1
|
Zou B, Wang D, Zhong J, He Z, Zhou Y, Yang H, Liu Y, Zeng G, Duan X. Mesenchymal stem cells attenuate hyperoxaluria-induced kidney injury and crystal depositions via inhibiting the activation of NLRP3 inflammasome. Life Sci 2025; 371:123608. [PMID: 40194762 DOI: 10.1016/j.lfs.2025.123608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
AIMS Calcium oxalate (CaOx) is the predominant form of kidney stones, associated with significant morbidity and recurrence rates. Mesenchymal stem cells (MSCs) have shown promise in treating renal injury, but their impact on CaOx stone formation remains unclear. MATERIALS AND METHODS We established a hyperoxaluria-induced AKI model in mice through intraperitoneal injection of glyoxylate. Two types of MSCs, bone marrow-derived MSCs (BMSCs) and umbilical cord-derived mesenchymal stem cells (UMSCs), were injected through tail vein injection. Histological evaluations and blood biochemical tests were performed to assess crystal deposition and kidney function. The inflammatory response and NLRP3 inflammasome activation were assessed using immunofluorescence, immunohistochemistry, TUNEL staining, and qPCR. In vitro, macrophages were cocultured in the presence of MSCs. ELISA was used to measure IL-1β and IL-18 release. MTS assays assessed renal epithelial cell protection. Western blotting evaluated NLRP3 inflammasome activation in macrophages. KEY FINDINGS Both BMSCs and UMSCs significantly inhibited CaOx crystal deposition and kidney injury by inhibiting NLRP3 inflammasome activation. In vitro, both MSC types suppressed NLRP3 inflammasome activation in macrophages through the NF-κB signaling pathway, leading to decreased release of IL-1β and IL-18 and enhanced protection of renal epithelial cells. This attenuation of renal tubular cell injury is a critical factor in preventing CaOx stone formation. SIGNIFICANCE Our findings reveal that Both BMSCs and UMSCs effectively attenuate hyperoxaluria-induced kidney injury and crystal deposition by inhibiting NLRP3 inflammasome activation. This discovery is helpful for developing new effective therapeutic means for nephrolithiasis.
Collapse
Affiliation(s)
- Bangyu Zou
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University; Department of Urology, Changhai Hospital, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ding Wang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinghua Zhong
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University
| | - Zhiqing He
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University
| | - Yuhao Zhou
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University
| | - Houmeng Yang
- Department of Urology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No.2 Hospital), Ningbo, China
| | - Yongda Liu
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University
| | - Guohua Zeng
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University.
| | - Xiaolu Duan
- Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Urological Diseases, Guangzhou, China; Guangdong Engineering Research Center of Urinary Minimally Invasive Surgery Robot and Intelligent Equipment, Guangzhou, China; Guangzhou Institute of Urology, Guangzhou Medical University.
| |
Collapse
|
2
|
Saadh MJ, Ahmed HH, Sanghvi G, Bin Awang Isa MZ, Singh P, Kaur K, Kumar MR, Husseen B. Recent advances in the delivery of microRNAs via exosomes derived from MSCs, and their role in regulation of ferroptosis. Pathol Res Pract 2025; 270:155984. [PMID: 40315562 DOI: 10.1016/j.prp.2025.155984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025]
Abstract
Mesenchymal stem cell (MSC) therapy, with its unique properties, has garnered interest in cancer treatment. Exosomes (EXOs)-derived from MSC retain the paracrine components of MSCs and demonstrate increased stability, minimal immunogenicity, and low risk of unintended tumorigenesis. Enhanced endocytosis methods make them versatile delivery vehicles for therapeutic cargo. MSC-EXOs can either promote or inhibit carcinogenesis, mediated by paracrine factors and various RNA molecules, particularly microRNAs (miRNAs). The prospect of using MSC-EXOs as a delivery tool for antitumor miRNAs in solid tumor therapy is promising. Exosomes' intrinsic tumor-targeting abilities and low immunogenicity make them ideal for delivering miRNAs, which have shown potential as cancer therapeutics. miRNAs within MSC-EXOs molecules can stimulate tumor growth or induce non-apoptotic cell death pathways, such as ferroptosis, depending on context. Ferroptosis is a kind of controlled cell death that is associated with the pathophysiology of several illnesses and includes iron metabolism. There is growing evidence that miRNAs carried by exosomes derived from MSCs may control ferroptosis in tumor cells by altering key genes related to antioxidant defense, lipid peroxidation, and iron metabolism. Understanding their complex mechanisms in the tumor microenvironment and optimizing their cargo are critical steps toward harnessing their full therapeutic potential. This review provides a comprehensive overview of MSC-EXOs and their role in cancer treatment. We also discuss the potential of MSC-EXOs as delivery vehicles for miRNAs to enhance therapeutic efficacy, as well as the role of exosomal miRNAs in the induction of ferroptosis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | | | - Priyanka Singh
- NIMS School of Allied Sciences and Technology, NIMS University, Jaipur, Rajasthan 303121, India
| | - Kiranjeet Kaur
- Chandigarh Pharmacy College, Chandigarh Group of colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Beneen Husseen
- Medical laboratory technique college, the Islamic University, Najaf, Iraq; Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| |
Collapse
|
3
|
Ruan T, Han J, Xue C, Wang F, Lin J. Mesenchymal stem cells protect the integrity of the alveolar epithelial barrier through extracellular vesicles by inhibiting MAPK-mediated necroptosis. Stem Cell Res Ther 2025; 16:250. [PMID: 40390004 PMCID: PMC12090679 DOI: 10.1186/s13287-025-04388-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Alveolar‒capillary barrier disruption is a hallmark of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). The contribution of necroptosis to the compromised alveolar-barrier in ALI remains unclear. Mesenchymal stem cells (MSCs) may contribute to tissue repair in ALI and ARDS. Here we evaluated the efficacy and explored the molecular mechanisms of menstrual blood-derived endometrial stem cells (MenSCs) and MenSC-derived extracellular vesicles (MenSC-EVs) in ALI-induced alveolar epithelial barrier dysfunction. METHODS Human lung epithelial cells were stimulated with endotoxin and treated with MenSCs or MenSC-EVs, and their barrier properties were evaluated. Lipopolysaccharide (LPS)-injured mice were treated with MenSCs or MSC-EVs, and the degree of lung injury and the alveolar epithelial barrier of the lung tissue were assessed. RESULTS We found that MenSCs reduced lung injury and restored alveolar-barrier integrity in lung tissue. In vitro, MenSCs reduced paracellular permeability and restored barrier integrity in human lung epithelial cells. MenSC-EVs replicated all these MenSC-mediated changes. Mechanistic research revealed that MenSCs inhibited MAPK signaling and necroptosis. JNK inhibition SP600125, and ERK inhibition U0126 or inhibition of necroptosis with Nec-1 or GSK872 diminished the beneficial anti-epithelial barrier dysfunction effects of MenSCs or MenSC-EVs. CONCLUSIONS Our results suggest that human menstrual blood-derived endometrial stem cells mitigate lung injury and improve alveolar barrier properties by inhibiting MAPK-mediated necroptosis through extracellular vesicles, supporting the application of MenSCs or MenSC-derived extracellular vesicles to treat ALI or ARDS.
Collapse
Affiliation(s)
- Tao Ruan
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jiaming Han
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chengxu Xue
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Fengyuan Wang
- Stem Cell and Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
4
|
Lan D, Zhang D, Dai X, Cai J, Zhou H, Song T, Wang X, Kong Q, Tang Z, Tan J, Zhang J. Mesenchymal stem cells and exosomes: A novel therapeutic approach for aging. Exp Gerontol 2025; 206:112781. [PMID: 40349806 DOI: 10.1016/j.exger.2025.112781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Mesenchymal stem cells (MSCs), a vital component of the adult stem cell repertoire, are distinguished by their dual capacity for self-renewal and multilineage differentiation. The therapeutic effects of MSCs are primarily mediated through mechanisms such as homing, paracrine signaling, and cellular differentiation. Exosomes (Exos), a type of extracellular vesicles (EVs) secreted by MSCs via the paracrine pathway, play a pivotal role in conveying the biological functions of MSCs. Accumulating evidence from extensive research underscores the remarkable anti-aging potential of both MSCs and their Exos. This review comprehensively explores the impact of MSCs and their Exos on key hallmarks of aging, including genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, impaired macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. Furthermore, this paper highlights emerging strategies and novel approaches for modulating the aging process, offering insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Dongfeng Lan
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Dan Zhang
- Zunyi Medical University Library, Zunyi 563000, China
| | - Xiaofang Dai
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Ji Cai
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - He Zhou
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Tao Song
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
| | - Qinghong Kong
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
| | - Zhengzhen Tang
- Department of Pediatrics, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi 563000, China.
| | - Jun Tan
- Department of Histology and Embryology, Zunyi Medical University, Zunyi 563000, China.
| | - Jidong Zhang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
5
|
Gao Q, Su Z, Pang X, Chen J, Luo R, Li X, Zhang C, Zhao Y. Overexpression of Heme Oxygenase 1 Enhances the Neuroprotective Effects of Exosomes in Subarachnoid Hemorrhage by Suppressing Oxidative Stress and Endoplasmic Reticulum Stress. Mol Neurobiol 2025; 62:6088-6101. [PMID: 39710823 DOI: 10.1007/s12035-024-04651-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
AIMS This study aims to elucidate the therapeutic effects and underlying mechanisms of exosomes derived from Heme oxygenase 1 (HO-1)-overexpressing human umbilical cord mesenchymal stem cells (ExoHO-1) in a subarachnoid hemorrhage (SAH) mouse model. METHODS In this study, exosomes were identified using Western blotting, particle analysis, and transmission electron microscopy. The effect of ExoHO-1 and ExoCtrl on the neurological function of SAH mice was assessed using the Garcia scoring system, Beam balance, Rotarod test, and Morris water maze test. Neuronal apoptosis and survival were evaluated through TUNEL and Nissl staining. Levels of oxidative and endoplasmic reticulum stress were measured via immunofluorescence, Western blotting, DHE staining, enzyme-linked immunosorbent assay, and commercial kits. RESULTS HO-1-overexpressing human umbilical cord mesenchymal stem cells encapsulated HO-1 into their exosomes. ExoHO-1 significantly enhanced both short-term and long-term neurological function protection. By reducing the activation of the PERK/CHOP/Caspase12 pathway and decreasing oxidative stress levels, ExoHO-1 effectively inhibited neuronal apoptosis in the ipsilateral temporal cortex. CONCLUSION ExoHO-1 enhances the therapeutic efficacy of exosomes in SAH mice by countering neuronal apoptosis, primarily through the suppression of oxidative and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Qiongqiong Gao
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Zhumin Su
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiangxiong Pang
- Translational Medicine Research Center, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China
| | - Jinshuo Chen
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ruixiang Luo
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiaoyang Li
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Chi Zhang
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Yun Zhao
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
- Translational Medicine Research Center, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
6
|
Fu H, Cheng J, Hu L, Heng BC, Zhang X, Deng X, Liu Y. Mitochondria-targeting materials and therapies for regenerative engineering. Biomaterials 2025; 316:123023. [PMID: 39708774 DOI: 10.1016/j.biomaterials.2024.123023] [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: 09/09/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
The hemostatic, inflammatory, proliferative, and remodeling phases of healing require precise spatiotemporal coordination and orchestration of numerous biological processes. As the primary energy generators in the cell, mitochondria play multifunctional roles in regulating metabolism, stress reactions, immunity, and cell density during the process of tissue regeneration. Mitochondrial dynamics involves numerous crucial processes, fusion, fission, autophagy, and translocation, which are all necessary for preserving mitochondrial function, distributing energy throughout cells, and facilitating cellular signaling. Tissue regeneration is specifically associated with mitochondrial dynamics due to perturbations of Ca2+, H2O2 and ROS levels, which can result in mitochondrial malfunction. Increasing evidence from multiple models suggests that clinical interventions or medicinal drugs targeting mitochondrial dynamics could be a promising approach. This review highlights significant advances in the understanding of mitochondrial dynamics in tissue regeneration, with specific attention on mitochondria-targeting biomaterials that accelerate multiple tissues' regeneration by regulating mitochondrial metabolism. The innovations in nanomaterials and nanosystems enhance mitochondrial-targeting therapies are critically examined with the prospects of modulating mitochondrial dynamics for new therapies in regenerative engineering.
Collapse
Affiliation(s)
- Hongying Fu
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Jingrong Cheng
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Le Hu
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Boon Chin Heng
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Xuliang Deng
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Yang Liu
- Department of Dental Materials & Dental Medical Devices Testing Center & NMPA Key Laboratory for Dental Materials & Beijing Key Laboratory of Digital Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & National Center for Stomatology & National Clinical Research Center for Oral Diseases & NHC Research Center of Engineering and Technology for Computerized Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, China; Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| |
Collapse
|
7
|
Chen FD, Zhang B, Wang LL, Jia YL, Zeng Q, Fan T, Wang HY, Xiong MF, Lin YX, Zhou JN, Yue W, Chen L, Xi JF. DSUP modified mesenchymal stem cells exert significant radiation protective effect by enhancing the hematopoietic niche. Stem Cell Res Ther 2025; 16:216. [PMID: 40312405 PMCID: PMC12045013 DOI: 10.1186/s13287-025-04300-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 04/01/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Radiation induced hematopoietic failure was the primary cause of death after exposure to a moderate or high dose of whole body irradiation, causing increased challenge for nuclear or radiological treatment, so it is an urgent need to develop radioprotectors for attenuating hematopoietic damage caused by acute radiation syndrome (ARS). Given the excellent therapeutic effects and special benefits of mesenchymal stem cells (MSCs) in radiation damaged hematopoietic stem/progenitor cells (HSPCs) recovery and hematopoietic niche reconstruction, enhancing the hematopoietic niche with the radiotolerance MSCs can be an alternative solution to prevent and attenuate hematopoietic radiation damage, which needs to be studied. METHODS Here, we constructed MSCs modified with Damage Suppressor Protein (DSUP), a radiotolerance gene identified from tardigrade Ramazzotius varieornatus, and verify its radiation protection effect in HSPCs-MSCs co-culture model in vitro and radiation damaged mice model in vivo. RESULTS Our results showed that DSUP protein had no significant toxic side effects on the basic stemness properties and differentiation potential of MSCs, and significantly enhanced the radiation tolerance and DNA protection ability of MSCs. Compared with the control (CON) group MSCs, the DSUP modified MSCs after radiation damage suffered less DNA damage, preserved most of proliferation activity and migration ability. In the HSPCs-MSCs co-culture model, DSUP modified MSCs have significant protective effect on HSPCs by providing a functional hematopoietic niche after radiation damage. The DSUP group irradiated HSPCs exhibited more rapid recovery, the higher HSPCs ratio and better hematopoietic differentiation potential. In animal studies, pre infusion of DSUP modified MSCs reduce irradiated mice mortality rate, reduce hematopoietic failure incidence, and provide a protective effect against radiation injury by protecting hematopoietic microenvironment and promoting HSCs recovery. DSUP modified MSCs can be used as a radioprotector and existed significant radiation protection effect for ARS at doses below 7 Gy total-body irradiation (TBI) of X-ray in both immunodeficient and immunocompetent mice models. CONCLUSIONS DSUP modified MSCs may serve as a new radioprotector for ARS. DSUP modified MSCs could attenuate radiation damage of HSPCs and promote HSPCs rapid recovery as well as hematopoietic reconstruction by providing a more functional niche after radiation, thereby reducing the occurrence of hematopoietic failure and improving survival rate.
Collapse
Affiliation(s)
- Fu-Dong Chen
- Medical School of Chinese PLA: Chinese, PLA General Hospital, Beijing, 100039, China
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
- Department of General Medicine, The First Center of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Biao Zhang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Li-Li Wang
- Department of General Medicine, The First Center of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Ya-Li Jia
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Quan Zeng
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Tao Fan
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Hai-Yang Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Ming-Fang Xiong
- Medical School of Chinese PLA: Chinese, PLA General Hospital, Beijing, 100039, China
| | - Ying-Xue Lin
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jun-Nian Zhou
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| | - Li Chen
- Medical School of Chinese PLA: Chinese, PLA General Hospital, Beijing, 100039, China.
- Department of General Medicine, The First Center of the Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jia-Fei Xi
- Medical School of Chinese PLA: Chinese, PLA General Hospital, Beijing, 100039, China.
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| |
Collapse
|
8
|
Wang W, Wang Y, Duan C, Tian W, Gao L. LncRNA NEAT1-206 regulates autophagy of human umbilical cord mesenchymal stem cells through the WNT5A/Ca 2+ signaling pathway under senescence stress. Noncoding RNA Res 2025; 11:234-248. [PMID: 39896347 PMCID: PMC11786084 DOI: 10.1016/j.ncrna.2024.12.013] [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: 10/12/2024] [Revised: 12/21/2024] [Accepted: 12/31/2024] [Indexed: 02/04/2025] Open
Abstract
Stem cells are crucial for maintaining bodily stability, but their regenerative abilities decline with age. This decline is marked by reduced proliferation and differentiation capacities of stem cells, as well as exhaustion of the stem cell pool. The accumulation of aged mesenchymal stem cells (MSCs) can reduce the tissue regeneration, but the molecular mechanisms influencing MSCs aging remain unclear. Moreover, collecting MSCs from elderly individuals is not suitable for observing the early response of MSCs to senescence stress, and the factors involved in early senescence remain unclear. In our previous study, we established a fast MSC aging model using D-galactose. We discovered that, while not affecting the "stemness" markers of mesenchymal stem cells, the expression of LncRNA NEAT1-206 was notably increased during the early stages of aging induction (within 4 days). And LncRNA NEAT1-206 was observed to be localized in the cytoplasmic matrix due to enhanced nuclear export. We found that the LncRNA NEAT1-206 could trigger autophagy through the WNT5A/Ca2+ signaling pathway, thereby decreasing senescence markers and enhancing the osteogenic differentiation of MSCs. This study elucidated the role that LncRNA NEAT1-206 as a potential key factor in conferring resistance to D-galactose-induced cell senescence at the early stage and promoting the osteogenic differentiation of MSCs. This study may provide a foundational understanding for delaying the MSCs aging process.
Collapse
Affiliation(s)
- Weili Wang
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Yongyu Wang
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Chunchun Duan
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Wenjing Tian
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Liyang Gao
- Life Science School, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| |
Collapse
|
9
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2025; 9:e2400297. [PMID: 39390651 PMCID: PMC12001015 DOI: 10.1002/adbi.202400297] [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: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
| | - Peiwen Wang
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Tianyu Wang
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Yanhui Chu
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
| |
Collapse
|
10
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2025; 70:103-124. [PMID: 38729561 PMCID: PMC11976416 DOI: 10.1016/j.jare.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
11
|
Lukacheva AV, Bogachev MI, Musorina AS, Kriger DV, Poljanskaya GG, Bobkov DE. It's Not Just About Speed: Single-Cell Tracking Reveals Changes in MSC Motility Associated with Replicative Senescence. Stem Cell Rev Rep 2025:10.1007/s12015-025-10868-x. [PMID: 40156639 DOI: 10.1007/s12015-025-10868-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Mesenchymal stem cells, cultured in 2D, are motile fibroblast-like cells with a well-developed actin cytoskeleton. In this study, we analyzed changes in cell motility during long-term cultivation accompanied by replicative senescence (RS) for DF2 and MSCWJ-1 cell lines derived from various sources and donors of different age under both normal and inflammatory conditions, the latter obtained by treatment with lysophosphatidic acid (LPA). Our results indicate that RS is associated with non-stationary alterations in the average migration speed: while the median speed derived from single-cell tracking is unaffected by the senescence stage, the average speed in young cells is enhanced due to the contribution of a subpopulation of fast-moving cells. The sensitivity of cell motility metrics to the impact of LPA varied depending on their origin, with the most pronounced effects observed during the initial passages. Using multivariate statistical analysis, we have shown explicitly that the common motility metrics (average and maximum speed, distance, sinuosity of trajectories, etc.) are associated with the passage, thus clearly reflecting senescence effect. Altogether, our results indicate that cell motility exhibits complex alterations with RS, with multiple metrics besides the average speed being affected and associated with their RS stage.
Collapse
Affiliation(s)
| | - Mikhail I Bogachev
- Biomedical Engineering Research Centre, St. Petersburg Electrotechnical University "LETI", St. Petersburg, 197022, Russia
| | | | - Darya V Kriger
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Galina G Poljanskaya
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Danila E Bobkov
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| |
Collapse
|
12
|
Wang JJ, Zheng Y, Li YL, Xiao Y, Ren YY, Tian YQ. Emerging role of mesenchymal stem cell-derived exosomes in the repair of acute kidney injury. World J Stem Cells 2025; 17:103360. [PMID: 40160687 PMCID: PMC11947899 DOI: 10.4252/wjsc.v17.i3.103360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/26/2024] [Accepted: 02/13/2025] [Indexed: 03/21/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid deterioration in kidney function and has a significant impact on patient health and survival. Mesenchymal stem cells (MSCs) have the potential to enhance renal function by suppressing the expression of cell cycle inhibitors and reducing the expression of senescence markers and microRNAs via paracrine and endocrine mechanisms. MSC-derived exosomes can alleviate AKI symptoms by regulating DNA damage, apoptosis, and other related signaling pathways through the delivery of proteins, microRNAs, long-chain noncoding RNAs, and circular RNAs. This technique is both safe and effective. MSC-derived exosomes may have great application prospects in the treatment of AKI. Understanding the underlying mechanisms will foster the development of new and promising therapeutic strategies against AKI. This review focused on recent advancements in the role of MSCs in AKI repair as well as the mechanisms underlying the role of MSCs and their secreted exosomes. It is anticipated that novel and profound insights into the functionality of MSCs and their derived exosomes will emerge.
Collapse
Affiliation(s)
- Juan-Juan Wang
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yu Zheng
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yan-Lin Li
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yin Xiao
- Department of Medical Imaging, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu Province, China
| | - Yang-Yang Ren
- Clinical Laboratory, Xinyi People's Hospital, Xuzhou 221000, Jiangsu Province, China
| | - Yi-Qing Tian
- Clinical Laboratory, Xuzhou Central Hospital, Xuzhou 221000, Jiangsu Province, China.
| |
Collapse
|
13
|
Zhang X, Guo Z, Li Y, Xu Y. Splicing to orchestrate cell fate. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102416. [PMID: 39811494 PMCID: PMC11729663 DOI: 10.1016/j.omtn.2024.102416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Alternative splicing (AS) plays a critical role in gene expression by generating protein diversity from single genes. This review provides an overview of the role of AS in regulating cell fate, focusing on its involvement in processes such as cell proliferation, differentiation, apoptosis, and tumorigenesis. We explore how AS influences the cell cycle, particularly its impact on key stages like G1, S, and G2/M. The review also examines AS in cell differentiation, highlighting its effects on mesenchymal stem cells and neurogenesis, and how it regulates differentiation into adipocytes, osteoblasts, and chondrocytes. Additionally, we discuss the role of AS in programmed cell death, including apoptosis and pyroptosis, and its contribution to cancer progression. Importantly, targeting aberrant splicing mechanisms presents promising therapeutic opportunities for restoring normal cellular function. By synthesizing recent findings, this review provides insights into how AS governs cellular fate and offers directions for future research into splicing regulatory networks.
Collapse
Affiliation(s)
- Xurui Zhang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, P.R. China
| | - Zhonghao Guo
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, P.R. China
| | - Yachen Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, P.R. China
| | - Yungang Xu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, P.R. China
| |
Collapse
|
14
|
Lee DR, Lee JE. Preservation of ovarian function using human pluripotent stem cell-derived mesenchymal progenitor cells. Clin Exp Reprod Med 2025; 52:30-37. [PMID: 38782038 PMCID: PMC11900672 DOI: 10.5653/cerm.2024.07003] [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: 02/24/2024] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 05/25/2024] Open
Abstract
Ovarian reserve diminishes with age, and older women experience a corresponding shift in sex hormone levels. These changes contribute to an age-dependent decrease in fertility and a decline in overall health. Furthermore, while survival rates following cancer treatment have improved for young female patients, a reduction in ovarian function due to the side effects of such treatments can be difficult to avoid. To date, no effective therapy has been recommended to preserve ovarian health in these patients. Mesenchymal progenitor cells (MPCs) are considered a promising option for cell therapy aimed at maintaining fertility and fecundity. Although MPCs derived from human adult tissues are recognized for their various protective effects against ovarian senescence, they are limited in quantity. Consequently, human pluripotent stem cell-derived MPCs (hPSC-MPCs), which exhibit high proliferative capacity and retain genetic stability during growth, have been utilized to delay reproductive aging. This review highlights the impact of hPSC-MPCs on preserving the functionality of damaged ovaries in female mouse models subjected to chemotherapy and natural aging. It also proposes their potential as a valuable cell source for fertility preservation in women with a variety of diseases.
Collapse
Affiliation(s)
- Dong Ryul Lee
- Department of Biomedical Science, College of Biological Science, CHA University, Seongnam, Republic of Korea
- Department of Biochemisty, CHA University School of Medicine, Seongnam, Republic of Korea
- CHA Advanced Research Institute, CHA Bundang Medical Center, Seongnam, Republic of Korea
| | - Jeoung Eun Lee
- CHA Advanced Research Institute, CHA Bundang Medical Center, Seongnam, Republic of Korea
| |
Collapse
|
15
|
Mizushina Y, Sun L, Nishio M, Nagata S, Kamakura T, Fukuda M, Tanaka K, Toguchida J, Jin Y. Hydroxycitric acid reconstructs damaged articular cartilages by modifying the metabolic cascade in chondrogenic cells. OSTEOARTHRITIS AND CARTILAGE OPEN 2025; 7:100564. [PMID: 39835169 PMCID: PMC11743121 DOI: 10.1016/j.ocarto.2024.100564] [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: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
Objective Osteoarthritis, a degenerative joint disease, requires innovative therapies due to the limited ability of cartilage to regenerate. Since mesenchymal stem cells (MSCs) provide a cell source for chondrogenic cells, we hypothesize that chemicals capable of enhancing the chondrogenic potential of MSCs with transforming growth factor-beta (TGFβ) in vitro may similarly promote chondrogenesis in articular cartilage in vivo. Design Chemical compounds that enhance the TGFβ signaling for chondrogenesis were investigated utilizing mesenchymal stem cells derived from human induced pluripotent stem cells. The mechanisms of action underlying the identified compound were explored in vitro, and its therapeutic effects were validated in vivo using a mouse model of exercise-induced osteoarthritis. Results Hydroxycitric acid (HCA) emerged as the lead compound. In vitro, HCA effectively enhanced chondrogenesis by inhibiting ATP citrate lyase, inducing citrate and alpha-ketoglutarate (α-KG), while reducing cytosolic acetyl coenzyme A (Ac-CoA). This induction of α-KG promoted collagen prolyl-4-hydroxylase activity, boosting hydroxyproline production and matrix formation. The reduction of Ac-CoA attenuated the inhibitory effect of β-catenin on mitochondrial activity by diminishing its acetylation. In vivo, orally administered HCA accumulated in joint tissues of mice and histological examination demonstrated newly synthesized cartilage tissues in damaged area. Analysis of joint tissue extracts revealed a downregulation of Ac-CoA and an upregulation of citrate and α-KG, accompanied by a systemic increase in an anabolic biomarker. Conclusions HCA demonstrates promise as an osteoarthritis therapy by enhancing chondrogenic differentiation. Its ability to modulate crucial metabolic pathways and facilitate cartilage repair suggests potential for clinical translation, addressing a critical need in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Yoshiyuki Mizushina
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd., 1-30-3 Toyokawa, Ibaraki, 567-0057, Japan
| | - Liping Sun
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Megumi Nishio
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Sanae Nagata
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Takeshi Kamakura
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masayuki Fukuda
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Kousuke Tanaka
- Central R & D Laboratory, Kobayashi Pharmaceutical Co., Ltd., 1-30-3 Toyokawa, Ibaraki, 567-0057, Japan
| | - Junya Toguchida
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Yonghui Jin
- Department of Regeneration Sciences and Engineering, Institute for Life and Medical Sciences, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
16
|
Saadh MJ, Jasim NY, Ahmed MH, Ballal S, Kumar A, Atteri S, Vashishth R, Rizaev J, Alhili A, Jawad MJ, Yazdi F, Salajegheh A, Akhavan-Sigari R. Critical roles of miR-21 in promotions angiogenesis: friend or foe? Clin Exp Med 2025; 25:66. [PMID: 39998742 PMCID: PMC11861128 DOI: 10.1007/s10238-025-01600-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
MiRNAs are small RNA strands that are managed following transcription and are of substantial importance in blood vessel formation. It is essential to oversee the growth, differentiation, death, movement and construction of tubes by angiogenesis-affiliated cells. If miRNAs are not correctly regulated in regard to angiogenesis, it can deteriorate the health and lead to various illnesses, which include cancer, cardiovascular disorder, critical limb ischemia, Crohn's disease, ocular diseases, diabetic microvascular complications, and more. Consequently, it is vital to understand the crucial part that miRNAs play in the development of blood vessels, so we can develop reliable treatment plans for vascular diseases. This write-up will assess the critical role of miR-21/exosomal miR-21 in managing angiogenesis associated with bone growth, wound recovery, and other pathological conditions like tumor growth, ocular illnesses, diabetes, and other diseases connected to formation of blood vessels. Previous investigations have demonstrated that miR-21 is present at higher amounts in certain cancerous cells, and it influences a multitude of genes that moderate the increased creation of blood vessels. Furthermore, studies demonstrated that exosomal miR-21 has the capacity to interact with endothelial cells to foster tumor angiogenesis. For that reason, this review explains the critical importance of miR-21/exosomal miR-21 in managing both healthy and diseased states of angiogenesis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Nisreen Yasir Jasim
- College of Nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Shikha Atteri
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali, Punjab, 140307, India
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | - Farzaneh Yazdi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | | | - Reza Akhavan-Sigari
- Dr. Schneiderhan GmbH and ISAR Klinikum, Munich, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw, Management University Warsaw, Warsaw, Poland
| |
Collapse
|
17
|
Mo J, Hu H, Li P, Ye Y, Chen W, Chen L, Qiao J, Zhao X, Yan Q, Chen C. Human hair follicle-derived mesenchymal stem cells improve ovarian function in cyclophosphamide-induced POF mice. Stem Cell Res Ther 2025; 16:67. [PMID: 39934888 PMCID: PMC11816600 DOI: 10.1186/s13287-024-04097-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/04/2024] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION Mesenchymal stem cell (MSCs) of different tissue origins have become a new option for the treatment of premature ovarian failure (POF) as they can recovery the ovarian function. However, there were rarely researches about human hair follicle-derived mesenchymal stem cells (HF-MSCs) in POF. OBJECTIVES In this study, we compared the effects of HF-MSCs and human umbilical cord mesenchymal stem cells (HU-MSCs) on POF models to explore the underlying molecular mechanisms. METHODS Female mice received intraperitoneal cyclophosphamid for 10 days to induce the POF mice model. One week after drug withdrawal, the mice were randomly divided into four groups according to the tail vein injection of drugs, which were: Control group (CON), Premature ovarian failure group (POF), HF-MSCs treatment group (P-H group) and HU-MSCs treatment group (P-U group). Which Treatment once a week for 4 consecutive times. Serum and ovarian tissues were collected 2 weeks after the last treatment, and fertility was performed by mating. ELISA, HE staining, transmission electron microscopy (TEM) were applied to evaluate the ovarian function, oocytes quantity and quality, and the mechanism was verified by qRT-PCR and western blot. In addition, the tumorigenic risk of organs was assessed by long-term observation. RESULTS The POF mice model was successfully established by intraperitoneal injection of cyclophosphamide 100 mg/kg/d for 10 days. Compared with POF group, two weeks after transplantation, serum FSH decreased, AMH and E2 increased in the P-H and P-U groups of mice (p < 0.05), but there was no significant difference between the P-H and P-U groups (p > 0.05). In addition, the number of primary follicles, secondary follicles and antral follicles in both P-H and P-U groups were significantly increased (p < 0.05), while the atretic follicles was significantly decreased (p < 0.05). The pups in POF group was significantly lower than that in P-H group and P-U group (p < 0. 01). Furthermore, those effects was more significant in P-H group than in P-U group (p < 0.05). In addition, the mitochondrial ultramicrostructure of the ovaries in the four groups showed a significant difference in the mitochondrial morphologies and number. In the POF group, the mitochondria presented a spheroids structure with fewer numbers, serious vacuolation and a disordered mitochondrial cristae arrangement. Nevertheless, after MSCs transplantation into the P-H and P-U group, we could observe ameliorative mitochondrial cristae alignment and vacuolation, as well as a small number of long rod-like structures. Mechanism study showed that KEAP1 protein expression was decreased in the P-H group, which increased the nuclear translocation of NRF2 and upregulated the expression of downstream HO-1 protein. At last, the possibility of tumor development after transplantation of HF-MSCs was excluded by long-term observation and organ anatomical examination. CONCLUSION HF-MSCs can improve ovarian function in cyclophosphamide-induced POF mice, and the effects were superior to HU-MSCs. The underlying mechanism may by inhibiting ferroptosis of granulosa cells through KEAP1/NRF2/HO-1 pathway.
Collapse
Affiliation(s)
- Jinhua Mo
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Hong Hu
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Pengdong Li
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Yang Ye
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Wanle Chen
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Lei Chen
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Jing Qiao
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Xiaoying Zhao
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China
| | - Qiuxia Yan
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China.
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China.
| | - Cairong Chen
- Center for Reproductive Medicine, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China.
- Guangdong Engineering Technology Research Center of Urinary Continence and Reproductive Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, No. 35, Yinquan North Road, Qingcheng District, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|
18
|
Zuo R, Li H, Cai C, Xia W, Liu J, Li J, Xu Y, Zhang Y, Li C, Wu Y, Zhang C. Autophagy modulates tenogenic differentiation of cartilage-derived stem cells in response to mechanical tension via FGF signaling. Stem Cells Transl Med 2025; 14:szae085. [PMID: 39673221 PMCID: PMC11878763 DOI: 10.1093/stcltm/szae085] [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: 07/20/2024] [Accepted: 10/01/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND In our previous study, we demonstrated that cartilage-derived stem cells (CDSCs) possess multi-differentiation potential, enabling direct bone-to-tendon structure regeneration after transplantation in a rat model. Therefore, the objective of this study is to investigate whether CDSCs are a suitable candidate for achieving biological regeneration of tendon injuries. METHODS Tenogenic differentiation was evaluated through cell morphology observation, PCR, and Western blot (WB) analysis. Autophagic flux, transmission electron microscopy, and WB analysis were employed to elucidate the role of autophagy during CDSC tenogenic differentiation. Cell survival and tenogenesis of transplanted CDSCs were assessed using fluorescence detection of gross and frozen section images. Heterotopic ossification and quality of tendon healing were evaluated by immunofluorescence, hematoxylin-eosin (H&E), and Safrinin O/Fast Green stains. RESULTS We found autophagy is activated in CDSCs when treated with cyclic tensile stress, which facilitates the preservation of their chondrogenic potential while impeding tenogenic differentiation. Inhibiting autophagy with chloroquine promoted tenogenic differentiation of CDSCs in response to cyclic tensile stress through activation of the Fgf2/Fgfr2 signaling pathway. This mechanism was further validated by 2 mouse transplantation models, revealed that autophagy inhibition could enhance the tendon regeneration efficacy of transplanted CDSCs at the patellar tendon resection site. CONCLUSION Our findings provide insights into CDSC transplantation for achieving biological regeneration of tendon injuries, and demonstrate how modulation of autophagy in CDSCs can promote tenogenic differentiation in response to tensile stress both in vivo and in vitro.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Haoke Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Chenhui Cai
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Wen Xia
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Jiabin Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Yuan Xu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Yi Zhang
- Chongqing International Institute for Immunology, Chongqing 401320, People’s Republic of China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| | - Yuzhang Wu
- Institute of Immunology, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, People’s Republic of China
| |
Collapse
|
19
|
Jing Y, Li B, Aierken A, Zhang Z, Han D, Lin Z, Gao J, Tian H, Hua J. Mesenchymal Stem Cells with Simultaneous Overexpression of GPX3 and CD47 for the Treatment of Drug-Induced Acute Liver Injury. Vet Sci 2025; 12:149. [PMID: 40005909 PMCID: PMC11861084 DOI: 10.3390/vetsci12020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The liver, as the largest metabolic and detoxification organ in mammals, metabolizes approximately 80-90% of drugs. However, drug-induced liver injury (DILI) is common and driven by factors such as individual variability, differences in liver metabolism, and improper drug use. Mesenchymal stem cells (MSCs), with their self-renewal and multipotent differentiation capabilities, offer therapeutic potential, but face challenges such as limited proliferation and increased apoptosis during in vitro expansion. Although MSCs exhibit low immunogenicity, they are often cleared by the host immune system, which limits their survival and engraftment. Glutathione peroxidase 3 (GPX3) is a key antioxidant enzyme that reduces reactive oxygen species (ROS), protecting cells from oxidative damage. CD47, also known as integrin-associated protein (IAP), helps cells evade immune clearance by binding to signal regulatory protein alpha (SIRPα) on the immune cells. Here, we used an acetaminophen (APAP)-induced DILI mouse model to evaluate the therapeutic efficacy of intravenously infused MSCs overexpressing GPX3 and CD47. Compared to unmodified MSCs, modified MSCs showed improved survival, reduced liver inflammation, and alleviated oxidative damage, offering enhanced protection against APAP-induced DILI.
Collapse
Affiliation(s)
- Yuanxiang Jing
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Balun Li
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Aili Aierken
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
- Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi 830000, China
| | - Zengyu Zhang
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Dongyao Han
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Zixi Lin
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Jiaqi Gao
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Hongkai Tian
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Jinlian Hua
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| |
Collapse
|
20
|
Fang JR, Chen CL, Chen YQ, Luo SK. Inhibition of Small Extracellular Vesicles by GW4869 Does not Disrupt the Paracrine Regulation of Adipose-Derived Mesenchymal Stem Cells Over Keloid Fibroblasts. Aesthetic Plast Surg 2025; 49:917-928. [PMID: 39496963 DOI: 10.1007/s00266-024-04477-1] [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: 06/30/2024] [Accepted: 10/14/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Keloid, scar caused by atypical wound repair, represents a significant difficulty for specialists in plastic surgery and dermatology. Adipose-derived mesenchymal stem cells (ADSCs) can regulate fibrotic phenotypes of keloid fibroblasts (KFs) in a paracrine fashion, but whether small extracellular vesicles (SEVs) are the key functional carrier in ADSC paracrine regulation of KFs remains unknown. This study aims to explore whether the regulatory effects of conditioned medium (CM) obtained from ADSCs on KFs can be impaired by decreased SEV content in the ADSC-CM. METHODS Clinical specimens were utilized to extract keloid fibroblasts (KFs), normal fibroblasts (NFs), and adipose-derived stem cells (ADSCs). Fibroblasts were cultured with CM obtained from ADSCs untreated or treated with the sphingomyelinase inhibitor GW4869. The features of SEVs derived from ADSC-CM were characterized, and fibroblast proliferation, migration, apoptosis, and expression of ECM proteins were analyzed. RESULTS The sphingomyelinase inhibitor GW4869 successfully reduced the SEV content in ADSC-CM, and both control ADSC-CM and ADSC-CM with reduced SEV content significantly inhibited KF proliferation, migration, and α-SMA synthesis but not KF apoptosis, whereas only NF proliferation was inhibited by ADSC-CM. The reduced SEV content only affected the inhibition of KF proliferation induced by ADSC-CM. CONCLUSION ADSC-CM inhibits various fibrotic phenotypes of KFs, but decreasing the SEV content in ADSC-CM did not significantly alter the antifibrotic effects of ADSC-CM. Thus, SEVs may not be the key mediator of ADSCs paracrine regulation of KFs. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors . www.springer.com/00266 .
Collapse
Affiliation(s)
- Jun-Ren Fang
- Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, China
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 466 Middle Xin Gang Road, Guangzhou City, 510317, Guangdong Province, China
| | - Chun-Lin Chen
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 466 Middle Xin Gang Road, Guangzhou City, 510317, Guangdong Province, China
| | - Yi-Qing Chen
- Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, China
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 466 Middle Xin Gang Road, Guangzhou City, 510317, Guangdong Province, China
| | - Sheng-Kang Luo
- Second School of Clinical Medicine, Southern Medical University, Guangzhou City, Guangdong Province, China.
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, 466 Middle Xin Gang Road, Guangzhou City, 510317, Guangdong Province, China.
| |
Collapse
|
21
|
Song J, Yang M, Xia L, Wang L, Wang K, Xiang Y, Cheng J, Chen J, Liu J, Zhao R, Liu F, Sun Z, Hou X, Zang N, Chen L. Aptamer-Conjugated Exosomes Ameliorate Diabetes-Induced Muscle Atrophy by Enhancing SIRT1/FoxO1/3a-Mediated Mitochondrial Function. J Cachexia Sarcopenia Muscle 2025; 16:e13717. [PMID: 39871746 PMCID: PMC11773161 DOI: 10.1002/jcsm.13717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 11/26/2024] [Accepted: 12/19/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Muscle atrophy is associated with Type 2 diabetes mellitus, which reduces the quality of life and lacks effective treatment strategies. Previously, it was determined that human umbilical cord mesenchymal stromal cell (hucMSC)-derived exosomes (EXOs) ameliorate diabetes-induced muscle atrophy. However, the systemic application of EXOs is less selective for diseased tissues, which reduces their efficacy and safety associated with their nonspecific biological distribution in vivo. Therefore, improving exosomal targeting is imperative. In this study, a skeletal muscle-specific aptamer (Apt) was used to explore the effects of Apt-functionalized EXOs derived from hucMSCs in diabetes-associated muscle atrophy and its specific mechanisms. METHODS Diabetic db/db mice and C2C12 myotubes were used to explore the effects of MSC-EXOs or Apt-EXOs in alleviating muscle atrophy. Grip strength, muscle weight and muscle fibre cross-sectional area (CSA) were used to evaluate skeletal muscle strength and muscle mass. Western blot analysis of muscle atrophy signalling, including MuRF1 and Atrogin 1 and the mitochondrial complex and Seahorse analysis were performed to investigate the underlying mechanisms of MSC-EXOs or Apt-EXOs on muscle atrophy. RESULTS MSC-EXOs increased grip strength (p = 0.0002) and muscle mass (p = 0.0044 for tibialis anterior (TA) muscle, p = 0.002 for soleus (SO) muscle) in db/db mice. It also increased the CSA of muscle fibres (p = 0.0011 for all fibres, p = 0.0036 for slow muscle fibres and p = 0.0089 for fast muscle fibres) and the percentage of slow-to-fast muscle fibres (p = 0.0109). However, Atrogin 1 (p = 0.0455) and MuRF1 expression (p = 0.0168) was reduced. MSC-EXOs activated SIRT1/FoxO1/3a signalling and enhanced mitochondrial function in db/db mice and C2C12 myotubes. SIRT1 knockdown decreased the beneficial antiatrophic effects of MSC-EXOs. Additionally, Apt conjugation increased the effect of MSC-EXOs on muscle atrophy and myofiber-type transition (p = 0.0133 for grip strength, p = 0.0124 for TA muscle weight, p = 0.0008 for SO muscle weight, p < 0.0001 for CSA of all muscle fibres, p = 0.0198 for CSA of slow muscle fibres, p = 0.0213 for CSA of fast muscle fibres, p = 0.011 for percentage of slow-fast muscle fibres, p = 0.0141 for Atrogin 1 expression and p = 0.005 for MuRF1 expression). CONCLUSIONS The results suggest that hucMSC-derived exosomes ameliorate diabetes-associated muscle atrophy by enhancing SIRT1/FoxO1/3a-mediated mitochondrial function and that Apt conjugation strengthens the effects of MSC-EXOs on muscle atrophy. These findings demonstrate the therapeutic potential of muscle-targeted MSC-EXOs for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Jia Song
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Mengmeng Yang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Longqing Xia
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Liming Wang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Kewei Wang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Yingyue Xiang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Jun Cheng
- Department of Clinical Laboratory, Shandong Engineering & Technology Research Center for Tumor Marker DetectionThe Second Hospital of Shandong UniversityJinanShandongChina
| | - Jun Chen
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Jidong Liu
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Ruxing Zhao
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Fuqiang Liu
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Zheng Sun
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
| | - Xinguo Hou
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases, Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Institute of Endocrine and Metabolic Diseases of Shandong UniversityJinanShandongChina
| | - Nan Zang
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases, Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Institute of Endocrine and Metabolic Diseases of Shandong UniversityJinanShandongChina
| | - Li Chen
- Department of Endocrinology and MetabolismQilu Hospital of Shandong UniversityJinanShandongChina
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases, Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Institute of Endocrine and Metabolic Diseases of Shandong UniversityJinanShandongChina
| |
Collapse
|
22
|
Narasimha RB, Shreya S, Jayabal VA, Yadav V, Rath PK, Mishra BP, Kancharla S, Kolli P, Mandadapu G, Kumar S, Mohanty AK, Jena MK. Stem Cell Therapy for Diseases of Livestock Animals: An In-Depth Review. Vet Sci 2025; 12:67. [PMID: 39852942 PMCID: PMC11768649 DOI: 10.3390/vetsci12010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Stem cells are unique, undifferentiated cells that have the ability to both replicate themselves and develop into specialized cell types. This dual capability makes them valuable in the development of regenerative medicine. Current development in stem cell research has widened their application in cell therapy, drug discovery, reproductive cloning in animals, and cell models for various diseases. Although there are substantial studies revealing the treatment of human degenerative diseases using stem cells, this is yet to be explored in livestock animals. Many diseases in livestock species such as mastitis, laminitis, neuromuscular disorders, autoimmune diseases, and some debilitating diseases are not covered completely by the existing drugs and treatment can be improved by using different types of stem cells like embryonic stem cells, adult stem cells, and induced pluripotent stem cells. This review mainly focuses on the use of stem cells for disease treatment in livestock animals. In addition to the diseases mentioned, the potential of stem cells can be helpful in wound healing, skin disease therapy, and treatment of some genetic disorders. This article explores the potential of stem cells from various sources in the therapy of livestock diseases and also their role in the conservation of endangered species as well as disease model preparation. Moreover, the future perspectives and challenges associated with the application of stem cells in livestock are discussed. Overall, the transformative impact of stem cell research on the livestock sector is comprehensively studied which will help researchers to design future research work on stem cells related to livestock diseases.
Collapse
Affiliation(s)
- Raghavendra B. Narasimha
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| | - Singireddy Shreya
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| | - Vijay Anand Jayabal
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai 600051, Tamil Nadu, India;
| | - Vikas Yadav
- Department of Clinical Sciences, Clinical Research Centre, Skåne University Hospital, Lund University, SE 20213 Malmö, Sweden
| | - Prasana Kumar Rath
- College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar 751003, Odisha, India; (P.K.R.); (B.P.M.)
| | - Bidyut Prava Mishra
- College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar 751003, Odisha, India; (P.K.R.); (B.P.M.)
| | - Sudhakar Kancharla
- Devansh Lab Werks, 234 Aquarius Drive, Homewood, AL 35209, USA; (S.K.); (G.M.)
| | - Prachetha Kolli
- Microgen Health Inc., 14225 Sullyfield Cir Suite E, Chantilly, VA 20151, USA;
| | - Gowtham Mandadapu
- Devansh Lab Werks, 234 Aquarius Drive, Homewood, AL 35209, USA; (S.K.); (G.M.)
| | - Sudarshan Kumar
- Cell, Molecular and Proteomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (ICAR-NDRI), Karnal 132001, Haryana, India;
| | - Ashok Kumar Mohanty
- ICAR-Central Institute for Research on Cattle (ICAR-CIRC), Meerut 250001, Uttar Pradesh, India;
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| |
Collapse
|
23
|
Wu SK, Tsai CL, Mir A, Hynynen K. MRI-guided focused ultrasound for treating Parkinson's disease with human mesenchymal stem cells. Sci Rep 2025; 15:2029. [PMID: 39815002 PMCID: PMC11735764 DOI: 10.1038/s41598-025-85811-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
Parkinson's disease (PD) is a progressive disorder that affects the nervous system and causes regions of the brain to deteriorate. In this study, we investigated the effects of MR-guided focused ultrasound (MRgFUS) for the delivery of human mesenchymal stem cells (MSCs) on the 6-hydroxydopamine (6-HODA)-induced PD rat model. MRgFUS-induced blood-brain barrier (BBB) permeability modulation was conducted using an acoustic controller with the targets at the striatum (ST) and SN. Human MSCs were injected immediately before sonication. Here, we show that we can deliver human MSCs into Parkinsonian rats through MRgFUS-induced BBB modulation using an acoustic controller. Stem cells were identified in the sonicated brain regions using surface markers, indicating the feasibility of MSC delivery via MRgFUS. MSCs + FUS treatment significantly improved the behavioural outcomes compared with control, FUS alone, and MSCs alone groups (p < 0.05). In the quantification analysis of the TH stain, a significant reservation of dopamine neurons was seen in the MSCs + FUS group as compared with the MSCs group (ST: p = 0.03; SN: p = 0.0005). Mesenchymal stem cell therapy may be a viable treatment option for neurodegenerative diseases such as Parkinson's. Transcranial MRgFUS serves as an efficacious and safe method for targeted and minimally invasive stem cell homing.
Collapse
Affiliation(s)
- Sheng-Kai Wu
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| | - Chia-Lin Tsai
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Aisha Mir
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
24
|
Kim JE, Lee JW, Cha GD, Yoon JK. The Potential of Mesenchymal Stem Cell-Derived Exosomes to Treat Diabetes Mellitus. Biomimetics (Basel) 2025; 10:49. [PMID: 39851765 PMCID: PMC11760843 DOI: 10.3390/biomimetics10010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/27/2024] [Accepted: 12/31/2024] [Indexed: 01/26/2025] Open
Abstract
Diabetes mellitus (DM) is a fatal metabolic disease characterized by persistent hyperglycemia. In recent studies, mesenchymal stem cell (MSC)-derived exosomes, which are being investigated clinically as a cell-free therapy for various diseases, have gained attention due to their biomimetic properties that closely resemble natural cellular communication systems. These MSC-derived exosomes inherit the regenerative and protective effects from MSCs, inducing pancreatic β-cell proliferation and inhibiting apoptosis, as well as ameliorating insulin resistance by suppressing the release of various inflammatory cytokines. Consequently, MSC-derived exosomes have attracted attention as a novel treatment for DM as an alternative to stem cell therapy. In this review, we will introduce the potential of MSC-derived exosomes for the treatment of DM by discussing the studies that have used MSC-derived exosomes to treat DM, which have shown therapeutic effects in both type 1 and type 2 DM.
Collapse
Affiliation(s)
| | | | | | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea (G.D.C.)
| |
Collapse
|
25
|
Chen Q, Su Y, Yang Z, Lin Q, Ke Y, Xing D, Li H. Bibliometric mapping of mesenchymal stem cell therapy for bone regeneration from 2013 to 2023. Front Med (Lausanne) 2025; 11:1484097. [PMID: 39835103 PMCID: PMC11743382 DOI: 10.3389/fmed.2024.1484097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have shown significant potential in bone regeneration and regenerative medicine in recent years. With the advancement of tissue engineering, MSCs have been increasingly applied in bone repair and regeneration, and their clinical application potential has grown through interdisciplinary approaches involving biomaterials and genetic engineering. However, there is a lack of systematic reviews summarizing their applications in bone regeneration. To address this gap, we analyzed the latest research on MSCs for bone regeneration published from 2013 to 2023. Using the Web of Science Core Collection, we conducted a literature search in December 2024 and employed bibliometric tools like CiteSpace and VOSviewer for a comprehensive analysis of the key research trends. Our findings focus on the development of cell engineering, highlighting the advantages, limitations, and future prospects of MSC applications in bone regeneration. These insights aim to enhance understanding of MSC-based bone regeneration, inspire new research directions, and facilitate the clinical translation of MSC research.
Collapse
Affiliation(s)
- Qianqian Chen
- Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Arthritis Clinic & Research Center, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yiqi Su
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
| | - Zhen Yang
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
| | - Qiyuan Lin
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
| | - Yan Ke
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Dan Xing
- Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Arthritis Clinic & Research Center, Zhejiang Chinese Medical University, Hangzhou, China
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| | - Hui Li
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing, China
- Arthritis Institute, Peking University, Beijing, China
| |
Collapse
|
26
|
Sotomayor-Lugo F, Iglesias-Barrameda N, Casado-Hernandez I, Villegas-Valverde CA, Ventura-Carmenate Y, Rivero-Jimenez RA. Aging: Disease or "natural" process? A glimpse from regenerative medicine. Rev Esp Geriatr Gerontol 2025; 60:101543. [PMID: 39369641 DOI: 10.1016/j.regg.2024.101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 10/08/2024]
Abstract
We explore aging as a global phenomenon, questioning whether it constitutes a treatable condition or follows a natural course. Acknowledging its multifactorial nature, we delve into the challenges and opportunities inherent in this intricate biological process. The inclusion of old age in the 11th International Classification of Diseases sparks debate, categorizing it as a disease based on mechanistic explanations, blood-based biomarkers, and anti-aging products. Ethical dilemmas arise, emphasizing the difficulty of defining the transition from normal to pathological states during this process. We suggest that aging should be regarded as a treatable condition without necessarily labeling it a 'disease.' While anti-aging research unveils promising interventions like Metformin, Rapamycin, and cellular therapy, achieving biological immortality remains a formidable challenge. The future promises to prolong life and enhance quality by comprehensively understanding aging's implications for human health.
Collapse
Affiliation(s)
| | | | | | | | - Yendry Ventura-Carmenate
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates; Yas Clinic Khalifa City Hospital, Abu Dhabi, United Arab Emirates; United Arab Emirates University, Office of Research and Graduate Studies, College of Medicine and Health Science, Abu Dhabi, United Arab Emirates
| | - Rene Antonio Rivero-Jimenez
- Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates; United Arab Emirates University, Office of Research and Graduate Studies, College of Medicine and Health Science, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
27
|
Mitani K, Ito Y, Takene Y, Inaba T. Evaluation of the quality of life-enhancing effect of allogeneic feline adipose mesenchymal stem cells in cats with osteoarthritis: A pilot study. Res Vet Sci 2025; 182:105470. [PMID: 39612738 DOI: 10.1016/j.rvsc.2024.105470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
Osteoarthritis (OA) is a progressive degenerative disease in older cats, and often leads to decreased quality of life (QOL). Mesenchymal stem cells (MSCs) have been used in novel therapies for inflammatory diseases. We aimed to evaluate quantitatively allogeneic adipose-derived MSC (ADSC) therapy in cats with naturally occurring OA, based on QOL assessment resources. To characterize the in vitro properties of ADSCs, we estimated ADSCs from four healthy cats with respect to morphology, differentiation potential, and immunomodulatory potential. Six cats with OA were administered a single intravenous injection of allogeneic ADSCs. Based on the feline musculoskeletal pain index (FMPI), the outcome measure was QOL. The cultured cells were adherent, exhibited a spindle shape without becoming flattened or large, and maintained doubling time until passage 5. After induction, the cells had osteogenic, adipogenic, and chondrogenic phenotypes. These cells expressed CD44 and CD90 and lacked expression of CD14 and CD45, had significantly suppressed the production of interferon -ɤ released from mitogen-stimulated lymphocytes (P < 0.05). The FMPI of all cats with OA significantly increased one month after ADSC therapy (P < 0.05). No adverse effects associated with ADSC administration were observed during follow-up in any of the cats. In conclusion, ADSC therapy with immunomodulatory potential could have beneficial effects on the QOL in cats with OA. Further research is necessary to carry out larger studies of the effectiveness of ADSC therapy.
Collapse
Affiliation(s)
- Kosuke Mitani
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Yuki Ito
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Yukio Takene
- Research and Development Department, J-ARM Co., Ltd., Osaka, Japan
| | - Toshio Inaba
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan.
| |
Collapse
|
28
|
Zheng B, Wang X, Guo M, Tzeng CM. Current Development of Mesenchymal Stem Cell-Derived Extracellular Vesicles. Cell Transplant 2025; 34:9636897241297623. [PMID: 39874070 PMCID: PMC11775985 DOI: 10.1177/09636897241297623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 01/30/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells with self-renewal. They play a critical role in cell therapy due to their powerful immunomodulatory and regenerative effects. Recent studies suggest that one of the key therapeutic mechanisms of MSCs seems to derive from their paracrine product, called extracellular vesicles (EVs). The EVs contain much DNA, messenger RNA (mRNA), microRNA, and protein components, which can exert intracellular communication to target cells. In clinical applications, the MSC-EVs have been widely used in tissue repair and immune disorder diseases. However, there are serval issues need to be considered such as how to accomplish the large-scale production of EVs and how to verify the exact mechanism of EVs. In this review, we summarize the current progress of MSC-EVs and discuss the challenges and future of MSC-EVs.
Collapse
Affiliation(s)
- Bingyi Zheng
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Xueting Wang
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Meizhai Guo
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Chi-Meng Tzeng
- Cells Good (Xiamen) Inc. Huli, Xiamen Torch Development Zone, Fujian, China
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
29
|
Roseti L, Cavallo C, Desando G, D’Alessandro M, Grigolo B. Forty Years of the Use of Cells for Cartilage Regeneration: The Research Side. Pharmaceutics 2024; 16:1622. [PMID: 39771600 PMCID: PMC11677864 DOI: 10.3390/pharmaceutics16121622] [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: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The treatment of articular cartilage damage has always represented a problem of considerable practical interest for orthopedics. Over the years, many surgical techniques have been proposed to induce the growth of repairing tissue and limit degeneration. In 1994, the turning point occurred: implanted autologous cells paved the way for a new treatment option based more on regeneration than repair. Objectives: This review aims to outline biological and clinical advances, from the use of mature adult chondrocytes to cell-derived products, going through progenitor cells derived from bone marrow or adipose tissue and their concentrates for articular cartilage repair. Moreover, it highlights the relevance of gene therapy as a valuable tool for successfully implementing current regenerative treatments, and overcoming the limitations of the local delivery of growth factors. Conclusions: Finally, this review concludes with an outlook on the importance of understanding the role and mechanisms of action of the different cell compounds with a view to implementing personalized treatments.
Collapse
Affiliation(s)
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.); (G.D.); (M.D.); (B.G.)
| | | | | | | |
Collapse
|
30
|
Rui Q, Li C, Rui Y, Zhang C, Xia C, Wang Q, Liu Y, Wang P. Human umbilical mesenchymal stem cells ameliorate atrophic gastritis in aging mice by participating in mitochondrial autophagy through Ndufs8 signaling. Stem Cell Res Ther 2024; 15:491. [PMID: 39707499 DOI: 10.1186/s13287-024-04094-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Chronic atrophic gastritis (CAG) is a chronic disease of the gastric mucosa characterized by a reduction or an absolute disappearance of the original gastric glands, possibly replaced by pseudopyloric fibrosis, intestinal metaplasia, or fibrosis. CAG develops progressively into intestinal epithelial metaplasia, dysplasia, and ultimately, gastric cancer. Epidemiological statistics have revealed a positive correlation between the incidence of CAG and age. Mesenchymal stem cells (MSCs) are a type of adult stem cells derived from mesoderm, with strong tissue repair capabilities. Therefore, the restoration of the gastric mucosa may serve as an efficacious strategy to ameliorate CAG and avert gastric cancer. However, the mechanisms by which MSCs inhibit the relentless progression of aging atrophic gastritis remain to be elucidated. This study endeavored to assess a novel approach utilizing MSCs to treat CAG and forestall carcinogenics. METHODS In this study, we selected mice with atrophic gastritis from naturally aging mice and administered human umbilical cord-derived mesenchymal stem cells (hUMSCs) via tail vein injection to evaluate the therapeutic effects of hUMSCs on age-related chronic atrophic gastritis. Initially, we employed methods such as ELISA, immunohistochemical analysis, and TUNEL assays to detect changes in the mice post-hUMSC injection. Proteomic and bioinformatics analyses were conducted to identify differentially expressed proteins, focusing on NADH: ubiquinone oxidoreductase core subunit S8 (Ndufs8). Co-culturing hUMSCs with Ndufs8 knockout gastric mucosal epithelial cells (GMECs), we utilized flow cytometry, Western blotting, real-time quantitative PCR, and immunofluorescence to investigate the mechanisms of action of hUMSCs. RESULTS We observed that hUMSCs are capable of migrating to and repairing damaged gastric mucosa. Initially, hUMSCs significantly enhanced the secretion of gastric proteins PG-1 and G17, while concurrently reducing inflammatory cytokines. Furthermore, hUMSCs mitigated gastric fibrosis and apoptosis in mucosal cells. Proteomic and bioinformatic analyses revealed alterations in the protein network involved in mitochondrial autophagy, with Ndufs8 playing a pivotal role. Upon knocking out Ndufs8 in GMECs, we noted mitochondrial damage and reduced autophagy, leading to an aged phenotype in GMECs. Co-culturing Ndufs8-knockout GMECs with hUMSCs demonstrated that hUMSCs could ameliorate mitochondrial dysfunction and restore the cell cycle in GMECs.
Collapse
Affiliation(s)
- Qiang Rui
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Chuyu Li
- State Key Laboratory of Reproductive Medicine, Center of Clinical Reproductive Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Yiqi Rui
- Department of General Surgery,Geriatric Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Chuanzhuo Zhang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Cunbing Xia
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Qing Wang
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yuanyuan Liu
- School of Medicine, Southeast University, Nanjing, 210096, China
| | - Peng Wang
- Department of General Surgery,Geriatric Hospital of Nanjing Medical University, Nanjing, 210000, China.
| |
Collapse
|
31
|
Wang Y, Luo P, Wuren T. Narrative Review of Mesenchymal Stem Cell Therapy in Renal Diseases: Mechanisms, Clinical Applications, and Future Directions. Stem Cells Int 2024; 2024:8658246. [PMID: 39698513 PMCID: PMC11655143 DOI: 10.1155/sci/8658246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Renal diseases, particularly acute kidney injury (AKI) and chronic kidney disease (CKD), are significant global health challenges. These conditions impair kidney function and can lead to serious complications, including cardiovascular diseases, which further exacerbate the public health burden. Currently, the global AKI mortality rate is alarmingly high (20%-50%); CKD is projected to emerge as a major global health burden by 2040. Existing treatments such as hemodialysis and kidney transplantation have limited effectiveness and are often associated with adverse effects. Mesenchymal stem cells (MSCs) offer considerable potential for treating renal diseases owing to their regenerative and immunomodulatory properties. Thus, this review focuses on the application of MSCs in renal disease, discusses fundamental research findings, and evaluates their application in clinical trials. Moreover, we discuss the impact and safety of MSCs as a therapeutic option and highlight challenges and potential directions for their clinical application. We selected research articles from PubMed published within the last 5 years (from 2019), focusing on high-impact journals and clinical trial data, and included a few key studies predating 2019. Considerations included the novelty of the research, sample size, experimental design, and data reliability. With advancements in single-cell sequencing, CRISPR/Cas9 gene editing, and other cutting-edge technologies, future MSC research will explore combination therapies and personalized treatments to provide more promising, safer treatments with reduced adverse reactions and enhanced therapeutic outcomes. These advances will improve kidney disease treatment methods, enhance patient quality of life, and maximize the benefits of MSC therapies.
Collapse
Affiliation(s)
- Yanjun Wang
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining 810001, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine (Qinghai-Utah Joint Key Laboratory for Plateau Medicine), Xining 810001, China
- Nephrology Department, Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Pengli Luo
- Nephrology Department, Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining 810001, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine (Qinghai-Utah Joint Key Laboratory for Plateau Medicine), Xining 810001, China
| |
Collapse
|
32
|
Chen Z, Pang Q, Zhan J, Liu J, Zhao W, Dong L, Huang W. MSCs-derived ECM functionalized hydrogel regulates macrophage reprogramming for osteoarthritis treatment by improving mitochondrial function and energy metabolism. Mater Today Bio 2024; 29:101340. [PMID: 39640869 PMCID: PMC11617891 DOI: 10.1016/j.mtbio.2024.101340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/20/2024] [Accepted: 11/10/2024] [Indexed: 12/07/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease that affects the entire joint, with synovial inflammation being a major pathological feature. Macrophages, as the most abundant immune cells in the synovium, have an M1/M2 imbalance that is closely related to the occurrence and development of OA. Mesenchymal stem cells (MSCs) have been shown to effectively suppress inflammation in the treatment of OA, but they still pose issues such as immune rejection and tumorigenicity. The extracellular matrix (ECM), as a major mediator of MSCs' immunoregulatory effects, offers a cell-free therapy to circumvent these risks. In this study, we developed an ECM-functionalized hydrogel by combining MSC-derived ECM with gelatin methacryloyl (GelMA). To enhance the immunomodulatory potential of MSCs, we pre-stimulated MSCs with the inflammatory factor interleukin-6 (IL-6) present in OA. In vitro results showed that the ECM-functionalized hydrogel promoted M2 macrophage polarization and inhibited the expression of various inflammatory genes, strongly indicating the hydrogel's powerful immunoregulatory capabilities. In an in vivo rat OA model, the ECM-functionalized hydrogel significantly reduced synovial inflammation and cartilage matrix degradation, alleviating the progression of OA. Furthermore, we utilized proteomics and transcriptomics analysis to reveal that the hydrogel accomplished macrophage metabolic reprogramming by regulating mitochondrial function and energy metabolism, thereby reducing inflammation. These findings suggest that the ECM-functionalized hydrogel is a promising biomaterial-based strategy for treating OA by targeting key pathological mechanisms.
Collapse
Affiliation(s)
- Zhuolin Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Qiming Pang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Jingdi Zhan
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Junyan Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Weikang Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Lili Dong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Orthopaedic Research Laboratory of Chongqing Medical University, Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Silva Couto P, Stibbs DJ, Sanchez BC, Khalife R, Panagopoulou TI, Barnes B, George V, Taghizadeh RR, Rafiq QA. Generating suspension-adapted human mesenchymal stromal cells (S-hMSCs) for the scalable manufacture of extracellular vesicles. Cytotherapy 2024; 26:1532-1546. [PMID: 39269403 DOI: 10.1016/j.jcyt.2024.06.011] [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: 03/29/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUD Human mesenchymal stromal cells (hMSCs) are a naturally adherent cell type and one of the most studied cellular agents used in cell therapy over the last 20 years. Their mechanism of action has been primarily associated with paracrine signaling, which has contributed to an increase in the number of studies focused on hMSC-related extracellular vesicles (EVs). METHODS In this study, we demonstrate for the first time that human telomerase reverse transcriptase (hTERT) immortalized hMSCs can be adapted to suspension culture, eliminating the need for microcarriers or other matrixes to support cell growth. RESULTS This novel cell line, named suspension hMSCs (S-hMSCs), has a doubling time of approximately 55 hours, with a growth rate of 0.423/d. Regarding its immunophenotype characteristics, S-hMSCs retained close to 90% of CD73 and CD105 expression levels, with the CD90 receptor being downregulated during the adherent to suspension adaptation process. An RNA sequencing analysis showed an upregulation of the transcripts coding for CD44, CD46 and CD47 compared to the expression levels in AT-hMSCs and hTERT-hMSCs. The cell line herein established was able to generate EVs using a chemically defined medium formulation with these nanoparticles averaging 150 nm in size and displaying the markers CD63, CD81, and TSG101, while not expressing the negative marker calnexin. CONCLUSION This body of evidence, combined with the visual confirmation of EV presence using transmission electron microscopy, demonstrates the EV-producing capabilities of the novel S-hMSCs. This cell line provides a platform for process development, drug discovery and translational studies in the EV field.
Collapse
Affiliation(s)
- Pedro Silva Couto
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Dale J Stibbs
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Braulio Carrillo Sanchez
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Rana Khalife
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Theano I Panagopoulou
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Benjamin Barnes
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | - Vaques George
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK
| | | | - Qasim A Rafiq
- Department of Biochemical Engineering, Advanced Centre for Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
34
|
Chen L, Zhang Y, Wang K, Jin M, Chen Q, Wang S, Hu W, Cai Z, Li Y, Li S, Gao Y, Zhou S, Peng Q. A patch comprising human umbilical cord-derived hydrogel and mesenchymal stem cells promotes pressure ulcer wound healing. ENGINEERED REGENERATION 2024; 5:433-442. [DOI: 10.1016/j.engreg.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
|
35
|
Ji X, Wang L, Zhong Y, Xu Q, Yan J, Pan D, Xu Y, Chen C, Wang J, Wang G, Yang M, Li T, Tang L, Wang X. Impact of mesenchymal stem cell size and adhesion modulation on in vivo distribution: insights from quantitative PET imaging. Stem Cell Res Ther 2024; 15:456. [PMID: 39609885 PMCID: PMC11606219 DOI: 10.1186/s13287-024-04078-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Successful engraftment and localization of mesenchymal stem cells (MSCs) within target tissues are critical factors influencing their therapeutic efficacy for tissue repair and regeneration. However, the relative contributions of biophysical factors like cell size and adhesion capacity in regulating MSC distribution in vivo remain incompletely understood. METHODS Cell adhesion peptides and hanging drop method were used to modify the adhesive capacity and size of MSCs. To quantitatively track the real-time biodistribution of transplanted MSCs with defined size and adhesion profiles in living mice and rats, the non-invasive positron emission tomography (PET) imaging was applied. RESULTS Surface modification with integrin binding peptides like RGD, GFOGER, and HAVDI reduced MSC adhesion capacity in vitro by up to 43.5% without altering cell size, but did not significantly decrease lung entrapment in vivo. In contrast, culturing MSCs as 3D spheroids for 48 h reduced their cell diameter by 34.6% and markedly enhanced their ability to pass through the lungs and migrate to other organs like the liver after intravenous administration. This size-dependent effect on MSC distribution was more pronounced in rats compared to mice, likely due to differences in pulmonary microvessel diameters between species. CONCLUSION Our findings reveal that cell size is a predominant biophysical regulator of MSC localization in vivo compared to adhesion capacity, providing crucial insights to guide optimization of MSC delivery strategies for enhanced therapeutic efficacy.
Collapse
Affiliation(s)
- Xin Ji
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, P.R. China
| | - Lizhen Wang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Yudan Zhong
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Qian Xu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Junjie Yan
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Yuping Xu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Chongyang Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Jing Wang
- Jiangsu Renocell Biotech Co., Ltd., Nanjing, 211100, P.R. China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Min Yang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China
| | - Tiannv Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, P.R. China
| | - Lijun Tang
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, P.R. China.
| | - Xinyu Wang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, P.R. China.
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, P.R. China.
| |
Collapse
|
36
|
Hassanpour Khodaei S, Sabetkam S, Kalarestaghi H, Dizaji Asl K, Mazloumi Z, Bahramloo M, Norouzi N, Naderali E, Rafat A. Mesenchymal stem cells and mesenchymal stem cell-derived exosomes: attractive therapeutic approaches for female reproductive dysfunction. Mol Biol Rep 2024; 52:10. [PMID: 39576370 DOI: 10.1007/s11033-024-10106-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024]
Abstract
Infertility is a reproductive health problem in the male or female reproductive system. Traditional assisted reproductive technology (ART) has been unable to solve various cases of infertility for years. Clinical researchers have sought to treat infertility using new methods that are more effective and noninvasive than the old methods. Recently, Mesenchymal stem cells (MSCs) and MSCs-derived Exosomes (MSC-Exos) via paracrine activity play an important role in treating various causes of infertility and improving pregnancy outcomes. In this review, we focus on the roles of MSCs and MSC-Exos cell therapy in female infertility in the different types of female reproductive disorders.
Collapse
Affiliation(s)
- Sepideh Hassanpour Khodaei
- Department of Dentistry, Eastern Mediterranean University (EMU) Famagusta, North Cyprus Mersin 10, Famagusta, Turkey
| | - Shahnaz Sabetkam
- Department of Anatomy, Faculty of Medicine, University of Kyrenia, Kyrenia, Northern Cyprus
| | - Hossein Kalarestaghi
- Research Laboratory for Embryology and Stem Cell, Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Khadijeh Dizaji Asl
- Department of Histopathology and Anatomy, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Zeinab Mazloumi
- Department of Medical Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadmahdi Bahramloo
- Department of Medical Sciences, Student Research Committee, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Nahid Norouzi
- Nursing Trauma Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Elahe Naderali
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Ali Rafat
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
37
|
Hu Z, Zhu L, Zhu Y, Xu Y. Mesenchymal Stem Extracellular Vesicles in Various Respiratory Diseases: A New Opportunity. J Inflamm Res 2024; 17:9041-9058. [PMID: 39583853 PMCID: PMC11586120 DOI: 10.2147/jir.s480345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024] Open
Abstract
Lung diseases are associated with high morbidity and mortality rates, thereby jeopardizing human health and imposing a great burden on society. Currently, lung diseases are mainly treated with medications, oxygen therapy and mechanical ventilation, but these approaches are unable to effectively reduce the mortality rate. Therefore, lung transplantation remains the ultimate treatment for various chronic lung diseases, but this treatment is also hindered by the limited availability of lung sources, immature technology and a low survival rate after transplantation. With constant changes in the environment, pathogens, type and amount of harmful substances and the prevalence of respiratory diseases, there is an urgent need to identify alternative treatment methods. Research on stem cell therapy has been very successful in recent years, and mesenchymal stem cells (MSCs), together with their secretory bodies, play a significant therapeutic role. Extracellular vesicles of MSCs (MSC-EVs) are also major components of the paracrine secretion of MSCs, including exosomes, microvesicles, and apoptotic bodies, among which exosomes are the most typical. MSC-EVs are believed to be present in various tissues of the human body where they can carry proteins, DNA, RNA and biologically active factors, just to name a few. They can also transmit various biological signals to participate in different biological activities, including the maintenance of homeostasis within the tissue. Several studies have further demonstrated that MSCs and their generated extracellular vesicles play an important role in the treatment of diseases. In this paper, the origin, properties and roles of MSCs and MSC-EVs are reviewed, the mechanisms of different lung diseases, the limitations of current therapeutic options and the roles of MSC-EVs in Chronic Obstructive Pulmonary Disease, asthma, infectious lung disease, lung cancer, pulmonary fibrosis, pulmonary arterial hypertension, and acute lung injury/ acute respiratory distress syndrome are also discussed (Figure 1). In addition, the current limitations and possible future research directions are also discussed in view of providing new ideas for the role of MSC-EVs in the treatment of lung diseases.
Collapse
Affiliation(s)
- Zijun Hu
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, People’s Republic of China
| | - Lujian Zhu
- Department of Infectious Disease, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, People’s Republic of China
| | - Yanglin Zhu
- Department of Hepatobiliary Pancreatic Gastrointestinal Surgery 2, Affiliated Jinhua Hospital of Wenzhou Medical University, Jinhua, Zhejiang, People’s Republic of China
| | - Yejin Xu
- Department of Infectious Disease, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, People’s Republic of China
| |
Collapse
|
38
|
Nie Z, Fan Q, Jiang W, Wei S, Luo R, Hu H, Liu G, Lei Y, Xie S. Placental mesenchymal stem cells suppress inflammation and promote M2-like macrophage polarization through the IL-10/STAT3/NLRP3 axis in acute lung injury. Front Immunol 2024; 15:1422355. [PMID: 39620220 PMCID: PMC11604576 DOI: 10.3389/fimmu.2024.1422355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 10/29/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Acute lung injury (ALI) is a clinically severe respiratory disorder that currently lacks specific and effective pharmacotherapy. The imbalance of M1/M2 macrophage polarization is pivotal in the initiation and progression of ALI. Shifting macrophage polarization from the proinflammatory M1 phenotype to the anti-inflammatory M2 phenotype could be a potential therapeutic strategy. The intratracheal administration of placental mesenchymal stem cells (pMSCs) has emerged as a novel and effective treatment for ALI. This study aimed to investigate the role and downstream mechanisms of pMSCs in reprogramming macrophage polarization to exert anti-inflammatory effects in ALI. METHODS The study used lipopolysaccharide (LPS) to induce inflammation in both cell and rat models of ALI. Intratracheal administration of pMSCs was tested as a therapeutic intervention. An expression dataset for MSCs cultured with LPS-treated macrophages was collected from the Gene Expression Omnibus database to predict downstream regulatory mechanisms. Experimental validation was conducted through in vitro and in vivo assays to assess pMSCs effects on macrophage polarization and inflammation. RESULTS Both in vitro and in vivo experiments validated that pMSCs promoted M2 macrophage polarization and reduced the release of inflammatory factors. Further analyses revealed that pMSCs activated the signal transducer and activator of transcription (STAT)3 signaling pathway by secreting interleukin (IL)-10, leading to increased STAT3 phosphorylation and nuclear translocation. This activation inhibited NLRP3 inflammasome activation, promoting M2 macrophage polarization and suppressing the inflammatory response. CONCLUSION The study concluded that pMSCs alleviated lung injury in an LPS-induced ALI model by inhibiting M1 macrophage polarization and proinflammatory factor secretion, while promoting M2 macrophage polarization. This effect was mediated via the IL-10/STAT3/NLRP3 axis, presenting a novel therapeutic pathway for ALI treatment.
Collapse
Affiliation(s)
- Zhihao Nie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinglu Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shujian Wei
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Renwei Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haifeng Hu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaoli Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yufei Lei
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Songping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
39
|
Wang D, Xu L, Liu Y, Wang C, Qi S, Li Z, Bai X, Liao Y, Wang Y. Role of mesenchymal stem cells in sepsis and their therapeutic potential in sepsis‑associated myopathy (Review). Int J Mol Med 2024; 54:92. [PMID: 39219272 PMCID: PMC11374154 DOI: 10.3892/ijmm.2024.5416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis‑induced myopathy (SIM) is one of the leading causes of death in critically ill patients. SIM mainly involves the respiratory and skeletal muscles of patients, resulting in an increased risk of lung infection, aggravated respiratory failure, and prolonged mechanical ventilation and hospital stay. SIM is also an independent risk factor associated with increased mortality in critically ill patients. At present, no effective treatment for SIM has yet been established. However, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach and have been utilized in the treatment of various clinical conditions. A significant body of basic and clinical research supports the efficacy of MSCs in managing sepsis and muscle‑related diseases. This literature review aims to explore the relationship between MSCs and sepsis, as well as their impact on skeletal muscle‑associated diseases. Additionally, the present review discusses the potential mechanisms and therapeutic benefits of MSCs in the context of SIM.
Collapse
Affiliation(s)
- Dongfang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ligang Xu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Chuntao Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yiliu Liao
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
40
|
Zhai R, Tai F, Ding K, Tan X, Li H, Cao Z, Ge C, Zheng X, Fu H. Comparative Analysis of the Therapeutic Effects of MSCs From Umbilical Cord, Bone Marrow, and Adipose Tissue and Investigating the Impact of Oxidized RNA on Radiation-Induced Lung Injury. Stem Cells Int 2024; 2024:7419270. [PMID: 39483952 PMCID: PMC11527546 DOI: 10.1155/2024/7419270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/16/2024] [Accepted: 09/22/2024] [Indexed: 11/03/2024] Open
Abstract
Radiation-induced lung injury (RILI) is frequently observed in patients undergoing radiotherapy for thoracic malignancies, constituting a significant complication that hampers the effectiveness and utilization of tumor treatments. Ionizing radiation exerts both direct and indirect detrimental effects on cellular macromolecules, including DNA, RNA and proteins, but the impact of oxidized RNA in RILI remains inadequately explored. Mesenchymal stem cells (MSCs) can repair injured tissues, and the reparative potential and molecular mechanism of MSCs in treating RILI remains incompletely understood. This study aimed to investigate the therapeutic effects and mechanisms of action of three distinct sources of MSCs, including human umbilical cord mesenchymal stem cells (UCMSCs), bone marrow mesenchymal stem cells (BMSCs), and adipose-derived stem cells (ADSCs), in thoracically irradiated mice. Comparative analysis revealed that all three types of MSCs exhibited the ability to mitigate radiation-induced inflammatory infiltration, alveolar hemorrhage, and alveolar wall thickening in the lung tissue of the mice. MSCs also attenuated RILI by decreasing inflammatory factors, upregulating anti-inflammatory factor expression, and reducing collagen accumulation. Immunohistochemical results showed that all three MSCs reduced radiation-induced cell apoptosis and promoted the regeneration of lung tissue cells. The analysis of malondialdehyde (MDA) and 8-hydroyguanosine (8-OHG) content indicated that MSCs possess reparative properties against radiation-induced oxidative damage in lung tissue. The study provides evidence that UCMSCs are a more appropriate therapeutic option for RILI compared to BMSCs and ADSCs. Additionally, MSCs effectively reduce the accumulation of oxidized RNA in RILI, thereby, presenting a unique avenue for investigating the underlying mechanism of MSC-based treatment for RILI.
Collapse
Affiliation(s)
- Rui Zhai
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Fumin Tai
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Kexin Ding
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xin Tan
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
- Department of Radiation Oncology, Chinese PLA General Hospital, Beijing 100853, China
| | - Hujie Li
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhengyue Cao
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Changhui Ge
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiaofei Zheng
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hanjiang Fu
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
41
|
Huang M, Liu Y, Zhang L, Wang S, Wang X, He Z. Advancements in Research on Mesenchymal Stem-Cell-Derived Exosomal miRNAs: A Pivotal Insight into Aging and Age-Related Diseases. Biomolecules 2024; 14:1354. [PMID: 39595531 PMCID: PMC11592330 DOI: 10.3390/biom14111354] [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: 09/12/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are capable of differentiating into various cell types and play a crucial role in repairing aging tissues and diseased organs. Aging manifests as a gradual loss of cellular, tissue, and organ function, leading to the progression of pathologies. Exosomes (Exos) are extracellular vesicles secreted by cells, which maintain cellular homeostasis, clear cellular debris, and facilitate communication between cells and organs. This review provides a comprehensive summary of the mechanisms for the synthesis and sorting of MSC-Exo miRNAs and summarizes the current research status of MSCs-Exos in mitigating aging and age-related diseases. It delves into the underlying molecular mechanisms, which encompass antioxidative stress, anti-inflammatory response, and the promotion of angiogenesis. Additionally, this review also discusses potential challenges in and future strategies for advancing MSC-Exo miRNA-based therapies in the treatment of aging and age-related diseases.
Collapse
Affiliation(s)
- Minglei Huang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Ye Liu
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Longze Zhang
- Scientific Research Center, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi 563000, China;
| | - Shuangmin Wang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Xianyao Wang
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China; (M.H.); (Y.L.); (S.W.)
| | - Zhixu He
- Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
42
|
Xu S, Zhang M, Wang R, Zhang J, Wang C, Xie L, Zhao W. Spatial dimension cues derived from fibrous scaffolds trigger mechanical activation to potentiate the paracrine and regenerative functions of MSCs via the FAK-PI3K/AKT axis. Acta Biomater 2024:S1742-7061(24)00631-7. [PMID: 39461692 DOI: 10.1016/j.actbio.2024.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/29/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Secretomes from mesenchymal stem cells (MSCs) have significant therapeutic potential and could be the basis for future MSCs treatments. Innovative design of the topology of biomaterials, which mechanically regulate cell behavior and function, can tremendously improve the efficacy of stem cell therapy. However, how spatial dimension cues derived from specific topology command cell mechanotransduction to regulate the paracrine function of MSCs remains unknown. In this study, the three-dimensional (3D) fibrous constructs with box-like pores and precise strand spacing from 150 µm down to only 40 µm were manufactured using melt electrowriting (MEW), which were used to systematically investigate the spatial dimension cues-triggered mechanotransduction of adipose-derived mesenchymal stem cells (Ad-MSCs) and their impact on the paracrine and regeneration function of Ad-MSCs. The results demonstrated that spatial instructions from the 3D fibrous constructs could influence the spatial reorganization of the cytoskeleton, resulting in cell elongation and augmented immunomodulatory and angiogenic paracrine effects of Ad-MSCs, which was most pronounced at a minimum strand spacing of 40 µm. Besides, mechanical activation of the FAK-PI3K/AKT axis significantly enhanced the paracrine function of Ad-MSCs. In vivo experiments demonstrated that the Ad-MSCs trained using well-defined 3D fibrous constructs with a strand spacing of 40 µm significantly promoted skin regeneration via paracrine signals. In conclusion, this study provides a new horizon for deciphering space dimension insights into the interactional mechanisms of mechanotransduction in regulating cell function, which has inspired innovations in biomaterials for improving tissue regeneration. STATEMENT OF SIGNIFICANCE: This study emphasized that designing cell-scale spatial dimension cues to command mechanical activation via the FAK-PI3K/AKT axis could significantly enhance the paracrine and regenerative functions of Ad-MSCs. Paracrine signals of Ad-MSCs triggered by mechanical activation promoted skin repair and regeneration via the immunomodulation and angiogenesis. The proposed mechanobiological signal transduction triggered by spatial dimensional cues, which potentiates the paracrine and regenerative functions of Ad-MSCs, is a promising engineering strategy and is expected to provide new inspirations for the development of biomaterials based on biophysical signals for cellular behavior modulation.
Collapse
Affiliation(s)
- Shixin Xu
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Miaomiao Zhang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Ruoying Wang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jinxin Zhang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chengwei Wang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Li Xie
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Wen Zhao
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
43
|
Muniz TDTP, Rossi MC, de Vasconcelos Machado VM, Alves ALG. Mesenchymal Stem Cells and Tissue Bioengineering Applications in Sheep as Ideal Model. Stem Cells Int 2024; 2024:5176251. [PMID: 39465229 PMCID: PMC11511598 DOI: 10.1155/2024/5176251] [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: 01/25/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
The most common technologies in tissue engineering include growth factor therapies; metal implants, such as titanium; 3D bioprinting; nanoimprinting for ceramic/polymer scaffolds; and cell therapies, such as mesenchymal stem cells (MSCs). Cell therapy is a promising alternative to organ grafts and transplants in the treatment of numerous musculoskeletal diseases. MSCs have increasingly been used in generative medicine due to their specialized self-renewal, immunomodulation, multiplication, and differentiation properties. To further expand the potential of these cells in tissue repair, significant efforts are currently dedicated to the production of biomaterials with desirable short- and long-term biophysical properties that can aid the differentiation and expansion of MSCs. Biomaterials support MSC differentiation by modulating their characteristics, such as composition, mechanical properties, porosity, and topography. This review aimed to describe recent MSC approaches, including those associated with biomaterials, from experimental, clinical, and preclinical studies with sheep models.
Collapse
Affiliation(s)
- Talita D'Paula Tavares Pereira Muniz
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Mariana Correa Rossi
- Materials Engineering Department (DEMa), São Carlos Federal University (UFSCar), 13.565-905, São Carlos, Sao Paulo, Brazil
| | - Vânia Maria de Vasconcelos Machado
- Department of Veterinary Surgery and Animal Reproduction, Imaging Diagnostic Sector, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| | - Ana Liz Garcia Alves
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), 18.618-681, Botucatu, Sao Paulo, Brazil
| |
Collapse
|
44
|
Li X, Si Y, Liang J, Li M, Wang Z, Qin Y, Sun L. Enhancing bone regeneration and immunomodulation via gelatin methacryloyl hydrogel-encapsulated exosomes from osteogenic pre-differentiated mesenchymal stem cells. J Colloid Interface Sci 2024; 672:179-199. [PMID: 38838627 DOI: 10.1016/j.jcis.2024.05.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) have emerged as promising candidates for cell-free therapy in tissue regeneration. However, the native osteogenic and angiogenic capacities of MSC-Exos are often insufficient to repair critical-sized bone defects, and the underlying immune mechanisms remain elusive. Furthermore, achieving sustained delivery and stable activity of MSC-Exos at the defect site is essential for optimal therapeutic outcomes. Here, we extracted exosomes from osteogenically pre-differentiated human bone marrow mesenchymal stem cells (hBMSCs) by ultracentrifugation and encapsulated them in gelatin methacryloyl (GelMA) hydrogel to construct a composite scaffold. The resulting exosome-encapsulated hydrogel exhibited excellent mechanical properties and biocompatibility, facilitating sustained delivery of MSC-Exos. Osteogenic pre-differentiation significantly enhanced the osteogenic and angiogenic properties of MSC-Exos, promoting osteogenic differentiation of hBMSCs and angiogenesis of human umbilical vein endothelial cells (HUVECs). Furthermore, MSC-Exos induced polarization of Raw264.7 cells from a pro-inflammatory phenotype to an anti-inflammatory phenotype under simulated inflammatory conditions, thereby creating an immune microenvironment conducive to osteogenesis. RNA sequencing and bioinformatics analysis revealed that MSC-Exos activate the p53 pathway through targeted delivery of internal microRNAs and regulate macrophage polarization by reducing DNA oxidative damage. Our study highlights the potential of osteogenic exosome-encapsulated composite hydrogels for the development of cell-free scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Xiaorong Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yunhui Si
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jingxian Liang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Mengsha Li
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Zhiwei Wang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Yinying Qin
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Litao Sun
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
45
|
Inchingolo AM, Inchingolo AD, Nardelli P, Latini G, Trilli I, Ferrante L, Malcangi G, Palermo A, Inchingolo F, Dipalma G. Stem Cells: Present Understanding and Prospects for Regenerative Dentistry. J Funct Biomater 2024; 15:308. [PMID: 39452606 PMCID: PMC11508604 DOI: 10.3390/jfb15100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Regenerative medicine in dentistry focuses on repairing damaged oral tissues using advanced tools like stem cells, biomaterials, and tissue engineering (TE). Mesenchymal stem cells (MSCs) from dental sources, such as dental pulp and periodontal ligament, show significant potential for tissue regeneration due to their proliferative and differentiative abilities. This systematic review, following PRISMA guidelines, evaluated fifteen studies and identified effective strategies for improving dental, periodontal, and bone tissue regeneration through scaffolds, secretomes, and bioengineering methods. Key advancements include the use of dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) to boost cell viability and manage inflammation. Additionally, pharmacological agents like matrine and surface modifications on biomaterials improve stem cell adhesion and promote osteogenic differentiation. By integrating these approaches, regenerative medicine and TE can optimize dental therapies and enhance patient outcomes. This review highlights the potential and challenges in this field, providing a critical assessment of current research and future directions.
Collapse
Affiliation(s)
- Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Paola Nardelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giulia Latini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Irma Trilli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| |
Collapse
|
46
|
Gong X, Jiao Y, Hu H, Zhang R, Jia W, Zhao J, Liu Z, Xin Y, Han W. A prospective randomized controlled study of multi-intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure and reduced ejection fraction (PRIME-HFrEF) trial: Rationale and design. Contemp Clin Trials Commun 2024; 41:101350. [PMID: 39246626 PMCID: PMC11377133 DOI: 10.1016/j.conctc.2024.101350] [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: 04/21/2024] [Revised: 07/12/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background and objective The use of mesenchymal stem cells for heart failure treatment has gained increasing interest. However, most studies have relied on a single injection approach, with no research yet confirming the effects of multiple administrations. The present trial aims to investigate the safety and efficacy of multi-intravenous infusion of umbilical cord-mesenchymal stem cells (UC-MSCs) in patients with heart failure and reduced ejection fraction (HFrEF). Methods The PRIME-HFrEF trial is a single-center, prospective, randomized, triple-blinded, placebo-controlled trial of multi-intravenous infusion of UC-MSCs in HFrEF patients. A total of 40 patients meeting the inclusion criteria for HFrEF were enrolled and randomized 1:1 to the MSC group or the placebo group. Patients enrolled will receive intravenous injections of either UC-MSCs or placebo every 6 weeks for three times. Both groups will be followed up for 12 months. The primary safety endpoint is the incidence of serious adverse events. The primary efficacy endpoint is a change in left ventricular ejection fraction (LVEF) measured by left ventricular opacification (LVO) with contrast echocardiography and magnetic resonance imaging (MRI) at 12 months. The secondary endpoints include a composite of the incidence of death and re-hospitalization caused by heart failure at the 12th month, serum NT-proBNP, growth stimulation expressed gene 2 (ST2), and a change of right ventricular structure and function. Conclusions The PRIME-HFrEF study is designed to shed new light on multiple UC-MSC administration regimens for heart failure treatment.
Collapse
Affiliation(s)
- Xin Gong
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuheng Jiao
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Hu
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongzhen Zhang
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Han
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| |
Collapse
|
47
|
Opretzka LCF, Pinto CD, Santos JRDJ, de Lima AA, Soares MBP, Villarreal CF. Mesenchymal stem cell-derived cell-free technologies: a patent landscape. Biotechnol Lett 2024; 46:907-924. [PMID: 38900338 DOI: 10.1007/s10529-024-03506-y] [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: 01/08/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
Mesenchymal stem/stromal cells (MSC) play a pivotal role in regenerative therapies. Recent studies show that factors secreted by MSC can replicate their biological activity, driving the emergence of cell-free therapy, likely to surpass stem cell therapy. Patents are an objective measure of R&D and innovation activities, and patent mapping allows us to verify the state of the art and technology, anticipate trends, and identify emerging lines of research. This review performed a search on Derwent World Patents Index™ and retrieved 269 patent families related to the MSC-derived cell-free products. Analysis reveals an exponential increase in patents from the mid-2010s, primarily focusing on exosomes. The patent's contents offer a great diversity of applications and associated technologies by using the products as medicinal agents or drug delivery systems. Nevertheless, numerous application branches remain unexplored, suggesting vast potential for cell-free technologies alone or combined with other approaches.
Collapse
Affiliation(s)
| | - Cláudio Damasceno Pinto
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, 40296-710, Brazil
| | | | - Alyne Almeida de Lima
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, 40296-710, Brazil
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, 40296-710, Brazil
- Institute of Advanced Systems in Health, SENAI CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Cristiane Flora Villarreal
- Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, 40170-115, Brazil.
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
48
|
Yuan Y, Liu T. Influence of mesenchymal stem cells from different origins on the therapeutic effectiveness of systemic lupus erythematosus. Exp Cell Res 2024; 442:114263. [PMID: 39307406 DOI: 10.1016/j.yexcr.2024.114263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is a chronic autoimmune inflammatory disorder characterized by alterations in the balance between inflammatory and regulatory cytokines. Mesenchymal stem cells (MSCs), which are non-hematopoietic stem cells with multipotent differentiation potential, due to their immunomodulatory, tissue repair, low immunogenicity, and chemotactic properties, have garnered increasing interest in SLE treatment. Studies increasingly reveal the heterogeneous nature of MSC populations. With sources including dental pulp, adipose tissue, bone marrow, and umbilical cord, the therapeutic effects of MSCs on SLE vary depending on their origin. This review consolidates clinical research on MSCs from different sources in treating SLE and analyzes the possible causes underlying these variable outcomes. Additionally, it elucidates five potential factors impacting the outcomes of MSC therapy in SLE: the influence of the microenvironment on MSCs, the complexity and paradoxical aspects of MSC mechanisms in SLE treatment, the heterogeneity of MSCs, the in vivo differentiation potential and post-transplant survival rates of MSCs, and disparities in MSC preparation conditions.
Collapse
Affiliation(s)
- Yuan Yuan
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China.
| | - Tong Liu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China
| |
Collapse
|
49
|
Xu T, Zhang K, Hu Y, Yang R, Tang J, Fu W. Comparison of the Therapeutic Efficacy and Autophagy-Mediated Mechanisms of Action of Urine-Derived and Adipose-Derived Stem Cells in Osteoarthritis. Am J Sports Med 2024; 52:3130-3146. [PMID: 39311500 DOI: 10.1177/03635465241277176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a prevalent and disabling disease that affects a significant proportion of the global population. Urine-derived stem cells (USCs) have shown great prospects in the treatment of OA, but there is no study that has compared them with traditional stem cells. PURPOSE This study aimed to compare the therapeutic efficacy and mechanisms of USCs and adipose-derived stem cells (ADSCs) for OA treatment. STUDY DESIGN Controlled laboratory study. METHODS We compared the biological properties of USCs and ADSCs using CCK-8, colony formation, EdU, adhesion, and apoptosis assays. We evaluated the protective effects of USCs and ADSCs on IL-1β-treated OA chondrocytes by chemical staining, immunofluorescence, and Western blotting. We assessed the effects of USCs and ADSCs on chondrocyte autophagy by transmission electron microscopy, immunofluorescence, and Western blotting. We also compared the therapeutic efficacy of intra-articular injections of USCs and ADSCs by gross, histological, micro-computed tomography, and immunohistochemical analyses in an OA rat model induced by anterior cruciate ligament transection. RESULTS USCs showed higher proliferation, colony formation, DNA synthesis, adhesion, and anti-apoptotic abilities than ADSCs. Both USCs and ADSCs increased the expression of cartilage-specific proteins and decreased the expression of matrix degradation-related proteins and inflammatory factors in OA chondrocytes. USCs had a greater advantage in suppressing MMP-13 and inflammatory factors than ADSCs. Both USCs and ADSCs enhanced autophagy in OA chondrocytes, with USCs being more effective than ADSCs. The autophagy inhibitor 3-MA reduced the enhanced autophagy and protective effects of USCs and ADSCs on OA chondrocytes. CONCLUSION To our knowledge, this is the first study to explore the efficacy of USCs in the treatment of knee OA and to compare them with ADSCs. Considering the superior properties of USCs in terms of noninvasive acquisition, a high cost-benefit ratio, and low ethical concerns, our study suggests that they may be a more promising therapeutic option than ADSCs for OA treatment under rigorous regulatory pathways. CLINICAL RELEVANCE USCs may be a superior cell source for stem cells to treat knee OA, and this study strengthens the evidence for the application of USCs.
Collapse
Affiliation(s)
- Tianhao Xu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaibo Zhang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yunan Hu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Runze Yang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiexi Tang
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weili Fu
- Sports Medicine Center, Department of Orthopedic Surgery/Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Zhang W, Uyemura R, Zhong K, Guo R, Zhong L. Current Advances and Future Perspectives on Mesenchymal Stem Cell-Derived Extracellular Vesicles in Alzheimer's Disease. Aging Dis 2024; 15:2015-2027. [PMID: 38270122 PMCID: PMC11346404 DOI: 10.14336/ad.2023.1206] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 01/26/2024] Open
Abstract
The incidence of Alzheimer's disease (AD) has been increasing in recent years as the world's population ages, which poses a significant challenge to public health. Due to the complexity of pathogenesis of AD, currently there is no effective treatment for it. In recent years, cell and gene therapy has attracted widespread attention in the treatment of neurodegenerative diseases. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) represent a novel cell-free therapy with numerous advantages over cell-based therapies owing to their low immunogenicity and high safety profile. We summarize recent progress in the application of EVs for treating AD and the specific mechanisms and outline the underlying mechanisms. We also explore various methods for optimizing the function of MSC-EVs, including gene editing, modifying stem cell culture conditions and peptide modification. In addition, we discuss the therapeutic potentials of MSC-EVs, as well as the obstacles that currently impede their clinical utilization.
Collapse
Affiliation(s)
- Wenjing Zhang
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Russell Uyemura
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Kun Zhong
- American Center of Stem Cell Research and Regenerative Medicine, Farmington Hills, Michigan 48336, USA
| | - Rui Guo
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Li Zhong
- College of Life Sciences, Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|