Protease-activated receptor

蛋白酶激活受体
  • 文章类型: Journal Article
    血液凝固和癌症有内在联系,高凝相关的血栓并发症通常在某些类型的癌症中观察到,往往导致癌症患者生存率下降。除了在凝血中的共同作用,凝血蛋白酶通常通过激活G蛋白偶联受体超家族蛋白酶:蛋白酶激活受体(PAR)在各种癌症中触发细胞内信号传导。尽管PAR在某些类型癌症的发展和进展中的作用已经确立,它们对癌症免疫反应的影响才刚刚出现。本综述强调了凝血蛋白酶驱动的PAR信号如何在调节先天和适应性免疫反应中发挥关键作用。接下来详细讨论了凝血蛋白酶诱导的信号传导在癌症免疫逃避中的作用,从而支持某些肿瘤的生长和发展。这篇综述的一个特殊部分展示了凝血蛋白酶的作用,凝血酶,因子VIIa,和Xa因子在癌症免疫逃避中的作用。靶向凝血蛋白酶诱导的信号可能是一种潜在的治疗策略,以增强宿主对抗癌症的免疫监视机制。从而增强靶向免疫治疗方案的临床后果。
    Blood coagulation and cancer are intrinsically connected, hypercoagulation-associated thrombotic complications are commonly observed in certain types of cancer, often leading to decreased survival in cancer patients. Apart from the common role in coagulation, coagulation proteases often trigger intracellular signaling in various cancers via the activation of a G protein-coupled receptor superfamily protease: protease-activated receptors (PARs). Although the role of PARs is well-established in the development and progression of certain types of cancer, their impact on cancer immune response is only just emerging. The present review highlights how coagulation protease-driven PAR signaling plays a key role in modulating innate and adaptive immune responses. This is followed by a detailed discussion on the contribution of coagulation protease-induced signaling in cancer immune evasion, thereby supporting the growth and development of certain tumors. A special section of the review demonstrates the role of coagulation proteases, thrombin, factor VIIa, and factor Xa in cancer immune evasion. Targeting coagulation protease-induced signaling might be a potential therapeutic strategy to boost the immune surveillance mechanism of a host fighting against cancer, thereby augmenting the clinical consequences of targeted immunotherapeutic regimens.
    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
    凝血酶抑制抑制肥胖,小鼠WAT炎症和代谢功能障碍。蛋白酶激活受体(PAR)1不能解释凝血酶驱动的肥胖,所以我们在这个背景下探讨了PAR4的罪魁祸首。雄性WT和PAR-4-/-小鼠接受高脂肪饮食(HFD)8周,WT对照组接受标准食物。通过NMR定量体脂肪。附睾WAT通过组织学评估,免疫组织化学,qPCR和脂肪酶活性测定。3T3-L1前脂肪细胞分化±凝血酶,急性刺激±PAR4激活肽(AP)并通过免疫印迹评估,qPCR和U937单核细胞粘附。通过免疫印迹评估肥胖和瘦患者的心外膜脂肪组织(EAT)。在HFD下,PAR4在小鼠WAT中上调。PAR4-/-小鼠在HFD下发生较少内脏肥胖和葡萄糖耐受不良,具有较小的脂肪细胞,较少的巨噬细胞和较低的脂肪表达(瘦素,PPARγ)和WAT中的促炎基因(CCL2,IL-1β)。PAR4缺失不影响HFD修饰的活性和脂肪酶或perilipin的表达。用凝血酶分化的3T3-L1脂肪细胞保留了Ki67表达,进一步上调IL-1β和CCL2,对单核细胞的粘附性更强。在成熟的脂肪细胞中,PAR4-AP增加磷酸化ERK1/2和AKT,上调Ki67、CCl2、IL-β和透明质酸合成酶1,但不上调TNF-αmRNA,和增加的透明质酸酶敏感的单核细胞粘附。肥胖的人EAT比瘦肉EAT表达更多的PAR4、CD68和CD54。肥胖中PAR4上调支持脂肪细胞肥大,WAT扩张和血栓炎症。新兴的PAR4拮抗剂在这种情况下提供了超出其经典抗血小板作用的治疗前景。
    Thrombin inhibition suppresses adiposity, WAT inflammation and metabolic dysfunction in mice. Protease-activated receptor (PAR)1 does not account for thrombin-driven obesity, so we explored the culprit role of PAR4 in this context. Male WT and PAR-4-/- mice received a high fat diet (HFD) for 8 weeks, WT controls received standard chow. Body fat was quantified by NMR. Epididymal WAT was assessed by histology, immunohistochemistry, qPCR and lipase activity assay. 3T3-L1 preadipocytes were differentiated ± thrombin, acutely stimulated ± PAR4 activating peptide (AP) and assessed by immunoblot, qPCR and U937 monocyte adhesion. Epicardial adipose tissue (EAT) from obese and lean patients was assessed by immunoblot. PAR4 was upregulated in mouse WAT under HFD. PAR4-/- mice developed less visceral adiposity and glucose intolerance under HFD, featuring smaller adipocytes, fewer macrophages and lower expression of adipogenic (leptin, PPARγ) and pro-inflammatory genes (CCL2, IL-1β) in WAT. HFD-modified activity and expression of lipases or perilipin were unaffected by PAR4 deletion. 3T3-L1 adipocytes differentiated with thrombin retained Ki67 expression, further upregulated IL-1β and CCL2 and were more adhesive for monocytes. In mature adipocytes, PAR4-AP increased phosphorylated ERK1/2 and AKT, upregulated Ki67, CCl2, IL-β and hyaluronan synthase 1 but not TNF-α mRNA, and augmented hyaluronidase-sensitive monocyte adhesion. Obese human EAT expressed more PAR4, CD68 and CD54 than lean EAT. PAR4 upregulated in obesity supports adipocyte hypertrophy, WAT expansion and thrombo-inflammation. The emerging PAR4 antagonists provide a therapeutic perspective in this context beyond their canonical antiplatelet action.
    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
    G蛋白偶联受体(GPCRs)是广泛表达的细胞表面受体,其介导许多生理反应并且是高度可药用的。在激活后,GPCR快速偶联至异三聚体G蛋白,然后磷酸化并从细胞表面内化。最近的研究表明,GPCRs不仅位于质膜上,而且还存在于细胞内区室中,在那里它们能够发出信号。GPCR的细胞内信号传导被最好地描述为发生在内体。一些研究已经很好地记录了内体GPCR-G蛋白和GPCR-β-抑制蛋白信号传导。除了磷酸化,GPCR也被泛素翻译后修饰。GPCR泛素化主要在受体内体-溶酶体运输的背景下进行研究。然而,新的研究表明,内皮细胞中表达的内源性GPCRs的泛素化引发细胞内p38丝裂原活化激酶信号复合物的组装,促进炎症反应.在这个小型审查中,我们讨论了新发现,这些发现为泛素化调节内体GPCR炎症信号的功能提供了关键见解。
    G protein-coupled receptors (GPCRs) are ubiquitously expressed cell surface receptors that mediate numerous physiological responses and are highly druggable. Upon activation, GPCRs rapidly couple to heterotrimeric G proteins and are then phosphorylated and internalized from the cell surface. Recent studies indicate that GPCRs not only localize at the plasma membrane but also exist in intracellular compartments where they are competent to signal. Intracellular signaling by GPCRs is best described to occur at endosomes. Several studies have elegantly documented endosomal GPCR-G protein and GPCR-β-arrestin signaling. Besides phosphorylation, GPCRs are also posttranslationally modified with ubiquitin. GPCR ubiquitination has been studied mainly in the context of receptor endosomal-lysosomal trafficking. However, new studies indicate that ubiquitination of endogenous GPCRs expressed in endothelial cells initiates the assembly of an intracellular p38 mitogen-activated kinase signaling complex that promotes inflammatory responses from endosomes. In this mini-review, we discuss emerging discoveries that provide critical insights into the function of ubiquitination in regulating GPCR inflammatory signaling at endosomes.
    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
    内皮蛋白C受体(EPCR)是天然抗凝血活化蛋白C(aPC)的受体。它通过蛋白酶激活受体(PAR)1/2的裂解介导aPC的抗炎和屏障保护功能。过敏性接触性皮炎是一种常见的皮肤病,其特征是炎症和皮肤屏障缺陷。本研究使用接触性超敏反应(CHS)模型研究了EPCR和3K3A-aPC对过敏性接触性皮炎的影响。在EPCR缺陷(KO)和匹配的野生型小鼠和用3K3A-aPC治疗的小鼠中,使用1-氟-2,4-二硝基苯诱导CHS,具有降低的抗凝血活性的aPC的突变形式。临床和组织学特征的变化,细胞因子,并检查了免疫细胞。EPCRKO小鼠表现出更严重的CHS,与野生型小鼠相比,皮肤中的免疫细胞浸润增加,炎症细胞因子和IgE水平更高。EPCR,aPC,PAR1/2表达于皮肤表皮,EPCR几乎只存在于基底层。EPCRKO增加了aPC和PAR1的表皮表达,而在CHS中,与野生型小鼠相比,它们的表达降低。3K3A-aPC通过抑制免疫细胞浸润/活化和炎性细胞因子降低了野生型和EPCRKO小鼠中的CHS严重程度。总之,EPCRKO加剧了CHS,而3K3A-aPC可以降低EPCRKO和野生型小鼠CHS的严重程度。
    Endothelial protein C receptor (EPCR) is a receptor for the natural anti-coagulant activated protein C (aPC). It mediates the anti-inflammatory and barrier-protective functions of aPC through the cleavage of protease-activated receptor (PAR)1/2. Allergic contact dermatitis is a common skin disease characterized by inflammation and defective skin barrier. This study investigated the effect of EPCR and 3K3A-aPC on allergic contact dermatitis using a contact hypersensitivity (CHS) model. CHS was induced using 1-Fluoro-2,4-dinitrobenzene in EPCR-deficient (KO) and matched wild-type mice and mice treated with 3K3A-aPC, a mutant form of aPC with diminished anti-coagulant activity. Changes in clinical and histological features, cytokines, and immune cells were examined. EPCRKO mice displayed more severe CHS, with increased immune cell infiltration in the skin and higher levels of inflammatory cytokines and IgE than wild-type mice. EPCR, aPC, and PAR1/2 were expressed by the skin epidermis, with EPCR presenting almost exclusively in the basal layer. EPCRKO increased the epidermal expression of aPC and PAR1, whereas in CHS, their expression was reduced compared to wild-type mice. 3K3A-aPC reduced CHS severity in wild-type and EPCRKO mice by suppressing immune cell infiltration/activation and inflammatory cytokines. In summary, EPCRKO exacerbated CHS, whereas 3K3A-aPC could reduce the severity of CHS in both EPCRKO and wild-type mice.
    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
    心房颤动(AF),以心脏心房的不规则高频收缩为特征,越来越重要的临床。原因是房颤患病率和血栓栓塞并发症的增加。所谓的心房重塑的特征是,除其他外,通过心房扩张和纤维化重塑。因此,AF是自持的并且形成促凝血状态。但是高凝不仅似乎是AF的结果。凝血因子可以通过蛋白酶激活的受体(PAR)对细胞产生影响,从而促凝血状态可能有助于AF的发展和维持。在这项工作中,FXa对心脏样1(HL-1)细胞的影响,是小鼠成年心房心肌细胞(永生化),被调查。检测PAR1、PAR2和PAR4的表达。用FXa(5-50nM;4-24小时)或PAR1-和PAR2-激动剂(20µM;4-24小时)孵育后,PAR表达或炎症信号级联均无变化.MAP激酶ERK1/2或NF-κB的p65亚基的磷酸化没有时间或浓度依赖性变化。此外,细胞粘附分子(ICAM-1,VCAM-1,纤连蛋白)的mRNA表达没有变化。因此,FXa对HL-1细胞没有直接的PAR依赖性作用。未来的研究应该研究FXa对人心肌细胞或其他心脏细胞类型如成纤维细胞的影响。
    Atrial fibrillation (AF), characterised by irregular high-frequency contractions of the atria of the heart, is of increasing clinical importance. The reasons are the increasing prevalence and thromboembolic complications caused by AF. So-called atrial remodelling is characterised, among other things, by atrial dilatation and fibrotic remodelling. As a result, AF is self-sustaining and forms a procoagulant state. But hypercoagulation not only appears to be the consequence of AF. Coagulation factors can exert influence on cells via protease-activated receptors (PAR) and thereby the procoagulation state could contribute to the development and maintenance of AF. In this work, the influence of FXa on Heart Like-1 (HL-1) cells, which are murine adult atrial cardiomyocytes (immortalized), was investigated. PAR1, PAR2, and PAR4 expression was detected. After incubations with FXa (5-50 nM; 4-24 h) or PAR1- and PAR2-agonists (20 µM; 4-24 h), no changes occurred in PAR expression or in the inflammatory signalling cascade. There were no time- or concentration-dependent changes in the phosphorylation of the MAP kinases ERK1/2 or the p65 subunit of NF-κB. In addition, there was no change in the mRNA expression of the cell adhesion molecules (ICAM-1, VCAM-1, fibronectin). Thus, FXa has no direct PAR-dependent effects on HL-1 cells. Future studies should investigate the influence of FXa on human cardiomyocytes or on other cardiac cell types like fibroblasts.
    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:内皮功能障碍在COVID-19的病理生理学中起着重要作用,并且与疾病的严重程度和死亡率密切相关。SARS-CoV-2感染的炎症反应可以改变内皮调节血管张力的能力,免疫反应,以及抗血栓形成和促血栓形成特性之间的平衡。然而,COVID-19过程中特定的内皮通路改变仍需要充分了解。
    目的:在本研究中,我们试图鉴定COVID-19特有的循环因子诱导的内皮细胞分子变化.
    结果:为此,我们用COVID-19或非COVID-19肺炎患者的血清培养内皮细胞。通过转录组学分析,我们能够从COVID-19患者中鉴定出一种由血清诱导的独特内皮表型.我们在体外证实并扩大了这一观察结果,表明COVID-19血清改变了内皮细胞的功能特性,导致细胞凋亡增加,屏障完整性的丧失,和高凝状态。此外,我们证明了这些内皮功能障碍是由蛋白酶激活受体2(PAR-2)介导的,正如转录组网络分析预测的那样,通过体外功能测定验证。
    结论:我们的发现为进一步研究提供了依据,以评估靶向PAR-2是否可能是对抗COVID-19内皮功能障碍的临床有效策略。
    BACKGROUND: Endothelial dysfunction plays a central role in the pathophysiology of COVID-19 and is closely linked to the severity and mortality of the disease. The inflammatory response to SARS-CoV-2 infection can alter the capacity of the endothelium to regulate vascular tone, immune responses, and the balance between anti-thrombotic and pro-thrombotic properties. However, the specific endothelial pathways altered during COVID-19 still need to be fully understood.
    OBJECTIVE: In this study, we sought to identify molecular changes in endothelial cells induced by circulating factors characteristic of COVID-19.
    RESULTS: To this aim, we cultured endothelial cells with sera from patients with COVID-19 or non-COVID-19 pneumonia. Through transcriptomic analysis, we were able to identify a distinctive endothelial phenotype that is induced by sera from COVID-19 patients. We confirmed and expanded this observation in vitro by showing that COVID-19 serum alters functional properties of endothelial cells leading to increased apoptosis, loss of barrier integrity, and hypercoagulability. Furthermore, we demonstrated that these endothelial dysfunctions are mediated by protease-activated receptor 2 (PAR-2), as predicted by transcriptome network analysis validated by in vitro functional assays.
    CONCLUSIONS: Our findings provide the rationale for further studies to evaluate whether targeting PAR-2 may be a clinically effective strategy to counteract endothelial dysfunction in COVID-19.
    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: Randomized Controlled Trial
    BMS-986141是一部小说,口服,蛋白酶激活,受体4(PAR4)-拮抗剂,在临床前研究中表现出强大的抗血栓活性和低出血风险。药代动力学,药效学,BMS-986141在健康参与者中的耐受性概况进行了随机评估,双盲,安慰剂对照,单剂量递增(SAD;N=60)研究;多剂量递增(MAD;N=32)研究;和日本MAD(JMAD;N=32)研究。BMS-9861412.5mg和150mg的暴露量与剂量成正比;最大浓度为17.6ng/mL和958ng/mL;曲线下面积(AUC)至无穷大分别为183h*×ng/mL和9207h*×ng/mL,分别。在整个剂量组中,平均半衰期为33.7至44.7小时。每日一次给药7天后的积累指数表明在稳态下AUC增加1.3至2倍。在SAD研究中,BMS-98614175和150mg对25-100µMPAR4激动剂肽(AP)诱导的血小板聚集产生≥80%的抑制作用,不影响PAR1-AP诱导的血小板聚集,通过≥24小时后给药。在MAD和JMAD研究中,BMS-986141剂量≥10mg在24小时内完全抑制12.5μM和25μMPAR4-AP诱导的血小板聚集。这项研究发现BMS-986141是安全的,耐受性良好。在广泛的剂量范围内,健康参与者具有剂量比例药代动力学和浓度依赖性药效学。ClinicalTrials.govID:NCT02341638。
    为什么要进行这项研究?抗血小板疗法的缺点限制了其临床应用,还有一个新的需求,安全,和有效的抗血小板药物,降低出血风险。PAR4拮抗剂是一类有前途的新型抗血小板药物,因为它可以后期抑制血栓生长,对血小板驱动的止血作用最小。BMS-986141是一部小说,强力,口服生物可利用性,PAR4特异性小分子拮抗剂。BMS-986141是安全和良好的耐受性,在广泛的剂量范围内,健康参与者具有剂量比例药代动力学和浓度依赖性药效学。BMS-986141具有强大的抗血栓活性和低出血风险。BMS-986141有可能改善动脉粥样硬化血栓形成患者抗血小板治疗的获益风险。
    BMS-986141 is a novel, oral, protease-activated, receptor 4 (PAR4)-antagonist that exhibited robust antithrombotic activity and low bleeding risk in preclinical studies. The pharmacokinetic, pharmacodynamic, and tolerability profiles of BMS-986141 in healthy participants were assessed in a randomized, double-blind, placebo-controlled, single-ascending-dose (SAD; N = 60) study; a multiple-ascending-dose (MAD; N = 32) study; and a Japanese MAD (JMAD; N = 32) study. Exposure was dose-proportional for BMS-986141 2.5 mg and 150 mg; maximum concentrations were 17.6 ng/mL and 958 ng/mL; and areas under the curve (AUC) to infinity were 183 h* × ng/mL and 9207 h* × ng/mL, respectively. Mean half-life ranged from 33.7 to 44.7 hours across dose panels. The accumulation index following once-daily administration for 7 days suggested a 1.3- to 2-fold AUC increase at steady state. In the SAD study, BMS-986141 75 and 150 mg produced ≥80% inhibition of 25-100 µM PAR4 agonist peptide (AP)-induced platelet aggregation, without affecting PAR1-AP-induced platelet aggregation, through ≥24 hours postdose. In the MAD and JMAD studies, BMS-986141 doses ≥10 mg completely inhibited 12.5 μM and 25 μM PAR4-AP-induced platelet aggregation through 24 hours. This study found BMS-986141 was safe and well tolerated, with dose-proportional pharmacokinetics and concentration-dependent pharmacodynamics in healthy participants over a wide dose range. ClinicalTrials.gov ID: NCT02341638.
    Why was the study done? Antiplatelet therapies have shortcomings that limit their clinical utility, and there is an unmet need for a new, safe, and effective antiplatelet agent with reduced bleeding risk.PAR4 antagonists are a promising novel class of antiplatelet drugs due to late-stage inhibition of thrombus growth with minimal effects on platelet-driven hemostasis.BMS-986141 is a novel, potent, orally bioavailable, small-molecule antagonist specific for PAR4.What is new? BMS-986141 is safe and well tolerated, with dose-proportional pharmacokinetics and concentration-dependent pharmacodynamics in healthy participants over a wide dose range.BMS-986141 has robust antithrombotic activity and low bleeding risk.What is the impact? BMS-986141 has the potential to improve the benefit–risk of antiplatelet therapy in patients with atherothrombosis.
    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:血栓弹力图(TEG)用于实时确定急性出血风险患者的止血状态。凝血酶被认为通过产生聚合的纤维蛋白和通过PAR激活血小板来驱动TEG中的凝血。然而,血小板激动剂受体和信号在TEG中的具体作用尚未见报道.
    目的:在这里,我们使用小鼠和人血液样品中血小板蛋白的遗传和药理抑制作用,研究了TEG中血小板功能所需的特异性受体和信号通路。
    方法:凝血参数(R,α,MA)在重新钙化中确定,使用TEG5000分析仪的高岭土触发柠檬酸盐的血样。
    结果:我们确认了血小板的需求,血小板收缩,和αIIbβ3整合素函数的正常α角(α)和最大振幅(MA)。巨核细胞/血小板(Talin1mKO)中整联蛋白衔接子Talin1的丢失也降低了α和MA,但在缺乏Rap1GTP酶信号的小鼠样品中仅观察到最小的缺陷。PAR4mKO样品显示α受损但MA正常。然而,在PAR4mKO小鼠的样本中观察到与血小板耗尽样品相似的TEG受损痕迹,这些样本在血小板上耗尽了GPVI或添加了Syk抑制剂.我们在人血液中复制了这些结果,同时抑制了PAR1,PAR4和Syk。
    结论:我们的结果表明,标准TEG对整合素内外激活和血小板止血功能至关重要的血小板信号通路不敏感。此外,我们提供了第一个证据,证明PAR和GPVI在TEG中血小板介导的凝块收缩中起冗余作用.
    Thromboelastography (TEG) is used for real-time determination of hemostatic status in patients with acute risk of bleeding. Thrombin is thought to drive clotting in TEG through generation of polymerized fibrin and activation of platelets through protease-activated receptors (PARs). However, the specific role of platelet agonist receptors and signaling in TEG has not been reported.
    Here, we investigated the specific receptors and signaling pathways required for platelet function in TEG using genetic and pharmacologic inhibition of platelet proteins in mouse and human blood samples.
    Clotting parameters (R time, α-angle [α], and maximum amplitude [MA]), were determined in recalcified, kaolin-triggered citrated blood samples using a TEG 5000 analyzer.
    We confirmed the requirement of platelets, platelet contraction, and αIIbβ3 integrin function for normal α and MA. Loss of the integrin adaptor Talin1 in megakaryocytes/platelets (Talin1mKO) also reduced α and MA, but only minimal defects were observed in samples from mice lacking Rap1 GTPase signaling. PAR4mKO samples showed impaired α but normal MA. However, impaired TEG traces similar to those in platelet-depleted samples were observed with samples from PAR4mKO mice depleted of glycoprotein VI on platelets or with addition of a Syk inhibitor. We reproduced these results in human blood with combined inhibition of PAR1, PAR4, and Syk.
    Our results demonstrate that standard TEG is not sensitive to platelet signaling pathways critical for integrin inside-out activation and platelet hemostatic function. Furthermore, we provide the first evidence that PARs and glycoprotein VI play redundant roles in platelet-mediated clot contraction in TEG.
    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
    在过去的几年中,已经取得了巨大的进展,以解释来自不同来源的看似无害的环境蛋白如何诱导有效的Th2偏向的炎症反应。趋同的发现已经显示了显示蛋白水解活性的过敏原在过敏反应的开始和进展中的关键作用。通过它们激活非IgE依赖性炎症途径的倾向,某些过敏原蛋白酶现在被认为是对自身和非蛋白酶过敏原致敏的引发剂。蛋白酶变应原降解角质形成细胞或气道上皮的连接蛋白,以促进变应原穿过上皮屏障的递送以及它们随后被抗原呈递细胞摄取。这些蛋白酶介导的上皮损伤以及蛋白酶激活受体(PAR)的感知引起有效的炎症反应,导致pro-Th2细胞因子(IL-6,IL-25,IL-1β,TSLP)和危险相关分子模式(DAMPs;IL-33,ATP,尿酸)。最近,蛋白酶过敏原显示切割IL-33的蛋白酶传感器结构域以产生超活性形式的alarmin。同时,纤维蛋白原的蛋白水解裂解可以触发TLR4信号,和各种细胞表面受体的裂解进一步形成Th2极化。值得注意的是,伤害性神经元对蛋白酶过敏原的感知可以代表过敏反应发展的主要步骤。这篇综述的目的是强调由蛋白酶过敏原触发的多种先天免疫机制,这些机制汇聚以引发过敏反应。
    Tremendous progress in the last few years has been made to explain how seemingly harmless environmental proteins from different origins can induce potent Th2-biased inflammatory responses. Convergent findings have shown the key roles of allergens displaying proteolytic activity in the initiation and progression of the allergic response. Through their propensity to activate IgE-independent inflammatory pathways, certain allergenic proteases are now considered as initiators for sensitization to themselves and to non-protease allergens. The protease allergens degrade junctional proteins of keratinocytes or airway epithelium to facilitate allergen delivery across the epithelial barrier and their subsequent uptake by antigen-presenting cells. Epithelial injuries mediated by these proteases together with their sensing by protease-activated receptors (PARs) elicit potent inflammatory responses resulting in the release of pro-Th2 cytokines (IL-6, IL-25, IL-1β, TSLP) and danger-associated molecular patterns (DAMPs; IL-33, ATP, uric acid). Recently, protease allergens were shown to cleave the protease sensor domain of IL-33 to produce a super-active form of the alarmin. At the same time, proteolytic cleavage of fibrinogen can trigger TLR4 signaling, and cleavage of various cell surface receptors further shape the Th2 polarization. Remarkably, the sensing of protease allergens by nociceptive neurons can represent a primary step in the development of the allergic response. The goal of this review is to highlight the multiple innate immune mechanisms triggered by protease allergens that converge to initiate the allergic response.
    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:心房肌病和心房颤动(AF)伴随血栓炎症。这促进疾病进展并促进主要不良心血管事件(MACE)。凝血酶受体(蛋白酶激活的受体1,PAR1)信号传导在介导血栓炎症中起重要作用。我们假设PAR1信号通过细胞毒性CD8+T淋巴细胞将凝血和炎症联系在首次诊断为AF(FDAF)的患者中。
    方法:共研究210例患者。我们纳入了FDAF患者的数据和血液样本(n=160),阵发性房颤患者的心脏组织(n=32)和20名对照。
    结果:在早期房颤期间,与患有慢性心血管疾病但无房颤的患者相比,T淋巴细胞的促炎和细胞毒性亚群(CD8+)循环频率更高。伴随着CD8+效应分子的血浆水平升高,这对应于不良心脏重塑和心房功能障碍的生物标志物。组织因子(TF)和PAR1的激活与促炎和细胞毒性效应功能有关。在发生MACE的FDAF患者中,PAR1相关的CD8+细胞活化更为频繁。
    结论:在FDAF患者中,TF因子Xa因子IIa轴通过CD8+T细胞中的PAR1促进血栓炎症。干预该级联可能是减少疾病进展和AF血管并发症的有希望的协同方法。
    Atrial myopathy and atrial fibrillation (AF) accompany thrombo-inflammation. This facilitates disease progression and promotes major adverse cardiovascular events (MACEs). Thrombin receptor (protease-activated receptor 1, PAR1) signalling is central in mediating thrombo-inflammation. We hypothesised that PAR1 signalling links coagulation and inflammation through cytotoxic CD8+ T lymphocytes in patients presenting with first-diagnosed AF (FDAF).
    A total of 210 patients were studied. We included data and blood samples from patients presenting with FDAF (n = 160), cardiac tissue from patients with paroxysmal AF (n = 32) and 20 controls.
    During early AF, a pro-inflammatory and cytotoxic subset of T lymphocytes (CD8+) circulated more frequently when compared to patients with chronic cardiovascular disease but without AF, accompanied by elevated plasma levels of CD8+ effector molecules, which corresponded to biomarkers of adverse cardiac remodelling and atrial dysfunction. Activation of tissue factor (TF) and PAR1 was associated with pro-inflammatory and cytotoxic effector functions. PAR1-related CD8+ cell activation was more frequent in FDAF patients that experienced a MACE.
    In patients with FDAF, the TF-factor Xa-factor IIa-axis contributes to thrombo-inflammation via PAR1 in CD8+ T cells. Intervening in this cascade might be a promising synergistic approach to reducing disease progression and the vascular complications of AF.
    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