首页 > 最新文献

Stem Cell Research & Therapy最新文献

英文 中文
Zebrafish radial glia orchestrate vascular regeneration: implications for bionic therapy of spinal cord injury. 斑马鱼放射状胶质管束血管再生:脊髓损伤仿生治疗的意义。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-09 DOI: 10.1186/s13287-026-04898-6
Xiaohui Wang, Yuyang Zhang, Shengnan Zhao, Hua Hui, Baorong He, Chao Jiang, Didier Y R Stainier, Hao Yang

Background: Bionic treatment is a strategy designed to facilitate functional recovery after clinical spinal cord injury (SCI) by emulating the natural morphological structure and regeneration process. We used zebrafish model, an animal with remarkable regenerative capabilities to investigate the regulatory pattern of spinal vascular regeneration following SCI, with the hope of providing inspirations for the development of bionic SCI treatment.

Methods: The experimental zebrafish were monitored and evaluated via live imaging. We first determined the formation time of the spinal perineural vessel plexus (PNVP) and used this as the timepoint to initiate SCI. Subsequently, a SCI model was established to observe the pattern of vascular repair without intervention; Furthermore, radial glial (RGs) of Tg(gfap: NTR-mCherry) report line fish were chemically ablated using metronidazole (Mtz) or nitrofuropyrinol (Nfp). We assessed the patterns of vascular repair, the vascular coverage of the injured area, and the number of vascular endothelial cells (ECs). Concomitantly, by analyzing the expression profile of vascular endothelial growth factor aa (Vegfaa) in the injured region following RGs ablation, and leveraging a public available single-cell sequencing dataset, we postulated the potential downstream pathways involved. The functional relevance of these pathways was finally evaluated by applying specific inhibitors.

Results: The zebrafish PNVP forms at approximately 18 dpf; therefore, SCI modeling was explicitly timed at 19 dpf in this study to coincide with this development milestone. In the Tg(gfap: NTR-mCherry) report line, RGs were successfully ablated using either Mtz or Nfp. Following ablation, both vascular coverage in the injured area and the number of ECs were significantly reduced in the Mtz/Nfp + SCI group compared to the DMSO + SCI group. Moreover, The vegfaa reporter line revealed a notable decline in vegfaa signal within the injured region post-ablation, suggesting its involvement in the repair process. This implication was further supported by inhibitor experiments, where intervention against the Notch and PI3K/Akt-mTOR pathways significantly altered the extend of vascular repair, indicating a potential correlation between these pathways and RGs-regulated vascular repair.

Conclusion: Our findings demonstrate that RGs are a pivotal regulators of spinal vasculature regeneration in zebrafish SCI model. The underlying mechanisms may involve the Vegfa-PI3K/Akt-mTOR and Notch signaling pathways. Therefore, it can be postulated that pro-vascular repair therapy in mammals following SCI could potentially be achieved by therapeutically mimicking pro-regenerative functions of RGs.

背景:仿生治疗是通过模拟自然形态结构和再生过程来促进临床脊髓损伤(SCI)后功能恢复的一种策略。我们利用具有显著再生能力的动物斑马鱼模型,研究脊髓损伤后脊髓血管再生的调控模式,以期为仿生脊髓损伤治疗的发展提供启示。方法:采用活体成像技术对实验斑马鱼进行监测和评价。我们首先确定了脊髓周围神经血管丛(PNVP)的形成时间,并以此作为起始脊髓损伤的时间点。随后,建立脊髓损伤模型,观察无干预的血管修复模式;此外,用甲硝唑(Mtz)或硝基呋喃吡啶醇(Nfp)化学消融Tg(gfap: NTR-mCherry)报告线鱼的径向胶质(RGs)。我们评估了血管修复的模式、损伤区域的血管覆盖范围和血管内皮细胞(ECs)的数量。同时,通过分析RGs消融后损伤区域血管内皮生长因子aa (Vegfaa)的表达谱,并利用公开的单细胞测序数据,我们假设了潜在的下游通路。这些途径的功能相关性最终通过应用特异性抑制剂进行评估。结果:斑马鱼PNVP形成于约18 dpf;因此,本研究明确将SCI建模时间定在19 dpf,以配合这一发展里程碑。在Tg(gfap: NTR-mCherry)报告线中,使用Mtz或Nfp均可成功消融RGs。消融后,与DMSO + SCI组相比,Mtz/Nfp + SCI组损伤区域的血管覆盖率和ECs数量均显著减少。此外,vegfaa报告线显示消融后损伤区域的vegfaa信号显著下降,提示其参与修复过程。抑制剂实验进一步支持了这一结论,对Notch和PI3K/Akt-mTOR通路的干预显著改变了血管修复的延伸,表明这些通路与rgs调节的血管修复之间存在潜在的相关性。结论:我们的研究结果表明RGs是斑马鱼脊髓损伤模型中脊髓血管再生的关键调节因子。潜在的机制可能涉及Vegfa-PI3K/Akt-mTOR和Notch信号通路。因此,可以假设,通过模拟RGs的促再生功能,可以实现对脊髓损伤后哺乳动物的促血管修复治疗。
{"title":"Zebrafish radial glia orchestrate vascular regeneration: implications for bionic therapy of spinal cord injury.","authors":"Xiaohui Wang, Yuyang Zhang, Shengnan Zhao, Hua Hui, Baorong He, Chao Jiang, Didier Y R Stainier, Hao Yang","doi":"10.1186/s13287-026-04898-6","DOIUrl":"https://doi.org/10.1186/s13287-026-04898-6","url":null,"abstract":"<p><strong>Background: </strong>Bionic treatment is a strategy designed to facilitate functional recovery after clinical spinal cord injury (SCI) by emulating the natural morphological structure and regeneration process. We used zebrafish model, an animal with remarkable regenerative capabilities to investigate the regulatory pattern of spinal vascular regeneration following SCI, with the hope of providing inspirations for the development of bionic SCI treatment.</p><p><strong>Methods: </strong>The experimental zebrafish were monitored and evaluated via live imaging. We first determined the formation time of the spinal perineural vessel plexus (PNVP) and used this as the timepoint to initiate SCI. Subsequently, a SCI model was established to observe the pattern of vascular repair without intervention; Furthermore, radial glial (RGs) of Tg(gfap: NTR-mCherry) report line fish were chemically ablated using metronidazole (Mtz) or nitrofuropyrinol (Nfp). We assessed the patterns of vascular repair, the vascular coverage of the injured area, and the number of vascular endothelial cells (ECs). Concomitantly, by analyzing the expression profile of vascular endothelial growth factor aa (Vegfaa) in the injured region following RGs ablation, and leveraging a public available single-cell sequencing dataset, we postulated the potential downstream pathways involved. The functional relevance of these pathways was finally evaluated by applying specific inhibitors.</p><p><strong>Results: </strong>The zebrafish PNVP forms at approximately 18 dpf; therefore, SCI modeling was explicitly timed at 19 dpf in this study to coincide with this development milestone. In the Tg(gfap: NTR-mCherry) report line, RGs were successfully ablated using either Mtz or Nfp. Following ablation, both vascular coverage in the injured area and the number of ECs were significantly reduced in the Mtz/Nfp + SCI group compared to the DMSO + SCI group. Moreover, The vegfaa reporter line revealed a notable decline in vegfaa signal within the injured region post-ablation, suggesting its involvement in the repair process. This implication was further supported by inhibitor experiments, where intervention against the Notch and PI3K/Akt-mTOR pathways significantly altered the extend of vascular repair, indicating a potential correlation between these pathways and RGs-regulated vascular repair.</p><p><strong>Conclusion: </strong>Our findings demonstrate that RGs are a pivotal regulators of spinal vasculature regeneration in zebrafish SCI model. The underlying mechanisms may involve the Vegfa-PI3K/Akt-mTOR and Notch signaling pathways. Therefore, it can be postulated that pro-vascular repair therapy in mammals following SCI could potentially be achieved by therapeutically mimicking pro-regenerative functions of RGs.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145946107","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
Human pluripotent stem cell models of Friedreich's ataxia: innovations, considerations, and future perspectives. 弗里德里希共济失调的人类多能干细胞模型:创新,考虑和未来展望。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-09 DOI: 10.1186/s13287-025-04861-x
Ha Thi Nguyen, Marek Napierala, Jill S Napierala

Friedreich's ataxia (FRDA) is an inherited, autosomal recessive, multisystem disorder that primarily manifests in children and affects the nervous system and the heart. FRDA is caused by an expansion of GAA repeats in the first intron of the frataxin (FXN) gene. The expansion disrupts transcription of FXN, resulting in significantly decreased FXN expression in FRDA patients' tissues. Frataxin is involved in biosynthesis of iron-sulfur (Fe-S) clusters, which are critical for the function of the electron transport chain and many metabolic enzymes. Frataxin deficiency leads to reduced energy production and accumulation of iron in mitochondria that exacerbates oxidative stress. Despite significant advancements in the field, FXN cellular functions and underlying pathological mechanisms of FXN deficiency in cell-type specific contexts remain to be elucidated. Inaccessibility to the most vulnerable cell types in FRDA patients, including neurons, cardiomyocytes, and β-cells, largely accounts for these limitations. Significant progress in recent years regarding the derivation and differentiation of human pluripotent stem cells (hPSCs), along with breakthroughs in gene editing technologies, enables the generation of patient-derived and isogenic control disease-relevant cell types and organoid-like structures as platforms for studying disease mechanisms and for drug discovery. Herein, we first provide an overview of hPSC derivation and intrinsic properties of these cells. We then discuss current advances and limitations of hiPSC-based cell models for FRDA. We also highlight the need to further refine and develop these in vitro cell models for pre-clinical advancement of therapeutic approaches for FRDA.

弗里德赖希共济失调症(FRDA)是一种遗传性常染色体隐性多系统疾病,主要表现在儿童,影响神经系统和心脏。FRDA是由frataxin (FXN)基因的第一个内含子中GAA重复序列的扩增引起的。这种扩增破坏了FXN的转录,导致FRDA患者组织中FXN的表达显著降低。Frataxin参与铁硫(Fe-S)簇的生物合成,这对电子传递链和许多代谢酶的功能至关重要。Frataxin缺乏导致线粒体中能量产生和铁积累减少,从而加剧氧化应激。尽管在该领域取得了重大进展,但FXN的细胞功能和FXN缺乏在细胞类型特异性背景下的潜在病理机制仍有待阐明。FRDA患者中最脆弱的细胞类型(包括神经元、心肌细胞和β细胞)难以接近,这在很大程度上解释了这些局限性。近年来,随着基因编辑技术的突破,人类多能干细胞(hPSCs)的衍生和分化取得了重大进展,使患者衍生和等基因控制疾病相关细胞类型和类器官结构的产生成为研究疾病机制和药物发现的平台。在这里,我们首先概述了hPSC的衍生和这些细胞的内在特性。然后,我们讨论了目前基于hipsc的FRDA细胞模型的进展和局限性。我们还强调需要进一步完善和发展这些体外细胞模型,以促进FRDA治疗方法的临床前进展。
{"title":"Human pluripotent stem cell models of Friedreich's ataxia: innovations, considerations, and future perspectives.","authors":"Ha Thi Nguyen, Marek Napierala, Jill S Napierala","doi":"10.1186/s13287-025-04861-x","DOIUrl":"https://doi.org/10.1186/s13287-025-04861-x","url":null,"abstract":"<p><p>Friedreich's ataxia (FRDA) is an inherited, autosomal recessive, multisystem disorder that primarily manifests in children and affects the nervous system and the heart. FRDA is caused by an expansion of GAA repeats in the first intron of the frataxin (FXN) gene. The expansion disrupts transcription of FXN, resulting in significantly decreased FXN expression in FRDA patients' tissues. Frataxin is involved in biosynthesis of iron-sulfur (Fe-S) clusters, which are critical for the function of the electron transport chain and many metabolic enzymes. Frataxin deficiency leads to reduced energy production and accumulation of iron in mitochondria that exacerbates oxidative stress. Despite significant advancements in the field, FXN cellular functions and underlying pathological mechanisms of FXN deficiency in cell-type specific contexts remain to be elucidated. Inaccessibility to the most vulnerable cell types in FRDA patients, including neurons, cardiomyocytes, and β-cells, largely accounts for these limitations. Significant progress in recent years regarding the derivation and differentiation of human pluripotent stem cells (hPSCs), along with breakthroughs in gene editing technologies, enables the generation of patient-derived and isogenic control disease-relevant cell types and organoid-like structures as platforms for studying disease mechanisms and for drug discovery. Herein, we first provide an overview of hPSC derivation and intrinsic properties of these cells. We then discuss current advances and limitations of hiPSC-based cell models for FRDA. We also highlight the need to further refine and develop these in vitro cell models for pre-clinical advancement of therapeutic approaches for FRDA.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145945640","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
Molecular and functional characterization of GMP-manufactured neural stem cells and their extracellular vesicles for innovative therapeutic applications. gmp制造的神经干细胞及其用于创新治疗的细胞外囊泡的分子和功能表征。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-09 DOI: 10.1186/s13287-026-04904-x
Martina Guzzetti, Letizia Mezzasoma, Davide Chiasserini, Lara Macchioni, Magdalena Davidescu, Alessandro di Michele, Marco Gargaro, Nicola Di-Iacovo, Giorgia Manni, Gianmarco Muzi, Ilaria Proietti, Giuseppina Bevacqua, Eleonora Becattini, Carlo Conti, Vincenzo Nicola Talesa, Rita Romani, Ilaria Bellezza, Valentina Grespi

Human neural stem cells (hNSCs) are promising candidates for regenerative medicine due to their self-renewal capacity, differentiation potential, and ability to modulate inflammation. However, several reports showed that the regenerative properties of stem cells are tied to the extracellular vesicles (EVs) they secrete. This study aimed at characterizing hNSCs produced under Good Manufacturing Practice (GMP) conditions and at elucidating the molecular and functional properties of their secreted extracellular vesicles (hNSC-EVs). hNSCs were first assessed for proliferation, and differentiation potential, showing a stable growth profile and expression of neural stem cell markers. High-resolution proteomic analysis identified over 5000 proteins, with about 40% overlap with previous NSCs studies. hNSCs expressed mostly markers for different cell lineage precursors. The molecular characterization of hNSC-derived EVs (hNSC-EVs) showed a size distribution, as measured by nanoparticle tracking analysis, ranging from 140 to 200 nm and an enrichment in EV markers, detected by western blotting. Functional analyses showed that hNSC-EVs, reduce nitric oxide generation and inducible nitric oxide expression in LPS-treated microglial cells and inhibit caspase-1 activation in monocytic cell models through uptake-dependent and independent mechanism, respectively. Our findings show that hNSC possess a strong stemness signature and secrete EVs with immunomodulatory properties, suggesting the worth of hNSC-EVs as either alternative to cell-based therapies or primer to boost anti-inflammatory properties of hNSCs in the treatment of neurological disorders.

人类神经干细胞(hNSCs)由于其自我更新能力、分化潜力和调节炎症的能力而成为再生医学的有希望的候选者。然而,一些报道表明,干细胞的再生特性与它们分泌的细胞外囊泡(EVs)有关。本研究旨在表征在GMP条件下生产的hNSCs,并阐明其分泌的细胞外囊泡(hnsc - ev)的分子和功能特性。首先评估hNSCs的增殖和分化潜力,显示出稳定的生长特征和神经干细胞标记物的表达。高分辨率蛋白质组学分析鉴定了5000多种蛋白质,与以前的NSCs研究有大约40%的重叠。hNSCs主要表达不同细胞系前体的标记物。通过纳米颗粒跟踪分析,hnsc -EV的分子特征显示出一个大小分布,范围在140 - 200 nm之间,通过western blotting检测到EV标记物的富集。功能分析显示,在lps处理的小胶质细胞中,hnsc - ev分别通过摄取依赖机制和独立机制减少一氧化氮的产生,诱导一氧化氮的表达,抑制单核细胞模型中caspase-1的激活。我们的研究结果表明,hNSC具有很强的干性特征,并分泌具有免疫调节特性的ev,这表明hNSC- ev作为基于细胞的治疗方法的替代方案或引物在神经系统疾病治疗中增强hNSC的抗炎特性的价值。
{"title":"Molecular and functional characterization of GMP-manufactured neural stem cells and their extracellular vesicles for innovative therapeutic applications.","authors":"Martina Guzzetti, Letizia Mezzasoma, Davide Chiasserini, Lara Macchioni, Magdalena Davidescu, Alessandro di Michele, Marco Gargaro, Nicola Di-Iacovo, Giorgia Manni, Gianmarco Muzi, Ilaria Proietti, Giuseppina Bevacqua, Eleonora Becattini, Carlo Conti, Vincenzo Nicola Talesa, Rita Romani, Ilaria Bellezza, Valentina Grespi","doi":"10.1186/s13287-026-04904-x","DOIUrl":"https://doi.org/10.1186/s13287-026-04904-x","url":null,"abstract":"<p><p>Human neural stem cells (hNSCs) are promising candidates for regenerative medicine due to their self-renewal capacity, differentiation potential, and ability to modulate inflammation. However, several reports showed that the regenerative properties of stem cells are tied to the extracellular vesicles (EVs) they secrete. This study aimed at characterizing hNSCs produced under Good Manufacturing Practice (GMP) conditions and at elucidating the molecular and functional properties of their secreted extracellular vesicles (hNSC-EVs). hNSCs were first assessed for proliferation, and differentiation potential, showing a stable growth profile and expression of neural stem cell markers. High-resolution proteomic analysis identified over 5000 proteins, with about 40% overlap with previous NSCs studies. hNSCs expressed mostly markers for different cell lineage precursors. The molecular characterization of hNSC-derived EVs (hNSC-EVs) showed a size distribution, as measured by nanoparticle tracking analysis, ranging from 140 to 200 nm and an enrichment in EV markers, detected by western blotting. Functional analyses showed that hNSC-EVs, reduce nitric oxide generation and inducible nitric oxide expression in LPS-treated microglial cells and inhibit caspase-1 activation in monocytic cell models through uptake-dependent and independent mechanism, respectively. Our findings show that hNSC possess a strong stemness signature and secrete EVs with immunomodulatory properties, suggesting the worth of hNSC-EVs as either alternative to cell-based therapies or primer to boost anti-inflammatory properties of hNSCs in the treatment of neurological disorders.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145946097","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
HucMSC-derived exosomes alleviate chemotherapy-induced premature ovarian insufficiency via SMURF1-mediated inhibition of ferroptosis in ovarian granulosa cells. hucmsc衍生的外泌体通过smurf1介导的卵巢颗粒细胞铁下沉抑制减轻化疗诱导的卵巢早衰。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-08 DOI: 10.1186/s13287-026-04893-x
Bo Xu, Hang Han, Yao Hao, Yitong Shang, Yu Deng, Zhen Zhang, Liyang Ding, Xiuying Pei, Xufeng Fu

Background: Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSC-Exo) have shown great therapeutic promise in the treatment of primary ovarian insufficiency (POI). Ferroptosis, a distinct form of cell death, has been associated with the pathogenesis of POI. However, whether HucMSC-Exo can mitigate POI by modulating ferroptosis remains unknown.

Methods: In a CTX-induced POI mouse model, HucMSC-Exo was administered. Ovarian function was assessed by monitoring the estrous cycle, hormone levels, ovarian index, fertility rate, and ovarian morphology. The molecular mechanisms underlying injury and repair were investigated through HucMSC-Exo tracing, immunohistochemical staining, western blot, and real-time polymerase chain reaction (PCR).

Results: HucMSC-Exo restored hormonal balance, preserved ovarian reserve, and reduced follicular atresia and developmental defects in a cyclophosphamide (CTX)-induced POI mouse model. Furthermore, HucMSC-Exo attenuated Fe²⁺ accumulation, oxidative stress, and ferroptosis in the granulosa cells (GCs) of atretic follicles in ovaries with POI. In vitro assays also demonstrated that HucMSC-Exo attenuated CTX-induced ferroptosis in GCs by alleviating Fe²-dependent oxidative damage. Interestingly, hucMSC-Exo specifically suppressed the CTX-induced upregulation of heme oxygenase-1 (HO-1), a key regulator of iron homeostasis, at the translational level. This suggests that post-translational modifications may play a regulatory role in HO-1 expression and iron homeostasis. Mechanistic studies revealed that HucMSC-Exo delivers SMURF1, an E3 ubiquitin ligase that promotes HO-1 degradation, thereby restoring iron homeostasis and inhibiting ferroptosis in GCs. Furthermore, HO-1 knockdown enhanced the protective effects of HucMSC-Exo against CTX-induced ferroptosis and cytotoxicity in GCs.

Conclusions: HucMSC-Exo delivers SMURF1 to promote HO-1 degradation, which in turn suppresses Fe2+ accumulation and lipid peroxidation, thereby preventing ferroptosis in GCs and ameliorating chemotherapy-induced POI.

背景:人脐带间充质干细胞衍生外泌体(HucMSC-Exo)在治疗原发性卵巢功能不全(POI)方面显示出巨大的治疗前景。铁下垂是一种独特的细胞死亡形式,与POI的发病机制有关。然而,HucMSC-Exo是否可以通过调节铁下垂来减轻POI仍然未知。方法:在ctx诱导的POI小鼠模型中,给予HucMSC-Exo。通过监测发情周期、激素水平、卵巢指数、生育率和卵巢形态来评估卵巢功能。通过HucMSC-Exo示踪、免疫组织化学染色、western blot和实时聚合酶链反应(PCR)研究损伤和修复的分子机制。结果:在环磷酰胺(CTX)诱导的POI小鼠模型中,hucscs - exo恢复了激素平衡,保留了卵巢储备,减少了卵泡闭锁和发育缺陷。此外,HucMSC-Exo减轻了POI卵巢闭锁卵泡颗粒细胞(GCs)中Fe +的积累、氧化应激和铁下垂。体外实验还表明,HucMSC-Exo通过减轻Fe²依赖性氧化损伤来减轻ctx诱导的GCs铁下垂。有趣的是,hucMSC-Exo在翻译水平特异性抑制ctx诱导的血红素氧合酶-1 (HO-1)的上调,HO-1是铁稳态的关键调节因子。这表明翻译后修饰可能在HO-1表达和铁稳态中起调节作用。机制研究表明,HucMSC-Exo传递SMURF1,一种促进HO-1降解的E3泛素连接酶,从而恢复铁稳态并抑制GCs中的铁凋亡。此外,HO-1敲低增强了HucMSC-Exo对ctx诱导的GCs铁下垂和细胞毒性的保护作用。结论:HucMSC-Exo传递SMURF1促进HO-1降解,进而抑制Fe2+积累和脂质过氧化,从而防止GCs中的铁下沉,改善化疗诱导的POI。
{"title":"HucMSC-derived exosomes alleviate chemotherapy-induced premature ovarian insufficiency via SMURF1-mediated inhibition of ferroptosis in ovarian granulosa cells.","authors":"Bo Xu, Hang Han, Yao Hao, Yitong Shang, Yu Deng, Zhen Zhang, Liyang Ding, Xiuying Pei, Xufeng Fu","doi":"10.1186/s13287-026-04893-x","DOIUrl":"https://doi.org/10.1186/s13287-026-04893-x","url":null,"abstract":"<p><strong>Background: </strong>Human umbilical cord mesenchymal stem cell-derived exosomes (HucMSC-Exo) have shown great therapeutic promise in the treatment of primary ovarian insufficiency (POI). Ferroptosis, a distinct form of cell death, has been associated with the pathogenesis of POI. However, whether HucMSC-Exo can mitigate POI by modulating ferroptosis remains unknown.</p><p><strong>Methods: </strong>In a CTX-induced POI mouse model, HucMSC-Exo was administered. Ovarian function was assessed by monitoring the estrous cycle, hormone levels, ovarian index, fertility rate, and ovarian morphology. The molecular mechanisms underlying injury and repair were investigated through HucMSC-Exo tracing, immunohistochemical staining, western blot, and real-time polymerase chain reaction (PCR).</p><p><strong>Results: </strong>HucMSC-Exo restored hormonal balance, preserved ovarian reserve, and reduced follicular atresia and developmental defects in a cyclophosphamide (CTX)-induced POI mouse model. Furthermore, HucMSC-Exo attenuated Fe²⁺ accumulation, oxidative stress, and ferroptosis in the granulosa cells (GCs) of atretic follicles in ovaries with POI. In vitro assays also demonstrated that HucMSC-Exo attenuated CTX-induced ferroptosis in GCs by alleviating Fe²-dependent oxidative damage. Interestingly, hucMSC-Exo specifically suppressed the CTX-induced upregulation of heme oxygenase-1 (HO-1), a key regulator of iron homeostasis, at the translational level. This suggests that post-translational modifications may play a regulatory role in HO-1 expression and iron homeostasis. Mechanistic studies revealed that HucMSC-Exo delivers SMURF1, an E3 ubiquitin ligase that promotes HO-1 degradation, thereby restoring iron homeostasis and inhibiting ferroptosis in GCs. Furthermore, HO-1 knockdown enhanced the protective effects of HucMSC-Exo against CTX-induced ferroptosis and cytotoxicity in GCs.</p><p><strong>Conclusions: </strong>HucMSC-Exo delivers SMURF1 to promote HO-1 degradation, which in turn suppresses Fe<sup>2+</sup> accumulation and lipid peroxidation, thereby preventing ferroptosis in GCs and ameliorating chemotherapy-induced POI.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935000","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
Immunomodulatory effect of mesenchymal stromal cell overexpressing HLA-G1 in cell-based therapy for myocardial infarction. 过表达HLA-G1的间充质间质细胞在心肌梗死细胞治疗中的免疫调节作用。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-08 DOI: 10.1186/s13287-026-04897-7
Wei Zhu, Jie Kong, Hong-Xia Li, Ting-Bo Jiang, Si-Jia Sun, Cao Zou

Background: Our previous study revealed that intravenous administration of mesenchymal stromal cells (MSCs) increases local cell engraftment and improves heart function. This study aims to investigate whether MSCs overexpressing HLA-G1 have further increased local transplanted cells engraftment and improved heart function.

Methods: The mice were intravenously administered saline or human umbilical cord blood-derived MSCs (hUCB-MSCs) 7 days before myocardial infarction (MI) induction. Then, the MI mouse model was established by ligating the left anterior descending coronary artery. The mice were then subjected to intramyocardial transplantation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) 30 min after MI induction. Echocardiographic analysis was carried out to assess heart function. Furthermore, in vivo fluorescent imaging analysis was performed to analyze cell engraftment. Moreover, flow cytometry of splenic regulatory T cells (Tregs) and natural killer (NK) cells was conducted to evaluate the immunomodulatory effect of hUCB-MSCs.

Results: The results showed that systemic intravenous administration of hUCB-MSCs substantially enhanced Tregs, decreased NK cells, and increased intramyocardially transplanted hiPSC-CMs' engraftment, thus improving heart function. Compared with hUCB-MSCs, HLA-G1 overexpressing hUCB-MSCs reduced systemic NK cells (7.13 ± 0.19% vs. 9.12 ± 0.06%, p < 0.05), increased Tregs (5.03 ± 0.17% vs. 3.36 ± 0.05%, p < 0.05), improved cell engraftment (Radiant efficiency: 3.01 ± 0.36 × 109 vs. 2.19 ± 0.27 × 109, p < 0.05) and heart function (LVEF: 73.00 ± 0.44 vs. 62.36 ± 1.01, p < 0.05). The in vitro assays revealed that HLA-G1 overexpressing hUCB-MSCs modulated the immune response by decreasing pro-inflammatory cytokines.

Conclusions: This study showed that systemic intravenous administration of HLA-G1 overexpressing hUCB-MSCs modulated immune response and increased transplanted hiPSC-CMs' engraftment to improve heart function following AMI.

背景:我们之前的研究表明,静脉注射间充质间质细胞(MSCs)可增加局部细胞植入并改善心脏功能。本研究旨在探讨过表达HLA-G1的MSCs是否进一步增加了局部移植细胞的植入,改善了心脏功能。方法:小鼠在心肌梗死(MI)诱导前7天静脉注射生理盐水或人脐带血来源的间充质干细胞(hub -MSCs)。结扎左冠状动脉前降支建立心肌梗死小鼠模型。然后在心肌梗死诱导后30分钟将小鼠进行人诱导多能干细胞来源的心肌细胞(hiPSC-CMs)的心肌内移植。超声心动图分析评估心功能。此外,采用体内荧光成像分析来分析细胞植入。此外,通过脾调节性T细胞(Tregs)和自然杀伤细胞(NK)的流式细胞术来评估hub - mscs的免疫调节作用。结果:结果显示全身静脉给药hub - mscs可显著增强Tregs,降低NK细胞,增加心内移植hiPSC-CMs的植入,从而改善心功能。与hUCB-MSCs相比,HLA-G1过表达的hUCB-MSCs减少了全身NK细胞(7.13±0.19% vs. 9.12±0.06%,p 9 vs. 2.19±0.27 × 109, p)结论:本研究表明,全身静脉注射HLA-G1过表达的hUCB-MSCs可调节免疫反应,增加移植的hiPSC-CMs的植入,改善AMI后心功能。
{"title":"Immunomodulatory effect of mesenchymal stromal cell overexpressing HLA-G1 in cell-based therapy for myocardial infarction.","authors":"Wei Zhu, Jie Kong, Hong-Xia Li, Ting-Bo Jiang, Si-Jia Sun, Cao Zou","doi":"10.1186/s13287-026-04897-7","DOIUrl":"https://doi.org/10.1186/s13287-026-04897-7","url":null,"abstract":"<p><strong>Background: </strong>Our previous study revealed that intravenous administration of mesenchymal stromal cells (MSCs) increases local cell engraftment and improves heart function. This study aims to investigate whether MSCs overexpressing HLA-G1 have further increased local transplanted cells engraftment and improved heart function.</p><p><strong>Methods: </strong>The mice were intravenously administered saline or human umbilical cord blood-derived MSCs (hUCB-MSCs) 7 days before myocardial infarction (MI) induction. Then, the MI mouse model was established by ligating the left anterior descending coronary artery. The mice were then subjected to intramyocardial transplantation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) 30 min after MI induction. Echocardiographic analysis was carried out to assess heart function. Furthermore, in vivo fluorescent imaging analysis was performed to analyze cell engraftment. Moreover, flow cytometry of splenic regulatory T cells (Tregs) and natural killer (NK) cells was conducted to evaluate the immunomodulatory effect of hUCB-MSCs.</p><p><strong>Results: </strong>The results showed that systemic intravenous administration of hUCB-MSCs substantially enhanced Tregs, decreased NK cells, and increased intramyocardially transplanted hiPSC-CMs' engraftment, thus improving heart function. Compared with hUCB-MSCs, HLA-G1 overexpressing hUCB-MSCs reduced systemic NK cells (7.13 ± 0.19% vs. 9.12 ± 0.06%, p < 0.05), increased Tregs (5.03 ± 0.17% vs. 3.36 ± 0.05%, p < 0.05), improved cell engraftment (Radiant efficiency: 3.01 ± 0.36 × 10<sup>9</sup> vs. 2.19 ± 0.27 × 10<sup>9</sup>, p < 0.05) and heart function (LVEF: 73.00 ± 0.44 vs. 62.36 ± 1.01, p < 0.05). The in vitro assays revealed that HLA-G1 overexpressing hUCB-MSCs modulated the immune response by decreasing pro-inflammatory cytokines.</p><p><strong>Conclusions: </strong>This study showed that systemic intravenous administration of HLA-G1 overexpressing hUCB-MSCs modulated immune response and increased transplanted hiPSC-CMs' engraftment to improve heart function following AMI.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145935033","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
Enhanced proliferation tracer reveals Dorsal-Ventral asymmetry in tracheal epithelial Renewal​. 增强的增殖示踪剂显示气管上皮更新的背腹不对称。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-07 DOI: 10.1186/s13287-025-04888-0
Haiyuan Chen, Yazhu Zhong, Hao Zhang, Wei Yu

To elucidate spatiotemporal dynamics of tissue renewal, we developed evProTracer, an enhanced dual-recombinase lineage tracing system for cumulative in vivo labeling of proliferating cells. Robust longitudinal tracing using evProTracer in murine tracheal epithelium revealed near-complete homeostatic turnover (91.6 ± 1.29% epithelial replacement over 25 weeks), while basal cell-specific ​Trp63-evProTracer​ uncovered a dorsally biased proliferation pattern​, contributing 33.88 ± 1.44% of total epithelial renewal over 6 months, with early differentiation bias toward club cells. These data demonstrate that ventral epithelial renewal is primarily mediated by non-basal facultative progenitors, revealing their constitutive activation during homeostasis. This study uncovers spatially stratified renewal hierarchies: dorsal basal stem cell reservoirs versus ventral facultative non-basal progenitors. evProTracer provides a versatile platform for investigating tissue plasticity hierarchies in regenerative organs.

为了阐明组织更新的时空动力学,我们开发了evProTracer,这是一种增强型双重组酶谱系追踪系统,用于增殖细胞的体内累积标记。使用evProTracer对小鼠气管上皮进行稳健的纵向追踪显示,近完全的稳态更新(25周内上皮替换率为91.6±1.29%),而基底细胞特异性Trp63-evProTracer发现了背侧偏向的增殖模式,在6个月内贡献了33.88±1.44%的总上皮更新,早期分化偏向于club细胞。这些数据表明,腹侧上皮的更新主要是由非基底兼性祖细胞介导的,揭示了它们在稳态过程中的组成激活。这项研究揭示了空间分层的更新层次:背侧基底干细胞库与腹侧兼性非基底祖细胞。evProTracer为研究再生器官的组织可塑性等级提供了一个通用的平台。
{"title":"Enhanced proliferation tracer reveals Dorsal-Ventral asymmetry in tracheal epithelial Renewal​.","authors":"Haiyuan Chen, Yazhu Zhong, Hao Zhang, Wei Yu","doi":"10.1186/s13287-025-04888-0","DOIUrl":"https://doi.org/10.1186/s13287-025-04888-0","url":null,"abstract":"<p><p>To elucidate spatiotemporal dynamics of tissue renewal, we developed evProTracer, an enhanced dual-recombinase lineage tracing system for cumulative in vivo labeling of proliferating cells. Robust longitudinal tracing using evProTracer in murine tracheal epithelium revealed near-complete homeostatic turnover (91.6 ± 1.29% epithelial replacement over 25 weeks), while basal cell-specific ​Trp63-evProTracer​ uncovered a dorsally biased proliferation pattern​, contributing 33.88 ± 1.44% of total epithelial renewal over 6 months, with early differentiation bias toward club cells. These data demonstrate that ventral epithelial renewal is primarily mediated by non-basal facultative progenitors, revealing their constitutive activation during homeostasis. This study uncovers spatially stratified renewal hierarchies: dorsal basal stem cell reservoirs versus ventral facultative non-basal progenitors. evProTracer provides a versatile platform for investigating tissue plasticity hierarchies in regenerative organs.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918490","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
Mesenchymal stromal cells alleviate pulmonary arterial hypertension by suppressing pulmonary arterial adventitial fibroblast activation and extracellular matrix remodeling via the SOCS3/STAT3 pathway. 间充质间质细胞通过SOCS3/STAT3通路抑制肺动脉内皮成纤维细胞活化和细胞外基质重塑,从而缓解肺动脉高压。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-05 DOI: 10.1186/s13287-025-04883-5
Jiaojiao Wang, Jing Jin, Mengni Zhang, Xinyuan Chen, Sheng Du, Xiaoxiao Mao, Changlei Bao, Jinsheng Zhu, Xinyu Song, Shiyue Li

Background: Pulmonary arterial hypertension (PAH) is a fatal condition characterized by progressive vascular remodeling in the pulmonary arteries, eventually leading to right heart failure and death. Dysregulated extracellular matrix (ECM) remodeling is central to PAH pathogenesis and represents a potential therapeutic target. Mesenchymal stromal cells (MSCs) have shown promise in preclinical studies; however, the optimal therapeutic window, dosing frequency, and mechanistic basis for their regulation of vascular ECM remain unclear.

Methods: We employed a monocrotaline (MCT)-induced rat model of PAH to evaluate different MSC treatment regimens, including early administration (day 1 post-MCT), delayed administration (days 7 and 14), and repeated dosing (days 1 and 11). Additionally, we combined in vivo and in vitro approaches to investigate how MSCs modulate the activation of pulmonary arterial adventitial fibroblasts (PAAFs) and influence ECM remodeling.

Results: Biodistribution studies indicated that MSC retention in lung tissue peaked within 24 h and gradually declined by day 21. A single early dose of MSCs (on day 1) significantly ameliorated PAH progression, increasing the 28-day survival rate, reducing right ventricular systolic pressure (RVSP), improving right ventricular function, and attenuating small pulmonary vascular remodeling, including reductions in medial thickening, excessive muscularization, and collagen deposition. Repeated MSC administration did not provide additive therapeutic benefit. Both in animal models and cell cultures, MSCs effectively suppressed PAAF activation and reduced ECM protein production. This anti-fibrotic effect was mediated, at least in part, via the pathway involving the upregulation of SOCS3 and consequent inhibition of STAT3 phosphorylation.

Conclusion: Our findings underscore the importance of early intervention in the PAH disease course for MSC-based therapy. MSCs attenuate vascular remodeling and disease progression, possibly through the SOCS3/STAT3 signaling pathway, by targeting PAAF activation and ECM dysregulation. These results offer a novel mechanistic foundation for MSC treatment in PAH.

背景:肺动脉高压(PAH)是一种以肺动脉进行性血管重构为特征的致命疾病,最终导致右心衰和死亡。细胞外基质(ECM)重塑失调是PAH发病机制的核心,是潜在的治疗靶点。间充质间质细胞(MSCs)在临床前研究中显示出前景;然而,它们调节血管ECM的最佳治疗窗口、给药频率和机制基础仍不清楚。方法:采用MCT诱导的PAH大鼠模型来评估不同的MSC治疗方案,包括早期给药(MCT后第1天)、延迟给药(第7天和第14天)和重复给药(第1天和第11天)。此外,我们结合体内和体外方法研究MSCs如何调节肺动脉外纤维母细胞(PAAFs)的激活和影响ECM重塑。结果:生物分布研究表明,骨髓间充质干细胞在肺组织中的滞留在24 h内达到峰值,并在第21天逐渐下降。单次早期剂量MSCs(第1天)显著改善PAH进展,增加28天存活率,降低右心室收缩压(RVSP),改善右心室功能,减轻小肺血管重构,包括减少内侧增厚、过度肌肉化和胶原沉积。重复给药MSC没有提供附加的治疗益处。在动物模型和细胞培养中,MSCs都能有效抑制PAAF的激活并减少ECM蛋白的产生。这种抗纤维化作用至少部分是通过SOCS3上调和STAT3磷酸化抑制的途径介导的。结论:我们的研究结果强调了早期干预PAH病程对msc治疗的重要性。MSCs可能通过靶向PAAF激活和ECM失调,通过SOCS3/STAT3信号通路减弱血管重塑和疾病进展。这些结果为MSC治疗PAH提供了新的机制基础。
{"title":"Mesenchymal stromal cells alleviate pulmonary arterial hypertension by suppressing pulmonary arterial adventitial fibroblast activation and extracellular matrix remodeling via the SOCS3/STAT3 pathway.","authors":"Jiaojiao Wang, Jing Jin, Mengni Zhang, Xinyuan Chen, Sheng Du, Xiaoxiao Mao, Changlei Bao, Jinsheng Zhu, Xinyu Song, Shiyue Li","doi":"10.1186/s13287-025-04883-5","DOIUrl":"https://doi.org/10.1186/s13287-025-04883-5","url":null,"abstract":"<p><strong>Background: </strong>Pulmonary arterial hypertension (PAH) is a fatal condition characterized by progressive vascular remodeling in the pulmonary arteries, eventually leading to right heart failure and death. Dysregulated extracellular matrix (ECM) remodeling is central to PAH pathogenesis and represents a potential therapeutic target. Mesenchymal stromal cells (MSCs) have shown promise in preclinical studies; however, the optimal therapeutic window, dosing frequency, and mechanistic basis for their regulation of vascular ECM remain unclear.</p><p><strong>Methods: </strong>We employed a monocrotaline (MCT)-induced rat model of PAH to evaluate different MSC treatment regimens, including early administration (day 1 post-MCT), delayed administration (days 7 and 14), and repeated dosing (days 1 and 11). Additionally, we combined in vivo and in vitro approaches to investigate how MSCs modulate the activation of pulmonary arterial adventitial fibroblasts (PAAFs) and influence ECM remodeling.</p><p><strong>Results: </strong>Biodistribution studies indicated that MSC retention in lung tissue peaked within 24 h and gradually declined by day 21. A single early dose of MSCs (on day 1) significantly ameliorated PAH progression, increasing the 28-day survival rate, reducing right ventricular systolic pressure (RVSP), improving right ventricular function, and attenuating small pulmonary vascular remodeling, including reductions in medial thickening, excessive muscularization, and collagen deposition. Repeated MSC administration did not provide additive therapeutic benefit. Both in animal models and cell cultures, MSCs effectively suppressed PAAF activation and reduced ECM protein production. This anti-fibrotic effect was mediated, at least in part, via the pathway involving the upregulation of SOCS3 and consequent inhibition of STAT3 phosphorylation.</p><p><strong>Conclusion: </strong>Our findings underscore the importance of early intervention in the PAH disease course for MSC-based therapy. MSCs attenuate vascular remodeling and disease progression, possibly through the SOCS3/STAT3 signaling pathway, by targeting PAAF activation and ECM dysregulation. These results offer a novel mechanistic foundation for MSC treatment in PAH.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145906920","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
Perspectives from the 2025 ISCBI/ISCI joint workshop on genetic stability, clonal monitoring, ethical data governance, and global inclusion in stem cell banking. 2025年ISCBI/ISCI基因稳定性、克隆监测、伦理数据治理和干细胞库全球包容联合研讨会的观点。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-03 DOI: 10.1186/s13287-025-04797-2
Jung-Hyun Kim, Andreas Kurtz, Ivana Barbaric, Maneesha S Inamdar, Martin F Pera, Nissim Benvenisty, Nika Shakiba, Rosario Isasi, Tadaaki Hanatani, Glyn Stacey

Two international stem cell consortia, the International Stem Cell Initiative (ISCI) and the International Stem Cell Biobanking Initiative (ISCBI, www.iscbi.org ) held a workshop on June 15th 2025 in Hong Kong on genetic variants in human pluripotent stem cell (hPSC) lines and accurate and standardized documentation of donor/hPSC genetic information including ethnicity. The occurrence and detection of genetic variants is a key issue for assuring reproducible stem cell research data and the safety of stem cell derived medicinal products. Presentations by leading experts addressed the nature of hPSC genetic variants, their detection and accurate recording of genetic data and ethnicity. The audience of stem cell researchers, cell banking directors and experts in ethic, policy and stem cell databases, from 13 countries across the globe, discussed progression of the ISCI consortium's efforts in providing further data and thought leadership on the management of genetic variants, and the challenges for standardizing biobanking approaches for hPSC genetic data including ethnicity. This paper records the key elements of this discussion and the conclusions and consensus reached and ongoing work to provide guidance for hPSC biobanks.

两个国际干细胞联盟,国际干细胞倡议组织(ISCI)和国际干细胞生物银行倡议组织(ISCBI, www.iscbi.org)于2025年6月15日在香港举行了一次研讨会,讨论人类多能干细胞(hPSC)系的遗传变异以及供体/hPSC遗传信息的准确和标准化记录,包括种族。遗传变异的发生和检测是确保干细胞研究数据可重复性和干细胞衍生药物安全性的关键问题。主要专家介绍了hPSC遗传变异的性质、它们的检测和遗传数据和种族的准确记录。来自全球13个国家的干细胞研究人员、细胞库主管和伦理、政策和干细胞数据库专家讨论了ISCI联盟在提供遗传变异管理方面的进一步数据和思想领导方面的进展,以及包括种族在内的hPSC遗传数据标准化生物库方法的挑战。本文记录了本次讨论的关键要素、达成的结论和共识以及正在进行的工作,为hPSC生物库提供指导。
{"title":"Perspectives from the 2025 ISCBI/ISCI joint workshop on genetic stability, clonal monitoring, ethical data governance, and global inclusion in stem cell banking.","authors":"Jung-Hyun Kim, Andreas Kurtz, Ivana Barbaric, Maneesha S Inamdar, Martin F Pera, Nissim Benvenisty, Nika Shakiba, Rosario Isasi, Tadaaki Hanatani, Glyn Stacey","doi":"10.1186/s13287-025-04797-2","DOIUrl":"10.1186/s13287-025-04797-2","url":null,"abstract":"<p><p>Two international stem cell consortia, the International Stem Cell Initiative (ISCI) and the International Stem Cell Biobanking Initiative (ISCBI, www.iscbi.org ) held a workshop on June 15th 2025 in Hong Kong on genetic variants in human pluripotent stem cell (hPSC) lines and accurate and standardized documentation of donor/hPSC genetic information including ethnicity. The occurrence and detection of genetic variants is a key issue for assuring reproducible stem cell research data and the safety of stem cell derived medicinal products. Presentations by leading experts addressed the nature of hPSC genetic variants, their detection and accurate recording of genetic data and ethnicity. The audience of stem cell researchers, cell banking directors and experts in ethic, policy and stem cell databases, from 13 countries across the globe, discussed progression of the ISCI consortium's efforts in providing further data and thought leadership on the management of genetic variants, and the challenges for standardizing biobanking approaches for hPSC genetic data including ethnicity. This paper records the key elements of this discussion and the conclusions and consensus reached and ongoing work to provide guidance for hPSC biobanks.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"17 1","pages":"2"},"PeriodicalIF":7.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12764013/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896844","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
Mesenchymal stem cell-derived extracellular vesicles in musculoskeletal regeneration: mechanisms, applications, and future prospects. 间充质干细胞来源的细胞外囊泡在肌肉骨骼再生中的作用:机制、应用和未来前景。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-03 DOI: 10.1186/s13287-025-04879-1
Fatemeh Aziziyan, Shiva Sarani Asl, Mohammadreza Mahdipour, Rahil Nasari Fard, Mohsen Sheykhhasan

Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as promising cell-free therapeutic strategies for musculoskeletal regeneration. MSC-EVs, which are enriched with diverse cargos, exert multifaceted biological effects, including the modulation of inflammation, the promotion of angiogenesis, and the regulation of immune responses. They also activate key regenerative signaling pathways, such as the PI3K/Akt, Wnt/β-catenin, TGF-β/Smad, and NF-κB pathways, thereby promoting osteogenesis, chondrogenesis, tenogenesis, and muscle repair to support the repair of bone, cartilage, tendon, and muscle tissues. In addition to their intrinsic activity, advances in bioengineering, including surface modification, cargo engineering, and integration with biomaterial scaffolds, have further increased their therapeutic potential and delivery. Preclinical studies consistently demonstrate efficacy across diverse musculoskeletal tissues, and early clinical trials highlight their translational promise. Nevertheless, clinical application remains constrained by challenges in large-scale production, standardization, and long-term safety evaluation. This review summarizes current knowledge on the mechanisms, therapeutic applications, engineering strategies, delivery systems, and clinical progress of the use of MSC-EVs in musculoskeletal regeneration while highlighting critical obstacles and future directions for their clinical implementation.

间充质干细胞衍生的细胞外囊泡(msc - ev)已成为肌肉骨骼再生的无细胞治疗策略。骨髓间充质干细胞具有丰富的功能,具有调节炎症、促进血管生成和调节免疫反应等多方面的生物学作用。它们还激活关键的再生信号通路,如PI3K/Akt、Wnt/β-catenin、TGF-β/Smad和NF-κB通路,从而促进骨生成、软骨生成、肌腱生成和肌肉修复,支持骨、软骨、肌腱和肌肉组织的修复。除了其固有的活性外,生物工程的进步,包括表面修饰,货物工程和与生物材料支架的整合,进一步增加了它们的治疗潜力和递送。临床前研究一致证明了不同肌肉骨骼组织的有效性,早期临床试验强调了它们的转化前景。然而,临床应用仍受到大规模生产、标准化和长期安全性评价等方面的挑战。本文综述了msc - ev在肌肉骨骼再生中的作用机制、治疗应用、工程策略、输送系统和临床进展,同时强调了其临床应用的关键障碍和未来方向。
{"title":"Mesenchymal stem cell-derived extracellular vesicles in musculoskeletal regeneration: mechanisms, applications, and future prospects.","authors":"Fatemeh Aziziyan, Shiva Sarani Asl, Mohammadreza Mahdipour, Rahil Nasari Fard, Mohsen Sheykhhasan","doi":"10.1186/s13287-025-04879-1","DOIUrl":"https://doi.org/10.1186/s13287-025-04879-1","url":null,"abstract":"<p><p>Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as promising cell-free therapeutic strategies for musculoskeletal regeneration. MSC-EVs, which are enriched with diverse cargos, exert multifaceted biological effects, including the modulation of inflammation, the promotion of angiogenesis, and the regulation of immune responses. They also activate key regenerative signaling pathways, such as the PI3K/Akt, Wnt/β-catenin, TGF-β/Smad, and NF-κB pathways, thereby promoting osteogenesis, chondrogenesis, tenogenesis, and muscle repair to support the repair of bone, cartilage, tendon, and muscle tissues. In addition to their intrinsic activity, advances in bioengineering, including surface modification, cargo engineering, and integration with biomaterial scaffolds, have further increased their therapeutic potential and delivery. Preclinical studies consistently demonstrate efficacy across diverse musculoskeletal tissues, and early clinical trials highlight their translational promise. Nevertheless, clinical application remains constrained by challenges in large-scale production, standardization, and long-term safety evaluation. This review summarizes current knowledge on the mechanisms, therapeutic applications, engineering strategies, delivery systems, and clinical progress of the use of MSC-EVs in musculoskeletal regeneration while highlighting critical obstacles and future directions for their clinical implementation.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896804","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
Bone marrow-derived mesenchymal stem cells alleviate hepatic lipid metabolism disorders after scald injury: integrating liver transcriptome and metabolome. 骨髓间充质干细胞缓解烫伤后肝脏脂质代谢紊乱:整合肝脏转录组和代谢组。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2026-01-02 DOI: 10.1186/s13287-025-04774-9
Zhian Chen, Ya'e Yang, Xiangwen Shi, Rensheng Yang, Wei Fang, Guangjin Liang, Yun Li, Jing Gao, Lihua Ma, Junchun Yang, Rongqing Pang

Previous studies have confirmed that scald injuries can lead to disturbances in hepatic lipid metabolism, and bone marrow-derived mesenchymal stem cells (BMSCs) have emerged as a promising therapeutic strategy for alleviating such disorders. However, research focusing on the regulation and restoration of liver lipid metabolic processes remains limited. In this study, we investigated the effects of BMSCs on hepatic lipid metabolism disorders induced by scald injury in rats through integrated transcriptomic and metabolomic analyses. The results demonstrated that portal vein infusion of BMSCs markedly improved body weight recovery, reduced hepatic lipid accumulation, normalized serum lipid profiles, and attenuated liver injury following scalding. Combined transcriptomic and metabolomic data further suggested that the therapeutic mechanism may involve inhibition of NF-κB/Gadd45a signaling in hepatocytes, restoration of sphingolipid metabolism, enhancement of hepatic lipid conversion, and suppression of adipocyte lipolysis. Overall, this study provides a theoretical basis for the potential clinical application of BMSCs in treating hepatic lipid metabolism disorders secondary to severe burn injury.

先前的研究证实,烫伤损伤可导致肝脏脂质代谢紊乱,骨髓间充质干细胞(BMSCs)已成为缓解此类疾病的一种有希望的治疗策略。然而,关于肝脏脂质代谢过程的调节和恢复的研究仍然有限。本研究通过综合转录组学和代谢组学分析,探讨骨髓间充质干细胞对烫伤大鼠肝脂质代谢紊乱的影响。结果表明,门静脉输注骨髓间充质干细胞可显著改善小鼠体重恢复,减少肝脏脂质积累,使血清脂质谱正常化,并减轻烫伤后的肝损伤。转录组学和代谢组学的综合数据进一步表明,其治疗机制可能涉及抑制肝细胞NF-κB/Gadd45a信号,恢复鞘脂代谢,增强肝脏脂质转化,抑制脂肪细胞脂解。综上所述,本研究为骨髓间充质干细胞治疗严重烧伤继发性肝脂质代谢紊乱的潜在临床应用提供了理论基础。
{"title":"Bone marrow-derived mesenchymal stem cells alleviate hepatic lipid metabolism disorders after scald injury: integrating liver transcriptome and metabolome.","authors":"Zhian Chen, Ya'e Yang, Xiangwen Shi, Rensheng Yang, Wei Fang, Guangjin Liang, Yun Li, Jing Gao, Lihua Ma, Junchun Yang, Rongqing Pang","doi":"10.1186/s13287-025-04774-9","DOIUrl":"10.1186/s13287-025-04774-9","url":null,"abstract":"<p><p>Previous studies have confirmed that scald injuries can lead to disturbances in hepatic lipid metabolism, and bone marrow-derived mesenchymal stem cells (BMSCs) have emerged as a promising therapeutic strategy for alleviating such disorders. However, research focusing on the regulation and restoration of liver lipid metabolic processes remains limited. In this study, we investigated the effects of BMSCs on hepatic lipid metabolism disorders induced by scald injury in rats through integrated transcriptomic and metabolomic analyses. The results demonstrated that portal vein infusion of BMSCs markedly improved body weight recovery, reduced hepatic lipid accumulation, normalized serum lipid profiles, and attenuated liver injury following scalding. Combined transcriptomic and metabolomic data further suggested that the therapeutic mechanism may involve inhibition of NF-κB/Gadd45a signaling in hepatocytes, restoration of sphingolipid metabolism, enhancement of hepatic lipid conversion, and suppression of adipocyte lipolysis. Overall, this study provides a theoretical basis for the potential clinical application of BMSCs in treating hepatic lipid metabolism disorders secondary to severe burn injury.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"17 1","pages":"1"},"PeriodicalIF":7.3,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12764114/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896835","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 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: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