cGAS–STING

cGAS - STING
  • 文章类型: Journal Article
    线粒体自噬,对线粒体健康至关重要,选择性降解受损的线粒体。它与cGAS-STING途径有着错综复杂的联系,这对先天免疫至关重要。该途径响应线粒体DNA并且与细胞应激反应相关。我们的综述探讨了线粒体自噬和cGAS-STING通路的分子细节和调控机制。我们批判性地评估了证明功能失调的线粒体自噬如何导致神经炎症的文献,主要是通过受损线粒体的积累,激活cGAS-STING途径。这种激活促使促炎细胞因子的产生,加剧神经炎症.这篇综述强调了线粒体自噬与cGAS-STING通路之间的相互作用。有效的线粒体自噬可以抑制cGAS-STING通路,提供对神经炎症的保护。相反,受损的线粒体自噬可能激活cGAS-STING通路,导致慢性神经炎症.此外,我们探索了这种相互作用如何影响神经退行性疾病,提示了这些疾病的共同机制。总之,需要进行更多的靶向研究,以揭示线粒体自噬-cGAS-STING相互作用的复杂性及其在神经变性中的作用.这篇综述强调了针对这些途径的潜在疗法,可能导致神经炎症和神经退行性疾病的新疗法。这种合成增强了我们对神经炎症的细胞和分子基础的理解,并为神经退行性疾病研究开辟了新的治疗途径。
    Mitophagy, essential for mitochondrial health, selectively degrades damaged mitochondria. It is intricately linked to the cGAS-STING pathway, which is crucial for innate immunity. This pathway responds to mitochondrial DNA and is associated with cellular stress response. Our review explores the molecular details and regulatory mechanisms of mitophagy and the cGAS-STING pathway. We critically evaluate the literature demonstrating how dysfunctional mitophagy leads to neuroinflammatory conditions, primarily through the accumulation of damaged mitochondria, which activates the cGAS-STING pathway. This activation prompts the production of pro-inflammatory cytokines, exacerbating neuroinflammation. This review emphasizes the interaction between mitophagy and the cGAS-STING pathways. Effective mitophagy may suppress the cGAS-STING pathway, offering protection against neuroinflammation. Conversely, impaired mitophagy may activate the cGAS-STING pathway, leading to chronic neuroinflammation. Additionally, we explored how this interaction influences neurodegenerative disorders, suggesting a common mechanism underlying these diseases. In conclusion, there is a need for additional targeted research to unravel the complexities of mitophagy-cGAS-STING interactions and their role in neurodegeneration. This review highlights potential therapies targeting these pathways, potentially leading to new treatments for neuroinflammatory and neurodegenerative conditions. This synthesis enhances our understanding of the cellular and molecular foundations of neuroinflammation and opens new therapeutic avenues for neurodegenerative disease research.
    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
    自然杀伤(NK)细胞在先天免疫和适应性免疫的激活中起着至关重要的作用。Mn2+对环GMP-AMP(cGAS)刺激因子的干扰素基因的激活作用(STING信号已经众所周知,但它对NK细胞的影响仍然难以捉摸。在这项研究中,我们确定了锰(Mn2+)在NK细胞活化中的重要作用。Mn2+直接增强NK细胞的细胞毒性,促进NK细胞分泌细胞因子,从而激活CD8+T细胞并增强其抗肿瘤活性。此外,Mn2+可以同时激活NK细胞内在cGAS和STING,从而增加X染色体上普遍转录的四三肽重复序列的表达(UTX促进NK细胞的反应性。我们的结果有助于更广泛地理解cGAS-STING如何调节NK细胞。作为cGAS-STING的有效激动剂,Mn2+为基于NK细胞的癌症免疫治疗提供了一个有希望的选择。
    Natural killer (NK) cells play a crucial role in both innate immunity and the activation of adaptive immunity. The activating effect of Mn2+ on cyclic GMP-AMP(cGAS)-stimulator of interferon genes (STING signaling has been well known, but its effect on NK cells remains elusive. In this study, we identified the vital role of manganese (Mn2+) in NK cell activation. Mn2+ directly boosts cytotoxicity of NK cells and promotes the cytokine secretion by NK cells, thereby activating CD8+ T cells and enhancing their antitumor activity. Furthermore, Mn2+ can simultaneously activate NK-cell intrinsic cGAS and STING and consequently augment the expression of ubiquitously transcribed tetratricopeptide repeat on chromosome X (UTX to promote the responsiveness of NK cells. Our results contribute to a broader comprehension of how cGAS-STING regulates NK cells. As a potent agonist of cGAS-STING, Mn2+ provides a promising option for NK cell-based immunotherapy of cancers.
    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
    以视网膜神经节细胞(RGC)的进行性变性和视力丧失为特征,青光眼是不可逆性失明的主要原因,无法治愈,影响超过7800万患者。然而,导致青光眼诱导的RGC丢失的致病机制尚不完全清楚.出乎意料的是,我们发现cGAS-STING(干扰素基因的2'3'-环GMP-AMP-刺激物)信号,它监测细胞溶质中的双链DNA(dsDNA)的移位,并启动先天免疫反应,在不同的小鼠模型中,在视网膜小胶质细胞的青光眼期间被强烈激活。STING的整体或小胶质细胞缺失显着缓解了青光眼症状,并保护了RGC变性和视力丧失,而具有遗传cGAS-STING超敏反应的小鼠加重了视网膜神经炎症和RGC丧失。机械上,来自组织损伤激活的小胶质细胞cGAS-STING信号的dsDNA,通过细胞因子介导的小胶质细胞-大胶质细胞相互作用在视网膜中引起有害的大胶质细胞反应,逐渐驱动RGCs的凋亡性死亡。值得注意的是,通过眼内注射TBK1i或抗IFNAR1抗体靶向cGAS-STING信号的临床前研究可预防青光眼诱导的RGC和视力丧失。因此,我们揭示了cGAS-STING信号传导在青光眼发病机制中的重要作用,并提出了治疗这种破坏性疾病的有希望的治疗策略。
    Characterized by progressive degeneration of retinal ganglion cells (RGCs) and vision loss, glaucoma is the primary cause of irreversible blindness, incurable and affecting over 78 million patients. However, pathogenic mechanisms leading to glaucoma-induced RGC loss are incompletely understood. Unexpectedly, we found that cGAS-STING (2\'3\'-cyclic GMP-AMP-stimulator of interferon genes) signaling, which surveils displaced double-stranded DNA (dsDNA) in the cytosol and initiates innate immune responses, was robustly activated during glaucoma in retinal microglia in distinct murine models. Global or microglial deletion of STING markedly relieved glaucoma symptoms and protected RGC degeneration and vision loss, while mice bearing genetic cGAS-STING supersensitivity aggravated retinal neuroinflammation and RGC loss. Mechanistically, dsDNA from tissue injury activated microglial cGAS-STING signaling, causing deleterious macroglia reactivity in retinas by cytokine-mediated microglia-macroglia interactions, progressively driving apoptotic death of RGCs. Remarkably, preclinical investigations of targeting cGAS-STING signaling by intraocular injection of TBK1i or anti-IFNAR1 antibody prevented glaucoma-induced losses of RGCs and vision. Therefore, we unravel an essential role of cGAS-STING signaling underlying glaucoma pathogenesis and suggest promising therapeutic strategies for treating this devastating disease.
    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
    细胞衰老,癌症的标志之一,其特征是细胞周期停滞和大多数正常细胞功能的丧失,同时获得过度分泌,促炎表型。癌细胞中衰老细胞的功能根据细胞状况而变化。在癌症发生之前,衰老细胞充当阻碍其发育的屏障。但是一旦癌症发生,衰老细胞起着促进肿瘤的作用。然而,目前的研究很少能充分解释癌症细胞衰老的多样性.在这里,我们详细总结了细胞衰老的最新内在机制,并强调衰老相关的分泌表型是肿瘤衰老细胞异质性的关键因素.我们还讨论了五种细胞衰老的诱导剂以及在癌症中的促衰老作用。这些药物倾向于清除衰老细胞。最后,我们总结了衰老细胞在不同癌症中的各种作用,并表明它们的功能在不同情况下可能是完全相反的。总之,本文有助于理解癌症细胞衰老的多样性,并为肿瘤治疗提供新的见解。
    Cellular senescence, one of the hallmarks of cancer, is characterized by cell cycle arrest and the loss of most normal cellular functions while acquiring a hypersecretory, proinflammatory phenotype. The function of senescent cells in cancer cells varies depending on the cellular conditions. Before the occurrence of cancer, senescent cells act as a barrier to prevent its development. But once cancer has occurred, senescent cells play a procancer role. However, few of the current studies have adequately explained the diversity of cellular senescence across cancers. Herein, we concluded the latest intrinsic mechanisms of cellular senescence in detail and emphasized the senescence-associated secretory phenotype as a key contributor to heterogeneity of senescent cells in tumor. We also discussed five kinds of inducers of cellular senescence and the advancement of senolytics in cancer, which are drugs that tend to clear senescent cells. Finally, we summarized the various effects of senescent cells in different cancers and manifested that their functions may be diametrically opposed under different circumstances. In short, this paper contributes to the understanding of the diversity of cellular senescence in cancers and provides novel insight for tumor therapy.
    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
    环GMP-AMP合酶(cGAS),哺乳动物细胞中一个突出的细胞内DNA传感器,控制先天免疫反应和干扰素基因(STING)介导的促炎细胞因子合成的刺激物,例如I型干扰素(IFN-I)。几十年来,IFN-I被认为在系统性红斑狼疮(SLE)的发展中至关重要,一种慢性多系统自身免疫,其特征是免疫复合物(IC)沉积在小血管中。最近的发现表明,自身DNA对cGAS-STING途径的激活将通过上调SLE中IFN-I的产生来传播自身免疫反应。在这次审查中,我们旨在全面展望cGAS-STING通路在SLE病理生物学中的作用,还有,更好地了解当前针对此轴的治疗机会。
    The cyclic GMP-AMP synthase (cGAS), a prominent intracellular DNA sensor in mammalian cells, controls the innate immune response and the stimulator of interferon genes (STING)-mediated synthesis of pro-inflammatory cytokines, such as type-I interferon (IFN-I). For decades, IFN-I has been hypothesized to be essential in the development of systemic lupus erythematosus (SLE), a chronic multisystem autoimmunity characterized by immune complex (IC) deposition in small vessels. Recent findings revealed that the activation of the cGAS-STING pathway by self-DNA would propagate the autoimmune responses via upregulating IFN-I production in SLE. In this review, we aimed to provide a comprehensive outlook of the role of the cGAS-STING pathway in SLE pathobiology, as well as, a better understanding of current therapeutic opportunities targeting this axis.
    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
    当细胞增殖时,对DNA复制的压力或暴露于内源性或外部损伤通常会导致DNA损伤。DNA损伤反应(DDR)网络是多细胞生物用于防止DNA损伤的复杂信号通路。根据破碎DNA的类型,各种途径,基本切除修复(BER),核苷酸切除修复(NER),不匹配修复(MMR),同源重组(HR),非同源末端连接(NHEJ),链间交联(ICL)修理,和其他直接修复途径,可以单独或组合激活以修复DNA损伤。为了保持体内平衡,先天和适应性免疫应答是抵抗内源性突变或外来病原体入侵的有效防御。有趣的是,新的研究不断显示DDR成分与免疫系统的关系。DDR和免疫应答通过免疫效应物例如环GMP-AMP合酶(cGAS)-干扰素基因刺激因子(STING)途径连接。这些效应物充当DNA损伤引起的免疫反应的传感器。此外,DDR组分本身在免疫应答中起作用以触发级联中的炎性细胞因子的产生或甚至触发程序性细胞死亡。已知有缺陷的DDR成分会破坏基因组稳定性并损害免疫反应。加重免疫失衡,导致癌症和自身免疫性疾病等严重疾病。这项研究探讨了DDR元件与免疫反应之间相互作用的最新进展。DDR网络的免疫调节剂的双重作用可能为治疗与DNA损伤相关的感染性疾病提供新的视角。包括癌症,以及靶向免疫治疗的发展。
    When cells proliferate, stress on DNA replication or exposure to endogenous or external insults frequently results in DNA damage. DNA-Damage Response (DDR) networks are complex signaling pathways used by multicellular organisms to prevent DNA damage. Depending on the type of broken DNA, the various pathways, Base-Excision Repair (BER), Nucleotide Excision Repair (NER), Mismatch Repair (MMR), Homologous Recombination (HR), Non-Homologous End-Joining (NHEJ), Interstrand Crosslink (ICL) repair, and other direct repair pathways, can be activated separately or in combination to repair DNA damage. To preserve homeostasis, innate and adaptive immune responses are effective defenses against endogenous mutation or invasion by external pathogens. It is interesting to note that new research keeps showing how closely DDR components and the immune system are related. DDR and immunological response are linked by immune effectors such as the cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway. These effectors act as sensors of DNA damage-caused immune response. Furthermore, DDR components themselves function in immune responses to trigger the generation of inflammatory cytokines in a cascade or even trigger programmed cell death. Defective DDR components are known to disrupt genomic stability and compromise immunological responses, aggravating immune imbalance and leading to serious diseases such as cancer and autoimmune disorders. This study examines the most recent developments in the interaction between DDR elements and immunological responses. The DDR network\'s immune modulators\' dual roles may offer new perspectives on treating infectious disorders linked to DNA damage, including cancer, and on the development of target immunotherapy.
    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
    自2013年发现环磷酸鸟苷-磷酸腺苷合成酶(cGAS)-干扰素基因刺激因子(STING)信号通路以来,在阐明其起源方面取得了很大进展,函数,cGAS-STING信号通路的调控机制。同时,触发和转导机制不断被阐明。cGAS-STING在人类疾病中起着关键作用,特别是DNA引发的炎性疾病,使其成为炎症相关疾病的潜在有效治疗靶点。这里,我们旨在总结cGAS-STING防御机制的古老起源,以及触发器,转导,和cGAS-STING的调节机制。我们还将关注cGAS-STING信号在病理条件下的重要作用,如感染,癌症,自身免疫性疾病,神经系统疾病,和内脏炎症,并综述了以cGAS-STING信号通路为靶点的药物研发进展。将讨论cGAS-STING信号通路在炎症性疾病和癌症的调节机制研究和治疗药物开发中的主要方向和潜在障碍。这些研究进展扩大了我们对cGAS-STING的理解,为进一步探讨cGAS-STING在疾病中的作用提供理论依据,并开辟了靶向cGAS-STING的新策略,将其作为多种疾病的有希望的治疗干预措施。
    Since cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway was discovered in 2013, great progress has been made to elucidate the origin, function, and regulating mechanism of cGAS-STING signaling pathway in the past decade. Meanwhile, the triggering and transduction mechanisms have been continuously illuminated. cGAS-STING plays a key role in human diseases, particularly DNA-triggered inflammatory diseases, making it a potentially effective therapeutic target for inflammation-related diseases. Here, we aim to summarize the ancient origin of the cGAS-STING defense mechanism, as well as the triggers, transduction, and regulating mechanisms of the cGAS-STING. We will also focus on the important roles of cGAS-STING signal under pathological conditions, such as infections, cancers, autoimmune diseases, neurological diseases, and visceral inflammations, and review the progress in drug development targeting cGAS-STING signaling pathway. The main directions and potential obstacles in the regulating mechanism research and therapeutic drug development of the cGAS-STING signaling pathway for inflammatory diseases and cancers will be discussed. These research advancements expand our understanding of cGAS-STING, provide a theoretical basis for further exploration of the roles of cGAS-STING in diseases, and open up new strategies for targeting cGAS-STING as a promising therapeutic intervention in multiple 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
    涉及远端穿支皮瓣的手术后,血管坏死通常是并发症。二氢辣椒素(DHC)可以保护组织免受缺血再灌注(I/R)损伤,但其在多区穿支皮瓣中的具体作用尚不清楚。在这项研究中,使用网络药理学分析预测I/R损伤背景下DHC的前瞻性目标.通过存活面积分析确定皮瓣活力,激光多普勒血流,血管造影,和组织学检查。血管生成的表现,凋亡,NLR家族pyrin结构域含3(NLRP3)炎性体,氧化应激,和分子相关的环磷酸鸟苷(GMP)-腺苷一磷酸合成酶(cGAS)-干扰素基因刺激(STING)途径使用蛋白质印迹进行评估,免疫荧光,TUNEL染色,和二氢乙锭(DHE)染色。我们的发现显示DHC促进了穿支皮瓣的存活,涉及到cGAS-STING途径,氧化应激,NLRP3炎性体,凋亡,和血管生成。DHC诱导抗氧化应激和抑制NLRP3炎性体,防止血管内皮细胞凋亡。通过STING途径的调节,DHC控制缺血皮瓣中内皮细胞的氧化应激和NLRP3水平。然而,cGAS-STING途径的激活导致活性氧(ROS)和NLRP3炎性体的积累,从而降低DHC的保护作用。DHC通过抑制cGAS-STING通路增强多域穿支皮瓣的存活,氧化应激,和NLRP3炎性体的形成。这些发现揭示了促进多域穿支皮瓣存活的潜在新机制,具有临床意义。
    Avascular necrosis frequently occurs as a complication following surgery involving the distal perforator flap. Dihydrocapsaicin (DHC) can protect tissue from ischemia-reperfusion (I/R) injury, but its specific role in multizone perforator flaps remains unclear. In this study, the prospective target of DHC in the context of I/R injury was predicted using network pharmacology analysis. Flap viability was determined through survival area analysis, laser Doppler blood flow, angiograms, and histological examination. The expressions of angiogenesis, apoptosis, NLR family pyrin domain containing 3 (NLRP3) inflammasome, oxidative stress, and molecules related to cyclic guanosine monophosphate (GMP)-adenosine monophosphate synthase (cGAS)-interferon gene stimulant (STING) pathway were assessed using western blotting, immunofluorescence, TUNEL staining, and dihydroethidium (DHE) staining. Our finding revealed that DHC promoted the perforator flap survival, which involves the cGAS-STING pathway, oxidative stress, NLRP3 inflammasome, apoptosis, and angiogenesis. DHC induced oxidative stress resistance and suppressed the NLRP3 inflammasome, preventing apoptosis in vascular endothelial cells. Through regulation of STING pathway, DHC controlled oxidative stress in endothelial cells and NLRP3 levels in ischemic flaps. However, activation of the cGAS-STING pathway led to the accumulation of reactive oxygen species (ROS) and NLRP3 inflammasome, thereby diminishing the protective role of DHC. DHC enhanced the survival of multidomain perforator flaps by suppressing the cGAS-STING pathway, oxidative stress, and the formation of NLRP3 inflammasome. These findings unveil a potentially novel mechanism with clinical significance for promoting the survival of multidomain perforator flaps.
    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:神经炎症在术后认知功能障碍(POCD)的发展中至关重要,小胶质细胞激活是这一过程的积极参与者。SS-31,一种线粒体靶向抗氧化剂,被广泛认为是治疗神经退行性疾病和炎症性疾病的潜在药物。在这项研究中,我们试图探讨SS-31是否发挥神经保护作用及其潜在机制.
    方法:对18月龄小鼠进行胫骨骨折内固定,以诱导手术相关的神经认知功能障碍。向BV2细胞施用LPS以诱导神经炎症。神经行为缺陷,海马损伤,蛋白质表达,在用SS-31、PHB2siRNA和STING激动剂处理后评估线粒体自噬水平和细胞状态。
    结果:我们的研究表明,SS-31与PHB2相互作用以激活线粒体自噬并改善手术老年小鼠的神经损伤,这归因于cGAS-STING途径的减少和M1小胶质细胞极化,原因是线粒体DNA(mtDNA)而不是核DNA(nDNA)的释放减少。体外,PHB2和STING激动剂的敲减消除了SS-31的保护作用。
    结论:SS-31通过促进PHB2介导的线粒体自噬激活以抑制mtDNA释放而赋予针对POCD的神经保护作用,这反过来又抑制了cGAS-STING途径和M1小胶质细胞极化。
    BACKGROUND: Neuroinflammation is crucial in the development of postoperative cognitive dysfunction (POCD), and microglial activation is an active participant in this process. SS-31, a mitochondrion-targeted antioxidant, is widely regarded as a potential drug for neurodegenerative diseases and inflammatory diseases. In this study, we sought to explore whether SS-31 plays a neuroprotective role and the underlying mechanism.
    METHODS: Internal fixation of tibial fracture was performed in 18-month-old mice to induce surgery-associated neurocognitive dysfunction. LPS was administrated to BV2 cells to induce neuroinflammation. Neurobehavioral deficits, hippocampal injury, protein expression, mitophagy level and cell state were evaluated after treatment with SS-31, PHB2 siRNA and an STING agonist.
    RESULTS: Our study revealed that SS-31 interacted with PHB2 to activate mitophagy and improve neural damage in surgically aged mice, which was attributed to the reduced cGAS-STING pathway and M1 microglial polarization by decreased release of mitochondrial DNA (mtDNA) but not nuclear DNA (nDNA). In vitro, knockdown of PHB2 and an STING agonist abolished the protective effect of SS-31.
    CONCLUSIONS: SS-31 conferred neuroprotection against POCD by promoting PHB2-mediated mitophagy activation to inhibit mtDNA release, which in turn suppressed the cGAS-STING pathway and M1 microglial polarization.
    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:化疗有可能在肿瘤微环境(TME)中诱导CD8+T细胞浸润,并在包括食管鳞状细胞癌(ESCC)在内的几种癌症中激活抗肿瘤免疫应答。已知肿瘤细胞固有的环状GMP-AMP合酶(cGAS)-干扰素基因刺激因子(STING)途径是调节TME中免疫细胞活化的关键组成部分。然而,其对ESCCTME中化疗诱导的免疫细胞浸润的影响尚未研究。
    方法:我们使用ESCC细胞系和接受新辅助化疗(NAC)的患者手术切除的ESCC标本,研究了肿瘤细胞内在cGAS-STING通路对化疗诱导的CD8+T细胞浸润的影响。
    结果:我们发现化疗药物,包括5-氟尿嘧啶(5-FU)和顺铂(CDDP),激活了cGAS-STING途径,因此诱导ESCC细胞中I型干扰素和T细胞吸引趋化因子的表达。此外,cGAS-STING的肿瘤细胞内在表达与NAC后ESCC中CD8T细胞密度显着正相关。然而,cGAS-STING的肿瘤细胞内在表达对NAC后ESCC患者的临床结局无显著影响.
    结论:我们的研究结果表明,肿瘤细胞固有的cGAS-STING途径可能有助于ESCCTME中化疗诱导的免疫细胞活化。
    BACKGROUND: Chemotherapy has the potential to induce CD8+ T-cell infiltration in the tumor microenvironment (TME) and activate the anti-tumor immune response in several cancers including esophageal squamous cell carcinoma (ESCC). The tumor cell-intrinsic cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has been known as a critical component for regulating immune cell activation in the TME. However, its effect on the infiltration of immune cells induced by chemotherapy in the ESCC TME has not been investigated.
    METHODS: We examined the effect of the tumor-cell intrinsic cGAS-STING pathway on the infiltration of CD8+ T cells induced by chemotherapy in ESCC using ESCC cell lines and surgically resected ESCC specimens from patients who received neoadjuvant chemotherapy (NAC).
    RESULTS: We found that chemotherapeutic agents, including 5-fluorouracil (5-FU) and cisplatin (CDDP), activated the cGAS-STING pathway, consequently inducing the expression of type I interferon and T-cell-attracting chemokines in ESCC cells. Moreover, the tumor cell-intrinsic expression of cGAS-STING was significantly and positively associated with the density of CD8+ T cells in ESCC after NAC. However, the tumor cell-intrinsic expression of cGAS-STING did not significantly impact clinical outcomes in patients with ESCC after NAC.
    CONCLUSIONS: Our findings suggest that the tumor cell-intrinsic cGAS-STING pathway might contribute to chemotherapy-induced immune cell activation in the ESCC TME.
    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