首页 > 最新文献

STEM CELLS最新文献

英文 中文
Advantages of cell proliferation and immune regulation in CD146+NESTIN+ HUMSCs: insights from single-cell RNA sequencing. CD146+NESTIN+ HUMSCs 的细胞增殖和免疫调节优势:单细胞 RNA 测序的启示。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-06-24 DOI: 10.1093/stmcls/sxae063
Peng Huang, Xiaofei Qin, Chuiqin Fan, Huifeng Zhong, Manna Wang, Fuyi Chen, Maochuan Liao, Nanpeng Zheng, Hongwu Wang, Bingchun Lin, Lian Ma

The heterogeneity of stem cells is a significant factor inhibiting their clinical application, as different cell subpopulations may exhibit substantial differences in biological functions. We performed single-cell sequencing on human umbilical cord mesenchymal stem cells (HUMSCs) from 3 donors of different gestational ages (22 + 5, 28, and 39 weeks). We also compared the data with single-cell sequencing data from BMSCs from 2 public databases. The content of CD146+Nestin+ MSCs in preterm HUMSCs (22 + 5W: 30.2%, 28W: 25.8%) was higher than that in full-term HUMSCs (39W: 0.5%) and BMSCs (BMSC1: 0, BMSC2: 0.9%). Cell cycle analysis indicated a higher proportion of cells in the proliferative G2M phase in CD146+Nestin+ MSCs (40.8%) compared to CD146+Nestin- MSCs (20%) and CD146-Nestin- MSCs (12.5%). The degree of differentiation assessment suggested that CD146+Nestin+ MSCs exhibited lower differentiation than other cell subpopulations. Differential gene analysis revealed that CD146+Nestin+ MSCs overexpressed immune regulation-related factors. GO and KEGG enrichment analysis of modules identified by weighted gene co-expression network analysis suggested enrichment in pathways related to cellular immune regulation, antimicrobial activity, and proliferation. Immune-related gene analysis indicated that CD146+Nestin+ MSCs exhibited expression of multiple immune-related genes associated with "antimicrobials," "cytokines," and "cytokine receptors." Gene regulatory network analysis revealed high expression of immune-related regulators RELB, GAPB1, and EHF in CD146+Nestin+ MSCs. Our study provides a single-cell atlas of preterm HUMSCs, demonstrating the expression of CD146+Nestin+ MSCs across different tissues and confirming their advantages in cellular proliferation, antimicrobial activity, immune regulation, and low differentiation at the RNA level. This contributes valuable insights for the clinical application of HUMSCs.

干细胞的异质性是阻碍其临床应用的一个重要因素,因为不同的细胞亚群可能在生物功能上表现出巨大差异。我们对来自三个不同胎龄(22+5周、28周、39周)供体的HUMSCs进行了单细胞测序。我们还将这些数据与两个公共数据库中 BMSCs 的单细胞测序数据进行了比较。早产 HUMSCs(22 + 5 周:30.2%;28 周:25.8%)中 CD146+Nestin+ 间充质干细胞的含量高于足月 HUMSCs(39 周:0.5%)和 BMSCs(BMSC1:0;BMSC2:0.9%)。细胞周期分析表明,CD146+Nestin+间充质干细胞(40.8%)与CD146+Nestin-间充质干细胞(20%)和CD146-Nestin-间充质干细胞(12.5%)相比,处于增殖G2M期的细胞比例更高。分化程度评估表明,CD146+Nestin+间充质干细胞的分化程度低于其他细胞亚群。差异基因分析显示,CD146+Nestin+间充质干细胞过度表达免疫调节相关因子。对WGCNA鉴定出的模块进行的GO和KEGG富集分析表明,与细胞免疫调节、抗菌活性和增殖相关的通路富集。免疫相关基因分析表明,CD146+Nestin+间充质干细胞表现出与 "抗菌素"、"细胞因子 "和 "细胞因子受体 "相关的多个免疫相关基因的表达。基因调控网络分析显示,CD146+Nestin+间充质干细胞中免疫相关调控因子RELB、GAPB1和EHF的表达量较高。我们的研究提供了早产儿HUMSCs的单细胞图谱,证明了CD146+Nestin+间充质干细胞在不同组织中的表达情况,并在RNA水平上证实了它们在细胞增殖、抗菌活性、免疫调节和低分化方面的优势。这为 HUMSCs 的临床应用提供了宝贵的见解。
{"title":"Advantages of cell proliferation and immune regulation in CD146+NESTIN+ HUMSCs: insights from single-cell RNA sequencing.","authors":"Peng Huang, Xiaofei Qin, Chuiqin Fan, Huifeng Zhong, Manna Wang, Fuyi Chen, Maochuan Liao, Nanpeng Zheng, Hongwu Wang, Bingchun Lin, Lian Ma","doi":"10.1093/stmcls/sxae063","DOIUrl":"10.1093/stmcls/sxae063","url":null,"abstract":"<p><p>The heterogeneity of stem cells is a significant factor inhibiting their clinical application, as different cell subpopulations may exhibit substantial differences in biological functions. We performed single-cell sequencing on human umbilical cord mesenchymal stem cells (HUMSCs) from 3 donors of different gestational ages (22 + 5, 28, and 39 weeks). We also compared the data with single-cell sequencing data from BMSCs from 2 public databases. The content of CD146+Nestin+ MSCs in preterm HUMSCs (22 + 5W: 30.2%, 28W: 25.8%) was higher than that in full-term HUMSCs (39W: 0.5%) and BMSCs (BMSC1: 0, BMSC2: 0.9%). Cell cycle analysis indicated a higher proportion of cells in the proliferative G2M phase in CD146+Nestin+ MSCs (40.8%) compared to CD146+Nestin- MSCs (20%) and CD146-Nestin- MSCs (12.5%). The degree of differentiation assessment suggested that CD146+Nestin+ MSCs exhibited lower differentiation than other cell subpopulations. Differential gene analysis revealed that CD146+Nestin+ MSCs overexpressed immune regulation-related factors. GO and KEGG enrichment analysis of modules identified by weighted gene co-expression network analysis suggested enrichment in pathways related to cellular immune regulation, antimicrobial activity, and proliferation. Immune-related gene analysis indicated that CD146+Nestin+ MSCs exhibited expression of multiple immune-related genes associated with \"antimicrobials,\" \"cytokines,\" and \"cytokine receptors.\" Gene regulatory network analysis revealed high expression of immune-related regulators RELB, GAPB1, and EHF in CD146+Nestin+ MSCs. Our study provides a single-cell atlas of preterm HUMSCs, demonstrating the expression of CD146+Nestin+ MSCs across different tissues and confirming their advantages in cellular proliferation, antimicrobial activity, immune regulation, and low differentiation at the RNA level. This contributes valuable insights for the clinical application of HUMSCs.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12199618/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142454402","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Impact of pro-inflammatory cytokine preconditioning on metabolism and extracellular vesicles in feline mesenchymal stromal cells: a preliminary study. 促炎细胞因子预处理对猫间充质间质细胞代谢和细胞外囊泡影响的初步研究
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-06-24 DOI: 10.1093/stmcls/sxaf014
Maria Soltero-Rivera, Boaz Arzi, Lynda Bourebaba, Krzysztof Marycz

Background: Extracellular vesicles (EVs) derived from mesenchymal stem cells have shown promise in treating inflammation. This study investigates whether preconditioning feline adipose-derived stem cells (FeASCs) with inflammatory cytokines, specifically IFN-γ and TNF-α, enhances the anti-inflammatory efficacy of MSC-derived EVs.

Objective: We hypothesize that cytokine-primed FeASCs will produce EVs with improved anti-inflammatory properties and that this preconditioning will affect mitochondrial dynamics to enhance EV therapy effectiveness.

Methods: FeASCs were exposed to a TNF-α/IFN-γ combination to mimic a pro-inflammatory milieu favoring ASCs' immunosuppressive phenotype. We analyzed morphological, metabolic, and immunomodulatory characteristics of native and cytokine-primed FeASCs. EVs were assessed for anti-inflammatory and mitochondrial-related markers. We also evaluated mitochondrial function and apoptosis markers in cytokine-primed cells.

Results: Cytokine priming led to significant morphological changes in FeASCs, including enhanced cell projections and increased apoptosis. EVs from cytokine-primed FeASCs exhibited a heightened immunomodulatory profile, with increased expression of both pro-inflammatory and anti-inflammatory mediators. Transcriptomic analysis of these EVs revealed the upregulation of genes associated with cell proliferation, survival, and apoptosis. Mitochondrial function was impaired in cytokine-primed cells, but mitochondrial morphology remained unchanged. EVs from these cells contained higher levels of mitochondrial-related transcripts, indicating a compensatory response.

Conclusions: Cytokine-primed FeASCs generate EVs with enhanced immunomodulatory potential, highlighting their therapeutic promise. However, further research is needed to validate their efficacy and safety and refine preconditioning strategies to optimize EV-based therapies for inflammatory conditions. These advancements could pave the way for broader applications in regenerative medicine.

背景:从间充质干细胞(MSCs)中提取的细胞外囊泡(EVs)已显示出治疗炎症的前景。本研究探讨了用炎症细胞因子(特别是 IFN-γ 和 TNF-α)预处理猫脂肪来源干细胞(FeASCs)是否能增强间充质干细胞衍生的 EVs 的抗炎功效:我们假设细胞因子刺激的 FeASCs 将产生具有更好抗炎特性的 EVs,并且这种预处理将影响线粒体动力学,从而提高 EV 治疗的效果:方法: 将 FeASCs 暴露于 TNF-α / IFN-γ 组合,以模拟有利于 ASCs 免疫抑制表型的促炎环境。我们分析了原生和细胞因子激发的 FeASCs 的形态、代谢和免疫调节特征。我们对EV进行了抗炎和线粒体相关标记物的评估。我们还评估了细胞因子激发细胞的线粒体功能和凋亡标志物:结果:细胞因子引物导致 FeASCs 发生了显著的形态学变化,包括细胞突起增强和细胞凋亡增加。细胞因子激发的 FeASCs 的 EVs 表现出更强的免疫调节特征,促炎和抗炎介质的表达都有所增加。对这些EVs的转录组分析表明,与细胞增殖、存活和凋亡相关的基因上调。细胞因子激发的细胞线粒体功能受损,但线粒体形态保持不变。这些细胞的EVs含有更高水平的线粒体相关转录本,表明存在补偿反应:结论:细胞因子刺激的 FeASCs 产生的 EVs 具有更强的免疫调节潜力,突显了其治疗前景。然而,还需要进一步的研究来验证其疗效和安全性,并完善预处理策略,以优化基于 EV 的炎症疗法。这些进展将为再生医学的更广泛应用铺平道路。
{"title":"Impact of pro-inflammatory cytokine preconditioning on metabolism and extracellular vesicles in feline mesenchymal stromal cells: a preliminary study.","authors":"Maria Soltero-Rivera, Boaz Arzi, Lynda Bourebaba, Krzysztof Marycz","doi":"10.1093/stmcls/sxaf014","DOIUrl":"10.1093/stmcls/sxaf014","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs) derived from mesenchymal stem cells have shown promise in treating inflammation. This study investigates whether preconditioning feline adipose-derived stem cells (FeASCs) with inflammatory cytokines, specifically IFN-γ and TNF-α, enhances the anti-inflammatory efficacy of MSC-derived EVs.</p><p><strong>Objective: </strong>We hypothesize that cytokine-primed FeASCs will produce EVs with improved anti-inflammatory properties and that this preconditioning will affect mitochondrial dynamics to enhance EV therapy effectiveness.</p><p><strong>Methods: </strong>FeASCs were exposed to a TNF-α/IFN-γ combination to mimic a pro-inflammatory milieu favoring ASCs' immunosuppressive phenotype. We analyzed morphological, metabolic, and immunomodulatory characteristics of native and cytokine-primed FeASCs. EVs were assessed for anti-inflammatory and mitochondrial-related markers. We also evaluated mitochondrial function and apoptosis markers in cytokine-primed cells.</p><p><strong>Results: </strong>Cytokine priming led to significant morphological changes in FeASCs, including enhanced cell projections and increased apoptosis. EVs from cytokine-primed FeASCs exhibited a heightened immunomodulatory profile, with increased expression of both pro-inflammatory and anti-inflammatory mediators. Transcriptomic analysis of these EVs revealed the upregulation of genes associated with cell proliferation, survival, and apoptosis. Mitochondrial function was impaired in cytokine-primed cells, but mitochondrial morphology remained unchanged. EVs from these cells contained higher levels of mitochondrial-related transcripts, indicating a compensatory response.</p><p><strong>Conclusions: </strong>Cytokine-primed FeASCs generate EVs with enhanced immunomodulatory potential, highlighting their therapeutic promise. However, further research is needed to validate their efficacy and safety and refine preconditioning strategies to optimize EV-based therapies for inflammatory conditions. These advancements could pave the way for broader applications in regenerative medicine.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12199603/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143762629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
E3 ligase Trim63 promotes the chondrogenic differentiation of mesenchymal stem cells by catalyzing K27-linked cysteine ubiquitination of Myh11. E3连接酶Trim63通过催化K27-linked半胱氨酸泛素化Myh11促进间充质干细胞的软骨分化。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-06-24 DOI: 10.1093/stmcls/sxaf017
Shanyu Ye, Yanqing Wang, Ziwei Luo, Aijun Liu, Xican Li, Jiasong Guo, Wei Zhao, Dongfeng Chen, Lin Yang, Helu Liu

Mesenchymal stem cells (MSCs) are multipotent stem cells that have a chondrogenic differentiation capacity. However, the molecular mechanism underlying the chondrogenic differentiation of MSCs has not been fully elucidated, which hinders further development of MSC-based cell therapies for cartilage repair in the clinic. Here, we showed that the E3 ubiquitin ligase Trim63 positively regulates the chondrogenic differentiation of MSCs by catalyzing the K27-linked cysteine ubiquitination of Myh11. Trim63 directly interacts with Myh11 and catalyzes K27-linked ubiquitination of cys382. Mutation of cys382 diminishes Trim63-catalyzed K27-linked ubiquitination and chondrogenic differentiation of MSCs. A deficiency in Trim63 significantly impairs the chondrogenic differentiation of MSCs. Trim63 enhances the repair of articular cartilage defects in vivo. Taken together, the results of our study demonstrated that Trim63 promotes the chondrogenic differentiation of MSCs by catalyzing K27-linked cysteine ubiquitination of Myh11, which provides an alternative therapeutic target for cartilage regeneration and repair.

间充质干细胞(MSCs)是具有软骨分化能力的多能干细胞。然而,MSCs成软骨分化的分子机制尚未完全阐明,这阻碍了临床中基于MSCs的软骨修复细胞疗法的进一步发展。在这里,我们发现E3泛素连接酶Trim63通过催化K27-linked半胱氨酸泛素化Myh11正向调节MSCs的软骨分化。Trim63直接与Myh11相互作用并催化k27连接的cys382泛素化。cys382突变减少了trim63催化的k27连接的泛素化和MSCs的软骨分化。Trim63缺乏会显著损害间充质干细胞的软骨分化。Trim63在体内促进关节软骨缺损的修复。综上所述,我们的研究结果表明,Trim63通过催化Myh11的K27-linked半胱氨酸泛素化促进MSCs的软骨分化,这为软骨再生和修复提供了另一种治疗靶点。
{"title":"E3 ligase Trim63 promotes the chondrogenic differentiation of mesenchymal stem cells by catalyzing K27-linked cysteine ubiquitination of Myh11.","authors":"Shanyu Ye, Yanqing Wang, Ziwei Luo, Aijun Liu, Xican Li, Jiasong Guo, Wei Zhao, Dongfeng Chen, Lin Yang, Helu Liu","doi":"10.1093/stmcls/sxaf017","DOIUrl":"10.1093/stmcls/sxaf017","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) are multipotent stem cells that have a chondrogenic differentiation capacity. However, the molecular mechanism underlying the chondrogenic differentiation of MSCs has not been fully elucidated, which hinders further development of MSC-based cell therapies for cartilage repair in the clinic. Here, we showed that the E3 ubiquitin ligase Trim63 positively regulates the chondrogenic differentiation of MSCs by catalyzing the K27-linked cysteine ubiquitination of Myh11. Trim63 directly interacts with Myh11 and catalyzes K27-linked ubiquitination of cys382. Mutation of cys382 diminishes Trim63-catalyzed K27-linked ubiquitination and chondrogenic differentiation of MSCs. A deficiency in Trim63 significantly impairs the chondrogenic differentiation of MSCs. Trim63 enhances the repair of articular cartilage defects in vivo. Taken together, the results of our study demonstrated that Trim63 promotes the chondrogenic differentiation of MSCs by catalyzing K27-linked cysteine ubiquitination of Myh11, which provides an alternative therapeutic target for cartilage regeneration and repair.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143956961","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SARS-CoV2 infection triggers inflammatory conditions and astrogliosis-related gene expression in long-term human cortical organoids. SARS-CoV2感染在长期的人类皮质类器官中引发炎症条件和星形胶质相关基因表达。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf010
Mathilde Colinet, Ioana Chiver, Antonela Bonafina, Gérald Masset, Daniel Almansa, Emmanuel Di Valentin, Jean-Claude Twizere, Laurent Nguyen, Ira Espuny-Camacho

SARS-CoV2, severe acute respiratory syndrome coronavirus 2, is frequently associated with neurological manifestations. Despite the presence of mild to severe CNS-related symptoms in a cohort of patients, there is no consensus whether the virus can infect directly brain tissue or if the symptoms in patients are a consequence of peripheral infectivity of the virus. Here, we use long-term human stem cell-derived cortical organoids to assess SARS-CoV2 infectivity of brain cells and unravel the cell-type tropism and its downstream pathological effects. Our results show consistent and reproducible low levels of SARS-CoV2 infection of astrocytes, deep projection neurons, upper callosal neurons, and inhibitory neurons in 6 months of human cortical organoids. Interestingly, astrocytes showed the highest infection rate among all infected cell populations which led to changes in their morphology and upregulation of SERPINA3, CD44, and S100A10 astrogliosis markers. Further, transcriptomic analysis revealed overall changes in expression of genes related to cell metabolism, astrogliosis and, inflammation and further, upregulation of cell survival pathways. Thus, local and minor infectivity of SARS-CoV2 in the brain may induce widespread adverse effects and lead to the resilience of dysregulated neurons and astrocytes within an inflammatory environment.

SARS-CoV2,即严重急性呼吸综合征冠状病毒2,通常与神经系统症状相关。尽管在一组患者中存在轻度至重度中枢神经系统相关症状,但该病毒是否可以直接感染脑组织,或者患者的症状是否是病毒外周感染性的结果,目前尚无共识。在这里,我们使用人类干细胞衍生的皮质类器官来评估SARS-CoV2对脑细胞的感染性,并揭示细胞的嗜型性及其下游病理效应。我们的结果显示,在6个月大的人类皮质类器官中,星形胶质细胞、深部投射神经元、上部胼胝体神经元和抑制性神经元的SARS-CoV2感染水平一致且可重复。有趣的是,星形胶质细胞在所有感染细胞群中显示出最高的感染率,导致其形态发生变化,SERPINA3、CD44和S100A10星形胶质细胞标记物上调。此外,转录组学分析揭示了与细胞代谢、星形胶质细胞形成、炎症以及细胞存活途径上调相关的基因表达的总体变化。因此,大脑中SARS-CoV2的局部和轻微感染性可能诱发广泛的不良反应,并导致炎症环境中失调的神经元和星形胶质细胞的恢复能力。
{"title":"SARS-CoV2 infection triggers inflammatory conditions and astrogliosis-related gene expression in long-term human cortical organoids.","authors":"Mathilde Colinet, Ioana Chiver, Antonela Bonafina, Gérald Masset, Daniel Almansa, Emmanuel Di Valentin, Jean-Claude Twizere, Laurent Nguyen, Ira Espuny-Camacho","doi":"10.1093/stmcls/sxaf010","DOIUrl":"10.1093/stmcls/sxaf010","url":null,"abstract":"<p><p>SARS-CoV2, severe acute respiratory syndrome coronavirus 2, is frequently associated with neurological manifestations. Despite the presence of mild to severe CNS-related symptoms in a cohort of patients, there is no consensus whether the virus can infect directly brain tissue or if the symptoms in patients are a consequence of peripheral infectivity of the virus. Here, we use long-term human stem cell-derived cortical organoids to assess SARS-CoV2 infectivity of brain cells and unravel the cell-type tropism and its downstream pathological effects. Our results show consistent and reproducible low levels of SARS-CoV2 infection of astrocytes, deep projection neurons, upper callosal neurons, and inhibitory neurons in 6 months of human cortical organoids. Interestingly, astrocytes showed the highest infection rate among all infected cell populations which led to changes in their morphology and upregulation of SERPINA3, CD44, and S100A10 astrogliosis markers. Further, transcriptomic analysis revealed overall changes in expression of genes related to cell metabolism, astrogliosis and, inflammation and further, upregulation of cell survival pathways. Thus, local and minor infectivity of SARS-CoV2 in the brain may induce widespread adverse effects and lead to the resilience of dysregulated neurons and astrocytes within an inflammatory environment.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12121356/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143655708","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MSC-derived exosomal miR-125b-5p suppressed retinal microvascular endothelial cell ferroptosis in diabetic retinopathy. msc来源的外泌体miR-125b-5p抑制糖尿病视网膜病变视网膜微血管内皮细胞铁下垂。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf023
Jun Tong, Yueqin Chen, Xinru Ling, Zhenping Huang, Genhong Yao, Zhenggao Xie

Progressive endothelial cell injury of retinal vascular is a vital factor in diabetic retinopathy (DR) pathogenesis. Mesenchymal stem cells-derived small extracellular vesicles (MSC-sEVs) showed beneficial effects on DR. However, the effects of MSC-sEVs on endothelial dysfunction of DR and the mechanism is still unclear. In this study, MSC-sEVs mitigated retinal blood-retina barrier (BRB) impairment in rats with streptozotocin (STZ)-induced DR by reducing ferroptosis in vivo and in vitro. MSC-sEVs miRNA sequencing analysis revealed that miR-125b-5p may mediate human retina microvascular endothelial cells (HRMECs) ferroptosis and P53 as a downstream target based on dual-luciferase reporter assays. Silencing miR-125b-5p in MSC-sEVs reversed the therapeutic effects of MSC-sEVs on rats with DR and advanced glycation end products (AGEs)-treated HRMECs. Additionally, overexpression of miR-125b-5p could diminish ferroptosis in HRMECs, and this effect could be effectively reversed by overexpressing P53. This study indicated the potential therapeutic effect of MSC-sEVs on vascular endothelial function maintenance and that the delivery of sEVs carrying miR-125b-5p could prevent endothelial cell ferroptosis by inhibiting P53, thereby protecting the BRB.

进行性视网膜血管内皮细胞损伤是糖尿病视网膜病变发病的重要因素。间充质干细胞衍生的小细胞外囊泡(MSC-sEVs)对DR有有益作用,但MSC-sEVs在DR内皮功能障碍中的作用及其机制尚不清楚。在本研究中,msc - sev通过减少体内和体外的铁上吊,减轻了链脲佐菌素(STZ)诱导的DR大鼠视网膜血视网膜屏障(BRB)损伤。MSC-sEVs miRNA测序分析显示,基于双荧光素酶报告基因检测,miR-125b-5p可能介导人视网膜微血管内皮细胞(HRMECs)铁下垂和P53作为下游靶点。在msc - sev中沉默miR-125b-5p逆转了msc - sev对DR和晚期糖基化终产物(AGEs)治疗的hrmec大鼠的治疗作用。此外,过表达miR-125b-5p可以减少HRMECs中的铁下垂,并且这种作用可以通过过表达P53有效逆转。本研究表明msc - sev对血管内皮功能维持的潜在治疗作用,并且携带miR-125b-5p的sev递送可以通过抑制P53来阻止内皮细胞铁下沉,从而保护BRB。
{"title":"MSC-derived exosomal miR-125b-5p suppressed retinal microvascular endothelial cell ferroptosis in diabetic retinopathy.","authors":"Jun Tong, Yueqin Chen, Xinru Ling, Zhenping Huang, Genhong Yao, Zhenggao Xie","doi":"10.1093/stmcls/sxaf023","DOIUrl":"10.1093/stmcls/sxaf023","url":null,"abstract":"<p><p>Progressive endothelial cell injury of retinal vascular is a vital factor in diabetic retinopathy (DR) pathogenesis. Mesenchymal stem cells-derived small extracellular vesicles (MSC-sEVs) showed beneficial effects on DR. However, the effects of MSC-sEVs on endothelial dysfunction of DR and the mechanism is still unclear. In this study, MSC-sEVs mitigated retinal blood-retina barrier (BRB) impairment in rats with streptozotocin (STZ)-induced DR by reducing ferroptosis in vivo and in vitro. MSC-sEVs miRNA sequencing analysis revealed that miR-125b-5p may mediate human retina microvascular endothelial cells (HRMECs) ferroptosis and P53 as a downstream target based on dual-luciferase reporter assays. Silencing miR-125b-5p in MSC-sEVs reversed the therapeutic effects of MSC-sEVs on rats with DR and advanced glycation end products (AGEs)-treated HRMECs. Additionally, overexpression of miR-125b-5p could diminish ferroptosis in HRMECs, and this effect could be effectively reversed by overexpressing P53. This study indicated the potential therapeutic effect of MSC-sEVs on vascular endothelial function maintenance and that the delivery of sEVs carrying miR-125b-5p could prevent endothelial cell ferroptosis by inhibiting P53, thereby protecting the BRB.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143956900","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unlocking the potential of regionally activated injury/ischemia-induced stem cells for neural regeneration. 释放区域激活损伤/缺血诱导干细胞用于神经再生的潜力。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf015
Takayuki Nakagomi

In the past, the mammal central nervous system (CNS) was assumed to lack the capacity for neural repair. However, increasing evidence shows that the CNS has repair capacity after injury. The migratory capacity of neural stem/progenitor cells (NSPCs) from subventricular zones (SVZ) is limited, and the precise repair mechanism active after ischemic stroke remains unknown. Consequently, it remains unclear how neural regeneration occurs in regions far from the SVZ, such as the cortex, especially given that these NSPCs can only migrate toward ischemic areas within specific brain regions. Nonetheless, using a mouse model of ischemic stroke with ischemic areas limited to the ipsilateral side of the cortex, we previously identified regionally-derived stem cells, injury/ischemia-induced stem cells (iSCs), within poststroke areas. Moreover, we showed that iSCs, which had the potential to differentiate into electrophysiologically functional neurons, were present within ischemic areas in poststroke human brains. This indicates that ischemic insult can activate locally-derived stem cells, even in nonneurogenic zones, and that iSCs can help achieve neural regeneration after ischemic stroke. However, inflammatory cells typically fill ischemic areas impairing neural regeneration in these areas. Here, we present the origin, characterization, and roles of iSCs based on our recent research. In addition, we discussed the potential of iSC-based therapies to achieve neural regeneration after ischemic stroke.

过去,人们认为哺乳动物中枢神经系统(CNS)缺乏神经修复能力。然而,越来越多的证据表明,中枢神经系统在损伤后具有修复能力。神经干/祖细胞(NSPCs)从脑室下区(SVZ)的迁移能力有限,缺血性卒中后激活的精确修复机制尚不清楚。因此,神经再生如何发生在远离SVZ的区域,如皮层,仍不清楚,特别是考虑到这些NSPCs只能迁移到特定大脑区域内的缺血区域。尽管如此,我们之前在脑卒中后区域发现了区域性来源的干细胞,即损伤/缺血诱导干细胞(iSCs)。此外,我们还发现,在中风后人类大脑的缺血区域中存在有分化为具有电生理功能的神经元的iSCs。这表明缺血损伤可以激活局部来源的干细胞,甚至在非神经源性区,iSCs可以帮助实现缺血性中风后的神经再生。然而,炎症细胞通常会填充缺血区域,损害这些区域的神经再生。在这里,我们根据我们最近的研究介绍了iSCs的起源、特征和作用。此外,我们还讨论了缺血性脑卒中后基于干细胞的神经再生疗法的潜力。
{"title":"Unlocking the potential of regionally activated injury/ischemia-induced stem cells for neural regeneration.","authors":"Takayuki Nakagomi","doi":"10.1093/stmcls/sxaf015","DOIUrl":"10.1093/stmcls/sxaf015","url":null,"abstract":"<p><p>In the past, the mammal central nervous system (CNS) was assumed to lack the capacity for neural repair. However, increasing evidence shows that the CNS has repair capacity after injury. The migratory capacity of neural stem/progenitor cells (NSPCs) from subventricular zones (SVZ) is limited, and the precise repair mechanism active after ischemic stroke remains unknown. Consequently, it remains unclear how neural regeneration occurs in regions far from the SVZ, such as the cortex, especially given that these NSPCs can only migrate toward ischemic areas within specific brain regions. Nonetheless, using a mouse model of ischemic stroke with ischemic areas limited to the ipsilateral side of the cortex, we previously identified regionally-derived stem cells, injury/ischemia-induced stem cells (iSCs), within poststroke areas. Moreover, we showed that iSCs, which had the potential to differentiate into electrophysiologically functional neurons, were present within ischemic areas in poststroke human brains. This indicates that ischemic insult can activate locally-derived stem cells, even in nonneurogenic zones, and that iSCs can help achieve neural regeneration after ischemic stroke. However, inflammatory cells typically fill ischemic areas impairing neural regeneration in these areas. Here, we present the origin, characterization, and roles of iSCs based on our recent research. In addition, we discussed the potential of iSC-based therapies to achieve neural regeneration after ischemic stroke.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143762665","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CD44: a key regulator of iron metabolism, redox balance, and therapeutic resistance in cancer stem cells. CD44:肿瘤干细胞铁代谢、氧化还原平衡和治疗抵抗的关键调节因子
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf024
Taiju Ando, Juntaro Yamasaki, Hideyuki Saya, Osamu Nagano

CD44, a multifunctional cell surface protein, has emerged as a pivotal regulator in cancer stem cell (CSC) biology, orchestrating processes such as stemness, metabolic reprogramming, and therapeutic resistance. Recent studies have identified a critical role of CD44 in ferroptosis resistance by stabilizing SLC7A11 (xCT), a key component of the antioxidant defense system, enabling CSCs to evade oxidative stress and sustain tumorigenic potential. Additionally, CD44 regulates intracellular iron metabolism and redox balance, further supporting CSC survival and adaptation to stressful microenvironments. Therapeutic strategies targeting CD44, including ferroptosis inducers and combination therapies, have shown significant potential in preclinical and early clinical settings. Innovations such as CD44-mediated nanocarriers and metabolic inhibitors present novel opportunities to disrupt CSC-associated resistance mechanisms. Furthermore, the dynamic plasticity of CD44 isoforms governed by transcriptional, post-transcriptional, and epigenetic regulation underscores the importance of context-specific therapeutic approaches. This review highlights the multifaceted roles of CD44 in CSC biology, focusing on its contribution to ferroptosis resistance, iron metabolism, and redox regulation. Targeting CD44 offers a promising avenue for overcoming therapeutic resistance and improving the outcomes of refractory cancers. Future studies are needed to refine these strategies and enable their clinical translation.

CD44是一种多功能细胞表面蛋白,已成为癌症干细胞(CSC)生物学中的关键调节因子,协调诸如干性、代谢重编程和治疗抗性等过程。最近的研究发现,CD44通过稳定SLC7A11 (xCT)在铁中毒抗性中发挥关键作用,SLC7A11是抗氧化防御系统的关键成分,使CSCs逃避氧化应激并维持致瘤潜能。此外,CD44调节细胞内铁代谢和氧化还原平衡,进一步支持CSC存活和适应应激微环境。针对CD44的治疗策略,包括铁下垂诱导剂和联合治疗,在临床前和早期临床环境中显示出巨大的潜力。cd44介导的纳米载体和代谢抑制剂等创新为破坏csc相关的耐药机制提供了新的机会。此外,受转录、转录后和表观遗传调控控制的CD44亚型的动态可塑性强调了环境特异性治疗方法的重要性。这篇综述强调了CD44在CSC生物学中的多方面作用,重点关注其对铁下沉抵抗、铁代谢和氧化还原调节的贡献。靶向CD44为克服治疗耐药和改善难治性癌症的预后提供了一条有希望的途径。未来的研究需要完善这些策略并使其临床转化。
{"title":"CD44: a key regulator of iron metabolism, redox balance, and therapeutic resistance in cancer stem cells.","authors":"Taiju Ando, Juntaro Yamasaki, Hideyuki Saya, Osamu Nagano","doi":"10.1093/stmcls/sxaf024","DOIUrl":"10.1093/stmcls/sxaf024","url":null,"abstract":"<p><p>CD44, a multifunctional cell surface protein, has emerged as a pivotal regulator in cancer stem cell (CSC) biology, orchestrating processes such as stemness, metabolic reprogramming, and therapeutic resistance. Recent studies have identified a critical role of CD44 in ferroptosis resistance by stabilizing SLC7A11 (xCT), a key component of the antioxidant defense system, enabling CSCs to evade oxidative stress and sustain tumorigenic potential. Additionally, CD44 regulates intracellular iron metabolism and redox balance, further supporting CSC survival and adaptation to stressful microenvironments. Therapeutic strategies targeting CD44, including ferroptosis inducers and combination therapies, have shown significant potential in preclinical and early clinical settings. Innovations such as CD44-mediated nanocarriers and metabolic inhibitors present novel opportunities to disrupt CSC-associated resistance mechanisms. Furthermore, the dynamic plasticity of CD44 isoforms governed by transcriptional, post-transcriptional, and epigenetic regulation underscores the importance of context-specific therapeutic approaches. This review highlights the multifaceted roles of CD44 in CSC biology, focusing on its contribution to ferroptosis resistance, iron metabolism, and redox regulation. Targeting CD44 offers a promising avenue for overcoming therapeutic resistance and improving the outcomes of refractory cancers. Future studies are needed to refine these strategies and enable their clinical translation.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12126136/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143954716","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cellular therapies for the prevention and treatment of acute graft-versus-host disease. 细胞疗法预防和治疗急性移植物抗宿主病。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf009
Daniel Peltier, Van Anh Do-Thi, Timothy Devos, Bruce R Blazar, Tomomi Toubai

Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT) that is caused by donor immune cells attacking and damaging host tissues. Immune suppressive small molecule and protein-based therapeutics targeting donor anti-host immune cells are currently used for GVHD prophylaxis and treatment. Even with these therapies, aGVHD progresses to life-threatening steroid-refractory aGVHD (SR-aGVHD) in up to 50% of cases and is a risk factor for the subsequent development of debilitating chronic GVHD. To improve aGVHD-related outcomes, donor graft engineering techniques and adoptive transfer of immune modulatory cells have been explored. Highly rigorous donor graft T-cell depletion approaches have revealed that mitigation of aGVHD can be accompanied by slow immune recovery post-allo-HCT and reduction in anti-microbial and anti-leukemia responses resulting in increased relapse and infection rates, respectively. Recent T-cell separation techniques allowing for precision graft engineering by selectively eliminating aGVHD-causing T-cells (eg, naïve T-cells) without loss of T-cells with beneficial functions and retaining and/or enriching immune regulatory populations (eg, regulatory T-cells (Tregs) or myeloid-derived suppressor cells) have been tested and will continue to improve. Clinical cell-based regulatory therapies have been employed for targeting SR-aGVHD, particularly mesenchymal stem cells (MSCs) and more recently, Tregs. In this review, we summarize aGVHD pathophysiology, highlight newly discovered aGVHD mechanisms, and discuss current and emerging cellular and graft manipulation approaches for aGVHD prevention and treatment.

急性移植物抗宿主病(aGVHD)是同种异体造血细胞移植(allo-HCT)的主要并发症,由供体免疫细胞攻击和破坏宿主组织引起。针对供体抗宿主免疫细胞的免疫抑制小分子和基于蛋白质的疗法目前用于GVHD的预防和治疗。即使采用这些治疗方法,高达50%的aGVHD病例仍会发展为危及生命的类固醇难治性aGVHD (SR-aGVHD),并且是随后发展为使人衰弱的慢性GVHD的危险因素。为了改善与agvhd相关的结果,研究人员探索了供体移植物工程技术和免疫调节细胞的过继转移。高度严格的供体移植物t细胞耗损方法表明,aGVHD的缓解可能伴随着同种异体造血干细胞移植后缓慢的免疫恢复,以及抗微生物和抗白血病反应的降低,分别导致复发率和感染率的增加。最近的t细胞分离技术允许通过选择性地消除引起agvhd的t细胞(例如naïve t细胞)而不损失具有有益功能的t细胞并保留和/或丰富免疫调节群体(例如调节性t细胞(Tregs)或髓源性抑制细胞)来进行精确移植物工程,已经经过测试并将继续改进。临床基于细胞的调节疗法已被用于靶向SR-aGVHD,特别是间充质干细胞(MSCs)和最近的Tregs。在这篇综述中,我们总结了aGVHD的病理生理,重点介绍了新发现的aGVHD机制,并讨论了目前和新兴的aGVHD预防和治疗的细胞和移植物操作方法。
{"title":"Cellular therapies for the prevention and treatment of acute graft-versus-host disease.","authors":"Daniel Peltier, Van Anh Do-Thi, Timothy Devos, Bruce R Blazar, Tomomi Toubai","doi":"10.1093/stmcls/sxaf009","DOIUrl":"10.1093/stmcls/sxaf009","url":null,"abstract":"<p><p>Acute graft-versus-host disease (aGVHD) is a major complication of allogeneic hematopoietic cell transplantation (allo-HCT) that is caused by donor immune cells attacking and damaging host tissues. Immune suppressive small molecule and protein-based therapeutics targeting donor anti-host immune cells are currently used for GVHD prophylaxis and treatment. Even with these therapies, aGVHD progresses to life-threatening steroid-refractory aGVHD (SR-aGVHD) in up to 50% of cases and is a risk factor for the subsequent development of debilitating chronic GVHD. To improve aGVHD-related outcomes, donor graft engineering techniques and adoptive transfer of immune modulatory cells have been explored. Highly rigorous donor graft T-cell depletion approaches have revealed that mitigation of aGVHD can be accompanied by slow immune recovery post-allo-HCT and reduction in anti-microbial and anti-leukemia responses resulting in increased relapse and infection rates, respectively. Recent T-cell separation techniques allowing for precision graft engineering by selectively eliminating aGVHD-causing T-cells (eg, naïve T-cells) without loss of T-cells with beneficial functions and retaining and/or enriching immune regulatory populations (eg, regulatory T-cells (Tregs) or myeloid-derived suppressor cells) have been tested and will continue to improve. Clinical cell-based regulatory therapies have been employed for targeting SR-aGVHD, particularly mesenchymal stem cells (MSCs) and more recently, Tregs. In this review, we summarize aGVHD pathophysiology, highlight newly discovered aGVHD mechanisms, and discuss current and emerging cellular and graft manipulation approaches for aGVHD prevention and treatment.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12111709/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143672869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ZIC1 transcription factor overexpression in segmental bone defects is associated with brown adipogenic and osteogenic differentiation. ZIC1转录因子在节段性骨缺损中的过表达与棕色脂肪形成和成骨分化有关。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf013
Neelima Thottappillil, Zhao Li, Xin Xing, Shreya Arondekar, Manyu Zhu, Masnsen Cherief, Qizhi Qin, Myles Zhou, Mary Archer, Kristen Broderick, Bruno Pèault, Min Lee, Aaron W James

Transcriptional factor regulation is central to the lineage commitment of stem/ progenitor cells. ZIC1 (ZIC family member 1) is a C2H2-type zinc finger transcription factor expressed during development, brown fat, and certain cancers. Previously, we observed that overexpression of ZIC1 induces osteogenic differentiation at the expense of white adipogenic differentiation. In the present study, the feasibility of ZIC1 overexpressed human progenitor cells in critical-sized bone defects was studied. To achieve this, human adipose stem/stromal cells with other without lentiviral ZIC1 overexpression were implanted in a femoral segmental defect model in NOD-SCIDγ mice. Results showed that ZIC1 overexpressed cells induced osteogenic differentiation by protein markers in a critical-sized femoral segment defect compared to empty lentiviral control, although bone union was not observed. The immunohistochemical evaluation showed that implantation of ZIC1 overexpression cells led to an increase in osteoblast antigen expression (RUNX2, OCN), activation of Hedgehog signaling (Patched1), and an increase in brown adipogenesis markers (ZIC1, EBF2). In contrast, no change in bone defect-associated vasculature was observed (CD31, Endomucin). Together, these data suggest that overexpression of the ZIC1 transcription factor in progenitor cells is associated with differentiation towards osteoblastic and brown adipogenic cell fates.

转录因子调控是干细胞/祖细胞谱系承诺的核心。ZIC1 (ZIC家族成员1)是一种c2h2型锌指转录因子,在发育、棕色脂肪和某些癌症中表达。之前,我们观察到ZIC1的过表达诱导成骨分化,以牺牲白色脂肪分化为代价。在本研究中,我们研究了ZIC1过表达人祖细胞在临界大小骨缺损中的可行性。为了实现这一目标,将其他没有慢病毒ZIC1过表达的人脂肪干细胞/基质细胞植入nod - scid γ小鼠股骨节段缺损模型。结果显示,与空慢病毒对照相比,ZIC1过表达细胞通过蛋白标记诱导了临界大小股骨节段缺损的成骨分化,尽管未观察到骨愈合。免疫组化评价显示,植入ZIC1过表达细胞导致成骨细胞抗原表达(RUNX2, OCN)增加,Hedgehog信号通路(Patched1)激活,棕色脂肪形成标志物(ZIC1, EBF2)增加。相比之下,未观察到骨缺损相关脉管系统的变化(CD31, Endomucin)。综上所述,这些数据表明祖细胞中ZIC1转录因子的过表达与成骨细胞和棕色脂肪细胞的分化有关。
{"title":"ZIC1 transcription factor overexpression in segmental bone defects is associated with brown adipogenic and osteogenic differentiation.","authors":"Neelima Thottappillil, Zhao Li, Xin Xing, Shreya Arondekar, Manyu Zhu, Masnsen Cherief, Qizhi Qin, Myles Zhou, Mary Archer, Kristen Broderick, Bruno Pèault, Min Lee, Aaron W James","doi":"10.1093/stmcls/sxaf013","DOIUrl":"10.1093/stmcls/sxaf013","url":null,"abstract":"<p><p>Transcriptional factor regulation is central to the lineage commitment of stem/ progenitor cells. ZIC1 (ZIC family member 1) is a C2H2-type zinc finger transcription factor expressed during development, brown fat, and certain cancers. Previously, we observed that overexpression of ZIC1 induces osteogenic differentiation at the expense of white adipogenic differentiation. In the present study, the feasibility of ZIC1 overexpressed human progenitor cells in critical-sized bone defects was studied. To achieve this, human adipose stem/stromal cells with other without lentiviral ZIC1 overexpression were implanted in a femoral segmental defect model in NOD-SCIDγ mice. Results showed that ZIC1 overexpressed cells induced osteogenic differentiation by protein markers in a critical-sized femoral segment defect compared to empty lentiviral control, although bone union was not observed. The immunohistochemical evaluation showed that implantation of ZIC1 overexpression cells led to an increase in osteoblast antigen expression (RUNX2, OCN), activation of Hedgehog signaling (Patched1), and an increase in brown adipogenesis markers (ZIC1, EBF2). In contrast, no change in bone defect-associated vasculature was observed (CD31, Endomucin). Together, these data suggest that overexpression of the ZIC1 transcription factor in progenitor cells is associated with differentiation towards osteoblastic and brown adipogenic cell fates.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143727129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inflammatory pathways and the bone marrow microenvironment in inherited bone marrow failure syndromes. 炎症途径和骨髓微环境在遗传性骨髓衰竭综合征。
IF 4 2区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-05-27 DOI: 10.1093/stmcls/sxaf021
Nicholas Neoman, Hye Na Kim, Jacob Viduya, Anju Goyal, Y Lucy Liu, Kathleen M Sakamoto

Inherited bone marrow failure syndromes (IBMFS) are a diverse group of genetic disorders characterized by insufficient hematopoietic cell production due to blood stem cell dysfunction. The most common syndromes are Fanconi Anemia, Diamond-Blackfan Anemia, and Shwachman-Diamond Syndrome. These conditions share a theme of chronically producing pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, TGF-β, IFN-I, and IFN-γ. Each of these cytokines can impact the bone marrow microenvironment and drive the pathophysiology of IBMFS. This review aims to provide the latest progress in the field regarding the mechanistic underpinnings of inflammation in these IBMFS, as well as the effect of inflammation on the bone marrow microenvironment. A comprehensive understanding of the inflammation in IBMFS will open new avenues for intervention to restore bone marrow stability and improve patient prognosis. Future research must include targeting these mechanisms to develop novel therapies that can potentially mitigate the effects of chronic inflammation in IBMFS.

遗传性骨髓衰竭综合征(IBMFS)是一组多样化的遗传性疾病,其特征是由于造血干细胞功能障碍导致造血细胞产生不足。最常见的综合征是范可尼贫血症、Diamond-Blackfan贫血症和Shwachman-Diamond综合征。这些疾病都有一个共同的主题,即慢性产生促炎细胞因子,如TNF-α、IL-1β、IL-6、TGF-β、IFN- i和IFN-γ。这些细胞因子都可以影响骨髓微环境并驱动IBMFS的病理生理。本文旨在就炎症在IBMFS中的机制基础以及炎症对骨髓微环境的影响等方面的最新进展进行综述。对IBMFS炎症的全面了解将为恢复骨髓稳定性和改善患者预后开辟新的干预途径。未来的研究必须包括针对这些机制来开发可能减轻IBMFS慢性炎症影响的新疗法。
{"title":"Inflammatory pathways and the bone marrow microenvironment in inherited bone marrow failure syndromes.","authors":"Nicholas Neoman, Hye Na Kim, Jacob Viduya, Anju Goyal, Y Lucy Liu, Kathleen M Sakamoto","doi":"10.1093/stmcls/sxaf021","DOIUrl":"10.1093/stmcls/sxaf021","url":null,"abstract":"<p><p>Inherited bone marrow failure syndromes (IBMFS) are a diverse group of genetic disorders characterized by insufficient hematopoietic cell production due to blood stem cell dysfunction. The most common syndromes are Fanconi Anemia, Diamond-Blackfan Anemia, and Shwachman-Diamond Syndrome. These conditions share a theme of chronically producing pro-inflammatory cytokines such as TNF-α, IL-1β, IL-6, TGF-β, IFN-I, and IFN-γ. Each of these cytokines can impact the bone marrow microenvironment and drive the pathophysiology of IBMFS. This review aims to provide the latest progress in the field regarding the mechanistic underpinnings of inflammation in these IBMFS, as well as the effect of inflammation on the bone marrow microenvironment. A comprehensive understanding of the inflammation in IBMFS will open new avenues for intervention to restore bone marrow stability and improve patient prognosis. Future research must include targeting these mechanisms to develop novel therapies that can potentially mitigate the effects of chronic inflammation in IBMFS.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2025-05-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12121359/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143952420","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
STEM CELLS
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:604180095
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