Pub Date : 2025-10-17eCollection Date: 2025-01-01DOI: 10.1155/sci/7889271
Yu Jing, Nan Yang, Shutao Yu, Huiran Qi, Tianqing Yu, Xingyu Chen, Shuyue Wei, Weiyan Zou, Junbin Wang
Objective: To evaluate the impact of exosomes derived from rat bone marrow mesenchymal stem cells (BM-MSCs) on the malignant properties of human colorectal cancer stem cells (CRC-CSCs) and the underlying mechanism involving epithelial-mesenchymal transition (EMT).
Methods: Exosomes were isolated and characterized from rat BM-MSCs. Human CRC-CSCs were enriched from HCT116 cells and subsequently treated with the exosomes. Cellular functions, including proliferation, apoptosis, cell cycle progression, migration, and invasion, were assessed using cell counting kit-8 (CCK-8), colony formation, flow cytometry, and Transwell assays, respectively. In vivo tumorigenicity and lung metastasis were evaluated using a xenograft mouse model. Expression levels of EMT markers (E-cadherin, N-cadherin, and Vimentin) were analyzed by western blot, qPCR, and immunofluorescence.
Results: BM-MSCs-derived exosomes were efficiently internalized by HCT116-CSCs. In vitro, exosome treatment significantly enhanced cell proliferation, migration, invasion, and cell cycle progression, while suppressing apoptosis. In vivo, exosomes promoted tumor growth and lung metastasis. Mechanistically, exosome exposure induced EMT, as evidenced by decreased E-cadherin expression and increased expression of N-cadherin and vimentin in both in vitro and in vivo models.
Conclusion: Exosomes derived from rat BM-MSCs enhance the malignant phenotype and suppress apoptosis in human CRC-CSCs through the activation of the EMT pathway. These findings underscore the potential role of BM-MSC-derived exosomes in tumor microenvironment (TME) regulation and highlight their relevance as a potential therapeutic target.
{"title":"Rat Bone Marrow Mesenchymal Stem Cells-Derived Exosomes Promote the Proliferation, Invasion, and Metastasis and Inhibit Apoptosis of Colorectal Cancer Stem Cells.","authors":"Yu Jing, Nan Yang, Shutao Yu, Huiran Qi, Tianqing Yu, Xingyu Chen, Shuyue Wei, Weiyan Zou, Junbin Wang","doi":"10.1155/sci/7889271","DOIUrl":"10.1155/sci/7889271","url":null,"abstract":"<p><strong>Objective: </strong>To evaluate the impact of exosomes derived from rat bone marrow mesenchymal stem cells (BM-MSCs) on the malignant properties of human colorectal cancer stem cells (CRC-CSCs) and the underlying mechanism involving epithelial-mesenchymal transition (EMT).</p><p><strong>Methods: </strong>Exosomes were isolated and characterized from rat BM-MSCs. Human CRC-CSCs were enriched from HCT116 cells and subsequently treated with the exosomes. Cellular functions, including proliferation, apoptosis, cell cycle progression, migration, and invasion, were assessed using cell counting kit-8 (CCK-8), colony formation, flow cytometry, and Transwell assays, respectively. In vivo tumorigenicity and lung metastasis were evaluated using a xenograft mouse model. Expression levels of EMT markers (E-cadherin, N-cadherin, and Vimentin) were analyzed by western blot, qPCR, and immunofluorescence.</p><p><strong>Results: </strong>BM-MSCs-derived exosomes were efficiently internalized by HCT116-CSCs. In vitro, exosome treatment significantly enhanced cell proliferation, migration, invasion, and cell cycle progression, while suppressing apoptosis. In vivo, exosomes promoted tumor growth and lung metastasis. Mechanistically, exosome exposure induced EMT, as evidenced by decreased E-cadherin expression and increased expression of N-cadherin and vimentin in both in vitro and in vivo models.</p><p><strong>Conclusion: </strong>Exosomes derived from rat BM-MSCs enhance the malignant phenotype and suppress apoptosis in human CRC-CSCs through the activation of the EMT pathway. These findings underscore the potential role of BM-MSC-derived exosomes in tumor microenvironment (TME) regulation and highlight their relevance as a potential therapeutic target.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"7889271"},"PeriodicalIF":3.3,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12552078/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145378711","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}
Periodontal ligament stem cell spheroids are reportedly valuable for tissue regeneration; however, their application in vivo requires the use of a scaffold, which may raise safety concerns. We moulded human periodontal ligament mesenchymal stem cell (hPDLMSC) spheroids into blocks as a scaffold-free method for handling spheroids. We then examined the characteristics and osteogenic capabilities of hPDLMSC spheroid blocks in vitro and in vivo. First, the hPDLMSCs were seeded into microwell chips to form homogeneous spheroids, which were then seeded into net mould devices and cultured with rotary shaking to form hPDLMSC spheroid blocks. Next, real-time polymerase chain reaction (PCR) analysis, a live/dead assay and histological analysis were performed to investigate the properties of the hPDLMSC spheroid blocks. Finally, bone defects were created in mouse calvaria, and the defects were transplanted with hPDLMSC spheroid blocks; the osteogenic potential of the spheroid blocks was evaluated using three-dimensional (3D) micro-X-ray computed tomography (µCT) analysis and histological techniques. We identified that the expression levels of stemness markers and osteogenesis-related genes were higher in hPDLMSC spheroid blocks than in monolayer and spheroid-cultured hPDLMSCs. The live/dead assay and histological analysis revealed that there was almost no central necrosis in the hPDLMSC spheroid blocks, and hPDLMSC spheroid blocks formed nodules under osteogenic differentiation induction. Furthermore, the hPDLMSC spheroid block treatment group (without the use of scaffolds) exhibited both the nearly complete closure of the mouse calvarial bone defect and significantly increased bone microstructural parameters compared with the group in which hPDLMSC spheroids were transplanted in combination with scaffolds. Together, these findings indicate that hPDLMSC spheroid blocks possess excellent stemness and osteogenic potential, and may contribute to the establishment of novel scaffold-free therapies.
{"title":"Evaluation of the Properties and Osteogenic Potential of a Novel Scaffold-Free Material, Spheroid Blocks Comprising Fused Spheroids of Human Periodontal Ligament Mesenchymal Stem Cells.","authors":"Kotaro Sano, Satoru Onizuka, Takenori Suga, Yuichiro Oka, Sho Hironaka, Kohji Nakazawa, Hisataka Kondo, Kazunori Hamamura, Wataru Ariyoshi, Michihiko Usui","doi":"10.1155/sci/2681140","DOIUrl":"10.1155/sci/2681140","url":null,"abstract":"<p><p>Periodontal ligament stem cell spheroids are reportedly valuable for tissue regeneration; however, their application in vivo requires the use of a scaffold, which may raise safety concerns. We moulded human periodontal ligament mesenchymal stem cell (hPDLMSC) spheroids into blocks as a scaffold-free method for handling spheroids. We then examined the characteristics and osteogenic capabilities of hPDLMSC spheroid blocks in vitro and in vivo. First, the hPDLMSCs were seeded into microwell chips to form homogeneous spheroids, which were then seeded into net mould devices and cultured with rotary shaking to form hPDLMSC spheroid blocks. Next, real-time polymerase chain reaction (PCR) analysis, a live/dead assay and histological analysis were performed to investigate the properties of the hPDLMSC spheroid blocks. Finally, bone defects were created in mouse calvaria, and the defects were transplanted with hPDLMSC spheroid blocks; the osteogenic potential of the spheroid blocks was evaluated using three-dimensional (3D) micro-X-ray computed tomography (µCT) analysis and histological techniques. We identified that the expression levels of stemness markers and osteogenesis-related genes were higher in hPDLMSC spheroid blocks than in monolayer and spheroid-cultured hPDLMSCs. The live/dead assay and histological analysis revealed that there was almost no central necrosis in the hPDLMSC spheroid blocks, and hPDLMSC spheroid blocks formed nodules under osteogenic differentiation induction. Furthermore, the hPDLMSC spheroid block treatment group (without the use of scaffolds) exhibited both the nearly complete closure of the mouse calvarial bone defect and significantly increased bone microstructural parameters compared with the group in which hPDLMSC spheroids were transplanted in combination with scaffolds. Together, these findings indicate that hPDLMSC spheroid blocks possess excellent stemness and osteogenic potential, and may contribute to the establishment of novel scaffold-free therapies.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"2681140"},"PeriodicalIF":3.3,"publicationDate":"2025-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12539984/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145347528","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}
Pub Date : 2025-10-13eCollection Date: 2025-01-01DOI: 10.1155/sci/5361754
Nadiar M Mussin, Kulyash R Zhilisbayeva, Akmaral Baspakova, Madina A Kurmanalina, Amin Tamadon
Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs), including exosomes, have garnered significant attention for their therapeutic potential in regenerative medicine and inflammatory disease management. This bibliometric analysis maps the global research landscape of MSC-derived EV studies from 2014 to 2025, utilizing data from Web of Science (WoS), Scopus, and PubMed. A total of 99 research articles were analyzed after screening 335 initial records, focusing on publication trends, citation metrics, collaboration networks, and thematic evolution. The field exhibited a 27.11% annual publication growth rate, with 841 authors contributing to 70 journals, reflecting its interdisciplinary and collaborative nature. Key findings include a surge in publications from 2018 onward, driven by clinical trials targeting conditions such as COVID-19, osteoarthritis, and spinal cord injury. The United States and China led international collaborations, with 22.68% of publications involving cross-border co-authorships. Keyword analysis revealed a shift from foundational terms like "exosomes" to clinical applications like "immunomodulation" and "osteoarthritis." Despite the field's promise, limitations such as partial 2025 data and exclusion of non-English studies suggest areas for broader inclusion. This study underscores the rapid growth and clinical potential of MSC-derived EV research, providing insights for researchers and policymakers to advance therapeutic development.
间充质干细胞(MSC)衍生的细胞外囊泡(EVs),包括外泌体,因其在再生医学和炎症性疾病管理中的治疗潜力而受到广泛关注。该文献计量分析利用Web of Science (WoS)、Scopus和PubMed的数据,绘制了2014年至2025年msc衍生EV研究的全球研究格局。在筛选了335篇初始记录后,共分析了99篇研究论文,重点关注出版趋势、引用指标、合作网络和主题演变。该领域的年发表量增长率为27.11%,共有841位作者在70种期刊上发表文章,反映了其跨学科和协作的性质。主要发现包括自2018年以来,在针对COVID-19、骨关节炎和脊髓损伤等疾病的临床试验的推动下,出版物激增。美国和中国在国际合作方面处于领先地位,有22.68%的出版物涉及跨境合作。关键词分析揭示了从“外泌体”等基础术语到“免疫调节”和“骨关节炎”等临床应用的转变。尽管该领域前景光明,但2025年的部分数据和排除非英语研究等局限性表明,该领域需要更广泛的纳入。这项研究强调了msc衍生的EV研究的快速增长和临床潜力,为研究人员和政策制定者提供了推动治疗发展的见解。
{"title":"Mapping the Landscape of Mesenchymal Stem Cell-Derived Extracellular Vesicles: From Bench to Bedside.","authors":"Nadiar M Mussin, Kulyash R Zhilisbayeva, Akmaral Baspakova, Madina A Kurmanalina, Amin Tamadon","doi":"10.1155/sci/5361754","DOIUrl":"10.1155/sci/5361754","url":null,"abstract":"<p><p>Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs), including exosomes, have garnered significant attention for their therapeutic potential in regenerative medicine and inflammatory disease management. This bibliometric analysis maps the global research landscape of MSC-derived EV studies from 2014 to 2025, utilizing data from Web of Science (WoS), Scopus, and PubMed. A total of 99 research articles were analyzed after screening 335 initial records, focusing on publication trends, citation metrics, collaboration networks, and thematic evolution. The field exhibited a 27.11% annual publication growth rate, with 841 authors contributing to 70 journals, reflecting its interdisciplinary and collaborative nature. Key findings include a surge in publications from 2018 onward, driven by clinical trials targeting conditions such as COVID-19, osteoarthritis, and spinal cord injury. The United States and China led international collaborations, with 22.68% of publications involving cross-border co-authorships. Keyword analysis revealed a shift from foundational terms like \"exosomes\" to clinical applications like \"immunomodulation\" and \"osteoarthritis.\" Despite the field's promise, limitations such as partial 2025 data and exclusion of non-English studies suggest areas for broader inclusion. This study underscores the rapid growth and clinical potential of MSC-derived EV research, providing insights for researchers and policymakers to advance therapeutic development.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"5361754"},"PeriodicalIF":3.3,"publicationDate":"2025-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12537182/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145347488","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}
Pub Date : 2025-10-02eCollection Date: 2025-01-01DOI: 10.1155/sci/4789882
Sema S Hakki, S Buket Bozkurt, Zehragul Ergul, Erdal Karaoz, Seyit Ali Kayis
The aim of this study was to compare whole-genome gene expressions of periodontal ligament (PDL) and pulp (P) mesenchymal stem cells (MSCs) isolated from third molar (m), premolar (p), and deciduous (dec) teeth. Total RNAs were isolated and used for cRNA synthesis. Human Expression Hybridization Assay was used for 47,000 probes. Data were subjected to quantile normalization before analysis. Based on the differentially expressed genes, immunomodulation properties of m/p/dec-MSCs were evaluated. Lymphocytes cocultured P/pdl-MSCs were investigated for apoptosis and cell survival of phytohemagglutinin-stimulated T cells. T cells and medium supernatants were collected on Days 1 and 4 of the experiments to evaluate T-cell proliferation by WST-1 and apoptotic markers by flow cytometry. Statistical analysis demonstrated that 291 genes were differentially expressed ≥2 fold in the cells isolated from p/m/dec, and pdl/P MSCs. The most significant difference was recognized in the proenkephalin (PENK) gene (24-fold) in pPDLMSCs, epidermal growth factor-like protein 6 (EGFL6), and complement factor D (CFD) genes were differentially expressed in decPMSCs 16.9-fold and 11-fold, respectively, when compared to other MSCs. A difference in PENK mRNA expression was also confirmed by RT-PCR. Findings of the study revealed that all dental MSCs cocultured with T cells suppressed the proliferation of T cells on Day 1 when compared to T cells alone (p=0.001). The suppression of T lymphocytes proliferation, PENK, and IL-10 mRNA expressions was higher in pPDLMSCs. Highest PENK and IL10 mRNA expressions and T-cell regulation in PDLMSCs suggested that PDLMSCs might be a promising candidate for immune regulation.
{"title":"Transcriptomic Profiling of Dental Tissue-Derived Mesenchymal Stem Cells.","authors":"Sema S Hakki, S Buket Bozkurt, Zehragul Ergul, Erdal Karaoz, Seyit Ali Kayis","doi":"10.1155/sci/4789882","DOIUrl":"10.1155/sci/4789882","url":null,"abstract":"<p><p>The aim of this study was to compare whole-genome gene expressions of periodontal ligament (PDL) and pulp (P) mesenchymal stem cells (MSCs) isolated from third molar (m), premolar (p), and deciduous (dec) teeth. Total RNAs were isolated and used for cRNA synthesis. Human Expression Hybridization Assay was used for 47,000 probes. Data were subjected to quantile normalization before analysis. Based on the differentially expressed genes, immunomodulation properties of m/p/dec-MSCs were evaluated. Lymphocytes cocultured P/pdl-MSCs were investigated for apoptosis and cell survival of phytohemagglutinin-stimulated T cells. T cells and medium supernatants were collected on Days 1 and 4 of the experiments to evaluate T-cell proliferation by WST-1 and apoptotic markers by flow cytometry. Statistical analysis demonstrated that 291 genes were differentially expressed ≥2 fold in the cells isolated from p/m/dec, and pdl/P MSCs. The most significant difference was recognized in the proenkephalin (PENK) gene (24-fold) in pPDLMSCs, epidermal growth factor-like protein 6 (EGFL6), and complement factor D (CFD) genes were differentially expressed in decPMSCs 16.9-fold and 11-fold, respectively, when compared to other MSCs. A difference in PENK mRNA expression was also confirmed by RT-PCR. Findings of the study revealed that all dental MSCs cocultured with T cells suppressed the proliferation of T cells on Day 1 when compared to T cells alone (<i>p</i>=0.001). The suppression of T lymphocytes proliferation, PENK, and IL-10 mRNA expressions was higher in pPDLMSCs. Highest PENK and IL10 mRNA expressions and T-cell regulation in PDLMSCs suggested that PDLMSCs might be a promising candidate for immune regulation.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"4789882"},"PeriodicalIF":3.3,"publicationDate":"2025-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12510763/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145281169","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}
Pub Date : 2025-09-24eCollection Date: 2025-01-01DOI: 10.1155/sci/8426444
Shang Li, Yinghui Wang, Zhiqun Wang, Shijing Deng, Lan Lv, Ying Jie
Objective: To report preliminary clinical outcomes of keratolimbal allograft (KLAL) transplantation with oral mucosal transplantation (OMT) for the treatment of limbal stem cell deficiency (LSCD) with symblepharon. Methods: This is a retrospective review of patients with LSCD and symblepharon who underwent KLAL transplantation with OMT at the Department of Ophthalmology, the Capital Medical University of Beijing Tongren Hospital between 2022 and 2024. Patients with at least 3 months of postoperative follow-up and adequate pre or postoperative records were enrolled. Grades of symblepharon, corneal conjunctivalisation, vascularization, opacification, fornix depth, and best corrected visual acuity (BCVA) were evaluated preoperatively and postoperatively. In five cases, in vivo confocal microscopy (IVCM), impression cytology (IC), and immunofluorescence (IF) were performed. Results: At a postoperative follow-up of 8.27 ± 5.80 months (3-22 months), 10 of 11 eyes (90.91%) maintained a successful outcome. The grades of symblepharon, corneal conjunctivalisation, vascularization, and opacification were significantly improved after surgery (p ≤ 0.01). Significant deepening of the fornix depth in the superior (p ≤ 0.01) and inferior conjunctival (p ≤ 0.05). Two-line improvement in BCVA was seen in 8 eyes (72.73%). Recurrence of LSCD occurred in 1 eye (9.09%). Morphology and structure of corneal epithelial cells and epithelial transition around the KLAL segments were detected by IVCM, IC, and IF. Conclusions: From the preliminary clinical results, KLAL with OMT is a safe and effective surgical technique for LSCD with symblepharon, maintaining epithelial stability, and restoring the patient's ocular anatomy.
{"title":"Clinical Outcomes of Keratolimbal Allograft Transplantation With Oral Mucosal Transplantation for Limbal Stem Cell Deficiency With Symblepharon.","authors":"Shang Li, Yinghui Wang, Zhiqun Wang, Shijing Deng, Lan Lv, Ying Jie","doi":"10.1155/sci/8426444","DOIUrl":"10.1155/sci/8426444","url":null,"abstract":"<p><p><b>Objective:</b> To report preliminary clinical outcomes of keratolimbal allograft (KLAL) transplantation with oral mucosal transplantation (OMT) for the treatment of limbal stem cell deficiency (LSCD) with symblepharon. <b>Methods:</b> This is a retrospective review of patients with LSCD and symblepharon who underwent KLAL transplantation with OMT at the Department of Ophthalmology, the Capital Medical University of Beijing Tongren Hospital between 2022 and 2024. Patients with at least 3 months of postoperative follow-up and adequate pre or postoperative records were enrolled. Grades of symblepharon, corneal conjunctivalisation, vascularization, opacification, fornix depth, and best corrected visual acuity (BCVA) were evaluated preoperatively and postoperatively. In five cases, in vivo confocal microscopy (IVCM), impression cytology (IC), and immunofluorescence (IF) were performed. <b>Results:</b> At a postoperative follow-up of 8.27 ± 5.80 months (3-22 months), 10 of 11 eyes (90.91%) maintained a successful outcome. The grades of symblepharon, corneal conjunctivalisation, vascularization, and opacification were significantly improved after surgery (<i>p</i> ≤ 0.01). Significant deepening of the fornix depth in the superior (<i>p</i> ≤ 0.01) and inferior conjunctival (<i>p</i> ≤ 0.05). Two-line improvement in BCVA was seen in 8 eyes (72.73%). Recurrence of LSCD occurred in 1 eye (9.09%). Morphology and structure of corneal epithelial cells and epithelial transition around the KLAL segments were detected by IVCM, IC, and IF. <b>Conclusions:</b> From the preliminary clinical results, KLAL with OMT is a safe and effective surgical technique for LSCD with symblepharon, maintaining epithelial stability, and restoring the patient's ocular anatomy.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"8426444"},"PeriodicalIF":3.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12488307/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145213754","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}
Pub Date : 2025-09-21eCollection Date: 2025-01-01DOI: 10.1155/sci/8825935
Chaoting Yan, Wen Sun, Zhi Chen, Liu Liu, Pin Zhou, Yueguang Gu, Geng Wu, Kunpeng Wang
Osteoradionecrosis of the jaw (ORNJ) is a complication of radiation therapy that can lead to hard-to-repair bone defects. Bone marrow mesenchymal stem cells (BMSCs) have been identified as potential "seeds" for restoring bone defects. In this study, we reported extracellular matrix protein cysteine-rich angiogenic inducer 61 (CYR61) to enhance the migratory and osteogenic functions of irradiated BMSCs (IR BMSCs) by migrasomes. Various assays, including alkaline phosphatase (ALP) activity assay, Cell Counting Kit-8 (CCK-8), apoptosis analysis, qRT-PCR, western blot, ALP staining, alizarin red S (ARS) staining, wound healing assay, transwell assay, and co-immunoprecipitation (co-IP) were conducted to assess the optimal radiation dose for generating IR BMSCs and migrasome functionality. Proteomics, bioinformatics analysis, gene transfection, and molecular docking were employed to identify key molecules mediating migration and osteoblastic differentiation and its downstream mechanisms. Furthermore, confocal microscopy, transmission electron microscopy (TEM), and western blot were utilized to identify migrasomes. Results showed that a radiation dose of 2 Gy inhibited migratory and osteogenic abilities of cells without significantly affecting viability. CYR61 emerged as a pivotal molecule regulating BMSC migration and osteoblastic differentiation through binding to integrin αvβ3 at the 125th aspartic acid and activating the ERK signaling pathway. We discovered that migrasomes are the key vehicle effectively delivering CYR61 to restore migration and osteogenesis of IR BMSCs. In conclusion, migrasomes-secreted CYR61 facilitating a promotional effect can regulate the migration and osteogenesis of IR BMSCs. Thus, migrasomes-origin CYR61 may serve as potential therapeutic agents for repairing ORNJ-related bone defects.
{"title":"ECM Protein CYR61 Promotes Migration and Osteoblastic Differentiation of Irradiation BMSCs via Migrasomes.","authors":"Chaoting Yan, Wen Sun, Zhi Chen, Liu Liu, Pin Zhou, Yueguang Gu, Geng Wu, Kunpeng Wang","doi":"10.1155/sci/8825935","DOIUrl":"10.1155/sci/8825935","url":null,"abstract":"<p><p>Osteoradionecrosis of the jaw (ORNJ) is a complication of radiation therapy that can lead to hard-to-repair bone defects. Bone marrow mesenchymal stem cells (BMSCs) have been identified as potential \"seeds\" for restoring bone defects. In this study, we reported extracellular matrix protein cysteine-rich angiogenic inducer 61 (CYR61) to enhance the migratory and osteogenic functions of irradiated BMSCs (IR BMSCs) by migrasomes. Various assays, including alkaline phosphatase (ALP) activity assay, Cell Counting Kit-8 (CCK-8), apoptosis analysis, qRT-PCR, western blot, ALP staining, alizarin red S (ARS) staining, wound healing assay, transwell assay, and co-immunoprecipitation (co-IP) were conducted to assess the optimal radiation dose for generating IR BMSCs and migrasome functionality. Proteomics, bioinformatics analysis, gene transfection, and molecular docking were employed to identify key molecules mediating migration and osteoblastic differentiation and its downstream mechanisms. Furthermore, confocal microscopy, transmission electron microscopy (TEM), and western blot were utilized to identify migrasomes. Results showed that a radiation dose of 2 Gy inhibited migratory and osteogenic abilities of cells without significantly affecting viability. CYR61 emerged as a pivotal molecule regulating BMSC migration and osteoblastic differentiation through binding to integrin αvβ3 at the 125th aspartic acid and activating the ERK signaling pathway. We discovered that migrasomes are the key vehicle effectively delivering CYR61 to restore migration and osteogenesis of IR BMSCs. In conclusion, migrasomes-secreted CYR61 facilitating a promotional effect can regulate the migration and osteogenesis of IR BMSCs. Thus, migrasomes-origin CYR61 may serve as potential therapeutic agents for repairing ORNJ-related bone defects.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"8825935"},"PeriodicalIF":3.3,"publicationDate":"2025-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12476934/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145193088","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}
Pub Date : 2025-09-18eCollection Date: 2025-01-01DOI: 10.1155/sci/9621027
Si-Lin Chen, Hang Zhou, Yu-Mei Li, Yun-Wen Zheng
Background: Circadian rhythms play a crucial role in the management of the temporal organization of various physiological and cellular processes in mammalian cell types. These rhythms are involved in the regulation of the cell cycle and metabolism and have implications for pathogenesis and physiological homeostasis. Therapeutic approaches that target circadian regulation are emerging for the treatment of digestive disorders, metabolic diseases, and cancer. The proper coordination of cellular clocks is essential for tissue homeostasis and metabolic health. Methods: The exact mechanisms governing the development and regulation of the circadian clock during embryo development are still unclear. However, embryo rhythms, such as those in the suprachiasmatic nucleus (SCN), liver, kidney, adrenal gland, and intestinal system, are believed to be influenced by maternal rhythms during different stages of embryo development. These rhythms then oscillate independently from the timing marked as the key to embryo development. In this review, we synthesize our laboratory experience and summarize current research to provide insight into how circadian rhythms regulate and synchronize organ functions for growth and differentiation during embryo development. Results: Our laboratory experience and current research suggest that circadian rhythms are involved in the regulation of organ functions during embryo development. Maternal rhythms may entrain embryo rhythms during specific developmental stages, leading to independent oscillation and coordination of organ functions. This knowledge has implications for regenerative medicine and potential clinical applications. Conclusions: Circadian rhythms play a crucial role in the coordination and synchronization of organ functions for growth and differentiation during embryo development. Understanding the regulation of circadian rhythms in embryos can provide valuable information for regenerative medicine and potential clinical applications. More research is needed to fully unravel the mechanisms underlying circadian clock development and regulation during embryo development.
{"title":"Embryonic Circadian Rhythm Establishment, Homeostasis, and Dysfunction During Organogenesis.","authors":"Si-Lin Chen, Hang Zhou, Yu-Mei Li, Yun-Wen Zheng","doi":"10.1155/sci/9621027","DOIUrl":"10.1155/sci/9621027","url":null,"abstract":"<p><p><b>Background:</b> Circadian rhythms play a crucial role in the management of the temporal organization of various physiological and cellular processes in mammalian cell types. These rhythms are involved in the regulation of the cell cycle and metabolism and have implications for pathogenesis and physiological homeostasis. Therapeutic approaches that target circadian regulation are emerging for the treatment of digestive disorders, metabolic diseases, and cancer. The proper coordination of cellular clocks is essential for tissue homeostasis and metabolic health. <b>Methods:</b> The exact mechanisms governing the development and regulation of the circadian clock during embryo development are still unclear. However, embryo rhythms, such as those in the suprachiasmatic nucleus (SCN), liver, kidney, adrenal gland, and intestinal system, are believed to be influenced by maternal rhythms during different stages of embryo development. These rhythms then oscillate independently from the timing marked as the key to embryo development. In this review, we synthesize our laboratory experience and summarize current research to provide insight into how circadian rhythms regulate and synchronize organ functions for growth and differentiation during embryo development. <b>Results:</b> Our laboratory experience and current research suggest that circadian rhythms are involved in the regulation of organ functions during embryo development. Maternal rhythms may entrain embryo rhythms during specific developmental stages, leading to independent oscillation and coordination of organ functions. This knowledge has implications for regenerative medicine and potential clinical applications. <b>Conclusions:</b> Circadian rhythms play a crucial role in the coordination and synchronization of organ functions for growth and differentiation during embryo development. Understanding the regulation of circadian rhythms in embryos can provide valuable information for regenerative medicine and potential clinical applications. More research is needed to fully unravel the mechanisms underlying circadian clock development and regulation during embryo development.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"9621027"},"PeriodicalIF":3.3,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12463512/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145186794","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}
Pub Date : 2025-09-15eCollection Date: 2025-01-01DOI: 10.1155/sci/9825165
[This corrects the article DOI: 10.1155/2023/5671809.].
[这更正了文章DOI: 10.1155/2023/5671809.]
{"title":"Corrigendum to \"lncRNA <i>CYTOR</i> Facilitates Osteogenic Differentiation of Human Periodontal Ligament Stem Cells by Modulating SOX11 via Sponging miR-6512-3p\".","authors":"","doi":"10.1155/sci/9825165","DOIUrl":"https://doi.org/10.1155/sci/9825165","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1155/2023/5671809.].</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"9825165"},"PeriodicalIF":3.3,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12453894/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145131954","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}
Pub Date : 2025-09-15eCollection Date: 2025-01-01DOI: 10.1155/sci/3218993
Haosen Ji, Zheming Xu, Leiting Shen, Sisi Yang, Jingyi Jin, Chengjie Lyu, Yichao Ren, Yi Xiao, Yuebai Zhang, Shu Fang, Xiaoxia Zhao, Xiang Yan, Dengming Lai, Jinfa Tou
Background: Mesenchymal stem cells (MSCs) inhibit macrophage inflammatory response and alleviate intestinal inflammation. However, the role of MSCs in Hirschsprung-associated enterocolitis (HAEC) remains uncertain. This study aims to investigate the effects of MSCs on HAEC and the mechanisms related to macrophages and MSCs. Methods: Immunofluorescence was used to measure CD68 and p-AKT in colonic tissues of HSCR patients with HAEC. Ednrb-/- mice was used as HSCR model. The proportion of colonic tissue macrophages in WT and Ednrb-/- mice was assessed by flow cytometry. The colonic tissues injury was evaluated with HE staining and the survival curves of mice were recorded. In vitro, macrophage-induced enterocyte death was induced by lipopolysaccharide (LPS). MSCs, MSC derived exosomes, miR-223, or MK2206 were added to macrophages, and the levels of miR-223 in macrophages after exosome treatment were measured by RT-qPCR. Flow cytometry was used to assess enterocyte death, western blot was performed to measure p-AKT expression in macrophages, and enzyme-linked immunosorbent assay (ELISA) was used to detect IL-1β concentration in macrophage supernatants and serum of Edrnb -/- mice. Results: Increased expression of CD68 and p-AKT was observed in the colonic tissues of HAEC patients. Colonic instillation of MSCs derived exosomes significantly reduce the inflammatory score of colonic tissues and prolong the survival time of HAEC mice. In vitro, LPS-stimulated macrophages induce the phosphorylation of AKT and enterocyte death. Stimulation of macrophages with MSC-derived exosomes increased the content of miR-223. MSC-derived exosomes, miR-223 and MK2206 significantly reduce macrophage-induced enterocyte death, attenuated AKT phosphorylation in macrophages, and decreased IL-1β concentration in macrophage supernatants. Conclusion: Macrophages accumulate in colonic tissues during HAEC and inflammatory macrophages drive enterocyte death. MSCs derived exosomes reduce enterocyte death by suppressing AKT phosphorylation and IL-1β secretion via miR-223, and subsequently mitigate HAEC in mice. These findings suggest that MSC-derived exosomes, particularly those enriched in miR-223, may serve as a promising therapeutic strategy for the prevention or treatment of HAEC.
{"title":"Mesenchymal Stem Cell Derived Exosomes Alleviates Hirschsprung-Associated Enterocolitis by Inhibiting AKT Phosphorylation in Macrophages Through miR-223.","authors":"Haosen Ji, Zheming Xu, Leiting Shen, Sisi Yang, Jingyi Jin, Chengjie Lyu, Yichao Ren, Yi Xiao, Yuebai Zhang, Shu Fang, Xiaoxia Zhao, Xiang Yan, Dengming Lai, Jinfa Tou","doi":"10.1155/sci/3218993","DOIUrl":"10.1155/sci/3218993","url":null,"abstract":"<p><p><b>Background:</b> Mesenchymal stem cells (MSCs) inhibit macrophage inflammatory response and alleviate intestinal inflammation. However, the role of MSCs in Hirschsprung-associated enterocolitis (HAEC) remains uncertain. This study aims to investigate the effects of MSCs on HAEC and the mechanisms related to macrophages and MSCs. <b>Methods:</b> Immunofluorescence was used to measure CD68 and p-AKT in colonic tissues of HSCR patients with HAEC. Ednrb<sup>-/-</sup> mice was used as HSCR model. The proportion of colonic tissue macrophages in WT and Ednrb<sup>-/-</sup> mice was assessed by flow cytometry. The colonic tissues injury was evaluated with HE staining and the survival curves of mice were recorded. In vitro, macrophage-induced enterocyte death was induced by lipopolysaccharide (LPS). MSCs, MSC derived exosomes, miR-223, or MK2206 were added to macrophages, and the levels of miR-223 in macrophages after exosome treatment were measured by RT-qPCR. Flow cytometry was used to assess enterocyte death, western blot was performed to measure p-AKT expression in macrophages, and enzyme-linked immunosorbent assay (ELISA) was used to detect IL-1β concentration in macrophage supernatants and serum of Edrnb <sup><i>-/-</i></sup> mice. <b>Results:</b> Increased expression of CD68 and p-AKT was observed in the colonic tissues of HAEC patients. Colonic instillation of MSCs derived exosomes significantly reduce the inflammatory score of colonic tissues and prolong the survival time of HAEC mice. In vitro, LPS-stimulated macrophages induce the phosphorylation of AKT and enterocyte death. Stimulation of macrophages with MSC-derived exosomes increased the content of miR-223. MSC-derived exosomes, miR-223 and MK2206 significantly reduce macrophage-induced enterocyte death, attenuated AKT phosphorylation in macrophages, and decreased IL-1β concentration in macrophage supernatants. <b>Conclusion:</b> Macrophages accumulate in colonic tissues during HAEC and inflammatory macrophages drive enterocyte death. MSCs derived exosomes reduce enterocyte death by suppressing AKT phosphorylation and IL-1β secretion via miR-223, and subsequently mitigate HAEC in mice. These findings suggest that MSC-derived exosomes, particularly those enriched in miR-223, may serve as a promising therapeutic strategy for the prevention or treatment of HAEC.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"3218993"},"PeriodicalIF":3.3,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12453914/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145131989","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}
Pub Date : 2025-09-12eCollection Date: 2025-01-01DOI: 10.1155/sci/8019494
Mohammad-Sadegh Lotfi, Fatemeh B Rassouli
Bone defects pose significant clinical challenges, necessitating the development of innovative strategies to effectively restore damaged bone and recover normal function. Mesenchymal stem cells (MSCs) have emerged as a promising tool for bone regeneration due to their accessibility from various sources, ease of isolation and expansion, and intrinsic ability to differentiate into osteogenic lineages with minimal ethical concerns. However, successful bone repair using MSCs requires the incorporation of biocompatible osteoinductive agents, preferably derived from natural sources. Natural polyphenols, particularly flavonoids, exhibit potent pharmaceutical properties that modulate MSC fate toward osteogenic differentiation. These secondary metabolites promote osteogenesis by interacting with key bone regulatory signaling pathways, including bone morphogenetic protein 2 (BMP2)/SMAD, wingless-related integration site (Wnt)/β-catenin, nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB), and mitogen-activated protein kinase (MAPK). Beyond their osteoinductive capacity, flavonoids possess anti-inflammatory, antibacterial, and pro-angiogenic effects, which synergistically enhance bone formation both in vitro and in vivo, thereby amplifying their therapeutic potential. This review synthesizes current insights into the interaction between MSCs and natural flavonoids, detailing the molecular mechanisms driving their synergistic effects. It also highlights recent advancements in nanoformulation-based delivery systems aimed at addressing challenges like poor solubility and bioavailability. Although preclinical data strongly support the bone-protective properties of these agents, their clinical translation remains forthcoming. Future studies must focus on optimizing delivery methods, ensuring long-term safety, and rigorously validating therapeutic efficacy across various bone disorders.
{"title":"Natural Polyphenols and Mesenchymal Stem Cells: A New Insight in Bone Regenerative Medicine.","authors":"Mohammad-Sadegh Lotfi, Fatemeh B Rassouli","doi":"10.1155/sci/8019494","DOIUrl":"10.1155/sci/8019494","url":null,"abstract":"<p><p>Bone defects pose significant clinical challenges, necessitating the development of innovative strategies to effectively restore damaged bone and recover normal function. Mesenchymal stem cells (MSCs) have emerged as a promising tool for bone regeneration due to their accessibility from various sources, ease of isolation and expansion, and intrinsic ability to differentiate into osteogenic lineages with minimal ethical concerns. However, successful bone repair using MSCs requires the incorporation of biocompatible osteoinductive agents, preferably derived from natural sources. Natural polyphenols, particularly flavonoids, exhibit potent pharmaceutical properties that modulate MSC fate toward osteogenic differentiation. These secondary metabolites promote osteogenesis by interacting with key bone regulatory signaling pathways, including bone morphogenetic protein 2 (BMP2)/SMAD, wingless-related integration site (Wnt)/β-catenin, nuclear factor kappa-light-chain-enhancer of activated B cell (NF-κB), and mitogen-activated protein kinase (MAPK). Beyond their osteoinductive capacity, flavonoids possess anti-inflammatory, antibacterial, and pro-angiogenic effects, which synergistically enhance bone formation both in vitro and in vivo, thereby amplifying their therapeutic potential. This review synthesizes current insights into the interaction between MSCs and natural flavonoids, detailing the molecular mechanisms driving their synergistic effects. It also highlights recent advancements in nanoformulation-based delivery systems aimed at addressing challenges like poor solubility and bioavailability. Although preclinical data strongly support the bone-protective properties of these agents, their clinical translation remains forthcoming. Future studies must focus on optimizing delivery methods, ensuring long-term safety, and rigorously validating therapeutic efficacy across various bone disorders.</p>","PeriodicalId":21962,"journal":{"name":"Stem Cells International","volume":"2025 ","pages":"8019494"},"PeriodicalIF":3.3,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12449111/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114212","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}