PAR-4

Par - 4
  • 文章类型: Journal Article
    哮喘中的2型炎症随着暴露于刺激而发展,包括来自屋尘螨(HDM)的吸入过敏原。特征包括粘液分泌过多和以基础Cl-电流升高为特征的促分泌离子转运的形成。使用用HDM提取物处理的人鼻窦上皮细胞的研究报告了更高的蛋白酶激活受体2(PAR-2)激动剂诱导的钙动员,这可能与变应原相关蛋白酶的气道致敏有关。在这里,本研究旨在探讨HDM对哮喘气道上皮细胞Ca2+信号和炎症反应的影响。在气-液界面培养的哮喘供体的原代支气管上皮细胞(hPBECs)用于评估电生理,Ca2+信号传导和炎症反应。在响应PAR-2激动剂2-Furoyl-LIGRLO-酰胺(2-FLI)的Ca2信号传导方面观察到差异,以及响应胰蛋白酶和2-FLI的等效短路电流(Ieq),在ALI哮喘和健康的hPBECs中。HDM处理导致细胞内阳离子水平增加(Ca2+,Na)并显着降低了2-FLI诱导的哮喘细胞中Ieq的变化。发现根尖HDM诱导的Ca2动员主要涉及PAR-2和PAR-4相关的存储操作的Ca2流入和TRPV1的激活。相比之下,PAR-2、PAR-4拮抗剂和TRPV1拮抗剂仅显示对基底外侧HDM诱导的Ca2+动员的轻微影响。HDM胰蛋白酶样丝氨酸蛋白酶是导致非阿米洛利敏感Ieq的主要成分,也增加了哮喘hPBEC的白介素33(IL-33)和胸腺基质淋巴细胞生成素(TSLP)。这些研究进一步深入了解与HDM诱导的细胞信号改变相关的复杂机制及其与哮喘病理变化的相关性。
    Type 2 inflammation in asthma develops with exposure to stimuli to include inhaled allergens from house dust mites (HDM). Features include mucus hypersecretion and the formation of pro-secretory ion transport characterised by elevated basal Cl- current. Studies using human sinonasal epithelial cells treated with HDM extract report a higher protease activated receptor-2 (PAR-2) agonist-induced calcium mobilisation that may be related to airway sensitisation by allergen-associated proteases. Herein, this study aimed to investigate the effect of HDM on Ca2+ signalling and inflammatory responses in asthmatic airway epithelial cells. Primary bronchial epithelial cells (hPBECs) from asthma donors cultured at air-liquid interface were used to assess electrophysiological, Ca2+ signalling and inflammatory responses. Differences were observed regarding Ca2+ signalling in response to PAR-2 agonist 2-Furoyl-LIGRLO-amide (2-FLI), and equivalent short-circuit current (Ieq) in response to trypsin and 2-FLI, in ALI-asthma and healthy hPBECs. HDM treatment led to increased levels of intracellular cations (Ca2+, Na+) and significantly reduced the 2-FLI-induced change of Ieq in asthma cells. Apical HDM-induced Ca2+ mobilisation was found to mainly involve the activation of PAR-2 and PAR-4-associated store-operated Ca2+ influx and TRPV1. In contrast, PAR-2, PAR-4 antagonists and TRPV1 antagonist only showed slight impact on basolateral HDM-induced Ca2+ mobilisation. HDM trypsin-like serine proteases were the main components leading to non-amiloride sensitive Ieq and also increased interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP) from asthma hPBECs. These studies add further insight into the complex mechanisms associated with HDM-induced alterations in cell signalling and their relevance to pathological changes within asthma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    肺癌是癌症相关死亡的主要原因。肺癌细胞通过抑制肿瘤抑制因子Par-4蛋白(也称为PAWR)的分泌和/或下调细胞表面上的Par-4受体GRP78(cGRsP78)来发展对凋亡的抗性。我们试图鉴定FDA批准的药物,这些药物可以提高肺癌细胞表面的csGRP78并诱导癌细胞和/或正常细胞的Par-4分泌,从而以自分泌或旁分泌的方式抑制癌症生长。在一个不偏不倚的屏幕上,我们确定了克唑替尼(CZT),激活的ALK/MET/ROS1受体酪氨酸激酶的抑制剂,作为CSGRP78在ALK阴性表达的诱导剂,KRAS或EGFR突变肺癌细胞。肺癌细胞中csGRP78的升高依赖于CZT对非受体酪氨酸激酶SRC的激活。抑制癌细胞中的SRC激活可防止csGRP78易位,但可促进CZT分泌Par-4,这意味着激活的SRC阻止了Par-4的分泌。在正常细胞中,CZT不会激活SRC和csGRP78升高,但会诱导Par-4分泌。因此,CZT诱导正常细胞分泌Par-4,并在ALK阴性肿瘤细胞中升高csGRP78,从而引起癌细胞培养物中的旁分泌凋亡和小鼠肿瘤异种移植物的生长抑制。因此,CZT诱导正常细胞和癌细胞中SRC的差异激活以触发促凋亡Par-4-GRP78轴。由于csGRP78是一种可靶向受体,CZT可用于提高csGRP78以抑制ALK阴性肺肿瘤。
    Lung cancer is the leading cause of cancer-related deaths. Lung cancer cells develop resistance to apoptosis by suppressing the secretion of the tumor suppressor Par-4 protein (also known as PAWR) and/or down-modulating the Par-4 receptor GRP78 on the cell surface (csGRP78). We sought to identify FDA-approved drugs that elevate csGRP78 on the surface of lung cancer cells and induce Par-4 secretion from the cancer cells and/or normal cells in order to inhibit cancer growth in an autocrine or paracrine manner. In an unbiased screen, we identified crizotinib (CZT), an inhibitor of activated ALK/MET/ROS1 receptor tyrosine kinase, as an inducer of csGRP78 expression in ALK-negative, KRAS or EGFR mutant lung cancer cells. Elevation of csGRP78 in the lung cancer cells was dependent on activation of the non-receptor tyrosine kinase SRC by CZT. Inhibition of SRC activation in the cancer cells prevented csGRP78 translocation but promoted Par-4 secretion by CZT, implying that activated SRC prevented Par-4 secretion. In normal cells, CZT did not activate SRC and csGRP78 elevation but induced Par-4 secretion. Consequently, CZT induced Par-4 secretion from normal cells and elevated csGRP78 in the ALK-negative tumor cells to cause paracrine apoptosis in cancer cell cultures and growth inhibition of tumor xenografts in mice. Thus, CZT induces differential activation of SRC in normal and cancer cells to trigger the pro-apoptotic Par-4-GRP78 axis. As csGRP78 is a targetable receptor, CZT can be repurposed to elevate csGRP78 for inhibition of ALK-negative lung tumors.
    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
    金黄色葡萄球菌感染是感染性心内膜炎(IE)的主要原因。该病原体对血小板的活化导致其聚集和血栓形成,这被认为是IE的发展和发病机理中的重要步骤。这里,我们发现一种分泌的半胱氨酸蛋白酶,staphoainA,激活人血小板并诱导其聚集。缺乏葡萄球菌蛋白酶A产生的scpA突变体的培养上清液触发血小板聚集的能力降低。纯化的staphostatinA抑制血小板激动剂活性,一种特定的抑制剂,从而暗示其蛋白酶活性的激动。在全血中,使用浓度的staphoppainA,否则不足以诱导血小板聚集,观察到与胶原和血栓形成的结合增加。使用对蛋白酶激活的受体1和4具有特异性的拮抗剂,我们证明了它们在介导staphoppainA诱导的血小板激活中的作用。
    Infection by Staphylococcus aureus is the leading cause of infective endocarditis (IE). Activation of platelets by this pathogen results in their aggregation and thrombus formation which are considered to be important steps in the development and pathogenesis of IE. Here, we show that a secreted cysteine protease, staphopain A, activates human platelets and induces their aggregation. The culture supernatant of a scpA mutant deficient in staphopain A production was reduced in its ability to trigger platelet aggregation. The platelet agonist activity of purified staphopain A was inhibited by staphostatin A, a specific inhibitor, thus implicating its protease activity in the agonism. In whole blood, using concentrations of staphopain A that were otherwise insufficient to induce platelet aggregation, increased binding to collagen and thrombus formation was observed. Using antagonists specific to protease-activated receptors 1 and 4, we demonstrate their role in mediating staphopain A induced platelet activation.
    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
    前列腺细胞凋亡反应-4(Par-4)是一种诱导癌细胞凋亡的肿瘤抑制因子。然而,Par-4的生理功能仍然未知。在这里,我们显示常规Par-4敲除(Par-4-/-)小鼠和脂肪细胞特异性Par-4敲除(AKO)小鼠,但不是肝细胞特异性Par-4基因敲除小鼠,标准饮食肥胖。Par-4-/-和AKO小鼠表现出脂肪细胞中脂肪的吸收和储存增加。机械上,Par-4损失与mdm2下调和p53的激活有关。我们确定补体因子c3是与脂肪细胞中脂肪储存相关的p53调节基因。脂肪细胞中的Par-4再表达或c3缺失逆转了肥胖小鼠的表型。此外,肥胖的人类受试者相对于瘦受试者显示出更低的Par-4表达,在纵向研究中,较低的基线Par-4水平表明在以后的生活中患肥胖症的风险增加.这些发现表明Par-4抑制p53及其靶标c3以调节肥胖。
    Prostate apoptosis response-4 (Par-4) is a tumor suppressor that induces apoptosis in cancer cells. However, the physiological function of Par-4 remains unknown. Here we show that conventional Par-4 knockout (Par-4-/-) mice and adipocyte-specific Par-4 knockout (AKO) mice, but not hepatocyte-specific Par-4 knockout mice, are obese with standard chow diet. Par-4-/- and AKO mice exhibit increased absorption and storage of fat in adipocytes. Mechanistically, Par-4 loss is associated with mdm2 downregulation and activation of p53. We identified complement factor c3 as a p53-regulated gene linked to fat storage in adipocytes. Par-4 re-expression in adipocytes or c3 deletion reversed the obese mouse phenotype. Moreover, obese human subjects showed lower expression of Par-4 relative to lean subjects, and in longitudinal studies, low baseline Par-4 levels denoted an increased risk of developing obesity later in life. These findings indicate that Par-4 suppresses p53 and its target c3 to regulate obesity.
    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
    For some cancer subtypes, such as triple-negative breast cancer, there are no specific therapies, which leads to a poor prognosis associated with invasion and metastases. Ruthenium complexes have been developed to act in all steps of tumor growth and its progression. In this study, we investigated the effects of Ruthenium (II) complexes coupled to the amino acids methionine (RuMet) and tryptophan (RuTrp) on the induction of cell death, clonogenic survival ability, inhibition of angiogenesis, and migration of MDA-MB-231 cells (human triple-negative breast cancer). The study also demonstrated that the RuMet and RuTrp complexes induce cell cycle blockage and apoptosis of MDA-MB-231 cells, as evidenced by an increase in the number of Annexin V-positive cells, p53 phosphorylation, caspase 3 activation, and poly(ADP-ribose) polymerase cleavage. Moreover, morphological changes and loss of mitochondrial membrane potential were detected. The RuMet and RuTrp complexes induced DNA damage probably due to reactive oxygen species production related to mitochondrial membrane depolarization. Therefore, the RuMet and RuTrp complexes acted directly on breast tumor cells, leading to cell death and inhibiting their metastatic potential; this reveals the potential therapeutic action of these drugs.
    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
    The last decade has witnessed a substantial expansion in the field of microRNA (miRNA) biology, providing crucial insights into the role of miRNAs in disease pathology, predominantly in cancer progression and its metastatic spread. The discovery of tumor-suppressing miRNAs represents a potential approach for developing novel therapeutics. In this context, through miRNA microarray analysis, we examined the consequences of Prostate apoptosis response-4 (Par-4), a well-established tumor-suppressor, stimulation on expression of different miRNAs in Panc-1 cells. The results strikingly indicated elevated miR-200c levels in these cells upon Par-4 overexpression. Intriguingly, the Reverse Phase Protein Array (RPPA) analysis revealed differentially expressed proteins (DEPs), which overlap between miR200c- and Par-4-transfected cells, highlighting the cross-talks between these pathways. Notably, Phospho-p44/42 MAPK; Bim; Bcl-xL; Rb Phospho-Ser807, Ser811; Akt Phospho-Ser473; Smad1/5 Phospho-Ser463/Ser465 and Zyxin scored the most significant DEPs among the two data sets. Furthermore, the GFP-Par-4-transfected cells depicted an impeded expression of critical mesenchymal markers viz. TGF-β1, TGF-β2, ZEB-1, and Twist-1, concomitant with augmented miR-200c and E-cadherin levels. Strikingly, while Par-4 overexpression halted ZEB-1 at the transcriptional level; contrarily, silencing of endogenous Par-4 by siRNA robustly augmented the Epithelial-mesenchymal transition (EMT) markers, along with declining miR-200c levels. The pharmacological Par-4-inducer, NGD16, triggered Par-4 expression which corresponded with increased miR-200c resulting in the ZEB-1 downregulation. Noteworthily, tumor samples obtained from the syngenic mouse pancreatic cancer model revealed elevated miR-200c levels in the NGD16-treated mice that positively correlated with the Par-4 and E-cadherin levels in vivo; while a negative correlation was evident with ZEB-1 and Vimentin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:我们最近报道了前列腺细胞凋亡反应4(Par-4),一种潜在的肿瘤抑制蛋白通过抑制Twist-1启动子活性抑制侵袭性癌细胞中的上皮-间质转化(EMT)特性并促进间质-上皮转化(MET).这里,我们证明了NGD16(一种小分子抗转移剂)对Par-4的遗传和药理学调节,通过抑制Twist-1的下游效应物MDM-2来限制侵袭性癌细胞中EMT诱导的化学抗性。
    方法:基质胶侵袭试验,明胶降解试验,细胞散射试验,采用MTT法和集落形成法研究侵袭性癌细胞的增殖和迁移能力。免疫印迹,免疫细胞化学,和免疫沉淀分析用于确定蛋白质表达和蛋白质-蛋白质相互作用。采用4T1侵袭性小鼠癌模型评估肿瘤生长和肺转移。
    结果:吉西他滨(核苷类似物抗癌剂)对胰腺癌(Panc-1,MiaPaca-2)和乳腺癌(MDA-MB-231)细胞的治疗可扩增MDM-2表达并增加EMT特性。相反,NGD16增强了肿瘤抑制因子Par-4的表达,并抑制了这些细胞的侵袭和迁移能力。此外,Par-4的诱导有效地减少了MDM-2以及pro-EMT标记,然而,增加了上皮标志物的表达。此外,siRNA介导的Par-4沉默表明NGD16以Par-4依赖性方式发挥其EMT抑制作用。机械上,Par-4激活通过破坏MDM-2-p53相互作用激发p53,恢复了癌细胞的上皮特征。此外,通过siRNA对MDM-2的部分敲除显著地显示了NGD16的抗增殖和抗侵袭作用。最后,NGD16有效抑制小鼠乳腺癌模型中的肿瘤生长和肺转移,而没有任何不良作用。
    结论:我们的发现揭示了Par-4是一个关键的治疗靶点,NGD16(Par-4的药理学调节剂)是抑制EMT和相关化疗耐药的潜在工具,可以在临床上用于治疗侵袭性癌症。
    BACKGROUND: We recently reported prostate apoptosis response 4 (Par-4), a potential tumor suppressor protein restrains epithelial-mesenchymal transition (EMT) properties and promotes mesenchymal-epithelial transition (MET) in invasive cancer cells by repressing Twist-1 promoter activity. Here, we demonstrate that genetic as well as pharmacological modulation of Par-4 by NGD16 (a small molecule antimetastatic agent), limits EMT-induced chemoresistance in aggressive cancer cells by suppressing MDM-2, a downstream effector of Twist-1.
    METHODS: Matrigel invasion assay, gelatin degradation assay, cell scattering assay, MTT assay and colony formation assay were used to study the proliferation and migration abilities of invasive cancer cells. Immunoblotting, immunocytochemistry, and immunoprecipitation analysis were utilized for determining protein expression and protein-protein interaction. 4T1 aggressive mouse carcinoma model was employed to evaluate tumor growth and lung metastasis.
    RESULTS: Treatment of gemcitabine (nucleoside analogue anticancer agent) to pancreatic cancer (Panc-1, MiaPaca-2) and breast cancer (MDA-MB-231) cells amplified MDM-2 expression along with increase in EMT properties. Conversely, NGD16 boosted expression of tumor suppressor Par-4 and inhibited invasion and migration abilities of these cells. Moreover, induction of Par-4 effectively diminished MDM-2 along with pro-EMT markers, whereas, augmented the expression of epithelial markers. Furthermore, siRNA-mediated silencing of Par-4 divulged that NGD16 exerts its EMT inhibitory effects in a Par-4-dependent manner. Mechanistically, Par-4 activation provokes p53 by disrupting MDM-2-p53 interaction, which restored epithelial characteristics in cancer cells. Additionally, partial knockdown of MDM-2 through siRNA pronounced the anti-proliferative and anti-invasive effects of NGD16. Finally, NGD16 efficiently inhibited tumor growth and lung metastasis in mouse mammary carcinoma model without showing any undesirable effects.
    CONCLUSIONS: Our findings unveil Par-4 as a key therapeutic target and NGD16 (the pharmacological modulator of Par-4) are potential tools to suppress EMT and associated chemoresistance, which could be exploited clinically for the treatment of aggressive cancers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Dual antiplatelet therapy reduces ischemic events in cardiovascular disease, but it increases bleeding risk. Thrombin receptors PAR1 and PAR4 are drug targets, but the role of thrombin in platelet aggregation remains largely unexplored in large populations. We performed a genome-wide association study (GWAS) of platelet aggregation in response to full-length thrombin, followed by clinical association analyses, Mendelian randomization, and functional characterization including iPSC-derived megakaryocyte and platelet experiments. We identified a single sentinel variant in the GRK5 locus (rs10886430-G, p = 3.0 × 10-42) associated with increased thrombin-induced platelet aggregation (β = 0.70, SE = 0.05). We show that disruption of platelet GRK5 expression by rs10886430-G is associated with enhanced platelet reactivity. The proposed mechanism of a GATA1-driven megakaryocyte enhancer is confirmed in allele-specific experiments. Utilizing further data, we demonstrate that the allelic effect is highly platelet- and thrombin-specific and not likely due to effects on thrombin levels. The variant is associated with increased risk of cardiovascular disease outcomes in UK BioBank, most strongly with pulmonary embolism. The variant associates with increased risk of stroke in the MEGASTROKE, UK BioBank, and FinnGen studies. Mendelian randomization analyses in independent samples support a causal role for rs10886430-G in increasing risk for stroke, pulmonary embolism, and venous thromboembolism through its effect on thrombin-induced platelet reactivity. We demonstrate that G protein-coupled receptor kinase 5 (GRK5) promotes platelet activation specifically via PAR4 receptor signaling. GRK5 inhibitors in development for the treatment of heart failure and cancer could have platelet off-target deleterious effects. Common variants in GRK5 may modify clinical outcomes with PAR4 inhibitors, and upregulation of GRK5 activity or signaling in platelets may have therapeutic benefits.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Deregulation of TGF-β signaling is intricately engrossed in the pathophysiology of pancreatic adenocarcinomas (PDACs). The role of TGF-β all through pancreatic cancer initiation and progression is multifarious and somewhat paradoxical. TGF-β plays a tumor suppressive role in early-stage pancreatic cancer by promoting apoptosis and inhibiting epithelial cell cycle progression, but incites tumor promotion in late-stage by modulating genomic instability, neo-angiogenesis, immune evasion, cell motility, and metastasis. Here, we provide evidences that Par-4 acts as one of the vital mediators to regulate TGF-β/Smad4 pathway, wherein, Par-4 induction/over-expression induced EMT which was later culminated in to apoptosis in presence of TGF-β via positive regulation of Smad4. Intriguingly, Par-4-/- cells were devoid of significant Smad4 induction compared to Par-4+/+ cells in presence of TGF-β and ectopic Par-4 steadily augmented Smad4 expression by restoring TGF-β/Smad4 axis in Panc-1 cells. Further, our FACS and western blotting results unveiled that Par-4 dragged the PDAC cells to G1 arrest in presence of TGF-β byelevating p21 and p27 levels while attenuating Cyclin E and A levels and augmenting caspase 3 cleavage triggering lethal EMT. Through restoration of Smad4, we further establish that in BxPC3 cell line (Smad4-/-), Smad4 is essential for Par-4 to indulge TGF-β dependent lethal EMT program. The mechanistic relevance of Par-4 mediated Smad4 activation was additionally validated by co-immunoprecipitation wherein disruption of NM23H1-STRAP interaction by Par-4 rescues TGF-β/Smad4 pathway in PDAC and mediates the tumor suppressive role of TGF-β, therefore serving as a vital cog to restore the apoptotic functions of TGF-β pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Prostate apoptosis response-4 (Par-4) is a tumor suppressor protein that selectively induces apoptosis in cancer cells. Although the mechanism of Par-4-mediated induction of apoptosis has been well studied, the involvement of Par-4 in other mechanisms of cell death such as autophagy is unclear. We investigated the mechanism involved in Par-4-mediated autophagic cell death in human malignant glioma. We demonstrate for the first time that the tumor suppressor lipid, ceramide (Cer), causes Par-4 induction, leading to autophagic cell death in human malignant glioma. Furthermore, we identified the tumor suppressor protein p53 and BCL2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3) as downstream targets of Par-4 during Cer-mediated autophagic cell death. RNAi-mediated down-regulation of Par-4 blocks Cer-induced p53-BNIP3 activation and autophagic cell death, while upregulation of Par-4 augmented p53-BNIP3 activation and autophagic cell death. Remarkably, in many instances, Par-4 overexpression alone was sufficient to induce cell death which is associated with features of autophagy. Interestingly, similar results were seen when glioma cells were exposed to classical autophagy inducers such as serum starvation, arsenic trioxide, and curcumin. Collectively, the novel Par-4-p53-BNIP3 axis plays a crucial role in autophagy-mediated cell death in human malignant glioma.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号