PAR1

PAR1
  • 文章类型: Journal Article
    背景:蛋白酶激活受体1(PAR1)在人血小板中表达,可以被低浓度的凝血酶激活。Vorapaxar,PAR1的选择性拮抗剂,抑制凝血酶诱导的人血小板钙动员,这与出血风险增加有关。相反,PAR1的正变构调节剂(PAM)的给药可能由于诱导过度的血小板活化而造成血栓形成的重大风险.在这项研究中,我们发现了一种新型的PAR1PAM,并研究了PAR1PAM增强PAR1活化对血小板活化的影响.方法:为了找到PAR1的PAMs,在HT29细胞中进行基于细胞的筛选,最后,gestodene,口服避孕药(OC),被鉴定为PAR1的新型PAM。在人巨核细胞白血病细胞系MEG-01细胞和人血小板中研究了孕二烯酮的作用机制及其对血小板活化的影响。结果:孕二烯酮以剂量依赖性方式增强了凝血酶和PAR1激活肽(AP)诱导的细胞内钙水平,而不改变PAR2和PAR4的活性。孕二烯酮显著增加PAR1-AP诱导的PAR1内化和ERK1/2磷酸化,且该增强作用被vorapaxar显著阻断。此外,孕二烯酮有效增加了PAR1-AP诱导的MEG-01细胞的形态学变化。值得注意的是,在人类血液中,孕二烯酮对PAR1-AP诱导的血小板聚集有强烈的增强作用,和vorapaxar有效减弱了孕二烯酮诱导的PAR1介导的血小板聚集增强。结论:孕二烯酮是PAR1的选择性PAM,提示与含孕二烯酮的OCs相关的静脉血栓栓塞风险增加的可能机制。
    Background: Protease-activated receptor 1 (PAR1) is expressed in human platelets and can be activated by low concentrations of thrombin. Vorapaxar, a selective antagonist of PAR1, inhibits thrombin-induced calcium mobilization in human platelet, which is associated with an increased risk of bleeding. Conversely, the administration of a positive allosteric modulator (PAM) of PAR1 may pose a substantial risk of thrombosis due to inducing excessive platelet activation. In this study, we discovered a novel PAM of PAR1 and investigated the effect of enhanced PAR1 activation by PAM of PAR1 on platelet activation. Methods: To find PAMs of PAR1, a cell-based screen was performed in HT29 cells, and finally, gestodene, an oral contraceptive drug (OC), was identified as a novel PAM of PAR1. The mechanism of action of gestodene and its effects on platelet activation were investigated in human megakaryocytic leukemia cell line MEG-01 cells and human platelet. Results: Gestodene enhanced both thrombin- and PAR1-activating peptide (AP)-induced intracellular calcium levels in a dose-dependent manner without altering PAR2 and PAR4 activity. Gestodene significantly increased PAR1-AP-induced internalization of PAR1 and phosphorylation of ERK1/2, and the enhancing effects were significantly blocked by vorapaxar. Furthermore, gestodene potently increased PAR1-AP induced morphological changes in MEG-01 cells. Remarkably, in human blood, gestodene exerted a robust augmentation of PAR1-AP-induced platelet aggregation, and vorapaxar effectively attenuated the gestodene-induced enhancement of platelet aggregation mediated by PAR1. Conclusion: Gestodene is a selective PAM of PAR1 and suggest one possible mechanism for the increased risk of venous thromboembolism associated with OCs containing gestodene.
    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
    重复创伤性脑损伤(RTBI)被认为是一种无声的被忽视的公共卫生危机,对其病理机制信号通路的理解不完全。越来越多的证据表明凝血酶及其受体的参与,蛋白酶激活受体(PAR)1,在TBI继发性损伤的发展中;然而,PAR1调节的后果及其对铁凋亡-氧化还原信号的影响,和RTBI中的NLRP3炎性体激活,仍然不清楚。Further,PAR1作为RTBI治疗靶点的功利性功能尚未阐明.为了研究这种相声,在Wistar大鼠中通过在右额叶区域每天减重5天诱导RTBI。包括三组:正常对照组,未经处理的RTBI,和RTBI+SCH79797(一种PAR1抑制剂,创伤后给药25μg/kg/天)。PAR1拮抗作用的伴随治疗改善了行为功能的改变,皮质组织结构,和神经元细胞存活。此外,受体阻滞剂下调PAR1的mRNA表达,但上调神经保护性受体PPAR-γ的mRNA表达。SCH79797的抗炎作用由低免疫表达/NF-κBp65、TNF-α、IL-1β,IL-18因此,PAR1阻断剂阻碍了炎症小体成分NLRP3、ASC、和活化的caspase-1。最终,SCH79797治疗通过激活抗氧化剂Nrf2/HO-1轴及其随后的抗氧化机制(GPX4,SOD)来限制脂质过氧化,从而减轻了铁凋亡依赖性铁氧化还原信号,铁积累,和转铁蛋白血清增量。总的来说,SCH79797通过激活PPAR-γ/Nrf2抗氧化剂提示来阻止铁中毒和NLRP3炎性体,从而提供了针对大鼠继发性RTBI后果的推定预防机制。
    Repetitive traumatic brain injury (RTBI) is acknowledged as a silent overlooked public health crisis, with an incomplete understanding of its pathomechanistic signaling pathways. Mounting evidence suggests the involvement of thrombin and its receptor, the protease-activated receptor (PAR)1, in the development of secondary injury in TBI; however, the consequences of PAR1 modulation and its impact on ferroptosis-redox signaling, and NLRP3 inflammasome activation in RTBI, remain unclear. Further, the utilitarian function of PAR1 as a therapeutic target in RTBI has not been elucidated. To study this crosstalk, RTBI was induced in Wistar rats by daily weight drops on the right frontal region for five days. Three groups were included: normal control, untreated RTBI, and RTBI+SCH79797 (a PAR1 inhibitor administered post-trauma at 25 μg/kg/day). The concomitant treatment of PAR1 antagonism improved altered behavior function, cortical histoarchitecture, and neuronal cell survival. Moreover, the receptor blockade downregulated mRNA expression of PAR1 but upregulatedthat of the neuroprotective receptor PPAR-γ. The anti-inflammatory impact of SCH79797 was signified by the low immune expression/levels of NF-κB p65,TNF-α, IL-1β, and IL-18. Consequently, the PAR1 blocker hindered the formation of inflammasome components NLRP3, ASC, and activated caspase-1. Ultimately, SCH79797 treatment abated ferroptosis-dependent iron redox signaling through the activation of the antioxidant Nrf2/HO-1 axis and its subsequent antioxidant machinery (GPX4, SOD) to limit lipid peroxidation, iron accumulation, and transferrin serum increment. Collectively, SCH79797 offered putative preventive mechanisms against secondary RTBI consequences in rats by impeding ferroptosis and NLRP3 inflammasome through activating the PPAR-γ/Nrf2 antioxidant cue.
    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
    细胞表面蛋白酶激活的受体1(PAR1)在多形性胶质母细胞瘤(GBM)中过表达。我们研究了微管蛋白介导的细胞内微管(MT)和PAR1的功能和结构。我们发现暴露于凝血酶会增加增殖的百分比,S,和M期细胞,受影响的形态,和增加的过程伸长率。PAR1拮抗剂反过来影响这些措施,增加C6细胞中微管蛋白末端结合蛋白3(EB3)mRNA的表达,并减少EB3彗星的长度,轨道长度,和神经母细胞瘤细胞的持续时间。此外,免疫荧光染色表明,在细胞分裂阶段,PAR1与MTα-微管蛋白和凝血级联蛋白密切相关。我们的发现支持PAR1参与MT动力学。
    The cell surface protease-activated receptor 1 (PAR1) is overexpressed in glioblastoma multiforme (GBM). We studied the function and structure of intracellular microtubule (MT) and PAR1 in a tubulin-mediated process. We found that exposure to thrombin increased the percentage of proliferative, S, and M phases cells, affected morphology, and increased process elongation. PAR1 antagonist inversely affects these measures, increases tubulin end-binding protein 3 (EB3) mRNA expression in C6 cells, and reduces EB3 comet length, track length, and duration in neuroblastoma cells. In addition, immunofluorescence staining suggests that PAR1 is in close association with the MT α-tubulin and with coagulation cascade proteins during cell division stages. Our findings support PAR1 involvement in MT dynamics.
    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
    对乙酰氨基酚(APAP)过量会发生肝内皮细胞(EC)功能障碍和小叶中央肝细胞坏死。蛋白酶凝血酶,这是在APAP过量期间急剧产生的,可以通过蛋白酶激活的受体1和4(PAR1/PAR4)发出信号。PAR1是一种高亲和力凝血酶受体,已知可在EC上发出信号,而PAR4是一种低亲和力的凝血酶受体,其在EC上的表达和功能的证据参差不齐。本研究旨在利用APAP过量期间产生的高水平凝血酶来确定(1)如果肝内皮PAR4是一种功能性受体,和(2)在高凝血酶背景下PAR1和PAR4的内皮特异性功能。
    我们产生了EC中Par1/Par4条件性缺失的小鼠,并用APAP过量给药。肝血管通透性,红细胞充血/出血,和肝功能在用药过量后进行评估。此外,我们使用内皮翻译核糖体亲和纯化和下一代测序(TRAPseq)研究了内皮PAR的表达水平以及它们如何影响APAP过量肝ECs中的转录.
    我们发现缺乏高表达内皮Par1或低表达Par4的小鼠在APAP诱导的肝血管不稳定中具有等效的降低,但对肝细胞坏死没有影响。此外,与单独缺乏ECs中任一受体的小鼠相比,APAP过量后,内皮Par1和Par4缺失的小鼠在较早时间点的通透性降低.我们还发现内皮PAR1-而不是PAR4-可以调节肝ECs中的转录。
    低表达PAR4可以与APAP过量肝ECs中的高表达PAR1反应相似,证明PAR4是一种有效的凝血酶受体。此外,这些受体在功能上是多余的,但在表达和影响肝脏ECs转录的能力方面作用不同.
    F2r和F2rl3分别是PAR1和PAR4的基因名称。为简单起见,以下我们将这些基因称为Par1和Par4。
    UNASSIGNED: Hepatic endothelial cell (EC) dysfunction and centrilobular hepatocyte necrosis occur with acetaminophen (APAP) overdose. The protease thrombin, which is acutely generated during APAP overdose, can signal through protease-activated receptors 1 and 4 (PAR1/PAR4). PAR1 is a high-affinity thrombin receptor that is known to signal on ECs, whereas PAR4 is a low-affinity thrombin receptor, and evidence for its expression and function on ECs is mixed. This study aims to exploit the high levels of thrombin generated during APAP overdose to determine (1) if hepatic endothelial PAR4 is a functional receptor, and (2) endothelial-specific functions for PAR1 and PAR4 in a high thrombin setting.
    UNASSIGNED: We generated mice with conditional deletion(s) of Par1/Par4 in ECs and overdosed them with APAP. Hepatic vascular permeability, erythrocyte congestion/bleeding, and liver function were assessed following overdose. Additionally, we investigated the expression levels of endothelial PARs and how they influence transcription in APAP-overdosed liver ECs using endothelial Translating Ribosome Affinity Purification followed by next-generation sequencing (TRAPseq).
    UNASSIGNED: We found that mice deficient in high-expressing endothelial Par1 or low-expressing Par4 had equivalent reductions in APAP-induced hepatic vascular instability but no effect on hepatocyte necrosis. Additionally, mice with loss of endothelial Par1 and Par4 had reduced permeability at an earlier time point after APAP overdose when compared to mice singly deficient in either receptor in ECs. We also found that endothelial PAR1-but not PAR4-can regulate transcription in hepatic ECs.
    UNASSIGNED: Low-expressing PAR4 can react similarly to high-expressing PAR1 in APAP-overdosed hepatic ECs, demonstrating that PAR4 is a potent thrombin receptor. Additionally, these receptors are functionally redundant but act divergently in their expression and ability to influence transcription in hepatic ECs.
    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
    星形胶质细胞在调节突触传递中起重要作用。这项研究描述了星形细胞G蛋白偶联受体(GPCRs)调节小鼠海马中兴奋性突触传递的一种新形式。我们之前已经描述了通过蛋白酶激活的受体-1(PAR1)激活的星形细胞谷氨酸释放,尽管这方面的监管机制很复杂。通过电生理分析和建模,我们发现PAR1激活持续增加突触间隙中谷氨酸的浓度和持续时间.这种作用不是由于突触前谷氨酸释放的变化或谷氨酸转运蛋白表达的变化。然而,阻断组II代谢型谷氨酸受体(mGluR2/3)取消了PAR1介导的突触谷氨酸浓度调节,提示此GPCR在介导PAR1激活对谷氨酸释放的影响中的作用。此外,mGluR2/3的激活导致海马星形胶质细胞通过TREK-1通道释放谷氨酸。这些数据表明,星形胶质细胞GPCRs参与了一种新的调节机制,以塑造海马兴奋性突触中突触释放的谷氨酸的时间过程。
    Astrocytes play an essential role in regulating synaptic transmission. This study describes a novel form of modulation of excitatory synaptic transmission in the mouse hippocampus by astrocytic G-protein-coupled receptors (GPCRs). We have previously described astrocytic glutamate release via protease-activated receptor-1 (PAR1) activation, although the regulatory mechanisms for this are complex. Through electrophysiological analysis and modeling, we discovered that PAR1 activation consistently increases the concentration and duration of glutamate in the synaptic cleft. This effect was not due to changes in the presynaptic glutamate release or alteration in glutamate transporter expression. However, blocking group II metabotropic glutamate receptors (mGluR2/3) abolished PAR1-mediated regulation of synaptic glutamate concentration, suggesting a role for this GPCR in mediating the effects of PAR1 activation on glutamate release. Furthermore, activation of mGluR2/3 causes glutamate release through the TREK-1 channel in hippocampal astrocytes. These data show that astrocytic GPCRs engage in a novel regulatory mechanism to shape the time course of synaptically-released glutamate in excitatory synapses of the hippocampus.
    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
    目的:胰凝乳蛋白酶是一种分泌到小肠腔以消化食物蛋白质的胰蛋白酶。我们假设胰凝乳蛋白酶活性可能在上皮细胞附近发现,并且胰凝乳蛋白酶通过蛋白酶激活的受体(PAR)向它们发出信号。我们破译了肠上皮细胞中胰凝乳蛋白酶信号传导的分子药理机制和基因表达调控。
    方法:通过Western印迹和酶活性测试在鼠和人肠道样品的腔和粘膜区室中评估胰凝乳蛋白酶的存在和活性。使用表达N末端标记的受体的细胞系评估胰凝乳蛋白酶切割PAR1或PAR2的胞外结构域的能力。通过HPLC-MS分析确定PAR1和PAR2上胰凝乳蛋白酶的切割位点。通过钙动员测定和(ERK1/2)磷酸化的Western印迹分析,在CMT93肠上皮细胞中研究了胰凝乳蛋白酶的信号传导机制。在结肠类器官上分析了胰凝乳蛋白酶信号的转录后果。
    结果:我们发现胰凝乳蛋白酶在结肠上皮附近存在并具有活性。分子药理学研究表明,胰凝乳蛋白酶可切割PAR1和PAR2受体。胰凝乳蛋白酶通过PAR2激活钙和ERK1/2信号通路,该通路促进结肠类器官中白细胞介素-10(IL-10)的上调。相比之下,胰凝乳蛋白酶解除了PAR1的武装,阻止了其经典激动剂的进一步激活,凝血酶。
    结论:我们的结果强调了胰凝乳蛋白酶通过PAR向肠上皮细胞发出信号的能力,这可能会对肠道稳态产生重要的生理影响。
    OBJECTIVE: Chymotrypsin is a pancreatic protease secreted into the lumen of the small intestine to digest food proteins. We hypothesized that chymotrypsin activity may be found close to epithelial cells and that chymotrypsin signals to them via protease-activated receptors (PARs). We deciphered molecular pharmacological mechanisms and gene expression regulation for chymotrypsin signalling in intestinal epithelial cells.
    METHODS: The presence and activity of chymotrypsin were evaluated by Western blot and enzymatic activity tests in the luminal and mucosal compartments of murine and human gut samples. The ability of chymotrypsin to cleave the extracellular domain of PAR1 or PAR2 was assessed using cell lines expressing N-terminally tagged receptors. The cleavage site of chymotrypsin on PAR1 and PAR2 was determined by HPLC-MS analysis. The chymotrypsin signalling mechanism was investigated in CMT93 intestinal epithelial cells by calcium mobilization assays and Western blot analyses of (ERK1/2) phosphorylation. The transcriptional consequences of chymotrypsin signalling were analysed on colonic organoids.
    RESULTS: We found that chymotrypsin was present and active in the vicinity of the colonic epithelium. Molecular pharmacological studies have shown that chymotrypsin cleaves both PAR1 and PAR2 receptors. Chymotrypsin activated calcium and ERK1/2 signalling pathways through PAR2, and this pathway promoted interleukin-10 (IL-10) up-regulation in colonic organoids. In contrast, chymotrypsin disarmed PAR1, preventing further activation by its canonical agonist, thrombin.
    CONCLUSIONS: Our results highlight the ability of chymotrypsin to signal to intestinal epithelial cells via PARs, which may have important physiological consequences in gut homeostasis.
    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
    首次诊断为心房颤动(FDAF)的患者在随访期间表现出主要的不良心血管事件(MACE)。临床前模型已证明血栓-炎症介导不良心脏重塑和动脉粥样硬化血栓形成事件。我们假设凝血酶活性(FIIa)将凝血与炎症和心脏纤维化/功能障碍联系起来。在FDAF中尚未表征血浆中血栓炎症反应的替代标志物。在这项前瞻性纵向研究中,出现FDAF的患者(n=80),和20个匹配的控件,包括在内。与无房颤的慢性心血管疾病患者相比,早期房颤患者的FIIa生成和血浆活性增加(对照;p<0.0001)。这种增加伴随着血浆中血小板和内皮活化的升高的生物标志物(ELISA)。与对照组相比,表达FIIa激活的蛋白酶激活的受体1(PAR1)(流式细胞术)的促炎外周免疫细胞(TNF-α+或IL-6+)在FDAF患者中更频繁地循环(p<0.0001)。FIIa活性与心脏纤维化(胶原更新)和心脏功能障碍(NT-proANP/NT-proBNP)替代标志物相关。发生MACE的FDAF患者血浆中的FIIa活性较高。通过FIIa的信号可能是凝血系统(组织因子-FXa/FIIa-PAR1轴)之间的假定链接,炎症,和促纤维化途径(血栓炎症)在FDAF。
    Patients with first-diagnosed atrial fibrillation (FDAF) exhibit major adverse cardiovascular events (MACEs) during follow-up. Preclinical models have demonstrated that thrombo-inflammation mediates adverse cardiac remodeling and atherothrombotic events. We have hypothesized that thrombin activity (FIIa) links coagulation with inflammation and cardiac fibrosis/dysfunction. Surrogate markers of the thrombo-inflammatory response in plasma have not been characterized in FDAF. In this prospective longitudinal study, patients presenting with FDAF (n = 80), and 20 matched controls, were included. FIIa generation and activity in plasma were increased in the patients with early AF compared to the patients with chronic cardiovascular disease without AF (controls; p < 0.0001). This increase was accompanied by elevated biomarkers (ELISA) of platelet and endothelial activation in plasma. Pro-inflammatory peripheral immune cells (TNF-α+ or IL-6+) that expressed FIIa-activated protease-activated receptor 1 (PAR1) (flow cytometry) circulated more frequently in patients with FDAF compared to the controls (p < 0.0001). FIIa activity correlated with cardiac fibrosis (collagen turnover) and cardiac dysfunction (NT-pro ANP/NT-pro BNP) surrogate markers. FIIa activity in plasma was higher in patients with FDAF who experienced MACE. Signaling via FIIa might be a presumed link between the coagulation system (tissue factor-FXa/FIIa-PAR1 axis), inflammation, and pro-fibrotic pathways (thrombo-inflammation) in FDAF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    心血管疾病(CVD)是世界范围内死亡的主要原因。尽管有许多变量有助于这种疾病的发展,主要是血小板的活性提供了这种疾病流行的机制。虽然有许多血小板受体表达在血小板的表面,在很大程度上,有10种主要的血小板受体对大多数血小板功能有贡献。了解这些关键的血小板受体对于患有心肌梗死的患者至关重要。CVD,以及由于这些受体的过度激活或突变而引起的许多其他疾病。本手稿的目的是回顾对血小板活性贡献最大的主要血小板受体。
    Cardiovascular disease (CVD) is a major cause of death worldwide. Although there are many variables that contribute to the development of this disease, it is predominantly the activity of platelets that provides the mechanisms by which this disease prevails. While there are numerous platelet receptors expressed on the surface of platelets, it is largely the consensus that there are 10 main platelet receptors that contribute to a majority of platelet function. Understanding these key platelet receptors is vitally important for patients suffering from myocardial infarction, CVD, and many other diseases that arise due to overactivation or mutations of these receptors. The goal of this manuscript is to review the main platelet receptors that contribute most to platelet activity.
    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
    瘙痒是一种令人不快的感觉,它唤起了抓挠的欲望。皮肤屏障不断暴露于微生物及其产品。然而,微生物在瘙痒产生中的作用是未知的。这里,我们发现金黄色葡萄球菌,一种与皮肤瘙痒疾病相关的细菌病原体,直接激活瘙痒感受器感觉神经元来驱动瘙痒。皮肤金黄色葡萄球菌暴露会导致强烈的瘙痒和划痕诱导的损伤。通过测试多个等基因细菌突变体的毒力因子,我们确定金黄色葡萄球菌丝氨酸蛋白酶V8是引起自发性瘙痒和异常的关键介质。V8裂解小鼠和人感觉神经元上的蛋白酶激活受体1(PAR1)。通过遗传缺陷靶向PAR1,小干扰RNA(siRNA)敲低,或药物阻断减少由V8和金黄色葡萄球菌暴露引起的瘙痒和皮肤损伤。因此,我们确定了一种致痒细菌因子的作用机制,并证明了抑制V8-PAR1信号传导治疗瘙痒的潜力.
    Itch is an unpleasant sensation that evokes a desire to scratch. The skin barrier is constantly exposed to microbes and their products. However, the role of microbes in itch generation is unknown. Here, we show that Staphylococcus aureus, a bacterial pathogen associated with itchy skin diseases, directly activates pruriceptor sensory neurons to drive itch. Epicutaneous S. aureus exposure causes robust itch and scratch-induced damage. By testing multiple isogenic bacterial mutants for virulence factors, we identify the S. aureus serine protease V8 as a critical mediator in evoking spontaneous itch and alloknesis. V8 cleaves proteinase-activated receptor 1 (PAR1) on mouse and human sensory neurons. Targeting PAR1 through genetic deficiency, small interfering RNA (siRNA) knockdown, or pharmacological blockade decreases itch and skin damage caused by V8 and S. aureus exposure. Thus, we identify a mechanism of action for a pruritogenic bacterial factor and demonstrate the potential of inhibiting V8-PAR1 signaling to treat itch.
    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
    G蛋白偶联受体(GPCRs)是高度可药用的,并涉及许多疾病,包括血管炎症。GPCR信号从质膜以及从核内体转导,并由翻译后修饰控制。凝血酶激活的GPCR蛋白酶激活的受体1(PAR1)被泛素修饰。PAR1的泛素化驱动转化生长因子-β激活的激酶-1结合蛋白2(TAB2)的募集和TAB1在内体的共缔合,在内皮细胞中触发p38丝裂原活化蛋白激酶(MAPK)依赖性炎症反应。其他内皮GPCR也通过非经典TAB1-TAB2依赖性途径诱导p38活化。然而,控制GPCR泛素驱动的p38炎症信号传导的调控过程仍知之甚少.我们发现了启动GPCR泛素依赖性p38信号传导的机制,然而,关闭路径的机制尚不清楚。我们假设去泛素化是调节泛素驱动的p38信号传导的重要步骤。为了鉴定控制GPCR-p38MAPK信号传导的特异性去泛素化酶(DUB),我们在内皮细胞和HeLa细胞中进行了针对96个DUB的siRNA文库筛选。我们确定了9个DUB,并验证了两种DUB的功能,包括圆柱瘤病(CYLD)和泛素特异性蛋白酶-34(USP34),它们特异性调节凝血酶诱导的p38磷酸化。通过siRNA增强凝血酶刺激的p38信号传导来耗尽CYLD表达,内皮屏障通透性和白细胞介素-6(IL-6)细胞因子表达增加。相反,siRNA敲低USP34表达降低凝血酶促进的IL-6表达,对凝血酶诱导的内皮屏障通透性无影响。这些研究表明,特定的DUB可以明显调节GPCR诱导的p38介导的炎症反应。
    G protein-coupled receptors (GPCRs) are highly druggable and implicated in numerous diseases, including vascular inflammation. GPCR signals are transduced from the plasma membrane as well as from endosomes and controlled by posttranslational modifications. The thrombin-activated GPCR protease-activated receptor-1 is modified by ubiquitin. Ubiquitination of protease-activated receptor-1 drives recruitment of transforming growth factor-β-activated kinase-1-binding protein 2 (TAB2) and coassociation of TAB1 on endosomes, which triggers p38 mitogen-activated protein kinase-dependent inflammatory responses in endothelial cells. Other endothelial GPCRs also induce p38 activation via a noncanonical TAB1-TAB2-dependent pathway. However, the regulatory processes that control GPCR ubiquitin-driven p38 inflammatory signaling remains poorly understood. We discovered mechanisms that turn on GPCR ubiquitin-dependent p38 signaling, however, the mechanisms that turn off the pathway are not known. We hypothesize that deubiquitination is an important step in regulating ubiquitin-driven p38 signaling. To identify specific deubiquitinating enzymes (DUBs) that control GPCR-p38 mitogen-activated protein kinase signaling, we conducted a siRNA library screen targeting 96 DUBs in endothelial cells and HeLa cells. We identified nine DUBs and validated the function two DUBs including cylindromatosis and ubiquitin-specific protease-34 that specifically regulate thrombin-induced p38 phosphorylation. Depletion of cylindromatosis expression by siRNA enhanced thrombin-stimulated p38 signaling, endothelial barrier permeability, and increased interleukin-6 cytokine expression. Conversely, siRNA knockdown of ubiquitin-specific protease-34 expression decreased thrombin-promoted interleukin-6 expression and had no effect on thrombin-induced endothelial barrier permeability. These studies suggest that specific DUBs distinctly regulate GPCR-induced p38-mediated inflammatory responses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号