STING1

STING1
  • 文章类型: Journal Article
    血管钙化是老年人和糖尿病个体心血管风险的普遍标志。衰老血管平滑肌细胞(VSMC)参与钙化;然而,相关的潜在机制仍然未知。由胞质DNA引起的干扰素基因1(STING1)的胞质DNA传感接头刺激物的异常激活,特别是从受损的线粒体泄漏出来的,是衰老相关疾病的催化剂。尽管油酰乙醇胺(OEA)是一种内源性生物活性脂质介质,具有脂质超负荷相关的血管保护作用,其在糖尿病血管钙化中的益处仍未表征。这项研究集中于STING1在糖尿病线粒体功能障碍介导的钙化和VMSC过早衰老中的作用以及OEA对这些病理过程的影响。在糖尿病大鼠/小鼠体内主动脉钙化模型和Nε-羧甲基赖氨酸(CML)诱导的体外VSMC钙化模型中,衰老水平,STING1信号激活,和线粒体损伤标记显著增强;然而,这些改变被OEA明显缓解,部分以核因子红系2相关因子2(Nrf2)依赖性方式,在STING1敲除小鼠和STING1敲除VSMC中观察到相似的抗钙化和衰老作用。机械上,线粒体DNA(mtDNA)损伤被CML以活性氧依赖的方式加重,随后是mtDNA泄漏到细胞质中,通过STING1途径激活促进VSMC衰老相关钙化。OEA治疗通过维持Nrf2易位到细胞核而增强细胞抗氧化能力,从而显著减弱上述CML的细胞毒性作用。总的来说,靶向STING1,一种新定义的VSMC衰老调节剂,有助于抗血管钙化作用。
    Vascular calcification is a prevalent hallmark of cardiovascular risk in elderly and diabetic individuals. Senescent vascular smooth muscle cells (VSMCs) participate in calcification; however, the associated underlying mechanisms remain unknown. Aberrant activation of the cytosolic DNA sensing adaptor stimulator of interferon gene 1 (STING1) caused by cytosolic DNA, particularly that leaked from damaged mitochondria, is a catalyst for aging-related diseases. Although oleoylethanolamide (OEA) is an endogenous bioactive lipid mediator with lipid overload-associated vasoprotective effects, its benefit in diabetic vascular calcification remains uncharacterized. This study focused on the role of STING1 in mitochondrial dysfunction-mediated calcification and premature VMSC senescence in diabetes and the effects of OEA on these pathological processes. In diabetic in vivo rat/mouse aorta calcification models and an in vitro VSMC calcification model induced by Nε-carboxymethyl-lysine (CML), senescence levels, STING1 signaling activation, and mitochondrial damage markers were significantly augmented; however, these alterations were markedly alleviated by OEA, partially in a nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent manner, and similar anti-calcification and senescence effects were observed in STING1-knockout mice and STING1-knockdown VSMCs. Mechanistically, mitochondrial DNA (mtDNA) damage was aggravated by CML in a reactive oxygen species-dependent manner, followed by mtDNA leakage into the cytosol, contributing to VSMC senescence-associated calcification via STING1 pathway activation. OEA treatment significantly attenuated the aforementioned cytotoxic effects of CML by enhancing cellular antioxidant capacity through the maintenance of Nrf2 translocation to the nucleus. Collectively, targeting STING1, a newly defined VSMC senescence regulator, contributes to anti-vascular calcification effects.
    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
    Autophagy is a crucial immune defense mechanism that controls the survival and pathogenesis of M. tb by maintaining cell physiology during stress and pathogen attack. The E3-Ub ligases (PRKN, SMURF1, and NEDD4) and autophagy receptors (SQSTM1, TAX1BP1, CALCOCO2, OPTN, and NBR1) play key roles in this process. Galectins (LGALSs), which bind to sugars and are involved in identifying damaged cell membranes caused by intracellular pathogens such as M. tb, are essential. These include LGALS3, LGALS8, and LGALS9, which respond to endomembrane damage and regulate endomembrane damage caused by toxic chemicals, protein aggregates, and intracellular pathogens, including M. tb. They also activate selective autophagy and de novo endolysosome biogenesis. LGALS3, LGALS9, and LGALS8 interact with various components to activate autophagy and repair damage, while CGAS-STING1 plays a critical role in providing immunity against M. tb by activating selective autophagy and producing type I IFNs with antimycobacterial functions. STING1 activates cGAMP-dependent autophagy which provides immunity against various pathogens. Additionally, cytoplasmic surveillance pathways activated by ds-DNA, such as inflammasomes mediated by NLRP3 and AIM2 complexes, control M. tb. Modulation of E3-Ub ligases with small regulatory molecules of LGALSs and TRIM proteins could be a novel host-based therapeutic approach for controlling TB.
    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: Case Reports
    在婴儿期发病的STING相关血管病变(SAVI)是一种罕见的,由STING1(TMEM173)中的功能获得变体引起的单基因干扰素病,其特征是全身性炎症,皮肤血管病变,和间质性肺病.我们报告了一例SAVI,该SAVI归因于一种新型STING1p.R284T变体,该变体表现出特征性皮肤特征,包括毛细血管扩张,面部和四肢上的livedo和肢端发青的变化,以及马鞍鼻畸形,未能茁壮成长,炎性关节炎和明显缺乏肺部疾病或自身抗体阳性。由于进行性和不可逆的肺和组织损伤的风险以及涉及使用Janus激酶抑制剂的不断发展的治疗前景,识别各种临床表型对于SAVI患者在病程早期诊断和考虑治疗方案至关重要.
    STING-associated vasculopathy with onset in infancy (SAVI) is a rare, monogenic interferonopathy caused by gain-of-function variants in STING1 (TMEM173) characterized by systemic inflammation, cutaneous vasculopathy, and interstitial lung disease. We report a case of SAVI attributed to a novel STING1 p.R284T variant who demonstrated characteristic cutaneous features including telangiectasias, livedo and acrocyanotic changes on face and extremities, as well as saddle nose deformity, failure to thrive, inflammatory arthritis and notable lack of pulmonary disease or autoantibody positivity. Due to the risk for progressive and irreversible lung and tissue damage and evolving therapeutic landscape involving the use of Janus kinase inhibitors, it is critical to recognize variable clinical phenotypes to diagnose and consider treatment options for SAVI patients early in their disease course.
    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
    哺乳动物细胞通过分泌抗病毒和促炎细胞因子对细胞溶胶中双链(ds)DNA的积累作出反应,特别是I型干扰素(IFN)。Tani等人报告的最新数据。证明了该途径的过度激活可以通过由I型IFN受体引起的适应性反馈机制来防止,并由外切核酸酶三主要修复外切核酸酶1(TREX1)执行。
    Mammalian cells react to the accumulation of double-stranded (ds)DNA in the cytosol by secreting antiviral and proinflammatory cytokines, notably type I interferon (IFN). Recent data reported by Tani et al. demonstrate that overactivation of this pathway is prevented by an adaptive feedback mechanism elicited by type I IFN receptors and executed by the exonuclease three prime repair exonuclease 1 (TREX1).
    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
    广泛的研究证明了STING1(也称为STING)蛋白作为信号中枢的重要性,该信号中枢协调对细胞质中异位DNA的免疫和自噬反应。这里,我们报道了STING1在驱动转录因子芳香烃受体(AHR)激活以控制肠道菌群组成和稳态中的核功能。该功能与DNA传感和自噬无关,并显示与细胞质环磷酸鸟苷(GMP)-AMP合酶(CGAS)-STING1信号传导的竞争性抑制。在结构上,STING1的环状二核苷酸结合结构域与AHRN末端结构域相互作用。蛋白质组学分析显示,STING1介导的AHR转录激活需要额外的核伙伴,包括正调节蛋白和负调节蛋白。尽管AHR配体可以挽救野生型小鼠的结肠炎病理和生态失调,这种保护被STING1的突变失活所废除.这些发现为理解微生物群和免疫系统之间的核分子串扰建立了关键框架。
    Extensive studies demonstrate the importance of the STING1 (also known as STING) protein as a signaling hub that coordinates immune and autophagic responses to ectopic DNA in the cytoplasm. Here, we report a nuclear function of STING1 in driving the activation of the transcription factor aryl hydrocarbon receptor (AHR) to control gut microbiota composition and homeostasis. This function was independent of DNA sensing and autophagy and showed competitive inhibition with cytoplasmic cyclic guanosine monophosphate (GMP)-AMP synthase (CGAS)-STING1 signaling. Structurally, the cyclic dinucleotide binding domain of STING1 interacted with the AHR N-terminal domain. Proteomic analyses revealed that STING1-mediated transcriptional activation of AHR required additional nuclear partners, including positive and negative regulatory proteins. Although AHR ligands could rescue colitis pathology and dysbiosis in wild-type mice, this protection was abrogated by mutational inactivation of STING1. These findings establish a key framework for understanding the nuclear molecular crosstalk between the microbiota and the immune system.
    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
    CGAS(环GMP-AMP合酶)-STING1(干扰素应答cGAMP相互作用因子1的刺激物)途径是在用>45bp的dsDNA刺激后诱导促炎细胞因子产生的重要的先天性免疫途径。我们最近鉴定了一类约20-40bp的小胞质dsDNA(scDNA),其阻断CGAS-STING1活化。在这个泪点,我们讨论了通过scDNA抑制CGAS-STING1激活的潜在机制。scDNA结合CGAS但不能激活其酶活性。它与>45bp的dsDNA竞争与CGAS结合以抑制CGAS-STING1活化。此外,scDNA激活巨自噬/自噬并诱导STING1和长dsDNA的自噬降解。自噬会增加scDNA水平,驱动反馈回路,加速STING1和长胞质dsDNA的降解。这些发现揭示了scDNA和自噬之间的相互通讯抑制了在用>45bp的dsDNA刺激后CGAS-STING1的激活。
    The CGAS (cyclic GMP-AMP synthase)-STING1 (stimulator of interferon response cGAMP interactor 1) pathway is an important innate immune pathway that induces proinflammatory cytokine production following stimulation with dsDNA > 45 bp. We recently identified a class of ~ 20-40 bp small cytosolic dsDNA (scDNA) that blocks CGAS-STING1 activation. In this punctum, we discuss the mechanism underlying the inhibition of CGAS-STING1 activation via scDNA. scDNA binds to CGAS but cannot activate its enzymatic activity. It competes with dsDNA > 45 bp for binding with CGAS to inhibit CGAS-STING1 activation. Moreover, scDNA activates macroautophagy/autophagy and induces the autophagic degradation of STING1 and long dsDNA. Autophagy then increases scDNA levels, driving a feedback loop that accelerates the degradation of STING1 and long cytosolic dsDNA. These findings reveal that mutual communication between scDNA and autophagy inhibits CGAS-STING1 activation following stimulation with dsDNA > 45 bp.
    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
    STING1(干扰素应答刺激因子cGAMP相互作用因子1)是CGAS-STING1信号通路中的典型蛋白,对于诱导I型IFN(干扰素)产生和引发先天免疫至关重要。然而,STING1的过度激活或持续激活与自身免疫性疾病的发作密切相关.值得注意的是,这些疾病中的大多数表现为I型干扰素和IFN刺激基因(ISGs)的上调表达。因此,严格调节STING1活性对于保持免疫稳态至关重要。这里,我们报道了CSNK1A1/CK1α,丝氨酸/苏氨酸蛋白激酶,对于通过STING1的自噬降解来防止STING1介导的I型IFN信号过度激活至关重要。机械上,CSNK1A1在CGAS-STING1途径激活时与STING1相互作用,并通过增强SQSTM1/p62在丝氨酸351(人类丝氨酸349)的磷酸化来促进STING1自噬降解,这对SQSTM1介导的STING1自噬降解至关重要。始终如一,SSTC3,一种选择性CSNK1A1激动剂,通过促进STING1自噬降解显着减弱CGAS-STING1信号传导的响应。重要的是,在Trex1-/-小鼠自身免疫性疾病模型中,使用SSTC3进行CSNK1A1的药理学激活显著抑制了全身自身炎症反应,并有效抑制了SLE患者PBMC中IFNs和ISGs的产生.一起来看,我们的研究揭示了CSNK1A1在STING1自噬降解以维持免疫稳态中的新调节作用。通过SSTC3操纵CSNK1A1可能是缓解自身免疫性疾病中STING1介导的异常I型IFN的潜在治疗策略。缩写:BMDMs:骨髓衍生的巨噬细胞;cGAMP:环GMP-AMP;CGAS:环GMP-AMP合酶;HTDNA:鲱鱼睾丸DNA;IFIT1:干扰素诱导的具有四三肽重复1的蛋白质;IFNA4:干扰素α4;IFNB:干扰素β;IRF3:干扰素调节因子3;ISD:干扰素刺激的DNA;ISGs:干扰素刺激的外周成纤维细胞1;含有IFN刺激
    STING1 (stimulator of interferon response cGAMP interactor 1) is the quintessential protein in the CGAS-STING1 signaling pathway, crucial for the induction of type I IFN (interferon) production and eliciting innate immunity. Nevertheless, the overactivation or sustained activation of STING1 has been closely associated with the onset of autoimmune disorders. Notably, the majority of these disorders manifest as an upregulated expression of type I interferons and IFN-stimulated genes (ISGs). Hence, strict regulation of STING1 activity is paramount to preserve immune homeostasis. Here, we reported that CSNK1A1/CK1α, a serine/threonine protein kinase, was essential to prevent the overactivation of STING1-mediated type I IFN signaling through autophagic degradation of STING1. Mechanistically, CSNK1A1 interacted with STING1 upon the CGAS-STING1 pathway activation and promoted STING1 autophagic degradation by enhancing the phosphorylation of SQSTM1/p62 at serine 351 (serine 349 in human), which was critical for SQSTM1-mediated STING1 autophagic degradation. Consistently, SSTC3, a selective CSNK1A1 agonist, significantly attenuated the response of the CGAS-STING1 signaling by promoting STING1 autophagic degradation. Importantly, pharmacological activation of CSNK1A1 using SSTC3 markedly repressed the systemic autoinflammatory responses in the trex1-/- mouse autoimmune disease model and effectively suppressed the production of IFNs and ISGs in the PBMCs of SLE patients. Taken together, our study reveals a novel regulatory role of CSNK1A1 in the autophagic degradation of STING1 to maintain immune homeostasis. Manipulating CSNK1A1 through SSTC3 might be a potential therapeutic strategy for alleviating STING1-mediated aberrant type I IFNs in autoimmune diseases.Abbreviations: BMDMs: bone marrow-derived macrophages; cGAMP: cyclic GMP-AMP; CGAS: cyclic GMP-AMP synthase; HTDNA: herring testes DNA; IFIT1: interferon induced protein with tetratricopeptide repeats 1; IFNA4: interferon alpha 4; IFNB: interferon beta; IRF3: interferon regulatory factor 3; ISD: interferon stimulatory DNA; ISGs: IFN-stimulated genes; MEFs: mouse embryonic fibroblasts; PBMCs: peripheral blood mononuclear cells; RSAD2: radical S-adenosyl methionine domain containing 2; SLE: systemic lupus erythematosus; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1.
    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
    Ⅰ型干扰素(IFN)是一类对恶性转化具有双重作用的促炎细胞因子,肿瘤进展,和对治疗的反应。一方面,健壮,急性,和解决I型IFN应答已被证明介导突出的抗癌作用,不仅反映了它们对(至少一些)恶性细胞的直接细胞抑制/细胞毒性活性,还有它们明显的免疫刺激功能。根据这个概念,I型IFN信号与各种免疫原性疗法的抗肿瘤作用有关,包括(但不限于)免疫原性细胞死亡(ICD)诱导剂和免疫检查点抑制剂(ICI)。另一方面,弱,惰性,和未解决的I型IFN反应已被证明支持肿瘤进展和对治疗的抵抗,反映了亚最佳I型IFN信号介导细胞保护活性的能力,促进干性,支持对染色体不稳定性的耐受性,并促进建立免疫耗尽的肿瘤微环境。这里,我们回顾了I型IFN信号的基本方面及其对恶性转化的环境依赖性影响,肿瘤进展,和对治疗的反应。
    Type I interferon (IFN) is a class of proinflammatory cytokines with a dual role on malignant transformation, tumor progression, and response to therapy. On the one hand, robust, acute, and resolving type I IFN responses have been shown to mediate prominent anticancer effects, reflecting not only their direct cytostatic/cytotoxic activity on (at least some) malignant cells, but also their pronounced immunostimulatory functions. In line with this notion, type I IFN signaling has been implicated in the antineoplastic effects of various immunogenic therapeutics, including (but not limited to) immunogenic cell death (ICD)-inducing agents and immune checkpoint inhibitors (ICIs). On the other hand, weak, indolent, and non-resolving type I IFN responses have been demonstrated to support tumor progression and resistance to therapy, reflecting the ability of suboptimal type I IFN signaling to mediate cytoprotective activity, promote stemness, favor tolerance to chromosomal instability, and facilitate the establishment of an immunologically exhausted tumor microenvironment. Here, we review fundamental aspects of type I IFN signaling and their context-dependent impact on malignant transformation, tumor progression, and response to therapy.
    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
    快速进展性/新月体肾小球肾炎(RPGN/CGN)涉及通过顶叶上皮细胞的适应不良分化而形成肾小球新月体,从而导致肾功能迅速丧失。对新月形成的分子机制知之甚少。因此,对分子机制的新见解可以确定RPGN/CGN的替代治疗靶标。对RPGN患者的肾活检分析显示间质增加,肾小球,和STING1的管状表达,STING1是c-GAS依赖性DNA传感途径的辅助蛋白,在抗GBM抗体诱导的鼠肾毒性肾炎中也观察到了这一点。STING1由参与RPGN和新月形成的关键细胞类型如肾小球壁上皮细胞表达,和肾小管细胞以及炎症辅助细胞。在体内功能研究中,Sting1-/-肾毒性肾炎小鼠肾脏细胞因子表达较低,先天和适应性免疫细胞的轻度肾脏浸润,降低疾病严重程度。药理学STING1抑制反映了这些发现。顶叶和肾小管细胞中的直接STING1激动激活了NF-κB依赖性细胞因子反应和干扰素诱导的基因(ISG)程序。这些反应也由促炎细胞因子TWEAK以STING1依赖性方式触发。这些结果确定STING1激活是RPGN/CGN中的病理机制,TWEAK是STING1的激活剂。因此,靶向STING1或上游调节剂的药理学策略可能是治疗RPGN的潜在替代品。©2023年英国和爱尔兰病理学会。
    Rapidly progressive/crescentic glomerulonephritis (RPGN/CGN) involves the formation of glomerular crescents by maladaptive differentiation of parietal epithelial cells that leads to rapid loss of renal function. The molecular mechanisms of crescent formation are poorly understood. Therefore, new insights into molecular mechanisms could identify alternative therapeutic targets for RPGN/CGN. Analysis of kidney biopsies from patients with RPGN revealed increased interstitial, glomerular, and tubular expression of STING1, an accessory protein of the c-GAS-dependent DNA-sensing pathway, which was also observed in murine nephrotoxic nephritis induced by an anti-GBM antibody. STING1 was expressed by key cell types involved in RPGN and crescent formation such as glomerular parietal epithelial cells, and tubular cells as well as by inflammation accessory cells. In functional in vivo studies, Sting1-/- mice with nephrotoxic nephritis had lower kidney cytokine expression, milder kidney infiltration by innate and adaptive immune cells, and decreased disease severity. Pharmacological STING1 inhibition mirrored these findings. Direct STING1 agonism in parietal and tubular cells activated the NF-κB-dependent cytokine response and the interferon-induced genes (ISGs) program. These responses were also triggered in a STING1-dependent manner by the pro-inflammatory cytokine TWEAK. These results identify STING1 activation as a pathological mechanism in RPGN/CGN and TWEAK as an activator of STING1. Pharmacological strategies targeting STING1, or upstream regulators may therefore be potential alternatives to treat RPGN. © 2023 The Pathological Society of Great Britain and Ireland.
    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
    SMAD4是一种肿瘤抑制因子,是胆管癌(CCA)中肿瘤免疫功能的重要调节因子。STING1是异常DNA的重要传感因子;然而,尚未评估SMAD4和STING1之间的相关性以及SMAD4-STING1相互作用在CCA进展中的作用。分析公共数据库以揭示SMAD4和STING1的表达。包括50个iCCA的队列,113名pCCA和119名dCCA患者进行了研究。免疫组织化学用于评估STING1和SMAD4的表达水平。体外transwell和CCK8测定,随着荧光素酶报告分析,分析SMAD4对STING1表达的潜在调控机制。CCA肿瘤中SMAD4和STING1的表达下调,STING1的表达与SMAD4的表达相关。发现SMAD4的过表达抑制了迁移,CCA细胞的侵袭和增殖能力;然而,SMAD4的击倒增强了这些能力。此外,观察到SMAD4在TGF-β1刺激后易位到细胞核中。SMAD4的敲低导致STING1转录活性的抑制,而SMAD4的过表达促进了STING1的转录活性。临床上,低STING1和SMAD4表达表明CCA预后不良,同时STING1和SMAD4的低表达预示着患者生存率较差。SMAD4通过其转录调节功能调节STING1的表达。STING1和SMAD4的双重低表达在预测患者生存率方面具有更大的功效。这些结果表明SMAD4沉默的CCA可能下调其STING1表达以适应免疫系统。
    SMAD4 is a tumour suppressor and an important regulator of tumour immune scape which is downregulated in cholangiocarcinoma (CCA). STING1 is a vital sensing factor of abnormal DNA; however, the correlation between SMAD4 and STING1 and the role of the SMAD4-STING1 interaction in the progression of CCA have not yet been evaluated. Public database was analysed to reveal the expression of SMAD4 and STING1. A cohort comprising 50 iCCA, 113 pCCA and 119 dCCA patients was assembled for the study. Immunohistochemistry was employed to evaluate the expression levels of STING1 and SMAD4. In vitro transwell and CCK8 assays, along with luciferase reporter assay, were conducted to analyse the potential regulatory mechanisms of SMAD4 on the expression of STING1. Expression of SMAD4 and STING1 were downregulated in CCA tumours and STING1 expression correlated with SMAD4 expression. The overexpression of SMAD4 was found to suppress the migration, invasion and proliferation capabilities of CCA cells; whereas, the knockdown of SMAD4 enhanced these abilities. Furthermore, it was observed that SMAD4 translocated into the nucleus following TGF-β1 stimulation. Knockdown of SMAD4 resulted in the inhibition of STING1 transcriptional activity, whereas the overexpression of SMAD4 promoted the transcriptional activity of STING1. Clinically, low STING1 and SMAD4 expression indicated poor prognosis in CCA, and simultaneously low expression of STING1 and SMAD4 predicts poorer patient survival. SMAD4 regulates the expression of STING1 through its transcription regulating function. Dual low expression of STING1 and SMAD4 had more power in predicting patient survival. These results indicate that SMAD4-silenced CCA may downregulate its STING1 expression to adapt to the immune system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号