{"title":"Emerging role of mesenchymal stem cell-derived exosomes in the repair of acute kidney injury.","authors":"Juan-Juan Wang, Yu Zheng, Yan-Lin Li, Yin Xiao, Yang-Yang Ren, Yi-Qing Tian","doi":"10.4252/wjsc.v17.i3.103360","DOIUrl":"10.4252/wjsc.v17.i3.103360","url":null,"abstract":"<p><p>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 <i>via</i> 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.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"103360"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947899/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754783","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yang-Fei Yi, Zi-Qi Fan, Can Liu, Yi-Tong Ding, Yao Chen, Jie Wen, Xiao-Hong Jian, Yu-Fei Li
{"title":"Immunomodulatory effects and clinical application of exosomes derived from mesenchymal stem cells.","authors":"Yang-Fei Yi, Zi-Qi Fan, Can Liu, Yi-Tong Ding, Yao Chen, Jie Wen, Xiao-Hong Jian, Yu-Fei Li","doi":"10.4252/wjsc.v17.i3.103560","DOIUrl":"10.4252/wjsc.v17.i3.103560","url":null,"abstract":"<p><p>Exosomes (Exos) are extracellular vesicles secreted by cells and serve as crucial mediators of intercellular communication. They play a pivotal role in the pathogenesis and progression of various diseases and offer promising avenues for therapeutic interventions. Exos derived from mesenchymal stem cells (MSCs) have significant immunomodulatory properties. They effectively regulate immune responses by modulating both innate and adaptive immunity. These Exos can inhibit excessive inflammatory responses and promote tissue repair. Moreover, they participate in antigen presentation, which is essential for activating immune responses. The cargo of these Exos, including ligands, proteins, and microRNAs, can suppress T cell activity or enhance the population of immunosuppressive cells to dampen the immune response. By inhibiting lymphocyte proliferation, acting on macrophages, and increasing the population of regulatory T cells, these Exos contribute to maintaining immune and metabolic homeostasis. Furthermore, they can activate immune-related signaling pathways or serve as vehicles to deliver microRNAs and other bioactive substances to target tumor cells, which holds potential for immunotherapy applications. Given the immense therapeutic potential of MSC-derived Exos, this review comprehensively explores their mechanisms of immune regulation and therapeutic applications in areas such as infection control, tumor suppression, and autoimmune disease management. This article aims to provide valuable insights into the mechanisms behind the actions of MSC-derived Exos, offering theoretical references for their future clinical utilization as cell-free drug preparations.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"103560"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947897/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754787","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Efficacy equivalence but hidden hurdles: Can serum-free human umbilical cord mesenchymal stem cells translate to clinically superior osteoarthritis therapy.","authors":"Fang Lin, Ke-Xin Ma, Yue Ding, Xiao-Ting Liang","doi":"10.4252/wjsc.v17.i3.104566","DOIUrl":"10.4252/wjsc.v17.i3.104566","url":null,"abstract":"<p><p>This article discusses the study by Xiao <i>et al</i>, which investigated the therapeutic efficacy of serum-free cultured human umbilical cord mesenchymal stem cells (N-hUCMSCs) in a mouse model of knee osteoarthritis. The results showed that N-hUCMSCs alleviated osteoarthritis-related cartilage damage and inflammation comparably to both serum-cultured hUCMSCs and hyaluronic acid. While these findings broaden the potential clinical utility of N-hUCMSCs by circumventing certain drawbacks of serum-based cultures, the equivalence in efficacy raises important questions. First, how do N-hUCMSCs differ phenotypically from serum-cultured hUCMSCs, particularly in terms of proliferation rate, replicative capacity, and senescence profile? Second, what advantages might N-hUCMSCs offer over hyaluronic acid - a well-established therapy - beyond avoiding xenogeneic components and ethical concerns? Future research should focus on long-term phenotypic stability, sustained functional benefits, safety profiles, and mechanistic insights to ascertain whether N-hUCMSCs can surpass current standards of care.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"104566"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jing-Jun Jin, Rong-Hua Liu, Jin-Yan Chen, Kun Wang, Jun-Yong Han, Dao-Shun Nie, Yu-Qing Gong, Bin Lin, Guo-Xing Weng
{"title":"MiR-21-5p-enriched exosomes from hiPSC-derived cardiomyocytes exhibit superior cardiac repair efficacy compared to hiPSC-derived exosomes in a murine MI model.","authors":"Jing-Jun Jin, Rong-Hua Liu, Jin-Yan Chen, Kun Wang, Jun-Yong Han, Dao-Shun Nie, Yu-Qing Gong, Bin Lin, Guo-Xing Weng","doi":"10.4252/wjsc.v17.i3.101454","DOIUrl":"10.4252/wjsc.v17.i3.101454","url":null,"abstract":"<p><strong>Background: </strong>Heart disease remains a leading cause of mortality worldwide, with existing treatments often failing to effectively restore damaged myocardium. Human-induced pluripotent stem cells (hiPSCs) and their derivatives offer promising therapeutic options; however, challenges such as low retention, engraftment issues, and tumorigenic risks hinder their clinical utility. Recent focus has shifted to exosomes (exos) - nanoscale vesicles that facilitate intercellular communication - as a safer and more versatile alternative. Understanding the specific mechanisms and comparative efficacy of exos from hiPSCs <i>vs</i> hiPSC-derived cardiomyocytes (hiPSC-CMs) is crucial for advancing cardiac repair therapies.</p><p><strong>Aim: </strong>To evaluate and compare the therapeutic efficacy of exos secreted by hiPSCs and hiPSC-CMs in cardiac repair, and to elucidate the role of microRNA 21-5p (miR-21-5p) in the observed effects.</p><p><strong>Methods: </strong>We differentiated hiPSCs into CMs using small molecule methods and characterized the cells and their exos.</p><p><strong>Results: </strong>Our findings indicate that hiPSC-CMs and their exos enhanced cardiac function, reduced infarct size, and decreased myocardial fibrosis in a murine myocardial infarction model. Notably, hiPSC-CM exos outperformed hiPSC-CM cell therapy, showing improved ejection fraction and reduced apoptosis. We identified miR-21-5p, a microRNA in hiPSC-CM exos, as crucial for CM survival. Exos with miR-21-5p were absorbed by AC16 cells, suggesting a mechanism for their cytoprotective effects.</p><p><strong>Conclusion: </strong>Overall, hiPSC-CM exos could serve as a potent therapeutic agent for myocardial repair, laying the groundwork for future research into exos as a treatment for ischemic heart disease.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"101454"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947891/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754789","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Clinical experience with cryopreserved mesenchymal stem cells for cardiovascular applications: A systematic review.","authors":"Moaz Safwan, Mariam Safwan Bourgleh, Khawaja Husnain Haider","doi":"10.4252/wjsc.v17.i3.102067","DOIUrl":"10.4252/wjsc.v17.i3.102067","url":null,"abstract":"<p><strong>Background: </strong>As living biodrugs, mesenchymal stem cells (MSCs) have progressed to phase 3 clinical trials for cardiovascular applications. However, their limited immediate availability hampers their routine clinical use.</p><p><strong>Aim: </strong>To validate our hypothesis that cryopreserved MSCs (<sup>Cryo</sup>MSCs) are as safe and effective as freshly cultured MSC counterparts but carry logistical advantages.</p><p><strong>Methods: </strong>Four databases were systematically reviewed for relevant randomized controlled trials (RCTs) evaluating the safety and efficacy of <sup>Cryo</sup>MSCs from various tissue sources in treating patients with heart disease. A subgroup analysis was performed based on MSC source and post-thaw cell viability to determine treatment effects across different <sup>Cryo</sup>MSCs sources and viability status. Weighted mean differences (WMDs) and odds ratios were calculated to measure changes in the estimated treatment effects. All statistical analyses were performed using RevMan version 5.4.1 software.</p><p><strong>Results: </strong>Seven RCTs (285 patients) met the eligibility criteria for inclusion in the meta-analysis. During short-term follow-up, <sup>Cryo</sup>MSCs demonstrated a significant 2.11% improvement in left ventricular ejection fraction (LVEF) [WMD (95%CI) = 2.11 (0.66-3.56), <i>P</i> = 0.004, <i>I</i> <sup>2</sup> = 1%], with umbilical cord-derived MSCs being the most effective cell type. However, the significant effect on LVEF was not sustained over the 12 months of follow-up. Subgroup analysis demonstrated a substantial 3.44% improvement in LVEF [WMD (95%CI) = 3.44 (1.46-5.43), <i>P</i> = 0.0007, <i>I</i> <sup>2</sup> = 0%] when using MSCs with post-thaw viability exceeding 80%. There was no statistically significant difference in the frequency of major cardiac adverse events observed in rehospitalization or mortality in patients treated with <sup>Cryo</sup>MSCs <i>vs</i> the control group.</p><p><strong>Conclusion: </strong><sup>Cryo</sup>MSCs are a promising option for heart failure patients, particularly considering the current treatment options for cardiovascular diseases. Our data suggest that <sup>Cryo</sup>MSCs could be a viable alternative or complementary treatment to the current options, potentially improving patient outcomes.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"102067"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754779","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cyclodextrin host-guest complex to facilitate sinomenine-based osteoporosis therapy.","authors":"Meng-Qin Guo, Ping Hu, Zheng-Wei Huang","doi":"10.4252/wjsc.v17.i3.101376","DOIUrl":"10.4252/wjsc.v17.i3.101376","url":null,"abstract":"<p><p>Xiao <i>et al</i> reported on the natural product sinomenine (SIN), which is a traditional Chinese medicine for treating osteoporosis <i>via</i> its modulation of autophagy; however, SIN was dissolved in dimethyl sulfoxide prior to administration, which is not conducive to the development of clinical injectables. By comparing solubilization techniques, including amorphisation, emulsification, micellisation, nano-crystallisation and host-guest inclusion, we found that the solubilization of SIN by host-guest inclusion can enhance solubility and improve stability and has an increased release rate and enhanced bioavailability. Therefore, we conclude that host-guest inclusion holds promise for SIN solubilization. To solubilise SIN, we selected β-cyclodextrin as the host agent considering its excellent biocompatibility, efficient encapsulation ability, mature preparation process and adequate drug stability. If the prerequisites of SIN-β-cyclodextrin complexes in terms of safety, efficacy, stability and the relevant laws and regulations are met, its clinical application for the treatment of osteoporosis may be achieved.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"101376"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947896/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754780","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Fat mass and obesity-associated protein in mesenchymal stem cells inhibits osteoclastogenesis <i>via</i> lnc NORAD/miR-4284 axis in ankylosing spondylitis.","authors":"Wen-Jie Liu, Jia-Xin Wang, Quan-Feng Li, Yun-Hui Zhang, Peng-Fei Ji, Jia-Hao Jin, Yi-Bin Zhang, Zi-Hao Yuan, Pei Feng, Yan-Feng Wu, Hui-Yong Shen, Peng Wang","doi":"10.4252/wjsc.v17.i3.98911","DOIUrl":"10.4252/wjsc.v17.i3.98911","url":null,"abstract":"<p><strong>Background: </strong>Ankylosing spondylitis (AS) is recognized as a long-term inflammatory disorder that leads to inflammation in the spine and joints, alongside abnormal bone growth. In previous studies, we reported that mesenchymal stem cells (MSCs) derived from individuals with AS demonstrated a remarkable inhibition in the formation of osteoclasts compared to those obtained from healthy donors. The mechanism through which MSCs from AS patients achieve this inhibition remains unclear.</p><p><strong>Aim: </strong>To investigate the potential underlying mechanism by which MSCs from individuals with ankylosing spondylitis (AS-MSCs) inhibit osteoclastogenesis.</p><p><strong>Methods: </strong>We analysed fat mass and obesity-associated (FTO) protein levels in AS-MSCs and MSCs from healthy donors and investigated the effects and mechanism by which FTO in MSCs inhibits osteoclastogenesis by coculturing and measuring the levels of tartrate-resistant acid phosphatase, nuclear factor of activated T cells 1 and cathepsin K.</p><p><strong>Results: </strong>We found that FTO, an enzyme responsible for removing methyl groups from RNA, was more abundantly expressed in MSCs from AS patients than in those from healthy donors. Reducing FTO levels was shown to diminish the capacity of MSCs to inhibit osteoclast development. Further experimental results revealed that FTO affects the stability of the long non-coding RNA activated by DNA damage (NORAD) by altering its N6-methyladenosine methylation status. Deactivating NORAD in MSCs significantly increased osteoclast formation by affecting miR-4284, which could regulate the MSC-mediated inhibition of osteoclastogenesis reported in our previous research.</p><p><strong>Conclusion: </strong>This study revealed elevated FTO levels in AS-MSCs and found that FTO regulated the ability of AS-MSCs to inhibit osteoclast formation through the long noncoding RNA NORAD/miR-4284 axis.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"98911"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947893/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754786","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"DNA methyltransferase 1/miR-342-3p/Forkhead box M1 signaling axis promotes self-renewal in cervical cancer stem-like cells <i>in vitro</i> and nude mice models.","authors":"Xiao-Zheng Cao, Yao-Feng Zhang, Yu-Wei Song, Lei Yuan, Hui-Li Tang, Jin-Yuan Li, Ye-Bei Qiu, Jia-Zhi Lin, Ying-Xia Ning, Xiao-Yu Wang, Yong Xu, Shao-Qiang Lin","doi":"10.4252/wjsc.v17.i3.99472","DOIUrl":"10.4252/wjsc.v17.i3.99472","url":null,"abstract":"<p><strong>Background: </strong>Cervical cancer (CC) stem cell-like cells (CCSLCs), defined by the capacity of differentiation and self-renewal and proliferation, play a significant role in the progression of CC. However, the molecular mechanisms regulating their self-renewal are poorly understood. Therefore, elucidation of the epigenetic mechanisms that drive cancer stem cell self-renewal will enhance our ability to improve the effectiveness of targeted therapies for cancer stem cells.</p><p><strong>Aim: </strong>To explore how DNA methyltransferase 1 (DNMT1)/miR-342-3p/Forkhead box M1 (FoxM1), which have been shown to have abnormal expression in CCSLCs, and their signaling pathways could stimulate self-renewal-related stemness in CCSLCs.</p><p><strong>Methods: </strong>Sphere-forming cells derived from CC cell lines HeLa, SiHa and CaSki served as CCSLCs. Self-renewal-related stemness was identified by determining sphere and colony formation efficiency, CD133 and CD49f protein level, and SRY-box transcription factor 2 and octamer-binding transcription factor 4 mRNA level. The microRNA expression profiles between HeLa cells and HeLa-derived CCSLCs or mRNA expression profiles that HeLa-derived CCSLCs were transfected with or without miR-342-3p mimic were compared using quantitative PCR analysis. The expression levels of <i>DNMT1</i> mRNA, miR-342-3p, and FoxM1 protein were examined by quantitative real-time PCR and western blotting. <i>In vivo</i> carcinogenicity was assessed using a mouse xenograft model. The functional effects of the DNMT1/miR-342-3p/FoxM1 axis were examined by <i>in vivo</i> and <i>in vitro</i> gain-of-activity and loss-of-activity assessments. Interplay among DNMT1, miR-342-3p, and FoxM1 was tested by methylation-specific PCR and a respective luciferase reporter assay.</p><p><strong>Results: </strong>CCSLCs derived from the established HeLa cell lines displayed higher self-renewal-related stemness, including enhanced sphere and colony formation efficiency, increased CD133 and CD49f protein level, and heightened transcriptional quantity of stemness-related factors SRY-box transcription factor 2 and octamer-binding transcription factor 4 <i>in vitro</i> as well as a stronger tumorigenic potential <i>in vivo</i> compared to their parental cells. Moreover, quantitative PCR showed that the <i>miR-342-3p</i> level was downregulated in HeLa-derived CCSLCs compared to HeLa cells. Its mimic significantly decreased <i>DNMT1</i> and <i>FoxM1</i> mRNA expression levels in CCSLCs. Knockdown of <i>DNMT1</i> or <i>miR-342-3p</i> mimic transfection suppressed <i>DNMT1</i> expression, increased <i>miR-342-3p</i> quantity by promoter demethylation, and inhibited CCSLC self-renewal. Inhibition of FoxM1 by shRNA transfection also resulted in the attenuation of CCSLC self-renewal but had little effect on the DNMT1 activity and miR-342-3p expression. Furthermore, the loss of CCSLC self-renewal exerted by miR-342-3p mimic was inverted by the overexp","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"99472"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947898/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754781","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yu Fu, Yao-Ting Han, Jun-Ling Xie, Rong-Qi Liu, Bo Zhao, Xing-Liao Zhang, Jun Zhang, Jing Zhang
{"title":"Mesenchymal stem cell exosomes enhance the development of hair follicle to ameliorate androgenetic alopecia.","authors":"Yu Fu, Yao-Ting Han, Jun-Ling Xie, Rong-Qi Liu, Bo Zhao, Xing-Liao Zhang, Jun Zhang, Jing Zhang","doi":"10.4252/wjsc.v17.i3.102088","DOIUrl":"10.4252/wjsc.v17.i3.102088","url":null,"abstract":"<p><strong>Background: </strong>Mesenchymal stem cells (MSCs) and their secretome have significant potential in promoting hair follicle development. However, the effects of MSC therapy have been reported to vary due to their heterogeneous characteristics. Different sources of MSCs or culture systems may cause heterogeneity of exosomes.</p><p><strong>Aim: </strong>To define the potential of human adipose-derived MSC exosomes (hADSC-Exos) and human umbilical cord-derived MSC exosomes (hUCMSC-Exos) for improving dermal papillary cell proliferation in androgenetic alopecia.</p><p><strong>Methods: </strong>We conducted liquid chromatography-mass spectrometry proteomic analysis of hADSC-Exos and hUCMSC-Exos. Liquid chromatography-mass spectrometry suggested that hADSC-Exos were related to metabolism and immunity. Additionally, the hADSC-Exo proteins regulated the cell cycle and other 9 functional groups.</p><p><strong>Results: </strong>We verified that hADSC-Exos inhibited glycogen synthase kinase-3β expression by activating the Wnt/β-catenin signaling pathway <i>via</i> cell division cycle protein 42, and enhanced dermal papillary cell proliferation and migration. Excess dihydrotestosterone caused androgenetic alopecia by shortening the hair follicle growth phase, but hADSC-Exos reversed these effects.</p><p><strong>Conclusion: </strong>This study indicated that hair development is influenced by hADSC-Exo-mediated cell-to-cell communication <i>via</i> the Wnt/β-catenin pathway.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"102088"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947894/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754788","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ye-Zhou Shen, Guang-Ping Yang, Qi-Min Ma, Yu-Song Wang, Xin Wang
{"title":"Regulation of lncRNA-ENST on Myc-mediated mitochondrial apoptosis in mesenchymal stem cells: <i>In vitro</i> evidence implicated for acute lung injury therapeutic potential.","authors":"Ye-Zhou Shen, Guang-Ping Yang, Qi-Min Ma, Yu-Song Wang, Xin Wang","doi":"10.4252/wjsc.v17.i3.100079","DOIUrl":"10.4252/wjsc.v17.i3.100079","url":null,"abstract":"<p><strong>Background: </strong>Acute lung injury (ALI) is a fatal and heterogeneous disease. While bone marrow mesenchymal stem cells (BMSCs) have shown promise in ALI repair, their efficacy is compromised by a high apoptotic percentage. Preliminary findings have indicated that long noncoding RNA (lncRNA)-ENST expression is markedly downregulated in MSCs under ischemic and hypoxic conditions, establishing a rationale for <i>in vitro</i> exploration.</p><p><strong>Aim: </strong>To elucidate the role of lncRNA-ENST00000517482 (lncRNA-ENST) in modulating MSC apoptosis.</p><p><strong>Methods: </strong>Founded on ALI in BEAS-2B cells with lipopolysaccharide, this study employed a transwell co-culture system to study BMSC tropism. BMSCs were genetically modified to overexpress or knockdown lncRNA-ENST. After analyzing the effects on autophagy, apoptosis and cell viability, the lncRNA-ENST/miR-539/c-MYC interaction was confirmed by dual-luciferase assays.</p><p><strong>Results: </strong>These findings have revealed a strong correlation between lncRNA-ENST levels and the apoptotic and autophagic status of BMSCs. On the one hand, the over-expression of lncRNA-ENST, as determined by Cell Counting Kit-8 assays, increased the expression of autophagy markers LC3B, ATG7, and ATG5. On the other hand, it reduced apoptosis and boosted BMSC viability. In co-cultures with BEAS-2B cells, lncRNA-ENST overexpression also improved cell vitality. Additionally, by downregulating miR-539 and upregulating c-MYC, lncRNA-ENST was found to influence mitochondrial membrane potential, enhance BMSC autophagy, mitigate apoptosis and lower the secretion of pro-inflammatory cytokines interleukin-6 and interleukin-1β. Collectively, within the <i>in vitro</i> framework, these results have highlighted the therapeutic potential of BMSCs in ALI and the pivotal regulatory role of lncRNA-ENST in miR-539 and apoptosis in lipopolysaccharide-stimulated BEAS-2B cells.</p><p><strong>Conclusion: </strong>Our <i>in vitro</i> results show that enhanced lncRNA ENST expression can promote BMSC proliferation and viability by modulating the miR-539/c-MYC axis, reduce apoptosis and induce autophagy, which has suggested its therapeutic potential in the treatment of ALI.</p>","PeriodicalId":23775,"journal":{"name":"World journal of stem cells","volume":"17 3","pages":"100079"},"PeriodicalIF":3.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11947895/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754790","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}