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)是一种常见的血管疾病,肺血管重构是PAH的重要病理生理机制。肺动脉重塑的主要病理变化,包括扩散,肥大和分泌活动增强,可发生于肺动脉平滑肌细胞(PASMC)。多种活性因子和细胞因子在PAH中起重要作用。然而,PAH中的活性因子和细胞因子的调节机制尚不清楚。本研究旨在揭示PASMC焦亡在PAH中的关键作用,并阐明其内在机制。建立PAH大鼠模型,向Sprague-Dawley大鼠腹膜内注射60mg/kg剂量的野百合碱(MCT)。蛋白质印迹法和ELISA法检测蛋白质和白细胞介素的表达。结果表明,在MCT诱导的PAH大鼠中,PASMCs的焦亡明显增加。值得注意的是,PASMC可分泌IL-1β和IL-18促进PASMC的增殖。在此基础上,抑制IL-1β和IL-18的分泌可以明显抑制PASMC的增殖。总的来说,本研究的结果表明,在MCT诱导的PAH大鼠中,PASMC焦亡具有关键作用,提示PAH的新预防和治疗策略。
    Pulmonary arterial hypertension (PAH) is a common vascular disease, and pulmonary vascular remodeling is a pivotal pathophysiological mechanism of PAH. Major pathological changes of pulmonary arterial remodeling, including proliferation, hypertrophy and enhanced secretory activity, can occur in pulmonary artery smooth muscle cells (PASMCs). Multiple active factors and cytokines play important roles in PAH. However, the regulatory mechanisms of the active factors and cytokines in PAH remain unclear. The present study aimed to reveal the crucial role of PASMC pyroptosis in PAH and to elucidate the intrinsic mechanisms. To establish the PAH rat models, Sprague-Dawley rats were injected intraperitoneally with monocrotaline (MCT) at a dose of 60 mg/kg. The expression of proteins and interleukins were detected by western blotting and ELISA assay. The results indicated that the pyroptosis of PASMCs is significantly increased in MCT-induced PAH rats. Notably, pyroptotic PASMCs can secret IL-1β and IL-18 to promote the proliferation of PASMCs. On this basis, inhibiting the secretion of IL-1β and IL-18 can markedly inhibit PASMC proliferation. Collectively, the findings of the present study indicate a critical role for PASMC pyroptosis in MCT-induced PAH rats, prompting a new preventive and therapeutic strategy for 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(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
    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
    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)

  • 文章类型: Journal Article
    肺动脉高压(PAH)引起肺血管重构,增加肺血管阻力(PVR)并导致右心衰竭和死亡。疾病早期的基质硬化促进肺动脉平滑肌细胞(PASMC)的重塑,有助于PAH的发病机制。我们的研究确定YAP和TAZ是PASMC机械生物学反馈回路的关键驱动因素,表明靶向它们可以减轻重塑。然而,YAP/TAZ广泛表达并执行多种功能,需要细胞特异性方法。我们先前的工作表明,靶向非规范IKB激酶TBK1降低了人肺成纤维细胞中YAP/TAZ的激活。这里,我们研究了非经典IKB激酶TBK1和IKKε在肺动脉高压(PH)中的作用,以及它们通过调节YAP/TAZ调节PASMC致病性重塑的潜力。我们显示,在大鼠PH模型中,TBK1和IKKε在PASMC中被激活。炎性细胞因子,在PAH中升高,激活人类PASMC中的这些激酶。抑制TBK1/IKKε表达/活性显著降低PAH相关的PASMC重塑,对YAP/TAZ的影响比曲前列环素更持久,批准的PAH疗法。这些结果表明,非经典IKB激酶可调节PASMC中的YAP/TAZ,并可能为减少PAH中的血管重塑提供新方法。
    Pulmonary arterial hypertension (PAH) causes pulmonary vascular remodeling, increasing pulmonary vascular resistance (PVR) and leading to right heart failure and death. Matrix stiffening early in the disease promotes remodeling in pulmonary artery smooth muscle cells (PASMCs), contributing to PAH pathogenesis. Our research identified YAP and TAZ as key drivers of the mechanobiological feedback loop in PASMCs, suggesting targeting them could mitigate remodeling. However, YAP/TAZ are ubiquitously expressed and carry out diverse functions, necessitating a cell-specific approach. Our previous work demonstrated that targeting non-canonical IKB kinase TBK1 reduced YAP/TAZ activation in human lung fibroblasts. Here, we investigate non-canonical IKB kinases TBK1 and IKKε in pulmonary hypertension (PH) and their potential to modulate PASMC pathogenic remodeling by regulating YAP/TAZ. We show that TBK1 and IKKε are activated in PASMCs in a rat PH model. Inflammatory cytokines, elevated in PAH, activate these kinases in human PASMCs. Inhibiting TBK1/IKKε expression/activity significantly reduces PAH-associated PASMC remodeling, with longer-lasting effects on YAP/TAZ than treprostinil, an approved PAH therapy. These results show that non-canonical IKB kinases regulate YAP/TAZ in PASMCs and may offer a novel approach for reducing vascular remodeling in 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
    N6-甲基腺苷(m6A)是转录后表观遗传变化,具有转录稳定性和功能,受特定m6A修饰酶调节。然而,在肺动脉高压(PAH)的背景下,m6A修饰的基因和m6A调控特异性酶的意义仍未被研究.应用MeRIP-seq和RNA-seq来探索对照和野百合碱诱导的PAH大鼠的肺动脉组织内m6A和RNA表达的差异。使用基因本体论和京都基因和基因组百科全书分析功能富集。要筛选候选m6A相关基因,使用STRING和Metascape数据库构建蛋白质-蛋白质相互作用网络,然后对其表达进行实时PCR验证.使用免疫组织化学染色进一步研究m6A调节剂的表达水平,免疫荧光,和蛋白质印迹技术。此外,对原代大鼠肺动脉平滑肌细胞(PASMC)进行增殖测定。我们鉴定了42个差异表达基因,这些基因表现出高甲基化或低甲基化的m6A。这些基因主要与细胞外基质结构有关,MAPK,和PI3K/AKT途径。一个候选基因,着丝粒蛋白F(CENPF),在PAH组中检测到表达增加。此外,我们首先确定了一个M6A阅读器,富含亮氨酸的五肽重复序列(LRPPRC),在PAH大鼠模型中下调。siRNA介导的Lrpprc的体外下调导致原代大鼠PASMC中CenpfmRNA的增殖增强和表达升高。我们的研究揭示了PAH大鼠肺动脉中修饰的全转录组m6A景观和相关调节机制,可能为未来的治疗策略提供新的目标。
    N6-methyladenosine (m6A) is a post-transcriptional epigenetic change with transcriptional stability and functionality regulated by specific m6A-modifying enzymes. However, the significance of genes modified by m6A and enzymes specific to m6A regulation in the context of pulmonary arterial hypertension (PAH) remains largely unexplored. MeRIP-seq and RNA-seq were applied to explore variances in m6A and RNA expression within the pulmonary artery tissues of control and monocrotaline-induced PAH rats. Functional enrichments were analyzed using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes. To screen candidate m6A-related genes, the STRING and Metascape databases were used to construct a protein-protein interaction network followed by a real-time PCR validation of their expression. The expression level of an m6A regulator was further investigated using immunohistochemical staining, immunofluorescence, and Western blot techniques. Additionally, proliferation assays were conducted on primary rat pulmonary artery smooth muscle cells (PASMCs). We identified forty-two differentially expressed genes that exhibited either hypermethylated or hypomethylated m6A. These genes are predominantly related to the extracellular matrix structure, MAPK, and PI3K/AKT pathways. A candidate gene, centromere protein F (CENPF), was detected with increased expression in the PAH group. Additionally, we first identified an m6A reader, leucine rich pentatricopeptide repeat containing (LRPPRC), which was downregulated in the PAH rat model. The in vitro downregulation of Lrpprc mediated by siRNA resulted in the enhanced proliferation and elevated expression of Cenpf mRNA in primary rat PASMCs. Our study revealed a modified transcriptome-wide m6A landscape and associated regulatory mechanisms in the pulmonary arteries of PAH rats, potentially offering a novel target for therapeutic strategies in the future.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号