1
|
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
|
2
|
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
|
3
|
Qiu M, Xie Y, Tan G, Wang X, Huang P, Hong L. Synovial mesenchymal stem cell-derived exosomal miR-485-3p relieves cartilage damage in osteoarthritis by targeting the NRP1-mediated PI3K/Akt pathway: Exosomal miR-485-3p relieves cartilage damage. Heliyon 2024; 10:e24042. [PMID: 38293485 PMCID: PMC10826677 DOI: 10.1016/j.heliyon.2024.e24042] [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: 06/28/2023] [Revised: 12/24/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Osteoarthritis (OA) is an age-related musculoskeletal disease that results in pain and functional disability. Stem cell therapy has been considered as a promising treatment for OA. In this study, the therapeutic action and potential mechanism of synovial mesenchymal stem cells (SMSCs)-derived exosomes (Exos) in OA cartilage damage were investigated. Cartilage cells were stimulated with IL-1β to establish an in vitro model of OA cartilage damage. Cartilage cell functions were detected by CCK-8, scratch assay, and flow cytometry, respectively. Inflammatory cytokine levels were assessed by ELISA. Target molecule levels were measured by qRT‒PCR and Western blotting. Exos-induced differential expression of miRNAs in cartilage cells were analyzed by microarray analysis. The interaction between miR-485-3p and neuropilin-1 (NRP1) was validated by dual luciferase reporter and RIP assays. We found that treatment with Exos promoted proliferation, migration, and ECM secretion, but restrained apoptosis and inflammation of IL-1β-exposed cartilage cells via up-regulation of miR-485-3p. Additionally, miR-485-3p directly targeted NRP1 to repress NRP1 expression, which subsequently caused inactivation of the PI3K/Akt pathway. The protective effect of Exos on cartilage damage was counteracted by NRP1 overexpression-mediated activation of the PI3K/Akt pathway. In conclusion, Exos delivered miR-485-3p to attenuate IL-1β-induced cartilage degradation by targeting NRP1 and succedent inactivation of the PI3K/Akt pathway. Our findings shed light on the novel protective mechanism of Exos in OA, which suggest that the restoration of miR-485-3p by Exos might be a novel approach for OA treatment.
Collapse
Affiliation(s)
- Mingjun Qiu
- Department of joint surgery, The Second Affiliated Hospital of University of South China, China
| | - Yanhua Xie
- Department of orthopedic, The Second Affiliated Hospital of University of South China, China
| | - Guanghua Tan
- Department of joint surgery, The Second Affiliated Hospital of University of South China, China
| | - Xiaoxu Wang
- Department of joint surgery, The Second Affiliated Hospital of University of South China, China
| | - Peiguan Huang
- Department of joint surgery, The Second Affiliated Hospital of University of South China, China
| | - Liang Hong
- Department of joint surgery, The Second Affiliated Hospital of University of South China, China
| |
Collapse
|
4
|
Dong L, Li X, Leng W, Guo Z, Cai T, Ji X, Xu C, Zhu Z, Lin J. Adipose stem cells in tissue regeneration and repair: From bench to bedside. Regen Ther 2023; 24:547-560. [PMID: 37854632 PMCID: PMC10579872 DOI: 10.1016/j.reth.2023.09.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
ADSCs are a large number of mesenchymal stem cells in Adipose tissue, which can be applied to tissue engineering. ADSCs have the potential of multi-directional differentiation, and can differentiate into bone tissue, cardiac tissue, urothelial cells, skin tissue, etc. Compared with other mesenchymal stem cells, ADSCs have a multitude of promising advantages, such as abundant number, accessibility in cell culture, stable function, and less immune rejection. There are two main methods to use ADSCs for tissue repair and regeneration. One is to implant the "ADSCs-scaffold composite" into the injured site to promote tissue regeneration. The other is cell-free therapy: using ADSC-exos or ADSC-CM alone to release a large number of miRNAs, cytokines and other bioactive substances to promote tissue regeneration. The tissue regeneration potential of ADSCs is regulated by a variety of cytokines, signaling molecules, and external environment. The differentiation of ADSCs into different tissues is also induced by growth factors, ions, hormones, scaffold materials, physical stimulation, and other factors. The specific mechanisms are complex, and most of the signaling pathways need to be further explored. This article reviews and summarizes the mechanism and clinical application of ADSCs in tissue injury repair so far, and puts forward further problems that need to be solved in this field, hoping to provide directions for further research in this field.
Collapse
Affiliation(s)
- Lei Dong
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xiaoyu Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Wenyuan Leng
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Zhenke Guo
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Tianyu Cai
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xing Ji
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Chunru Xu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Zhenpeng Zhu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| |
Collapse
|
5
|
Gao H, Di J, Yin M, He T, Wu D, Chen Z, Li S, He L, Rong L. Identification of chondrocyte subpopulations in osteoarthritis using single-cell sequencing analysis. Gene 2023; 852:147063. [PMID: 36427677 DOI: 10.1016/j.gene.2022.147063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/20/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease. Previous studies were focused on general functions of chondrocyte population in OA without elucidating the existence of chondrocyte subpopulations. To investigate the heterogeneity of chondrocyte, here we conducted detailed analysis on the single-cell sequencing data of cartilage cells from OA patients. After quality control, unsupervised K-mean clustering identified seven different subpopulations of chondrocytes in OA. Those subpopulations of chondrocytes were nominated based on Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis: stress-metabolizing chondrocytes (cluster 1), rhythmic chondrocytes (cluster 2), apoptotic chondrocytes (cluster 3), matrix-synthesis-related chondrocytes (cluster 4), developmental chondrocytes (cluster 5), protein-synthesis-related chondrocytes (cluster 6 and 8), and osteogenesis chondrocytes (cluster 7). We further noticed that the stress-metabolizing chondrocytes (cluster 1) were dominant in early stages of cartilage damage with increased metabolic levels inhibiting cartilage tissue degeneration, while the matrix-synthesis-related chondrocytes (cluster 4) were mainly existed in the late stages of cartilage damage which reorganized collagen fibers with type III collagen disrupting the extracellular matrix and further cartilage damages. Besides, we identified genes NFKBIA and TUBB2B as potential markers for the stress-metabolizing chondrocytes and the matrix synthesis related chondrocytes, respectively. Our study identifies different chondrocyte subpopulations in OA, and highlights the potential different functions of chondrocyte subpopulations in the early versus late stages of cartilage damage.
Collapse
Affiliation(s)
- Han Gao
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China
| | - Jiawei Di
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China
| | - Mingyu Yin
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China
| | - Tianwei He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China
| | - Depeng Wu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China
| | - Zihao Chen
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China
| | - Shangfu Li
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China
| | - Lei He
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China.
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, 510630 Guangzhou, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, 510630 Guangzhou, China; Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, 510630 Guangzhou, China.
| |
Collapse
|
6
|
Li C, Li W, Pu G, Wu J, Qin F. Exosomes derived from miR-338-3p-modified adipose stem cells inhibited inflammation injury of chondrocytes via targeting RUNX2 in osteoarthritis. J Orthop Surg Res 2022; 17:567. [PMID: 36572886 PMCID: PMC9791748 DOI: 10.1186/s13018-022-03437-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease that is one of the main causes of disability in middle-aged and elderly people. Adipose stem cell (ASC)-derived exosomes (ASC-Exo) could repair cartilage damage and treat OA. MiRNA-338-3p expression was confirmed to play a role in inhibiting proinflammatory cytokines. Herein, we aimed to explore the mechanism by which exosomes derived from miR-338-3p overexpressing ASCs protects chondrocytes from interleukin (IL)-1β-induced chondrocyte change. METHODS Exosomes were extracted from ASCs transfected with miR-338-3p or its antisense inhibitor. The ASC-Exos (miR-338-3p silencing/overexpression) were incubated with IL-1β-induced ATDC5 cells, followed by evaluation of the chondrocyte proliferation, degradation, and inflammation injury. RESULTS In vitro results revealed that ASC-Exos inhibited the expression of prostaglandin E2 (PGE2), IL-6, IL-1β, and TNF-α, as well as promoted the proliferation of ATDC5 cells. Moreover, ASC-Exos inhibited inflammation injury and degradation of ATDC5 cells by transferring miR-338-3p. Luciferase reporter assays showed that RUNX2 was a target gene of miR-338-3p. Additionally, RUNX2 overexpression in ATDC5 cells reversed the protective effect of miR-338-3p on chondrocytes. Taken together, this study demonstrated that exosomes secreted from miR-338-3p-modified ASCs were effective in the repair of IL-1β-induced chondrocyte change by inhibiting RUNX2 expression. CONCLUSIONS Our result provided valuable data for understanding the mechanism of ASC-Exos in OA treatment.
Collapse
Affiliation(s)
- ChunLiang Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Wei Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - GengZang Pu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - JingWen Wu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Feng Qin
- grid.459333.bDepartment of Endocrinology, Qinghai University Affiliated Hospital, Chengxi District, No. 6, Xichuan South Road, Xining, 810006 Qinghai China
| |
Collapse
|
7
|
The Effectiveness and Safety of Mesenchymal Stem Cells in the Treatment of Osteoarthritis: A Systematic Review and Meta-analysis of 28 Randomized Controlled Trials. Stem Cells Int 2022; 2022:6151866. [PMID: 36277037 PMCID: PMC9581629 DOI: 10.1155/2022/6151866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/08/2022] [Accepted: 07/21/2022] [Indexed: 11/18/2022] Open
Abstract
Objective To evaluate the effectiveness and safety of mesenchymal stem cells (MSCs) in the treatment of osteoarthritis (OA). Methods Chinese databases (such as CNKI and SinoMed) and English databases (such as PubMed and Embase) were searched to collect randomized controlled trials (RCTs) of MSCs in the treatment of OA. The retrieval time is from inception to October 10, 2021. The literature was strictly selected according to the inclusion and exclusion criteria, data was extracted, and the quality was evaluated. RevMan 5.3 software was used for meta-analysis. STATA was used to evaluate publication bias. The registration number of this systematic review and meta-analysis is CRD42021277145. Results A total of 28 RCTs involving 1494 participants were included. The primary outcomes showed that MSCs may reduce WOMAC pain and VAS at the 3rd-month follow-up [WOMAC pain: -3.81 (-6.95, -0.68), P = 0.02. VAS: -1.11 (-1.53, -0.68), P < 0.00001], and the effect lasts for at least 12 months [WOMAC pain: -4.29 (-7.12, -1.47), P = 0.003. VAS: -1.77 (-2.43, -1.12), P < 0.00001]. MSCs may also reduce WOMAC stiffness and physical function at the 6th-month follow-up [WOMAC stiffness: -1.12 (-2.09, -0.14), P = 0.03. WOMAC physical function: -4.40 (-6.84, -1.96), P = 0.0004], and the effect lasts for at least 12 months [WOMAC stiffness: -0.99 (-1.95, -0.03), P = 0.04. WOMAC physical function: -3.26 (-5.91, -0.61), P = 0.02]. The improvement of WOMAC pain, VAS, WOMAC stiffness, and WOMAC physical function may be clinically significant. Meanwhile, after the MSC injection, Lequesne had been reduced compared with the control group [-4.49 (-8.21, -0.77), P = 0.002]. For adverse events, there is no significant difference in the safety of MSC injection and the control group [1.20 (0.97, 1.48), P = 0.09]. The quality of WOMAC physical function and adverse events were moderate. Conclusion Based on current evidence, MSCs may be a safety therapy that have a good curative effect in the treatment of OA, the onset time is no later than 3 months, and the time to maintain the curative effect is no less than 12 months. However, these results should be generalized with caution due to the generally low quality of evidence and RCTs.
Collapse
|
8
|
Liu W, Lin H, Nie W, Wan J, Jiang Q, Zhang A. Exosomal miR-221-3p Derived from Bone Marrow Mesenchymal Stem Cells Alleviates Asthma Progression by Targeting FGF2 and Inhibiting the ERK1/2 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5910874. [PMID: 35990834 PMCID: PMC9385294 DOI: 10.1155/2022/5910874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 12/02/2022]
Abstract
Exosomes derived from human bone marrow mesenchymal stem cells (BMSCs) play potential protective roles in asthma. However, the underlying mechanisms remain not fully elucidated. Herein, exosomes were isolated from BMSCs, and the morphology, particle size, and exosome marker proteins were identified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot, respectively. Then airway smooth muscle cells (ASMCs) were treated with transforming growth factor-β1 (TGF-β1) to construct a proliferation model and then incubated with BMSCs-derived exosomes. We found that exosome incubation increased miR-221-3p expression and inhibited proliferation, migration, and the levels of extracellular matrix (ECM) proteins including fibronectin and collagen III. Moreover, FGF2 was identified as a target gene of miR-221-3p. FGF2 overexpression reversed the inhibitory effects of exosomal miR-221-3p on ASMC progression. Besides, the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) is inhibited by exosomal miR-221-3p, which was reversed by FGF2 overexpression. And ERK1/2 signaling activator reversed the effects of exosomal miR-221-3p on ASMC progression. Additionally, an ovalbumin (OVA)-induced asthmatic mice model was established, and exosome treatment alleviated airway hyper-responsiveness (AHR), histopathological damage, and ECM deposition in asthmatic mice. Taken together, our findings indicated that exosomal miR-221-3p derived from BMSCs inhibited FGF2 expression and the ERK1/2 signaling, thus attenuating proliferation, migration, and ECM deposition in ASMCs and alleviating asthma progression in OVA-induced asthmatic mice. Our findings may provide a novel therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Weike Liu
- Department of Pediatrics, Chengyang District People's Hospital of Qingdao, Qingdao 266000, Shandong, China
| | - Hui Lin
- Department of Pediatrics, Chengyang District People's Hospital of Qingdao, Qingdao 266000, Shandong, China
| | - Wuhui Nie
- Department of Pediatrics, Chengyang District People's Hospital of Qingdao, Qingdao 266000, Shandong, China
| | - Jieting Wan
- Department of Haemodialysis, Jimo District People's Hospital of Qingdao, Qingdao 266000, Shandong, China
| | - Qian Jiang
- Department of Pediatrics, Chengyang District People's Hospital of Qingdao, Qingdao 266000, Shandong, China
| | - Aimei Zhang
- Department of Pediatrics, Chengyang District People's Hospital of Qingdao, Qingdao 266000, Shandong, China
| |
Collapse
|
9
|
Karagergou E, Ligomenou T, Chalidis B, Kitridis D, Papadopoulou S, Givissis P. Evaluation of Adipose Cell-Based Therapies for the Treatment of Thumb Carpometacarpal Joint Osteoarthritis. Biomolecules 2022; 12:473. [PMID: 35327665 PMCID: PMC8946069 DOI: 10.3390/biom12030473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/04/2022] [Accepted: 03/18/2022] [Indexed: 11/16/2022] Open
Abstract
Adipose tissue and its regenerative products which are isolated with enzymatic or mechanical processing of the harvested fat have been studied in a wide range of degenerative diseases, including osteoarthritis of the knee and hip. Intra-articular injection of these products can provide symptomatic relief of pain and postpone surgery. However, their use in the treatment of thumb carpometacarpal joint (CMCJ) osteoarthritis is limited and just a few studies have been published on that topic. For this reason, a review of the literature was performed by a thorough search of eight terms using the Pubmed database. In total, seven human studies met the selection criteria, including case-control studies, case-series and one case report. In all studies, intra-articular injection of autologous fat in osteoarthritic thumb CMCJ provided reduction in pain and improvement in hand function. Grip and pinch strength showed variable results, from no change to significant improvement. Fat-processing techniques were based on centrifugation and mechanical homogenization but biological characterization of the injected cells was not performed in any study. Although the results are encouraging, a uniformly standardized method of fat processing and the conduction of randomized controlled trials in the future could better evaluate the effectiveness of this procedure for thumb CMCJ osteoarthritis.
Collapse
Affiliation(s)
- Eleni Karagergou
- Department of Burns, Plastic Surgery and Hand Surgery, Georgios Papanikolaou Hospital, 57010 Thessaloniki, Greece; (T.L.); (S.P.)
| | - Theodora Ligomenou
- Department of Burns, Plastic Surgery and Hand Surgery, Georgios Papanikolaou Hospital, 57010 Thessaloniki, Greece; (T.L.); (S.P.)
| | - Byron Chalidis
- 1st Orthopaedic Department, School of Medicine, Georgios Papanikolaou Hospital, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (B.C.); (D.K.); (P.G.)
| | - Dimitrios Kitridis
- 1st Orthopaedic Department, School of Medicine, Georgios Papanikolaou Hospital, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (B.C.); (D.K.); (P.G.)
| | - Sophia Papadopoulou
- Department of Burns, Plastic Surgery and Hand Surgery, Georgios Papanikolaou Hospital, 57010 Thessaloniki, Greece; (T.L.); (S.P.)
| | - Panagiotis Givissis
- 1st Orthopaedic Department, School of Medicine, Georgios Papanikolaou Hospital, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; (B.C.); (D.K.); (P.G.)
| |
Collapse
|
10
|
Yin B, Ni J, Witherel CE, Yang M, Burdick JA, Wen C, Wong SHD. Harnessing Tissue-derived Extracellular Vesicles for Osteoarthritis Theranostics. Theranostics 2022; 12:207-231. [PMID: 34987642 PMCID: PMC8690930 DOI: 10.7150/thno.62708] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/29/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent chronic whole-joint disease characterized by low-grade systemic inflammation, degeneration of joint-related tissues such as articular cartilage, and alteration of bone structures that can eventually lead to disability. Emerging evidence has indicated that synovium or articular cartilage-secreted extracellular vesicles (EVs) contribute to OA pathogenesis and physiology, including transporting and enhancing the production of inflammatory mediators and cartilage degrading proteinases. Bioactive components of EVs are known to play a role in OA include microRNA, long non-coding RNA, and proteins. Thus, OA tissues-derived EVs can be used in combination with advanced nanomaterial-based biosensors for the diagnostic assessment of OA progression. Alternatively, mesenchymal stem cell- or platelet-rich plasma-derived EVs (MSC-EVs or PRP-EVs) have high therapeutic value for treating OA, such as suppressing the inflammatory immune microenvironment, which is often enriched by pro-inflammatory immune cells and cytokines that reduce chondrocytes apoptosis. Moreover, those EVs can be modified or incorporated into biomaterials for enhanced targeting and prolonged retention to treat OA effectively. In this review, we explore recently reported OA-related pathological biomarkers from OA joint tissue-derived EVs and discuss the possibility of current biosensors for detecting EVs and EV-related OA biomarkers. We summarize the applications of MSC-EVs and PRP-EVs and discuss their limitations for cartilage regeneration and alleviating OA symptoms. Additionally, we identify advanced therapeutic strategies, including engineered EVs and applying biomaterials to increase the efficacy of EV-based OA therapies. Finally, we provide our perspective on the future of EV-related diagnosis and therapeutic potential for OA treatment.
Collapse
Affiliation(s)
- Bohan Yin
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Junguo Ni
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | | | - Mo Yang
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, PA 16802, USA.,✉ Corresponding authors: Jason A. Burdick: . Chunyi Wen: . Siu Hong Dexter Wong:
| | - Chunyi Wen
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China.,Research Institute of Smart Ageing, the Hong Kong Polytechnic University, Hong Kong, 999077, China.,✉ Corresponding authors: Jason A. Burdick: . Chunyi Wen: . Siu Hong Dexter Wong:
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, the Hong Kong Polytechnic University, Hong Kong, 999077, China.,✉ Corresponding authors: Jason A. Burdick: . Chunyi Wen: . Siu Hong Dexter Wong:
| |
Collapse
|
11
|
Sharma S, Muthu S, Jeyaraman M, Ranjan R, Jha SK. Translational products of adipose tissue-derived mesenchymal stem cells: Bench to bedside applications. World J Stem Cells 2021; 13:1360-1381. [PMID: 34786149 PMCID: PMC8567449 DOI: 10.4252/wjsc.v13.i10.1360] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/02/2021] [Accepted: 08/30/2021] [Indexed: 02/06/2023] Open
Abstract
With developments in the field of tissue engineering and regenerative medicine, the use of biological products for the treatment of various disorders has come into the limelight among researchers and clinicians. Among all the available biological tissues, research and exploration of adipose tissue have become more robust. Adipose tissue engineering aims to develop by-products and their substitutes for their regenerative and immunomodulatory potential. The use of biodegradable scaffolds along with adipose tissue products has a major role in cellular growth, proliferation, and differentiation. Adipose tissue, apart from being the powerhouse of energy storage, also functions as the largest endocrine organ, with the release of various adipokines. The progenitor cells among the heterogeneous population in the adipose tissue are of paramount importance as they determine the capacity of regeneration of these tissues. The results of adipose-derived stem-cell assisted fat grafting to provide numerous growth factors and adipokines that improve vasculogenesis, fat graft integration, and survival within the recipient tissue and promote the regeneration of tissue are promising. Adipose tissue gives rise to various by-products upon processing. This article highlights the significance and the usage of various adipose tissue by-products, their individual characteristics, and their clinical applications.
Collapse
Affiliation(s)
- Shilpa Sharma
- Department of Pediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
| | - Sathish Muthu
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul, Tamil Nadu 624304, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
| | - Madhan Jeyaraman
- Indian Stem Cell Study Group, Lucknow, Uttar Pradesh 226010, India
- Research Scholar, Department of Biotechnology, School of Engineering and Technology, Greater Noida, Sharda University, Uttar Pradesh 201306, India
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India.
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201306, India
| |
Collapse
|
12
|
Lu X, Wu J, Qin Y, Liang J, Qian H, Song J, Qu C, Liu R. Identification of N-glycoproteins of hip cartilage in patients with osteonecrosis of femoral head using quantitative glycoproteomics. Int J Biol Macromol 2021; 187:892-902. [PMID: 34331982 DOI: 10.1016/j.ijbiomac.2021.07.159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/04/2021] [Accepted: 07/23/2021] [Indexed: 10/20/2022]
Abstract
N-glycosylation is a major post-translational modification of proteins and involved in many diseases, however, the state and role of N-glycosylation in cartilage degeneration of osteonecrosis of femoral head (ONFH) remain unclear. The aim of this study is to identify the glycoproteins of ONFH hip cartilage. Cartilage tissues were collected from nine patients with ONFH and nine individuals with traumatic femoral neck fracture. Cartilage glycoproteins were identified by glycoproteomics based on LC-MS/MS. The differentially N-glycoproteins including glycosites were identified in ONFH and controls. A total of 408 N-glycoproteins with 444 N-glycosites were identified in ONFH and control cartilage. Among them, 104 N-glycoproteins with 130 N-glycosites were significantly differential in ONFH and control cartilage, which including matrix-remodeling-associated protein 5, prolow-density lipoprotein receptor-related protein 1, clusterin and lysosome-associated membrane glycoprotein 2. Gene Ontology analysis revealed the significantly differential glycoproteins mainly belonged to protein metabolic process, single-multicellular organism process, proteolysis, biological adhesion and cell adhesion. KEGG pathway and protein-protein interaction analysis suggested that the significantly differential glycoproteins were associated with PI3K-Akt signalling pathway, ECM-receptor interaction, protein processing in the endoplasmic reticulum and N-glycan biosynthesis. This information provides substantial insight into the role of protein glycosylation in the development of cartilage degeneration of ONFH patients.
Collapse
Affiliation(s)
- Xueliang Lu
- Department of Orthopedics, the Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, China; Department of Orthopedics, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan Province 471003, China
| | - Junlong Wu
- Department of Orthopedics, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan Province 471009, China
| | - Yannan Qin
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jialin Liang
- Department of Orthopedics, the Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, China
| | - Hang Qian
- Department of Orthopedics, the Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, China
| | - Jidong Song
- Department of Orthopedics, the Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, China
| | - Chengjuan Qu
- Department of Odontology, Umeå University, Umeå 90185, Sweden
| | - Ruiyu Liu
- Department of Orthopedics, the Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 Xiwu Road, Xi'an 710004, China.
| |
Collapse
|
13
|
Wu HY, Zhang XC, Jia BB, Cao Y, Yan K, Li JY, Tao L, Jie ZG, Liu QW. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate acetaminophen-induced acute liver failure through activating ERK and IGF-1R/PI3K/AKT signaling pathway. J Pharmacol Sci 2021; 147:143-155. [PMID: 34294366 DOI: 10.1016/j.jphs.2021.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the therapeutic potential of human umbilical cord mesenchymal stem cells derived exosomes (hUCMSC-Exo) in acute liver failure (ALF) in mice as well as its underlying mechanism. We found that a single tail vein administration of hucMSC-Exo effectively enhanced the survival rate, inhibited apoptosis in hepatocytes, and improved liver function in APAP-induced mouse model of ALF. Furthermore, the deletion of glutathione (GSH) and superoxide dismutase (SOD), generation of malondialdehyde (MDA), and the over production of cytochrome P450 E1 (CYP2E1) and 4-hydroxynonenal (4-HNE) caused by APAP were also inhibited by hucMSC-Exo, indicating that hucMSC-Exo inhibited APAP-induced apoptosis of hepatocytes by reducing oxidative stress. Moreover, hucMSC-Exo significantly down-regulated the levels of inflammatory cytokines IL-6, IL-1β, and TNF-α in APAP-treated livers. Western blot showed that hucMSC-Exo significantly promoted the activation of ERK1/2 and IGF-1R/PI3K/AKT signaling pathways in APAP-injured LO2 cells, resulting in the inhibition of apoptosis of LO2 cells. Importantly, PI3K inhibitor LY294002 and ERK1/2 inhibitor PD98059 could reverse the function of hucMSC-Exo on APAP-injured LO2 cells in some extent. Our results suggest that hucMSC-Exo offer antioxidant hepatoprotection against APAP in vitro and in vivo by inhibitiing oxidative stress-induced apoptosis via upregulation of ERK1/2 and PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Han-You Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, PR China
| | - Xiang-Cheng Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Bing-Bing Jia
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310013, PR China
| | - Ye Cao
- Information Engineering School of NanChang University, Nanchang, 330031, PR China
| | - Kai Yan
- Department of Pediatrics, The First Affiliated Hospital of NanChang University, NanChang, 330006, PR China
| | - Jing-Yuan Li
- School of Chemistry, Biology and Material Science, East China University of Technology, Nanchang, 330013, China
| | - Li Tao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Zhi-Gang Jie
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Quan-Wen Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, PR China.
| |
Collapse
|
14
|
Cai Q, Yin F, Hao L, Jiang W. Research Progress of Mesenchymal Stem Cell Therapy for Severe COVID-19. Stem Cells Dev 2021; 30:459-472. [PMID: 33715385 DOI: 10.1089/scd.2020.0198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Corona virus disease 2019 (COVID-19) refers to a type of pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Sixty million confirmed cases have been reported worldwide until November 29, 2020. Unfortunately, the novel coronavirus is extremely contagious and the mortality rate of severe and critically ill patients is high. Thus, there is no definite and effective treatment in clinical practice except for antiviral therapy and supportive therapy. Mesenchymal stem cells (MSCs) are not only characterized by low immunogenicity and homing but also have anti-inflammatory and immunomodulation characteristics. Furthermore, they can inhibit the occurrence and development of a cytokine storm, inhibit lung injury, and exert antipulmonary fibrosis and antioxidative stress, therefore MSC therapy is expected to become one of the effective therapies to treat severe COVID-19. This article will review the possible mechanisms of MSCs in the treatment of severe COVID-19.
Collapse
Affiliation(s)
- Qiqi Cai
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, China
| | - Fei Yin
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, China
| | - Liming Hao
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, China
| | - Wenhua Jiang
- Department of Histology and Embryology, Basic Medical College of Jilin University, Changchun, China
| |
Collapse
|
15
|
Li D, Gupta P, Sgaglione NA, Grande DA. Exosomes Derived from Non-Classic Sources for Treatment of Post-Traumatic Osteoarthritis and Cartilage Injury of the Knee: In Vivo Review. J Clin Med 2021; 10:jcm10092001. [PMID: 34066986 PMCID: PMC8124969 DOI: 10.3390/jcm10092001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis of the knee is one of the most common chronic, debilitating musculoskeletal conditions. Current conservative treatment modalities such as weight loss, non-steroidal anti-inflammatory drugs, and intra-articular steroid injections often only provide temporary pain relief and are unsatisfactory for long-term management. Though end stage osteoarthritis of the knee can be managed with total knee arthroplasty (TKA), finding alternative non-surgical options to delay or prevent the need for TKA are needed due to the increased healthcare costs and expenditures associated with TKA. Exosomes have been of particular interest given recent findings highlighting that stem cells may at least partially mediate some of their effects through the release of extracellular vesicles, such as exosomes. As such, better understanding the biological mechanisms and potential therapeutic effects of these exosomes is necessary. Here, we review in vivo studies that highlight the potential clinical use of exosomes derived from non-classical sources (not bone marrow or adipose derived MSCs derived MSCs) for osteoarthritis of the knee.
Collapse
Affiliation(s)
- Dan Li
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, NY 11030, USA;
| | - Puneet Gupta
- Department of Orthopaedic Surgery, George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA;
| | - Nicholas A. Sgaglione
- Department of Orthopaedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA;
| | - Daniel A. Grande
- Orthopedic Research Laboratory, The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, NY 11030, USA;
- Department of Orthopaedic Surgery, Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY 11040, USA;
- Correspondence: ; Tel.: +1-(516)-562-1138
| |
Collapse
|
16
|
Li GQ, Fang YX, Liu Y, Meng FR, Wu X, Zhang CW, Zhang Y, Liu YQ, Liu D. MicroRNA-21 from bone marrow mesenchymal stem cell-derived extracellular vesicles targets TET1 to suppress KLF4 and alleviate rheumatoid arthritis. Ther Adv Chronic Dis 2021; 12:20406223211007369. [PMID: 33995992 PMCID: PMC8072656 DOI: 10.1177/20406223211007369] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/15/2021] [Indexed: 12/31/2022] Open
Abstract
Background Accumulating evidence has demonstrated that bone marrow mesenchymal stem cells (BMSCs)-derived extracellular vesicles (EVs) can be used effectively to transfer drugs and biomolecules to target lesions. Meanwhile, BMSCs have been reported to be beneficial in the treatment of rheumatoid arthritis (RA). In this study, we employ gain- and loss-of-function experiments to determine how BMSCs-derived EVs alleviate RA in vitro and in vivo. Methods We isolated EVs from BMSCs and characterized them by transmission electron microscopy and western blot analysis. The regulatory relationship between miR-21 and TET1 was predicted by bioinformatics analysis and validated by dual luciferase assay. Next, we utilized bisulfite sequencing PCR to decipher how TET1 promoted KLF4 transcription. Then, we established an RA mouse model and determined the role of miR-21 in RA progression. Functional assays were used to validate the role the miR-21-TET1-KLF4 regulatory axis in controlling mouse fibroblast-like synoviocytes (mFLS) cell proliferation and inflammatory cytokines secretion in vitro. Results RT-qPCR results revealed that miR-21 was highly expressed in BMSCs-derived EVs, and confirmed that BMSCs-derived EVs transferred miR-21 into mFLS cells. Bioinformatic analysis predicted that TET1 was the directly downstream target of miR-21, which was further validated by dual luciferase assay. TET1 promoted KLF4 promoter methylation to increase its expression. Collectively, BMSCs-derived EVs relieved RA by delivering miR-21, while the exosomal miR-21 alleviated RA through targeting the TET1/KLF4 regulatory axis. Conclusion miR-21 from BMSCs-derived EVs suppresses KLF4 to relive RA by targeting TET1.
Collapse
Affiliation(s)
- Guo-Qing Li
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, P. R. China
| | - Yu-Xuan Fang
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, P. R. China
| | - Ying Liu
- Clinical Medical College, Dalian Medical University, Dalian, P. R. China
| | - Fan-Ru Meng
- Clinical Medical College, Dalian Medical University, Dalian, P. R. China
| | - Xia Wu
- Clinical Medical College, Dalian Medical University, Dalian, P. R. China
| | - Chun-Wang Zhang
- Clinical Medical College, Dalian Medical University, Dalian, P. R. China
| | - Yu Zhang
- Medical College of Yangzhou University, Yangzhou, P. R. China
| | - Yan-Qing Liu
- Medical College of Yangzhou University, Yangzhou, P. R. China
| | - Dan Liu
- Department of Pathology, Clinical Medical College, Yangzhou University, No. 98, Nantong West Road, Yangzhou, Jiangsu Province 225001, P. R. China
| |
Collapse
|
17
|
Xiong M, Zhang Q, Hu W, Zhao C, Lv W, Yi Y, Wu Y, Wu M. Exosomes From Adipose-Derived Stem Cells: The Emerging Roles and Applications in Tissue Regeneration of Plastic and Cosmetic Surgery. Front Cell Dev Biol 2020; 8:574223. [PMID: 33015067 PMCID: PMC7511773 DOI: 10.3389/fcell.2020.574223] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/20/2020] [Indexed: 12/21/2022] Open
Abstract
Adipose-derived stem cells (ASCs) are an important stem cell type separated from adipose tissue, with the properties of multilineage differentiation, easy availability, high proliferation potential, and self-renewal. Exosomes are novel frontiers of intercellular communication regulating the biological behaviors of cells, such as angiogenesis, immune modulation, proliferation, and migration. ASC-derived exosomes (ASC-exos) are important components released by ASCs paracrine, possessing multiple biological activities. Tissue regeneration requires coordinated “vital networks” of multiple growth factors, proteases, progenitors, and immune cells producing inflammatory cytokines. Recently, as cell-to-cell messengers, ASC-exos have received much attention for the fact that they are important paracrine mediators contributing to their suitability for tissue regeneration. ASC-exos, with distinct properties by encapsulating various types of bioactive cargoes, are endowed with great application potential in tissue regeneration, mechanically via the migration and proliferation of repair cells, facilitation of the neovascularization, and other specific functions in different tissues. Here, this article elucidated the research progress of ASC-exos about tissue regeneration in plastic and cosmetic surgery, including skin anti-aging therapy, dermatitis improvement, wound healing, scar removal, flap transplantation, bone tissue repair and regeneration, obesity prevention, fat grafting, breast cancer, and breast reconstruction. Deciphering the biological properties of ASC-exos will provide further insights for exploring novel therapeutic strategies of tissue regeneration in plastic and cosmetic surgery.
Collapse
Affiliation(s)
- Mingchen Xiong
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhang
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijie Hu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chongru Zhao
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenchang Lv
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yi
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiping Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|