PMVECs

  • 文章类型: Journal Article
    背景:肥胖是导致各种呼吸系统疾病发展的主要危险因素,如哮喘和肺动脉高压。肺微血管内皮细胞(PMVECs)在肺部疾病的发生发展中起着重要作用。乌头酸脱羧酶1(Acod1)介导衣康酸的生产,据报道,Acod1/衣康酸轴在多种疾病中起保护作用。然而,Acod1/衣康酸轴在肥胖小鼠PMVECs中的作用尚不清楚.
    方法:进行mRNA-seq以鉴定高脂饮食(HFD)诱导的小鼠PMVECs和食物喂养的PMVECs之间的差异表达基因(DEGs)(|log2倍数变化|≥1,p≤0.05)。游离脂肪酸(FFA)用于诱导细胞损伤,用Acod1过表达的质粒或4-辛基衣酯(4-OI)给药后,小鼠PMVEC中的炎症和线粒体氧化应激。此外,我们研究了核因子类红细胞2样2(Nrf2)通路是否参与了Acod1/衣康酸在FFA诱导的PMVECs中的作用.
    结果:通过mRNA-seq在HFD小鼠PMVEC中鉴定出下调的Acod1。在FFA处理的PMVEC中Acod1表达也降低。Acod1过表达抑制细胞损伤,FFA诱导的小鼠PMVECs炎症和线粒体氧化应激。4-OI施用在FFA处理的小鼠PMVEC中显示一致的结果。此外,沉默Nrf2逆转了FFA处理的PMVECs中Acod1过表达和4-OI给药的效果,表明Nrf2激活是Acod1/衣康酸的保护作用所必需的。
    结论:我们的结果表明,Acod1/衣酯轴可能保护小鼠PMVECs免受FFA诱导的损伤,炎症和线粒体氧化应激经由过程激活Nrf2通路。这对肥胖引起的肺微血管内皮病的治疗具有重要意义。
    BACKGROUND: Obesity is the main risk factor leading to the development of various respiratory diseases, such as asthma and pulmonary hypertension. Pulmonary microvascular endothelial cells (PMVECs) play a significant role in the development of lung diseases. Aconitate decarboxylase 1 (Acod1) mediates the production of itaconate, and Acod1/itaconate axis has been reported to play a protective role in multiple diseases. However, the roles of Acod1/itaconate axis in the PMVECs of obese mice are still unclear.
    METHODS: mRNA-seq was performed to identify the differentially expressed genes (DEGs) between high-fat diet (HFD)-induced PMVECs and chow-fed PMVECs in mice (|log2 fold change| ≥ 1, p ≤ 0.05). Free fatty acid (FFA) was used to induce cell injury, inflammation and mitochondrial oxidative stress in mouse PMVECs after transfection with the Acod1 overexpressed plasmid or 4-Octyl Itaconate (4-OI) administration. In addition, we investigated whether the nuclear factor erythroid 2-like 2 (Nrf2) pathway was involved in the effects of Acod1/itaconate in FFA-induced PMVECs.
    RESULTS: Down-regulated Acod1 was identified in HFD mouse PMVECs by mRNA-seq. Acod1 expression was also reduced in FFA-treated PMVECs. Acod1 overexpression inhibited cell injury, inflammation and mitochondrial oxidative stress induced by FFA in mouse PMVECs. 4-OI administration showed the consistent results in FFA-treated mouse PMVECs. Moreover, silencing Nrf2 reversed the effects of Acod1 overexpression and 4-OI administration in FFA-treated PMVECs, indicating that Nrf2 activation was required for the protective effects of Acod1/itaconate.
    CONCLUSIONS: Our results demonstrated that Acod1/Itaconate axis might protect mouse PMVECs from FFA-induced injury, inflammation and mitochondrial oxidative stress via activating Nrf2 pathway. It was meaningful for the treatment of obesity-caused pulmonary microvascular endotheliopathy.
    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
    最近的研究表明,STS通过抑制肺血管重塑和抑制PASMC异常升高的增殖和迁移,在治疗肺动脉高压中具有有益的作用。然而,STS对肺血管内皮的作用仍是众所周知的。
    在这项研究中,我们利用慢性低氧诱导的肺动脉高压(HPH)大鼠模型研究了STS对肺血管内皮功能障碍的影响和机制。以及在原代培养的大鼠PMVECs和人ESC-ECs细胞模型中。
    首先,STS的21天治疗通过使右心室收缩压和右心室肥大正常化来显著预防HPH的疾病发展,改善心输出量.然后,STS治疗可显着抑制缺氧引起的远端肺内动脉内壁增厚。值得注意的是,STS显著抑制缺氧诱导的HPH大鼠和原代培养的PMVECs肺内皮细胞凋亡,通过稳定BMPR2蛋白和保护减弱的BMP9-BMPR2-Smad1/5/9信号通路。在机制上,STS治疗通过稳定BMPR2蛋白恢复BMPR2的低氧下调,通过溶酶体系统抑制BMPR2蛋白降解,并促进BMPR2的质膜定位,所有这些共同增强了PMVEC和人ESC-EC中BMP9诱导的信号转导。然而,这些效应在表达杂合功能失调BMPR2蛋白(BMPR2+/R899X)的hESC-ECs中不存在。
    STS可能发挥抗凋亡作用,至少部分地,通过诱导肺内皮和PMVECs中的BMP9-BMPR2-Smad1/5/9信号转导。
    Recent studies have demonstrated the beneficial effects of STS in treating pulmonary hypertension by inhibiting the pulmonary vascular remodeling and suppressing the abnormally elevated proliferation and migration of PASMCs. However, the roles of STS on pulmonary vascular endothelium remain largely known.
    In this study, we investigated the effects and mechanisms of STS on pulmonary vascular endothelial dysfunction by using a chronic hypoxia-induced pulmonary hypertension (HPH) rat model, as well as in primarily cultured rat PMVECs and human ESC-ECs cell models.
    Firstly, a 21-day treatment of STS significantly prevents the disease development of HPH by normalizing the right ventricular systolic pressure and right ventricular hypertrophy, improving the cardiac output. Then, STS treatment markedly inhibits the hypoxia-induced medial wall thickening of the distal intrapulmonary arteries. Notably, STS significantly inhibits the hypoxia-induced apoptosis in both the pulmonary endothelium of HPH rats and primarily cultured PMVECs, through the stabilization of BMPR2 protein and protection of the diminished BMP9-BMPR2-Smad1/5/9 signaling pathway. In mechanism, STS treatment retrieves the hypoxic downregulation of BMPR2 by stabilizing the BMPR2 protein, inhibiting the BMPR2 protein degradation via lysosome system, and promoting the plasma membrane localization of BMPR2, all of which together reinforcing the BMP9-induced signaling transduction in both PMVECs and human ESC-ECs. However, these effects are absent in hESC-ECs expressing heterozygous dysfunctional BMPR2 protein (BMPR2+/R899X).
    STS may exert anti-apoptotic roles, at least partially, via induction of the BMP9-BMPR2-Smad1/5/9 signaling transduction in pulmonary endothelium and PMVECs.
    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
    急性肺毁伤(ALI)主要介导肺微血管内皮细胞(PMVECs)的毁伤。LPS是导致ALI微循环异常的致病因素之一。阿魏酸(FA)对各种疾病具有治疗作用。在脂多糖诱导的急性呼吸窘迫综合征期间,FA,如果事先给出,可以抑制炎症和氧化应激。然而,FA在ALI中的具体作用和潜在机制尚未得到很好的表征。使用10μg/mL脂多糖(LPS)治疗大鼠PMVEC24小时。qRT-PCR用于检测miR-17和第10号染色体上缺失的磷酸酶和张力蛋白同源物(PTEN)的水平。Westernblot用于分析PI3K/Akt通路中的相关蛋白,和凋亡相关蛋白。进行流式细胞术分析以检测PMVECs的凋亡。构建MTT法检测细胞活力。荧光素酶法检测miR-17和PTEN的靶基因。使用PMVECs建立了用于体外研究FA在ALI中的作用的细胞模型。我们的数据表明FA上调miR-17并降低LPS诱导的细胞凋亡。FA通过上调miR-17抑制细胞凋亡。此外,我们发现miR-17靶向PTEN呈阴性.FA通过上调miR-17介导的PI3K/Akt途径抑制裂解的caspase-3和Bax表达。与miR-17抑制剂转染的细胞相比,PTEN的过表达可能有助于PI3K/Akt信号通路蛋白的相似表达趋势。FA通过miR-17/PTEN抑制LPS诱导的PMVECs凋亡,进一步调控ALI中PI3K/Akt通路的激活。我们预计我们的数据将引发更多的ALI临床治疗研究。
    Acute lung injury (ALI) is mainly mediated by the damage of pulmonary microvascular endothelial cells (PMVECs). LPS is one of the pathogenic factors leading to microcirculatory abnormalities of ALI. Ferulic acid (FA) exhibits therapeutic effects against various diseases. During lipopolysaccharide-induced acute respiratory distress syndrome, FA, when given beforehand, could depress inflammation and oxidative stress. However, the concrete role and underlying mechanism of FA in ALI have not been well characterized. Ten μg/mL Lipopolysaccharide (LPS) was used to treat rat PMVECs for 24 hr. qRT-PCR was used to detect the level of miR-17 and phosphatase and tensin homolog deleted on chromosome ten (PTEN). Western blot was used to analyze the associated proteins in the PI3K/Akt pathway, and the apoptosis-related proteins. Flow cytometric analysis was performed to detect the apoptosis of PMVECs. MTT assay was constructed to detect the cell viability. Luciferase assay was conducted to detect the target gene of miR-17 and PTEN. A cell model for in vitro studying the role of FA in ALI was established using PMVECs. Our data demonstrate that FA up-regulates miR-17 and declines apoptosis induced by LPS. FA inhibits apoptosis mediated by up-regulating miR-17. Furthermore, we found miR-17 targeted PTEN negatively. FA inhibits cleaved caspase-3 and Bax expression through the PI3K/Akt pathway mediated by up-regulating miR-17. Over-expression of PTEN could contribute to the similar expression trend of the PI3K/Akt signal pathway protein compared to miR-17 inhibitor transfected cells. FA inhibits PMVECs apoptosis induced by LPS via miR-17/PTEN to further regulate the activation of the PI3K/Akt pathway in ALI. We anticipate that our data will provoke additional studies for ALI clinical therapy.
    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
    Inhalation or systemic administration of lipopolysaccharide (LPS) can induce acute pulmonary inflammation and lung injury. The pulmonary vasculature is composed of pulmonary microvascular endothelial cells (PMVECs), which form a semiselective membrane for gas exchange. The miRNA miR-642a-5p has previously been reported to be up-regulated in patients with acute respiratory distress syndrome; thus, here, we examined whether this miRNA is involved in the effects of LPS on PMVECs. The levels of miR-642a-5p and mRNA encoding eukaryotic elongation factor 2 (eEF2) were detected by quantitative RT-PCR. Moesin and eEF2 protein levels were tested by western blot assay. Dual-luciferase reporter assay was used to examine the relationship between miR-642a-5p and eEF2. Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and cell permeability was analyzed using the transendothelial electrical resistance assay. We report that miR-642a-5p levels are significantly up-regulated in LPS-stimulated PMVECs, and miR-642a-5p contributes to LPS-induced hyperpermeability and apoptosis of PMVECs. LPS treatment results in down-regulation of eEF2 in PMVECs. Overexpression of eEF2, a direct target of miR-642a-5p, inhibited the effect of LPS on PMVECs. miR-642a-5p promoted LPS-induced hyperpermeability and apoptosis by targeting eEF2. Thus, miR-642a-5p and eEF2 may serve as potential targets for acute lung injury/acute respiratory distress syndrome diagnosis or treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    坏死是一种新定义的调节坏死形式,并参与各种人类炎症性疾病。目前尚不清楚中暑引起的肺损伤是否存在坏死。我们表明,热应激(HS)以时间依赖性方式触发了受体相互作用蛋白1(RIP1)和混合谱系激酶结构域样蛋白(MLKL)表达的显着上调,受体相互作用蛋白3(RIP3)无明显变化。此外,免疫共沉淀实验表明,RIP1与RIP3结合,在热应激诱导的PMVEC中形成坏死体。体外,necrostatin-1(Nec-1)预处理减少热应激诱导的PMVECs坏死凋亡,这也抑制了HMGB1从细胞核转位到细胞质中。同样,ERK抑制(PD98059),NF-κB(BAY11-7082)和c-Jun(c-Jun肽),分别,还抑制了HMGB1细胞质易位。此外,siRNA介导的RIP1/RIP3敲低通过ERK负调控HS诱导的细胞凋亡中HMGB1的释放,NF-κB,和c-Jun信号通路。我们的研究表明,HS通过MAPK诱导RIP1/RIP3依赖性坏死,NF-κB,和PMVECs中的c-Jun信号通路。
    Necroptosis represents a newly defined form of regulated necrosis and participates in various human inflammatory diseases. It remains unclear whether necroptosis is presented in heatstroke-induced lung injury. We show that heat stress(HS) triggered an significant upregulation of receptor-interacting protein 1 (RIP1) and mixed lineage kinase domain-like protein (MLKL) expression in a time-dependent manner, without a significant change of receptor-interacting protein 3 (RIP3). Furthermore, co-immunoprecipitation assays showed that RIP1 binds to RIP3 to form the necrosome in heat stress-induced PMVECs. In vitro, necrostatin-1 (Nec-1) pre-treatment reduced heat stress-induced PMVECs necroptosis, which also inhibited HMGB1 translocation from the nucleus into the cytoplasm. Similarly, inhibition for ERK (PD98059), NF-κB (BAY11-7082) and c-Jun (c-Jun peptide), respectively, also suppressed the HMGB1 cytoplasm translocation. Furthermore, siRNA-mediated RIP1/RIP3 knockdown negatively regulated the release of HMGB1 in HS-induced necroptosis through the ERK, NF-κB, and c-Jun signaling pathways. Our study reveals that HS induces RIP1/RIP3-dependent necroptosis through the MAPK, NF-κB, and c-Jun signaling pathways in PMVECs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Transfusion-related acute lung injury (TRALI) remains the leading cause of transfusion-related mortality. Endothelium semipermeable barrier function plays a critical role in the pathophysiology of transfusion-related acute lung injury (TRALI). Recently, Roundabout protein 4 (Robo4), interaction with its ligand Slit 2, was appreciated as a modulator of endothelial permeability and integrity. However, not much is known about the role of Slit2/Robo4 signaling pathway in the pathophysiology of TRALI. In this study, the TRALI model was performed by the \"two-event\" model of polymorphonuclear neutrophils (PMN)-mediated pulmonary microvascular endothelial cells (PMVECs) damage. We investigated the expression of Slit2/Robo4 and VE-cadherin and examined the pulmonary endothelial hyper-permeability in TRALI model. We found that the expression of Slit2/Robo4 and VE-cadherin were significantly decreased in a time-dependent manner, whereas the PMVECs permeability was gradually increased over time in TRALI model. Moreover, the treatment with Slit2-N, an active fragment of Slit2, increased the expression of Slit2/Robo4 and VE-cadherin to protect PMVECs from PMN-mediated pulmonary endothelial hyper-permeability. These results indicate that targeting Slit2/Robo4 signaling pathway may modulate the permeability as well as protect the integrity of endothelial barrier. In addition, Slit2-N appears to be a promising candidate for developing novel therapies against TRALI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The endotoxin lipopolysaccharide (LPS)-induced pulmonary endothelial barrier disruption is a key pathogenesis of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). However, the molecular mechanisms underlying LPS-impaired permeability of pulmonary microvascular endothelial cells (PMVECs) are not fully understood.
    Rat PMVECs were isolated and monolayered cultured, then challenged with different doses of LPS (0.1mg/L, 1mg/L, and 10mg/L). Trans-endothelial electrical resistance (TER) was utilized to measure the integrity of the endothelial barrier. Ras-related C3 botulinum toxin substrate 1 (Rac1) activity and the phosphorylation of Ezrin/Radixin/Moesin proteins (ERM) were assessed by pulldown assay and Western Blotting. Small interfering RNA (siRNA) inhibition of Rac1 and Moesin were applied to evaluate the effect of PMVEs permeability and related pathway.
    LPS induced dose and time-dependent decreases in TER and increase in ERM threonine phosphorylation, while inactivated Rac1 activity in PMVEC. siRNA study demonstrated that both Rac1 and Moesin were involved in the mediation of the LPS-induced hyperpermeability in PMVECs monolayers, and Rac1 and Moesin could regulate each other.
    Phosphorylated ERM mediates LPS induced PMVECs permeability through negatively regulating Rac1 activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Acute lung injury (ALI) is a critical complication of the severe acute pancreatitis (SAP), characterized by increased pulmonary permeability with high mortality. Pulmonary microvascular endothelial cells (PMVECs) injury and apoptosis play a key role in ALI. Previous studies indicated that store-operated calcium entry (SOCE) could regulate a variety of cellular processes. The present study was to investigate the effects of SOCE inhibition on ALI induced by SAP in Sprague-Dawley rats, and PMVECs injury induced by lipopolysaccharide (LPS). Rat model of SAP-associated ALI were established by the retrograde infusion of sodium deoxycholate. Serum levels of amylase, TNF-α, and IL-6, histological changes, water content of the lung, oxygenation index, and ultrastructural changes of PMVECs were examined in ALI rats with or without store-operated Ca(2+) channels (SOCs) pharmacological inhibitor (2-aminoethoxydiphenyl borate, 2-APB) pretreatment. For in vitro studies, PMVECs were transiently transfected with or without small interfering RNA (siRNA) against calcium release-activated calcium channel protein1 (Orai1) and stromal interaction molecule1 (STIM1), the two main molecular constituents of SOCs, then exposed to LPS. The viability of PMVECs was determined. The expression of STIM1, Orai1, Bax, and caspase3, both in lung tissue and in PMVECs, were assessed by quantitative real-time PCR and western blot. Administration of sodium deoxycholate upregulated the expression of SOCs proteins in lung tissue. Similarly, the SOCs proteins were increased in PMVECs induced by LPS. 2-APB reduced the serum levels of amylase, TNF-α, and IL-6, and attenuated lung water content and histological findings. In addition, the decreased oxygenation index and ultrastructural damage in PMVECs associated with SAP were ameliorated after administration of 2-APB. Knockdown of STIM1 and Orai1 inhibited LPS-induced PMVECs death. Furthermore, blockade of SOCE significantly suppressed Orai1, STIM1, Bax, and caspase3 expression both in vivo and in vitro. These results suggest that SOCE may play a critical role in SAP-associated ALI and the protective effects of inhibition of SOCs could be mediated, at least partially, by restraining mitochondrial associated apoptosis of PMVECs.
    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