Endothelial Progenitor Cells

内皮祖细胞
  • 文章类型: Journal Article
    脑动脉瘤(CA)是一种重要的健康问题,它是由大脑中血管的病理性扩张引起的,并可能导致严重的和潜在的危及生命的疾病。虽然CA的发病机制复杂,新兴的研究表明,内皮祖细胞(EPCs)起着至关重要的作用。在本文中,我们进行了全面的文献综述,以探讨EPCs在CA发病机制和治疗中的潜在作用。目前的研究表明,EPCs的数量减少和功能障碍破坏了内皮功能障碍和修复之间的平衡,从而增加CA形成的风险。逆转这些EPCs异常可能会减少动脉瘤诱导后血管变性的进展,表明EPC是开发新的治疗策略以促进CA修复的有希望的靶标。这促使研究人员开发新的治疗方案,包括药物应用,血管内联合治疗和组织工程治疗。尽管临床前研究显示出了有希望的结果,在临床转化和患者最终受益之前,还有相当长的路要走。尽管如此,这些发现为改善这种疾病的治疗和管理提供了希望。
    Cerebral aneurysm (CA) is a significant health concern that results from pathological dilations of blood vessels in the brain and can lead to severe and potentially life-threatening conditions. While the pathogenesis of CA is complex, emerging studies suggest that endothelial progenitor cells (EPCs) play a crucial role. In this paper, we conducted a comprehensive literature review to investigate the potential role of EPCs in the pathogenesis and treatment of CA. Current research indicates that a decreased count and dysfunction of EPCs disrupt the balance between endothelial dysfunction and repair, thus increasing the risk of CA formation. Reversing these EPCs abnormalities may reduce the progression of vascular degeneration after aneurysm induction, indicating EPCs as a promising target for developing new therapeutic strategies to facilitate CA repair. This has motivated researchers to develop novel treatment options, including drug applications, endovascular-combined and tissue engineering therapies. Although preclinical studies have shown promising results, there is still a considerable way to go before clinical translation and eventual benefits for patients. Nonetheless, these findings offer hope for improving the treatment and management of this condition.
    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
    内皮祖细胞(EPCs)在维持血管健康和帮助修复受损血管中起着至关重要的作用。然而,EPCs来源的外泌体对脂多糖(LPS)引起的血管内皮细胞损伤的具体影响尚不清楚.本研究旨在探讨EPC外泌体在减轻LPS诱导的血管损伤中的潜在益处,并阐明其潜在机制。最初,从小鼠外周血中分离EPCs,并通过流式细胞术和免疫细胞化学证实了它们的身份。随后,使用透射电子显微镜(TEM)和蛋白质印迹分析鉴定源自EPCs的外泌体.通过对脑微血管内皮细胞(BMEC)进行LPS诱导的损伤来诱导脓毒症模型。EPC及其外泌体均显示出BMECs增殖的显着增加,减少细胞凋亡,降低促炎因子(TNF-α,IL-6和caspase-3),并增强了BMEC的发芽和血管生成。值得注意的,外泌体对这些参数有更显著的影响.此外,EPCs和外泌体均表现出显著增加的miR-126a-5p水平,外泌体显示出更实质性的增强。这些发现表明,补充来自EPCs的外泌体miR-126a-5p可以对BMECs提供保护作用,为治疗脓毒症诱导的微血管内皮细胞损伤提供了潜在的治疗选择。
    Endothelial progenitor cells (EPCs) play a crucial role in maintaining vascular health and aiding in the repair of damaged blood vessels. However, the specific impact of EPCs-derived exosomes on vascular endothelial cell injury caused by lipopolysaccharide (LPS) remains inadequately understood. This study aims to explore the potential benefits of EPC-exosomes in mitigating LPS-induced vascular injury and to elucidate the underlying mechanism. Initially, EPCs were isolated from mouse peripheral blood, and their identity was confirmed through flow cytometry and immunocytochemistry. Subsequently, the exosomes derived from EPCs were identified using transmission electron microscopy (TEM) and western blot analysis. A sepsis model was induced by subjecting brain microvascular endothelial cells (BMECs) to LPS-induced injury. Both EPC and their exosomes demonstrated a significant increase in BMECs proliferation, reduced apoptosis, decreased levels of pro-inflammatory factors (TNF-α, IL-6, and caspase-3), and enhanced sprouting and angiogenesis of BMECs. Notable, the Exosomes demonstrated a more pronounced impact on these parameters. Furthermore, both EPCs and Exosomes exhibited significantly increased levels of miR-126a-5p, with the Exosomes showing a more substantial enhancement. These findings suggest that supplementing exosomal miR-126a-5p from EPCs can provide protective effects on BMECs, offering a potential therapeutic option for treating sepsis-induced microvascular endothelial cell injury.
    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
    腿部静脉性溃疡(VLU)是普遍存在的慢性伤口,治疗选择有限。本研究旨在探讨小檗碱在VLU愈合中增强内皮祖细胞(EPC)功能的潜力。
    使用HE染色和ELISA测定评估深静脉血栓形成(DVT)小鼠模型中的组织病理学变化和炎性细胞因子水平。采用荧光素酶报告基因测定来鉴定miR-21-3p和RRAGB靶向关系。EPC增殖,迁移,通过CCK-8、Transwell、和小管形成测定,而mTOR通路和自噬相关蛋白通过免疫荧光染色和免疫印迹分析。
    小檗碱显著改善了EPC功能,如扩散,迁移,和体外试管形成,并在DVT小鼠模型中增强体内EPC介导的伤口愈合。此外,miR-21-3p在VLU患者的EPCs中下调,及其过表达改进了模型EPC功能。机械上,RRAGB,调节mTOR通路,被鉴定为EPCs中潜在的miR-21-3p靶标。RRAGB过表达抑制自噬活性和EPC功能受损。
    小檗碱在改善EPC功能和促进VLU伤口愈合方面显示出希望。小檗碱对miR-21-3p/RRAGB轴的调节可以为管理VLU提供有希望的治疗方法。
    UNASSIGNED: Venous leg ulcers (VLUs) are prevalent chronic wounds with limited treatment options. This study aimed to investigate the potential of berberine to enhance endothelial progenitor cell (EPC) function in VLU healing.
    UNASSIGNED: Histopathological changes and inflammatory cytokine levels in a deep venous thrombosis (DVT) mouse model were assessed using HE staining and ELISA assays. A luciferase reporter assay was employed to identify the miR-21-3p and RRAGB targeting relationship. EPC proliferation, migration, and tube formation were evaluated through CCK-8, Transwell, and tubule formation assays, while the mTOR pathway and autophagy-related proteins were analyzed by immunofluorescence staining and western blotting.
    UNASSIGNED: Berberine significantly improved EPC functions, such as proliferation, migration, and tube formation in vitro, and enhanced in vivo EPC-mediated wound healing in a DVT mouse model. Furthermore, miR-21-3p was downregulated in EPCs from VLU patients, and its overexpression improved model EPC functions. Mechanistically, RRAGB, which regulates the mTOR pathway, was identified as a potential miR-21-3p target in EPCs. Overexpression of RRAGB inhibited autophagic activity and impaired EPC function.
    UNASSIGNED: Berberine shows promise in ameliorating EPC function and promoting wound healing in VLUs. The regulation of the miR-21-3p/RRAGB axis by berberine could offer a promising therapeutic approach for managing VLUs.
    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
    Delta样非规范Notch配体1(DLK1),作为表皮生长因子家族的一员,在体细胞生长中起着关键作用,组织发育和可能的组织更新。尽管以前的研究表明DLK1有助于脂肪生成和肌肉生成,DLK1是否影响血管生成以及它如何与Notch信号相互作用仍有争议,来自不同模型的许多相互矛盾的报道。根据我们的初步发现,DLK1在小鼠缺血腓肠肌和梗死心肌边界区表达上调,在建立后肢缺血(HLI)和心肌梗死(MI)后,我们在C57BL/6小鼠中施用重组DLK1(rDLK1)或PBS,分别。外源性rDLK1显著改善小鼠缺血后肢的血流灌注和肌肉运动功能,HLI之后的第7天,通过促进新血管形成。在MI后第28天在小鼠中证实了对新血管形成的类似作用以及心力衰竭的改善。相应地,CD34+KDR+细胞的数量,表示为内皮祖细胞(EPCs),通过rDLK1给药在小鼠缺血腓肠肌中显著,DAPT作为Notch细胞内结构域(NICD)的特异性抑制剂被废除。此外,从C57BL/6小鼠获得骨髓单核细胞并体外分化为EPCs。暴露于缺氧和血清剥夺时,用rDLK1孵育可触发EPCs中Notch1mRNA和NICD蛋白的表达,促进EPC增殖,迁移,抗凋亡和管形成。否则,rDLK1孵育显著降低细胞内和线粒体活性氧,增加ATP含量和线粒体膜电位,OPA-1表达的短同工型下调,而通过Notch1信号在EPCs中上调mitofusin(-1,-2)表达,都被DAPT废除了。总之,本研究揭示了通过激活内皮祖细胞中的Notch1信号,rDLK1的促血管生成及其机制。
    Delta like non-canonical Notch ligand 1 (DLK1), as a member of epidermal growth factor-like family, plays a critical role in somatic growth, tissue development and possibly tissue renewal. Though previous studies had indicated that DLK1 contributed to adipogenesis and myogenesis, it\'s still controversial whether DLK1 affects angiogenesis and how it interacts with Notch signaling with numerous conflicting reports from different models. Based on our preliminary finding that DLK1 expression was up-regulated in mice ischemic gastrocnemius and in the border zone of infarcted myocardium, we administered either recombinant DLK1 (rDLK1) or PBS in C57BL/6 mice after establishment of hindlimb ischemia (HLI) and myocardial infarction (MI), respectively. Exogenous rDLK1 administration significantly improved both blood perfusion of mice ischemic hindlimbs and muscle motor function on the 3rd, 7th day after HLI, by promoting neovascularization. Similar effect on neovascularization was verified in mice on the 28th day after MI as well as improvement of cardiac failure. Correspondingly, the number of CD34+KDR+ cells, indicated as endothelial progenitor cells (EPCs), was significantly in mice ischemic gastrocnemius by rDLK1 administration, which was abrogated by DAPT as the specific inhibitor of Notch intracellular domain (NICD). Furthermore, bone marrow mononuclear cells were obtained from C57BL/6 mice and differentiated to EPCs ex vivo. Incubation with rDLK1 triggered Notch1 mRNA and NICD protein expressions in EPCs as exposed to hypoxia and serum deprivation, promoting EPCs proliferation, migration, anti-apoptosis and tube formation. Otherwise, rDLK1 incubation significantly decreased intracellular and mitochondrial reactive oxygen species, increased ATP content and mitochondrial membrane potential, downregulated short isoform of OPA-1 expression whereas upregulated mitofusin (-1, -2) expression in EPCs by Notch1 signaling, which were all abrogated by DAPT. In summary, the present study unveils the pro-angiogenesis and its mechanism of rDLK1 through activation of Notch1 signaling in endothelial progenitor cells.
    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:内皮祖细胞(EPCs)在获得性血管生成和内皮损伤修复中起着至关重要的作用。瞬时受体电位典型通道4(TRPC4),商店操作的钙通道的关键组成部分,对于EPC功能至关重要。虽然已经阐明了TRPCs在血管疾病中的作用,TRPC4和EPC功能之间的关系,以及潜在的分子机制,尚不清楚,需要进一步阐明。
    方法:从犬骨髓中分离EPCs,并通过形态学和流式细胞术进行鉴定。使用慢病毒或阴性对照将TRPC4转染到EPC中,并使用实时聚合酶链反应(RT-PCR)评估其表达。扩散,迁移,使用细胞计数试剂盒-8(CCK-8)评估管的形成,Transwell,和Matrigel分析,分别。使用酶联免疫吸附测定(ELISA)测量血管内皮生长因子(VEGF)和基质细胞衍生因子-1(SDF-1)的水平。
    结果:与正常对照(NC)-shRNA组相比,TRPC4-短发夹RNA(shRNA)转染的EPC中的TRPC4mRNA表达显着降低。TRPC4沉默后迁移和成管显著减少,而增殖没有差异。此外,TRPC4沉默后,EPCs中SDF-1和VEGF的水平显着降低。
    结论:TRPC4在调节内皮祖细胞的血管生成中起着至关重要的作用。沉默TRPC4可通过抑制VEGF和SDF-1表达导致血管生成减少,提示TRPC4敲低可能是血管疾病的一种新的治疗策略。
    OBJECTIVE: Endothelial progenitor cells (EPCs) play a crucial role in acquired angiogenesis and endothelial injury repair. Transient receptor potential canonical channel 4 (TRPC4), a key component of store-operated calcium channels, is essential for EPC function. While the role of TRPCs has been clarified in vascular diseases, the relationship between TRPC4 and EPC function, along with the underlying molecular mechanisms, remains unclear and requires further elucidation.
    METHODS: EPCs were isolated from canine bone marrow and identified by morphology and flow cytometry. TRPC4 was transfected into EPCs using lentivirus or negative control, and its expression was assessed using real-time polymerase chain reaction (RT-PCR). Proliferation, migration, and tube formation were evaluated using Cell Counting Kit-8 (CCK-8), Transwell, and Matrigel assays, respectively. Levels of vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1 (SDF-1) were measured using enzyme-linked immunosorbent assay (ELISA).
    RESULTS: TRPC4 mRNA expression was significantly reduced in TRPC4-short hairpin RNA (shRNA) transfected EPCs compared to the normal control (NC)-shRNA groups. Migration and tube formation were significantly decreased after TRPC4 silencing, while proliferation showed no difference. Additionally, levels of SDF-1 and VEGF in EPCs were markedly reduced following TRPC4 silencing.
    CONCLUSIONS: TRPC4 plays a crucial role in regulating angiogenesis in EPCs. Silencing of TRPC4 can lead to decreased angiogenesis by inhibiting VEGF and SDF-1 expression, suggesting that TRPC4 knockdown might be a novel therapeutic strategy for vascular diseases.
    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
    由于骨骼血管系统的降解和骨质疏松微环境内局部骨代谢的破坏,骨质疏松性骨缺损的治疗提出了挑战。然而,通过增强血管化调节局部骨代谢失衡是可行的,一种称为“血管化-骨代谢平衡”的理论。这项研究开发了用EPLQLKM和SVVYGLR肽(PCL-SE)修饰的3D打印聚己内酯(PCL)支架。EPLQLKM肽吸引骨髓间充质干细胞(BMSCs),而SVVYGLR肽增强内皮祖细胞(EPCs)的血管分化,从而通过EPCs的旁分泌作用调节骨代谢和促进骨再生。进一步的机制研究表明,PCL-SE促进了EPCs的血管化,激活BMSCs的Notch信号通路,导致成骨相关基因的上调和破骨细胞相关基因的下调,从而恢复骨代谢平衡。此外,PCL-SE促进EPCs向“H”型血管的分化和BMSCs的募集以协同增强成骨,导致骨质疏松SD大鼠股骨髁骨缺损病例中正常微血管和骨组织再生。这项研究表明PCL-SE支持原位血管化,重塑骨代谢平移平衡,并为骨质疏松性骨缺损提供了有希望的治疗方案。
    The treatment of osteoporotic bone defects poses a challenge due to the degradation of the skeletal vascular system and the disruption of local bone metabolism within the osteoporotic microenvironment. However, it is feasible to modulate the disrupted local bone metabolism imbalance through enhanced vascularization, a theory termed \"vascularization-bone metabolic balance\". This study developed a 3D-printed polycaprolactone (PCL) scaffold modified with EPLQLKM and SVVYGLR peptides (PCL-SE). The EPLQLKM peptide attracts bone marrow-derived mesenchymal stem cells (BMSCs), while the SVVYGLR peptide enhances endothelial progenitor cells (EPCs) vascular differentiation, thus regulating bone metabolism and fostering bone regeneration through the paracrine effects of EPCs. Further mechanistic research demonstrated that PCL-SE promoted the vascularization of EPCs, activating the Notch signaling pathway in BMSCs, leading to the upregulation of osteogenesis-related genes and the downregulation of osteoclast-related genes, thereby restoring bone metabolic balance. Furthermore, PCL-SE facilitated the differentiation of EPCs into \"H\"-type vessels and the recruitment of BMSCs to synergistically enhance osteogenesis, resulting in the regeneration of normal microvessels and bone tissues in cases of femoral condylar bone defects in osteoporotic SD rats. This study suggests that PCL-SE supports in-situ vascularization, remodels bone metabolic translational balance, and offers a promising therapeutic regimen for osteoporotic bone defects.
    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
    脑血管畸形(CCM)主要在大脑中发现,它们会导致中风的风险增加,癫痫发作,和局灶性神经功能缺损.脑血管系统的独特特征是由脑神经血管单元形成的血脑屏障。最近的研究表明,CCM基因的缺失会导致血脑屏障完整性的破坏,这是CCM发育的诱因。CCM病变最初是由血管生成性静脉毛细血管内皮细胞(EC)和相应的常驻内皮祖细胞(EPC)亚群的单克隆扩增而来。然而,脑ECs/EPCs亚类中CCM病变发生和进展的关键信号事件尚不清楚.
    通过使Pdcd10fl/fl小鼠与Mfsd2a-CreERT2小鼠交叉产生脑EC特异性CCM3缺陷型(Pdcd10BECKO)小鼠。通过铬单细胞平台(10X基因组学)进行单细胞RNA测序分析。基于视觉检查和GO分析将细胞簇注释为EC亚型。通过2光子体内成像和组织免疫荧光分析可视化脑血管。通过细胞生物学和生物化学方法进行CCM3和Cav1(caveolin-1)对mTOR(雷帕霉素的机械靶标)信号传导的调节。
    对携带可见CCM病变的P10Pdcd10BECKO小鼠进行单细胞RNA测序分析,发现CCM病变特征和有丝分裂EC簇上调,但血脑屏障相关EC簇减少。然而,从P6Pdcd10BECKO脑的早期阶段发现了一个独特的EPC簇,该簇具有富含mTOR信号的干细胞标志物的高表达水平.的确,mTOR信号在小鼠和人CCM病变中均上调。Raptor(mTOR的调节相关蛋白)的遗传缺陷,但不是Rictor(mTOR的雷帕霉素不敏感伴侣),在Pdcd10BECKO模型中预防CCM病变形成。重要的是,在Pdcd10BECKO小鼠中,mTORC1(mTOR复合物1)药物抑制剂雷帕霉素抑制EPC增殖并改善CCM发病机制。机制研究表明,Cav1/caveolae在CCM3耗尽的EPC介导的细胞内运输和mTORC1信号蛋白的复合物形成中增加。
    CCM3对于维持血脑屏障完整性至关重要,而在脑EPC中CCM3丢失诱导的mTORC1信号传导启动并促进CCM发病机制。
    UNASSIGNED: Cerebral vascular malformations (CCMs) are primarily found within the brain, where they result in increased risk for stroke, seizures, and focal neurological deficits. The unique feature of the brain vasculature is the blood-brain barrier formed by the brain neurovascular unit. Recent studies suggest that loss of CCM genes causes disruptions of blood-brain barrier integrity as the inciting events for CCM development. CCM lesions are proposed to be initially derived from a single clonal expansion of a subset of angiogenic venous capillary endothelial cells (ECs) and respective resident endothelial progenitor cells (EPCs). However, the critical signaling events in the subclass of brain ECs/EPCs for CCM lesion initiation and progression are unclear.
    UNASSIGNED: Brain EC-specific CCM3-deficient (Pdcd10BECKO) mice were generated by crossing Pdcd10fl/fl mice with Mfsd2a-CreERT2 mice. Single-cell RNA-sequencing analyses were performed by the chromium single-cell platform (10× genomics). Cell clusters were annotated into EC subtypes based on visual inspection and GO analyses. Cerebral vessels were visualized by 2-photon in vivo imaging and tissue immunofluorescence analyses. Regulation of mTOR (mechanistic target of rapamycin) signaling by CCM3 and Cav1 (caveolin-1) was performed by cell biology and biochemical approaches.
    UNASSIGNED: Single-cell RNA-sequencing analyses from P10 Pdcd10BECKO mice harboring visible CCM lesions identified upregulated CCM lesion signature and mitotic EC clusters but decreased blood-brain barrier-associated EC clusters. However, a unique EPC cluster with high expression levels of stem cell markers enriched with mTOR signaling was identified from early stages of the P6 Pdcd10BECKO brain. Indeed, mTOR signaling was upregulated in both mouse and human CCM lesions. Genetic deficiency of Raptor (regulatory-associated protein of mTOR), but not of Rictor (rapamycin-insensitive companion of mTOR), prevented CCM lesion formation in the Pdcd10BECKO model. Importantly, the mTORC1 (mTOR complex 1) pharmacological inhibitor rapamycin suppressed EPC proliferation and ameliorated CCM pathogenesis in Pdcd10BECKO mice. Mechanistic studies suggested that Cav1/caveolae increased in CCM3-depleted EPC-mediated intracellular trafficking and complex formation of the mTORC1 signaling proteins.
    UNASSIGNED: CCM3 is critical for maintaining blood-brain barrier integrity and CCM3 loss-induced mTORC1 signaling in brain EPCs initiates and facilitates CCM pathogenesis.
    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:外周动脉疾病(PAD)是一种缺血性疾病,在全球范围内发病率不断上升。lncRNAH19(H19)富集在内皮祖细胞(EPCs)中,和焦凋亡抗性H19过表达EPCs(oe-H19-EPCs)的移植可能促进PAD的血管生成和血流恢复,尤其是严重肢体缺血(CLI)。
    方法:使用免疫荧光和流式细胞术对从人外周血中分离的EPC进行表征。用CCK8和EdU测定法测定细胞增殖。通过Transwell和伤口愈合测定评估细胞迁移。使用管形成测定评价血管生成潜力。蛋白质印迹法检测EPCs中的焦亡途径相关蛋白。使用荧光素酶测定法分析miR-107上H19和FADD的结合位点。在体内,Oe-H19-EPCs移植到小鼠缺血肢体模型中,通过激光多普勒成像检测血流。使用全转录组测序检查了oe-H19-EPCs对缺血性肢体的治疗作用背后的转录景观。
    结果:H19在EPC中的过表达导致增殖增加,迁移,和管形成能力。这些作用是通过焦亡途径介导的,受H19/miR-107/FADD轴调控。在小鼠缺血肢体模型中移植oe-H19-EPCs促进血管生成和血流恢复。全转录组测序表明,oe-H19-EPCs治疗后,缺血肢体血管发生途径显著激活。
    结论:H19过表达通过竞争性结合miR-107增加FADD水平,导致增殖增强,迁移,血管生成,和抑制EPCs的焦亡。这些作用最终促进CLI中血流的恢复。
    BACKGROUND: Peripheral artery disease (PAD) is an ischemic disease with a rising incidence worldwide. The lncRNA H19 (H19) is enriched in endothelial progenitor cells (EPCs), and transplantation of pyroptosis-resistant H19-overexpressed EPCs (oe-H19-EPCs) may promote vasculogenesis and blood flow recovery in PAD, especially with critical limb ischemia (CLI).
    METHODS: EPCs isolated from human peripheral blood was characterized using immunofluorescence and flow cytometry. Cell proliferation was determined with CCK8 and EdU assays. Cell migration was assessed by Transwell and wound healing assays. The angiogenic potential was evaluated using tube formation assay. The pyroptosis pathway-related protein in EPCs was detected by western blot. The binding sites of H19 and FADD on miR-107 were analyzed using Luciferase assays. In vivo, oe-H19-EPCs were transplanted into a mouse ischemic limb model, and blood flow was detected by laser Doppler imaging. The transcriptional landscape behind the therapeutic effects of oe-H19-EPCs on ischemic limbs were examined with whole transcriptome sequencing.
    RESULTS: Overexpression of H19 in EPCs led to an increase in proliferation, migration, and tube formation abilities. These effects were mediated through pyroptosis pathway, which is regulated by the H19/miR-107/FADD axis. Transplantation of oe-H19-EPCs in a mouse ischemic limb model promoted vasculogenesis and blood flow recovery. Whole transcriptome sequencing indicated significant activation of vasculogenesis pathway in the ischemic limbs following treatment with oe-H19-EPCs.
    CONCLUSIONS: Overexpression of H19 increases FADD level by competitively binding to miR-107, leading to enhanced proliferation, migration, vasculogenesis, and inhibition of pyroptosis in EPCs. These effects ultimately promote the recovery of blood flow in CLI.
    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
    背景:内皮祖细胞(EPCs)的移植已被证明可以减少动脉损伤后的新生内膜增生。然而,这种方法的有效性受到有限的EPC归巢到损伤部位的阻碍。此外,未连续监测移植EPCs的体内募集和代谢活性.
    方法:用吲哚菁绿(ICG)结合的超顺磁性氧化铁纳米颗粒(SPIONs)标记EPC,并进行外部磁场靶向,以增强其对Sprague-Dawley大鼠颈动脉球囊损伤(BI)模型的传递。磁性粒子成像(MPI)/荧光成像(FLI)多模态活体成像,损伤后进行3DMPI/CT成像和MPI/FLI离体成像。收集颈动脉并进行病理和免疫荧光染色分析。通过酶联免疫吸附试验分析旁分泌效应。
    结果:磁场的应用显着增强了SPIONs@PEG-ICG-EPCs在动脉损伤部位的定位和保留,体内连续监测和离体观察都证明了这一点。这种靶向递送方法有效地抑制了新生内膜增生并增加了CD31阳性细胞在损伤部位的存在。此外,血清SDF-1α水平,VEGF,IGF-1和TGF-β1显著升高,表明旁分泌活性增强。
    结论:我们的研究结果表明,向动脉损伤区域外磁场定向递送SPIONs@PEG-ICG-EPCs可显著增强其治疗效果。这种增强可能是通过增加的旁分泌信号传导介导的。这些结果强调了磁性引导的SPIONs@PEG-ICG-EPC递送作为治疗动脉损伤的有希望的策略的潜力。
    BACKGROUND: The transplantation of endothelial progenitor cells (EPCs) has been shown to reduce neointimal hyperplasia following arterial injury. However, the efficacy of this approach is hampered by limited homing of EPCs to the injury site. Additionally, the in vivo recruitment and metabolic activity of transplanted EPCs have not been continuously monitored.
    METHODS: EPCs were labeled with indocyanine green (ICG)-conjugated superparamagnetic iron oxide nanoparticles (SPIONs) and subjected to external magnetic field targeting to enhance their delivery to a carotid balloon injury (BI) model in Sprague-Dawley rats. Magnetic particle imaging (MPI)/ fluorescence imaging (FLI) multimodal in vivo imaging, 3D MPI/CT imaging and MPI/FLI ex vivo imaging was performed after injury. Carotid arteries were collected and analyzed for pathology and immunofluorescence staining. The paracrine effects were analyzed by enzyme-linked immunosorbent assay.
    RESULTS: The application of a magnetic field significantly enhanced the localization and retention of SPIONs@PEG-ICG-EPCs at the site of arterial injury, as evidenced by both in vivo continuous monitoring and ex vivo by observation. This targeted delivery approach effectively inhibited neointimal hyperplasia and increased the presence of CD31-positive cells at the injury site. Moreover, serum levels of SDF-1α, VEGF, IGF-1, and TGF-β1 were significantly elevated, indicating enhanced paracrine activity.
    CONCLUSIONS: Our findings demonstrate that external magnetic field-directed delivery of SPIONs@PEG-ICG-EPCs to areas of arterial injury can significantly enhance their therapeutic efficacy. This enhancement is likely mediated through increased paracrine signaling. These results underscore the potential of magnetically guided SPIONs@PEG-ICG-EPCs delivery as a promising strategy for treating arterial injuries.
    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
    背景:深静脉血栓形成(DVT)是由深静脉血液凝结引起的常见血管外科疾病,主要发生在下肢。内皮祖细胞(EPCs)是一种多功能干细胞,它们是血管内皮细胞的前体。EPCs已逐渐发展成为一种有前途的治疗策略,通过各种物理和化学因素的刺激促进深静脉血栓溶解和再通。
    方法:在本研究中,我们利用小鼠DVT模型,并进行了一些实验,包括qRT-PCR,蛋白质印迹,管形成,伤口愈合,Transwell分析,免疫荧光,流式细胞术分析,和免疫沉淀研究HOXD9在EPCs细胞功能中的作用。还探讨了过表达HOXD9的EPCs对DVT模型的治疗作用及其机制。
    结果:过表达HOXD9显著增强EPCs的血管生成和迁移能力,同时抑制细胞凋亡。此外,结果表明,HOXD9特异性靶向HRD1启动子区并调节下游PINK1介导的线粒体自噬。有趣的是,小鼠静脉注射过表达HOXD9的EPCs促进血栓溶解和再通,显著减少静脉血栓形成。
    结论:这项研究的结果表明,HOXD9在刺激内皮祖细胞血管形成中起关键作用,表明其作为DVT管理的治疗目标的潜力。
    BACKGROUND: Deep vein thrombosis (DVT) is a common vascular surgical disease caused by the coagulation of blood in the deep veins, and predominantly occur in the lower limbs. Endothelial progenitor cells (EPCs) are multi-functional stem cells, which are precursors of vascular endothelial cells. EPCs have gradually evolved into a promising treatment strategy for promoting deep vein thrombus dissolution and recanalization through the stimulation of various physical and chemical factors.
    METHODS: In this study, we utilized a mouse DVT model and performed several experiments including qRT-PCR, Western blot, tube formation, wound healing, Transwell assay, immunofluorescence, flow cytometry analysis, and immunoprecipitation to investigate the role of HOXD9 in the function of EPCs cells. The therapeutic effect of EPCs overexpressing HOXD9 on the DVT model and its mechanism were also explored.
    RESULTS: Overexpression of HOXD9 significantly enhanced the angiogenesis and migration abilities of EPCs, while inhibiting cell apoptosis. Additionally, results indicated that HOXD9 specifically targeted the HRD1 promoter region and regulated the downstream PINK1-mediated mitophagy. Interestingly, intravenous injection of EPCs overexpressing HOXD9 into mice promoted thrombus dissolution and recanalization, significantly decreasing venous thrombosis.
    CONCLUSIONS: The findings of this study reveal that HOXD9 plays a pivotal role in stimulating vascular formation in endothelial progenitor cells, indicating its potential as a therapeutic target for DVT management.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号