Sting

Sting
  • 文章类型: Journal Article
    cGAS-STING先天免疫途径和SREBP激活的胆固醇和脂肪酸合成途径在神经退行性疾病中异常地共同调节。STING信号的激活发生在内质网(ER)膜上,当甾醇丰富时,STING被INSIG1锚定,与SREBP和固醇结合的SREBP裂解激活蛋白(SCAP)一起。当甾醇含量低时,INSIG依赖性STING途径失活,SREBP-SCAP复合物易位到高尔基体,在那里SREBP被切割并易位到细胞核,以反式激活胆固醇和脂肪酸合成的基因。因此,STING与STING的反向激活呼吸:当先天免疫活跃时,胆固醇和脂肪酸合成的途径被抑制,反之亦然。STING途径由外源病毒细胞质核酸与环GMP-AMP合酶(cGAS)DNA传感器或RIG-I和MDA5dsRNA传感器相互作用刺激,但是在神经变性时,先天免疫也被自身DNA和随着神经元死亡而积累的双链RNA激活。下游,激活的STING招募TBK1并刺激干扰素刺激的基因和自噬途径的反式激活,两者都是保护性的。然而,先天免疫的慢性激活有助于小胶质细胞的激活,神经炎症和自噬失败导致神经变性。STING也是质子通道,其在被激活时刺激质子从STING囊泡离开,导致细胞死亡。在这里,我们回顾了先天免疫和胆固醇和脂肪酸合成途径的显着特征,观察到神经退行性疾病中的STING和SREBP信号异常,和相关的治疗方法。
    The cGAS-STING innate immunity pathway and the SREBP-activated cholesterol and fatty acid synthesis pathway are abnormally co-regulated in neurodegenerative disease. Activation of STING signaling occurs at the endoplasmic reticulum (ER) membrane with STING anchored by INSIG1 along with SREBP and the sterol-bound SREBP cleavage activating protein (SCAP) when sterols are in abundance. When sterols are low, the INSIG-dependent STING pathway is inactivated and the SREBP-SCAP complex is translocated to the Golgi where SREBP is cleaved and translocated to the nucleus to transactivate genes for cholesterol and fatty acid synthesis. Thus, there is inverse activation of STING vs. SREBP: when innate immunity is active, pathways for cholesterol and fatty acid synthesis are suppressed, and vice versa. The STING pathway is stimulated by foreign viral cytoplasmic nucleic acids interacting with the cyclic GMP-AMP synthase (cGAS) DNA sensor or RIG-I and MDA5 dsRNA sensors, but with neurodegeneration innate immunity is also activated by self-DNAs and double-stranded RNAs that accumulate with neuronal death. Downstream, activated STING recruits TBK1 and stimulates the transactivation of interferon stimulated genes and the autophagy pathway, which are both protective. However, chronic activation of innate immunity contributes to microglia activation, neuroinflammation and autophagy failure leading to neurodegeneration. STING is also a proton channel that when activated stimulates proton exit from STING vesicles leading to cell death. Here we review the salient features of the innate immunity and cholesterol and fatty acid synthesis pathways, observations of abnormal STING and SREBP signaling in neurodegenerative disease, and relevant therapeutic approaches.
    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
    肝细胞癌(HCC)是肝癌的最常见形式,预后不良。用于晚期HCC的可用药物是有限的,并且仍然迫切需要实质性的治疗进展,包括新药和新的联合疗法。在这项研究中,我们发现罗伊氏乳杆菌的主要代谢产物(L.reuteri),罗伊林显示出巨大的抗HCC潜力,可以帮助索拉非尼治疗。罗伊林治疗会损害线粒体自噬,并导致线粒体核苷酸的异常聚集,从而阻断线粒体DNA(mtDNA)复制和线粒体裂变,可以促进肝癌细胞mtDNA的泄漏和随后的STING激活。STING可以激活焦亡和坏死性凋亡,而reuterin处理还诱导caspase8表达,通过裂解RIPK3抑制HCC细胞的坏死。因此,焦亡是reuterin处理的HCC细胞的主要死亡形式,STING抑制显着挽救了reuterin的生长抑制作用,同时敲低caspase8协同抑制了焦亡的诱导。总之,我们的研究解释了罗伊特林抗肿瘤作用的详细分子机制,并揭示了其作为HCC治疗组合药物的潜力.
    Hepatocellular carcinoma (HCC) is the most common form of liver cancer with poor prognosis. The available drugs for advanced HCC are limited and substantial therapeutic advances including new drugs and new combination therapies are still in urgent need. In this study, we found that the major metabolite of Lactobacillus reuteri (L. reuteri), reuterin showed great anti-HCC potential and could help in sorafenib treatment. Reuterin treatment impaired mitophagy and caused the aberrant clustering of mitochondrial nucleoids to block mitochondrial DNA (mtDNA) replication and mitochondrial fission, which could promote mtDNA leakage and subsequent STING activation in HCC cells. STING could activate pyroptosis and necroptosis, while reuterin treatment also induced caspase 8 expression to inhibit necroptosis through cleaving RIPK3 in HCC cells. Thus, pyroptosis was the main death form in reuterin-treated HCC cells and STING suppression remarkably rescued the growth inhibitory effect of reuterin and concurrently knockdown caspase 8 synergized to restrain the induction of pyroptosis. In conclusion, our study explains the detailed molecular mechanisms of the antitumor effect of reuterin and reveals its potential to perform as a combinational drug for HCC 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
    已知同时滥用酒精-可卡因会导致肝脏中更强,更不可预测的细胞损伤,心,和大脑。然而,可卡因和酒精在肝损伤中的作用机制仍不清楚.这些发现揭示了可卡因诱导的小鼠和小鼠的肝损伤和炎症。值得注意的是,与单独治疗相比,在小鼠中同时服用可卡因和乙醇会导致更严重的肝损伤。代谢组学分析证实,马尿酸(HA)是可卡因消耗后血清中最丰富的代谢物,并且在原代猴肝细胞中形成。HA,可卡因的代谢产物,增加线粒体DNA泄漏,随后通过Kupffer细胞(KCs)中的STING信号增加促炎因子的产生。此外,可卡因处理的KC通过酒精诱导的TNFR1诱导的肝细胞坏死的条件培养基。最后,体内STING信号传导的中断改善了酒精和可卡因诱导的肝损伤和炎症的共同给药。这些发现假设HA-STING-TNFR1轴的干预作为治疗酒精和可卡因诱导的过度肝损伤的新策略。
    The simultaneous abuse of alcohol-cocaine is known to cause stronger and more unpredictable cellular damage in the liver, heart, and brain. However, the mechanistic crosstalk between cocaine and alcohol in liver injury remains unclear. The findings revealed cocaine-induced liver injury and inflammation in both marmosets and mice. Of note, co-administration of cocaine and ethanol in mice causes more severe liver damage than individual treatment. The metabolomic analysis confirmed that hippuric acid (HA) is the most abundant metabolite in marmoset serum after cocaine consumption and that is formed in primary marmoset hepatocytes. HA, a metabolite of cocaine, increases mitochondrial DNA leakage and subsequently increases the production of proinflammatory factors via STING signaling in Kupffer cells (KCs). In addition, conditioned media of cocaine-treated KC induced hepatocellular necrosis via alcohol-induced TNFR1. Finally, disruption of STING signaling in vivo ameliorated co-administration of alcohol- and cocaine-induced liver damage and inflammation. These findings postulate intervention of HA-STING-TNFR1 axis as a novel strategy for treatment of alcohol- and cocaine-induced excessive liver damage.
    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
    少突胶质前体细胞(OPCs)在发育中的中枢神经系统中使用血管作为物理支架广泛迁移。尽管OPCs与脉管系统的关联对于迁移至关重要,OPCs增殖和少突胶质细胞发育的重要调控机制尚不清楚.这里,在大脑发育过程中,血管发育与OPCs反应之间存在相关性。干扰素基因(STING)的内皮刺激物的缺失通过抑制法尼基-二磷酸法尼基转移酶1(FDFT1)来破坏血管生成,从而减少胆固醇的合成。此外,内皮细胞代谢稳态的扰动增加了白细胞介素17D的产生,白细胞介素17D介导了从内皮细胞到OPCs的信号转导,抑制少突胶质细胞发育和髓鞘形成,并导致成年小鼠行为异常。总的来说,这些发现表明内皮STING如何维持代谢稳态,并有助于发育中的新皮质中的少突胶质细胞前体细胞反应。
    Oligodendrocyte precursor cells (OPCs) migrate extensively using blood vessels as physical scaffolds in the developing central nervous system. Although the association of OPCs with the vasculature is critical for migration, the regulatory mechanisms important for OPCs proliferative and oligodendrocyte development are unknown. Here, a correlation is demonstrated between the developing vasculature and OPCs response during brain development. Deletion of endothelial stimulator of interferon genes (STING) disrupts angiogenesis by inhibiting farnesyl-diphosphate farnesyltransferase 1 (FDFT1) and thereby reducing cholesterol synthesis. Furthermore, the perturbation of metabolic homeostasis in endothelial cells increases interleukin 17D production which mediates the signal transduction from endothelial cells to OPCs, which inhibits oligodendrocyte development and myelination and causes behavioral abnormalities in adult mice. Overall, these findings indicate how the endothelial STING maintains metabolic homeostasis and contributes to oligodendrocyte precursor cells response in the developing neocortex.
    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:结肠癌是胃肠道系统常见的恶性肿瘤,其特点是发病率和死亡率高。目的分析miR-181a-2-3p在结肠癌中的表达及生物学作用,探讨其通过干扰素刺激基因(STING)调控结肠癌的分子机制。
    方法:采用实时逆转录聚合酶链反应(qRT-PCR)法检测miR-181a-2-3p在结肠癌细胞株和正常肠上皮细胞中的表达。在结肠癌细胞系SW480和HT29中过表达miR-181a-2-3p后,通过CCK8,Transwell,和流式细胞术检测增殖的改变,迁移,凋亡,和细胞周期。通过生物信息学预测miR-181a-2-3p的靶基因,并通过双荧光素酶测定进行验证。进行挽救实验以探索STING在miR-181a-2-3p的作用中的作用。miR-181a-2-3p对结肠癌体内增殖的影响通过裸鼠致瘤性试验进行验证。
    结果:miR-181a-2-3p在结肠癌组织和细胞系中均低表达。miR-181a-2-3p的过表达导致增殖和迁移减少,细胞凋亡增加,结肠癌细胞系SW480和HT29的细胞周期改变。STING是miR-181a-2-3p的靶基因。STING表达的增加部分抵消了miR-181a-2-3p过表达对结肠癌细胞系的影响。miR-181a-2-3p还在体内抑制结肠癌增殖。
    结论:miR-181a-2-3p抑制结肠癌的增殖和致癌作用,STING可以抑制其分子机制。
    OBJECTIVE: Colon cancer is a common malignant tumor of the gastrointestinal system, which is characterized by high morbidity and mortality. The purpose of this study was to analyze the expression and biological role of miR-181a-2-3p in colon cancer and to investigate the molecular mechanism of its regulatory effect on colon cancer through stimulator of interferon genes (STING).
    METHODS: Real-time reverse transcription polymerase chain reaction (qRT-PCR) assay was used to detect the expression of miR-181a-2-3p in colon cancer cell lines and normal intestinal epithelial cells. After overexpression of miR-181a-2-3p in colon cancer cell lines SW480 and HT29, cells were examined by CCK8, Transwell, and flow cytometry assays for alterations in proliferation, migration, apoptosis, and cell cycle. Target genes of miR-181a-2-3p were predicted by bioinformatics and validated by dual luciferase assays. Rescue experiments were performed to explore the role of STING in the effect of miR-181a-2-3p. The effect of miR-181a-2-3p on colon cancer proliferation in vivo was validated by nude mouse tumorigenicity assay.
    RESULTS: miR-181a-2-3p was lowly expressed in both colon cancer tissues and cell lines. Overexpression of miR-181a-2-3p led to reduced proliferation and migration, increased apoptosis, and altered cell cycle in colon cancer cell lines SW480 and HT29. STING was a target gene of miR-181a-2-3p. Increased STING expression partially counteracted the effect of overexpression of miR-181a-2-3p on colon cancer cell lines. miR-181a-2-3p also suppressed colon cancer proliferation in vivo.
    CONCLUSIONS: miR-181a-2-3p inhibits the proliferation and oncogenicity of colon cancer, and its molecular mechanism could be inhibited by STING.
    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
    有机阴离子转运多肽(OATPs)在胆汁酸和胆红素的转运中起着至关重要的作用。在我们之前的研究中,白细胞介素6(IL-6)降低胆汁淤积性疾病中的OATP1B3水平。然而,目前尚不清楚IL-6是否抑制胆汁淤积性疾病中OATP1B1的表达.本研究旨在研究IL-6能否抑制OATP1B1的表达并探讨其作用机制。
    使用RT-qPCR和酶联免疫吸附测定在胆汁淤积小鼠模型中研究了干扰素基因刺激因子(STING)信号传导对炎症因子的影响。探讨炎症因子对肝细胞癌中OATP1B1表达的影响,我们用TNF-α处理PLC/PRF/5细胞后,通过RT-qPCR和WesternBlot分析OATP1B1的表达,IL-1β,IL-6为了阐明IL-6抑制OATP1B1表达的机制,我们使用RT-qPCR和Western印迹检测了OATP1B1调节因子TCF4在PLC/PRF/5和HepG2细胞中的表达。通过siRNA转染敲低β-catenin/TCF4,研究了β-catenin/TCF4与OATP1B1之间的相互作用机制。
    STING抑制剂降低胆汁淤积小鼠模型的炎症因子水平,IL-6对OATP1B1表现出最有效的抑制作用。IL-6下调β-catenin/TCF4,导致OATP1B1表达降低。敲低β-连环蛋白/TCF4抵消了β-连环蛋白/TCF4介导的OATP1B1抑制。
    STING介导的IL-6上调可抑制OATP1B1,导致OATP1B1对胆汁酸和胆红素的转运减少。这可能有助于改变患有与IL-6产生增加相关的疾病的患者的药代动力学。
    UNASSIGNED: Organic anion-transporting polypeptides (OATPs) play a crucial role in the transport of bile acids and bilirubin. In our previous study, interleukin 6 (IL-6) reduced OATP1B3 levels in cholestatic disease. However, it remains unclear whether IL-6 inhibits OATP1B1 expression in cholestatic diseases. This study aimed to investigate whether IL-6 can inhibit OATP1B1 expression and explore the underlying mechanisms.
    UNASSIGNED: The effect of stimulator of interferon genes (STING) signaling on inflammatory factors was investigated in a cholestatic mouse model using RT-qPCR and enzyme-linked immunosorbent assay. To assess the impact of inflammatory factors on OATP1B1 expression in hepatocellular carcinoma, we analyzed OATP1B1 expression by RT-qPCR and Western Blot after treating PLC/PRF/5 cells with TNF-α, IL-1β, and IL-6. To elucidate the mechanism by which IL-6 inhibits OATP1B1 expression, we examined the expression of the OATP1B1 regulator TCF4 in PLC/PRF/5 and HepG2 cells using RT-qPCR and Western Blot. The interaction mechanism between β-catenin/TCF4 and OATP1B1 was investigated by knocking down β-catenin/TCF4 through siRNA transfection.
    UNASSIGNED: The STING inhibitor decreased inflammatory factor levels in the cholestatic mouse model, with IL-6 exhibiting the most potent inhibitory effect on OATP1B1. IL-6 downregulated β-catenin/TCF4, leading to decreased OATP1B1 expression. Knocking-down β-catenin/TCF4 counteracted the β-catenin/TCF4-mediated repression of OATP1B1.
    UNASSIGNED: STING-mediated IL-6 up-regulation may inhibit OATP1B1, leading to reduced transport of bile acids and bilirubin by OATP1B1. This may contribute to altered pharmacokinetics in patients with diseases associated with increased IL-6 production.
    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
    铁凋亡的激活提出了一种在癌症治疗中增强抗肿瘤免疫反应的通用策略。然而,开发结合高生物相容性和治疗效率的铁凋亡诱导物仍然具有挑战性。在这项研究中,我们提出了一种新的方法,使用来自大肠杆菌外膜囊泡(OMV)的生物纳米颗粒用于肿瘤治疗,旨在激活铁细胞凋亡并刺激免疫反应。具体来说,我们通过静电相互作用将OMV与亚铁离子锚定并加载STING激动剂-4,然后进行肿瘤靶向DSPE-PEG-FA修饰,此后称为OMV/SaFeFA。亚铁离子的锚定赋予OMV过氧化物酶样活性,能够通过催化H2O2至·OH来诱导细胞脂质过氧化。此外,OMV/SaFeFA表现出亚铁离子和激动剂的pH响应性释放,以及肿瘤靶向能力,使肿瘤特异性治疗,同时尽量减少副作用。值得注意的是,STING通路的同时激活和铁凋亡在结肠荷瘤小鼠模型中引起强烈的抗肿瘤反应,导致特殊的治疗效果和延长生存期。重要的是,在接受OMV/SaFeFA治疗的小鼠中未观察到急性毒性,强调其未来肿瘤治疗和临床转化的潜力。
    The activation of ferroptosis presents a versatile strategy for enhancing the antitumor immune responses in cancer therapy. However, developing ferroptosis inducers that combine high biocompatibility and therapeutic efficiency remains challenging. In this study, we propose a novel approach using biological nanoparticles derived from outer membrane vesicles (OMVs) of Escherichia coli for tumor treatment, aiming to activate ferroptosis and stimulate the immune responses. Specifically, we functionalize the OMVs by anchoring them with ferrous ions via electrostatic interactions and loading them with the STING agonist-4, followed by tumor-targeting DSPE-PEG-FA decoration, henceforth referred to as OMV/SaFeFA. The anchoring of ferrous ions endows the OMVs with peroxidase-like activity, capable of inducing cellular lipid peroxidation by catalyzing H2O2 to •OH. Furthermore, OMV/SaFeFA exhibits pH-responsive release of ferrous ions and the agonist, along with tumor-targeting capabilities, enabling tumor-specific therapy while minimizing side effects. Notably, the concurrent activation of the STING pathway and ferroptosis elicits robust antitumor responses in colon tumor-bearing mouse models, leading to exceptional therapeutic efficacy and prolonged survival. Importantly, no acute toxicity was observed in mice receiving OMV/SaFeFA treatments, underscoring its potential for future tumor therapy and clinical translation.
    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:骨关节炎(OA)是一种退行性疾病,其特征在于炎症和细胞外基质(ECM)降解,会导致严重的个人残疾和社会负担。日本金鱼草。(LJT)是一种具有抗炎和镇痛作用的柳科植物。在传统的东方医学中,LJT通常用于治疗各种疾病,包括骨关节炎和腰背痛。
    目的:为了研究抗凋亡,LJT在IL-1β诱导的小鼠软骨细胞中的抗炎和抗衰老特性,并澄清所涉及的潜在机制。此外,该研究还通过建立骨关节炎小鼠模型来检查LJT的作用。最终目标是确定LJT作为抗骨关节炎药物的机制。
    方法:在这项研究中,进行分子对接和网络药理学分析以确定LJT作用的潜伏途径和关键靶标。CCK-8试剂盒用于评估LJT对软骨细胞活力的影响。西方印迹,免疫荧光,TUNEL染色试剂盒,和SA-β-gal染色用于验证LJT对软骨细胞的影响。此外,所以,他,免疫组织化学用于评估LJT对小鼠骨关节炎的影响。进行了体外和体内实验以验证LJT在OA中的潜在机制。
    结果:网络药理学分析显示AKT1、PTGS2和ESR1是LJT治疗OA的关键候选靶点。分子对接结果表明,AKT1对LJT的主要成分具有较低的结合亲和力。因此,我们选择了STING,PTGS2的上游监管机构,作为我们的调查目标。分子对接显示谷甾醇,福蒙素,豆甾醇和α-Onocerin,LJT的主要组成部分,与STING具有良好的结合活性。体外实验表明LJT抑制IL-1β介导的炎症介质分泌,软骨细胞的凋亡和衰老。结果表明,LJT消除了小鼠不稳定内侧半月板(DMM)引起的软骨退变。机制研究表明,LJT通过抑制STING/NF-κB信号通路,下调NF-κB激活,从而抑制OA的发展。
    结论:LJT通过抑制炎症逆转OA的进展,动物模型和软骨细胞的凋亡和衰老。LJT的作用是通过STING/NF-κB途径介导的。
    BACKGROUND: Osteoarthritis (OA) is a degenerative disease, its characteristic lies in the inflammation and extracellular matrix (ECM) degradation, can lead to significant personal disability and social burden. Lycopodium japonicum Thunb. (LJT) is a lycopinaceae plant with anti-inflammatory and analgesic effects. In traditional Oriental medicine, LJT is commonly used to treat a variety of conditions, including osteoarthritis and low back pain.
    OBJECTIVE: To investigate the anti-apoptotic, anti-inflammatory and anti-senescence properties of LJT in IL-1β-induced mouse chondrocytes, and to clarify the underlying mechanisms involved. In addition, the study also examined the effects of LJT by establishing a mouse model of osteoarthritis. The ultimate goal is to identify the mechanism of LJT as an anti-osteoarthritis agent.
    METHODS: In this research, molecular docking and network pharmacology analysis were performed to identify the latent pathways and key targets of LJT action. The CCK-8 kit was used to evaluate LJT\'s effect on chondrocyte viability. Western blotting, Immunofluorescence, TUNEL staining kit, and SA-β-gal staining were employed to verify LJT\'s impact on chondrocytes. Additionally, SO, HE, and Immunohistochemical were utilized to assess LJT\'s effects on osteoarthritis in mice. In vitro and in vivo experiments were performed to verify the potential mechanism of LJT in OA.
    RESULTS: Network pharmacology analysis revealed that AKT1, PTGS2, and ESR1 were the key candidate targets for the treatment of OA with LJT. The results of molecular docking indicated that AKT1 exhibited a low binding affinity to the principal constituents of LJT. Hence, we have chosen STING, an upstream regulator of PTGS2, as our target for investigation. Molecular docking revealed that sitosterol, formononetin, stigmasterol and alpha-Onocerin, the main components of LJT, have good binding activity with STING. In vitro experiments showed that LJT inhibited IL-1β-mediated secretion of inflammatory mediators, apoptosis and senescence of chondrocytes. The results showed that LJT abolished cartilage degeneration induced by unstable medial meniscus (DMM) in mice. Mechanism research has shown that LJT by inhibiting the STING/NF-κB signaling pathways, down-regulating the NF-κB activation, so as to inhibit the development of OA.
    CONCLUSIONS: LJT reversed the progression of OA by inhibiting inflammation, apoptosis and senescence in animal models and chondrocytes. The effects of LJT are mediated through the STING/NF-κB pathway.
    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
    非小细胞肺癌(NSCLC)中的肿瘤相关巨噬细胞(TAMs)通过与癌细胞相互作用促进肿瘤细胞转移。人参皂苷Re能够调节宿主免疫系统并通过多种途径发挥抗癌作用。AMPK和STING都参与了MΦ极化的调节,从而影响肿瘤进展。然而,它们之间是否存在调节关系及其对MΦ极化和肿瘤进展的影响尚不清楚。本研究的目的是提供机制证据,证明人参皂苷Re通过抑制AMPKα1/STING正反馈回路调节MΦ表型,从而在NSCLC免疫治疗中发挥抗转移作用。构建了细胞培养模型和条件培养基(CM)系统,并通过数据库分析对处理后的MΦ进行分析,RT-PCR,西方印迹,流式细胞术,和免疫荧光来确定AMPK和STING之间的调节关系以及人参皂苷Re对MΦ极化和肿瘤细胞迁移的影响。通过HE染色评估人参皂苷Re(10,20mg/kg/天)对小鼠TAMs表型以及肿瘤进展的影响。免疫组织化学染色,和西方印迹。在这项研究中,AMPKα1/STING正反馈回路在NSCLCTAMs诱导M2型极化中,进而促进NSCLC细胞迁移。此外,人参皂苷Re被发现抑制M2样MΦ极化,从而抑制NSCLC细胞迁移。机械上,Re能够抑制AMPKα1/STING正反馈回路的形成,从而抑制其对M2样MΦ的诱导,从而抑制NSCLC细胞的上皮-间质转化(EMT)过程。此外,在老鼠模型中,发现Re通过抑制TAM的M2型极化来抑制LLC肿瘤的生长和定植。我们的发现表明人参皂苷Re可以有效地调节MΦ极化,因此在NSCLC的抗转移免疫治疗中发挥重要作用。
    Tumor-associated macrophages (TAMs) in non-small cell lung cancer (NSCLC) promote tumor cell metastasis by interacting with cancer cells. Ginsenoside Re is capable of modulating the host immune system and exerts anticancer effects through multiple pathways. Both AMPK and STING are involved in the regulation of MΦ polarization, thereby affecting tumor progression. However, whether there is a regulatory relationship between them and its effect on MΦ polarization and tumor progression is unclear. The aim of this study was to provide mechanistic evidence that ginsenoside Re modulates MΦ phenotype through inhibition of the AMPKα1/STING positive feedback loop and thus exerts an antimetastatic effect in NSCLC immunotherapy. Cell culture models and conditioned media (CM) systems were constructed, and the treated MΦ were analyzed by database analysis, RT-PCR, Western blotting, flow cytometry, and immunofluorescence to determine the regulatory relationship between AMPK and STING and the effects of ginsenoside Re on MΦ polarization and tumor cells migration. The effects of ginsenoside Re (10, 20 mg/kg/day) on TAMs phenotype as well as tumor progression in mice were assessed by HE staining, immunohistochemical staining, and Western blotting. In this study, AMPKα1/STING positive feedback loop in NSCLC TAMs induced M2 type polarization, which in turn promoted NSCLC cell migration. In addition, ginsenoside Re was discovered to inhibit M2-like MΦ polarization, thereby inhibiting NSCLC cell migration. Mechanistically, Re was able to inhibit the formation of the AMPKα1/STING positive feedback loop, thereby inhibiting its induction of M2-like MΦ and consequently inhibiting the epithelial-mesenchymal transition (EMT) process of NSCLC cells. Furthermore, in mouse models, Re was found to suppress LLC tumor growth and colonization by inhibiting M2-type polarization of TAMs. Our finding indicates that ginsenoside Re can effectively modulate MΦ polarization and thus play an important role in antimetastatic immunotherapy of NSCLC.
    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
    有效的皮肤伤口愈合需要由白细胞亚群的动态变化介导的炎症阶段之间的协调过渡。这里,我们确定STING是通过调节皮肤修复过程中的巨噬细胞动力学来控制炎症及时消退的关键先天免疫介质。使用鼠标模型,我们显示STING缺乏导致伤口闭合延迟与TNF-α+白细胞的异常持续相关。这是由受损的巨噬细胞募集引起的。STING控制了骨髓骨髓细胞进入血液和伤口的运输,通过STAT3激活内在增强巨噬细胞迁移能力。具体来说,STING调节单核细胞趋化因子及其受体CCR2/CCR5的产生,以实现有效的外出和伤口浸润。因此,破坏的全身和局部STING-STAT3-趋化因子信号结合延迟巨噬细胞流入。这项研究阐明了STING是通过STAT3调节巨噬细胞反应以协调有效伤口愈合所必需的炎症消退的关键变阻器。我们的发现对于在再生医学和炎性疾病背景下靶向治疗STING具有广泛的意义。STING在伤口愈合中通过STAT3调节巨噬细胞运输。
    Efficient cutaneous wound healing requires a coordinated transition between inflammatory phases mediated by dynamic changes in leukocyte subset populations. Here, we identify STING as a key innate immune mediator governing timely resolution of inflammation by regulating macrophage dynamics during skin repair. Using a mouse model, we show STING deficiency caused delayed wound closure associated with abnormal persistence of TNF-α+ leukocytes. This resulted from the impaired macrophage recruitment. STING controlled the trafficking of bone marrow myeloid cells into blood and wounds, intrinsically enhancing macrophage migratory capacity through STAT3 activation. Specifically, STING modulated the production of monocyte chemokines and their receptors CCR2/CCR5 to enable efficient egress and wound infiltration. Consequently, disrupted systemic and local STING-STAT3-chemokine signaling combine to delay macrophage influx. This study elucidates STING as a critical rheostat tuning macrophage responses through STAT3 to orchestrate inflammatory resolution necessary for efficient wound healing. Our findings have broad implications for targeting STING therapeutically in both regenerative medicine and inflammatory disease contexts. STING regulates the macrophage trafficking through STAT3 in wound healing.
    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