Endothelial cells

内皮细胞
  • 文章类型: Journal Article
    甲型流感病毒(IAV)感染主要影响肺部,它通常与心血管并发症有关。然而,这种关联的潜在机制尚未完全理解.这里,我们调查了FBXL19的潜在作用,FBXL19是E3泛素连接酶的Skp1-Cullin-F-box家族的成员,在IAV诱导的心脏炎症中。我们证明了FBXL19在内皮细胞(ECs)中的过表达降低了病毒滴度和IAV基质蛋白1(M1)水平,在增加抗病毒基因表达的同时,包括干扰素(IFN)-α,β,IAV感染小鼠心脏组织中的γ和RANTES。此外,FBXL19的EC特异性过表达减弱了IAV感染减少的干扰素调节因子3(IRF3)水平,而不改变其mRNA水平,抑制心脏炎症.此外,IAV感染引发心脏细胞衰老程序,如p16和p21mRNA水平上调所示,以及羔羊B1水平的下调,在ECs中被FBXL19过表达部分逆转。我们的发现表明,FBXL19的EC特异性过表达通过增强干扰素介导的抗病毒信号来保护IAV诱导的心脏损伤,减少心脏炎症,并抑制细胞衰老程序。
    Influenza A virus (IAV) infection while primarily affects the lungs, it is often associated with cardiovascular complications. However, the mechanisms underlying this association are not fully understood. Here, we investigated the potential role of FBXL19, a member of the Skp1-Cullin-F-box family of E3 ubiquitin ligase, in IAV-induced cardiac inflammation. We demonstrated that FBXL19 overexpression in endothelial cells (ECs) reduced viral titers and IAV matrix protein 1 (M1) levels, while increasing antiviral genes expression, including interferon (IFN)-α, β, γ and RANTES in the cardiac tissue of IAV-infected mice. Moreover, EC-specific overexpression of FBXL19 attenuated the IAV infection-reduced interferon regulatory factor 3 (IRF3) level without altering its mRNA level, and suppressed cardiac inflammation. Furthermore, IAV infection triggered cellular senescence programs in the heart as indicated by the upregulation of p16 and p21 mRNA levels, and the downregulation of lamin B1 levels, which were partially reversed by FBXL19 overexpression in ECs. Our findings indicate that EC-specific overexpression of FBXL19 protects against IAV-induced cardiac damage by enhancing interferon-mediated antiviral signaling, reducing cardiac inflammation, and suppressing cellular senescence programs.
    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
    血管内皮细胞(EC)在血管床中表现出异质性,预计会导致特定部位的血管病变。使用体内转录组学评估这种异质性,和体外功能测定,但是,在体外培养的人类EC中,蛋白质组如何比较仍未完全表征。我们在稳态和IFNγ诱导的炎症后,在六个器官和两个体外模型中产生了深入的人类EC蛋白质组景观(>8000蛋白质)。EC蛋白质组表现出高度相似性,器官特异性蛋白有限。ECs之间的变化主要基于增殖和分化过程,其中血液生长内皮细胞(BOEC)和人脐静脉细胞(HUVEC)代表蛋白质组表型的极端。IFNγ应答在所有样品中是高度保守的。利用蛋白质丰度的动力学,我们描绘了VWF和VE-Cadherin相关网络。这种EC景观为从体外角度研究EC生物学和异质性提供了广泛的蛋白质组学补充。
    Blood vessel endothelial cells (EC) display heterogeneity across vascular beds, which is anticipated to drive site-specific vascular pathology. This heterogeneity is assessed using transcriptomics in vivo, and functional assays in vitro, but how proteomes compare across human in vitro cultured ECs remains incompletely characterized. We generated an in-depth human EC proteomic landscape (>8000 proteins) across six organs and two in vitro models in steady-state and upon IFNγ-induced inflammation. EC proteomes displayed a high similarity and organ-specific proteins were limited. Variation between ECs was mainly based on proliferation and differentiation processes in which Blood outgrowth endothelial cells (BOEC) and Human umbilical vein cells (HUVEC) represented the extremes of proteomic phenotypes. The IFNγ response was highly conserved across all samples. Harnessing dynamics in protein abundances we delineated VWF and VE-Cadherin correlation networks. This EC landscape provides an extensive proteomic addition in studying EC biology and heterogeneity from an in vitro perspective.
    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
    小胶质细胞,中枢神经系统的固有免疫细胞,与大脑最基本的过程密切相关,从在发育过程中修剪神经突触到防止整个生命中过度的神经元活动。研究报道了在疾病背景下,小胶质细胞在血脑屏障(BBB)中的有益和有害作用。然而,对健康大脑中小胶质细胞-内皮细胞相互作用的了解较少。为了研究小胶质细胞在健康BBB中的作用,我们使用集落刺激因子1受体(CSF1R)抑制剂PLX5622消耗小胶质细胞,并分析BBB超微结构,渗透性,和转录组。有趣的是,我们发现,尽管它们与内皮细胞直接接触,小胶质细胞不是维持BBB结构所必需的,函数,或健康大脑中的基因表达。然而,我们发现PLX5622治疗改变了脑内皮胆固醇代谢.这种效应与小胶质细胞耗竭无关,表明PLX5622对脑血管系统有脱靶效应。
    Microglia, the resident immune cells of the central nervous system, are intimately involved in the brain\'s most basic processes, from pruning neural synapses during development to preventing excessive neuronal activity throughout life. Studies have reported both helpful and harmful roles for microglia at the blood-brain barrier (BBB) in the context of disease. However, less is known about microglia-endothelial cell interactions in the healthy brain. To investigate the role of microglia at a healthy BBB, we used the colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 to deplete microglia and analyzed the BBB ultrastructure, permeability, and transcriptome. Interestingly, we found that, despite their direct contact with endothelial cells, microglia are not necessary for the maintenance of BBB structure, function, or gene expression in the healthy brain. However, we found that PLX5622 treatment alters brain endothelial cholesterol metabolism. This effect was independent from microglial depletion, suggesting that PLX5622 has off-target effects on brain vasculature.
    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
    暂无摘要。
    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:本研究的目的是研究CRISPR/Cas9介导的A4GALT抑制在挽救源自人诱导多能干细胞(hiPSCs)的法布里病(FD)内皮细胞(FD-ECs)的内皮功能障碍中的功效。
    方法:我们分化了hiPSC(WT(野生型),WTC-11),GLA-突变型hiPSCs(GLA-KO,CMC-Fb-002),和CRISPR/Cas9介导的A4GALT-KOhiPSC(GLA/A4GALT-KO,Fb-002-A4GALT-KO)进入ECs,并比较FD表型和内皮功能障碍。我们还通过RNA测序分析了A4GALT抑制对活性氧(ROS)形成和转录组谱的影响。
    结果:GLA-突变型hiPSC-EC(GLA-KO和CMC-Fb-002)显示EC标记表达下调,α-GalA表达显著降低,同时Gb-3沉积和溶酶体内包涵体增加。然而,GLA/A4GALT-KO和Fb-002-A4GALT-KOhiPSC-EC中CRISPR/Cas9介导的A4GALT抑制增加了EC标志物的表达水平并拯救了这些FD表型。GLA-突变型hiPSC-EC在管形成测定中未能形成管状结构,显示细胞向划伤区域的迁移显著减少。相比之下,A4GALT抑制改善了管形成和细胞迁移能力。Westernblot分析显示GLA-KOhiPSC-EC中MAPK和AKT磷酸化水平下调,而SOD和过氧化氢酶上调。然而,A4GALT的抑制恢复了这些蛋白质的改变。RNA测序分析表明GLA突变体EC的显著转录组变化,尤其是在血管生成中,细胞死亡,和细胞对氧化应激的反应。然而,这些在GLA/A4GALT-KOhiPSC-ECs中有效恢复。
    结论:CRISPR/Cas9介导的A4GALT抑制挽救了GLA突变hiPSC-ECs的FD表型和内皮功能障碍,为FD血管病变提供了一种潜在的治疗方法。
    OBJECTIVE: The objective of this study was to investigate the efficacy of CRISPR/Cas9-mediated A4GALT suppression in rescuing endothelial dysfunction in Fabry disease (FD) endothelial cells (FD-ECs) derived from human induced pluripotent stem cells (hiPSCs).
    METHODS: We differentiated hiPSCs (WT (wild-type), WTC-11), GLA-mutant hiPSCs (GLA-KO, CMC-Fb-002), and CRISPR/Cas9-mediated A4GALT-KO hiPSCs (GLA/A4GALT-KO, Fb-002-A4GALT-KO) into ECs and compared FD phenotypes and endothelial dysfunction. We also analyzed the effect of A4GALT suppression on reactive oxygen species (ROS) formation and transcriptome profiles through RNA sequencing.
    RESULTS: GLA-mutant hiPSC-ECs (GLA-KO and CMC-Fb-002) showed downregulated expression of EC markers and significantly reduced α-GalA expression with increased Gb-3 deposition and intra-lysosomal inclusion bodies. However, CRISPR/Cas9-mediated A4GALT suppression in GLA/A4GALT-KO and Fb-002-A4GALT-KO hiPSC-ECs increased expression levels of EC markers and rescued these FD phenotypes. GLA-mutant hiPSC-ECs failed to form tube-like structure in tube formation assays, showing significantly decreased migration of cells into the scratched wound area. In contrast, A4GALT suppression improved tube formation and cell migration capacity. Western blot analysis revealed that MAPK and AKT phosphorylation levels were downregulated while SOD and catalase were upregulated in GLA-KO hiPSC-ECs. However, suppression of A4GALT restored these protein alterations. RNA sequencing analysis demonstrated significant transcriptome changes in GLA-mutant EC, especially in angiogenesis, cell death, and cellular response to oxidative stress. However, these were effectively restored in GLA/A4GALT-KO hiPSC-ECs.
    CONCLUSIONS: CRISPR/Cas9-mediated A4GALT suppression rescued FD phenotype and endothelial dysfunction in GLA-mutant hiPSC-ECs, presenting a potential therapeutic approach for FD-vasculopathy.
    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
    我们提出了一个创新的体外模型,旨在研究组织刚度和剪切应力对内皮细胞(EC)功能的联合影响,这对于了解血管健康和动脉粥样硬化等疾病的发作至关重要。传统上,研究探讨了剪切应力和基底刚度对EC的影响,独立。然而,该集成系统结合了这些因素,以提供更精确的模拟的机械环境的脉管系统。目的是使用人EC检查跨各种组织硬度水平和流量条件的EC机械传导。我们详细介绍了合成具有可调刚度的明胶甲基丙烯酸酯(GelMA)水凝胶并用EC播种以实现汇合的方案。此外,我们描述了具有成本效益的流动室的设计和组装,辅以计算流体动力学模拟,以产生以层流和适当的剪切应力水平为特征的生理流动条件。该协议还纳入了荧光标记的共聚焦显微镜,能够评估EC对组织顺应性和流量状况的反应。通过对培养的EC进行多种综合机械刺激,该模型能够全面研究高血压和衰老等因素如何影响EC功能和EC介导的血管疾病.从这些研究中获得的见解将有助于阐明血管疾病的潜在机制和制定有效的治疗策略。
    We present an innovative in vitro model aimed at investigating the combined effects of tissue rigidity and shear stress on endothelial cell (EC) function, which are crucial for understanding vascular health and the onset of diseases such as atherosclerosis. Traditionally, studies have explored the impacts of shear stress and substrate stiffness on ECs, independently. However, this integrated system combines these factors to provide a more precise simulation of the mechanical environment of the vasculature. The objective is to examine EC mechanotransduction across various tissue stiffness levels and flow conditions using human ECs. We detail the protocol for synthesizing gelatin methacrylate (GelMA) hydrogels with tunable stiffness and seeding them with ECs to achieve confluency. Additionally, we describe the design and assembly of a cost-effective flow chamber, supplemented by computational fluid dynamics simulations, to generate physiological flow conditions characterized by laminar flow and appropriate shear stress levels. The protocol also incorporates fluorescence labeling for confocal microscopy, enabling the assessment of EC responses to both tissue compliance and flow conditions. By subjecting cultured ECs to multiple integrated mechanical stimuli, this model enables comprehensive investigations into how factors such as hypertension and aging may affect EC function and EC-mediated vascular diseases. The insights gained from these investigations will be instrumental in elucidating the mechanisms underlying vascular diseases and in developing effective treatment strategies.
    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
    鞘脂是由内质网(ER)中的非鞘脂前体合成的十八碳醇脂质。鞘脂作为我们细胞中发现的大量部分的前体,在各种细胞过程中发挥关键作用。包括细胞分裂,衰老,迁移,分化,凋亡,焦亡,自噬,营养摄入,新陈代谢,和蛋白质合成。在CVD中,不同亚类的鞘脂和其他衍生分子,如鞘磷脂(SM),神经酰胺(CER),鞘氨醇-1-磷酸(S1P)与糖尿病心肌病直接相关,扩张型心肌病,心肌炎,缺血性心脏病(IHD),高血压,和动脉粥样硬化。几项全基因组关联研究表明,鞘脂途径基因的遗传变异与CVD风险之间存在关联。鞘脂途径在外泌体的生物发生和分泌中起重要作用。最近发现小的细胞外囊泡(sEV)/外泌体是CVD发作的可能指标,连接有助于疾病进展的各种细胞信号传导途径。电动汽车的重要特征,如生物相容性,和跨越生物屏障可以改善药物的药代动力学,并将被用于开发下一代药物递送系统。在这次审查中,我们已经全面讨论了鞘脂的作用,和鞘脂代谢产物在CVD的发展中。此外,进行了简洁的讨论,讨论了sEV/外泌体在调节CVD的病理生理过程中的作用,并将外泌体作为治疗靶标.
    Sphingolipids are eighteen carbon alcohol lipids synthesized from non-sphingolipid precursors in the endoplasmic reticulum (ER). The sphingolipids serve as precursors for a vast range of moieties found in our cells that play a critical role in various cellular processes, including cell division, senescence, migration, differentiation, apoptosis, pyroptosis, autophagy, nutrition intake, metabolism, and protein synthesis. In CVDs, different subclasses of sphingolipids and other derived molecules such as sphingomyelin (SM), ceramides (CERs), and sphingosine-1-phosphate (S1P) are directly related to diabetic cardiomyopathy, dilated cardiomyopathy, myocarditis, ischemic heart disease (IHD), hypertension, and atherogenesis. Several genome-wide association studies showed an association between genetic variations in sphingolipid pathway genes and the risk of CVDs. The sphingolipid pathway plays an important role in the biogenesis and secretion of exosomes. Small extracellular vesicles (sEVs)/ exosomes have recently been found as possible indicators for the onset of CVDs, linking various cellular signaling pathways that contribute to the disease progression. Important features of EVs like biocompatibility, and crossing of biological barriers can improve the pharmacokinetics of drugs and will be exploited to develop next-generation drug delivery systems. In this review, we have comprehensively discussed the role of sphingolipids, and sphingolipid metabolites in the development of CVDs. In addition, concise deliberations were laid to discuss the role of sEVs/exosomes in regulating the pathophysiological processes of CVDs and the exosomes as therapeutic targets.
    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:肌萎缩侧索硬化症(ALS)是一种快速发展的神经退行性疾病,具有最低限度的有效治疗选择。ALS药物开发中的一个重要障碍是目前受血脑屏障(BBB)存在限制的运动皮质的非侵入性治疗性通路。聚焦超声和微泡(FUS+MB)治疗是一项新兴技术,已成功用于ALS患者暂时打开皮质BBB。然而,FUS+MB介导的跨ALS患者BBB的药物递送尚未报道。同样,FUS+MB对人ALSBBB细胞的影响仍未被探索。
    方法:在这里,我们建立了第一个FUS+MB兼容,基于诱导的脑内皮样细胞(iBECs)的完全人ALS患者细胞衍生的BBB模型,以研究体外抗TDP-43抗体递送和FUSMB生物效应。
    结果:生成的ALSiBECs概括了BBB病理的疾病特异性标志,包括降低BBB完整性和渗透性,和TDP-43蛋白病。结果还确定了散发性ALS和家族性(携带C9orf72扩增)ALSiBECs之间的差异,反映了与疾病亚组相关的患者异质性。这些模型中的研究揭示了在体外成功的ALSiBEC单层开放,没有FUS+MB的不利细胞作用,如通过乳酸脱氢酶(LDH)释放活力测定所反映的,并且在FUS+MB处理的细胞中缺乏可见的单层损伤或形态变化。这伴随着ALSiBECs中FUSMB的分子生物学效应,包括紧密和粘附连接标记表达的变化,以及药物转运蛋白和炎症介质,零星和C9orf72ALSiBECs产生瞬时特异性反应。此外,我们证明,在C9orf72(2.7倍)和散发性(1.9倍)ALSiBECs中,FUS+MB有效增加了抗TDP-43抗体的递送量,这首次提供了概念证据,证明FUS+MB可用于增强大分子疗法在人ALS体外模型中跨BBB的通透性.
    结论:一起,这项研究首次描述了ALSiBECs对FUS+MB的细胞和分子反应,并为ALSBBB体外模型上FUS+MB介导的药物递送筛选提供了完全人类的平台.
    BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disorder with minimally effective treatment options. An important hurdle in ALS drug development is the non-invasive therapeutic access to the motor cortex currently limited by the presence of the blood-brain barrier (BBB). Focused ultrasound and microbubble (FUS+ MB) treatment is an emerging technology that was successfully used in ALS patients to temporarily open the cortical BBB. However, FUS+ MB-mediated drug delivery across ALS patients\' BBB has not yet been reported. Similarly, the effects of FUS+ MB on human ALS BBB cells remain unexplored.
    METHODS: Here we established the first FUS+ MB-compatible, fully-human ALS patient-cell-derived BBB model based on induced brain endothelial-like cells (iBECs) to study anti-TDP-43 antibody delivery and FUS+ MB bioeffects in vitro.
    RESULTS: Generated ALS iBECs recapitulated disease-specific hallmarks of BBB pathology, including reduced BBB integrity and permeability, and TDP-43 proteinopathy. The results also identified differences between sporadic ALS and familial (C9orf72 expansion carrying) ALS iBECs reflecting patient heterogeneity associated with disease subgroups. Studies in these models revealed successful ALS iBEC monolayer opening in vitro with no adverse cellular effects of FUS+ MB as reflected by lactate dehydrogenase (LDH) release viability assay and the lack of visible monolayer damage or morphology change in FUS+ MB treated cells. This was accompanied by the molecular bioeffects of FUS+ MB in ALS iBECs including changes in expression of tight and adherens junction markers, and drug transporter and inflammatory mediators, with sporadic and C9orf72 ALS iBECs generating transient specific responses. Additionally, we demonstrated an effective increase in the delivery of anti-TDP-43 antibody with FUS+ MB in C9orf72 (2.7-fold) and sporadic (1.9-fold) ALS iBECs providing the first proof-of-concept evidence that FUS+ MB can be used to enhance the permeability of large molecule therapeutics across the BBB in a human ALS in vitro model.
    CONCLUSIONS: Together, this study describes the first characterisation of cellular and molecular responses of ALS iBECs to FUS+ MB and provides a fully-human platform for FUS+ MB-mediated drug delivery screening on an ALS BBB in vitro model.
    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
    肾上腺皮质功能不全的细胞疗法可以潜在地提供响应于生理刺激的类固醇替代。以前,我们报道了脂肪组织来源的基质细胞(ADSC)通过核受体亚家族5组A成员1(NR5A1)的过表达而转化为类固醇生成细胞.类固醇生成细胞的特征在于产生肾上腺和性腺类固醇。肾上腺皮质功能不全的细胞治疗需要具有更多肾上腺皮质特征的细胞。考虑到肾上腺皮质内高度发达的血管网络,所有肾上腺皮质细胞与血管内皮细胞(VEC)相邻并相互作用.在这项研究中,将源自小鼠ADSC的NR5A1诱导的类固醇生成细胞(NR5A1-ADSC)与小鼠VEC共培养。NR5A1-ADSCs的睾酮分泌没有改变;然而,在皮质酮合成途径中,皮质酮分泌显着增加,而类固醇生成酶的水平显着增加。与淋巴内皮细胞(LECs)或ADSCs共培养,或与NR5A1-ADSCs和VECs的transwell培养没有改变皮质酮的产生。VEC比LEC表达更高水平的胶原蛋白和层粘连蛋白。IV型胶原蛋白和层粘连蛋白包被的培养皿中的培养增加了NR5A1-ADSC的皮质酮分泌。这些结果表明,VECs可能将ADSC衍生的类固醇细胞表征为更多的皮质酮产生表型,和VEC可用于从干细胞产生肾上腺类固醇细胞。
    Cell therapy for adrenocortical insufficiency can potentially provide steroid replacement in response to physiological stimuli. Previously, we reported that adipose tissue-derived stromal cells (ADSCs) are transformed into steroid-producing cells by overexpression of nuclear receptor subfamily 5 group A member 1 (NR5A1). The steroidogenic cells are characterized by the production of both adrenal and gonadal steroids. Cytotherapy for adrenocortical insufficiency requires cells with more adrenocortical characteristics. Considering the highly developed vascular network within the adrenal cortex, all adrenocortical cells are adjacent to and interact with vascular endothelial cells (VECs). In this study, NR5A1-induced steroidogenic cells derived from mouse ADSCs (NR5A1-ADSCs) were co-cultured with mouse VECs. Testosterone secretion in NR5A1-ADSCs was not altered; however, corticosterone secretion significantly increased while levels of steroidogenic enzymes significantly increased in the corticosterone synthesis pathway. Co-culture with lymphatic endothelial cells (LECs) or ADSCs, or transwell culture with NR5A1-ADSCs and VECs did not alter corticosterone production. VECs expressed higher levels of collagen and laminin than LECs. Culture in type-IV collagen and laminin-coated dishes increased corticosterone secretion in NR5A1-ADSCs. These results suggest that VECs may characterize ADSC-derived steroidogenic cells into a more corticosterone-producing phenotype, and VECs may be useful for generating adrenal steroidogenic cells from stem cells.
    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
    双膦酸盐是有效的骨吸收抑制剂,其中阿仑膦酸钠(ALN)是大多数骨质疏松症患者的常用处方,但长期应用ALN可导致双膦酸盐相关性颌骨坏死(BRONJ),其发病机制尚不清楚。先前的研究表明,双膦酸盐通过影响血管内皮细胞的生物学行为而引起颌骨缺血,导致BRONJ.然而,ALN对血管内皮细胞的影响及其机制尚不清楚。这项工作的目的是评估ALN对人脐静脉内皮细胞(HUVECs)的影响,并阐明所涉及的分子途径。我们发现高浓度的ALN诱导HUVECsG1期阻滞,通过细胞周期蛋白D1和细胞周期蛋白D3的下调来证明。此外,高浓度ALN处理对HUVECs有促凋亡作用,通过裂解的caspase-3,裂解的PARP和Bax的水平增加来证明,随着抗凋亡蛋白Bcl-2水平的降低。进一步试验显示ERK1/2磷酸化降低。此外,ALN引起HUVEC中活性氧(ROS)的积累,导致ERK1/2途径抑制。N-乙酰-L-半胱氨酸(NAC),ROS清除剂,有效促进ERK1/2磷酸化,减轻ALN在HUVECs中引发的G1期阻滞和凋亡。PD0325901,一种ERK1/2抑制剂,可减少ERK1/2磷酸化,增强ALN诱导的HUVECsG1期停滞和凋亡。这些发现表明ALN通过ROS介导的ERK1/2通路抑制在HUVECs中诱导G1期阻滞和凋亡,提供对致病过程的新见解,在接受长期使用ALN的个体中预防和治疗BRONJ。
    Bisphosphonates are potent bone resorption inhibitors, among which alendronate sodium (ALN) is commonly prescribed for most osteoporosis patients, but long-term application of ALN can cause bisphosphonate-related osteonecrosis of jaw (BRONJ), the pathogenesis of which remains unclear. Previous studies have suggested that bisphosphonates cause jaw ischemia by affecting the biological behavior of vascular endothelial cells, leading to BRONJ. However, the impacts of ALN on vascular endothelial cells and its mechanism remain unclear. The purpose of this work is to assess the influence of ALN on human umbilical vein endothelial cells (HUVECs) and clarify the molecular pathways involved. We found that high concentration of ALN induced G1 phase arrest in HUVECs, demonstrated by downregulation of Cyclin D1 and Cyclin D3. Moreover, high concentration of ALN treatment showed pro-apoptotic effect on HUVECs, demonstrated by increased levels of the cleaved caspase-3, the cleaved PARP and Bax, along with decreased levels of anti-apoptotic protein Bcl-2. Further experiments showed that ERK1/2 phosphorylation was decreased. Additionally, ALN provoked the build-up of reactive oxygen species (ROS) in HUVECs, leading to ERK1/2 pathway suppression. N-acetyl-L-cysteine (NAC), a ROS scavenger, efficiently promoted the ERK1/2 phosphorylation and mitigated the G1 phase arrest and apoptosis triggered by ALN in HUVECs. PD0325901, an inhibitor of ERK1/2 that diminishes the ERK1/2 phosphorylation enhanced the ALN-induced G1 phase arrest and apoptosis in HUVECs. These findings show that ALN induces G1 phase arrest and apoptosis through ROS-mediated ERK1/2 pathway inhibition in HUVECs, providing novel insights into the pathogenic process, prevention and treatment of BRONJ in individuals receiving extended use of ALN.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号