cGAS-Sting pathway

cGAS - STING 通路
  • 文章类型: Journal Article
    背景:锰离子(Mn2+)与能够损伤和裂解肿瘤细胞的佐剂结合形成抗肿瘤纳米调节剂,通过环GMP-AMP干扰素基因合成酶刺激因子(cGAS-STING)途径的级联激活,增强癌症治疗的免疫功效,这强调了开发抗肿瘤纳米调节剂的重要性,诱导DNA损伤并增强cGAS-STING活性,作为未来重要的研究方向。方法和结果:我们成功合成了一种抗肿瘤纳米调节剂,具有良好的分散性和生物安全性。这种纳米调制器是通过加载二氧化锰纳米片(M-NS)与zebularine(Zeb),以其免疫原性增强作用而闻名,并使用透明质酸(HA)进行靶向表面改性。体循环到肿瘤部位后,Mn2+,Zeb,H+和H2O2在肿瘤微环境中催化释放活性氧(ROS)。这些成分可以直接或间接地损伤肿瘤细胞的DNA或线粒体,从而诱导程序性细胞死亡。此外,它们促进细胞质中双链DNA(dsDNA)的积累,增强cGAS-STING信号通路的激活并促进I型干扰素的产生和促炎细胞因子的分泌。此外,Zeb@MH-NS增强树突状细胞的成熟,细胞毒性T淋巴细胞的浸润,以及在肿瘤部位招募自然杀伤细胞。
    结论:这种HA修饰的基于锰的杂合纳米调节剂可以通过增强先天免疫活性来增强抗肿瘤治疗,并可能为癌症的免疫治疗和临床转化提供新的方向。
    BACKGROUND: Manganese ions (Mn2+) combined with adjuvants capable of damaging and lysing tumor cells form an antitumor nano-modulator that enhances the immune efficacy of cancer therapy through the cascade activation of the cyclic GMP-AMP interferon gene synthase-stimulator (cGAS-STING) pathway, which underscores the importance of developing antitumor nano-modulators, which induce DNA damage and augment cGAS-STING activity, as a critical future research direction. METHODS AND RESULTS: We have successfully synthesized an antitumor nano-modulator, which exhibits good dispersibility and biosafety. This nano-modulator is engineered by loading manganese dioxide nanosheets (M-NS) with zebularine (Zeb), known for its immunogenicity-enhancing effects, and conducting targeted surface modification using hyaluronic acid (HA). After systemic circulation to the tumor site, Mn2+, Zeb, and reactive oxygen species (ROS) are catalytically released in the tumor microenvironment by H+ and H2O2. These components can directly or indirectly damage the DNA or mitochondria of tumor cells, thereby inducing programmed cell death. Furthermore, they promote the accumulation of double-stranded DNA (dsDNA) in the cytoplasm, enhancing the activation of the cGAS-STING signalling pathway and boosting the production of type I interferon and the secretion of pro-inflammatory cytokines. Additionally, Zeb@MH-NS enhances the maturation of dendritic cells, the infiltration of cytotoxic T lymphocytes, and the recruitment of natural killer cells at the tumor site.
    CONCLUSIONS: This HA-modified manganese-based hybrid nano-regulator can enhance antitumor therapy by boosting innate immune activity and may provide new directions for immunotherapy and clinical translation in cancer.
    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
    肿瘤免疫医治在肿瘤医治中获得了愈来愈多的存眷。然而,乳酸在肿瘤组织中的积累抑制了免疫细胞的反应以形成免疫抑制微环境(ISME)。为了扭转ISME,据报道,酸响应性纳米平台(称为MLLN@HA)用于协同增强的肿瘤免疫疗法.MLLN@HA是通过将乳酸氧化酶(LOX)和DNA修复抑制剂(NU7441)共负载在锰掺杂的层状双氢氧化物(Mn-LDH)中构建的,然后用透明质酸(HA)修饰用于肿瘤靶向递送。肿瘤细胞内吞作用后,MLLN@HA分解并释放LOX,酸性肿瘤微环境中的NU7441和Mn2+离子。释放的LOX催化乳酸转化为过氧化氢(H2O2),这不仅缓解了ISME,但也为Mn2介导的Fenton样反应提供了反应物,以增强化学动力学疗法(CDT)。释放NU7441可防止CDT诱导的DNA损伤被修复,从而增加肿瘤细胞内的双链DNA(dsDNA)片段。重要的是,释放的Mn2+离子增强了环GMP-AMP合酶(cGAS)对dsDNA片段的敏感性,并激活干扰素基因(STING)的刺激物以诱导抗肿瘤免疫反应。这种精心策划的免疫增强策略最终实现了有效的肿瘤生长抑制并防止了肿瘤肺转移。重要声明:为了提高肿瘤免疫治疗的疗效,开发了一种创新的酸响应性MLLN@HA纳米平台,用于协同增强的肿瘤免疫治疗。MLLN@HA通过HA与CD44的相互作用主动靶向肿瘤细胞,然后降解释放LOX,酸性肿瘤微环境中的NU7441和Mn2+离子。释放的LOX为Mn2介导的Fenton反应产生H2O2,并通过消耗乳酸来逆转ISME。NU7441防止DNA损伤修复,导致游离DNA片段的浓度增加,而Mn2+离子激活cGAS-STING途径,增强全身抗肿瘤免疫反应。精心策划的免疫增强纳米平台有效抑制肿瘤生长和肺转移,提出了一种有希望的癌症治疗策略。
    Tumor immunotherapy has gained more and more attention in tumor treatment. However, the accumulation of lactic acid in tumor tissue inhibits the response of immune cells to form an immunosuppressive microenvironment (ISME). To reverse the ISME, an acid-responsive nanoplatform (termed as MLLN@HA) is reported for synergistically enhanced tumor immunotherapy. MLLN@HA is constructed by the co-loading of lactate oxidase (LOX) and DNA repair inhibitor (NU7441) in a manganese-doped layered double hydroxide (Mn-LDH), and then modified with hyaluronic acid (HA) for tumor-targeted delivery. After endocytosis by tumor cells, MLLN@HA decomposes and releases LOX, NU7441 and Mn2+ ions in the acidic tumor microenvironment. The released LOX catalyzes the conversion of lactic acid into hydrogen peroxide (H2O2), which not only alleviates the ISME, but also provides reactants for the Mn2+-mediated Fenton-like reaction to enhance chemodynamic therapy (CDT). Released NU7441 prevents CDT-induced DNA damage from being repaired, thereby increasing double-stranded DNA (dsDNA) fragments within tumor cells. Importantly, the released Mn2+ ions enhance the sensitivity of cyclic GMP-AMP synthase (cGAS) to dsDNA fragments, and activate the stimulator of interferon genes (STING) to induce an anti-tumor immune response. Such an orchestrated immune-boosting strategy ultimately achieves effective tumor growth inhibition and prevents tumor lung metastasis. STATEMENT OF SIGNIFICANCE: To improve the efficacy of tumor immunotherapy, an innovative acid-responsive MLLN@HA nanoplatform was developed for synergistically enhanced tumor immunotherapy. The MLLN@HA actively targets to tumor cells through the interaction of HA with CD44, and then degrades to release LOX, NU7441 and Mn2+ ions in the acidic tumor microenvironment. The released LOX generates H2O2 for the Mn2+-mediated Fenton reaction and reverses the ISME by consuming lactate. NU7441 prevents DNA damage repair, leading to an increased concentration of free DNA fragments, while Mn2+ ions activate the cGAS-STING pathway, enhancing the systemic anti-tumor immune response. The orchestrated immune-boosting nanoplatform effectively inhibits tumor growth and lung metastasis, presenting a promising strategy for cancer 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
    砷是农药中广泛使用的有毒类金属元素,防腐剂和半导体行业。然而,砷通过食物链积累会对动物和人类健康造成严重损害。然而,砷对鸡肝的毒性机制尚不清楚,本研究旨在探讨cGAS-STING和NF-κB通路在鸡肝脏炎症损伤中的潜在作用。在这项研究中,将75只白羽肉鸡分为对照组,低剂量砷组(4mg/kg)和高剂量砷组(8mg/kg)研究砷对鸡肝的毒性作用。在这项研究中,我们发现暴露于ATO时肝脏会发生炎症细胞浸润和囊泡变性等病理变化。至关重要的是,暴露于ATO触发了cGAS-STING途径,并显着提高了cGAS的mRNA和蛋白表达水平,STING,TBK1和IRF7。I型干扰素应答也被触发。同时,STING诱导常规NF-κB信号通路的激活,刺激炎症相关基因的表达,如IL-6、TNF-α和IL-1β。总之,在高ATO暴露下通过cGAS-STING和NF-κB信号通路诱导炎症反应,为进一步研究砷的毒理机制提供了新的思路。
    Arsenic is a toxic metal-like element widely used in the pesticide, preservative and semiconductor industries. However, accumulation of arsenic through the food chain can cause serious damage to animal and human health. However, the toxic mechanism of arsenic-induced hepatotoxicity in chickens is not clear, and the present study aimed to investigate the potential role of cGAS-STING and NF-κB pathways on inflammatory injury in chicken liver. In this study, 75 white-feathered broilers were divided into a control group, a low-dose arsenic group (4 mg/kg) and a high-dose arsenic group (8 mg/kg) to investigate the toxic effects of arsenic on chicken liver. In this study, we found that pathological changes such as inflammatory cell infiltration and vesicular degeneration occurred in the liver when exposed to ATO. Crucially, exposure to ATO triggered the cGAS-STING pathway and markedly raised the levels of mRNA and protein expression of cGAS, STING, TBK1, and IRF7. The type I interferon response was also triggered. Simultaneously, STING induced the activation of the conventional NF-κB signaling pathway and stimulated the expression of genes associated with inflammation, such as IL-6, TNF-α and IL-1β. In summary, the induction of inflammatory responses via cGAS-STING and NF-κB signaling pathways under high ATO exposure provides new ideas for further studies on the toxicological mechanisms of arsenic.
    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
    激活循环肿瘤细胞簇(CTC簇)的cGAS-STING途径代表了减轻转移的有希望的策略。为了充分利用胆固醇调节剂激活CTC的STING水平的潜力,我们开发了由金属复合物脂质(MCL)组成的纳米颗粒(NP)。此设计包括MCL-米利铂,以增加NP硬度和负荷洛米他必特(lomi)调节胆固醇水平,导致PLT@Pt-脂质@LomiNPs的产生。MCL-米利铂不仅增强了Lomi对STING途径的诱导功效,而且还增加了NPs的刚度,因此是影响CTC集群渗透以进一步提高Lomi能力的重要因素。通过cy5跟踪实验证明,PLT@Pt-脂质@lomiNPs通过血小板膜锚定快速附着于癌细胞,深入球体,到达亚细胞内质网,洛米调节胆固醇。此外,这些NP已经被证明可以追踪血液中的CTC,自由药物没有证明的能力。与游离lomi相比,PLTs@Pt-脂质@lomiNP更有效地激活STING途径并降低CTC干性。最终,在手术后动物模型中,PLT@Pt-脂质@lomiNP减少转移。虽然胆固醇调节剂在重新定位为免疫调节剂时功效有限,这种MCL组成的NP策略证明了有效地将这些药物递送到靶向CTC簇的潜力。
    Activating the cGAS-STING pathway of circulating tumor cell clusters (CTC clusters) represents a promising strategy to mitigate metastases. To fully exploit the potential of cholesterol-regulating agents in activating CTCs\' STING levels, we developed a nanoparticle (NP) composed of metal complex lipid (MCL). This design includes MCL-miriplatin to increase NP stiffness and loads lomitapide (lomi) modulating cholesterol levels, resulting in the creation of PLTs@Pt-lipid@lomi NPs. MCL-miriplatin not only enhances lomi\'s eliciting efficacy on STING pathway but also increases NPs\' stiffness, thus a vital factor affecting the penetration into CTC clusters to further boost lomi\'s ability. Demonstrated by cy5 tracking experiments, PLTs@Pt-lipid@lomi NPs quickly attach to cancer cell via platelet membrane anchorage, penetrate deep into the spheres, and reach the subcellular endoplasmic reticulum where lomi regulates cholesterol. Additionally, these NPs have been shown to track CTCs in the bloodstream, a capability not demonstrated by the free drug. PLTs@Pt-lipid@lomi NPs more efficiently activate the STING pathway and reduce CTC stemness compared to free lomi. Ultimately, PLTs@Pt-lipid@lomi NPs reduce metastasis in a post-surgery animal model. While cholesterol-regulating agents are limited in efficacy when being repositioned as immunomodulatory agents, this MCL-composing NP strategy demonstrates the potential to effectively deliver these agents to target CTC clusters.
    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
    随着对免疫学认识的不断深入,越来越多的免疫疗法正在癌症治疗领域中被探索和实施。cGAS-STING途径,先天免疫反应的关键因素,已被确定为癌症免疫疗法的关键。我们评估了五味子木酚素成分五味子乙素C(SC)在4T1和MC38荷瘤小鼠中的抗肿瘤作用,并通过RNA测序研究了SC对cGAS-STING通路和抗肿瘤免疫的增强作用,qRT-PCR,和流式细胞术。我们的发现表明,SC在乳腺癌和结肠癌模型中均显着抑制肿瘤生长。肿瘤生长的这种抑制归因于I型IFN应答的激活和肿瘤内T细胞和NK细胞的增强存在。此外,SC显著促进顺铂诱导的cGAS-STING通路激活。与顺铂单一疗法相比,SC和顺铂的联合治疗对肿瘤生长具有更大的抑制作用。增强的化疗疗效与增强的I型IFN反应和增强的抗肿瘤免疫力有关。SC被证明可以通过增强I型IFN反应激活和增强抗肿瘤免疫力来减少肿瘤生长和增加化疗敏感性。丰富了对中国南方黄连抗肿瘤免疫的研究,为其在乳腺癌和结肠癌中的应用奠定了理论基础。
    With the advancing comprehension of immunology, an increasing number of immunotherapies are being explored and implemented in the field of cancer treatment. The cGAS-STING pathway, a crucial element of the innate immune response, has been identified as pivotal in cancer immunotherapy. We evaluated the antitumor effects of Schisandra chinensis lignan component Schisandrin C (SC) in 4T1 and MC38 tumor-bearing mice, and studied the enhancing effects of SC on the cGAS-STING pathway and antitumor immunity through RNA sequencing, qRT-PCR, and flow cytometry. Our findings revealed that SC significantly inhibited tumor growth in models of both breast and colon cancer. This suppression of tumor growth was attributed to the activation of type I IFN response and the augmented presence of T cells and NK cells within the tumor. Additionally, SC markedly promoted the cGAS-STING pathway activation induced by cisplatin. In comparison to cisplatin monotherapy, the combined treatment of SC and cisplatin exhibited a greater inhibitory effect on tumor growth. The amplified chemotherapeutic efficacy was associated with an enhanced type I IFN response and strengthened antitumor immunity. SC was shown to reduce tumor growth and increase chemotherapy sensitivity by enhancing the type I IFN response activation and boosting antitumor immunity, which enriched the research into the antitumor immunity of S. chinensis and laid a theoretical basis for its application in combating breast and colon cancer.
    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:双丹解毒汤(SJD)是由六种具有解热解毒功能的中药组成的配方,抗菌和抗炎作用,临床上用于治疗包括COVID-19在内的各种肺部炎症性疾病,但其作用的治疗物质基础及其分子机制尚不清楚。
    目的:本研究试图确定SJD对LPS诱导的急性肺损伤(ALI)的治疗作用,以及研究其作用机制并评估其在临床上治愈炎症相关疾病的治疗潜力。
    方法:我们通过气管滴注LPS建立了ALI模型,在SJD管理之后,收集小鼠支气管肺泡灌洗液和肺组织,检测其中炎症因子的表达。此外,我们通过免疫印迹评估了SJD对环磷酸鸟苷-磷酸腺苷合成酶-干扰素基因(cGAS-STING)和炎性体的刺激物的影响,和实时定量聚合酶链反应(RT-qPCR)。
    结果:我们证明了SJD通过抑制支气管肺泡灌洗液(BALF)中的促炎细胞因子水平,可有效缓解LPS诱导的ALI,改善肺组织病理学水平和中性粒细胞数量,以及降低炎症因子相关基因的表达。重要的是,我们发现SJD可以抑制多种刺激驱动的cGAS-STING和炎性小体的激活。进一步的研究表明,在配方剂量下,SJD中的中草药对cGAS-STING途径和单独的炎性小体没有影响。通过增加这些草药的浓度,我们观察到对cGAS-STING途径和炎症小体的抑制作用,当六种草药混合使用时,效果最大,表明这些草药之间的协同作用在SJD的抗炎作用中起着至关重要的作用。
    结论:我们的研究表明SJD对ALI具有良好的保护作用,其作用机制可能与6种中药抑制cGAS-STING和炎性小体异常激活的协同作用有关。这些结果有利于SJD在临床上的广泛应用以及从草药配方中开发ALI药物。
    BACKGROUND: Shuangdan Jiedu Decoction (SJD) is a formula composed of six Chinese herbs with heat-removing and detoxifying, antibacterial, and anti-inflammatory effects, which is clinically used in the therapy of various inflammatory diseases of the lungs including COVID-19, but the therapeutic material basis of its action as well as its molecular mechanism are still unclear.
    OBJECTIVE: The study attempted to determine the therapeutic effect of SJD on LPS-induced acute lung injury (ALI), as well as to investigate its mechanism of action and assess its therapeutic potential for the cure of inflammation-related diseases in the clinical setting.
    METHODS: We established an ALI model by tracheal drip LPS, and after the administration of SJD, we collected the bronchoalveolar lavage fluid (BALF) and lung tissues of mice and examined the expression of inflammatory factors in them. In addition, we evaluated the effects of SJD on the cyclic guanosine monophosphate-adenosine monophosphate synthase -stimulator of interferon genes (cGAS-STING) and inflammasome by immunoblotting and real-time quantitative polymerase chain reaction (RT-qPCR).
    RESULTS: We demonstrated that SJD was effective in alleviating LPS-induced ALI by suppressing the levels of pro-inflammatory cytokines in the BALF, improving the level of lung histopathology and the number of neutrophils, as well as decreasing the inflammatory factor-associated gene expression. Importantly, we found that SJD could inhibit multiple stimulus-driven activation of cGAS-STING and inflammasome. Further studies showed that the Chinese herbal medicines in SJD had no influence on the cGAS-STING pathway and inflammasome alone at the formulated dose. By increasing the concentration of these herbs, we observed inhibitory effects on the cGAS-STING pathway and inflammasome, and the effect exerted was maximal when the six herbs were combined, indicating that the synergistic effects among these herbs plays a crucial role in the anti-inflammatory effects of SJD.
    CONCLUSIONS: Our research demonstrated that SJD has a favorable protective effect against ALI, and its mechanism of effect may be associated with the synergistic effect exerted between six Chinese medicines to inhibit the cGAS-STING and inflammasome abnormal activation. These results are favorable for the wide application of SJD in the clinic as well as for the development of drugs for ALI from herbal formulas.
    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
    cGAS-STING途径是先天免疫系统的关键元件,识别胞质DNA以启动I型干扰素和促炎细胞因子的产生。这项研究调查了24和108周龄小鼠皮质和海马中cGAS-STING信号传导成分的变化。在老老鼠的皮层中,观察到dsDNA传感器蛋白cGAS及其产物2\'3\'-cGAMP的增加,没有相应的下游信令激活,表明cGAS活性与STING激活的解偶联。这种现象可能归因于EC神经元中dsDNA浓度的增加,可能是由核DNA损伤引起的。相反,海马体没有表现出随着衰老而增加的cGAS活性,但是STING水平有明显的升高,特别是在小胶质细胞中,神经元和星形胶质细胞。STING的增加与IRF3激活增强无关,表明cGAS-STING途径诱导的脑部炎症可能在衰老过程中表现得极其晚。此外,我们强调了自噬的作用及其与cGAS-STING通路的相互作用,有证据表明老年海马神经元的自噬功能障碍导致STING积累。这些发现强调了cGAS-STING通路参与脑老化的复杂性,活动的区域差异和对神经退行性疾病的潜在影响。
    The cGAS-STING pathway is a pivotal element of the innate immune system, recognizing cytosolic DNA to initiate the production of type I interferons and pro-inflammatory cytokines. This study investigates the alterations of the cGAS-STING signaling components in the cortex and hippocampus of mice aged 24 and 108 weeks. In the cortex of old mice, an increase in the dsDNA sensor protein cGAS and its product 2\'3\'-cGAMP was observed, without corresponding activation of downstream signaling, suggesting an uncoupling of cGAS activity from STING activation. This phenomenon may be attributed to increased dsDNA concentrations in the EC neurons, potentially arising from nuclear DNA damage. Contrastingly, the hippocampus did not exhibit increased cGAS activity with aging, but there was a notable elevation in STING levels, particularly in microglia, neurons and astrocytes. This increase in STING did not correlate with enhanced IRF3 activation, indicating that brain inflammation induced by the cGAS-STING pathway may manifest extremely late in the aging process. Furthermore, we highlight the role of autophagy and its interplay with the cGAS-STING pathway, with evidence of autophagy dysfunction in aged hippocampal neurons leading to STING accumulation. These findings underscore the complexity of the cGAS-STING pathway\'s involvement in brain aging, with regional variations in activity and potential implications for neurodegenerative diseases.
    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
    具有细胞器级精度的新一代纳米材料为线粒体的针对性攻击提供了重要的前景。表现出显著的治疗效力。这里,我们报道了一种用于线粒体靶向光动力疗法(PDT)的新型两亲性酚类聚合物(PF),它可以通过氧化应激和呋喃介导的DNA交联的协同作用引发过度的线粒体DNA(mtDNA)损伤。此外,PF上的酚类单元能够通过金属-酚类配位与Mn2+进一步自组装,形成金属-酚类纳米材料(PFM)。我们专注于肿瘤相关巨噬细胞(TAMs)中Mn2+和肿瘤衍生的mtDNA对cGAS-STING途径的协同激活,随后将M2样TAM重新极化为M1表型。我们强调,PFM在细胞器水平上促进cGAS-STING依赖性免疫,以获得有效的抗肿瘤功效。
    New generation of nanomaterials with organelle-level precision provide significant promise for targeted attacks on mitochondria, exhibiting remarkable therapeutic potency. Here, we report a novel amphiphilic phenolic polymer (PF) for the mitochondria-targeted photodynamic therapy (PDT), which can trigger excessive mitochondrial DNA (mtDNA) damages by the synergistic action of oxidative stress and furan-mediated DNA cross-linking. Moreover, the phenolic units on PF enable further self-assembly with Mn2+ via metal-phenolic coordination to form metal-phenolic nanomaterial (PFM). We focus on the synergistic activation of the cGAS-STING pathway by Mn2+ and tumor-derived mtDNA in tumor-associated macrophages (TAMs), and subsequently repolarizing M2-like TAMs to M1 phenotype. We highlight that PFM facilitates the cGAS-STING-dependent immunity at the organelle level for potent antitumor efficacy.
    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
    前列腺癌是一种具有挑战性的疾病,因为它通常被描述为免疫学上的“冷”肿瘤,导致临床环境中当前免疫治疗方法的次优结果。光动力疗法(PDT)利用光敏剂(PS)产生的活性氧来破坏细胞内氧化还原平衡。这个过程诱导线粒体和细胞核的DNA损伤,激活免疫原性细胞死亡(ICD)和cGAS-STING途径的过程。最终,这种级联事件导致抗肿瘤免疫反应的启动。然而,现有的PS面临挑战,包括次优的肿瘤靶向,聚集诱导猝灭,肿瘤区域的氧气水平不足。为此,设计了一种通用的仿生纳米平台,用于同时递送聚集诱导的发射PSTPAQ-Py-PF6和紫杉醇(PTX)。纳米平台的细胞膜伪装导致其在肿瘤靶向和细胞内化方面的显着能力。激光照射后,TPAQ-Py-PF6与PTX联合使用显示出明显且增强的协同抗肿瘤作用。此外,纳米平台具有启动cGAS-STING途径的能力,导致细胞因子的产生。由ICD诱导的损伤相关分子模式的存在与这些上述细胞因子协作导致树突状细胞成熟的募集和促进。因此,这引发了针对肿瘤的全身免疫反应。总之,这种有前途的策略突出了多功能仿生纳米平台的使用,联合化疗,PDT,和免疫疗法,以增强抗肿瘤治疗的有效性。
    Prostate cancer presents as a challenging disease, as it is often characterized as an immunologically \"cold\" tumor, leading to suboptimal outcomes with current immunotherapeutic approaches in clinical settings. Photodynamic therapy (PDT) harnesses reactive oxygen species generated by photosensitizers (PSs) to disrupt the intracellular redox equilibrium. This process induces DNA damage in both the mitochondria and nucleus, activating the process of immunogenic cell death (ICD) and the cGAS-STING pathway. Ultimately, this cascade of events leads to the initiation of antitumor immune responses. Nevertheless, existing PSs face challenges, including suboptimal tumor targeting, aggregation-induced quenching, and insufficient oxygen levels in the tumor regions. To this end, a versatile bionic nanoplatform has been designed for the simultaneous delivery of the aggregation-induced emission PS TPAQ-Py-PF6 and paclitaxel (PTX). The cell membrane camouflage of the nanoplatform leads to its remarkable abilities in tumor targeting and cellular internalization. Upon laser irradiation, the utilization of TPAQ-Py-PF6 in conjunction with PTX showcases a notable and enhanced synergistic antitumor impact. Additionally, the nanoplatform has the capability of initiating the cGAS-STING pathway, leading to the generation of cytokines. The presence of damage-associated molecular patterns induced by ICD collaborates with these aforementioned cytokines lead to the recruitment and facilitation of dendritic cell maturation. Consequently, this elicits a systemic immune response against tumors. In summary, this promising strategy highlights the use of a multifunctional biomimetic nanoplatform, combining chemotherapy, PDT, and immunotherapy to enhance the effectiveness of antitumor 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
    慢性阻塞性肺疾病(COPD)是一种以气流受限和肺功能日益下降为特征的逐渐恶化和致命的异质性肺部疾病。目前,它是全世界死亡的主要原因之一。COPD的一致特征是气道炎症。已知几种炎症因子参与COPD的发病机制;然而,抗炎治疗不是COPD的一线治疗.虽然支气管扩张剂,皮质类固醇和罗氟司特可以改善气流和控制症状,他们无法逆转这种疾病。干扰素基因的环GMP-AMP合酶-刺激物(cGAS-STING)信号通路在免疫系统中起着重要的新作用,并已被证实是感染过程中炎症的关键介质。细胞应激,和组织损伤。最近的研究强调cGAS-STING的异常激活有助于COPD,为我们迫切需要开发的新疗法提供了方向。这里,我们专注于cGAS-STING途径,深入了解其分子机制,总结目前关于cGAS-STING通路在COPD中的作用的知识。此外,我们探索了cGAS和STING的拮抗剂,以确定针对cGAS-STING通路的COPD的潜在治疗策略.
    Chronic obstructive pulmonary disease(COPD) is a gradually worsening and fatal heterogeneous lung disease characterized by airflow limitation and increasingly decline in lung function. Currently, it is one of the leading causes of death worldwide. The consistent feature of COPD is airway inflammation. Several inflammatory factors are known to be involved in COPD pathogenesis; however, anti-inflammatory therapy is not the first-line treatment for COPD. Although bronchodilators, corticosteroids and roflumilast could improve airflow and control symptoms, they could not reverse the disease. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway plays an important novel role in the immune system and has been confirmed to be a key mediator of inflammation during infection, cellular stress, and tissue damage. Recent studies have emphasized that abnormal activation of cGAS-STING contributes to COPD, providing a direction for new treatments that we urgently need to develop. Here, we focused on the cGAS-STING pathway, providing insight into its molecular mechanism and summarizing the current knowledge on the role of the cGAS-STING pathway in COPD. Moreover, we explored antagonists of cGAS and STING to identify potential therapeutic strategies for COPD that target the cGAS-STING pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号