STING pathway

STING 通路
  • 文章类型: Journal Article
    生物膜诱导的“相对免疫受损区”产生了免疫抑制微环境,这是骨科内生菌难治性感染的重要原因。因此,操纵免疫细胞共抑制或共激活信号是生物膜管理的关键策略。这项研究报道了将Mn2掺入含有干扰素基因刺激因子(STING)途径激活剂cGAMP(Mncp)的介孔多巴胺纳米颗粒(Mnp)中,并由M1样巨噬细胞细胞膜(m-Mncp)包裹。细胞膜增强了材料对生物膜的靶向能力,允许它在感染灶局部积累。此外,m-Mncp通过光热疗法机械地破坏生物膜,并通过光动力疗法产生的活性氧(ROS)诱导抗原暴露。重要的是,免疫抑制和免疫激活的调节导致抗原呈递细胞(APC)的增强和抗原呈递的开始,从而诱导生物膜特异性体液免疫和记忆反应。此外,这种方法有效抑制髓源性抑制细胞(MDSC)的活化,同时增强T细胞的活性.我们的研究展示了利用m-Mncp免疫疗法与光热和光动力疗法结合使用的功效,以有效减轻在提取受感染的植入物后的残留和复发性感染。因此,这项研究提出了一个可行的替代传统的抗生素治疗生物膜,这是一个具有挑战性的管理。
    The biofilm-induced \"relatively immune-compromised zone\" creates an immunosuppressive microenvironment that is a significant contributor to refractory infections in orthopedic endophytes. Consequently, the manipulation of immune cells to co-inhibit or co-activate signaling represents a crucial strategy for the management of biofilm. This study reports the incorporation of Mn2+ into mesoporous dopamine nanoparticles (Mnp) containing the stimulator of interferon genes (STING) pathway activator cGAMP (Mncp), and outer wrapping by M1-like macrophage cell membrane (m-Mncp). The cell membrane enhances the material\'s targeting ability for biofilm, allowing it to accumulate locally at the infectious focus. Furthermore, m-Mncp mechanically disrupts the biofilm through photothermal therapy and induces antigen exposure through photodynamic therapy-generated reactive oxygen species (ROS). Importantly, the modulation of immunosuppression and immune activation results in the augmentation of antigen-presenting cells (APCs) and the commencement of antigen presentation, thereby inducing biofilm-specific humoral immunity and memory responses. Additionally, this approach effectively suppresses the activation of myeloid-derived suppressor cells (MDSCs) while simultaneously boosting the activity of T cells. Our study showcases the efficacy of utilizing m-Mncp immunotherapy in conjunction with photothermal and photodynamic therapy to effectively mitigate residual and recurrent infections following the extraction of infected implants. As such, this research presents a viable alternative to traditional antibiotic treatments for biofilm that are challenging to manage.
    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
    PD-1治疗在非小细胞肺癌(NSCLC)患者中的疗效仍不令人满意。激活STING途径是改善PD-1抑制剂功效的有希望的策略。这里,我们发现了粉防己碱(TET),一种从中药中常用的药用植物中提取的抗肿瘤化合物,具有抑制NSCLC肿瘤生长的能力。机械上,TET诱导核DNA损伤并增加胞质dsDNA,从而激活STING/TBK1/IRF3途径,进而促进树突状细胞(DC)的肿瘤浸润,巨噬细胞,以及小鼠的CD8+T细胞。体内成像动态监测TET治疗后STING途径的活性增加,并预测肿瘤免疫微环境的激活。我们进一步发现,TET与αPD-1单克隆抗体(αPD-1mAb)的组合可通过促进CD8T细胞浸润并增强其细胞杀伤作用而产生显着的抗癌作用。这反过来又减少了肿瘤的生长和延长NSCLC小鼠的生存期。因此,TET通过激活STING途径有效消除NSCLC细胞并增强免疫治疗功效,TET与抗PD-1免疫疗法的联合应用值得进一步探索。
    The efficacy of PD-1 therapy in non-small cell lung cancer (NSCLC) patients remains unsatisfactory. Activating the STING pathway is a promising strategy to improve PD-1 inhibitor efficacy. Here, we found tetrandrine (TET), an anti-tumor compound extracted from a medicinal plant commonly used in traditional Chinese medicine, has the ability to inhibit NSCLC tumor growth. Mechanistically, TET induces nuclear DNA damage and increases cytosolic dsDNA, thereby activating the STING/TBK1/IRF3 pathway, which in turn promotes the tumor infiltration of dendritic cells (DCs), macrophages, as well as CD8+ T cells in mice. In vivo imaging dynamically monitored the increased activity of the STING pathway after TET treatment and predicted the activation of the tumor immune microenvironment. We further revealed that the combination of TET with αPD-1 monoclonal antibody (αPD-1 mAb) yields significant anti-cancer effects by promoting CD8+ T cell infiltration and enhancing its cell-killing effect, which in turn reduced the growth of tumors and prolonged survival of NSCLC mice. Therefore, TET effectively eliminates NSCLC cells and enhances immunotherapy efficacy through the activation of the STING pathway, and combining TET with anti-PD-1 immunotherapy deserves further exploration for applications.
    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
    顺铂耐药是针对晚期膀胱癌(BC)的全身治疗的主要挑战。关于顺铂耐药性调节的信息很少,其潜在机制需要阐明。这里,我们检测到肿瘤抑制因子的下调,PPP2R2B(丝氨酸/苏氨酸蛋白磷酸酶2A调节亚基),在BC中促进细胞增殖和迁移。更重要的是,PPP2R2B低表达与顺铂耐药相关。体外和体内实验证实PPP2R2B可以促进BC对顺铂的敏感性。在机制方面,我们确定了PPP2R2B作为核质转运分子的新功能。PPP2R2B通过介导IPO5与ISG15的结合促进ISG15进入细胞核。ISG15的核易位抑制DNA修复,通过激活STING途径进一步增加ISG15表达。此外,PPP2R2B被SUV39H1介导的组蛋白3赖氨酸9三甲基化下调,可以通过SUV39H1特异性抑制剂恢复,Chaetocin.我们的数据表明,PPP2R2B表达水平是化疗反应的潜在生物标志物,化疗联合chaetocin可能是BC患者可行的治疗策略。
    Cisplatin resistance is a major challenge for systemic therapy against advanced bladder cancer (BC). Little information is available on the regulation of cisplatin resistance and the underlying mechanisms require elucidation. Here, we detected that downregulation of the tumor suppressor, PPP2R2B (a serine/threonine protein phosphatase 2 A regulatory subunit), in BC promoted cell proliferation and migration. What\'s more, low PPP2R2B expression was correlated with cisplatin resistance. In vitro and in vivo experiments verified that PPP2R2B could promote BC sensitivity to cisplatin. In terms of mechanism, we identified a novel function of PPP2R2B as a nucleocytoplasmic transport molecule. PPP2R2B promoted ISG15 entry into the nucleus by mediating binding of IPO5 with ISG15. Nuclear translocation of ISG15 inhibited DNA repair, further increasing ISG15 expression through activation of the STING pathway. Besides, PPP2R2B was down-regulated by SUV39H1-mediated histone 3 lysine 9 trimethylation, which could be restored by the SUV39H1-specific inhibitor, chaetocin. Our data suggest that PPP2R2B expression level is a potential biomarker for chemotherapy response and that chemotherapy in combination with chaetocin may be a feasible treatment strategy for patients with BC.
    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
    I型干扰素(IFN-Is)在系统性红斑狼疮(SLE)发病机理中起重要作用。双重过滤血浆置换(DFPP)是SLE的一种治疗选择;然而,其对IFN-Is的影响尚不清楚。因此,我们研究了DFPP对IFN-Is的影响。使用基于细胞的报告系统分析了定期接受DFPP的SLE患者(n=11)的血浆,以检测IFN-I的生物利用度和诱导活性。测量血浆dsDNA的浓度,和蛋白质印迹分析用于评估STING途径的磷酸化。与健康对照相比,在患者中观察到更高的IFN-I生物利用度和诱导活性。DFPP后两个参数均下降。在具有高疾病活动性的患者中,IFN-I诱导活性的降低尤其显著。值得注意的是,在STING敲除的报告细胞中未观察到这种减少.此外,DFPP治疗后血浆dsDNA水平下降,这表明STING途径的抑制是观察到的活性降低的原因。Western印迹分析揭示DFPP后STING途径磷酸化的抑制。DFPP降低了IFN-I的生物利用度和血浆的诱导活性。这种减少可能归因于通过消除dsDNA对STING途径的抑制。
    Type I interferons (IFN-Is) play a significant role in systemic lupus erythematosus (SLE) pathogenesis. Double-filtration plasmapheresis (DFPP) is a treatment option for SLE; however, its effect on IFN-Is remains unclear. Therefore, we investigated the effects of DFPP on IFN-Is. Plasma from patients with SLE (n = 11) who regularly underwent DFPP was analysed using a cell-based reporter system to detect the bioavailability and inducing activity of IFN-I. The concentration of plasma dsDNA was measured, and western blotting analysis was used to assess the phosphorylation of the STING pathway. A higher IFN-I bioavailability and inducing activity were observed in patients compared to healthy controls, and both parameters decreased after DFPP. The reduction in IFN-I-inducing activity was particularly prominent in patients with high disease activity. Notably, this reduction was not observed in STING-knockout reporter cells. Additionally, plasma dsDNA levels decreased after DFPP treatment, suggesting that inhibition of the STING pathway was responsible for the observed decrease in activity. Western blotting analysis revealed suppression of STING pathway phosphorylation after DFPP. DFPP reduced IFN-I bioavailability and the inducing activity of plasma. This reduction is likely attributable to the inhibition of the STING pathway through the elimination of dsDNA.
    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
    摘要-本研究探讨了非典型STING-PERK信号通路在脓毒症相关急性肾损伤(SA-AKI)中的作用。分析了来自GEO数据库的基因表达数据和SA-AKI患者的血清STING蛋白水平。使用LPS诱导的小鼠模型和使用HK-2细胞的体外模型来研究STING在SA-AKI中的作用。使用shRNA沉默技术和STING抑制剂C176抑制STING表达。肾功能,炎症标志物,凋亡,并测量衰老。通过沉默HK-2细胞中的PERK并施用PERK抑制剂GSK2606414来研究STING-PERK途径的作用。SA-AKI患者STINGmRNA表达和血清STING蛋白水平明显升高。抑制STING表达改善肾功能,减少炎症,并抑制细胞凋亡和衰老。沉默PERK或给予GSK2606414抑制炎症反应,细胞凋亡,和衰老,提示PERK是STING信号通路的下游效应子。STING-PERK信号通路加剧SA-AKI中的细胞衰老和凋亡。抑制该途径可以为SA-AKI治疗提供潜在的治疗靶标。
    Abstract-This study explored the role of the non-canonical STING-PERK signaling pathway in sepsis-associated acute kidney injury (SA-AKI). Gene expression data from the GEO database and serum STING protein levels in patients with SA-AKI were analyzed. An LPS-induced mouse model and an in vitro model using HK-2 cells were used to investigate the role of STING in SA-AKI. STING expression was suppressed using shRNA silencing technology and the STING inhibitor C176. Kidney function, inflammatory markers, apoptosis, and senescence were measured. The role of the STING-PERK pathway was investigated by silencing PERK in HK-2 cells and administering the PERK inhibitor GSK2606414. STING mRNA expression and serum STING protein levels were significantly higher in patients with SA-AKI. Suppressing STING expression improved kidney function, reduced inflammation, and inhibited apoptosis and senescence. Silencing PERK or administering GSK2606414 suppressed the inflammatory response, cell apoptosis, and senescence, suggesting that PERK is a downstream effector in the STING signaling pathway. The STING-PERK signaling pathway exacerbates cell senescence and apoptosis in SA-AKI. Inhibiting this pathway could provide potential therapeutic targets for SA-AKI treatment.
    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
    细胞周期蛋白依赖性激酶4和6(CDK4/6)抑制剂通过减少经典CDK4/6底物视网膜母细胞瘤(Rb)蛋白的磷酸化,显示出对几种实体瘤的显着活性,而CDK4/6抑制剂对Rb缺陷型肿瘤的抗肿瘤作用尚不清楚。大多数小细胞肺癌(SCLC)缺乏Rb,尽管最近在使用免疫疗法方面取得了进展,但对免疫检查点阻断(ICB)的反应非常温和。这里,我们旨在研究CDK4/6抑制对SCLC细胞的直接作用,并确定其在SCLC联合治疗中的疗效.
    CDK4/6抑制剂abemaciclib对细胞周期的直接影响,最初检查了四种SCLC细胞系中的细胞活力和凋亡。探讨abemaciclib对双链DNA(ds-DNA)损伤诱导的影响以及abemaciclib与放疗(RT)的联合作用。westernblot,免疫荧光(IF)和定量实时聚合酶链反应(qRT-PCR)。建立了Rb缺陷的免疫活性鼠SCLC模型,以评估abemaciclib在联合治疗中的疗效。组织学染色,流式细胞术分析和RNA测序分析肿瘤微环境(TME)中浸润免疫细胞的变化。
    这里,我们证明了abemaciclib在Rb缺陷型SCLC细胞中诱导增加的ds-DNA损伤。abemaciclib和RT联合诱导更多的细胞质ds-DNA,并协同激活STING途径。我们进一步表明,低剂量的abemaciclib与低剂量RT(LDRT)加抗程序性细胞死亡蛋白-1(抗PD-1)抗体的组合可显著增强CD8+T细胞浸润,并显著抑制肿瘤生长和延长在Rb缺陷型免疫活性鼠SCLC模型中的存活。
    我们的结果定义了先前不确定的CDK4/6抑制剂abemaciclib在Rb缺陷型SCLC中的DNA损伤诱导特性,并证明低剂量的abemaciclib联合LDRT可以刺激TME并增强抗PD-1免疫疗法在SCLC模型中的疗效,这代表了SCLC的潜在新治疗策略。
    UNASSIGNED: Cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors have shown significant activity against several solid tumors by reducing the phosphorylation of the canonical CDK4/6 substrate retinoblastoma (Rb) protein, while the anti-tumor effect of CDK4/6 inhibitors on Rb-deficient tumors is not clear. Most small cell lung cancers (SCLCs) are Rb-deficient and show very modest response to immune checkpoint blockade (ICB) despite recent advances in the use of immunotherapy. Here, we aimed to investigate the direct effect of CDK4/6 inhibition on SCLC cells and determine its efficacy in combination therapy for SCLC.
    UNASSIGNED: The immediate impact of CDK4/6 inhibitor abemaciclib on cell cycle, cell viability and apoptosis in four SCLC cell lines was initially checked. To explore the effect of abemaciclib on double-strand DNA (ds-DNA) damage induction and the combination impact of abemaciclib coupled with radiotherapy (RT), western blot, immunofluorescence (IF) and quantitative real-time polymerase chain reaction (qRT-PCR) were performed. An Rb-deficient immunocompetent murine SCLC model was established to evaluate efficacy of abemaciclib in combination therapy. Histological staining, flow cytometry analysis and RNA sequencing were performed to analyze alteration of infiltrating immune cells in tumor microenvironment (TME).
    UNASSIGNED: Here, we demonstrated that abemaciclib induced increased ds-DNA damage in Rb-deficient SCLC cells. Combination of abemaciclib and RT induced more cytosolic ds-DNA, and activated the STING pathway synergistically. We further showed that combining low doses of abemaciclib with low-dose RT (LDRT) plus anti-programmed cell death protein-1 (anti-PD-1) antibody substantially potentiated CD8+ T cell infiltration and significantly inhibited tumor growth and prolonged survival in an Rb-deficient immunocompetent murine SCLC model.
    UNASSIGNED: Our results define previously uncertain DNA damage-inducing properties of CDK4/6 inhibitor abemaciclib in Rb-deficient SCLCs, and demonstrate that low doses of abemaciclib combined with LDRT inflame the TME and enhance the efficacy of anti-PD-1 immunotherapy in SCLC model, which represents a potential novel therapeutic strategy for SCLC.
    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
    STING(干扰素基因刺激因子)途径是调节先天免疫的途径之一,胞外水解酶外核苷酸焦磷酸酶/磷酸二酯酶1(ENPP1)已被确定为其主要的负调节因子。由于激活先天免疫系统是治疗各种传染病和癌症的有前途的策略,ENPP1抑制剂作为候选药物引起了极大的关注。我们先前已经通过使用荧光探针的化学筛选鉴定了具有[1,2,4]三唑并[1,5-a]嘧啶支架的小分子ENPP1抑制剂,TG-mAMP。在这项研究中,我们详细评估了命中和先导化合物的结构-活性关系,并成功开发出不仅在体外强烈且选择性地抑制ENPP1的化合物,而且在蜂窝系统中。
    The STING (stimulator of interferon genes) pathway is one of the pathways that regulate innate immunity, and the extracellular hydrolytic enzyme ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) has been identified as its dominant negative regulator. Since activation of the innate immune system is a promising strategy for the treatment of various infectious diseases and cancers, ENPP1 inhibitors have attracted great attention as candidate drugs. We have previously identified small-molecule ENPP1 inhibitors having a [1,2,4]triazolo[1,5-a]pyrimidine scaffold by means of chemical screening using a fluorescence probe, TG-mAMP. In this study, we evaluated the structure-activity relationships of the hit and lead compounds in detail, and succeeded in developing compounds that strongly and selectively inhibit ENPP1 not only in vitro, but also in cellular systems.
    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
    肿瘤疫苗已显示出适度的反应率,主要归因于它们向树突状细胞(DC)的低效递送,低交叉陈述,DC-内源性免疫抑制信号,和免疫抑制肿瘤微环境(TME)。这里,引流淋巴结(DLN)靶向和肿瘤靶向纳米疫苗被提出来解决这些限制,合成杂环类脂质(A18)和聚酯(BR647)以实现双靶向癌症免疫治疗。同时,掺入寡透明质酸(HA)和DMG-PEG2000-甘露糖,以制备用STAT3siRNA和模型抗原包封的双靶向纳米疫苗。纳米疫苗被设计为靶向DLN和肿瘤,促进货物进入细胞质。这些双靶向纳米疫苗改善了抗原呈递和DC成熟,激活干扰素基因刺激因子(STING)途径,增强了促凋亡作用,并刺激抗肿瘤免疫反应。此外,这些双靶向纳米疫苗克服了免疫抑制TME,减少免疫抑制细胞,并促进肿瘤相关中性粒细胞从N2向N1的极化。在四种诱导强大抗肿瘤反应的双靶向纳米疫苗中,杂环类脂质@聚酯杂化纳米疫苗(MALO@HBNS)显示出最有希望的结果。此外,涉及MALO@HBNS和抗PD-L1抗体的组合策略显示出非常强大的抗癌作用.这项工作介绍了一种用于抗肿瘤治疗的双靶向纳米疫苗平台,提示其与免疫检查点阻断的潜在组合作为一种全面的抗癌策略。
    Tumor vaccines have demonstrated a modest response rate, primarily attributed to their inefficient delivery to dendritic cells (DCs), low cross-presentation, DC-intrinsic immunosuppressive signals, and an immunosuppressive tumor microenvironment (TME). Here, draining lymph node (DLN)-targeted and tumor-targeted nanovaccines were proposed to address these limitations, and heterocyclic lipidoid (A18) and polyester (BR647) were synthesized to achieve dual-targeted cancer immunotherapy. Meanwhile, oligo hyaluronic acid (HA) and DMG-PEG2000-Mannose were incorporated to prepare dual-targeted nanovaccines encapsulated with STAT3 siRNA and model antigens. The nanovaccines were designed to target the DLN and the tumor, facilitating the delivery of cargo into the cytoplasm. These dual-targeted nanovaccines improved antigen presentation and DC maturation, activated the stimulator of interferon genes (STING) pathway, enhanced the pro-apoptotic effect, and stimulated antitumor immune responses. Additionally, these dual-targeted nanovaccines overcame immunosuppressive TME, reduced immunosuppressive cells, and promoted the polarization of tumor-associated neutrophils from N2 to N1. Among the four dual-targeted nanovaccines that induced robust antitumor responses, the heterocyclic lipidoid@polyester hybrid nanovaccines (MALO@HBNS) demonstrated the most promising results. Furthermore, a combination strategy involving MALO@HBNS and an anti-PD-L1 antibody exhibited an immensely powerful anticancer role. This work introduced a dual-targeted nanovaccine platform for antitumor treatment, suggesting its potential combination with an immune checkpoint blockade as a comprehensive anticancer strategy.
    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
    尽管抗PD-L1抗体用于免疫治疗的巨大潜力,由于免疫抑制的肿瘤微环境,它们的低反应率阻碍了它们的应用。为了解决这个问题,我们构建了一种细胞膜包被的纳米系统(mB4S),以将免疫抑制微环境逆转为免疫支持环境,以增强抗肿瘤作用。在这个系统中,作为免疫原性细胞死亡(ICD)诱导剂的表柔比星(EPI)通过腙键与支链的糖共聚物偶联,作为干扰素基因(STING)激动剂的刺激剂的diABZI封装到mB4S中。在mB4S内化后,EPI是酸性响应性释放以诱导ICD,其特征是钙网蛋白(CRT)暴露水平增加和ATP分泌增强。同时,DiABZI有效激活STING途径。mB4S与抗PD-L1抗体联合治疗通过增加成熟树突状细胞(DCs)和CD8+T细胞的比例引起有效的免疫应答。促进细胞因子分泌,上调M1样肿瘤相关巨噬细胞(TAMs)和下调免疫抑制髓源性抑制细胞(MDSC)。因此,这种用于ICD诱导物和STING激动剂共递送的纳米系统实现了促进DC成熟和CD8+T细胞浸润,创造一个免疫支持的微环境,从而增强抗PD-L1抗体在4T1乳腺和CT26结肠肿瘤小鼠中的治疗效果。
    Despite the great potential of anti-PD-L1 antibodies for immunotherapy, their low response rate due to an immunosuppressive tumor microenvironment has hampered their application. To address this issue, we constructed a cell membrane-coated nanosystem (mB4S) to reverse an immunosuppressive microenvironment to an immuno-supportive one for strengthening the anti-tumor effect. In this system, Epirubicin (EPI) as an immunogenic cell death (ICD) inducer was coupled to a branched glycopolymer via hydrazone bonds and diABZI as a stimulator of interferon genes (STING) agonist was encapsulated into mB4S. After internalization of mB4S, EPI was acidic-responsively released to induce ICD, which was characterized by an increased level of calreticulin (CRT) exposure and enhanced ATP secretion. Meanwhile, diABZI effectively activated the STING pathway. Treatment with mB4S in combination with an anti-PD-L1 antibody elicited potent immune responses by increasing the ratio of matured dendritic cells (DCs) and CD8+ T cells, promoting cytokines secretion, up-regulating M1-like tumor-associated macrophages (TAMs) and down-regulating immunosuppressive myeloid-derived suppressor cells (MDSCs). Therefore, this nanosystem for co-delivery of an ICD inducer and a STING agonist achieved promotion of DCs maturation and CD8+ T cells infiltration, creating an immuno-supportive microenvironment, thus potentiating the therapy effect of the anti-PD-L1 antibody in both 4T1 breast and CT26 colon tumor 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
    背景:微血栓形成常见于脓毒症和COVID-19。紫雪散(ZXP)是一种传统的中草药配方,具有治疗微血管和感染性疾病的潜力。然而,ZXP在脓毒症相关血栓形成中的作用和机制尚不清楚.
    目的:研究ZXP在脓毒症血栓形成中的治疗效果和潜在机制。
    方法:用UPLC-QTOF-MS检查ZXP的成分。通过各种方法评估ZXP对脓毒症诱导的血栓形成的疗效:使用苏木精-伊红染色检查肝组织病理学,血小板计数由血细胞分析仪测定,酶联免疫吸附试验(ELISA)检测血清组织因子(TF)水平,血栓烷B2(TXB2),D-二聚体,和纤溶酶原激活物抑制剂-1(PAI-1)。中性粒细胞胞外陷阱(NETs)通过免疫荧光在肝组织中定位和表达,通过ELISA评估外周血中的NETs数量,测定血清中cf-DNA和MPO-DNA的含量。流式细胞术检测血小板P-选择素表达和血小板-中性粒细胞聚集,ELISA法测定血浆P-选择素表达。此外,使用STING激动剂研究了干扰素基因刺激因子(STING)信号通路在ZXP抗脓毒症血栓形成作用中的作用机制,蛋白质印迹实验,和免疫沉淀实验。
    结果:UPLC-QTOF-MS鉴定出ZXP的40种化学成分。ZXP的管理导致肝血栓形成的显着改善,血小板计数,和TXB2,TF,脓毒症大鼠的PAI-1和D-二聚体。此外,ZXP抑制肝组织和外周血中的NETs形成。此外,ZXP降低了血小板和血浆中P-选择素的水平,以及血小板中性粒细胞聚集体的形成,从而抑制P-选择素介导的NETs释放。免疫沉淀和免疫荧光染色实验表明,ZXP通过抑制STING介导的血小板可溶性N-乙基马来酰亚胺敏感因子附着蛋白受体(SNAREs)复合物的组装来减弱P-选择素的分泌,最终阻止NETs形成的抑制。
    结论:我们的研究表明,ZXP主要通过调节STING途径有效减轻血小板颗粒分泌,因此阻碍脓毒症中NET相关的血栓形成。这些发现为ZXP的开发和应用的未来研究提供了宝贵的见解。
    BACKGROUND: Microthrombosis is commonly seen in sepsis and COVID-19. Zixue Powder (ZXP) is a traditional Chinese herbal formula with the potential to treat microvascular and infectious diseases. However, the role and mechanism of ZXP in sepsis-associated thrombosis remain unclear.
    OBJECTIVE: Investigating the therapeutic effectiveness and underlying mechanisms of ZXP in septic thrombosis.
    METHODS: ZXP\'s compositions were examined with UPLC-QTOF-MS. The efficacy of ZXP on sepsis-induced thrombosis was assessed through various methods: liver tissue pathology was examined using hematoxylin-eosin staining, platelet count was determined by a blood cell analyzer, and an enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of serum tissue factor (TF), thromboxane B2 (TXB2), D-Dimer, and plasminogen activator inhibitor-1 (PAI-1). Neutrophil extracellular traps (NETs) were localized and expressed in liver tissues by immunofluorescence, and the number of NETs in peripheral blood was evaluated by ELISA, which measured the quantity of cf-DNA and MPO-DNA in serum. Platelet P-selectin expression and platelet-neutrophil aggregation were measured by flow cytometry, and plasma P-selectin expression was measured by ELISA. Furthermore, the mechanism of the stimulator of interferon genes (STING) signaling pathway in ZXP\'s anti-sepsis thrombosis effect was investigated using the STING agonist, Western blot experiments, and immunoprecipitation experiments.
    RESULTS: UPLC-QTOF-MS identified 40 chemical compositions of ZXP. Administration of ZXP resulted in significant improvements in liver thrombosis, platelet counts, and levels of TXB2, TF, PAI-1, and D-Dimer in septic rats. Moreover, ZXP inhibited NETs formation in both liver tissue and peripheral blood. Additionally, ZXP decreased the levels of P-selectin in both platelets and plasma, as well as the formation of platelet-neutrophil aggregates, thereby suppressing P-selectin-mediated NETs release. Immunoprecipitation and immunofluorescence staining experiments revealed that ZXP attenuated P-selectin secretion by inhibiting STING-mediated assembly of platelet soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) complex, ultimately preventing inhibition of NETs formation.
    CONCLUSIONS: Our study showed that ZXP effectively mitigates platelet granule secretion primarily through modulation of the STING pathway, consequently impeding NET-associated thrombosis in sepsis. These findings offer valuable insights for future research on the development and application of ZXP.
    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