pulmonary artery smooth muscle cells

肺动脉平滑肌细胞
  • 文章类型: Journal Article
    肺动脉高压(PAH)是一种阻塞性血管病变,如果不及时治疗,最终导致右心衰竭.因此,临床前研究需要支持和优化PAH的治疗方法.这里,我们通过调节TRAF6探讨七氟醚在实验性PAH中的预期功能。野百合碱(MCT)诱导的PAH大鼠接受七氟烷吸入和气管内滴注过表达TRAF6的慢病毒。将血小板衍生的生长因子(PDGF)处理的肺动脉平滑肌细胞(PASMC)暴露于七氟醚,并对TRAF6过表达进行遗传操作。发现MCT和PDGF攻击上调大鼠肺组织和PASMC中TRAF6的水平,但七氟醚治疗导致TRAF6表达降低。在MCT诱导的大鼠中吸入七氟醚导致减轻肺血管重塑,减轻右心室功能障碍和肥大,改善线粒体功能和动力学,以及NF-κB通路的失活。体外研究证实暴露于七氟醚抑制PDGF诱导的增殖,迁移,和PASMC的表型转换,并抑制PDGF刺激的PASMC中的线粒体功能障碍和NF-κB激活。TRAF6的过表达逆转了七氟醚对肺组织病理学改变和PASMCs细胞表型的有益影响。总之,我们的研究提示七氟醚通过下调TRAF6表达靶向PAH的保护特性,为PAH的管理提供了一条新颖的途径。
    Pulmonary arterial hypertension (PAH) is an obstructive vasculopathy that, if not promptly treated, culminates in right heart failure. Therefore, pre-clinical studies are needed to support and optimize therapeutic approaches of PAH. Here, we explore a prospective function of sevoflurane in experimental PAH through regulating TRAF6. Monocrotaline (MCT)-induced PAH rats were subjected to sevoflurane inhalation and intratracheal instillation of lentivirus overexpressing TRAF6. Platelet-derived growth factor (PDGF)-treated pulmonary artery smooth muscle cells (PASMCs) were exposed to sevoflurane and genetically manipulated for TRAF6 overexpression. It was found that MCT and PDGF challenge upregulated the levels of TRAF6 in rat lung tissues and PASMCs, but sevoflurane treatment led to reduced TRAF6 expression. Sevoflurane inhalation in MCT-induced rats resulted in alleviative pulmonary vascular remodeling, mitigated right ventricular dysfunction and hypertrophy, improved mitochondrial function and dynamics, and inactivation of NF-κB pathway. In vitro studies confirmed that exposure to sevoflurane repressed PDGF-induced proliferation, migration, and phenotype switching of PASMCs, and suppressed mitochondrial dysfunction and NF-κB activation in PDGF-stimulated PASMCs. The beneficial impact of sevoflurane on pathological changes of lung and cell phenotype of PASMCs were reversed by overexpression of TRAF6. In summary, our study suggested the protective properties of sevoflurane in targeting PAH by downregulating TRAF6 expression, providing a novel avenue for the management of PAH.
    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
    肺动脉高压(PAH)是一种严重的肺血管疾病,其特征是由于血管重塑和血管收缩而导致的肺血管阻力增加。随后,PAH导致右心室肥大和心力衰竭。细胞死亡机制在发育和组织稳态中起着重要作用,调节细胞增殖和分化之间的平衡。一些基础和临床研究表明,细胞死亡的多种机制,包括焦亡,凋亡,自噬,铁性凋亡,Anoikis,Parthanatos,和衰老,与PAH的发病机制密切相关。本文综述了肺动脉平滑肌细胞(PASMCs)和肺动脉内皮细胞(PAECs)死亡的不同细胞死亡机制。PAH中的主要靶细胞。本文综述了这些细胞死亡机制的作用,相关的信号通路,独特的效应分子,和各种促生存或重编程机制。这篇综述的目的是总结目前已知的PAH潜在的分子机制。对细胞死亡机制的进一步研究可能为预防和治疗PAH开辟新的途径。
    Pulmonary arterial hypertension (PAH) is a severe pulmonary vascular disease characterized by increased pulmonary vascular resistance because of vascular remodeling and vasoconstriction. Subsequently, PAH leads to right ventricular hypertrophy and heart failure. Cell death mechanisms play a significant role in development and tissue homeostasis, and regulate the balance between cell proliferation and differentiation. Several basic and clinical studies have demonstrated that multiple mechanisms of cell death, including pyroptosis, apoptosis, autophagy, ferroptosis, anoikis, parthanatos, and senescence, are closely linked with the pathogenesis of PAH. This review summarizes different cell death mechanisms involved in the death of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs), the primary target cells in PAH. This review summarizes the role of these cell death mechanisms, associated signaling pathways, unique effector molecules, and various pro-survival or reprogramming mechanisms. The aim of this review is to summarize the currently known molecular mechanisms underlying PAH. Further investigations of the cell death mechanisms may unravel new avenues for the prevention and treatment of PAH.
    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
    肺动脉高压(PH)是一种持续进行性、无法治愈,以肺血管重塑为特征的多因素相关致命性肺血管疾病。长链非编码RNA(lncRNAs)参与调节病理过程,如肺血管收缩,增厚,重塑,和炎症细胞浸润在PH通过作用于不同的细胞类型。由于它们在PH患者中的差异表达,如观察所证明的,在PH患者中,一些lncRNAs显著上调,而另一些则显著下调,lncRNAs是评估PH患者疾病进展和诊断或预后的潜在有用的生物标志物。本文概述了lncRNAs参与PH发病的不同机制。
    Pulmonary hypertension (PH) is a persistently progressive, incurable, multifactorial associated fatal pulmonary vascular disease characterized by pulmonary vascular remodeling. Long noncoding RNAs (lncRNAs) are involved in regulating pathological processes such as pulmonary vasoconstriction, thickening, remodeling, and inflammatory cell infiltration in PH by acting on different cell types. Because of their differential expression in PH patients, as demonstrated by the observation that some lncRNAs are significantly upregulated while others are significantly downregulated in PH patients, lncRNAs are potentially useful biomarkers for assessing disease progression and diagnosis or prognosis in PH patients. This article provides an overview of the different mechanisms by which lncRNAs are involved in the pathogenesis of PH.
    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
    为了在低氧环境中生存,牦牛通过血管重塑有效避免低氧诱导的肺动脉高压。TGF-β/BMP信号通路在维持肺动脉平滑肌细胞(PASMC)的稳态中起关键作用。然而,TGF-β/BMP信号通路促进牦牛PASMCs增殖的分子调控机制知之甚少。在这项研究中,牦牛PASMCs体外培养,构建低氧模型,探讨TGFβ/BMP信号通路对牦牛PASMC增殖的影响。低氧处理显著增加牦牛PASMCs的增殖。随着缺氧持续时间的增加,TGF-β1的表达水平和Smad2/3的磷酸化水平明显上调。低氧瞬时激活BMP信号通路,随着BMPR2表达和Smad1/5磷酸化的增加,随着低氧暴露时间的延长,这些变化逐渐逆转。此外,外源性TGF-β1激活TGF-β信号通路,提高下游蛋白Smad2和Smad3的磷酸化水平,显著提高牦牛PASMCs的增殖和迁移速率。最后,用noggin(BMP信号的抑制剂)治疗可显着降低BMPR2蛋白表达水平和Smad1/5磷酸化水平,并增加牦牛PASMC的增殖和迁移率。总之,这些结果表明,在缺氧条件下,TGF-β/BMP信号通路的动态调控促进牦牛PASMCs增殖。
    To survive in low-oxygen environments, yaks effectively avoid hypoxia-induced pulmonary arterial hypertension through vascular remodeling. The TGF-β/BMP signaling pathway plays a key role in maintaining the homeostasis of pulmonary artery smooth muscle cells (PASMCs). However, little is known about the molecular regulatory mechanisms by which the TGF-β/BMP signaling pathway contributes to the proliferation of yak PASMCs. In this study, yak PASMCs were cultured in vitro, and a hypoxia model was constructed to investigate the effect of TGFβ/BMP signaling on yak PASMC proliferation. Hypoxia treatment increased the proliferation of yak PASMCs significantly. As the duration of hypoxia increased, the expression levels of TGF-β1 and the phosphorylation levels of Smad2/3 were upregulated significantly. The BMP signaling pathway was transiently activated by hypoxia, with increases in BMPR2 expression and Smad1/5 phosphorylation, and these changes were gradually reversed with prolonged hypoxia exposure. In addition, exogenous TGF-β1 activated the TGF-β signaling pathway, increased the phosphorylation levels of the downstream proteins Smad2 and Smad3, and increased the proliferation and migration rates of yak PASMCs significantly. Finally, treatment with noggin (an inhibitor of BMP signaling) significantly reduced BMPR2 protein expression levels and Smad1/5 phosphorylation levels and increased yak PASMC proliferation and migration rates. In summary, these results revealed that under hypoxic conditions, the dynamic regulation of the TGF-β/BMP signaling pathway promotes the proliferation of yak PASMCs.
    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:低氧性肺血管重构(HPVR)是低氧性肺动脉高压(HPH)的关键病理特征。肺动脉平滑肌细胞(PASMCs)中的氧敏感钾(K)通道在HPVR中起着至关重要的作用。木犀草素(Lut)是一种植物来源的黄酮类化合物,具有多种药理作用。我们先前的研究发现Lut减轻了HPH大鼠的HPVR。
    目的:为了阐明Lut减轻HPVR的机制,关注氧敏感电压依赖性钾通道1.5(Kv1.5)。
    方法:采用低压舱模拟海拔5000m建立HPH大鼠模型。隔离灌注/通气大鼠肺,利用分离的肺小动脉环来研究Lut对K通道活性的影响。评估了HPH大鼠肺组织和肺小动脉中的Kv1.5水平。CyclinD1,CDK4,PCNA,Bax,检测HPH大鼠肺组织中Bcl-2、caspase-3的水平。Lut对Kv1.5,细胞质游离钙浓度([Ca2]cyt)的影响,CyclinD1,CDK4,PCNA,Bax/Bcl-2在缺氧条件下检测PASMCs,DPO-1作为Kv1.5特异性抑制剂。通过药物亲和力响应性靶标稳定性(DARTS)检测PASMC中Lut与Kv1.5之间的结合亲和力。利用KCNA5基因(编码Kv1.5)在HEK293T细胞中的过表达来证实Lut和Kv1.5之间的相互作用。此外,Lut对线粒体结构的影响,SOD,GSH,GSH-Px,测定缺氧条件下HPH大鼠和PASMCs肺组织中MDA和HIF-1α的含量。
    结果:Lut通过直接激活平滑肌中的Kv和Ca2激活的K通道(KCa)来扩张肺动脉。Lut上调了HPH大鼠肺组织和肺小动脉中的Kv1.5水平。Lut下调HPH大鼠肺组织中的CyclinD1,CDK4,PCNA,同时上调Bax/Bcl-2/caspase-3轴。Lut降低了[Ca2+]cyt,减少CDK4,CyclinD1,PCNA,Bax/Bcl-2比值增加,在缺氧条件下的PASMC中,通过上调Kv1.5。在PASMC和HEK293T细胞中验证了Lut和Kv1.5之间的结合亲和力和相互作用。Lut还通过在缺氧下靶向HEK293T细胞的Kv1.5降低[Ca2]cyt并抑制增殖。此外,Lut保护的线粒体结构,增加SOD,GSH,GSH-Px,MDA降低,在HPH大鼠的肺组织中。Lut下调缺氧条件下HPH大鼠和PASMC肺组织HIF-1α水平。
    结论:Lut通过促进肺动脉舒张减轻HPVR,减少细胞增殖,并通过上调PASMC中Kv1.5诱导细胞凋亡。
    BACKGROUND: Hypoxic pulmonary vascular remodeling (HPVR) is a key pathological feature of hypoxic pulmonary hypertension (HPH). Oxygen-sensitive potassium (K+) channels in pulmonary artery smooth muscle cells (PASMCs) play a crucial role in HPVR. Luteolin (Lut) is a plant-derived flavonoid compound with variety of pharmacological actions. Our previous study found Lut alleviated HPVR in HPH rat.
    OBJECTIVE: To elucidate the mechanism by which Lut mitigated HPVR, focusing on oxygen-sensitive voltage-dependent potassium channel 1.5 (Kv1.5).
    METHODS: HPH rat model was established using hypobaric chamber to simulate 5000 m altitude. Isolated perfused/ventilated rat lung, isolated pulmonary arteriole ring was utilized to investigate the impact of Lut on K+ channels activity. Kv1.5 level in lung tissue and pulmonary arteriole of HPH rat was assessed. CyclinD1, CDK4, PCNA, Bax, Bcl-2, cleaved caspase-3 levels in lung tissue of HPH rat were tested. The effect of Lut on Kv1.5, cytoplasmic free calcium concentration ([Ca2+]cyt), CyclinD1, CDK4, PCNA, Bax/Bcl-2 was examined in PASMCs under hypoxia, with DPO-1 as a Kv1.5 specific inhibitor. The binding affinity between Lut and Kv1.5 in PASMCs was detected by drug affinity responsive target stability (DARTS). The overexpression of KCNA5 gene (encoding Kv1.5) in HEK293T cells was utilized to confirm the interaction between Lut and Kv1.5. Furthermore, the impact of Lut on mitochondrial structure, SOD, GSH, GSH-Px, MDA and HIF-1α levels were evaluated in lung tissue of HPH rat and PASMCs under hypoxia.
    RESULTS: Lut dilated pulmonary artery by directly activating Kv and Ca2+-activated K+ channels (KCa) in smooth muscle. Kv1.5 level in lung tissue and pulmonary arteriole of HPH rat was upregulated by Lut. Lut downregulated CyclinD1, CDK4, PCNA while upregulating Bax/Bcl-2/caspase-3 axis in lung tissue of HPH rat. Lut decreased [Ca2+]cyt, reduced CDK4, CyclinD1, PCNA, increased Bax/Bcl-2 ratio, in PASMCs under hypoxia, by upregulating Kv1.5. The binding affinity and the interaction between Lut and Kv1.5 was verified in PASMCs and in HEK293T cells. Lut also decreased [Ca2+]cyt and inhibited proliferation via targeting Kv1.5 of HEK293T cells under hypoxia. Furthermore, Lut protected mitochondrial structure, increased SOD, GSH, GSH-Px, decreased MDA, in lung tissue of HPH rat. Lut downregulated HIF-1α level in both lung tissue of HPH rat and PASMCs under hypoxia.
    CONCLUSIONS: Lut alleviated HPVR by promoting vasodilation of pulmonary artery, reducing cellular proliferation, and inducing apoptosis through upregulating of Kv1.5 in PASMCs.
    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
    肺动脉平滑肌细胞(PASMC)功能与肺动脉高压(PH)的发病机理有关,肺动脉高压是急性肺栓塞(APE)的一种危及生命的并发症。本研究旨在探讨微小RNA(miR)-221-3p在APE-PH患者中的表达模式及其在PASMCs增殖和迁移中的作用。收集APE-PH患者的临床资料及静脉血。检测血清中miR-221-3p和磷酸酶和张力蛋白同源物(PTEN)的表达水平,其次是miR-221-3p诊断效能的受试者特征曲线分析。用miR-221-3p模拟物和PTEN过表达的载体转染PASMC,然后评估细胞活力,扩散,通过细胞计数试剂盒-8,5-乙炔基-2'-脱氧尿苷,Transwell,和伤口愈合试验。miR-221-3p与PTEN3'UTR区之间的结合通过双荧光素酶测定来证明。miR-221在APE-PH患者血清中上调,显示出良好的诊断效能,临界值为1.155,灵敏度66.25%,和67.50%的特异性。miR-221与APE-PH患者PTEN呈负相关。miR-221过表达在体外促进PASMCs增殖和迁移。miR-221-3p结合PTEN3'UTR区以降低PTEN蛋白水平。PTEN过表达取消了miR-221-3p在PASMC中的促进作用。总的来说,miR-221-3p靶向PTEN以促进PASMC增殖和迁移。
    Pulmonary arterial smooth muscle cells (PASMCs) functions are associated with the pathogenesis of pulmonary hypertension (PH) which is a life-threatening complication of acute pulmonary embolism (APE). This study sought to explore the expression pattern of microRNA (miR)-221-3p in APE-PH patients and its role in PASMCs proliferation and migration. The clinical data and venous blood of APE-PH patients were collected. The expression levels of miR-221-3p and phosphatase and tensin homolog (PTEN) in serum were determined, followed by receiver operator characteristic curve analysis of miR-221-3p diagnostic efficacy. PASMCs were transfected with miR-221-3p mimics and PTEN-overexpressed vector, followed by assessment of cell viability, proliferation, and migration through cell counting kit-8, 5-ethynyl-2\'-deoxyuridine, Transwell, and wound healing assays. The binding between miR-221-3p and PTEN 3\'UTR region was testified by the dual-luciferase assay. miR-221 was upregulated in the serum of APE-PH patients and presented with good diagnostic efficacy with 1.155 cutoff value, 66.25% sensitivity, and 67.50% specificity. miR-221 was negatively correlated with PTEN in APE-PH patients. miR-221 overexpression facilitated PASMCs proliferation and migration in vitro. miR-221-3p bound to PTEN 3\'UTR region to decrease PTEN protein levels. PTEN overexpression abolished the promotive role of miR-221-3p in PASMCs. Overall, miR-221-3p targeted PTEN to facilitate PASMC proliferation and migration.
    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
    肺动脉平滑肌细胞(PASMCs)的异常增殖是肺动脉高压(PH)的重要病理机制之一,因此逐渐被采用作为治疗PH的重要方向。据报道,金属硫蛋白(MT)与PH有关,但是潜在的机制还没有完全理解。这里,我们证明了PH患者和慢性缺氧诱导的大鼠和小鼠PH模型的肺小动脉中金属硫蛋白3(MT3)的表达水平显着增加,以及在缺氧处理的人PASMC中。敲除MT3可显著抑制人PASMCs的增殖,将细胞周期阻滞在G1期,而MT3的过表达则有相反的作用。机械上,我们发现MT3通过增加细胞内锌(Zn2+)浓度来增强金属调节转录因子1(MTF1)的转录活性,促进自噬相关基因5(ATG5)的表达,促进自噬体形成。更重要的是,MT3诱导的人PASMCs的自噬和增殖在很大程度上被MTF1和ATG5的敲低所阻止。因此,在这项研究中,我们确定MT3-锌-MTF1-ATG5是一种通过调节自噬体形成影响PASMC增殖的新通路,提示MT3可能是治疗PH的新靶点。
    Abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) is one of the critical pathological mechanisms of pulmonary hypertension (PH), and therefore is gradually being adopted as an important direction for the treatment of PH. Metallothioneins (MTs) have been reported to be associated with PH, but the underlying mechanisms are not fully understood. Here, we demonstrated that the expression level of metallothionein 3 (MT3) was significantly increased in pulmonary arterioles from PH patients and chronic hypoxia-induced rat and mouse PH models, as well as in hypoxia-treated human PASMCs. Knockdown of MT3 significantly inhibited the proliferation of human PASMCs by arresting the cell cycle in the G1 phase, while overexpression of MT3 had the opposite effect. Mechanistically, we found that MT3 increased the intracellular zinc (Zn2+) concentration to enhance the transcriptional activity of metal-regulated transcription factor 1 (MTF1), which promoted the expression of autophagy-related gene 5 (ATG5), facilitating autophagosome formation. More importantly, MT3-induced autophagy and proliferation of human PASMCs were largely prevented by knockdown of MTF1 and ATG5. Therefore, in this study, we identified MT3-Zinc-MTF1-ATG5 as a novel pathway that affects PASMC proliferation by regulating autophagosome formation, suggesting that MT3 may be a novel target for the treatment of PH.
    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
    肺动脉平滑肌细胞(PASMCs)的增殖增加和凋亡减少被认为是肺动脉高压(PAH)的普遍标志,部分与丙酮酸脱氢酶(PDH)活性降低有关,导致葡萄糖的氧化磷酸化减少和有氧糖酵解增加(Warburg效应)。Perhexiline是公认的用于心脏疾病的肉碱棕榈酰转移酶-1(CPT1)抑制剂,反过来增加PDH活性,但与与细胞色素P4502D6(CYP2D6)酶的多态性变异相关的可变药代动力学相关,除非监测血液水平并调整剂量,否则会导致慢代谢者的神经和肝毒性风险。我们先前已经报道了一种新型的全氟辛宁衍生物(FPER-1)具有与全氟辛宁相同的治疗方案,但不被CYP2D6代谢,导致比母体药物更可预测的药代动力学。我们试图研究帕西汀和FPER-1对PAH患者PASMC中PDH通量的影响。我们首先证实PAHPASMCs表现出增加的细胞增殖,AKTSer473,ERK1/2和PDH-E1αSer293的磷酸化增强,表明与健康的PASMC相比具有Warburg效应。使用通己素或FPER-1预处理以浓度依赖性方式显着减弱PAHPASMC的增殖,并抑制AKTSer473的激活,但对ERK途径没有影响。Perhexiline和FPER-1显著激活PDH(被视为PDH-E1αSer293的去磷酸化),糖酵解减少,并通过海马分析检测到这些PAHPASMC中的线粒体呼吸上调。然而,通过caspase3/7活性测定,perhexiline和FPER-1均未诱导细胞凋亡.我们首次表明,perhexiline和FPER-1可以代表减少人PAHPASMC细胞增殖的治疗剂,通过逆转Warburg生理学。
    Increased proliferation and reduced apoptosis of pulmonary artery smooth muscle cells (PASMCs) is recognised as a universal hallmark of pulmonary arterial hypertension (PAH), in part related to the association with reduced pyruvate dehydrogenase (PDH) activity, resulting in decreased oxidative phosphorylation of glucose and increased aerobic glycolysis (Warburg effect). Perhexiline is a well-recognised carnitine palmitoyltransferase-1 (CPT1) inhibitor used in cardiac diseases, which reciprocally increases PDH activity, but is associated with variable pharmacokinetics related to polymorphic variation of the cytochrome P450-2D6 (CYP2D6) enzyme, resulting in the risk of neuro and hepatotoxicity in \'slow metabolisers\' unless blood levels are monitored and dose adjusted. We have previously reported that a novel perhexiline fluorinated derivative (FPER-1) has the same therapeutic profile as perhexiline but is not metabolised by CYP2D6, resulting in more predictable pharmacokinetics than the parent drug. We sought to investigate the effects of perhexiline and FPER-1 on PDH flux in PASMCs from patients with PAH. We first confirmed that PAH PASMCs exhibited increased cell proliferation, enhanced phosphorylation of AKTSer473, ERK 1/2Thr202/Tyr204 and PDH-E1αSer293, indicating a Warburg effect when compared to healthy PASMCs. Pre-treatment with perhexiline or FPER-1 significantly attenuated PAH PASMC proliferation in a concentration-dependent manner and suppressed the activation of the AKTSer473 but had no effect on the ERK pathway. Perhexiline and FPER-1 markedly activated PDH (seen as dephosphorylation of PDH-E1αSer293), reduced glycolysis, and upregulated mitochondrial respiration in these PAH PASMCs as detected by Seahorse analysis. However, both perhexiline and FPER-1 did not induce apoptosis as measured by caspase 3/7 activity. We show for the first time that both perhexiline and FPER-1 may represent therapeutic agents for reducing cell proliferation in human PAH PASMCs, by reversing Warburg physiology.
    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
    暴露于缺氧导致肺动脉高压(PAH)的发展。肺动脉平滑肌细胞(PASMC)中细胞内Ca2浓度([Ca2]i)的增加是肺血管收缩和增殖的主要触发因素。这项研究调查了具有磷酸二酯酶抑制活性的黄嘌呤衍生物KMUP-1的机制,抑制缺氧诱导的典型瞬时受体电位通道1(TRPC1)蛋白过表达,并通过储存操作的钙通道(SOC)调节[Ca2]i。将Sprague-Dawley大鼠的体外PASMC在模块化培养箱中在1%O2/5%CO2下培养24小时,以阐明TRPC1的过表达并观察Ca2的释放和进入。KMUP-1(1μM)抑制缺氧诱导的TRPC家族蛋白编码SOC过表达,特别是TRPC1。蛋白激酶G(PKG)抑制剂KT5823(1μM)和蛋白激酶A(PKA)抑制剂KT5720(1μM)恢复了KMUP-1对TRPC1蛋白的抑制作用。KMUP-1减弱蛋白激酶C(PKC)激活剂佛波醇12-肉豆蔻酸酯13-乙酸酯(PMA,1μM)-上调的TRPC1。我们建议KMUP-1对TRPC1的作用可能涉及激活环磷酸鸟苷(cGMP)/PKG和环磷酸腺苷(cAMP)/PKA途径并抑制PKC途径。我们还使用了Fura2-乙酰氧基甲酯(Fura2-AM,5μM),以测量在thapsigargin(1μM)和硝苯地平(5μM)治疗下,低氧PASMC中从肌浆网(SR)释放的钙和通过SOCs进入的钙。在缺氧条件下,PASMC中的储存操作钙进入(SOCE)活性得到增强,KMUP-1减少了这种活动。总之,在低氧PASMC中,KMUP-1抑制TRPC1蛋白的表达和SOC介导的Ca2进入的活性。
    Exposure to hypoxia results in the development of pulmonary arterial hypertension (PAH). An increase in the intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMCs) is a major trigger for pulmonary vasoconstriction and proliferation. This study investigated the mechanism by which KMUP-1, a xanthine derivative with phosphodiesterase inhibitory activity, inhibits hypoxia-induced canonical transient receptor potential channel 1 (TRPC1) protein overexpression and regulates [Ca2+]i through store-operated calcium channels (SOCs). Ex vivo PASMCs were cultured from Sprague-Dawley rats in a modular incubator chamber under 1% O2/5% CO2 for 24 h to elucidate TRPC1 overexpression and observe the Ca2+ release and entry. KMUP-1 (1 μM) inhibited hypoxia-induced TRPC family protein encoded for SOC overexpression, particularly TRPC1. KMUP-1 inhibition of TRPC1 protein was restored by the protein kinase G (PKG) inhibitor KT5823 (1 μM) and the protein kinase A (PKA) inhibitor KT5720 (1 μM). KMUP-1 attenuated protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA, 1 μM)-upregulated TRPC1. We suggest that the effects of KMUP-1 on TRPC1 might involve activating the cyclic guanosine monophosphate (cGMP)/PKG and cyclic adenosine monophosphate (cAMP)/PKA pathways and inhibiting the PKC pathway. We also used Fura 2-acetoxymethyl ester (Fura 2-AM, 5 μM) to measure the stored calcium release from the sarcoplasmic reticulum (SR) and calcium entry through SOCs in hypoxic PASMCs under treatment with thapsigargin (1 μM) and nifedipine (5 μM). In hypoxic conditions, store-operated calcium entry (SOCE) activity was enhanced in PASMCs, and KMUP-1 diminished this activity. In conclusion, KMUP-1 inhibited the expression of TRPC1 protein and the activity of SOC-mediated Ca2+ entry upon SR Ca2+ depletion in hypoxic PASMCs.
    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:先前的研究表明,中性粒细胞胞外陷阱(NETs)在肺动脉高压(PAH)的发病机制中起关键作用。然而,尚未确定NETs对肺动脉平滑肌细胞(PASMC)影响的具体机制。这项研究的目的是阐明NETs促进PAH进展的潜在机制。
    方法:本研究采用生物信息学分析来筛选与PAH发生和发展相关的潜在分子和机制。这些发现随后在人体样本中得到了验证,含有25个(CCDC25)敲低PASMC的卷曲螺旋结构域,以及野百合碱诱导的PAH大鼠模型。
    结果:NET促进了PASMC的增殖,从而促进PAH的发病机制。这种现象是由跨膜受体CCDC25在PASMC上的激活介导的,随后激活ILK/β-parvin/RAC1通路。因此,PASMC中发生细胞骨架重塑和表型转化。此外,NETs水平可作为PAH严重程度的指标,也可作为缓解PAH的潜在治疗目标.
    结论:这项研究阐明了NETs通过影响PASMC的功能而参与PAH的发病机制。从而突出了它们作为PAH评估和治疗的有希望的目标的潜力。
    BACKGROUND: Previous studies have indicated that neutrophil extracellular traps (NETs) play a pivotal role in pathogenesis of pulmonary arterial hypertension (PAH). However, the specific mechanism underlying the impact of NETs on pulmonary artery smooth muscle cells (PASMCs) has not been determined. The objective of this study was to elucidate underlying mechanisms through which NETs contribute to progression of PAH.
    METHODS: Bioinformatics analysis was employed in this study to screen for potential molecules and mechanisms associated with occurrence and development of PAH. These findings were subsequently validated in human samples, coiled-coil domain containing 25 (CCDC25) knockdown PASMCs, as well as monocrotaline-induced PAH rat model.
    RESULTS: NETs promoted proliferation of PASMCs, thereby facilitating pathogenesis of PAH. This phenomenon was mediated by the activation of transmembrane receptor CCDC25 on PASMCs, which subsequently activated ILK/β-parvin/RAC1 pathway. Consequently, cytoskeletal remodeling and phenotypic transformation occur in PASMCs. Furthermore, the level of NETs could serve as an indicator of PAH severity and as potential therapeutic target for alleviating PAH.
    CONCLUSIONS: This study elucidated the involvement of NETs in pathogenesis of PAH through their influence on the function of PASMCs, thereby highlighting their potential as promising targets for the evaluation and treatment of PAH.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号