1
|
Zhang WH, Xiang WY, Yi L, Fang R. The status and hotspot analysis of research on extracellular vesicles and osteoarthritis: a bibliometric analysis. Front Pharmacol 2025; 16:1484437. [PMID: 40230694 PMCID: PMC11994722 DOI: 10.3389/fphar.2025.1484437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
Background Degenerative joint disease, known as osteoarthritis (OA), is characterized by pain, swelling, and decreased mobility. The illness has a major negative influence on patients' quality of life and is common around the world, especially among older people. Nevertheless, there are insufficient possibilities for early diagnosis and therapy. Extracellular vesicles, or EVs, control the immune response, tissue healing, and cellular communication. Methods This work offers a bibliometric representation of the areas of focus and correlations between extracellular vesicles and osteoarthritis. We searched for osteoarthritis and extracellular vesicles in publications in the Web of Science Core Collection (WoSCC) database. Bibliometrics, an R package, CiteSpace 6.1. R2, and VOSviewer 1.6.17 were used to perform bibliometric analyses of concentration fields, trends, and relevant factors. Results 944 papers from 59 nations were published; the countries that contributed the most to the field were China, the USA, and Italy. Professors Laura and Enrico are the top contributors. Sichuan University, Istituto Ortopedico Galeazzi, and Shanghai Jiao Tong University are the top three universities. The International Journal of Molecular Sciences is an excellent publication. Exosome, expression, knee osteoarthritis, extracellular vesicle, mesenchymal stem cell, osteoarthritis, and inflammation are the most often occurring keywords. Conclusion These results suggest areas of interest and focus for future research on EVs and OA. This trend suggests that the volume of literature on OA and EVs will continue to rise, with more research being published in the future. This study helps scholars understand current research hotspots in the field and may inspire future research.
Collapse
Affiliation(s)
- Wen Hao Zhang
- The Fourth Clinical College of Xinjiang Medical University, Urumqi, China
| | - Wen Yuan Xiang
- The Fourth Clinical College of Xinjiang Medical University, Urumqi, China
- Department of Orthopaedic, Institute of Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Department of Orthopaedic, Xinjiang Uygur Autonomous Region Institute of Traditional Chinese Medicine, Urumqi, China
- Department of orthopaedic, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Lin Yi
- The Fourth Clinical College of Xinjiang Medical University, Urumqi, China
- Department of Orthopaedic, Institute of Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Department of Orthopaedic, Xinjiang Uygur Autonomous Region Institute of Traditional Chinese Medicine, Urumqi, China
- Department of orthopaedic, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Rui Fang
- The Fourth Clinical College of Xinjiang Medical University, Urumqi, China
- Department of Orthopaedic, Institute of Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- Department of Orthopaedic, Xinjiang Uygur Autonomous Region Institute of Traditional Chinese Medicine, Urumqi, China
- Department of orthopaedic, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
2
|
Zubair M, Abouelnazar FA, Iqbal MA, Pan J, Zheng X, Chen T, Shen W, Yin J, Yan Y, Liu P, Mao F, Chu Y. Mesenchymal stem cell-derived exosomes as a plausible immunomodulatory therapeutic tool for inflammatory diseases. Front Cell Dev Biol 2025; 13:1563427. [PMID: 40129569 PMCID: PMC11931156 DOI: 10.3389/fcell.2025.1563427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs), especially, exosomes are considered to have diverse therapeutic effects for various significant diseases. MSC-derived exosomes (MSCex) offer substantial advantages over MSCs due to their long-term preservation, stability, absence of nuclei and fewer adverse effects such as infusion toxicity, thereby paving the way towards regenerative medicine and cell-free therapeutics. These exosomes harbor several cellular contents such as DNA, RNA, lipids, metabolites, and proteins, facilitating drug delivery and intercellular communication. MSCex have the ability to immunomodulate and trigger the anti-inflammatory process hence, playing a key role in alleviating inflammation and enhancing tissue regeneration. In this review, we addressed the anti-inflammatory effects of MSCex and the underlying immunomodulatory pathways. Moreover, we discussed the recent updates on MSCex in treating specific inflammatory diseases, including arthritis, inflammatory bowel disease, inflammatory eye diseases, and respiratory diseases such as asthma and acute respiratory distress syndrome (ARDS), as well as neurodegenerative and cardiac diseases. Finally, we highlighted the challenges in using MSCex as the successful therapeutic tool and discussed future perspectives.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Fatma A. Abouelnazar
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, Egypt
| | | | - Jingyun Pan
- Department of Traditional Chinese Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Xuwen Zheng
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Tao Chen
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Wenming Shen
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Jinnan Yin
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Pengjun Liu
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Chu
- Wujin Clinical College, Xuzhou Medical University, Changzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
3
|
Chen X, Tian B, Wang Y, Zheng J, Kang X. Potential and challenges of utilizing exosomes in osteoarthritis therapy (Review). Int J Mol Med 2025; 55:43. [PMID: 39791222 PMCID: PMC11759586 DOI: 10.3892/ijmm.2025.5484] [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: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025] Open
Abstract
Exosomes are integral to the pathophysiology of osteoarthritis (OA) due to their roles in mediating intercellular communication and regulating inflammatory processes. Exosomes are integral to the transport of bioactive molecules, such as proteins, lipids and nucleic acids, which can influence chondrocyte behavior and joint homeostasis. Given their properties of regeneration and ability to target damaged tissues, exosomes represent a promising therapeutic avenue for OA treatment. Exosomes have potential in promoting cartilage repair, reducing inflammation and improving overall joint function. However, several challenges remain, including the need for standardized isolation and characterization methods, variability in exosomal content, and regulatory hurdles. The present review aims to provide a comprehensive overview of the current understanding of exosome mechanisms in OA and their therapeutic potential, while also addressing the ongoing challenges faced in translating these findings into clinical practice. By consolidating existing research, the present review aims to pave the way for future studies aimed at optimizing exosome‑based therapies for effective OA management.
Collapse
Affiliation(s)
| | | | | | - Jiang Zheng
- Department of Joint Surgery, Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| | - Xin Kang
- Department of Joint Surgery, Sports Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shanxi 710054, P.R. China
| |
Collapse
|
4
|
Pakdaman Kolour SS, Nematollahi S, Dehbozorgi M, Fattahi F, Movahed F, Esfandiari N, Kahrizi MS, Ghavamikia N, Hajiagha BS. Extracecellulr vesicles (EVs) microRNAs (miRNAs) derived from mesenchymal stem cells (MSCs) in osteoarthritis (OA); detailed role in pathogenesis and possible therapeutics. Heliyon 2025; 11:e42258. [PMID: 40007782 PMCID: PMC11850152 DOI: 10.1016/j.heliyon.2025.e42258] [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: 04/02/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
The primary cause of pain and disability in the world is osteoarthritis (OA), a common joint disease characterized by the primary pathological alteration in articular cartilage deterioration. The general outcome of treatment is not acceptable despite current interventions. Therefore, joint replacement surgery is frequently needed by patients with severe OA. Mesenchymal stem cells (MSCs) have become a practical treatment choice for preclinical and clinical OA palliation in recent years, mainly due to their unique immunomodulatory attributes. Further, attractive candidates for cell-free therapy for OA are MSC-derived extracecellulr vesicles (EVs) that convey bioactive molecules of the original cells, such as microRNAs. These EVs have been shown to significantly influence the regulation of various physiological activities of cells in the joint cavity. Dysregulated miRNAs upregulate the synthesis of enzymes that degrade cartilage, downregulate the expression of components in the cartilage matrix, promote the production of proinflammatory cytokines, induce programmed cell death in chondrocytes, inhibit the process of autophagy in chondrocytes, and participate in pathways related to pain. MiRNAs are also found in extracellular membranous vesicles (EVs), such as exosomes, and play a role in intercellular communication in osteoarthritic joints. Thus, the biosynthesis, chemical makeup, and mechanism of action of miRNAs-enriched EVs in OA are all thoroughly covered in this review. We additionally discussed how miRNA-enriched MSC-EVs might be used therapeutically to change intercellular interaction in OA.
Collapse
Affiliation(s)
| | - Saeide Nematollahi
- Department of Radiology, Kerman University of Paramedical Sciences, Kerman, Iran
| | | | | | - Fatemeh Movahed
- Department of Gynecology, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Nima Ghavamikia
- Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Salmanian Hajiagha
- Department of Cellular and Molecular Biology, Faculty of Basic Science, East Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
5
|
Huang J, Zeng N, Xu S, Lv Y, Li X, Yang P, Liu Y. The study on bone marrow mesenchymal stem cell-derived extracellular matrix promoting the repair of damaged chondrocytes by regulating the Notch1/RBPJ pathway. Cytotechnology 2025; 77:35. [PMID: 39764424 PMCID: PMC11700074 DOI: 10.1007/s10616-024-00702-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/25/2024] [Indexed: 03/08/2025] Open
Abstract
Cartilage and joint damage can lead to cartilage degeneration. Bone marrow mesenchymal stem cells (BMSCs) have the potential to address cartilage damage. Hence, this study probed the mechanism of BMSC-extracellular matrix (BMSC-ECM) in promoting damaged chondrocyte repair by regulating the Notch1/RBPJ pathway. Human immortalized chondrocytes were cultured in vitro and treated with Notch1 small interfering (si)RNA, pCDNA3.1-Notch1, RBPJ siRNA and their negative controls (NCs). Damaged chondrocytes were constructed. Damaged chondrocyte-BMSC co-culture system was established and treated with lentiviral vector carrying short hairpin-Notch1 and its NC. Cell viability and apoptosis were assessed by CCK-8 and flow cytometry assays. Levels of glycosaminoglycan (GAG), Notch1 and RBPJ mRNA, and Notch1, RBPJ, Col2α1, mmp3, Hes1 and Hey1 were determined by a kit, RT-qPCR and Western blot. NICD nuclear translocation was detected by immunofluorescence. Damaged chondrocytes exhibited down-regulated Notch1 expression, reduced cell viability, and enhanced apoptosis. Further Notch1 knockdown aggravated chondrocyte damage, whereas its overexpression enhanced chondrocyte viability and decreased apoptosis. NICD translocated into the nucleus and bound to RBPJ to activate the Notch1 pathway. RBPJ silencing partly annulled Notch1-regulated damaged chondrocyte apoptosis. BMSC-damaged chondrocyte co-culture up-regulated Notch1, RBPJ, Col2α1, mmp3, Hes1, Hey1 and GAG levels, enhanced cell viability, and reduced apoptosis in chondrocytes, which were partly negated by Notch1 suppression, indicating that BMSC-ECM facilitated damaged chondrocyte repair by activating the Notch1/RBPJ pathway. BMSC-ECM promoted the repair of damaged chondrocytes by promoting NICD translocation into the nucleus and binding to RBPJ to activate the Notch1 pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00702-6.
Collapse
Affiliation(s)
- Jiangfa Huang
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Ningjing Zeng
- The Second Clinical College of Guangzhou, University of Chinese Medicine, Guangzhou, Guangdong China
| | - Shuchai Xu
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Yang Lv
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Xing Li
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Peng Yang
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| | - Yan Liu
- Department of Sports Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 261 Datong Road, Yuexiu District, Guangzhou, 510105 Guangdong China
| |
Collapse
|
6
|
He Y, Wang Y, Yu S, Wang L, Dou Y, Ma R, Cao S, Song W, Ma P. BMP2 enhance the osteogenic effect of BMSCs-derived exosomes in skull defect of diabetic rats. MATERIALS & DESIGN 2024; 248:113517. [DOI: 10.1016/j.matdes.2024.113517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
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
|
8
|
José Alcaraz M. Control of articular degeneration by extracellular vesicles from stem/stromal cells as a potential strategy for the treatment of osteoarthritis. Biochem Pharmacol 2024; 228:116226. [PMID: 38663683 DOI: 10.1016/j.bcp.2024.116226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024]
Abstract
Osteoarthritis (OA) is a degenerative joint condition that contributes to years lived with disability. Current therapeutic approaches are limited as there are no disease-modifying interventions able to delay or inhibit the progression of disease. In recent years there has been an increasing interest in the immunomodulatory and regenerative properties of mesenchymal stem/stromal cells (MSCs) to develop new OA therapies. Extracellular vesicles (EVs) mediate many of the biological effects of these cells and may represent an alternative avoiding the limitations of cell-based therapy. There is also a growing interest in EV modifications to enhance their efficacy and applications. Recent preclinical studies have provided strong evidence supporting the potential of MSC EVs for the development of OA treatments. Thus, MSC EVs may regulate chondrocyte functions to avoid cartilage destruction, inhibit abnormal subchondral bone metabolism and synovial tissue alterations, and control pain behavior. EV actions may be mediated by the transfer of their cargo to target cells, with an important role for proteins and non-coding RNAs modulating signaling pathways relevant for OA progression. Nevertheless, additional investigations are needed concerning EV optimization, and standardization of preparation procedures. More research is also required for a better knowledge of possible effects on different OA phenotypes, pharmacokinetics, mechanism of action, long-term effects and safety profile. Furthermore, MSC EVs have a high potential as vehicles for drug delivery or as adjuvant therapy to potentiate or complement the effects of other approaches.
Collapse
Affiliation(s)
- María José Alcaraz
- Department of Pharmacology, University of Valencia, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| |
Collapse
|
9
|
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
|
10
|
Zhu F, Ye Y, Shao Y, Xue C. MEG3 shuttled by exosomes released from human bone marrow mesenchymal stem cells promotes TP53 stability to regulate MCM5 transcription in keloid fibroblasts. J Gene Med 2024; 26:e3688. [PMID: 38686583 DOI: 10.1002/jgm.3688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Despite the interest in mesenchymal stem cells (MSC), their potential to treat abnormal scarring, especially keloids, is yet to be described. The present study aimed to investigate the therapeutic potential of exosomes derived from human bone marrow MSCs (hBMSC-Exos) in alleviating keloid formation. METHODS Exosomes were isolated from hBMSC, and keloid fibroblasts (KFs) were treated with hBMSC-Exos. Cell counting kit-8, wound healing, transwell invasion, immunofluorescence, and western blot assays were conducted to study the malignant phenotype of KFs. Mice were induced with keloids and treated with hBMSC-Exos. The effect of hBMSC-Exos on keloid formation in vivo was evaluated by hematoxylin and eosin staining, Masson staining, immunohistochemistry, and western blotting. The GSE182192 dataset was screened for differentially expressed long non-coding RNA during keloid formation. Next, maternally expressed gene 3 (MEG3) was knocked down in hBMSC to obtain hBMSC-Exossh-MEG3. The molecular mechanism of MEG3 was investigated by bioinformatic screening, and the relationship between MEG3 and TP53 or MCM5 was verified. RESULTS hBMSC-Exos inhibited the malignant proliferation, migration, and invasion of KFs at same time as promoting their apoptosis, Moreover, hBMSC-Exos reduced the expression of fibrosis- and collagen-related proteins in the cells and the formation of keloids caused by KFs. The reduction in MEG3 enrichment in hBMSC-Exos weakened the inhibitory effect of hBMSC-Exos on KF activity. hBMSC-Exos delivered MEG3 to promote MCM5 transcription by TP53 in KFs. Overexpression of MCM5 in KFs reversed the effects of hBMSC-Exossh-MEG3, leading to reduced KF activity. CONCLUSIONS hBMSC-Exos delivered MEG3 to promote the protein stability of TP53, thereby activating MCM5 and promoting KF activity.
Collapse
Affiliation(s)
- Feibin Zhu
- Department of Burn Surgery, Huizhou Central People's Hospital, Huizhou, China
| | - Yuanjian Ye
- Hand and Foot Microsurgery & Wound Repair Department, Huizhou First Hospital, Huizhou, China
| | - Ying Shao
- Department of Tumor Radiotherapy, Huizhou Central People's Hospital, Huizhou, China
| | - Chunli Xue
- Department of Burn Surgery, Huizhou Central People's Hospital, Huizhou, China
| |
Collapse
|
11
|
Cheng S, Xu X, Wang R, Chen W, Qin K, Yan J. Chondroprotective effects of bone marrow mesenchymal stem cell-derived exosomes in osteoarthritis. J Bioenerg Biomembr 2024; 56:31-44. [PMID: 38012335 DOI: 10.1007/s10863-023-09991-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/21/2023] [Indexed: 11/29/2023]
Abstract
Chondrocyte ferroptosis constitutes a major cause of the development of osteoarthritis (OA). Bone marrow mesenchymal stem cell-derived exosomes (BMSC-Exos) have a protective role against ferroptosis in various diseases. Hence, we aimed to determine whether BMSC-Exos alleviated chondrocyte ferroptosis and its effect on OA, and to dissect out the possible mechanisms. An OA rat chondrocyte model was established by interleukin-1β (IL-1β) exposure, and treated with BMSC-Exos/ferroptosis inhibitor Ferrostatin-1. Cell viability/ferroptosis-related index levels [reactive oxygen species (ROS)/malondialdehyde (MDA)/glutathione (GSH)]/cell death/ACSL4 mRNA and protein levels and METTL3 levels were assessed by MTT/kits/immunohistochemical method and TUNEL staining/RT-qPCR and Western blot. METTL3/ACSL4 were overexpressed in rat chondrocytes to evaluate their role in BMSC-Exo-produced repression on chondrocyte ferroptosis. Bioinformatics website predicted the presence of m6A modification sites on ACSL4 mRNA, with the m6A level enriched on it assessed by MeRIP/RT-qPCR. ACSL4 mRNA stability was detected by actinomycin D assay. A surgical destabilized medial meniscus rat OA model was also established, followed by injection with BMSC-Exos to verify their function. IL-1β stimulation in rat chondrocytes inhibited cell viability, elevated Fe2+/ROS/MDA levels, declined GSH levels and increased TUNEL positive cell number and ACSL4 level, which were neutralized by BMSC-Exos. BMSC-Exos limited chondrocyte ferroptosis by down-regulating METTL3, with the effect abrogated by METTL3 overexpression. METTL3 regulated the m6A modification of ACSL4 mRNA, increasing ACSL4 mRNA stability and ACSL4 expression. BMSC-Exos reduced chondrocyte ferroptosis and prevented OA progression via disruption of the METTL3-m6A-ACSL4 axis. BMSC-Exos might exert a chondroprotective effect by attenuating chondrocyte ferroptosis and alleviate OA progression.
Collapse
Affiliation(s)
- Shi Cheng
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province, 150010, China
| | - Xiangning Xu
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province, 150010, China
| | - Ren Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province, 150010, China
| | - Weijie Chen
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province, 150010, China
| | - Kunhan Qin
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province, 150010, China
| | - Jinglong Yan
- Department of Orthopedic Surgery, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin City, Heilongjiang Province, 150010, China.
| |
Collapse
|
12
|
Somadder R, Faraj L, Datta S, Kanapathipillai M, Ghosh G. Effect of extracellular matrices on production and potency of mesenchymal stem cell-derived exosomes. Biotechnol J 2024; 19:e2300474. [PMID: 38403471 DOI: 10.1002/biot.202300474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/17/2023] [Accepted: 12/23/2023] [Indexed: 02/27/2024]
Abstract
Mesenchymal stem cell (MSC) derived exosomes have emerged as potential acellular therapeutics for various tissue regenerative applications. However, successful clinical translation of exosome-based therapy is limited by lack of a structured production platform. Thus, in this study, the effect of decellularized extracellular matrix (dECM) was assessed on the production and potency of exosomes secreted by bone marrow-derived human MSCs. The results indicate that there was a ∼2-fold increase in MSC-exosome production when MSCs were cultured on dECM compared to TCP. Further, our study revealed that dECM generation induced by ascorbic acid (AA) up to 100 µg mL-1 highly increased exosome yield thereby indicating a potential scale up method for MSC exosome production. The bioactivity of exosomes was investigated by their ability to improve the healing of wounded human skin explants. Wound closure was enhanced in the presence of exosomes isolated from MSCs cultured on ascorbic acid-induced dECM compared to TCP generated MSC-exosomes. In summary, this study suggests a promising solution to a major bottleneck in large-scale production of MSC exosomes for cell-free therapy.
Collapse
Affiliation(s)
- Rittika Somadder
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Lina Faraj
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Saurav Datta
- Amgen Bioprocessing Center, Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, USA
| | - Mathumai Kanapathipillai
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Gargi Ghosh
- Bioengineering Program, Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, Michigan, USA
- Amgen Bioprocessing Center, Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, California, USA
| |
Collapse
|
13
|
Luo D, Zhu H, Li S, Wang Z, Xiao J. Mesenchymal stem cell-derived exosomes as a promising cell-free therapy for knee osteoarthritis. Front Bioeng Biotechnol 2024; 12:1309946. [PMID: 38292826 PMCID: PMC10824863 DOI: 10.3389/fbioe.2024.1309946] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Osteoarthritis (OA), as a degenerative disease, leads to high socioeconomic burdens and disability rates. The knee joint is typically the most affected and is characterized by progressive destruction of articular cartilage, subchondral bone remodeling, osteophyte formation and synovial inflammation. The current management of OA mainly focuses on symptomatic relief and does not help to slow down the advancement of disease. Recently, mesenchymal stem cells (MSCs) and their exosomes have garnered significant attention in regenerative therapy and tissue engineering areas. Preclinical studies have demonstrated that MSC-derived exosomes (MSC-Exos), as bioactive factor carriers, have promising results in cell-free therapy of OA. This study reviewed the application of various MSC-Exos for the OA treatment, along with exploring the potential underlying mechanisms. Moreover, current strategies and future perspectives for the utilization of engineered MSC-Exos, alongside their associated challenges, were also discussed.
Collapse
Affiliation(s)
| | | | | | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Bordon G, Berenbaum F, Distler O, Luciani P. Harnessing the multifunctionality of lipid-based drug delivery systems for the local treatment of osteoarthritis. Biomed Pharmacother 2023; 168:115819. [PMID: 37939613 DOI: 10.1016/j.biopha.2023.115819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/10/2023] Open
Abstract
Osteoarthritis (OA) is a widespread joint condition affecting millions globally, presenting a growing socioeconomic burden thus making the development of more effective therapeutic strategies crucial. This review emphasizes recent advancements in lipid-based drug delivery systems (DDSs) for intra-articular administration of OA therapeutics, encompassing non-steroidal anti-inflammatory drugs, corticosteroids, small molecule disease-modifying OA drugs, and RNA therapeutics. Liposomes, lipid nanoparticles, lipidic mesophases, extracellular vesicles and composite systems exhibit enhanced stability, targeted delivery, and extended joint retention, which contribute to improved therapeutic outcomes and minimized systemic drug exposure. Although active targeting strategies hold promise, further research is needed to assess their targeting efficiency in physiologically relevant conditions. Simultaneously, multifunctional DDSs capable of delivering combinations of distinct therapeutic classes offer synergistic effects and superior OA treatment outcomes. The development of such long-acting systems that resist rapid clearance from the joint space is crucial, where particle size and targeting capabilities emerge as vital factors. Additionally, combining cartilage lubrication properties with sustained drug delivery has demonstrated potential in animal models, meriting further investigation in human clinical trials. This review highlights the crucial need for direct, head-to-head comparisons of novel DDSs with standard treatments, particularly within the same drug class. These comparisons are essential in accurately evaluating their effectiveness, safety, and clinical applicability, and are set to significantly shape the future of OA therapy.
Collapse
Affiliation(s)
- Gregor Bordon
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Francis Berenbaum
- Sorbonne University, INSERM CRSA, AP-HP Saint-Antoine Hospital, Paris, France
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
15
|
Long L, Zou G, Cheng Y, Li F, Wu H, Shen Y. MATN3 delivered by exosome from synovial mesenchymal stem cells relieves knee osteoarthritis: Evidence from in vitro and in vivo studies. J Orthop Translat 2023; 41:20-32. [PMID: 37635810 PMCID: PMC10448336 DOI: 10.1016/j.jot.2023.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 08/29/2023] Open
Abstract
Background Synovial mesenchymal stem cell (SMSC) exerts chondroprotective effects in osteoarthritis (OA) clinical models. However, the regulatory potentials of SMSC-derived exosomes (SMSC-Exo) in OA still need to be discovered, which attracted our attention. Methods The destabilization of the medial meniscus surgery was performed on the knee joints of a mouse OA model, followed by injection of SMSC-Exo. In addition, SMSC-Exo was administrated to mouse chondrocytes to observe the functional and molecular alterations. Results Both of SMSC-Exo and overexpression of Matrilin-3 (MATN3) alleviated cartilage destruction and suppressed degradation of extracellular matrix (ECM) in the OA rat model. In addition, assays concerning the in vitro OA model induced by IL-1β showed that SMSC-Exo could promote chondrocyte viability and inhibit autophagy defects. Furthermore, SMSC-Exo achieved the chondroprotective effects through the delivery of MATN3/IL-17A, and MATN3 could suppress the activation of PI3K/AKT/mTOR signaling through IL-17A. Conclusion SMSC-Exo exerts beneficial therapeutic effects on OA by preventing ECM degradation and autophagy defects by delivering MATN3/IL-17A. The Translational Potential of this Article The translational potential of this study is not only limited to the treatment of knee osteoarthritis but also provides new insights for the treatment of other joint diseases by exploring the mechanism of MATN3. In addition, SMSCExo, as a novel drug carrier, has great potential for treating and diagnosing other diseases. With further research, these findings will provide new directions for developing personalized and innovative treatment options.
Collapse
Affiliation(s)
- Long Long
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, China
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, 224001, Jiangsu Province, China
| | - Guoyou Zou
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, 224001, Jiangsu Province, China
| | - Yi Cheng
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, 224001, Jiangsu Province, China
| | - Feng Li
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, 224001, Jiangsu Province, China
| | - Hao Wu
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, 224001, Jiangsu Province, China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, Jiangsu Province, China
| |
Collapse
|