VSMC

VSMC
  • 文章类型: Journal Article
    目的:血管平滑肌细胞(VSMC)可塑性是一种状态,其中VSMC经历从静止收缩表型到其他功能不同表型的表型转换。尽管新出现的证据表明VSMC可塑性在血管疾病的发展中起关键作用,对控制VSMC可塑性和命运的关键决定因素知之甚少。
    结果:我们发现在他莫昔芬诱导的Lkb1flox/flox中,平滑肌细胞特异性缺失Lkb1;Myh11-Cre/ERT2小鼠自发和逐渐诱导主动脉/动脉扩张,动脉瘤,破裂,过早死亡。单细胞RNA测序和基于成像的谱系追踪显示,Lkb1缺陷型VSMC从早期调节的VSMC逐渐转分化为成纤维细胞样和软骨细胞样细胞,导致骨化和血管破裂.机械上,Lkb1调节聚嘧啶束结合蛋白1(Ptbp1)的表达并控制丙酮酸激酶肌肉(PKM)亚型1和2的选择性剪接。VSMC中的Lkb1损失导致PKM2/PKM1比率增加,并通过促进有氧糖酵解改变代谢谱。用PKM2激活剂TEPP-46治疗可挽救Lkb1flox/flox;Myh11-Cre/ERT2小鼠的VSMC转化和主动脉扩张。此外,我们发现与对照组织相比,人主动脉瘤组织中Lkb1的表达降低,随着VSMC命运标志物的变化。
    结论:Lkb1通过调节PKM的Ptbp1依赖性可变剪接,通过抑制VSMC可塑性将VSMC维持在收缩状态。
    OBJECTIVE: Vascular smooth muscle cell (VSMC) plasticity is a state in which VSMCs undergo phenotypic switching from a quiescent contractile phenotype into other functionally distinct phenotypes. Although emerging evidence suggest that VSMC plasticity plays critical roles in the development of vascular diseases, little is known about the key determinant for controlling VSMC plasticity and fate.
    RESULTS: We found that smooth muscle cell-specific deletion of Lkb1 in tamoxifen-inducible Lkb1flox/flox; Myh11-Cre/ERT2 mice spontaneously and progressively induced aortic/arterial dilation, aneurysm, rupture, and premature death. Single-cell RNA sequencing and imaging-based lineage tracing showed that Lkb1-deficient VSMCs transdifferentiated gradually from early modulated VSMCs to fibroblast-like and chondrocyte-like cells, leading to ossification and blood-vessel rupture. Mechanistically, Lkb1 regulates polypyrimidine tract binding protein 1 (Ptbp1) expression and controls alternative splicing of pyruvate kinase muscle (PKM) isoforms 1 and 2. Lkb1 loss in VSMC results in an increased PKM2/PKM1 ratio and alters the metabolic profile by promoting aerobic glycolysis. Treatment with PKM2 activator TEPP-46 rescues VSMC transformation and aortic dilation in Lkb1flox/flox; Myh11-Cre/ERT2 mice. Furthermore, we found that Lkb1 expression decreased in human aortic aneurysm tissue compared to control tissue, along with changes in markers of VSMC fate.
    CONCLUSIONS: Lkb1, via its regulation of Ptbp1-dependent alterative splicing of PKM, maintains VSMC in contractile states by suppressing VSMC plasticity.
    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
    血管平滑肌细胞(VSMC),以其非凡的终生表型可塑性而闻名,通过它们在不同表型之间转换的能力,在血管病变中发挥关键作用。我们小组发现线粒体蛋白Poldip2的缺乏在体内和体外诱导VSMC分化。进一步的全面生化研究揭示了Poldip2与线粒体ATPase酪蛋白分解蛋白酶分子伴侣X亚基(CLPX)的特异性相互作用,它是酪蛋白水解蛋白酶蛋白水解亚基(ClpP)的调节亚基,形成ClpXP复合物的一部分-从细菌到人类进化保守的蛋白酶体样蛋白酶。这种相互作用限制了蛋白酶的活性,和降低Poldip2水平导致ClpXP复杂激活。这一发现提示了线粒体内ClpXP复合物活性可能调节VSMC表型的假设。采用功能增益和功能丧失策略,我们证明ClpXP活性显著影响VSMC表型。值得注意的是,ClpXP的遗传和药理激活都会抑制VSMC的可塑性并促进静止,差异化,和抗炎VSMC表型。使用TIC10的ClpP的药理激活,目前在癌症的III期临床试验中,在弹性蛋白酶诱导的主动脉瘤小鼠模型中,成功地在体外和体内复制了这种表型,并显着减少了动脉瘤的发展。我们的机理探索表明,ClpP激活通过改变细胞NAD/NADH比率和激活Sirtuin1来调节VSMC表型。我们的研究结果揭示了线粒体蛋白稳定在VSMC表型调节中的关键作用,并提出了ClpP蛋白酶作为一种新的,用于操纵VSMC表型的可操作靶标。
    Vascular smooth muscle cells (VSMCs), known for their remarkable lifelong phenotypic plasticity, play a pivotal role in vascular pathologies through their ability to transition between different phenotypes. Our group discovered that the deficiency of the mitochondrial protein Poldip2 induces VSMC differentiation both in vivo and in vitro. Further comprehensive biochemical investigations revealed Poldip2\'s specific interaction with the mitochondrial ATPase caseinolytic protease chaperone subunit X (CLPX), which is the regulatory subunit for the caseinolytic protease proteolytic subunit (ClpP) that forms part of the ClpXP complex - a proteasome-like protease evolutionarily conserved from bacteria to humans. This interaction limits the protease\'s activity, and reduced Poldip2 levels lead to ClpXP complex activation. This finding prompted the hypothesis that ClpXP complex activity within the mitochondria may regulate the VSMC phenotype. Employing gain-of-function and loss-of-function strategies, we demonstrated that ClpXP activity significantly influences the VSMC phenotype. Notably, both genetic and pharmacological activation of ClpXP inhibits VSMC plasticity and fosters a quiescent, differentiated, and anti-inflammatory VSMC phenotype. The pharmacological activation of ClpP using TIC10, currently in phase III clinical trials for cancer, successfully replicates this phenotype both in vitro and in vivo and markedly reduces aneurysm development in a mouse model of elastase-induced aortic aneurysms. Our mechanistic exploration indicates that ClpP activation regulates the VSMC phenotype by modifying the cellular NAD+/NADH ratio and activating Sirtuin 1. Our findings reveal the crucial role of mitochondrial proteostasis in the regulation of the VSMC phenotype and propose the ClpP protease as a novel, actionable target for manipulating the VSMC phenotype.
    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
    血管重塑是血管壁对生理和病理生理变化的适应性反应,与血管疾病密切相关。血管平滑肌细胞(VSMC)在这一过程中起着至关重要的作用。焦亡,一种以过度释放炎症因子为特征的程序性细胞死亡形式,可引起VSMC的表型转化,导致它们的扩散,迁移,和钙化-所有这些加速血管重塑。抑制VSMC焦亡可以延迟该过程。本文综述了焦亡对VSMC的影响以及VSMC焦亡在血管重构中的致病作用。我们还讨论了焦亡途径中关键蛋白的抑制剂及其对VSMC焦亡的影响。这些发现增强了我们对血管重塑的发病机理的理解,并为开发靶向控制VSMC焦亡作为血管疾病潜在治疗策略的新型药物奠定了基础。
    Vascular remodeling is the adaptive response of the vessel wall to physiological and pathophysiological changes, closely linked to vascular diseases. Vascular smooth muscle cells (VSMCs) play a crucial role in this process. Pyroptosis, a form of programmed cell death characterized by excessive release of inflammatory factors, can cause phenotypic transformation of VSMCs, leading to their proliferation, migration, and calcification-all of which accelerate vascular remodeling. Inhibition of VSMC pyroptosis can delay this process. This review summarizes the impact of pyroptosis on VSMCs and the pathogenic role of VSMC pyroptosis in vascular remodeling. We also discuss inhibitors of key proteins in pyroptosis pathways and their effects on VSMC pyroptosis. These findings enhance our understanding of the pathogenesis of vascular remodeling and provide a foundation for the development of novel medications that target the control of VSMC pyroptosis as a potential treatment strategy for vascular diseases.
    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:交感神经支配在调节血管平滑肌细胞(VSMC)表型和血管疾病中起重要作用,但其在腹主动脉瘤(AAA)中的作用尚不清楚。
    目的:本研究旨在探讨交感神经支配在促进AAA发展中的作用及其潜在机制。
    方法:采用蛋白质印迹和免疫化学染色检测交感神经支配过度。我们通过腹腔神经节切除术(CGX)和6-OHDA给药进行了交感神经支配,以了解交感神经支配在AAA中的作用,并通过转录组和功能研究研究了潜在的机制。利用Sema4D敲除(Sema4D-/-)小鼠来确定Sema4D在诱导交感神经支配过度和AAA发育中的参与。
    结果:我们观察到交感神经支配过度,交感神经重塑最重要的形式,在小鼠AAA模型和AAA患者中。CGX或6-OHDA消除交感神经支配能显著抑制AAA的发展和进展。我们进一步揭示了交感神经支配通过释放细胞外ATP(eATP)和激活eATP-P2rx4-p38信号传导促进AAA中的VSMC表型转换。此外,单细胞RNA测序显示,破骨细胞样细胞分泌的Sema4D通过与Plxnb1结合诱导交感神经扩散和神经支配过度。我们一致观察到在Sema4D缺陷小鼠中AAA进展显著改善。
    结论:由破骨细胞样细胞来源的Sema4D驱动的交感神经支配通过激活eATP/P2rx4/p38通路促进VSMC表型转换并加速病理性动脉瘤进展。抑制交感神经支配过度是预防和治疗AAA的潜在新治疗策略。
    BACKGROUND: Sympathetic hyperinnervation plays an important role in modulating the vascular smooth muscle cell (VSMC) phenotype and vascular diseases, but its role in abdominal aortic aneurysm (AAA) is still unknown.
    OBJECTIVE: This study aimed to investigate the role of sympathetic hyperinnervation in promoting AAA development and the underlying mechanism involved.
    METHODS: Western blotting and immunochemical staining were used to detect sympathetic hyperinnervation. We performed sympathetic denervation through coeliac ganglionectomy (CGX) and 6-OHDA administration to understand the role of sympathetic hyperinnervation in AAA and investigated the underlying mechanisms through transcriptome and functional studies. Sema4D knockout (Sema4D-/-) mice were utilized to determine the involvement of Sema4D in inducing sympathetic hyperinnervation and AAA development.
    RESULTS: We observed sympathetic hyperinnervation, the most important form of sympathetic neural remodeling, in both mouse AAA models and AAA patients. Elimination of sympathetic hyperinnervation by CGX or 6-OHDA significantly inhibited AAA development and progression. We further revealed that sympathetic hyperinnervation promoted VSMC phenotypic switching in AAA by releasing extracellular ATP (eATP) and activating eATP-P2rx4-p38 signaling. Moreover, single-cell RNA sequencing revealed that Sema4D secreted by osteoclast-like cells induces sympathetic nerve diffusion and hyperinnervation through binding to Plxnb1. We consistently observed that AAA progression was significantly ameliorated in Sema4D-deficient mice.
    CONCLUSIONS: Sympathetic hyperinnervation driven by osteoclast-like cell-derived Sema4D promotes VSMC phenotypic switching and accelerates pathological aneurysm progression by activating the eATP/P2rx4/p38 pathway. Inhibition of sympathetic hyperinnervation emerges as a potential novel therapeutic strategy for preventing and treating AAA.
    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:血管平滑肌细胞(VSMC)功能失调导致多种疾病,如动脉粥样硬化和损伤后增生。然而,靶向VSMC的抗增殖药物表现出较差的特异性。因此,迫切需要开发高度特异性的抗增殖药物来预防和治疗VSMC去分化相关的动脉硬化。康乐欣(KLX),我们团队设计的一种新的蒽醌化合物,具有根据理化性质调节VSMC表型的潜力。
    目的:该项目旨在评估KLX在VSMC去分化和动脉粥样硬化中的治疗作用,新内膜形成并说明了潜在的分子机制。
    方法:体内,用高脂饮食(HFD)喂养ApoE-/-小鼠,持续13周,建立动脉粥样硬化模型.采用大鼠颈动脉损伤模型建立新生内膜形成模型。体外,PDGF-BB用于诱导VSMC去分化。
    结果:我们发现KLX改善了动脉粥样硬化的进展,包括动脉粥样硬化病变的形成,动脉粥样硬化小鼠模型主动脉和主动脉窦中的脂质沉积和胶原沉积。此外,KLX的给药有效地改善了SD大鼠球囊损伤后颈动脉中的新内膜形成。来自分子对接和表面等离子体共振(SPR)实验的发现明确表明,KLX具有结合PDGFR-β的潜力。研究工作证明,KLX阻止VSMC增殖,通过激活PDGFR-β-MEK-ERK-ELK-1/KLF4信号通路进行迁移和去分化。
    结论:总的来说,我们证明,KLX通过抑制PDGFR-β-MEK-ERK-ELK-1/KLF4信号传导的VSMC表型转化,有效地减弱了ApoE-/-小鼠动脉粥样硬化的进展和SD大鼠颈动脉新内膜的形成。KLX显示出作为治疗VSMC表型转化相关动脉硬化的可行治疗剂的有希望的潜力。
    BACKGROUND: Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties.
    OBJECTIVE: This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism.
    METHODS: In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation.
    RESULTS: We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-β. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-β-MEK -ERK-ELK-1/KLF4 signaling pathway.
    CONCLUSIONS: Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-β-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.
    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
    颈动脉粥样硬化是缺血性卒中的主要原因,缺血性中风是全球发病率和死亡率的主要原因。不幸的是,动脉粥样硬化斑块形成的原因尚不清楚.miRNA-29c可促进糖尿病小鼠血管平滑肌细胞(VSMCs)的表型转化,最终导致斑块形成和出血。然而,这种研究很少见,仅限于动物实验。
    在我们的研究中,根据是否诊断为DM,将40例患者分为糖尿病(DM)组和非DM组。然后,应用实时定量PCR检测来自40例接受颈动脉内膜切除术的受试者的人颈动脉斑块组织中的miRNA-29c水平.
    简而言之,与非DM受试者相比,糖尿病患者的miRNA-29c水平降低,这种比较具有统计学意义(P=0.02)。值得注意的是,可变miRNA-29c水平与HbA1c水平呈负相关,尽管没有观察到统计学意义。此外,卒中患者的miRNA-29c水平升高.
    集体,颈动脉斑块中miRNA-29c水平与DM和脑卒中密切相关,这可能有助于动脉粥样硬化的形成。
    UNASSIGNED: Carotid artery atherosclerosis is a major cause of ischemic stroke, and ischemic stroke is the leading cause of morbidity and mortality worldwide. Unfortunately, the reason for the build-up of atherosclerosis plaque is unknown. The miRNA-29c was reported to promote the phenotype transformation of vascular smooth muscle cells (VSMCs) in diabetes mice, eventually leading to plaque formation and bleeding. However, such studies are rare and limited to animal experiments.
    UNASSIGNED: In our study, 40 patients were divided into a diabetic mellitus (DM) group and a non-DM group according to whether they were diagnosed with DM. Then, the real-time quantitative PCR was applied to examine the miRNA-29c level in human carotid plaque tissue derived from 40 subjects receiving carotid endarterectomy.
    UNASSIGNED: Briefly, diabetes patients had a decreased miRNA-29c level as compared with non-DM subjects, and this comparison was statistically significant (P = 0.02). Notably, variable miRNA-29c level was negatively associated with HbA1c level, although no statistical significance was observed. Moreover, there was an increased miRNA-29c level in patients with cerebral stroke.
    UNASSIGNED: Collectively, the miRNA-29c level in the carotid plaque is closely associated with DM and cerebral stroke, which may contribute to atherosclerosis formation.
    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
    本研究旨在探讨外核苷酸焦磷酸酶/磷酸二酯酶1(ENPP1)对大鼠血管钙化的影响。研究ENPP1在血管钙化中的作用的基本原理在于其调节钙化过程的潜力。了解这种关系可以为解决血管钙化相关疾病的新治疗途径提供见解。在这个实验中,β-甘油磷酸诱导血管平滑肌细胞(VSMC)钙化。随后,将携带ENPP1的重组AAV9导入VSMC以实现ENPP1的体外和体内过表达。结果表明,ENPP1过表达显著降低了主动脉中钙和磷的含量(P<0.05)。茜素红和vonKossa染色显示VSMC和主动脉中钙盐沉积显着减少,分别。值得注意的是,BMP-2,PINP的表达水平,OC,VSMC和BALP显著下降(P<0.05),强调ENPP1在阻碍VSMCs成骨细胞样转分化中的作用。此外,与对照大鼠相比,ENPP1过表达导致焦磷酸盐(PPi)水平显著增加(P<0.05)。总之,本研究提示ENPP1通过升高PPi水平和抑制VSMC的表型转化,有助于缓解血管钙化.这些发现揭示了ENPP1在减轻血管钙化相关并发症方面的潜在治疗作用。
    This study aims to investigate the impact of ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) on vascular calcification in rats. The rationale behind studying ENPP1\'s role in vascular calcification lies in its potential to modulate calcification processes. Understanding this relationship can offer insights into novel therapeutic avenues for addressing vascular calcification-related disorders. In this experiment, vascular smooth muscle cell (VSMC) calcification was induced using β-glycerophosphoric acid. Subsequently, recombinant AAV9-carrying ENPP1 was introduced into VSMCs to achieve both in vitro and in vivo overexpression of ENPP1. The findings indicate that ENPP1 overexpression significantly reduces calcium and phosphorus content in the aorta (P < 0.05). Alizarin red and von Kossa staining reveal notable reductions in calcium salt deposits in VSMCs and aorta, respectively. Notably, the expression levels of BMP-2, PINP, OC, and BALP were substantially decreased in VSMCs (P < 0.05), underscoring ENPP1\'s role in impeding osteoblast-like transdifferentiation of VSMCs. Additionally, ENPP1 overexpression led to a significant increase in pyrophosphate (PPi) levels compared to control rats (P < 0.05). In conclusion, this study suggests that ENPP1 contributes to alleviating vascular calcification by elevating PPi levels and inhibiting the phenotypic transformation of VSMCs. These findings shed light on the potential therapeutic role of ENPP1 in mitigating vascular calcification-related complications.
    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
    放射性血管损伤(RIBV)是放射治疗的一种严重的长期并发症,以动脉粥样硬化的发展为特征。血管平滑肌细胞(VSMC)衰老参与辐射诱导的动脉粥样硬化的发病机制,然而,控制VSMCs衰老的确切机制仍未得到充分理解。在这项研究中,通过使用SA-β-gal染色并评估体内和体外p16和p21的表达来检查VSMC的衰老。我们的发现表明,电离辐射(IR)具有增强细胞衰老的潜力。此外,IR显著激活NF-κB通路,正如p65核易位增加所证明的那样,磷酸化p65表达,和增强p65(EMSA)的结合能力。此外,通过蛋白质印迹分析观察到暴露于IR后HMGB2表达的减少,而CTCF表达保持不变。有趣的是,在超分辨率荧光显微镜下检测到CTCF空间聚类的形成。同时,ChIP技术通过IR鉴定了CTCF与p16基因相互作用的促进作用。CTCF的抑制或通过慢病毒的HMGB2的过表达有效地消除了CTCF簇的形成以及IR后p16和p21的上调。PDTC(100μM)抑制IR诱导的NF-κB激活导致SA-β-gal染色减少,p16表达减少,HMGB2蛋白表达的增加和CTCF簇形成的减少。本研究通过调控NF-κB/CTCF/p16通路,对IR在VSMCs衰老中的作用和机制提供了重要的见解。
    Radiation injury of blood vessels (RIBV) is a serious long-term complication of radiotherapy, characterized by the development of atherosclerosis. The involvement of vascular smooth muscle cells (VSMCs) senescence in the pathogenesis of radiation-induced atherosclerosis has been implicated, yet the precise mechanisms governing VSMCs senescence remain inadequately comprehended. In this study, the senescence of VSMCs was examined by employing SA-β-gal staining and assessing the expression of p16 and p21, both in vivo and in vitro. Our findings revealed that ionizing radiation (IR) has the potential to augment cellular senescence. In addition, IR significantly activated the NF-κB pathway, as evidenced by increased p65 nuclear translocation, phospho-p65 expression, and enhanced binding ability of p65 (EMSA). Furthermore, a decrease in HMGB2 expression following exposure to IR was observed via Western blot analysis, while CTCF expression remained unchanged. Interestingly, the formation of CTCF spatial clustering was detected under super-resolution fluorescence microscopy. Concurrently, the ChIP technique identified the facilitation of the interaction between CTCF and p16 gene through IR. The inhibition of CTCF or the overexpression of HMGB2 through lentiviruses effectively eliminates the formation of CTCF clusters and the upregulation of p16 and p21 after IR. Inhibition of NF-κB activation induced by IR by PDTC (100 μM) led to a decrease in the staining of SA-β-gal, a reduction in p16 expression, an increase in HMGB2 protein expression and a decrease in CTCF clusters formation. This study provided significant insights into the role and mechanism of IR in VSMCs senescence by regulating NF-κB/CTCF/p16 pathway.
    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
    为了应对伤害,动脉壁的血管平滑肌细胞(VSMC)去分化为增殖和迁移表型,导致内膜增生。ERK1/2通路参与细胞增殖和迁移,而双特异性磷酸酶6(DUSP6,也称为MKP3)可以去磷酸化激活的ERK1/2。我们发现DUSP6在正常动脉中表达较低的基线水平;然而,动脉损伤显著增加血管壁中DUSP6的水平。与野生型小鼠相比,Dusp6缺陷小鼠的新内膜较小。体外,IL-1β诱导DUSP6表达并增加VSMC增殖和迁移。缺乏DUSP6减少了IL-1β诱导的VSMC增殖和迁移。DUSP6缺乏不影响IL-1β刺激的ERK1/2激活。相反,ERK1/2抑制剂U0126阻止IL-1β诱导DUSP6,表明ERK1/2在DUSP6的上游起作用以调节VSMC中的DUSP6表达,而不是在下游作为DUSP6底物。IL-1β降低VSMCs中细胞周期抑制剂p27和细胞粘附分子N-cadherin的水平,而缺乏DUSP6保持了高水平,揭示了DUSP6在调节这两种分子中的新功能。一起来看,我们的结果表明,缺乏DUSP6通过减少VSMC增殖和迁移来减弱动脉损伤后的新内膜形成,这可能是通过维持p27和N-cadherin水平介导的。
    In response to injury, vascular smooth muscle cells (VSMCs) of the arterial wall dedifferentiate into a proliferative and migratory phenotype, leading to intimal hyperplasia. The ERK1/2 pathway participates in cellular proliferation and migration, while dual-specificity phosphatase 6 (DUSP6, also named MKP3) can dephosphorylate activated ERK1/2. We showed that DUSP6 was expressed in low baseline levels in normal arteries; however, arterial injury significantly increased DUSP6 levels in the vessel wall. Compared with wild-type mice, Dusp6-deficient mice had smaller neointima. In vitro, IL-1β induced DUSP6 expression and increased VSMC proliferation and migration. Lack of DUSP6 reduced IL-1β-induced VSMC proliferation and migration. DUSP6 deficiency did not affect IL-1β-stimulated ERK1/2 activation. Instead, ERK1/2 inhibitor U0126 prevented DUSP6 induction by IL-1β, indicating that ERK1/2 functions upstream of DUSP6 to regulate DUSP6 expression in VSMCs rather than downstream as a DUSP6 substrate. IL-1β decreased the levels of cell cycle inhibitor p27 and cell-cell adhesion molecule N-cadherin in VSMCs, whereas lack of DUSP6 maintained their high levels, revealing novel functions of DUSP6 in regulating these two molecules. Taken together, our results indicate that lack of DUSP6 attenuated neointima formation following arterial injury by reducing VSMC proliferation and migration, which were likely mediated via maintaining p27 and N-cadherin levels.
    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
    血管平滑肌细胞(VSMC)的巨噬细胞样转化是动脉粥样硬化(AS)进展的危险因素。转录因子同源异型盒A1(HOXA1)在分化和发育中起着功能作用。本研究旨在探讨HOXA1在VSMC转化中的作用,从而为AS发病的潜在机制提供证据。
    将高脂饮食(HFD)喂养的载脂蛋白E基因敲除(ApoE-/-)小鼠作为体内模型来模仿AS,而1-棕榈酰-2-(5-氧代戊酰)-sn-甘油-3-磷酸胆碱(POV-PC)处理的VSMC用作体外模型。产生表达靶向HOXA1的短发夹RNA的重组腺相关病毒-1(AAV-1)载体,本文称为AAV1-shHOXA1,用于整个研究的功能丧失实验。
    在AS小鼠的主动脉根部,脂质沉积严重,HOXA1表达高于正常饮食或HFD喂养的宽型小鼠。沉默HOXA1抑制AS诱导的体重增加,炎症反应,血清和肝脏脂质代谢紊乱和动脉粥样硬化斑块形成。此外,HOXA1敲低AS小鼠的病变显示VSMC向巨噬细胞样细胞的转分化较少,同时抑制krüppel样因子4(KLF4)和核因子(NF)-κBRelA(p65)的表达。体外实验一致证实,HOXA1敲低抑制脂质积累,POV-PC处理的VSMC到巨噬细胞的表型转换和炎症。机制研究进一步表明,HOXA1转录激活RelA和KLF4参与了VSMC的病理表现。
    HOXA1通过调节NF-κBp65和KLF4调节VSMCs可塑性参与AS进展。HOXA1有可能成为AS的生物标志物或治疗靶标。
    Macrophage-like transformation of vascular smooth muscle cells (VSMCs) is a risk factor of atherosclerosis (AS) progression. Transcription factor homeobox A1 (HOXA1) plays functional roles in differentiation and development. This study aims to explore the role of HOXA1 in VSMC transformation, thereby providing evidence for the potential mechanism of AS pathogenesis.
    High fat diet (HFD)-fed apolipoprotein E knockout (ApoE-/-) mice were applied as an in vivo model to imitate AS, while 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POV-PC)-treated VSMCs were applied as an in vitro model. Recombinant adeno-associated-virus-1 (AAV-1) vectors that express short-hairpin RNAs targeting HOXA1, herein referred as AAV1-shHOXA1, were generated for the loss-of-function experiments throughout the study.
    In the aortic root of AS mice, lipid deposition was severer and HOXA1 expression was higher than the wide-type mice fed with normal diet or HFD. Silencing of HOXA1 inhibited the AS-induced weight gain, inflammatory response, serum and liver lipid metabolism disorder and atherosclerotic plaque formation. Besides, lesions from AS mice with HOXA1 knockdown showed less trans-differentiation of VSMCs to macrophage-like cells, along with a suppression of krüppel-like factor 4 (KLF4) and nuclear factor (NF)-κB RelA (p65) expression. In vitro experiments consistently confirmed that HOXA1 knockdown suppressed lipid accumulation, VSMC-to-macrophage phenotypic switch and inflammation in POV-PC-treated VSMCs. Mechanism investigations further illustrated that HOXA1 transcriptionally activated RelA and KLF4 to participate in the pathological manifestations of VSMCs.
    HOXA1 participates in AS progression by regulating VSMCs plasticity via regulation of NF-κB p65 and KLF4. HOXA1 has the potential to be a biomarker or therapeutic target for AS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号