, Liu B, Ni Z. Mesenchymal stem cell-derived extracellular vesicles in joint diseases: Therapeutic effects and underlying mechanisms. J Orthop Translat 2024; 48:53-69. [PMID: 39170747 PMCID: PMC11338158 DOI: 10.1016/j.jot.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/09/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024] Open
Abstract
Joint diseases greatly impact the daily lives and occupational functioning of patients globally. However, conventional treatments for joint diseases have several limitations, such as unsatisfatory efficacy and side effects, necessitating the exploration of more efficacious therapeutic strategies. Mesenchymal stem cell (MSC)-derived EVs (MSC-EVs) have demonstrated high therapeutic efficacyin tissue repair and regeneration, with low immunogenicity and tumorigenicity. Recent studies have reported that EVs-based therapy has considerable therapeutic effects against joint diseases, including osteoarthritis, tendon and ligament injuries, femoral head osteonecrosis, and rheumatoid arthritis. Herein, we review the therapeutic potential of various types of MSC-EVs in the aforementioned joint diseases, summarise the mechanisms underlying specific biological effects of MSC-EVs, and discuss future prospects for basic research on MSC-EV-based therapeutic modalities and their clinical translation. In general, this review provides an in-depth understanding of the therapeutic effects of MSC-EVs in joint diseases, as well as the underlying mechanisms, which may be beneficial to the clinical translation of MSC-EV-based treatment. The translational potential of this article: MSC-EV-based cell-free therapy can effectively promote regeneration and tissue repair. When used to treat joint diseases, MSC-EVs have demonstrated desirable therapeutic effects in preclinical research. This review may supplement further research on MSC-EV-based treatment of joint diseases and its clinical translation.
Collapse
Affiliation(s)
- Jinhui Wu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Jiangyi Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Zheng Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yunquan Gong
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Daibo Feng
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Wei Xiang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Shunzheng Fang
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Ran Chen
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, China
| | - Yaran Wu
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University, Gantaoyan Street, Shapinba District, Chongqing, 400038, China
| | - Shu Huang
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Yizhao Zhou
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Ningning Liu
- Department of Laboratory Medicine, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People's Hospital), Zhengzhou, 450003, China
| | - Hao Xu
- Department of Laboratory Medicine, the Third Affiliated Hospital of Zhengzhou University Zhengzhou, 450003, China
| | - Siru Zhou
- War Trauma Medical Center, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center, Daping Hospital, Army Medical University, Chongqing, 40038, China
| | - Baorong Liu
- Department of Joint Surgery and Sport Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410000, China
| | - Zhenhong Ni
- Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| |
Collapse
|
25
|
Gonçalves MO, Di Iorio JF, Marin GV, Meneghetti P, Negreiros NGS, Torrecilhas AC. Extracellular vesicles. CURRENT TOPICS IN MEMBRANES 2024; 94:1-31. [PMID: 39370203 DOI: 10.1016/bs.ctm.2024.06.008] [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: 10/08/2024]
Abstract
Cells, pathogens, and other systems release extracellular vesicles (EVs). The particles promote intercellular communication and contain proteins, lipids, RNA and DNA. Initially considered to be cellular waste in the twentieth century, EVs were becoming recognized for their function in biological communication and control. EVs are divided into many subtypes: exosomes, microvesicles, and apoptotic bodies. Exosomes form in the late endosome/multivesicular body and are released when the compartments fuse with the plasma membrane. Microvesicles are generated by direct budding of the plasma membrane, whereas apoptotic bodies are formed after cellular apoptosis. The new guideline for EVs that describes alternate nomenclature for EVs. The particles modulate the immune response by affecting both innate and adaptive immunity, and their specific the structure allows them to be used as biomarkers to diagnose a variety of diseases. EVs have a wide range of applications, for example, delivery systems for medications and genetic therapies because of their ability to convey specific cellular material. In anti-tumor therapy, EVs deliver therapeutic chemicals to tumor cells. The EVs promote transplant compatibility and reduce organ rejection. Host-parasite interactions, therapeutic and diagnostic for cancer, cardiovascular disease, cardiac tissue regeneration, and the treatment of neurological diseases such as Alzheimer's and Parkinson's. The study of EVs keeps on expanding, revealing new functions and beneficial options. EVs have the potential to change drug delivery, diagnostics, and specific therapeutics, creating a new frontier in biomedical.
Collapse
Affiliation(s)
- Mariana Ottaiano Gonçalves
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Juliana Fortes Di Iorio
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Gabriela Villa Marin
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Paula Meneghetti
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Náthani Gabrielly Silva Negreiros
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Ana Claudia Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
26
|
Mazahir F, Yadav AK. Recent progress in engineered extracellular vesicles and their biomedical applications. Life Sci 2024; 350:122747. [PMID: 38797364 DOI: 10.1016/j.lfs.2024.122747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
AIMS To present the recent update on the isolation, engineering techniques for extracellular vesicles, limitations associated with different isolation techniques, different biomedical applications, and challenges of engineered extracellular vesicles for the benefit of researchers from academic, industry, etc. MATERIALS AND METHODS: Peer-reviewed articles from most recognized journals were collected, and presented information was analyzed to discuss collection, chemical, electroporation, cellular, and membrane surface engineering to design extracellular vesicles for various therapeutic applications. In addition, we present the applications and limitations of techniques for the collection of extracellular vesicles. KEY FINDINGS There is a need for isolation techniques with the gold standard. However, advanced extracellular vesicle isolation techniques showed improved recovery, and purity of extracellular vesicles. Tumor therapy is a major part of the therapy section that illustrates the role of engineered extracellular vesicles in synergetic therapy such as phototherapy, theragnostic, and delivery of genetic materials. In addition, extracellular vesicles have shown their potential in the treatment of retinal disorders, neurodegenerative disease, tuberculosis, osteoporosis, inflammatory bowel disease, vaccine production, and wound healing. SIGNIFICANCE Engineered extracellular vesicles can deliver cargo to the specific cells, elicit an immune response and could be used for the development of the vaccines in the future. However, the progress is at the initial stage. Overall, this review will provide a comprehensive understanding and could serve as a reference for researchers in the clinical translation of engineered extracellular vesicles in different biomedical fields.
Collapse
Affiliation(s)
- Farhan Mazahir
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India.
| |
Collapse
|
27
|
Yang L, Li W, Zhao Y, Shang L. Magnetic Polysaccharide Mesenchymal Stem Cells Exosomes Delivery Microcarriers for Synergistic Therapy of Osteoarthritis. ACS NANO 2024. [PMID: 39039744 DOI: 10.1021/acsnano.4c01406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease that afflicts more than 250 million people worldwide, impairing their mobility and quality of life. However, conventional drug therapy is palliative. Exosomes (Exo), although with the potential to fundamentally repair cartilage, face challenges in their efficient enrichment and delivery. In this study, we developed magnetic polysaccharide hydrogel particles as microcarriers for synergistic therapy of OA. The microcarriers were composed of modified natural polysaccharides, hyaluronic acid (HAMA), and chondroitin sulfate (CSMA), and were generated from microfluidic electrospray in combination with a cryogelation process. Magnetic nanoparticles with spiny structures capable of capturing stem cell Exo were encapsulated within the microcarriers together with an anti-inflammatory drug diclofenac sodium (DS). The released DS and Exo from the microcarriers had a synergistic effect in alleviating the OA symptoms and promoting cartilage repair. The in vitro and in vivo results demonstrated the excellent performance of the microcarrier for OA treatment. We believe this work has potential for Exo therapy of OA and other related diseases.
Collapse
Affiliation(s)
- Lei Yang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Wenzhao Li
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Luoran Shang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
28
|
Sankaranarayanan J, Lee SC, Kim HK, Kang JY, Kuppa SS, Seon JK. Cinnamaldehyde-Treated Bone Marrow Mesenchymal-Stem-Cell-Derived Exosomes via Aqueous Two-Phase System Attenuate IL-1β-Induced Inflammation and Catabolism via Modulation of Proinflammatory Signaling Pathways. Int J Mol Sci 2024; 25:7263. [PMID: 39000370 PMCID: PMC11242605 DOI: 10.3390/ijms25137263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/21/2024] [Accepted: 06/29/2024] [Indexed: 07/16/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disorder that is distinguished by inflammation and chronic cartilage damage. Interleukin-1β (IL-1β) is a proinflammatory cytokine that plays an important role in the catabolic processes that underlie the pathogenesis of OA. In this study, we investigate the therapeutic efficacy of exosomes derived from untreated bone-marrow-derived mesenchymal stem cells (BMMSC-Exo) and those treated with cinnamaldehyde (BMMSC-CA-Exo) for preventing the in vitro catabolic effects of IL-1β on chondrocytes. We stimulated chondrocytes with IL-1β to mimic the inflammatory microenvironment of OA. We then treated these chondrocytes with BMMSC-Exo and BMMSC-CA-Exo isolated via an aqueous two-phase system and evaluated their effects on the key cellular processes using molecular techniques. Our findings revealed that treatment with BMMSC-Exo reduces the catabolic effects of IL-1β on chondrocytes and alleviates inflammation. However, further studies directly comparing treatments with BMMSC-Exo and BMMSC-CA-Exo are needed to determine if CA preconditioning can provide additional anti-inflammatory benefits to the exosomes beyond those of CA preconditioning or treatment with regular BMMSC-Exo. Through a comprehensive molecular analysis, we elucidated the regulatory mechanisms underlying this protective effect. We found a significant downregulation of proinflammatory signaling pathways in exosome-infected chondrocytes, suggesting the potential modulation of the NF-κB and MAPK signaling cascades. Furthermore, our study identified the molecular cargo of BMMSC-Exo and BMMSC-CA-Exo, determining the key molecules, such as anti-inflammatory cytokines and cartilage-associated factors, that may contribute to their acquisition of chondroprotective properties. In summary, BMMSC-Exo and BMMSC-CA-Exo exhibit the potential as therapeutic agents for OA by antagonizing the in vitro catabolic effects of IL-1β on chondrocytes. The regulation of the proinflammatory signaling pathways and bioactive molecules delivered by the exosomes suggests a multifaceted mechanism of action. These findings highlight the need for further investigation into exosome-based therapies for OA and joint-related diseases.
Collapse
Affiliation(s)
- Jaishree Sankaranarayanan
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.S.); (S.S.K.)
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Seok Cheol Lee
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Hyung Keun Kim
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Ju Yeon Kang
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Sree Samanvitha Kuppa
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.S.); (S.S.K.)
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| | - Jong Keun Seon
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun 58128, Republic of Korea; (J.S.); (S.S.K.)
- Department of Orthopaedic Surgery, Center for Joint Disease of Chonnam National University Hwasun Hospital, 322 Seoyang-ro, Hwasun-eup 519763, Republic of Korea; (S.C.L.); (H.K.K.); (J.Y.K.)
- Korea Biomedical Materials and Devices Innovation Research Center, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 501757, Republic of Korea
| |
Collapse
|
29
|
Yang J, Gong X, Li T, Xia Z, He R, Song X, Wang X, Wu J, Chen J, Wang F, Xiong R, Lin Y, Chen G, Yang L, Cai K. Tantalum Particles Promote M2 Macrophage Polarization and Regulate Local Bone Metabolism via Macrophage-Derived Exosomes Influencing the Fates of BMSCs. Adv Healthc Mater 2024; 13:e2303814. [PMID: 38497832 DOI: 10.1002/adhm.202303814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/07/2024] [Indexed: 03/19/2024]
Abstract
In this study, the regulatory role and mechanisms of tantalum (Ta) particles in the bone tissue microenvironment are explored. Ta particle deposition occurs in both clinical samples and animal tissues following porous Ta implantation. Unlike titanium (Ti) particles promoting M1 macrophage (Mϕ) polarization, Ta particles regulating calcium signaling pathways and promoting M2 Mϕ polarization. Ta-induced M2 Mϕ enhances bone marrow-derived mesenchymal stem cells (BMSCs) proliferation, migration, and osteogenic differentiation through exosomes (Exo) by upregulating miR-378a-3p/miR-221-5p and downregulating miR-155-5p/miR-212-5p. Ta particles suppress the pro-inflammatory and bone resorption effects of Ti particles in vivo and in vitro. In a rat femoral condyle bone defect model, artificial bone loaded with Ta particles promotes endogenous Mϕ polarization toward M2 differentiation at the defect site, accelerating bone repair. In conclusion, Ta particles modulate Mϕ polarization toward M2 and influence BMSCs osteogenic capacity through Exo secreted by M2 Mϕ, providing insights for potential bone repair applications.
Collapse
Affiliation(s)
- Junjun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Xiaoyuan Gong
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Tao Li
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Zengzilu Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Rui He
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiongbo Song
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xin Wang
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jiangyi Wu
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Jiajia Chen
- Center of Biomedical Analysis, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Fangzheng Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ran Xiong
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yangjing Lin
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Guangxing Chen
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Liu Yang
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
30
|
Feng K, Wang F, Chen H, Zhang R, Liu J, Li X, Xie X, Kang Q. Cartilage progenitor cells derived extracellular vesicles-based cell-free strategy for osteoarthritis treatment by efficient inflammation inhibition and extracellular matrix homeostasis restoration. J Nanobiotechnology 2024; 22:345. [PMID: 38890638 PMCID: PMC11186174 DOI: 10.1186/s12951-024-02632-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease which currently lacks of effective agents. It is therefore urgent and necessary to seek an effective approach that can inhibit inflammation and promote cartilage matrix homeostasis. Cartilage progenitor cells (CPCs) are identified as a cell population of superficial zone in articular cartilage which possess strong migration ability, proliferative capacity, and chondrogenic potential. Recently, the application of CPCs may represent a novel cell therapy strategy for OA treatment. There is growing evidence that extracellular vesicles (EVs) are primary mediators of the benefits of stem cell-based therapy. In this study, we explored the protective effects of CPCs-derived EVs (CPCs-EVs) on IL-1β-induced chondrocytes. We found CPCs-EVs exhibited chondro-protective effects in vitro. Furthermore, our study demonstrated that CPCs-EVs promoted matrix anabolism and inhibited inflammatory response at least partially via blocking STAT3 activation. In addition, liquid chromatography-tandem mass spectrometry analysis identified 991 proteins encapsulated in CPCs-EVs. By bioinformatics analysis, we showed that STAT3 regulatory proteins were enriched in CPCs-EVs and could be transported to chondrocytes. To promoting the protective function of CPCs-EVs in vivo, CPCs-EVs were modified with cationic peptide ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) for surface charge reverse. In posttraumatic OA mice, our results showed PPD modified CPCs-EVs (PPD-EVs) effectively inhibited extracellular matrix catabolism and attenuated cartilage degeneration. Moreover, PPD-EVs down-regulated inflammatory factors expressions and reduced OA-related pain in OA mice. In ex-vivo cultured OA cartilage explants, PPD-EVs successfully promoted matrix anabolism and inhibited inflammation. Collectively, CPCs-EVs-based cell-free therapy is a promising strategy for OA treatment.
Collapse
Affiliation(s)
- Kai Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongfang Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Rui Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiashuo Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaodong Li
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xuetao Xie
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
31
|
Varela L, van de Lest CH, van Weeren PR, Wauben MH. Synovial fluid extracellular vesicles as arthritis biomarkers: the added value of lipid-profiling and integrated omics. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:276-296. [PMID: 39698533 PMCID: PMC11648409 DOI: 10.20517/evcna.2024.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/18/2024] [Accepted: 05/29/2024] [Indexed: 12/20/2024]
Abstract
Arthritis, a diverse group of inflammatory joint disorders, poses great challenges in early diagnosis and targeted treatment. Timely intervention is imperative, yet conventional diagnostic methods are not able to detect subtle early symptoms. Hence, there is an urgent need for specific biomarkers that discriminate between different arthritis forms and for early diagnosis. The pursuit of such precise diagnostic tools has prompted a growing interest in extracellular vesicles (EVs). EVs, released by cells in a regulated fashion, are detectable in body fluids, including synovial fluid (SF), which fills the joint space. They provide insights into the intricate molecular landscapes of arthritis, and this has stimulated the search for minimally invasive EV-based diagnostics. As such, the analysis of EVs in SF has become a focus for identifying EV-based biomarkers for joint disease endotyping, prognosis, and progression. EVs are composed of a lipid bilayer and a wide variety of different cargo types, of which proteins and RNAs are widely investigated. In contrast, membrane lipids of EVs, especially the abundance, presence, or absence of specific lipids and their contribution to the biological activity of EVs, are largely overlooked in EV research. Furthermore, the identification of specific combinations of different EV components acting in concert in EVs can fuel the definition of composite biomarkers. We here provide a state-of-the-art overview of the knowledge on SF-derived EVs with emphasis on lipid analysis and we give an example of the added value of integrated proteomics and lipidomics analysis in the search for composite EV-associated biomarkers.
Collapse
Affiliation(s)
- Laura Varela
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, the Netherlands
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, the Netherlands
| | - Chris H.A. van de Lest
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, the Netherlands
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, the Netherlands
| | - P. René van Weeren
- Division Equine Sciences, Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, the Netherlands
| | - Marca H.M. Wauben
- Division Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CM, the Netherlands
| |
Collapse
|
32
|
Lin Y, Wang Z, Liu S, Liu J, Zhang Z, Ouyang Y, Su Z, Chen D, Guo L, Luo T. Roles of extracellular vesicles on macrophages in inflammatory bone diseases. Mol Cell Biochem 2024; 479:1401-1414. [PMID: 37436653 DOI: 10.1007/s11010-023-04809-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
Inflammatory bone disease is a general term for a series of diseases caused by chronic inflammation, which leads to the destruction of bone homeostasis, that is, the osteolytic activity of osteoclasts increases, and the osteogenic activity of osteoblasts decreases, leading to osteolysis. Macrophages are innate immune cell with plasticity, and their polarization is related to inflammatory bone diseases. The dynamic balance of macrophages between the M1 phenotype and the M2 phenotype affects the occurrence and development of diseases. In recent years, an increasing number of studies have shown that extracellular vesicles existing in the extracellular environment can act on macrophages, affecting the progress of inflammatory diseases. This process is realized by influencing the physiological activity or functional activity of macrophages, inducing macrophages to secrete cytokines, and playing an anti-inflammatory or pro-inflammatory role. In addition, by modifying and editing extracellular vesicles, the potential of targeting macrophages can be used to provide new ideas for developing new drug carriers for inflammatory bone diseases.
Collapse
Affiliation(s)
- Yifan Lin
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Wang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shirong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaohong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiyi Zhang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuanting Ouyang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhikang Su
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ding Chen
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lvhua Guo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Tao Luo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
33
|
Yuan J, Wang Y, Huang Y, Li S, Zhang X, Wu Z, Zhao W, Zhu J, Zhang J, Huang G, Yu P, Cheng X, Wang X, Liu X, Jia J. Investigating Novel Therapeutic Approaches for Idiopathic Short Stature: Targeting siRNA and Growth Hormone Delivery to the Growth Plate Using Exosome Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309559. [PMID: 38639394 PMCID: PMC11200009 DOI: 10.1002/advs.202309559] [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: 12/07/2023] [Revised: 03/26/2024] [Indexed: 04/20/2024]
Abstract
Idiopathic short stature (ISS) is a common childhood condition with largely unknown underlying causes. Recent research highlights the role of circulating exosomes in the pathogenesis of various disorders, but their connection to ISS remains unexplored. In the experiments, human chondrocytes are cocultured with plasma exosomes from ISS patients, leading to impaired chondrocyte growth and bone formation. Elevated levels of a specific long non-coding RNA (lncRNA), ISSRL, are identified as a distinguishing factor in ISS, boasting high specificity and sensitivity. Silencing ISSRL in ISS plasma exosomes reverses the inhibition of chondrocyte proliferation and bone formation. Conversely, overexpression of ISSRL in chondrocytes impedes their growth and bone formation, revealing its mechanism of action through the miR-877-3p/GZMB axis. Subsequently, exosomes (CT-Exo-siISSRL-oeGH) with precise cartilage-targeting abilities are engineered, loaded with customized siRNA for ISSRL and growth hormone. This innovative approach offers a therapeutic strategy to address ISS by rectifying abnormal non-coding RNA expression in growth plate cartilage and delivering growth hormone with precision to promote bone growth. This research provides valuable insights into ISS diagnosis and treatment, highlighting the potential of engineered exosomes.
Collapse
Affiliation(s)
- Jinghong Yuan
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Yameng Wang
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Yanzhe Huang
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Shengqin Li
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Xiaowen Zhang
- Department of PediatricsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
| | - Zhiwen Wu
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Wenrui Zhao
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Junchao Zhu
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Junqiu Zhang
- Department of PediatricsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
| | - Guowen Huang
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Peng Yu
- Department of Endocrinology and MetabolismThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
| | - Xigao Cheng
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| | - Xinhui Wang
- Division of Gastrointestinal and Oncologic SurgeryDepartment of SurgeryMassachusetts General HospitalHarvard Medical SchoolBostonMA02114USA
| | - Xijuan Liu
- Department of PediatricsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
| | - Jingyu Jia
- Department of OrthopaedicsThe Second Affiliated Hospital of Nanchang UniversityNanchang330006P. R. China
- Institute of Orthopaedics of Jiangxi ProvinceNanchang330006P. R. China
| |
Collapse
|
34
|
Yuan Q, Yang M, Zheng H, Cai Y, Luo P, Wang X, Xu P. M2 Macrophage-Derived Extracellular Vesicles Encapsulated in Hyaluronic Acid Alleviate Osteoarthritis by Modulating Macrophage Polarization. ACS Biomater Sci Eng 2024; 10:3355-3377. [PMID: 38563817 DOI: 10.1021/acsbiomaterials.3c01833] [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: 04/04/2024]
Abstract
An imbalance between M1 and M2 macrophage polarization is critical in osteoarthritis (OA) development. We investigated the effect of M2 macrophage-derived extracellular vesicles (M2-EVs) to reprogramme macrophages from the M1 to M2 phenotype for OA treatment. M1 macrophages and mouse OA models were treated with M2-EVs. Proteomic analysis was performed to evaluate macrophage polarization in vitro. The OA models were as follows: destabilization of the medial meniscus (DMM) surgery-induced OA and collagenase-induced OA (CIOA). Hyaluronic acid (HA) was used to deliver M2-EVs. M2-EVs decreased macrophage accumulation, repolarized macrophages from the M1 to M2 phenotype, mitigated synovitis, reduced cartilage degradation, alleviated subchondral bone damage, and improved gait abnormalities in the CIOA and DMM models. Moreover, HA increased the retention time of M2-EVs and enhanced the efficiency of M2-EVs in OA treatment. Furthermore, proteomic analysis demonstrated that M2-EVs exhibited a macrophage reprogramming ability similar to IL-4, and the pathways might be the NOD-like receptor (NLR), TNF, NF-κB, and Toll-like receptor (TLR) signaling pathways. M2-EVs reprogrammed macrophages from the M1 to M2 phenotype, which resulted in beneficial effects on cartilage and attenuation of OA severity. In summary, our study indicated that M2-EV-guided reprogramming of macrophages is a promising treatment strategy for OA.
Collapse
Affiliation(s)
- Qiling Yuan
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Mingyi Yang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Haishi Zheng
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Yongsong Cai
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Pan Luo
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Xinyi Wang
- Department of Rehabilitation, Shaanxi Provincial Rehabilitation Hospital, Xi'an, Shaanxi 710065, China
| | - Peng Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| |
Collapse
|
35
|
Li Y, Qian M, Liu Y, Qiu X. APPROACH: Sensitive Detection of Exosomal Biomarkers by Aptamer-Mediated Proximity Ligation Assay and Time-Resolved Förster Resonance Energy Transfer. BIOSENSORS 2024; 14:233. [PMID: 38785707 PMCID: PMC11117858 DOI: 10.3390/bios14050233] [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: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
Exosomal biomarker detection holds great importance in the field of in vitro diagnostics, offering a non-invasive and highly sensitive approach for early disease detection and personalized treatment. Here, we proposed an "APPROACH" strategy, combining aptamer-mediated proximity ligation assay (PLA) with rolling circle amplification (RCA) and time-resolved Förster resonance energy transfer (TR-FRET) for the sensitive and semi-homogenous detection of exosomal biomarkers. PLA probes consisted of a cholesterol-conjugated oligonucleotide, which anchored to the membrane of an exosome, and a specific aptamer oligonucleotide that recognized a target protein of the exosome; the proximal binding of pairs of PLA probes to the same exosome positioned the oligonucleotides in the vicinity of each other, guiding the hybridization and ligation of two subsequently added backbone and connector oligonucleotides to form a circular DNA molecule. Circular DNA formed from PLA underwent rolling circle amplification (RCA) for signal amplification, and the resulting RCA products were subsequently quantified by TR-FRET. The limits of detection provided by APPROACH for the exosomal biomarkers CD63, PD-L1, and HER2 were 0.46 ng∙μL-1, 0.77 ng∙μL-1, and 1.1 ng∙μL-1, respectively, demonstrating excellent analytical performance with high sensitivity and quantification accuracy. Furthermore, the strategy afforded sensitive detection of exosomal CD63 with a LOD of 1.56 ng∙μL-1 in complex biological matrices, which underscored its anti-interference capability and potential for in vitro detection. The proposed strategy demonstrates wide-ranging applicability in quantifying diverse exosomal biomarkers while exhibiting robust analytical characteristics, including high sensitivity and accuracy.
Collapse
Affiliation(s)
- Ying Li
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.L.); (M.Q.)
| | - Meiqi Qian
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.L.); (M.Q.)
| | | | - Xue Qiu
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.L.); (M.Q.)
| |
Collapse
|
36
|
Chen B, Sun Y, Xu G, Jiang J, Zhang W, Wu C, Xue P, Cui Z. Role of crosstalk between synovial cells and chondrocytes in osteoarthritis (Review). Exp Ther Med 2024; 27:201. [PMID: 38590580 PMCID: PMC11000048 DOI: 10.3892/etm.2024.12490] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 04/10/2024] Open
Abstract
Osteoarthritis (OA) is a low-grade, nonspecific inflammatory disease that affects the entire joint. This condition is characterized by synovitis, cartilage erosion, subchondral bone defects, and subpatellar fat pad damage. There is mounting evidence demonstrating the significance of crosstalk between synovitis and cartilage destruction in the development of OA. To comprehensively explore the phenotypic alterations of synovitis and cartilage destruction, it is important to elucidate the crosstalk mechanisms between chondrocytes and synovial cells. Furthermore, the updated iteration of single-cell sequencing technology reveals the interaction between chondrocyte and synovial cells. In the present review, the histological and pathological alterations between cartilage and synovium during OA progression are described, and the mode of interaction and molecular mechanisms between synovial cells and chondrocytes in OA, both of which affect the OA process mainly by altering the inflammatory environment and cellular state, are elucidated. Finally, the current OA therapeutic approaches are summarized and emerging therapeutic targets are reviewed in an attempt to provide potential insights into OA treatment.
Collapse
Affiliation(s)
- Baisen Chen
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yuyu Sun
- Department of Orthopedics, Nantong Third People's Hospital, Nantong, Jiangsu 226003, P.R. China
| | - Guanhua Xu
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jiawei Jiang
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Wenhao Zhang
- Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chunshuai Wu
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Pengfei Xue
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Zhiming Cui
- Department of Orthopedics, Nantong City No. 1 People's Hospital and Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
37
|
Agnihotram R, Dhar R, Dhar D, Purushothaman K, Narasimhan AK, Devi A. Fusion of Exosomes and Nanotechnology: Cutting-Edge Cancer Theranostics. ACS APPLIED NANO MATERIALS 2024; 7:8489-8506. [DOI: 10.1021/acsanm.4c01033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Rohan Agnihotram
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Debolina Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Kaavya Purushothaman
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Ashwin Kumar Narasimhan
- Department of Biomedical Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu-603203, India
| |
Collapse
|
38
|
Reis IL, Lopes B, Sousa P, Sousa AC, Caseiro AR, Mendonça CM, Santos JM, Atayde LM, Alvites RD, Maurício AC. Equine Musculoskeletal Pathologies: Clinical Approaches and Therapeutical Perspectives-A Review. Vet Sci 2024; 11:190. [PMID: 38787162 PMCID: PMC11126110 DOI: 10.3390/vetsci11050190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Musculoskeletal injuries such as equine osteoarthritis, osteoarticular defects, tendonitis/desmitis, and muscular disorders are prevalent among sport horses, with a fair prognosis for returning to exercise or previous performance levels. The field of equine medicine has witnessed rapid and fruitful development, resulting in a diverse range of therapeutic options for musculoskeletal problems. Staying abreast of these advancements can be challenging, prompting the need for a comprehensive review of commonly used and recent treatments. The aim is to compile current therapeutic options for managing these injuries, spanning from simple to complex physiotherapy techniques, conservative treatments including steroidal and non-steroidal anti-inflammatory drugs, hyaluronic acid, polysulfated glycosaminoglycans, pentosan polysulfate, and polyacrylamides, to promising regenerative therapies such as hemoderivatives and stem cell-based therapies. Each therapeutic modality is scrutinized for its benefits, limitations, and potential synergistic actions to facilitate their most effective application for the intended healing/regeneration of the injured tissue/organ and subsequent patient recovery. While stem cell-based therapies have emerged as particularly promising for equine musculoskeletal injuries, a multidisciplinary approach is underscored throughout the discussion, emphasizing the importance of considering various therapeutic modalities in tandem.
Collapse
Affiliation(s)
- Inês L. Reis
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana C. Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana R. Caseiro
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Veterinary Sciences Department, University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
- Vasco da Gama Research Center (CIVG), University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
| | - Carla M. Mendonça
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Jorge M. Santos
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís M. Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Rui D. Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana C. Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| |
Collapse
|
39
|
Wang Y, Wen J, Lu T, Han W, Jiao K, Li H. Mesenchymal Stem Cell-Derived Extracellular Vesicles in Bone-Related Diseases: Intercellular Communication Messengers and Therapeutic Engineering Protagonists. Int J Nanomedicine 2024; 19:3233-3257. [PMID: 38601346 PMCID: PMC11005933 DOI: 10.2147/ijn.s441467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/23/2024] [Indexed: 04/12/2024] Open
Abstract
Extracellular vesicles (EVs) can deliver various bioactive molecules among cells, making them promising diagnostic and therapeutic alternatives in diseases. Mesenchymal stem cell-derived EVs (MSC-EVs) have shown therapeutic potential similar to MSCs but with drawbacks such as lower yield, reduced biological activities, off-target effects, and shorter half-lives. Improving strategies utilizing biotechniques to pretreat MSCs and enhance the properties of released EVs, as well as modifying MSC-EVs to enhance targeting abilities and achieve controlled release, shows potential for overcoming application limitations and enhancing therapeutic effects in treating bone-related diseases. This review focuses on recent advances in functionalizing MSC-EVs to treat bone-related diseases. Firstly, we underscore the significance of MSC-EVs in facilitating crosstalk between cells within the skeletal environment. Secondly, we highlight strategies of functional-modified EVs for treating bone-related diseases. We explore the pretreatment of stem cells using various biotechniques to enhance the properties of resulting EVs, as well as diverse approaches to modify MSC-EVs for targeted delivery and controlled release. Finally, we address the challenges and opportunities for further research on MSC-EVs in bone-related diseases.
Collapse
Affiliation(s)
- Yanyi Wang
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Juan Wen
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Medical School of Nanjing University, Nanjing, People’s Republic of China
- Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, 4006, Australia
| | - Tong Lu
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Medical School of Nanjing University, Nanjing, People’s Republic of China
| | - Wei Han
- Medical School of Nanjing University, Nanjing, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital & State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, People’s Republic of China
- Medical School of Nanjing University, Nanjing, People’s Republic of China
| |
Collapse
|
40
|
Liu B, Xian Y, Chen X, Shi Y, Dong J, Yang L, An X, Shen T, Wu W, Ma Y, He Y, Gong W, Peng R, Lin J, Liu N, Guo B, Jiang Q. Inflammatory Fibroblast-Like Synoviocyte-Derived Exosomes Aggravate Osteoarthritis via Enhancing Macrophage Glycolysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307338. [PMID: 38342630 PMCID: PMC11005727 DOI: 10.1002/advs.202307338] [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: 10/03/2023] [Revised: 01/23/2024] [Indexed: 02/13/2024]
Abstract
The severity of osteoarthritis (OA) and cartilage degeneration is highly associated with synovial inflammation. Although recent investigations have revealed a dysregulated crosstalk between fibroblast-like synoviocytes (FLSs) and macrophages in the pathogenesis of synovitis, limited knowledge is available regarding the involvement of exosomes. Here, increased exosome secretion is observed in FLSs from OA patients. Notably, internalization of inflammatory FLS-derived exosomes (inf-exo) can enhance the M1 polarization of macrophages, which further induces an OA-like phenotype in co-cultured chondrocytes. Intra-articular injection of inf-exo induces synovitis and exacerbates OA progression in murine models. In addition, it is demonstrated that inf-exo stimulation triggers the activation of glycolysis. Inhibition of glycolysis using 2-DG successfully attenuates excessive M1 polarization triggered by inf-exo. Mechanistically, HIF1A is identified as the determinant transcription factor, inhibition of which, both pharmacologically or genetically, relieves macrophage inflammation triggered by inf-exo-induced hyperglycolysis. Furthermore, in vivo administration of an HIF1A inhibitor alleviates experimental OA. The results provide novel insights into the involvement of FLS-derived exosomes in OA pathogenesis, suggesting that inf-exo-induced macrophage dysfunction represents an attractive target for OA therapy.
Collapse
|
41
|
Chen Y, Zhang H. [Research progress in targeted delivery of inner ear using nanocarriers]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2024; 38:348-353. [PMID: 38563182 PMCID: PMC11387296 DOI: 10.13201/j.issn.2096-7993.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Indexed: 04/04/2024]
Abstract
Various inner ear diseases such as sensorineural deafness and Meniere's disease bring about problems such as speech communication disorders and decreased work efficiency, which seriously affect the life quality of patients. Due to the special anatomical structure and blood-labyrinth barrier in the inner ear, the current drug administration methods are often unable to achieve satisfactory results. Nanocarriers are the forefront and hot spot of nanotechnology research. In recent years, a lot of research progress has been made in the field of targeted delivery of the inner ear, which is expected to be eventually applied to the treatment of clinical diseases of the inner ear. This review focuses on the advantages, main research achievements and limitations of various nanocarriers in the targeted delivery of the inner ear, hoping to provide new ideas for related research.
Collapse
Affiliation(s)
- Yaoheng Chen
- Department of Otolaryngology Head and Neck Surgery,Affiliated Zhujiang Hospital of Southern Medical University;Department of Southern Medical University Hearing Research Center,Guangzhou,510220,China
| | - Hongzheng Zhang
- Department of Otolaryngology Head and Neck Surgery,Affiliated Zhujiang Hospital of Southern Medical University;Department of Southern Medical University Hearing Research Center,Guangzhou,510220,China
| |
Collapse
|
42
|
Chu H, Zhang S, Zhang Z, Yue H, Liu H, Li B, Yin F. Comparison studies identify mesenchymal stromal cells with potent regenerative activity in osteoarthritis treatment. NPJ Regen Med 2024; 9:14. [PMID: 38561335 PMCID: PMC10984924 DOI: 10.1038/s41536-024-00358-y] [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: 06/05/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
Osteoarthritis affects 15% of people over 65 years of age. It is characterized by articular cartilage degradation and inflammation, leading to joint pain and disability. Osteoarthritis is incurable and the patients may eventually need joint replacement. An emerging treatment is mesenchymal stromal cells (MSCs), with over two hundred clinical trials being registered. However, the outcomes of these trials have fallen short of the expectation, due to heterogeneity of MSCs and uncertain mechanisms of action. It is generally believed that MSCs exert their function mainly by secreting immunomodulatory and trophic factors. Here we used knee osteoarthritis mouse model to assess the therapeutic effects of MSCs isolated from the white adipose or dermal adipose tissue of Prrx1-Cre; R26tdTomato mice and Dermo1-Cre; R26tdTomato mice. We found that the Prrx1-lineage MSCs from the white adipose tissues showed the greatest in vitro differentiation potentials among the four MSC groups and single cell profiling showed that the Prrx1-lineage MSCs contained more stem cells than the Dermo1 counterpart. Only the Prrx1-lineage cells isolated from white adipose tissues showed long-term therapeutic effectiveness on early-stage osteoarthritis models. Mechanistically, Prrx1-lineage MSCs differentiated into Col2+ chondrocytes and replaced the damage cartilage, activated Col1 expressing in resident chondrocytes, and inhibited synovial inflammation. Transcriptome analysis showed that the articular chondrocytes derived from injected MSCs expressed immunomodulatory cytokines, trophic factors, and chondrocyte-specific genes. Our study identified a MSC population genetically marked by Prrx1 that has great multipotentiality and can differentiate into chondrocytes to replace the damaged cartilage.
Collapse
Affiliation(s)
- Hongshang Chu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shaoyang Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhenlin Zhang
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Hua Yue
- Department of Osteoporosis and Bone Diseases, Shanghai Clinical Research Center of Bone Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| | - Feng Yin
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
- Department of Joint and Sports Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
43
|
Liu Y, Jiang P, Qu Y, Liu C, Zhang D, Xu B, Zhang Q. Exosomes and exosomal miRNAs: A new avenue for the future treatment of rheumatoid arthritis. Heliyon 2024; 10:e28127. [PMID: 38533025 PMCID: PMC10963384 DOI: 10.1016/j.heliyon.2024.e28127] [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: 08/28/2023] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Rheumatoid arthritis is a chronic systemic autoimmune disease that involves mainly synovitis and joint injury and is one of the main causes of disability. The pathogenesis of rheumatoid arthritis is complicated, and the treatment cycle is long. The traditional methods of inhibiting inflammation and immunosuppression are no longer sufficient for treatment of the disease, so there is an urgent need to seek new treatments. The exocrine microenvironment is a kind of microvesicle with a lipid bilayer membrane structure that can be secreted by most cells in the body. This structure contains cell-specific proteins, lipids and nucleic acids that can transmit this information from one cell to another. To achieve cell-to-cell communication. Exocrine microRNAs can be contained in exocrine cells and can be selectively transferred to target receptor cells via exocrine signaling, thus regulating the physiological function of target cells. This article focuses on the pathological changes that occur during the development of rheumatoid arthritis and the biological regulation of exocrine and exocrine microRNAs in rheumatoid joints. Research on the roles of exocrine and exocrine microRNAs in regulating the inflammatory response, cell proliferation/apoptosis, autophagy, effects on fibroblast-like synoviocytes and immune regulation in rheumatoid arthritis was reviewed. In addition, the challenges faced by this new treatment are discussed.
Collapse
Affiliation(s)
- Yuan Liu
- The First Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Ping Jiang
- The First Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Qu
- The First Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Zhang
- Science and Technology Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
44
|
Jonoush ZA, Mahdavi R, Farahani M, Zeinali F, Shayan E, Amari A. The implications of exosomes in psoriasis: disease: emerging as new diagnostic markers and therapeutic targets. Mol Biol Rep 2024; 51:465. [PMID: 38551769 DOI: 10.1007/s11033-024-09449-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/14/2024] [Indexed: 04/02/2024]
Abstract
As the largest human organ, the skin is continuously exposed to various external and internal triggers that affect body homeostasis. Psoriasis is a persistent inflammatory skin condition that has a major bearing on patients' physiological functioning as well as their mental well-being. It is an autoimmune disorder and has been the focus of extensive research efforts in recent years. Cells secrete exosomes into the environment surrounding them, which comprises a lipid bilayer. The movement of cellular components like microRNAs, mRNAs, DNA, lipids, metabolites, and cell-surface proteins is mediated by exosomes. Exosomes are crucial for inducing communication between cells. There has been extensive study of exosomes, both preclinical and clinical, looking at their potential role in autoimmune diseases. Besides the role that they play in the body's basic processes, exosomes are also considered an increasingly essential part as diagnostic and therapeutic agents. In the following article, we conduct a literature review of current studies related to molecular and structural aspects of exosomes. We emphasis on the function of exosomes in pathogenesis, as well as the possibility of their usage in medicinal applications and as biomarkers.
Collapse
Affiliation(s)
- Zahra Akbari Jonoush
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Roya Mahdavi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Zeinali
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elahe Shayan
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Afshin Amari
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
45
|
Safaei S, Fadaee M, Farzam OR, Yari A, Poursaei E, Aslan C, Samemaleki S, Shanehbandi D, Baradaran B, Kazemi T. Exploring the dynamic interplay between exosomes and the immune tumor microenvironment: implications for breast cancer progression and therapeutic strategies. Breast Cancer Res 2024; 26:57. [PMID: 38553754 PMCID: PMC10981336 DOI: 10.1186/s13058-024-01810-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
Breast cancer continues to pose a substantial worldwide health concern, demanding a thorough comprehension of the complex interaction between cancerous cells and the immune system. Recent studies have shown the significant function of exosomes in facilitating intercellular communication and their participation in the advancement of cancer. Tumor-derived exosomes have been identified as significant regulators in the context of breast cancer, playing a crucial role in modulating immune cell activity and contributing to the advancement of the illness. This study aims to investigate the many effects of tumor-derived exosomes on immune cells in the setting of breast cancer. Specifically, we will examine their role in influencing immune cell polarization, facilitating immunological evasion, and modifying the tumor microenvironment. Furthermore, we explore the nascent domain of exosomes produced from immune cells and their prospective involvement in the prevention of breast cancer. This paper focuses on new research that emphasizes the immunomodulatory characteristics of exosomes produced from immune cells. It also explores the possibility of these exosomes as therapeutic agents or biomarkers for the early identification and prevention of breast cancer. The exploration of the reciprocal connections between exosomes formed from tumors and immune cells, together with the rising significance of exosomes derived from immune cells, presents a potential avenue for the advancement of novel approaches in the field of breast cancer therapy and prevention.
Collapse
Affiliation(s)
- Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Manouchehr Fadaee
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Elham Poursaei
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cynthia Aslan
- Research Center for Integrative Medicine in Aging, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Samemaleki
- Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Gholghasht Ave, Tabriz, Iran.
| |
Collapse
|
46
|
Hazrati A, Malekpour K, Khorramdelazad H, Rajaei S, Hashemi SM. Therapeutic and immunomodulatory potentials of mesenchymal stromal/stem cells and immune checkpoints related molecules. Biomark Res 2024; 12:35. [PMID: 38515166 PMCID: PMC10958918 DOI: 10.1186/s40364-024-00580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are used in many studies due to their therapeutic potential, including their differentiative ability and immunomodulatory properties. These cells perform their therapeutic functions by using various mechanisms, such as the production of anti-inflammatory cytokines, growth factors, direct cell-to-cell contact, extracellular vesicles (EVs) production, and mitochondrial transfer. However, mechanisms related to immune checkpoints (ICPs) and their effect on the immunomodulatory ability of MSCs are less discussed. The main function of ICPs is to prevent the initiation of unwanted responses and to regulate the immune system responses to maintain the homeostasis of these responses. ICPs are produced by various types of immune system regulatory cells, and defects in their expression and function may be associated with excessive responses that can ultimately lead to autoimmunity. Also, by expressing different types of ICPs and their ligands (ICPLs), tumor cells prevent the formation and durability of immune responses, which leads to tumors' immune escape. ICPs and ICPLs can be produced by MSCs and affect immune cell responses both through their secretion into the microenvironment or direct cell-to-cell interaction. Pre-treatment of MSCs in inflammatory conditions leads to an increase in their therapeutic potential. In addition to the effect that inflammatory environments have on the production of anti-inflammatory cytokines by MSCs, they can increase the expression of various types of ICPLs. In this review, we discuss different types of ICPLs and ICPs expressed by MSCs and their effect on their immunomodulatory and therapeutic potential.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Samira Rajaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Qian Y, Chu G, Zhang L, Wu Z, Wang Q, Guo JJ, Zhou F. M2 macrophage-derived exosomal miR-26b-5p regulates macrophage polarization and chondrocyte hypertrophy by targeting TLR3 and COL10A1 to alleviate osteoarthritis. J Nanobiotechnology 2024; 22:72. [PMID: 38374072 PMCID: PMC10877765 DOI: 10.1186/s12951-024-02336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/09/2024] [Indexed: 02/21/2024] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent chronic musculoskeletal diseases among the elderly population. In this study, macrophage-derived exosomes were isolated and identified. Exosomes were subjected to microRNA (miRNA) sequencing and bioinformatic analysis, and differentially expressed miRNAs were verified. miR-26b-5p target genes were confirmed through target-site mutation combined with a dual-luciferase reporter assay. The effects of miR-26b-5p on macrophage polarization and chondrocyte hypertrophy were assessed in vitro. miR-26b-5p agomir was applied to mice with OA induced by anterior cruciate ligament transection (ACLT). The therapeutic effects of miR-26b-5p were evaluated via pain behavior experiments and histological observations. In vitro, miR-26b-5p repolarized M1 macrophages to an anti-inflammatory M2 type by targeting the TLR3 signaling pathway. miR-26b-5p could target COL10A1, further inhibiting chondrocyte hypertrophy induced by M1 macrophage-conditioned medium (M1-CM). In vivo, miR-26b-5p agomir ameliorated gait abnormalities and mechanical allodynia in OA mice. miR-26b-5p treatment attenuated synovitis and cartilage degeneration, thereby delaying OA progression. In conclusion, M2 macrophage-derived exosomal miR-26b-5p could protect articular cartilage and ameliorate gait abnormalities in OA mice by targeting TLR3 and COL10A1. miR-26b-5p further affected macrophage polarization and chondrocyte hypertrophy. Thus, this exosomal miR-26b-5p-based strategy might be a potential method for OA treatment.
Collapse
Affiliation(s)
- Yufan Qian
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Genglei Chu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Lei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Zhikai Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Qiuyuan Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China
| | - Jiong Jiong Guo
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China.
| | - Feng Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, No. 899 Ping Hai Road, Suzhou, Jiangsu, China.
- Orthopedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
48
|
Fang S, Zhang B, Xiang W, Zheng L, Wang X, Li S, Zhang T, Feng D, Gong Y, Wu J, Yuan J, Wu Y, Zhu Y, Liu E, Ni Z. Natural products in osteoarthritis treatment: bridging basic research to clinical applications. Chin Med 2024; 19:25. [PMID: 38360724 PMCID: PMC10870578 DOI: 10.1186/s13020-024-00899-w] [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: 11/21/2023] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative musculoskeletal disease, severely impacting the function of patients and potentially leading to disability, especially among the elderly population. Natural products (NPs), obtained from components or metabolites of plants, animals, microorganisms etc., have gained significant attention as important conservative treatments for various diseases. Recently, NPs have been well studied in preclinical and clinical researches, showing promising potential in the treatment of OA. In this review, we summed up the main signaling pathways affected by NPs in OA treatment, including NF-κB, MAPKs, PI3K/AKT, SIRT1, and other pathways, which are related to inflammation, anabolism and catabolism, and cell death. In addition, we described the therapeutic effects of NPs in different OA animal models and the current clinical studies in OA patients. At last, we discussed the potential research directions including in-depth analysis of the mechanisms and new application strategies of NPs for the OA treatment, so as to promote the basic research and clinical transformation in the future. We hope that this review may allow us to get a better understanding about the potential bioeffects and mechanisms of NPs in OA therapy, and ultimately improve the effectiveness of NPs-based clinical conservative treatment for OA patients.
Collapse
Affiliation(s)
- Shunzheng Fang
- School of Pharmacy, Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400022, China
- Rehabilitation Center, Key Specialty of Neck and Low Back Pain Rehabilitation, Strategic Support Force Xingcheng Special Duty Sanatorium, Liaoning, 125100, China
| | - Wei Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Liujie Zheng
- Department of Orthopaedic Surgery, The Fourth Hospital of Wuhan, Wuhan, 430000, Hubei, China
| | - Xiaodong Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Song Li
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, Laboratory for Prevention and Rehabilitation of Training Injuries, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Tongyi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Daibo Feng
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yunquan Gong
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Jinhui Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Jing Yuan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yaran Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Yizhen Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China
| | - Enli Liu
- School of Pharmacy, Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China.
| | - Zhenhong Ni
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Rehabilitation Medicine, Daping Hospital, Army Medical University, Chongqing, 400022, China.
| |
Collapse
|
49
|
Li S, Wu Y, Peng X, Chen H, Zhang T, Chen H, Yang J, Xie Y, Qi H, Xiang W, Huang B, Zhou S, Hu Y, Tan Q, Du X, Huang J, Zhang R, Li X, Luo F, Jin M, Su N, Luo X, Huang S, Yang P, Yan X, Lian J, Zhu Y, Xiong Y, Xiao G, Liu Y, Shen C, Kuang L, Ni Z, Chen L. A Novel Cargo Delivery System-AnCar-Exo LaIMTS Ameliorates Arthritis via Specifically Targeting Pro-Inflammatory Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306143. [PMID: 38083984 PMCID: PMC10870055 DOI: 10.1002/advs.202306143] [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: 08/28/2023] [Revised: 10/31/2023] [Indexed: 02/17/2024]
Abstract
Macrophages are heterogenic phagocytic cells that play distinct roles in physiological and pathological processes. Targeting different types of macrophages has shown potent therapeutic effects in many diseases. Although many approaches are developed to target anti-inflammatory macrophages, there are few researches on targeting pro-inflammatory macrophages, which is partially attributed to their non-s pecificity phagocytosis of extracellular substances. In this study, a novel recombinant protein is constructed that can be anchored on an exosome membrane with the purpose of targeting pro-inflammatory macrophages via antigen recognition, which is named AnCar-ExoLaIMTS . The data indicate that the phagocytosis efficiencies of pro-inflammatory macrophages for different AnCar-ExoLaIMTS show obvious differences. The AnCar-ExoLaIMTS3 has the best targeting ability for pro-inflammatory macrophages in vitro and in vivo. Mechanically, AnCar-ExoLaIMTS3 can specifically recognize the leucine-rich repeat domain of the TLR4 receptor, and then enter into pro-inflammatory macrophages via the TLR4-mediated receptor endocytosis pathway. Moreover, AnCar-ExoLaIMTS3 can efficiently deliver therapeutic cargo to pro-inflammatory macrophages and inhibit the synovial inflammatory response via downregulation of HIF-1α level, thus ameliorating the severity of arthritis in vivo. Collectively, the work established a novel gene/drug delivery system that can specifically target pro-inflammatory macrophages, which may be beneficial for the treatments of arthritis and other inflammatory diseases.
Collapse
|
50
|
Wang P, Shao W, Li Z, Wang B, Lv X, Huang Y, Feng Y. Non-bone-derived exosomes: a new perspective on regulators of bone homeostasis. Cell Commun Signal 2024; 22:70. [PMID: 38273356 PMCID: PMC10811851 DOI: 10.1186/s12964-023-01431-7] [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: 09/10/2023] [Accepted: 12/09/2023] [Indexed: 01/27/2024] Open
Abstract
Accumulating evidence indicates that exosomes help to regulate bone homeostasis. The roles of bone-derived exosomes have been well-described; however, recent studies have shown that some non-bone-derived exosomes have better bone targeting ability than bone-derived exosomes and that their performance as a drug delivery vehicle for regulating bone homeostasis may be better than that of bone-derived exosomes, and the sources of non-bone-derived exosomes are more extensive and can thus be better for clinical needs. Here, we sort non-bone-derived exosomes and describe their composition and biogenesis. Their roles and specific mechanisms in bone homeostasis and bone-related diseases are also discussed. Furthermore, we reveal obstacles to current research and future challenges in the practical application of exosomes, and we provide potential strategies for more effective application of exosomes for the regulation of bone homeostasis and the treatment of bone-related diseases. Video Abstract.
Collapse
Affiliation(s)
- Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wang
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiyao Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|