首页 > 最新文献

Current stem cell research & therapy最新文献

英文 中文
Molecular Mechanisms and Pathways of Mesenchymal Stem Cell-mediated Therapy in Brain Cancer.
Pub Date : 2025-01-23 DOI: 10.2174/011574888X341525250116052000
Kuldeep Singh, Geetanjali Singh, Jeetendra Kumar Gupta, Alka N Choudhary, Arun Kumar, Divya Jain, Mukesh Chandra Sharma, Shivendra Kumar

Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach in the treatment of brain cancer due to their unique biological properties, including their ability to home tumor sites, modulate the tumor microenvironment, and exert anti-tumor effects. This review delves into the molecular mechanisms and pathways underlying MSC-mediated therapy in brain cancer. We explore the various signalling pathways activated by MSCs that contribute to their therapeutic efficacy, such as the PI3K/Akt, Wnt/β-catenin, and Notch pathways. Additionally, we discuss the role of exosomes and microRNAs secreted by MSCs in mediating anti-tumor effects. The review also addresses the challenges and future directions in optimizing MSC-based therapies for brain cancer, including issues related to MSC sourcing, delivery methods, and potential side effects. Through a comprehensive understanding of these mechanisms and pathways, we aim to highlight the potential of MSCs as a viable therapeutic option for brain cancer and to guide future research in this field.

{"title":"Molecular Mechanisms and Pathways of Mesenchymal Stem Cell-mediated Therapy in Brain Cancer.","authors":"Kuldeep Singh, Geetanjali Singh, Jeetendra Kumar Gupta, Alka N Choudhary, Arun Kumar, Divya Jain, Mukesh Chandra Sharma, Shivendra Kumar","doi":"10.2174/011574888X341525250116052000","DOIUrl":"https://doi.org/10.2174/011574888X341525250116052000","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach in the treatment of brain cancer due to their unique biological properties, including their ability to home tumor sites, modulate the tumor microenvironment, and exert anti-tumor effects. This review delves into the molecular mechanisms and pathways underlying MSC-mediated therapy in brain cancer. We explore the various signalling pathways activated by MSCs that contribute to their therapeutic efficacy, such as the PI3K/Akt, Wnt/β-catenin, and Notch pathways. Additionally, we discuss the role of exosomes and microRNAs secreted by MSCs in mediating anti-tumor effects. The review also addresses the challenges and future directions in optimizing MSC-based therapies for brain cancer, including issues related to MSC sourcing, delivery methods, and potential side effects. Through a comprehensive understanding of these mechanisms and pathways, we aim to highlight the potential of MSCs as a viable therapeutic option for brain cancer and to guide future research in this field.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143034762","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Anti-arthritic Effects of Undifferentiated and Chondrogenic Differentiated MSCs in MIA-induced Osteoarthritis in Wistar Rats: Involvement of Oxidative Stress and Immune Modulation.
Pub Date : 2025-01-23 DOI: 10.2174/011574888X348230241209072307
Ablaa S Saleh, Mohammed Abdel-Gabbar, Hala Gabr, Anwar Shams, Shadi Tamur, Emad A Mahdi, Osama M Ahmed

Introduction: Osteoarthritis (OA) is a degenerative joint disease that can affect the many tissues of the joint. There are no officially recognized disease-modifying therapies for clinical use at this time probably due to a lack of complete comprehension of the pathogenesis of the disease. In recent years, emerging regenerative therapy and treatments with stem cells both undifferentiated and differentiated cells have gained much attention as they can efficiently promote tissue repair and regeneration.

Methods: To determine how bone marrow-derived mesenchymal stem cells (BM-MSCs) and chondrogenic differentiated MSCs (CD-MSCs) can treat OA in rats, OA was induced in Wistar rats by injecting three doses of 100 μL physiological saline containing 1 mg of MIA into rat ankle joint of the right hind leg for three consecutive days. Following the induction, the osteoarthritic rats were injected weekly with BM-MSCs or CD-MSCs at a dose of 1x106 cells/rat/dose for three weeks. In addition to morphological and histological investigations of the ankle, spectrophotometric, ELISA, and Western blot analyses were applied to detect various immunological and molecular parameters in serum and ankle.

Results: The results of the study showed that in osteoarthritic rats, BM-MSCs and CD-MSCs significantly reduced right hind paw circumference, total leucocyte count (TLC), differential leukocyte count (DLC) of neutrophils, monocytes, lymphocytes, and eosinophils, serum rheumatoid factor (RF), prostaglandin E2 (PGE2) and interleukin (IL-) 1β levels, while they elevated serum IL-10 level. Additionally, BM-MSCs and CD-MSCs markedly reduced lipid peroxides (LPO) levels while they elevated superoxide dismutase (SOD) and glutathione-S-transferase (GST) activities. The monocyte chemoattractant protein-1 (MCP-1) level was significantly downregulated in ankle joint articular tissues by treatment with BM-MSCs or CD-MSCs while nuclear factor erythroid 2-related factor 2 (Nrf2) was upregulated; CD-MSCs treatment was more effective.

Conclusion: According to these findings, it can be inferred that BM-MSCs and CD-MSCs have anti-arthritic potential in MIA-induced OA; CD-MSCs therapy is more effective than MSCs. The ameliorative anti-arthritic effects may be mediated by suppressing inflammation and oxidative stress through the downregulation of MCP-1 and upregulation of Nrf2. Based on the obtained results, BM-MSCs and CD-MSCs therapies are promising new options that can be associated with other clinical treatments to improve cartilage regeneration and joint healing. However, more preclinical and clinical research is required to assess the benefits and safety of treating osteoarthritic patients with BM-MSCs and CD-MSCs.

{"title":"Anti-arthritic Effects of Undifferentiated and Chondrogenic Differentiated MSCs in MIA-induced Osteoarthritis in Wistar Rats: Involvement of Oxidative Stress and Immune Modulation.","authors":"Ablaa S Saleh, Mohammed Abdel-Gabbar, Hala Gabr, Anwar Shams, Shadi Tamur, Emad A Mahdi, Osama M Ahmed","doi":"10.2174/011574888X348230241209072307","DOIUrl":"https://doi.org/10.2174/011574888X348230241209072307","url":null,"abstract":"<p><strong>Introduction: </strong>Osteoarthritis (OA) is a degenerative joint disease that can affect the many tissues of the joint. There are no officially recognized disease-modifying therapies for clinical use at this time probably due to a lack of complete comprehension of the pathogenesis of the disease. In recent years, emerging regenerative therapy and treatments with stem cells both undifferentiated and differentiated cells have gained much attention as they can efficiently promote tissue repair and regeneration.</p><p><strong>Methods: </strong>To determine how bone marrow-derived mesenchymal stem cells (BM-MSCs) and chondrogenic differentiated MSCs (CD-MSCs) can treat OA in rats, OA was induced in Wistar rats by injecting three doses of 100 μL physiological saline containing 1 mg of MIA into rat ankle joint of the right hind leg for three consecutive days. Following the induction, the osteoarthritic rats were injected weekly with BM-MSCs or CD-MSCs at a dose of 1x106 cells/rat/dose for three weeks. In addition to morphological and histological investigations of the ankle, spectrophotometric, ELISA, and Western blot analyses were applied to detect various immunological and molecular parameters in serum and ankle.</p><p><strong>Results: </strong>The results of the study showed that in osteoarthritic rats, BM-MSCs and CD-MSCs significantly reduced right hind paw circumference, total leucocyte count (TLC), differential leukocyte count (DLC) of neutrophils, monocytes, lymphocytes, and eosinophils, serum rheumatoid factor (RF), prostaglandin E2 (PGE2) and interleukin (IL-) 1β levels, while they elevated serum IL-10 level. Additionally, BM-MSCs and CD-MSCs markedly reduced lipid peroxides (LPO) levels while they elevated superoxide dismutase (SOD) and glutathione-S-transferase (GST) activities. The monocyte chemoattractant protein-1 (MCP-1) level was significantly downregulated in ankle joint articular tissues by treatment with BM-MSCs or CD-MSCs while nuclear factor erythroid 2-related factor 2 (Nrf2) was upregulated; CD-MSCs treatment was more effective.</p><p><strong>Conclusion: </strong>According to these findings, it can be inferred that BM-MSCs and CD-MSCs have anti-arthritic potential in MIA-induced OA; CD-MSCs therapy is more effective than MSCs. The ameliorative anti-arthritic effects may be mediated by suppressing inflammation and oxidative stress through the downregulation of MCP-1 and upregulation of Nrf2. Based on the obtained results, BM-MSCs and CD-MSCs therapies are promising new options that can be associated with other clinical treatments to improve cartilage regeneration and joint healing. However, more preclinical and clinical research is required to assess the benefits and safety of treating osteoarthritic patients with BM-MSCs and CD-MSCs.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143034796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Semaphorin 3A Confers Protection against Oxidative Stress-induced Damage in Periodontal Ligament Stem Cells through the Activation of the Wnt/β-catenin Signaling Pathway.
Pub Date : 2025-01-17 DOI: 10.2174/011574888X343230250107145153
Haipeng He, Xueshun Yuan, Yanyan Chen, Tianyong Hu, Baohui Cheng, Ruitian Du, Jiamin Huang, Xiaorui Geng, Hongwen Li, Senqing Liu, Zhiqiang Liu

Objectives: The osteogenic potential of periodontal ligament stem cells (PDLSCs) is crucial for periodontal tissue regeneration. Prolonged and excessive oxidative stress (OS) impairs the osteogenic function of PDLSCs. Recently, Semaphorin 3A (Sema3A) has been reported to have multiple roles in bone protection. This study aimed to investigate the protective effect of Sema3A on the osteogenic differentiation of PDLSCs under OS conditions induced by hydrogen peroxide (H2O2).

Methods: PDLSCs were subjected to H2O2 treatment to induce OS. The OS indices in PDLSCs were evaluated by analyzing levels of reactive oxygen species (ROS), cell viability, and expression of antioxidant factors using relevant assay kits. A small molecule inhibitor, XAV-939, was employed to block the Wnt/β-catenin pathway. Osteogenic differentiation was assessed using alkaline phosphatase (ALP) activity staining and Alizarin Red S (ARS) staining for mineralized nodules. Expression levels of osteogenic gene markers and β-catenin were determined via real-time quantitative polymerase chain reaction (RT-qPCR) or western blot (WB) analysis.

Results: The stimulation of H2O2 induced OS in PDLSCs, resulting in a downregulation of Sema3A expression and a decrease in osteogenic markers, including ALP activity, mineralized nodule formation, and the expression of osteogenic genes (RUNX2 and ALP). However, the application of recombinant human Sema3A (rhSema3A) counteracted H2O2-induced OS and restored these osteogenic markers in PDLSCs under OS induced by H2O2. Mechanistic studies revealed that these effects were associated with an upregulation of β-catenin levels. Moreover, inhibiting β-- catenin expression compromised the protective effect of Sema3A on osteogenesis in PDLSCs under OS.

Conclusion: Sema3A exerts a protective effect against H2O2-induced OS and activates the Wnt/β-- catenin pathway to restore osteogenic differentiation impaired by OS in PDLSCs.

{"title":"Semaphorin 3A Confers Protection against Oxidative Stress-induced Damage in Periodontal Ligament Stem Cells through the Activation of the Wnt/β-catenin Signaling Pathway.","authors":"Haipeng He, Xueshun Yuan, Yanyan Chen, Tianyong Hu, Baohui Cheng, Ruitian Du, Jiamin Huang, Xiaorui Geng, Hongwen Li, Senqing Liu, Zhiqiang Liu","doi":"10.2174/011574888X343230250107145153","DOIUrl":"https://doi.org/10.2174/011574888X343230250107145153","url":null,"abstract":"<p><strong>Objectives: </strong>The osteogenic potential of periodontal ligament stem cells (PDLSCs) is crucial for periodontal tissue regeneration. Prolonged and excessive oxidative stress (OS) impairs the osteogenic function of PDLSCs. Recently, Semaphorin 3A (Sema3A) has been reported to have multiple roles in bone protection. This study aimed to investigate the protective effect of Sema3A on the osteogenic differentiation of PDLSCs under OS conditions induced by hydrogen peroxide (H2O2).</p><p><strong>Methods: </strong>PDLSCs were subjected to H2O2 treatment to induce OS. The OS indices in PDLSCs were evaluated by analyzing levels of reactive oxygen species (ROS), cell viability, and expression of antioxidant factors using relevant assay kits. A small molecule inhibitor, XAV-939, was employed to block the Wnt/β-catenin pathway. Osteogenic differentiation was assessed using alkaline phosphatase (ALP) activity staining and Alizarin Red S (ARS) staining for mineralized nodules. Expression levels of osteogenic gene markers and β-catenin were determined via real-time quantitative polymerase chain reaction (RT-qPCR) or western blot (WB) analysis.</p><p><strong>Results: </strong>The stimulation of H2O2 induced OS in PDLSCs, resulting in a downregulation of Sema3A expression and a decrease in osteogenic markers, including ALP activity, mineralized nodule formation, and the expression of osteogenic genes (RUNX2 and ALP). However, the application of recombinant human Sema3A (rhSema3A) counteracted H2O2-induced OS and restored these osteogenic markers in PDLSCs under OS induced by H2O2. Mechanistic studies revealed that these effects were associated with an upregulation of β-catenin levels. Moreover, inhibiting β-- catenin expression compromised the protective effect of Sema3A on osteogenesis in PDLSCs under OS.</p><p><strong>Conclusion: </strong>Sema3A exerts a protective effect against H2O2-induced OS and activates the Wnt/β-- catenin pathway to restore osteogenic differentiation impaired by OS in PDLSCs.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143054564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Current Applications and Future Directions for Vascular Stents with Stem Cells in the Treatment of Intracranial Aneurysms: A Mini-Review. 干细胞血管支架在颅内动脉瘤治疗中的应用现状及未来发展方向
Pub Date : 2025-01-09 DOI: 10.2174/011574888X329158241015053444
Kaustav Chattopadhyay, Sanjeev Sreenivasan, Gaurav Gupta, Arevik Abramyan, Idiberto Jose Zotarelli Filho, Srihari Sundararajan, Anil Nanda, Sudipta Roychowdhury

Vascular stents and stem cells have been used in high-acuity cases for many decades, particularly in cardiology. Providing the physician with another avenue of treatment, they have had a reasonable amount of success. However, there has been very little research conducted on seeding vascular stents with stem cells when treating intracranial aneurysms. Our work aims to understand the current literature available on the viability of such stents and the future directions one should take when choosing stents seeded with stem cells. Three computerized searches in PubMed were used. Four papers met the criteria, and two were excluded. There have been some experiments where the efficacy of vascular stents seeded with different materials was tested. G/PLL- coated stents provided multiple advantages and bioactive benefits to the mesenchymal stem cells. On the other hand, SF/SDF-1α also promoted similar benefits but provoked multiple unwanted inflammatory responses. G/PLL and SF/SDF-1α coated stents were able to provide satisfactory results but still require more extensive research to thoroughly understand their efficacies and safety. Future directions may include researching and discovering a wider array of biocompatible materials to seed the stents.

血管支架和干细胞在高敏度病例中已经使用了几十年,特别是在心脏病学中。为医生提供了另一种治疗途径,他们取得了相当大的成功。然而,目前关于干细胞植入血管支架治疗颅内动脉瘤的研究还很少。我们的工作旨在了解目前关于此类支架可行性的文献,以及在选择干细胞植入支架时应该采取的未来方向。在PubMed中使用了三种计算机搜索。4篇论文符合标准,2篇被排除。有一些实验测试了不同材料植入血管支架的效果。G/PLL包被支架对间充质干细胞具有多种优势和生物活性。另一方面,SF/SDF-1α也促进了类似的益处,但引发了多种不必要的炎症反应。G/PLL和SF/SDF-1α涂层支架能够提供令人满意的结果,但仍需要更广泛的研究来彻底了解其有效性和安全性。未来的方向可能包括研究和发现更广泛的生物相容性材料来植入支架。
{"title":"Current Applications and Future Directions for Vascular Stents with Stem Cells in the Treatment of Intracranial Aneurysms: A Mini-Review.","authors":"Kaustav Chattopadhyay, Sanjeev Sreenivasan, Gaurav Gupta, Arevik Abramyan, Idiberto Jose Zotarelli Filho, Srihari Sundararajan, Anil Nanda, Sudipta Roychowdhury","doi":"10.2174/011574888X329158241015053444","DOIUrl":"https://doi.org/10.2174/011574888X329158241015053444","url":null,"abstract":"<p><p>Vascular stents and stem cells have been used in high-acuity cases for many decades, particularly in cardiology. Providing the physician with another avenue of treatment, they have had a reasonable amount of success. However, there has been very little research conducted on seeding vascular stents with stem cells when treating intracranial aneurysms. Our work aims to understand the current literature available on the viability of such stents and the future directions one should take when choosing stents seeded with stem cells. Three computerized searches in PubMed were used. Four papers met the criteria, and two were excluded. There have been some experiments where the efficacy of vascular stents seeded with different materials was tested. G/PLL- coated stents provided multiple advantages and bioactive benefits to the mesenchymal stem cells. On the other hand, SF/SDF-1α also promoted similar benefits but provoked multiple unwanted inflammatory responses. G/PLL and SF/SDF-1α coated stents were able to provide satisfactory results but still require more extensive research to thoroughly understand their efficacies and safety. Future directions may include researching and discovering a wider array of biocompatible materials to seed the stents.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142981029","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bone Marrow Mesenchymal Stem Cells Ameliorate Diabetes and Diabetic Renal Fibrosis by Modulating the Inflammatory Factor IL-11. 骨髓间充质干细胞通过调节炎症因子IL-11改善糖尿病和糖尿病肾纤维化。
Pub Date : 2024-12-30 DOI: 10.2174/011574888X348254241216171655
Li-Lan Huang, Ji Yang, Yue-Yuan Hou, Yi-Hua Bai, Hong-Ying Jiang

Objective: This study aims to explore the therapeutic potential of mesenchymal stem cells (MSC) in treating diabetic nephropathy (DN) by investigating their effect on IL-11 modulation in a mouse model.

Methods: The effects of MSC therapy on DN were examined both in vivo and in vitro. Sixty adult male C57BL/6 mice were divided into the streptozotocin (STZ) diabetes (T1D) and the high-fat diet diabetes (T2D) models, with both groups receiving MSC treatment or saline for 4 or 8 weeks. Blood glucose, serum urea, interleukin-11 (IL-11), and kidney fibrosis markers were measured. Additionally, western blotting was used to assess levels of Type I and III collagen, E-Cadherin, α- smooth muscle actin (α-SMA), Vimentin, and ferroptosis suppressor protein 1 (FSP-1).

Results: MSC-treated T1D and T2D mice showed reduced blood glucose, serum urea, IL-11, TGF-β, and fibrosis markers (type I and III collagen, α-SMA, Vimentin, FSP-1), alongside increased E-Cadherin expression. Similar effects were observed in vitro using mouse glomerular epithelial cells, confirming MSC-mediated suppression of fibrosis pathways.

Conclusion: MSC therapy improves nephropathy, likely by inhibiting IL-11 and reducing fibrosis- related markers, making it a promising treatment for DN.

目的:通过研究间充质干细胞(MSC)在小鼠模型中对IL-11调节的影响,探讨其治疗糖尿病肾病(DN)的潜力。方法:在体内和体外观察MSC治疗DN的作用。将60只成年雄性C57BL/6小鼠分为链脲霉素(STZ)型糖尿病(T1D)和高脂饮食型糖尿病(T2D)模型,两组分别给予MSC治疗或生理盐水治疗4周和8周。测定血糖、血清尿素、白细胞介素-11 (IL-11)、肾纤维化指标。此外,western blotting检测I型和III型胶原蛋白、E-Cadherin、α-平滑肌肌动蛋白(α- sma)、Vimentin和铁下垂抑制蛋白1 (FSP-1)的水平。结果:mscs处理的T1D和T2D小鼠血糖、血清尿素、IL-11、TGF-β和纤维化标志物(I型和III型胶原、α-SMA、Vimentin、FSP-1)降低,E-Cadherin表达增加。使用小鼠肾小球上皮细胞在体外观察到类似的效果,证实了msc介导的纤维化途径的抑制。结论:MSC治疗可能通过抑制IL-11和降低纤维化相关标志物来改善肾病,使其成为DN的一种有希望的治疗方法。
{"title":"Bone Marrow Mesenchymal Stem Cells Ameliorate Diabetes and Diabetic Renal Fibrosis by Modulating the Inflammatory Factor IL-11.","authors":"Li-Lan Huang, Ji Yang, Yue-Yuan Hou, Yi-Hua Bai, Hong-Ying Jiang","doi":"10.2174/011574888X348254241216171655","DOIUrl":"https://doi.org/10.2174/011574888X348254241216171655","url":null,"abstract":"<p><strong>Objective: </strong>This study aims to explore the therapeutic potential of mesenchymal stem cells (MSC) in treating diabetic nephropathy (DN) by investigating their effect on IL-11 modulation in a mouse model.</p><p><strong>Methods: </strong>The effects of MSC therapy on DN were examined both in vivo and in vitro. Sixty adult male C57BL/6 mice were divided into the streptozotocin (STZ) diabetes (T1D) and the high-fat diet diabetes (T2D) models, with both groups receiving MSC treatment or saline for 4 or 8 weeks. Blood glucose, serum urea, interleukin-11 (IL-11), and kidney fibrosis markers were measured. Additionally, western blotting was used to assess levels of Type I and III collagen, E-Cadherin, α- smooth muscle actin (α-SMA), Vimentin, and ferroptosis suppressor protein 1 (FSP-1).</p><p><strong>Results: </strong>MSC-treated T1D and T2D mice showed reduced blood glucose, serum urea, IL-11, TGF-β, and fibrosis markers (type I and III collagen, α-SMA, Vimentin, FSP-1), alongside increased E-Cadherin expression. Similar effects were observed in vitro using mouse glomerular epithelial cells, confirming MSC-mediated suppression of fibrosis pathways.</p><p><strong>Conclusion: </strong>MSC therapy improves nephropathy, likely by inhibiting IL-11 and reducing fibrosis- related markers, making it a promising treatment for DN.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934016","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Potential Future Therapeutic Application of Mesenchymal Stem Cell-derived Exosomes in Ulcerative Colitis. 间充质干细胞衍生外泌体在溃疡性结肠炎中的潜在未来治疗应用。
Pub Date : 2024-12-30 DOI: 10.2174/011574888X340609241220053638
Huiting Qu, Shoukai He, Jie He, Chengfei Wang, Kewei Wang, Chao Deng, Ting Liu, Haibing Hua, Youyi Liu

Exosomes, a subclass of Extracellular Vesicles (EVs), are pivotal mediators of intercellular communication. Exosomes derived from Mesenchymal Stem Cells (MSCs) exhibit anti-inflammatory and immunomodulatory activities similar to that of their parental cells, which makes them a cell-free treatment strategy against Ulcerative Colitis (UC). Engineered MSC Exosomes (MSC-Exos) hold the potential to impart multifunctionality to MSCs and optimize their therapeutic effectiveness. This study provides a comprehensive overview of the research progress, mechanisms of action, and potential applications of MSC-Exos and engineered MSC-Exos in the treatment of UC.

外泌体是细胞外囊泡(EVs)的一个亚类,是细胞间通讯的关键介质。来自间充质干细胞(MSCs)的外泌体表现出与其亲本细胞相似的抗炎和免疫调节活性,这使其成为治疗溃疡性结肠炎(UC)的无细胞治疗策略。工程化间充质干细胞外泌体(MSC- exos)具有赋予间充质干细胞多功能性和优化其治疗效果的潜力。本文综述了MSC-Exos和工程化MSC-Exos在UC治疗中的研究进展、作用机制和潜在应用。
{"title":"Potential Future Therapeutic Application of Mesenchymal Stem Cell-derived Exosomes in Ulcerative Colitis.","authors":"Huiting Qu, Shoukai He, Jie He, Chengfei Wang, Kewei Wang, Chao Deng, Ting Liu, Haibing Hua, Youyi Liu","doi":"10.2174/011574888X340609241220053638","DOIUrl":"https://doi.org/10.2174/011574888X340609241220053638","url":null,"abstract":"<p><p>Exosomes, a subclass of Extracellular Vesicles (EVs), are pivotal mediators of intercellular communication. Exosomes derived from Mesenchymal Stem Cells (MSCs) exhibit anti-inflammatory and immunomodulatory activities similar to that of their parental cells, which makes them a cell-free treatment strategy against Ulcerative Colitis (UC). Engineered MSC Exosomes (MSC-Exos) hold the potential to impart multifunctionality to MSCs and optimize their therapeutic effectiveness. This study provides a comprehensive overview of the research progress, mechanisms of action, and potential applications of MSC-Exos and engineered MSC-Exos in the treatment of UC.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934061","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Knowledge Mapping of Stem Cell Therapy for Premature Ovarian Insufficiency: A Bibliometric Analysis (2000-2023). 干细胞治疗卵巢早衰的知识图谱:文献计量学分析(2000-2023)。
Pub Date : 2024-12-27 DOI: 10.2174/011574888X329310241206105808
Yuting Cao, Jinyuan Huang, Xiaoyin Fan, Yinmei Dai

Background: Premature Ovarian Failure (POI), a prevalent gynecological, endocrine disease, significantly impairs the reproductive health of women of childbearing age and presents a formidable challenge to clinicians. Until now, there has been a lack of effective treatments to fundamentally improve ovarian function in patients with POI. Stem cell therapy has emerged as a promising treatment in the field of POI, with notable research progress achieved to date.

Objective: This review sought to analyze the current status and hotspots of research on stem cell therapy for POI, forecasting future directions through bibliometrics.

Methods: Research related to stem cell therapy for POI from 2000 to 2023 was searched in the Web of Science Core Collection (WOSCC) database by setting subject-term, and the literature was analyzed econometrically using VOSviewer, CiteSpace, and the R package "bibliometrix."

Results: According to our search and screening strategy, 203 pieces of literature related to stem cell therapy for POI were obtained and analyzed. There is a marked annual increase in publications, with a particularly rapid ascent in recent years. China has become the most prolific country in this field, with 136 publications. Shanghai Jiao Tong University ranked first among many universities and institutions in terms of the number of publications and citations. Stem Cell Research & Therapy was the most popular and influential journal in the field of stem cell therapy for POI. Lai Dongmei has published the most papers, while Liu Te boasts the highest frequency of co-citations. Investigation into the mechanisms of exosomes derived from stem cells and their associated signaling pathways is anticipated to be a crucial research topic in stem cell therapy for POI.

Conclusion: This review offers the first comprehensive and systematic analysis of the field of stem cell therapy for POI, with a visual representation of the findings. By summarizing the current status and projecting forthcoming trends, this study aims to offer guidance and a reference for scholars in the field.

背景:卵巢早衰(POI)是一种常见的妇科内分泌疾病,严重损害了育龄妇女的生殖健康,对临床医生提出了巨大的挑战。到目前为止,还缺乏从根本上改善POI患者卵巢功能的有效治疗方法。干细胞治疗已成为POI领域的一种有前景的治疗方法,迄今为止取得了显著的研究进展。目的:通过文献计量学分析POI干细胞治疗的研究现状及热点,预测未来发展方向。方法:设置主题词,在Web of Science Core Collection (WOSCC)数据库中检索2000 - 2023年与POI干细胞治疗相关的研究,使用VOSviewer、CiteSpace和R软件包“bibliometrix”对文献进行计量分析。“结果:根据我们的搜索和筛选策略,我们获得并分析了203篇与POI干细胞治疗相关的文献。出版物每年都有显著的增长,近年来增长尤其迅速。中国已成为该领域最多产的国家,发表了136篇论文。上海交通大学在众多高校和机构中发表论文数量和被引次数均排名第一。《干细胞研究与治疗》是POI干细胞治疗领域最受欢迎和最具影响力的期刊。赖冬梅发表论文最多,刘特共被引频次最高。研究来自干细胞的外泌体及其相关信号通路的机制有望成为POI干细胞治疗的重要研究课题。结论:本综述首次对POI干细胞治疗领域进行了全面和系统的分析,并对研究结果进行了直观的描述。本研究旨在通过总结现状和预测未来的发展趋势,为该领域的学者提供指导和参考。
{"title":"Knowledge Mapping of Stem Cell Therapy for Premature Ovarian Insufficiency: A Bibliometric Analysis (2000-2023).","authors":"Yuting Cao, Jinyuan Huang, Xiaoyin Fan, Yinmei Dai","doi":"10.2174/011574888X329310241206105808","DOIUrl":"https://doi.org/10.2174/011574888X329310241206105808","url":null,"abstract":"<p><strong>Background: </strong>Premature Ovarian Failure (POI), a prevalent gynecological, endocrine disease, significantly impairs the reproductive health of women of childbearing age and presents a formidable challenge to clinicians. Until now, there has been a lack of effective treatments to fundamentally improve ovarian function in patients with POI. Stem cell therapy has emerged as a promising treatment in the field of POI, with notable research progress achieved to date.</p><p><strong>Objective: </strong>This review sought to analyze the current status and hotspots of research on stem cell therapy for POI, forecasting future directions through bibliometrics.</p><p><strong>Methods: </strong>Research related to stem cell therapy for POI from 2000 to 2023 was searched in the Web of Science Core Collection (WOSCC) database by setting subject-term, and the literature was analyzed econometrically using VOSviewer, CiteSpace, and the R package \"bibliometrix.\"</p><p><strong>Results: </strong>According to our search and screening strategy, 203 pieces of literature related to stem cell therapy for POI were obtained and analyzed. There is a marked annual increase in publications, with a particularly rapid ascent in recent years. China has become the most prolific country in this field, with 136 publications. Shanghai Jiao Tong University ranked first among many universities and institutions in terms of the number of publications and citations. Stem Cell Research & Therapy was the most popular and influential journal in the field of stem cell therapy for POI. Lai Dongmei has published the most papers, while Liu Te boasts the highest frequency of co-citations. Investigation into the mechanisms of exosomes derived from stem cells and their associated signaling pathways is anticipated to be a crucial research topic in stem cell therapy for POI.</p><p><strong>Conclusion: </strong>This review offers the first comprehensive and systematic analysis of the field of stem cell therapy for POI, with a visual representation of the findings. By summarizing the current status and projecting forthcoming trends, this study aims to offer guidance and a reference for scholars in the field.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934055","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immunomodulatory Roles of Mesenchymal Stem Cell-derived Extracellular Vesicles: A Promising Therapeutic Approach for Autoimmune Diseases. 间充质干细胞来源的细胞外囊泡的免疫调节作用:一种有前途的自身免疫性疾病治疗方法。
Pub Date : 2024-12-27 DOI: 10.2174/011574888X341781241216044130
Prince Ahad Mir, Md Sadique Hussain, Murtaza Ahmad Khanday, Roohi Mohi-Ud-Din, Faheem Hyder Pottoo, Reyaz Hasssan Mir

Autoimmune diseases pose a significant challenge due to their complex pathogenesis and rising prevalence. Traditional therapies are often limited by systemic side effects, immunosuppression, and lack of long-term efficacy. Mesenchymal stem cells (MSCs) have demonstrated immunomodulatory properties, primarily through the secretion of extracellular vesicles (EVs), which are now recognized as potent mediators of immune regulation. MSC-derived EVs carry bioactive molecules such as microRNAs, proteins, and lipids that influence key immune pathways, making them a promising therapeutic avenue for autoimmune diseases. This review critically examines the immunomodulatory mechanisms of MSC-derived EVs, focusing on their role in regulating T cells, B cells, and macrophages, which are central to autoimmune pathology. We explore recent preclinical and clinical studies that highlight the ability of MSC-derived EVs to reduce inflammation, promote immune tolerance, and restore tissue homeostasis in autoimmune settings. Furthermore, we discuss the advantages of EV-based therapy over MSC-based therapies, including improved safety profiles, lower immunogenicity, and scalability for clinical application. By evaluating the current landscape of MSC-derived EV research, we identify key gaps and propose innovative strategies to optimize EVbased therapies for autoimmune diseases. These strategies include engineering EVs to enhance their specificity and therapeutic efficacy, as well as integrating them with biomaterials for targeted delivery. Our review aims to provide a forward-looking perspective on the potential of MSC-derived EVs as a novel therapeutic approach, moving beyond traditional cell-based therapies to offer more precise and personalized treatment options for autoimmune diseases.

自身免疫性疾病由于其复杂的发病机制和不断上升的患病率,对治疗提出了重大挑战。传统的治疗方法往往受到全身副作用、免疫抑制和缺乏长期疗效的限制。间充质干细胞(MSCs)主要通过分泌细胞外囊泡(EVs)表现出免疫调节特性,现在认为细胞外囊泡是免疫调节的有效介质。msc衍生的ev携带生物活性分子,如影响关键免疫途径的microrna、蛋白质和脂质,使其成为自身免疫性疾病的有希望的治疗途径。本文综述了msc衍生的ev的免疫调节机制,重点关注它们在调节T细胞、B细胞和巨噬细胞中的作用,这些细胞是自身免疫病理的核心。我们探索了最近的临床前和临床研究,这些研究强调了msc衍生的ev在自身免疫环境下减少炎症、促进免疫耐受和恢复组织稳态的能力。此外,我们讨论了基于ev的治疗相对于基于msc的治疗的优势,包括更高的安全性、更低的免疫原性和临床应用的可扩展性。通过评估msc衍生EV研究的现状,我们确定了关键差距,并提出了创新策略,以优化基于EV的自身免疫性疾病治疗。这些策略包括改造电动汽车以增强其特异性和治疗效果,以及将其与生物材料结合以靶向递送。我们的综述旨在为msc衍生的ev作为一种新的治疗方法的潜力提供前瞻性的观点,超越传统的基于细胞的治疗方法,为自身免疫性疾病提供更精确和个性化的治疗选择。
{"title":"Immunomodulatory Roles of Mesenchymal Stem Cell-derived Extracellular Vesicles: A Promising Therapeutic Approach for Autoimmune Diseases.","authors":"Prince Ahad Mir, Md Sadique Hussain, Murtaza Ahmad Khanday, Roohi Mohi-Ud-Din, Faheem Hyder Pottoo, Reyaz Hasssan Mir","doi":"10.2174/011574888X341781241216044130","DOIUrl":"https://doi.org/10.2174/011574888X341781241216044130","url":null,"abstract":"<p><p>Autoimmune diseases pose a significant challenge due to their complex pathogenesis and rising prevalence. Traditional therapies are often limited by systemic side effects, immunosuppression, and lack of long-term efficacy. Mesenchymal stem cells (MSCs) have demonstrated immunomodulatory properties, primarily through the secretion of extracellular vesicles (EVs), which are now recognized as potent mediators of immune regulation. MSC-derived EVs carry bioactive molecules such as microRNAs, proteins, and lipids that influence key immune pathways, making them a promising therapeutic avenue for autoimmune diseases. This review critically examines the immunomodulatory mechanisms of MSC-derived EVs, focusing on their role in regulating T cells, B cells, and macrophages, which are central to autoimmune pathology. We explore recent preclinical and clinical studies that highlight the ability of MSC-derived EVs to reduce inflammation, promote immune tolerance, and restore tissue homeostasis in autoimmune settings. Furthermore, we discuss the advantages of EV-based therapy over MSC-based therapies, including improved safety profiles, lower immunogenicity, and scalability for clinical application. By evaluating the current landscape of MSC-derived EV research, we identify key gaps and propose innovative strategies to optimize EVbased therapies for autoimmune diseases. These strategies include engineering EVs to enhance their specificity and therapeutic efficacy, as well as integrating them with biomaterials for targeted delivery. Our review aims to provide a forward-looking perspective on the potential of MSC-derived EVs as a novel therapeutic approach, moving beyond traditional cell-based therapies to offer more precise and personalized treatment options for autoimmune diseases.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934020","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Intraarticular Injection of Stem Cell and Related Exosome Targeting Synovial Macrophages in Osteoarthritis. 骨关节炎关节内注射针对滑膜巨噬细胞的干细胞和相关外泌体。
Pub Date : 2024-12-27 DOI: 10.2174/011574888X338318241213055616
Zheng Li, Yuanchi Huang, Weisong Zhang, Wensen Jing

Osteoarthritis is a costly and debilitating condition, especially as the population ages and more people are affected. The primary osteoarthritis targets in the joint cavity are chondrocytes and synovial cells. Researchers are increasingly convinced that macrophages play a crucial role in the development or therapy of osteoarthritis despite being largely ignored in earlier studies due to their capacity to switch from a pro-inflammatory to an anti-inflammatory phenotype. Stem cell or similar extracellular vesicle intraarticular injection offers fresh promise for treating osteoarthritis. However, the mechanism by which this works needs further investigation. It is important to investigate the intricate cellular interactions between mesenchymal stem cells (MSCs) and macrophages. Emerging routes using extracellular vesicles (EVs) are garnering more and more attention in intercellular communication, which has historically focused on cytokines and soluble mediators. Therefore, we focus on the polarization of macrophages as a primary consideration in our study of stem cells and associated EVs utilization in treating knee osteoarthritis.

骨关节炎是一种昂贵且使人衰弱的疾病,尤其是随着人口老龄化和越来越多的人受到影响。关节腔骨关节炎的主要目标是软骨细胞和滑膜细胞。研究人员越来越相信巨噬细胞在骨关节炎的发展或治疗中起着至关重要的作用,尽管在早期的研究中由于巨噬细胞能够从促炎表型转变为抗炎表型而在很大程度上被忽视。干细胞或类似的细胞外囊泡关节内注射为治疗骨关节炎提供了新的希望。然而,其工作机制还需要进一步研究。研究间充质干细胞(MSCs)和巨噬细胞之间复杂的细胞相互作用是很重要的。在细胞间通讯中,利用细胞外囊泡(EVs)的新途径越来越受到关注,历史上主要集中在细胞因子和可溶性介质上。因此,我们关注巨噬细胞的极化作为我们研究干细胞和相关ev在治疗膝关节骨关节炎中的应用的主要考虑因素。
{"title":"Intraarticular Injection of Stem Cell and Related Exosome Targeting Synovial Macrophages in Osteoarthritis.","authors":"Zheng Li, Yuanchi Huang, Weisong Zhang, Wensen Jing","doi":"10.2174/011574888X338318241213055616","DOIUrl":"https://doi.org/10.2174/011574888X338318241213055616","url":null,"abstract":"<p><p>Osteoarthritis is a costly and debilitating condition, especially as the population ages and more people are affected. The primary osteoarthritis targets in the joint cavity are chondrocytes and synovial cells. Researchers are increasingly convinced that macrophages play a crucial role in the development or therapy of osteoarthritis despite being largely ignored in earlier studies due to their capacity to switch from a pro-inflammatory to an anti-inflammatory phenotype. Stem cell or similar extracellular vesicle intraarticular injection offers fresh promise for treating osteoarthritis. However, the mechanism by which this works needs further investigation. It is important to investigate the intricate cellular interactions between mesenchymal stem cells (MSCs) and macrophages. Emerging routes using extracellular vesicles (EVs) are garnering more and more attention in intercellular communication, which has historically focused on cytokines and soluble mediators. Therefore, we focus on the polarization of macrophages as a primary consideration in our study of stem cells and associated EVs utilization in treating knee osteoarthritis.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142934039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Aerobic Training Alleviates Muscle Atrophy by Promoting the Proliferation of Skeletal Muscle Satellite Cells in Myotonic Dystrophy Type 1 by Inhibiting Glycolysis via the Upregulation of MBNL1. 有氧训练通过上调MBNL1抑制糖酵解,促进1型肌强直性营养不良骨骼肌卫星细胞增殖,从而缓解肌肉萎缩。
Pub Date : 2024-12-20 DOI: 10.2174/011574888X360503241214045130
Hui-Qi Wang, Min Guo, Jie-Qiong Lu, Ling-Yun Chen, Feng Liang, Peng-Peng Huang, Kai-Yi Song

Background: Skeletal muscle atrophy in myotonic dystrophy type 1 (DM1) is caused by abnormal skeletal muscle satellite cell (SSC) proliferation due to increased glycolysis, which impairs muscle regeneration. In DM1, RNA foci sequester muscleblind-like protein 1 (MBNL1) in the nucleus, inhibiting its role in regulating SSC proliferation. Aerobic training reduces glycolysis and increases SSC proliferation and muscle fiber volume. This study aimed to investigate whether aerobic training prevents muscle atrophy in DM1 through the regulation of glycolysis via MBNL1.

Methods: In this study, we used the HSALR transgenic mice (DM1 mice model) to investigate the effects of aerobic training on skeletal muscle atrophy and its molecular mechanisms. HSALR mice were subjected to 4 weeks of aerobic training. After aerobic training, hindlimb grip, and myofiber mean cross-sectional area (CSA) detected by haematoxylin and eosin (HE) staining were performed. In DM1 primary SSCs, cell proliferation was assessed using Pax7 and MyoD immunofluorescence and CCK-8 assays, RNA foci were detected by RNA fluorescence in situ hybridization, and total MBNL1 expression was measured by western blot. We also used lentivirus to knock down MBNL1 in DM1 primary SSCs and performed RNA sequencing and extracellular acidification rate (ECAR). Furthermore, glycolysis detected by ECAR and oxygen consumption rate (OCR) assays were performed in WT, Sedentary, and Training group SSCs. Glycolysis was inhibited with shikonin, a glycolysis inhibitor, and the proliferation of DM1 SSCs was subsequently evaluated. Finally, we engineered an adeno-associated virus specifically targeting MBNL1 to knock down MBNL1 in DM1 mice. Subsequently, we assessed hindlimb grip strength and CSA in vivo, as well as the glycolytic capacity and proliferative capacity of DM1 SSCs in vitro.

Results: Aerobic training increased hindlimb grip strength and the average myofiber CSA in DM1 mice. Additionally, aerobic training reduced RNA foci, upregulated MBNL1, and promoted SSC proliferation. Gene-set enrichment analysis (GSEA) indicated that glycolytic processes were enriched following the knockdown of MBNL1. Furthermore, ECAR showed glycolysis was enhanced after the knockdown of MBNL1. Aerobic training reduced elevated glycolysis in DM1 mice and primary SSCs. Treatment with shikonin promoted DM1 SSC proliferation. However, MBNL1 knockdown was shown to abolish the reduced glycolysis and increased proliferation capability of SSCs due to aerobic training.

Conclusion: Taken together, aerobic training suppresses glycolysis in SSCs via the upregulation of MBNL1, thereby enhancing SSC proliferation and alleviating muscle atrophy.

背景:1型肌强直性营养不良(DM1)的骨骼肌萎缩是由于糖酵解增加导致骨骼肌卫星细胞(SSC)增殖异常引起的,这损害了肌肉再生。在DM1中,RNA在细胞核中隔离了肌盲样蛋白1 (MBNL1),抑制了其调节SSC增殖的作用。有氧训练减少糖酵解,增加SSC增殖和肌纤维体积。本研究旨在探讨有氧训练是否通过MBNL1调节糖酵解来防止DM1中的肌肉萎缩。方法:采用HSALR转基因小鼠(DM1小鼠模型),研究有氧训练对骨骼肌萎缩的影响及其分子机制。HSALR小鼠进行4周的有氧训练。有氧训练后,后肢握力和肌纤维平均横截面积(CSA)通过血红素和伊红(HE)染色检测。在DM1原代ssc中,采用Pax7、MyoD免疫荧光和CCK-8检测细胞增殖,采用RNA荧光原位杂交检测RNA灶,western blot检测MBNL1总表达。我们还使用慢病毒敲除DM1初级ssc中的MBNL1,并进行RNA测序和细胞外酸化率(ECAR)。此外,在WT组、久坐组和训练组的ssc中,通过ECAR和耗氧量(OCR)检测糖酵解。糖酵解抑制剂紫草素抑制糖酵解,随后对DM1 ssc的增殖进行了评估。最后,我们设计了一种特异性靶向MBNL1的腺相关病毒,以敲除DM1小鼠中的MBNL1。随后,我们评估了DM1 SSCs的后肢握力和体内CSA,以及体外糖酵解能力和增殖能力。结果:有氧训练增加DM1小鼠后肢握力和平均肌纤维CSA。此外,有氧训练减少RNA聚焦,上调MBNL1,促进SSC增殖。基因集富集分析(GSEA)表明,MBNL1敲低后糖酵解过程富集。此外,ECAR显示MBNL1敲除后糖酵解增强。有氧训练降低DM1小鼠和原发性ssc糖酵解升高。紫草素促进DM1 SSC增殖。然而,MBNL1敲低被证明可以消除有氧训练导致的SSCs糖酵解减少和增殖能力增加。结论:综上所述,有氧训练通过上调MBNL1抑制SSC的糖酵解,从而促进SSC增殖,缓解肌肉萎缩。
{"title":"Aerobic Training Alleviates Muscle Atrophy by Promoting the Proliferation of Skeletal Muscle Satellite Cells in Myotonic Dystrophy Type 1 by Inhibiting Glycolysis via the Upregulation of MBNL1.","authors":"Hui-Qi Wang, Min Guo, Jie-Qiong Lu, Ling-Yun Chen, Feng Liang, Peng-Peng Huang, Kai-Yi Song","doi":"10.2174/011574888X360503241214045130","DOIUrl":"10.2174/011574888X360503241214045130","url":null,"abstract":"<p><strong>Background: </strong>Skeletal muscle atrophy in myotonic dystrophy type 1 (DM1) is caused by abnormal skeletal muscle satellite cell (SSC) proliferation due to increased glycolysis, which impairs muscle regeneration. In DM1, RNA foci sequester muscleblind-like protein 1 (MBNL1) in the nucleus, inhibiting its role in regulating SSC proliferation. Aerobic training reduces glycolysis and increases SSC proliferation and muscle fiber volume. This study aimed to investigate whether aerobic training prevents muscle atrophy in DM1 through the regulation of glycolysis via MBNL1.</p><p><strong>Methods: </strong>In this study, we used the HSALR transgenic mice (DM1 mice model) to investigate the effects of aerobic training on skeletal muscle atrophy and its molecular mechanisms. HSALR mice were subjected to 4 weeks of aerobic training. After aerobic training, hindlimb grip, and myofiber mean cross-sectional area (CSA) detected by haematoxylin and eosin (HE) staining were performed. In DM1 primary SSCs, cell proliferation was assessed using Pax7 and MyoD immunofluorescence and CCK-8 assays, RNA foci were detected by RNA fluorescence in situ hybridization, and total MBNL1 expression was measured by western blot. We also used lentivirus to knock down MBNL1 in DM1 primary SSCs and performed RNA sequencing and extracellular acidification rate (ECAR). Furthermore, glycolysis detected by ECAR and oxygen consumption rate (OCR) assays were performed in WT, Sedentary, and Training group SSCs. Glycolysis was inhibited with shikonin, a glycolysis inhibitor, and the proliferation of DM1 SSCs was subsequently evaluated. Finally, we engineered an adeno-associated virus specifically targeting MBNL1 to knock down MBNL1 in DM1 mice. Subsequently, we assessed hindlimb grip strength and CSA in vivo, as well as the glycolytic capacity and proliferative capacity of DM1 SSCs in vitro.</p><p><strong>Results: </strong>Aerobic training increased hindlimb grip strength and the average myofiber CSA in DM1 mice. Additionally, aerobic training reduced RNA foci, upregulated MBNL1, and promoted SSC proliferation. Gene-set enrichment analysis (GSEA) indicated that glycolytic processes were enriched following the knockdown of MBNL1. Furthermore, ECAR showed glycolysis was enhanced after the knockdown of MBNL1. Aerobic training reduced elevated glycolysis in DM1 mice and primary SSCs. Treatment with shikonin promoted DM1 SSC proliferation. However, MBNL1 knockdown was shown to abolish the reduced glycolysis and increased proliferation capability of SSCs due to aerobic training.</p><p><strong>Conclusion: </strong>Taken together, aerobic training suppresses glycolysis in SSCs via the upregulation of MBNL1, thereby enhancing SSC proliferation and alleviating muscle atrophy.</p>","PeriodicalId":93971,"journal":{"name":"Current stem cell research & therapy","volume":" ","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142879186","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Current stem cell research & therapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1