Neointima

新内膜
  • 文章类型: Journal Article
    血管平滑肌细胞(VSMC)的增殖和迁移能力在血管损伤后的新内膜形成中起着至关重要的作用。Skp2通过细胞周期调节促进细胞的增殖和迁移,提出了动脉粥样硬化的重要治疗靶点,肺动脉高压,血管再狭窄.这项研究旨在鉴定能够抑制血管损伤后新内膜形成的天然产物。这里,我们证明了特洛克鲁丁,一种类黄酮,显著降低VSMC中的生存力和下调Skp2。此外,曲克鲁丁对VSMC具有抗增殖作用,并减轻了新内膜的形成。这些发现共同阐明了曲克鲁丁治疗动脉粥样硬化的内在机制,肺动脉高压,通过靶向E3连接酶Skp2和血管再狭窄。
    The proliferative and migratory abilities of vascular smooth muscle cells (VSMCs) play a crucial role in neointima formation following vascular injury. Skp2 facilitates proliferation and migration in cells through cell cycle regulation, presenting an important therapeutic target for atherosclerosis, pulmonary hypertension, and vascular restenosis. This study aimed to identify a natural product capable of inhibiting neointima formation post vascular injury. Here, we demonstrate that troxerutin, a flavonoid, significantly reduced viability and downregulated Skp2 in VSMCs. Moreover, troxerutin exhibited anti-proliferative effects on VSMCs and mitigated neointima formation. These findings collectively elucidate the intrinsic mechanism of troxerutin in treating atherosclerosis, pulmonary hypertension, and vascular restenosis by targeting the E3-linked enzyme Skp2.
    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
    冠状动脉旁路移植术中使用的常见动脉移植物包括胸廓内动脉(ITA),桡动脉(RA)和右胃表皮动脉(RGA)移植物;其中,ITA具有最好的临床结果。这里,通过分析不同动脉移植物的单细胞转录组,我们建议以ITA为参考的RA和RGA的优化策略。与ITA相比,RA有更多的脂质处理相关的CD36+内皮细胞.来自RGA的血管平滑肌细胞更容易发生痉挛,其次是RA;与ITA比较表明钾通道开放剂可以抵消血管痉挛。来自RA和RGA的成纤维细胞分别高度表达GDF10和CREB5;GDF10和CREB5均与细胞外基质沉积相关。细胞-细胞通讯分析显示RA中高水平的巨噬细胞迁移抑制因子信号传导。对部分颈动脉结扎的小鼠施用巨噬细胞移动抑制因子抑制剂可阻断由血流紊乱引起的新生内膜增生。确定的目标的调节可能对动脉移植物具有保护作用。
    Common arterial grafts used in coronary artery bypass grafting include internal thoracic artery (ITA), radial artery (RA) and right gastroepiploic artery (RGA) grafts; of these, the ITA has the best clinical outcome. Here, by analyzing the single-cell transcriptome of different arterial grafts, we suggest optimization strategies for the RA and RGA based on the ITA as a reference. Compared with the ITA, the RA had more lipid-handling-related CD36+ endothelial cells. Vascular smooth muscle cells from the RGA were more susceptible to spasm, followed by those from the RA; comparison with the ITA suggested that potassium channel openers may counteract vasospasm. Fibroblasts from the RA and RGA highly expressed GDF10 and CREB5, respectively; both GDF10 and CREB5 are associated with extracellular matrix deposition. Cell-cell communication analysis revealed high levels of macrophage migration inhibitory factor signaling in the RA. Administration of macrophage migration inhibitory factor inhibitor to mice with partial carotid artery ligation blocked neointimal hyperplasia induced by disturbed flow. Modulation of identified targets may have protective effects on arterial grafts.
    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
    平滑肌细胞(SMC)表型调制,主要由PDGFRβ信号驱动,与闭塞性心血管疾病有关。然而,PDGFRβ的促进和限制性调节机制以及蛋白酪氨酸磷酸酶非受体14(PTPN14)在新生内膜增生中的作用尚不清楚。我们的研究观察到新内膜增生期间SMC中PTPN14的显着上调。PTPN14过表达以磷酸酶活性依赖性方式加剧新生内膜增生,而SMC特异性PTPN14缺乏减轻小鼠的这一过程。RNA-seq表明PTPN14缺陷抑制PDGFRβ信号传导诱导的SMC表型调节。此外,PTPN14与PDGFRβ的胞内区域相互作用并介导其在Y692位点上的去磷酸化。PDGFRβY692的磷酸化负调控PDGFRβ信号传导激活。PTPN14和磷酸-PDGFRβY692的水平与人冠状动脉的狭窄程度相关。我们的发现表明PTPN14是SMC的关键调节剂,促进新生内膜增生。由PTPN14去磷酸化的PDGFRβY692作为控制PDGFRβ活化的自我抑制位点。
    Smooth muscle cell (SMC) phenotypic modulation, primarily driven by PDGFRβ signaling, is implicated in occlusive cardiovascular diseases. However, the promotive and restrictive regulation mechanism of PDGFRβ and the role of protein tyrosine phosphatase non-receptor type 14 (PTPN14) in neointimal hyperplasia remain unclear. Our study observes a marked upregulation of PTPN14 in SMCs during neointimal hyperplasia. PTPN14 overexpression exacerbates neointimal hyperplasia in a phosphatase activity-dependent manner, while SMC-specific deficiency of PTPN14 mitigates this process in mice. RNA-seq indicates that PTPN14 deficiency inhibits PDGFRβ signaling-induced SMC phenotypic modulation. Moreover, PTPN14 interacts with intracellular region of PDGFRβ and mediates its dephosphorylation on Y692 site. Phosphorylation of PDGFRβY692 negatively regulates PDGFRβ signaling activation. The levels of both PTPN14 and phospho-PDGFRβY692 are correlated with the degree of stenosis in human coronary arteries. Our findings suggest that PTPN14 serves as a critical modulator of SMCs, promoting neointimal hyperplasia. PDGFRβY692, dephosphorylated by PTPN14, acts as a self-inhibitory site for controlling PDGFRβ activation.
    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
    血清反应因子(SRF)控制血管平滑肌细胞(VSMC)中的基因转录,并调节VSMC从收缩状态到合成状态的表型转换,在心血管疾病(CVD)的发病机制中起着关键作用。不知道翻译后SUMO化如何调节CVD中的SRF活性。在这里,我们显示,在VSMCSenp1缺乏增加SUMOylatedSRF和SRF-ELK复合物,导致小鼠血管重塑和新内膜形成增强。机械上,VSMC中的SENP1缺乏会增加赖氨酸143处的SRFSUMO化,从而减少SRF溶酶体定位,同时增加核积累,并将收缩表型响应性SRF-myocardin复合物转换为合成表型响应性SRF-ELK1复合物。来自CVD患者冠状动脉的VSMC中SUMO化SRF和磷酸-ELK1增加。重要的是,ELK抑制剂AZD6244可防止SRF-myocardin转变为SRF-ELK复合物,在Senp1缺陷小鼠中减弱VSMC合成表型和新内膜形成。因此,靶向SRF复合物可能具有治疗CVD的治疗潜力.
    Serum response factor (SRF) controls gene transcription in vascular smooth muscle cells (VSMCs) and regulates VSMC phenotypic switch from a contractile to a synthetic state, which plays a key role in the pathogenesis of cardiovascular diseases (CVD). It is not known how post-translational SUMOylation regulates the SRF activity in CVD. Here we show that Senp1 deficiency in VSMCs increased SUMOylated SRF and the SRF-ELK complex, leading to augmented vascular remodeling and neointimal formation in mice. Mechanistically, SENP1 deficiency in VSMCs increases SRF SUMOylation at lysine 143, reducing SRF lysosomal localization concomitant with increased nuclear accumulation and switching a contractile phenotype-responsive SRF-myocardin complex to a synthetic phenotype-responsive SRF-ELK1 complex. SUMOylated SRF and phospho-ELK1 are increased in VSMCs from coronary arteries of CVD patients. Importantly, ELK inhibitor AZD6244 prevents the shift from SRF-myocardin to SRF-ELK complex, attenuating VSMC synthetic phenotypes and neointimal formation in Senp1-deficient mice. Therefore, targeting the SRF complex may have a therapeutic potential for the treatment of CVD.
    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:血管平滑肌细胞(VSMC)增殖与许多类型的动脉疾病有关,包括新内膜增生,其中Ca2+被认为是关键角色。然而,内质网通过肌醇1,4,5-三磷酸受体(IP3Rs)释放Ca2在调节VSMC增殖中的生理作用尚未得到很好的确定。
    结果:建立了体外细胞培养模型和体内小鼠模型,以研究IP3R在调节VSMC增殖中的作用。在血小板衍生的生长因子-BB和FBS刺激后,培养的VSMC以及血管闭塞后经历内膜增厚的左颈动脉中,所有3种IP3R亚型的表达均增加。所有3种IP3R亚型的遗传消融消除了培养的VSMCs中的内质网Ca2释放,血小板源性生长因子-BB和FBS刺激诱导的细胞增殖显着降低,并降低了VSMC的细胞迁移。此外,成年小鼠中所有IP3R亚型的平滑肌特异性缺失可显着减弱左颈动脉结扎诱导的新内膜形成,伴随着损伤血管中细胞增殖和基质金属蛋白酶-9表达的显着降低。机械上,IP3R介导的Ca2+释放可激活cAMP反应元件结合蛋白,控制VSMC扩散的关键角色,通过Ca2+/钙调蛋白依赖性蛋白激酶II和Akt。IP3R的丢失抑制了培养的VSMC和损伤血管中Ser133的cAMP反应元件结合蛋白磷酸化,而Ca2+可渗透的离子载体的应用,离子霉素,可以逆转IP3R三重敲除VSMC中cAMP反应元件结合蛋白的磷酸化。
    结论:我们的结果证明了IP3R介导的内质网释放Ca2+在调节cAMP反应元件结合蛋白激活中的重要作用,VSMC增殖,和小鼠动脉中的新内膜形成。
    BACKGROUND: Vascular smooth muscle cell (VSMC) proliferation is involved in many types of arterial diseases, including neointima hyperplasia, in which Ca2+ has been recognized as a key player. However, the physiological role of Ca2+ release via inositol 1,4,5-trisphosphate receptors (IP3Rs) from endoplasmic reticulum in regulating VSMC proliferation has not been well determined.
    RESULTS: Both in vitro cell culture models and in vivo mouse models were generated to investigate the role of IP3Rs in regulating VSMC proliferation. Expression of all 3 IP3R subtypes was increased in cultured VSMCs upon platelet-derived growth factor-BB and FBS stimulation as well as in the left carotid artery undergoing intimal thickening after vascular occlusion. Genetic ablation of all 3 IP3R subtypes abolished endoplasmic reticulum Ca2+ release in cultured VSMCs, significantly reduced cell proliferation induced by platelet-derived growth factor-BB and FBS stimulation, and also decreased cell migration of VSMCs. Furthermore, smooth muscle-specific deletion of all IP3R subtypes in adult mice dramatically attenuated neointima formation induced by left carotid artery ligation, accompanied by significant decreases in cell proliferation and matrix metalloproteinase-9 expression in injured vessels. Mechanistically, IP3R-mediated Ca2+ release may activate cAMP response element-binding protein, a key player in controlling VSMC proliferation, via Ca2+/calmodulin-dependent protein kinase II and Akt. Loss of IP3Rs suppressed cAMP response element-binding protein phosphorylation at Ser133 in both cultured VSMCs and injured vessels, whereas application of Ca2+ permeable ionophore, ionomycin, can reverse cAMP response element-binding protein phosphorylation in IP3R triple knockout VSMCs.
    CONCLUSIONS: Our results demonstrated an essential role of IP3R-mediated Ca2+ release from endoplasmic reticulum in regulating cAMP response element-binding protein activation, VSMC proliferation, and neointima formation in mouse arteries.
    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:血管闭塞患者经皮血管成形术后经常出现再狭窄,严重威胁其健康。大量证据表明,使用药物洗脱支架预防血管平滑肌细胞增殖是改善再狭窄的有效方法。已经证明葫芦素在各种肿瘤中发挥抗增殖作用和降压作用。本研究旨在探讨从甜瓜中提取的葫芦素(CuECs)和葫芦素B(CuB)对再狭窄的作用。
    方法:C57BL/6小鼠进行左颈动脉结扎并皮下注射CuECs或CuB4周。苏木精-伊红,免疫荧光和免疫组织化学染色用于评估CuECs和CuB对新生内膜增生的影响。蛋白质印迹,实时PCR,流式细胞术分析,采用EdU染色和细胞免疫荧光法检测CuECs和CuB对细胞增殖和细胞周期的影响。CuEC与细胞周期蛋白A2的潜在相互作用通过分子对接进行。
    结果:结果表明,CuECs和CuB均对血管平滑肌细胞的新生内膜增生和增殖具有明显的抑制作用。此外,CuECs和CuB介导的细胞周期阻滞在S期。自动对接分析表明,CuB,CuD,CuE和CuI对细胞周期蛋白A2具有高结合能。我们的研究还显示CuECs和CuB显著抑制FBS诱导的细胞周期蛋白A2表达。此外,cyclinA2在CuEC和CuB处理的新内膜中的表达下调。
    结论:CuEC,尤其是CuB,在VSMC中发挥抗增殖作用,可能是预防再狭窄的潜在药物。
    BACKGROUND: Restenosis frequently occurs after percutaneous angioplasty in patients with vascular occlusion and seriously threatens their health. Substantial evidence has revealed that preventing vascular smooth muscle cell proliferation using a drug-eluting stent is an effective approach to improve restenosis. Cucurbitacins have been demonstrated to exert an anti-proliferation effect in various tumors and a hypotensive effect. This study aims to investigate the role of cucurbitacins extracted from Cucumis melo L. (CuECs) and cucurbitacin B (CuB) on restenosis.
    METHODS: C57BL/6 mice were subjected to left carotid artery ligation and subcutaneously injected with CuECs or CuB for 4 weeks. Hematoxylin-Eosin, immunofluorescence and immunohistochemistry staining were used to evaluate the effect of CuECs and CuB on neointimal hyperplasia. Western blot, real-time PCR, flow cytometry analysis, EdU staining and cellular immunofluorescence assay were employed to measure the effects of CuECs and CuB on cell proliferation and the cell cycle in vitro. The potential interactions of CuECs with cyclin A2 were performed by molecular docking.
    RESULTS: The results demonstrated that both CuECs and CuB exhibited significant inhibitory effects on neointimal hyperplasia and proliferation of vascular smooth muscle cells. Furthermore, CuECs and CuB mediated cell cycle arrest at the S phase. Autodocking analysis demonstrated that CuB, CuD, CuE and CuI had high binding energy for cyclin A2. Our study also showed that CuECs and CuB dramatically inhibited FBS-induced cyclin A2 expression. Moreover, the expression of cyclin A2 in CuEC- and CuB-treated neointima was downregulated.
    CONCLUSIONS: CuECs, especially CuB, exert an anti-proliferation effect in VSMCs and may be potential drugs to prevent restenosis.
    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)是高度可塑的。血管损伤诱导从分化到去分化的VSMC的表型转化,这涉及收缩蛋白的表达减少和细胞外基质和炎性细胞因子的产生增加。这种转变在动脉粥样硬化等多种心血管疾病中起着重要作用。高血压,和主动脉瘤。TGF-β(转化生长因子-β)对于VSMC分化和平衡去分化因子的作用至关重要。然而,在体内条件下控制TGF-β活性和VSMC表型调节的机制知之甚少。最近已显示细胞外基质蛋白TN-X(生腱蛋白-X)结合TGF-β并阻止其激活其受体。
    我们使用他莫昔芬诱导的SMC特异性敲除和腺相关病毒介导的敲除研究了TN-X在各种鼠疾病模型中的VSMC中的作用。
    在高血压和高脂肪饮食的小鼠中,在颈动脉结扎以及人类动脉瘤主动脉后,Tnxb的表达,编码TN-X的基因,在VSMC中增加了。平滑肌细胞特异性丢失TN-X(SMC-Tnxb-KO)的小鼠显示VSMC中TGF-β信号传导增加,与对照组相比,血管重塑过程中VSMC分化标记基因的表达也上调。SMC特异性TN-X缺乏减少了颈动脉结扎后的新内膜形成,并减少了AngII(血管紧张素II)引起的高血压期间的血管壁增厚。缺乏ApoE的SMC-Tnxb-KO小鼠在高脂饮食下显示出减少的动脉粥样硬化和AngII诱导的动脉瘤形成。针对Tnxb的短发夹RNA的腺相关病毒介导的SMC特异性表达显示出类似的有益效果。用抗TGF-β抗体或额外的SMC特异性TGF-β受体缺失的治疗逆转了SMC特异性TN-X缺乏的作用。
    总之,TN-X通过抑制TGF-β信号传导在血管损伤期间关键调节VSMC可塑性。我们的数据表明,抑制血管平滑肌TN-X可能代表预防和治疗病理性血管重塑的策略。
    UNASSIGNED: Vascular smooth muscle cells (VSMCs) are highly plastic. Vessel injury induces a phenotypic transformation from differentiated to dedifferentiated VSMCs, which involves reduced expression of contractile proteins and increased production of extracellular matrix and inflammatory cytokines. This transition plays an important role in several cardiovascular diseases such as atherosclerosis, hypertension, and aortic aneurysm. TGF-β (transforming growth factor-β) is critical for VSMC differentiation and to counterbalance the effect of dedifferentiating factors. However, the mechanisms controlling TGF-β activity and VSMC phenotypic regulation under in vivo conditions are poorly understood. The extracellular matrix protein TN-X (tenascin-X) has recently been shown to bind TGF-β and to prevent it from activating its receptor.
    UNASSIGNED: We studied the role of TN-X in VSMCs in various murine disease models using tamoxifen-inducible SMC-specific knockout and adeno-associated virus-mediated knockdown.
    UNASSIGNED: In hypertensive and high-fat diet-fed mice, after carotid artery ligation as well as in human aneurysmal aortae, expression of Tnxb, the gene encoding TN-X, was increased in VSMCs. Mice with smooth muscle cell-specific loss of TN-X (SMC-Tnxb-KO) showed increased TGF-β signaling in VSMCs, as well as upregulated expression of VSMC differentiation marker genes during vascular remodeling compared with controls. SMC-specific TN-X deficiency decreased neointima formation after carotid artery ligation and reduced vessel wall thickening during Ang II (angiotensin II)-induced hypertension. SMC-Tnxb-KO mice lacking ApoE showed reduced atherosclerosis and Ang II-induced aneurysm formation under high-fat diet. Adeno-associated virus-mediated SMC-specific expression of short hairpin RNA against Tnxb showed similar beneficial effects. Treatment with an anti-TGF-β antibody or additional SMC-specific loss of the TGF-β receptor reverted the effects of SMC-specific TN-X deficiency.
    UNASSIGNED: In summary, TN-X critically regulates VSMC plasticity during vascular injury by inhibiting TGF-β signaling. Our data indicate that inhibition of vascular smooth muscle TN-X may represent a strategy to prevent and treat pathological vascular remodeling.
    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:内皮祖细胞(EPCs)的移植已被证明可以减少动脉损伤后的新生内膜增生。然而,这种方法的有效性受到有限的EPC归巢到损伤部位的阻碍。此外,未连续监测移植EPCs的体内募集和代谢活性.
    方法:用吲哚菁绿(ICG)结合的超顺磁性氧化铁纳米颗粒(SPIONs)标记EPC,并进行外部磁场靶向,以增强其对Sprague-Dawley大鼠颈动脉球囊损伤(BI)模型的传递。磁性粒子成像(MPI)/荧光成像(FLI)多模态活体成像,损伤后进行3DMPI/CT成像和MPI/FLI离体成像。收集颈动脉并进行病理和免疫荧光染色分析。通过酶联免疫吸附试验分析旁分泌效应。
    结果:磁场的应用显着增强了SPIONs@PEG-ICG-EPCs在动脉损伤部位的定位和保留,体内连续监测和离体观察都证明了这一点。这种靶向递送方法有效地抑制了新生内膜增生并增加了CD31阳性细胞在损伤部位的存在。此外,血清SDF-1α水平,VEGF,IGF-1和TGF-β1显著升高,表明旁分泌活性增强。
    结论:我们的研究结果表明,向动脉损伤区域外磁场定向递送SPIONs@PEG-ICG-EPCs可显著增强其治疗效果。这种增强可能是通过增加的旁分泌信号传导介导的。这些结果强调了磁性引导的SPIONs@PEG-ICG-EPC递送作为治疗动脉损伤的有希望的策略的潜力。
    BACKGROUND: The transplantation of endothelial progenitor cells (EPCs) has been shown to reduce neointimal hyperplasia following arterial injury. However, the efficacy of this approach is hampered by limited homing of EPCs to the injury site. Additionally, the in vivo recruitment and metabolic activity of transplanted EPCs have not been continuously monitored.
    METHODS: EPCs were labeled with indocyanine green (ICG)-conjugated superparamagnetic iron oxide nanoparticles (SPIONs) and subjected to external magnetic field targeting to enhance their delivery to a carotid balloon injury (BI) model in Sprague-Dawley rats. Magnetic particle imaging (MPI)/ fluorescence imaging (FLI) multimodal in vivo imaging, 3D MPI/CT imaging and MPI/FLI ex vivo imaging was performed after injury. Carotid arteries were collected and analyzed for pathology and immunofluorescence staining. The paracrine effects were analyzed by enzyme-linked immunosorbent assay.
    RESULTS: The application of a magnetic field significantly enhanced the localization and retention of SPIONs@PEG-ICG-EPCs at the site of arterial injury, as evidenced by both in vivo continuous monitoring and ex vivo by observation. This targeted delivery approach effectively inhibited neointimal hyperplasia and increased the presence of CD31-positive cells at the injury site. Moreover, serum levels of SDF-1α, VEGF, IGF-1, and TGF-β1 were significantly elevated, indicating enhanced paracrine activity.
    CONCLUSIONS: Our findings demonstrate that external magnetic field-directed delivery of SPIONs@PEG-ICG-EPCs to areas of arterial injury can significantly enhance their therapeutic efficacy. This enhancement is likely mediated through increased paracrine signaling. These results underscore the potential of magnetically guided SPIONs@PEG-ICG-EPCs delivery as a promising strategy for treating arterial injuries.
    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
    血管内损伤后的内膜增生(IH)和血管重塑,例如,在血管成形术后再狭窄,导致下游缺血和进行性终末器官损伤。已知干扰素γ(IFNγ)在该过程中起关键作用。在小鼠模型中,我们先前已经表明表达组织因子(TF)的纤维细胞被早期募集到损伤部位。通过凝血酶生成和蛋白酶激活受体-1(PAR-1)的激活,纤维细胞分泌血管生成素-2,刺激新内膜细胞增殖,抑制细胞凋亡并诱导CXCL-12的产生,所有这些都有助于随后发展的渐进式IH。在这项研究中,我们调查了TF,血管生成素-2和IFNγ。
    在腔内损伤4周后,在野生型小鼠的颈动脉中发育的IH含有相当比例的IFNγ纤维细胞和巨噬细胞,我们展示的是,使用先前定义的过继转移模型,来源于循环CD34+细胞。IFNγ缺陷小鼠损伤后未出现IH,除WT骨髓移植或WTCD34+细胞过继转移后。体外,从损伤后小鼠分离的CD34+细胞不表达IFNγ,但这是在提供FVIIa和FX时引起的,并在提供凝血酶原时增强:在两种情况下,IFNγ分泌都是TF依赖性的,主要通过蛋白酶激活的PAR-1介导。IFNγ主要由纤维细胞表达。在体内,WT小鼠中的所有IFNγ新内膜细胞共表达血管生成素2,IFNγ-/-小鼠中招募的少量新内膜细胞也是如此。过继转移的WTCD34+细胞用抗TIE-2抗体处理,或抗血管生成素-2的siRNA抑制IFNγ的表达和IH的发展。
    新招募的纤维细胞产生TF依赖性血管生成素-2,和较小程度的巨噬细胞,打开IFNγ表达,这是IH发展所必需的。这些新发现增强了我们对IH病理生理学的理解,并揭示了治疗干预的潜在目标。
    UNASSIGNED: The intimal hyperplasia (IH) and vascular remodelling that follows endovascular injury, for instance after post-angioplasty re-stenosis, results in downstream ischaemia and progressive end organ damage. Interferon gamma (IFNγ) is known to play a critical role in this process. In mouse models we have previously shown that fibrocytes expressing tissue factor (TF) are recruited early to the site of injury. Through thrombin generation and protease activated receptor-1 (PAR-1) activation, fibrocytes secrete angiopoietin-2, stimulate neointimal cell proliferation, inhibit apoptosis and induce CXCL-12 production, all of which contribute to the progressive IH that then develops. In this study we investigated the relationship between TF, angiopoietin-2 and IFNγ.
    UNASSIGNED: IH developing in carotid arteries of wild-type mice 4 weeks after endoluminal injury contained a significant proportion of IFNγ+ fibrocytes and macrophages, which we show, using a previously defined adoptive transfer model, were derived from circulating CD34+ cells. IH did not develop after injury in IFNγ-deficient mice, except after transplantation of WT bone marrow or adoptive transfer of WT CD34+ cells. In vitro, CD34+ cells isolated from post-injury mice did not express IFNγ, but this was induced when provided with FVIIa and FX, and enhanced when prothrombin was also provided: In both cases IFNγ secretion was TF-dependent and mediated mainly through protease activated PAR-1. IFNγ was predominantly expressed by fibrocytes. In vivo, all IFNγ+ neointimal cells in WT mice co-expressed angiopoietin-2, as did the small numbers of neointimal cells recruited in IFNγ-/- mice. Adoptively transferred WT CD34+ cells treated with either an anti-TIE-2 antibody, or with siRNA against angiopoetin-2 inhibited the expression of IFNγ and the development of IH.
    UNASSIGNED: TF-dependent angiopoietin-2 production by newly recruited fibrocytes, and to a lesser extent macrophages, switches on IFNγ expression, and this is necessary for the IH to develop. These novel findings enhance our understanding of the pathophysiology of IH and expose potential targets for therapeutic intervention.
    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
    目前尚不清楚A激酶锚定蛋白1(AKAP1)在血管平滑肌细胞(VSMC)表型调节和新内膜形成中的作用和机制。AKAP1是线粒体PKA锚定蛋白并维持线粒体稳态。本研究旨在探讨AKAP1/PKA信号如何通过调节线粒体裂变在抑制VSMC表型转化和新内膜形成中发挥保护作用。结果表明,PDGF-BB处理和球囊损伤均降低了转录,表达式,和AKAP1的线粒体锚定。体外,过表达AKAP1显著抑制PDGF-BB介导的VSMC增殖和迁移,而AKAP1敲低进一步加重了VSMC的表型转化。此外,在体内球囊损伤模型中,AKAP1过表达减少新内膜形成,肌纤维面积比,和大鼠VSMC增殖和迁移。此外,PDGF-BB和球囊损伤抑制了Ser637的Drp1磷酸化,并促进了Drp1活性和线粒体中区裂变;AKAP1过表达逆转了这些作用。AKAP1过表达还抑制了线粒体在质膜的分布和PDGF-BB诱导的PKARIIβ表达的降低,线粒体-质膜距离以及PKARIIβ蛋白水平的增加证明了这一点。此外,PKA激动剂促进Drp1磷酸化(Ser637)并抑制PDGF-BB介导的线粒体裂变,细胞增殖,和移民。PKA拮抗剂逆转了AKAP1过表达引起的Drp1磷酸化(Ser637)的增加以及线粒体中区裂变和VSMC表型转化的下降。这项研究的结果表明,AKAP1通过PKA改善Ser637的Drp1磷酸化并抑制线粒体裂变来保护VSMCs免受表型调节,从而防止新内膜形成。
    The roles and mechanisms of A-kinase anchoring protein 1 (AKAP1) in vascular smooth muscle cell (VSMC) phenotypic modulation and neointima formation are currently unknown. AKAP1 is a mitochondrial PKA-anchored protein and maintains mitochondrial homeostasis. This study aimed to investigate how AKAP1/PKA signaling plays a protective role in inhibiting VSMC phenotypic transformation and neointima formation by regulating mitochondrial fission. The results showed that both PDGF-BB treatment and balloon injury reduced the transcription, expression, and mitochondrial anchoring of AKAP1. In vitro, the overexpression of AKAP1 significantly inhibited PDGF-BB mediated VSMC proliferation and migration, whereas AKAP1 knockdown further aggravated VSMC phenotypic transformation. Additionally, in the balloon injury model in vivo, AKAP1 overexpression reduced neointima formation, the muscle fiber area ratio, and rat VSMC proliferation and migration. Furthermore, PDGF-BB and balloon injury inhibited Drp1 phosphorylation at Ser637 and promoted Drp1 activity and mitochondrial midzone fission; AKAP1 overexpression reversed these effects. AKAP1 overexpression also inhibited the distribution of mitochondria at the plasma membrane and the reduction of PKARIIβ expression induced by PDGF-BB, as evidenced by an increase in mitochondria-plasma membrane distance as well as PKARIIβ protein levels. Moreover, the PKA agonist promoted Drp1 phosphorylation (Ser637) and inhibited PDGF-BB-mediated mitochondrial fission, cell proliferation, and migration. The PKA antagonist reversed the increase in Drp1 phosphorylation (Ser637) and the decline in mitochondrial midzone fission and VSMC phenotypic transformation caused by AKAP1 overexpression. The results of this study reveal that AKAP1 protects VSMCs against phenotypic modulation by improving Drp1 phosphorylation at Ser637 through PKA and inhibiting mitochondrial fission, thereby preventing neointima formation.
    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