Human umbilical cord mesenchymal stem cells

人脐带间充质干细胞
  • 文章类型: Journal Article
    由于水凝胶在再生医学中的巨大潜力,通过使用聚乙二醇二缩水甘油醚作为交联剂的绿色工艺开发了新的生物相容性明胶基混合水凝胶,向网络中加入角叉菜胶和壳聚糖多糖,以更好地模拟天然细胞外基质的混合组成。总的来说,水凝胶显示出合适的结构稳定性,高孔隙率和孔隙互连性,良好的膨胀性,最后,生物相容性。它们的力学行为,通过拉伸和压缩试验进行研究,似乎具有高顺应性值的非线性弹性,快速应力松弛,和良好的应变可逆性,在相对较高的变形水平为50%的压缩加载-卸载循环中没有机械破坏的迹象。降解试验证实了水凝胶通过逐渐水解的生物可吸收性,在此期间,两种材料的结构完整性得到保持,而他们的力学行为变得越来越柔顺。人脐带来源的间充质干细胞(hUC-MSC)用于测试水凝胶作为组织工程中细胞递送的潜在载体。在水凝胶内培养的hUC-MSC显示出均匀分布,并在至少21天的培养中保持其生长和活力。具有日益扩散的趋势。因此,这项研究有助于进一步了解混合水凝胶和hUC-MSCs在广泛的生物医学应用中的潜在用途,特别是在软组织工程中。
    Motivated by the enormous potential of hydrogels in regenerative medicine, new biocompatible gelatin-based hybrid hydrogels were developed through a green process using poly(ethylene glycol) diglycidyl ether as a cross-linking agent, adding carrageenan and chitosan polysaccharides to the network to better mimic the hybrid composition of native extracellular matrix. Overall, the hydrogels show suitable structural stability, high porosity and pore interconnectivity, good swellability, and finally, biocompatibility. Their mechanical behavior, investigated by tensile and compression tests, appears to be characterized by nonlinear elasticity with high compliance values, fast stress-relaxation, and good strain reversibility with no sign of mechanical failure for compressive loading-unloading cycles at relatively high deformation levels of 50%. Degradation tests confirm the hydrogel bioresorbability by gradual hydrolysis, during which the structural integrity of both materials is maintained, while their mechanical behavior becomes more and more compliant. Human Umbilical Cord-derived Mesenchymal Stem Cells (hUC-MSCs) were used to test the hydrogels as potential carriers for cell delivery in tissue engineering. hUC-MSCs cultured inside the hydrogels show a homogenous distribution and maintain their growth and viability for at least 21 days of culture, with an increasing proliferation trend. Hence, this study contributes to a further understanding of the potential use of hybrid hydrogels and hUC-MSCs for a wide range of biomedical applications, particularly in soft tissue engineering.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    系统性红斑狼疮(SLE)是一种慢性炎症性自身免疫性疾病,缺乏有效的治疗方法。二甲基氧烯丙基甘氨酸(DMOG)增强间充质干细胞(MSC)的能力,但目前尚不清楚MSCs的DMOG预处理如何增强其SLE治疗。这里,我们探讨了DMOG预处理的人脐带间充质干细胞(hUC-MSCs)在小鼠狼疮性肾炎(LN)模型中的治疗潜力.体外实验表明,DMOG可以减轻肿瘤坏死因子(TNF)-α的mRNA水平,干扰素(IFN)-γ,和白细胞介素(IL)-6,并增加IL-13在脂多糖(LPS)诱导的hUC-MSCs炎症中的mRNA水平。DMOG增强了hUC-MSCs的迁移和侵袭能力。体内动物研究表明,与单独的MSC相比,DMOG预处理的hUC-MSC对淋巴结肿大的抑制作用更为明显,肾脏重量和尿蛋白含量降低。DMOG预处理的hUC-MSCs改善肾脏形态结构,减轻炎症细胞浸润和肾脏纤维化,纤维化标志物的mRNA水平降低证明,包括纤连蛋白(Fn),胶原蛋白α-1链(Colα1),胶原蛋白α-3链(Colα3),和TNF-α,IFN-γ,和IL-6细胞因子。进一步研究发现,DMOG预处理的hUC-MSCs下调转化生长因子(Tgf)-β1及其下游效应子Smad2和Smad3的表达,被认为是肾脏纤维化的主要介质(P<0.05)。研究结果表明,DMOG预处理的hUC-MSCs可以通过增强其抗炎和抗纤维化作用来增强hUC-MSCs在LN中的治疗功效,TGF-β/Smad信号通路可能参与了这一过程。
    Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease lacking effective treatments without adverse effects. Dimethyloxallyl glycine (DMOG) enhanced mesenchymal stem cells (MSC) capabilities, but it remains unclear how DMOG-pretreatment of MSCs augments their SLE treatment. Here, we explore the therapeutic potential of DMOG-pretreated human umbilical cord MSCs (hUC-MSCs) in a mouse lupus nephritis (LN) model. In vitro experiments showed that DMOG could alleviate the mRNA levels of tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and interleukin (IL)-6 and increase the mRNA level of IL-13 in lipopolysaccharide (LPS)-induced inflammation in hUC-MSCs. DMOG enhanced the migratory and invasive abilities of the hUC-MSCs. In vivo animal studies revealed that DMOG-pretreated hUC-MSCs exhibited more pronounced inhibition of lymphadenectasis and reduced kidney weight and urinary protein content than MSCs alone. DMOG-pretreated hUC-MSCs improved renal morphological structure and alleviated inflammatory cell infiltration and renal fibrosis, evidenced by the reduced mRNA levels of fibrosis markers, including fibronectin (Fn), collagen alpha-1 chain (Colα1), collagen alpha-3 chain (Colα3), and TNF-α, IFN-γ, and IL-6 cytokines. Further investigation revealed that DMOG-pretreated hUC-MSCs down-regulated the expressions of transforming growth factor (Tgf)-β1 and its downstream effectors Smad2 and Smad3, recognized as central mediators in renal fibrosis (P < 0.05). The findings suggest that DMOG-pretreated hUC-MSCs can augment the therapeutic efficacy of hUC-MSCs in LN by enhancing their anti-inflammatory and antifibrotic effects, and the TGF-β/Smad signaling pathway may be involved in this process.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:观察人脐带间充质干细胞(hUCMSCs)分泌对小鼠视网膜星形胶质细胞相关因子的影响。探讨hUCMSCs对血管内皮生长因子-A(VEGF-A)表达的影响,并观察其对早产儿视网膜病变(ROP)小鼠模型的治疗作用。
    方法:培养hUCMSCs并从中提取外泌体,然后将视网膜星形胶质细胞分为对照组和缺氧组。MTT测定,流式细胞术,采用逆转录-聚合酶链反应(RT-PCR)和Westernblot检测相关指标。探讨了hUCMSCs外泌体影响缺氧诱导的小鼠视网膜星形胶质细胞VEGF-A表达的可能机制。最后,研究了UCMSCs外泌体在小鼠ROP模型中的功效。Graphpad6用于综合处理数据信息。
    结果:通过梯度超速离心从hUCMSCs的培养上清液中成功提取分泌物。不同缺氧时间下小鼠视网膜星形胶质细胞的活性氧(ROS)和缺氧诱导因子-1α(HIF-1α)表达水平及VEGF-A蛋白和VEGF-AmRNA表达水平升高,缺氧6h后建立ROP细胞模型。中、高浓度hUCMSCs的分泌可降低ROS和HIF-1α,VEGF-A蛋白和VEGF-AmRNA的表达水平具有统计学意义和浓度依赖性。与ROP细胞模型组相比,磷脂酰肌醇3-激酶(PI3K)/蛋白激酶B(AKT)/哺乳动物雷帕霉素靶蛋白(mTOR)信号通路相关因子在hUCMSCs外分泌组中的表达显著降低。玻璃体内注射中、高浓度的hUCMSCs可以减少ROP模型组织中的VEGF-A和HIF-1α。HE染色显示ROP小鼠视网膜新生血管的数量随着hUCMSCs分泌剂量的增加而减少。
    结论:在缺氧诱导的小鼠视网膜星形胶质细胞模型中,发现hUCMSCs外泌体能有效降低HIF-1α和VEGF-A的表达,与hUCMSCs外泌体浓度呈正相关。HUCMSCs外泌体能有效降低ROP小鼠视网膜新生血管的数量及HIF-1α和VEGF-A蛋白的表达,与药物剂量呈正相关。此外,能降低PI3K/AKT/mTOR信号通路的相关因子。
    OBJECTIVE: To observe the effect of human umbilical cord mesenchymal stem cells (hUCMSCs) secretions on the relevant factors in mouse retinal astrocytes, and to investigate the effect of hUCMSCs on the expression of vascular endothelial growth factor-A (VEGF-A) and to observe the therapeutic effect on the mouse model of retinopathy of prematurity (ROP).
    METHODS: Cultured hUCMSCs and extracted exosomes from them and then retinal astrocytes were divided into control group and hypoxia group. MTT assay, flow cytometry, reverse transcription-polymerase chain reaction (RT-PCR) and Western blot were used to detect related indicators. Possible mechanisms by which hUCMSCs exosomes affect VEGF-A expression in hypoxia-induced mouse retinal astrocytes were explored. At last, the efficacy of exosomes of UCMSCs in a mouse ROP model was explored. Graphpad6 was used to comprehensively process data information.
    RESULTS: The secretion was successfully extracted from the culture supernatant of hUCMSCs by gradient ultracentrifugation. Reactive oxygen species (ROS) and hypoxia inducible factor-1α (HIF-1α) of mice retinal astrocytes under different hypoxia time and the expression level of VEGF-A protein and VEGF-A mRNA increased, and the ROP cell model was established after 6h of hypoxia. The secretions of medium and high concentrations of hUCMSCs can reduce ROS and HIF-1α, the expression levels of VEGF-A protein and VEGF-A mRNA are statistically significant and concentration dependent. Compared with the ROP cell model group, the expression of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signal pathway related factors in the hUCMSCs exocrine group is significantly decreased. The intravitreal injection of the secretions of medium and high concentrations of hUCMSCs can reduce VEGF-A and HIF-1α in ROP model tissues. HE staining shows that the number of retinal neovascularization in ROP mice decreases with the increase of the dose of hUCMSCs secretion.
    CONCLUSIONS: In a hypoxia induced mouse retinal astrocyte model, hUCMSCs exosomes are found to effectively reduce the expression of HIF-1α and VEGF-A, which are positively correlated with the concentration of hUCMSCs exosomes. HUCMSCs exosomes can effectively reduce the number of retinal neovascularization and the expression of HIF-1α and VEGF-A proteins in ROP mice, and are positively correlated with drug dosage. Besides, they can reduce the related factors on the PI3K/AKT/mTOR signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:血友病B是一种X连锁出血性疾病,由编码凝血因子IX(FIX)的基因突变引起。基因治疗为治愈这种疾病提供了有希望的潜力。然而,目前相对高剂量的病毒注射方法具有固有的风险。本研究的目的是介绍一种新型的scAAV-DJ/8-LP1-hFIXco载体转导的人脐带血来源的间充质干细胞(HUCMSCs),作为血友病B的常规基因治疗的替代方法。
    方法:我们通过从NCBI检索文献设计了LP1-hFIXco基因结构,并由FIXco-8公司构建了商业载体。以常规方法培养HUCMSC并用scAAV-DJ/8-LP1-hFIXco载体转导。人FIX活化系统用于检测hFIXco活性。使用PCR和蛋白质印迹技术评估hFIXco的RNA和蛋白质表达水平。在动物研究中,NSG和F9-KO小鼠均用于实验,其中凝血时间被用作出血评估的参数。免疫组织化学分析用于评估HUCMSC在小鼠组织切片中的分布。通过苏木精-伊红染色后的病理观察来评估这种基于细胞的基因治疗的致瘤性安全性。
    结果:用scAAV-DJ/8-LP1-hFIXco载体转导HUCMSCs导致人FIXco在体外和小鼠模型中在5个月期间的一致和可持续分泌。分泌水平(hFIXco活性:第7天97.1±2.3%至第5个月时48.8±4.5%)与静脉内注射高剂量病毒载体后观察到的水平相当(hFIXco活性:95.2±2.2%至40.8±4.3%)。经过5个月的观察期,在所研究的任何小鼠中均未观察到组织中转导细胞的克隆扩增。
    结论:我们发现了一种新型且更安全的HUCMSCs介导的方法,可能对血友病B的基因治疗有效。
    BACKGROUND: Hemophilia B is an X-linked bleeding disorder caused by a mutation in the gene responsible for encoding coagulation factor IX (FIX). Gene therapy offers promising potential for curing this disease. However, the current method of relatively high dosage of virus injection carries inherent risks. The purpose of this study was to introduce a novel scAAV-DJ/8-LP1-hFIXco vector transduced human umbilical cord blood derived mesenchymal stem cells (HUCMSCs) as an alternative cell-based gene therapy to conventional gene therapy for Hemophilia B.
    METHODS: The LP1-hFIXco gene structure was designed by us through searching the literature from NCBI and the scAAV-DJ/8-LP1-hFIXco vector was constructed by a commercial company. The HUCMSCs were cultivated in routine approach and transduced with scAAV-DJ/8-LP1-hFIXco vector. The human FIX activation system was employed for detection of hFIXco activity. The RNA and protein expression levels of the hFIXco were evaluated using PCR and western blot techniques. In animal studies, both NSG and F9-KO mice were used for the experiment, in which clotting time was utilized as a parameter for bleeding assessment. The immunohistochemical analysis was used to assess the distribution of HUCMSCs in mouse tissue sections. The safety for tumorigenicity of this cell-based gene therapy was evaluated by pathological observation after hematoxylin-eosin staining.
    RESULTS: The transduction of HUCMSCs with the scAAV-DJ/8-LP1-hFIXco vector results in consistent and sustainable secretion of human FIXco during 5 months period both in vitro and in mouse model. The secretion level (hFIXco activity: 97.1 ± 2.3% at day 7 to 48.8 ± 4.5% at 5 months) was comparable to that observed following intravenous injection with a high dose of the viral vector (hFIXco activity: 95.2 ± 2.2% to 40.8 ± 4.3%). After a 5-month observation period, no clonal expansions of the transduced cells in tissues were observed in any of the mice studied.
    CONCLUSIONS: We have discovered a novel and safer HUCMSCs mediated approach potentially effective for gene therapy in hemophilia B.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    对于急性缺血性中风治疗,治疗方法的局限性和围手术期并发症的高发生率严重影响患者的生存率和术后恢复。人脐带间充质干细胞(hucMSCs)具有多向分化潜能和免疫调节功能,这是一种潜在的细胞疗法。本研究涉及通过大鼠大脑中动脉阻塞(MCAO)后90分钟的血栓切除术建立大鼠脑缺血再灌注损伤模型,并利用综合多系统评估方法,包括检测脑组织缺血,术后生存率,神经评分,麻醉恢复监测,疼痛评估,应激反应,术后肺部并发症,目的探讨尾静脉注射hucMSCs治疗MCAO围手术期并发症的疗效。根据我们的研究,已经确定hucMSCs治疗可以减少脑组织缺血的体积,促进神经功能的恢复,提高MCAO大鼠术后存活率。同时,hucMSCs治疗可延长麻醉恢复时间,缓解麻醉恢复期间谵妄的发生,并且对术后体重减轻也有很好的控制作用,面部疼痛的表情,和肺损伤。它还可以通过调节血糖和包括TNF-α在内的应激相关蛋白的血清水平来减少术后应激反应。IL-6,CRP,NE,皮质醇,β-内啡肽,并最终促进MCAO围手术期并发症的恢复。
    For acute ischemic stroke treatment, the limitations of treatment methods and the high incidence of perioperative complications seriously affect the survival rate and postoperative recovery of patients. Human umbilical cord mesenchymal stem cells (hucMSCs) have multi-directional differentiation potential and immune regulation function, which is a potential cell therapy. The present investigation involved developing a model of cerebral ischemia-reperfusion injury by thrombectomy after middle cerebral artery occlusion (MCAO) for 90 min in rats and utilizing comprehensive multi-system evaluation methods, including the detection of brain tissue ischemia, postoperative survival rate, neurological score, anesthesia recovery monitoring, pain evaluation, stress response, and postoperative pulmonary complications, to elucidate the curative effect of tail vein injection of hucMSCs on MCAO\'s perioperative complications. Based on our research, it has been determined that hucMSCs treatment can reduce the volume of brain tissue ischemia, promote the recovery of neurological function, and improve the postoperative survival rate of MCAO in rats. At the same time, hucMSCs treatment can prolong the time of anesthesia recovery, relieve the occurrence of delirium during anesthesia recovery, and also have a good control effect on postoperative weight loss, facial pain expression, and lung injury. It can also reduce postoperative stress response by regulating blood glucose and serum levels of stress-related proteins including TNF-α, IL-6, CRP, NE, cortisol, β-endorphin, and IL-10, and ultimately promote the recovery of MCAO\'s perioperative complications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    急性肝衰竭(ALF)是一种威胁生命的临床问题,治疗选择有限。人脐带间充质干细胞(hUC-MSC)的给药可能是ALF的一种有希望的方法。本研究旨在探讨hUC-MSCs在ALF治疗中的作用及其机制。
    通过脂多糖和d-半乳糖胺给药诱导ALF的小鼠模型。通过评估血清酶活性来评估hUC-MSCs的治疗效果,组织学外观,和肝组织中的细胞凋亡。在AML12细胞中分析凋亡率。检测与hUC-MSCs共培养的RAW264.7细胞的炎性细胞因子水平和表型。研究了C-Jun氨基末端激酶/核因子-κB信号通路。
    hUC-MSCs治疗降低了血清丙氨酸氨基转移酶和天冬氨酸氨基转移酶的水平,减少病理损伤,减轻肝细胞凋亡,降低体内死亡率。hUC-MSCs共培养可降低AML12细胞的凋亡率。此外,脂多糖刺激的RAW264.7细胞具有较高水平的肿瘤坏死因子-α,白细胞介素-6和白细胞介素-1β显示更多的CD86阳性细胞,而hUC-MSCs共培养降低了三种炎性细胞因子的水平,并增加了CD206阳性细胞的比例。hUC-MSC处理不仅在肝组织中而且在与hUC-MSC共培养的AML12和RAW264.7细胞中抑制磷酸化(p)-C-JunN-末端激酶和p-核因子-κB的活化。
    hUC-MSCs可通过调节肝细胞凋亡和巨噬细胞极化减轻ALF,因此,基于hUC-MSC的细胞治疗可能是ALF患者的替代选择.
    UNASSIGNED: Acute liver failure (ALF) is a life-threatening clinical problem with limited treatment options. Administration of human umbilical cord mesenchymal stem cells (hUC-MSCs) may be a promising approach for ALF. This study aimed to explore the role of hUC-MSCs in the treatment of ALF and the underlying mechanisms.
    UNASSIGNED: A mouse model of ALF was induced by lipopolysaccharide and d-galactosamine administration. The therapeutic effects of hUC-MSCs were evaluated by assessing serum enzyme activity, histological appearance, and cell apoptosis in liver tissues. The apoptosis rate was analyzed in AML12 cells. The levels of inflammatory cytokines and the phenotype of RAW264.7 cells co-cultured with hUC-MSCs were detected. The C-Jun N-terminal kinase/nuclear factor-kappa B signaling pathway was studied.
    UNASSIGNED: The hUC-MSCs treatment decreased the levels of serum alanine aminotransferase and aspartate aminotransferase, reduced pathological damage, alleviated hepatocyte apoptosis, and reduced mortality in vivo. The hUC-MSCs co-culture reduced the apoptosis rate of AML12 cells in vitro. Moreover, lipopolysaccharide-stimulated RAW264.7 cells had higher levels of tumor necrosis factor-α, interleukin-6, and interleukin-1β and showed more CD86-positive cells, whereas the hUC-MSCs co-culture reduced the levels of the three inflammatory cytokines and increased the ratio of CD206-positive cells. The hUC-MSCs treatment inhibited the activation of phosphorylated (p)-C-Jun N-terminal kinase and p-nuclear factor-kappa B not only in liver tissues but also in AML12 and RAW264.7 cells co-cultured with hUC-MSCs.
    UNASSIGNED: hUC-MSCs could alleviate ALF by regulating hepatocyte apoptosis and macrophage polarization, thus hUC-MSC-based cell therapy may be an alternative option for patients with ALF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    由MSC产生的细胞外囊泡(EV)目前被认为是用于皮肤组织再生和修复的新型治疗剂。预处理干细胞可以激活更多的分子途径并释放更多的生物活性剂。在这项研究中,我们从正常(N-EV)和血清和葡萄糖剥夺(SGD-EV)人脐带间充质干细胞(HUCMSCs)中获得EV,并表明SGD-EV促进了迁移,扩散,和体外HUVECs的管形成。利用大鼠模型的体内实验表明,N-EV和SGD-EV均可促进皮肤缺损的血管生成并加速皮肤伤口愈合。而用SGD-EV治疗伤口导致更快的皮肤愈合和增强的血管生成。miRNA测序显示miR-29a-3p在SGD-EV中含量丰富,过表达miR-29a-3p增强了HUVECs的血管生成能力,而抑制miR-29a-3p呈现相反的效果。进一步研究表明,miR-29a-3p直接靶向CTNNBIP1,通过抑制CTNNBIP1激活Wnt/β-catenin信号通路,介导HUCMSCs来源的EV血管生成。一起来看,这些发现表明SGD-EV通过转移miR-29a-3p促进血管生成,Wnt/β-catenin信号通路的激活在SGD-EVs诱导伤口血管生成过程中VEGFA的产生中起关键作用。我们的研究结果为改进电动汽车以增强组织血管生成和增强其在皮肤修复中的作用提供了新的途径。
    Extracellular vesicles (EVs) produced from MSCs were currently considered as a novel therapeutic agent for skin tissue regeneration and repair. Preconditioning stem cells may activate more molecular pathways and release more bioactive agents. In this study, we obtained EVs from normal (N-EVs) and serum- and glucose-deprived (SGD-EVs) human umbilical cord mesenchymal stem cells (HUCMSCs), and showed that SGD-EVs promoted the migration, proliferation, and tube formation of HUVECs in vitro. In vivo experiments utilizing a rat model show that both N-EVs and SGD-EVs boosted angiogenesis of skin defects and accelerated skin wound healing, while treating wounds with SGD-EVs led to faster skin healing and enhanced angiogenesis. miRNA sequencing showed that miR-29a-3p was abundant in SGD-EVs, and overexpressing miR-29a-3p enhanced the angiogenic ability of HUVECs, while inhibiting miR-29a-3p presented the opposite effect. Further studies demonstrated that miR-29a-3p directly targeted CTNNBIP1, which mediated angiogenesis of HUCMSCs-derived EVs through inhibiting CTNNBIP1 to activate Wnt/β-catenin signaling pathway. Taken together, these findings suggested that SGD-EVs promote angiogenesis via transferring miR-29a-3p, and activation of Wnt/β-catenin signaling pathway played a crucial role in SGD-EVs-induced VEGFA production during wound angiogenesis. Our results offered a new avenue for modifying EVs to enhance tissue angiogenesis and augment its role in skin repair.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    白癜风是一种以表皮黑素细胞破坏为特征的自身免疫性疾病,异常的自身免疫反应和过度的氧化应激是两个主要机制。人脐带间充质干细胞来源的外泌体(hUMSCs-Exos)由于具有有效的免疫抑制和抗氧化特性,被认为是自身免疫性疾病的有希望的治疗选择。可以在3D细胞培养条件下增强。然而,hUMSCs3D球状体来源的外泌体(3D-Exos)是否对白癜风具有相当大的治疗效果,其潜在的机制仍然难以捉摸。在这项研究中,3D-Exos的全身给药在治疗白癜风小鼠中显示出显着的效果,如改善皮肤色素脱失所揭示的,CD8+T细胞浸润减少,并在皮肤中扩增Treg细胞,3D-Exos比2D-Exos效果更好。机械上,3D-Exos可以显著促进白癜风皮损中Treg细胞的扩增,并抑制H2O2诱导的黑素细胞凋亡。正向miRNA谱分析和分子实验已经证明,在3D-Exos中富集的miR-132-3p和miR-125b-5p通过分别靶向Sirt1和Bak1极大地促进了这些生物学效应。总的来说,3D-Exos可以通过递送miR-132-3p和miR-125b-5p同时增强Treg细胞介导的免疫抑制和抑制氧化应激诱导的黑素细胞损伤,从而有效改善白癜风。3D-Exos的使用将是白癜风的有希望的治疗方法。
    Vitiligo is an autoimmune disease characterized by epidermal melanocyte destruction, with abnormal autoimmune responses and excessive oxidative stress as two cardinal mechanisms. Human umbilical mesenchymal stem cells-derived exosomes (hUMSCs-Exos) are regarded as promising therapeutic choice for autoimmune diseases due to potent immunosuppressive and anti-oxidative properties, which can be potentiated under 3D cell culture condition. Nevertheless, whether exosomes derived from 3D spheroids of hUMSCs (3D-Exos) exhibit considerable therapeutic effect on vitiligo and the underlying mechanism remain elusive. In this study, systemic administration of 3D-Exos showed a remarkable effect in treating mice with vitiligo, as revealed by ameliorated skin depigmentation, less CD8+T cells infiltration, and expanded Treg cells in skin, and 3D-Exos exerted a better effect than 2D-Exos. Mechanistically, 3D-Exos can prominently facilitate the expansion of Treg cells in vitiligo lesion and suppress H2O2-induced melanocytes apoptosis. Forward miRNA profile analysis and molecular experiments have demonstrated that miR-132-3p and miR-125b-5p enriched in 3D-Exos greatly contributed to these biological effects by targeting Sirt1 and Bak1 respectively. In aggregate, 3D-Exos can efficiently ameliorate vitiligo by simultaneously potentiating Treg cells-mediated immunosuppression and suppressing oxidative stress-induced melanocyte damage via the delivery of miR-132-3p and miR-125b-5p. The employment of 3D-Exos will be a promising treament for vitiligo.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    开发有效的牙槽骨缺损再生方法是骨科的重大挑战。来自人脐带间充质干细胞(HUMSC-Exos)的外泌体已显示出在骨修复中的潜力,但由于不确定的应用方法和机制而面临局限性。为了解决这个问题,将HUMSC-Exos封装在聚乙烯醇(PVA)水凝胶(Exo@PVA)中以创建用于牙槽骨修复的新型材料。这种组合比单独的Exos更有效地增强了骨髓间充质干细胞(BMSCs)和人脐静脉内皮细胞(HUVECs)的成骨分化。此外,Exo@PVA显著改良年夜鼠牙槽骨再生和缺损修复。Exo@PVA中的microRNA-21-5p(miR-21-5p)通过GEO数据库识别并通过计算机模拟方法进行分析,发挥了至关重要的作用。miR-21-5p通过抑制WWP1介导的KLF5泛素化促进BMSC成骨分化,并通过靶向ATP2B4增强HUVEC血管生成。这些发现强调了基于Exo的PVA水凝胶支架修复骨缺损的潜力,通过miR-21-5p/WWP1/ATP2B4信号轴操作。
    Developing effective methods for alveolar bone defect regeneration is a significant challenge in orthopedics. Exosomes from human umbilical cord mesenchymal stem cells (HUMSC-Exos) have shown potential in bone repair but face limitations due to undefined application methods and mechanisms. To address this, HUMSC-Exos were encapsulated in polyvinyl alcohol (PVA) hydrogel (Exo@PVA) to create a novel material for alveolar bone repair. This combination enhanced the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) more effectively than Exos alone. Additionally, Exo@PVA significantly improved alveolar bone regeneration and defect repair in rats. The microRNA-21-5p (miR-21-5p) in Exo@PVA, identified through the GEO database and analyzed via in silico methods, played a crucial role. miR-21-5p promoted BMSC osteogenic differentiation by inhibiting WWP1-mediated KLF5 ubiquitination and enhanced HUVEC angiogenesis by targeting ATP2B4. These findings underscore the potential of an Exo-based approach with PVA hydrogel scaffolds for bone defect repair, operating through the miR-21-5p/WWP1/ATP2B4 signaling axis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    最近,通过髓芯减压(CD)使用细胞疗法治疗股骨头坏死(ONFH)的兴趣日益浓厚.我们的研究目的是探讨人脐带间充质干细胞(hUCMSCs)治疗激素诱导的ONFH的有效性和机制。我们构建了类固醇诱导的ONFH兔模型以及地塞米松(Dex)处理的人股骨头骨微血管内皮细胞(BMECs)模型。我们通过CD将hUCMSCs注射到兔股骨头中。通过显微CT评估hUCMSCs对激素诱导的ONFH兔模型和Dex处理的BMECs的影响。微血管造影,组织学,免疫组织化学,伤口愈合,管形成,和蛋白质印迹分析。此外,我们进行了单细胞RNA测序(scRNA-seq)来检查内皮细胞的特征,信号通路的激活,以及ONFH中的蜂窝间通信。我们的数据表明,hUCMSCs改善了激素诱导的ONFH兔模型的股骨头微结构和骨修复,并促进了血管生成。重要的是,hUCMSCs通过分泌COL6A2通过整合素α1β1激活FAK/PI3K/AKT信号通路,改善Dex处理的BMECs的迁移能力和血管成形术。
    Recently, there has been growing interest in using cell therapy through core decompression (CD) to treat osteonecrosis of the femoral head (ONFH). Our study aimed to investigate the effectiveness and mechanism of human umbilical cord mesenchymal stem cells (hUCMSCs) in treating steroid-induced ONFH. We constructed a steroid-induced ONFH rabbit model as well as dexamethasone (Dex)-treated bone microvascular endothelial cells (BMECs) model of human femoral head. We injected hUCMSCs into the rabbit femoral head via CD. The effects of hUCMSCs on steroid-induced ONFH rabbit model and Dex-treated BMECs were evaluated via micro-CT, microangiography, histology, immunohistochemistry, wound healing, tube formation, and western blotting assay. Furthermore, we conducted single-cell RNA sequencing (scRNA-seq) to examine the characteristics of endothelial cells, the activation of signaling pathways, and inter-cellular communication in ONFH. Our data reveal that hUCMSCs improved the femoral head microstructure and bone repair and promoted angiogenesis in the steroid-induced ONFH rabbit model. Importantly, hUCMSCs improved the migration ability and angioplasty of Dex-treated BMECs by secreting COL6A2 to activate FAK/PI3K/AKT signaling pathway via integrin α1β1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号