STAT1

Stat1
  • 文章类型: Journal Article
    急性胰腺炎(AP)是一种由外分泌腺泡细胞死亡引发的炎症性疾病,但其发病机制尚不清楚。信号转导和转录激活因子3(STAT3)是调节免疫和炎症反应的多功能因子。报道了STAT3在柯萨奇病毒B3(CVB3)诱导的心脏纤维化中的保护作用,然而,STAT3在调节胰腺中病毒诱导的STAT1激活和I型干扰素(IFN)刺激的基因(ISG)转录中的确切作用仍不清楚.在这项研究中,我们测试了STAT3是否调节病毒诱导的STAT1易位.我们发现CVB3,特别是衣壳VP1蛋白,显著上调STAT3(p-STAT3)的磷酸化和核输入,同时显著阻碍p-STAT1在感染后第3天(p.i.)在小鼠胰腺和心脏中的核转位.免疫印迹和免疫荧光分析表明,在CVB3感染的腺泡266-6细胞中,p-STAT3的表达和核易位增加,但p-STAT1核易位减弱。STAT3shRNA敲低或STAT3抑制剂通过拯救STAT1核易位并增加体外ISRE活性和ISG转录来减少病毒复制。STAT1的敲低阻断了STAT3抑制剂的抗病毒作用。STAT3通过病毒诱导IFN信号的有效抑制剂来抑制STAT1的激活,细胞因子信号传导抑制因子-3((SOCS)-3)。在SOCS3敲低细胞中诱导持续pSTAT1和ISGs的表达升高。体内施用HJC0152,一种药物STAT3抑制剂,通过在第3天p.i.增加IFNβ和ISG表达并减少多器官中的病毒载量来减轻病毒诱导的AP和心肌炎病理。这些发现将STAT3定义为I型IFN应答的负调节因子,其通过阻止核STAT1易位,否则在感染的胰腺和心脏中触发ISG诱导。我们的发现将STAT3确定为IFN-STAT1信号通路的拮抗因子,并为病毒诱导的AP和心肌炎提供了潜在的治疗靶点。
    Acute pancreatitis (AP) is an inflammatory disease initiated by the death of exocrine acinar cells, but its pathogenesis remains unclear. Signal transducer and activator of transcription 3 (STAT3) is a multifunctional factor that regulates immunity and the inflammatory response. The protective role of STAT3 is reported in Coxsackievirus B3 (CVB3)-induced cardiac fibrosis, yet the exact role of STAT3 in modulating viral-induced STAT1 activation and type I interferon (IFN)-stimulated gene (ISG) transcription in the pancreas remains unclarified. In this study, we tested whether STAT3 regulated viral-induced STAT1 translocation. We found that CVB3, particularly capsid VP1 protein, markedly upregulated the phosphorylation and nuclear import of STAT3 (p-STAT3) while it significantly impeded the nuclear translocation of p-STAT1 in the pancreases and hearts of mice on day 3 postinfection (p.i.). Immunoblotting and an immunofluorescent assay demonstrated the increased expression and nuclear translocation of p-STAT3 but a blunted p-STAT1 nuclear translocation in CVB3-infected acinar 266-6 cells. STAT3 shRNA knockdown or STAT3 inhibitors reduced viral replication via the rescue of STAT1 nuclear translocation and increasing the ISRE activity and ISG transcription in vitro. The knockdown of STAT1 blocked the antiviral effect of the STAT3 inhibitor. STAT3 inhibits STAT1 activation by virally inducing a potent inhibitor of IFN signaling, the suppressor of cytokine signaling-3 ((SOCS)-3). Sustained pSTAT1 and the elevated expression of ISGs were induced in SOCS3 knockdown cells. The in vivo administration of HJC0152, a pharmaceutical STAT3 inhibitor, mitigated the viral-induced AP and myocarditis pathology via increasing the IFNβ as well as ISG expression on day 3 p.i. and reducing the viral load in multi-organs. These findings define STAT3 as a negative regulator of the type I IFN response via impeding the nuclear STAT1 translocation that otherwise triggers ISG induction in infected pancreases and hearts. Our findings identify STAT3 as an antagonizing factor of the IFN-STAT1 signaling pathway and provide a potential therapeutic target for viral-induced AP and myocarditis.
    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
    严重再生障碍性贫血(SAA)是一种危及生命的骨髓衰竭综合征,其发展可由环境引发,自身免疫,和/或遗传因素。后者包括基因中的种系致病性变体,这些变体会导致习惯性易感综合征以及仅偶尔发生的免疫缺陷。这些疾病之一是慢性粘膜皮肤念珠菌病(CMC)的常染色体显性形式,其由种系STAT1功能获得(GOF)致病变体定义。STAT1的过度表达和组成型激活导致Janus激酶/信号转导子和转录激活因子1(STAT)信号通路失调,它通常组织免疫和造血系统不同成分的发育和适当的相互作用。尽管SAA是这种疾病中极为罕见的并发症,当很明显潜在的致病机制可能,以类似的方式,也有助于至少一些特发性SAA病例。基于这些前提,我们在此介绍的是CMC家族中历史上最可能的首例脐带血移植SAA病例,该家族有STAT1GOF致病变异.此外,我们概述了迄今为止报道的6例CMCSAA病例的特征,并讨论了STAT1GOF致病变异和其他STAT1信号紊乱在这些特定类型的骨髓衰竭综合征中的意义.因为一个组成型激活的STAT1信号,无论是由STAT1GOF种系致病变异或任何其他致病变异独立事件驱动,显然对于启动和维持SAA疾病过程很重要,我们建议承认SAA是STAT1突变的CMC病例中明确的疾病表现之一.出于同样的原因,我们认为有必要将STAT1的分子和功能分析纳入SAA病例的诊断工作.
    Severe aplastic anemia (SAA) is a life-threatening bone marrow failure syndrome whose development can be triggered by environmental, autoimmune, and/or genetic factors. The latter comprises germ line pathogenic variants in genes that bring about habitually predisposing syndromes as well as immune deficiencies that do so only occasionally. One of these disorders is the autosomal dominant form of chronic mucocutaneous candidiasis (CMC), which is defined by germ line STAT1 gain-of-function (GOF) pathogenic variants. The resultant overexpression and constitutive activation of STAT1 dysregulate the Janus kinase/signal transducer and activator of transcription 1 (STAT) signaling pathway, which normally organizes the development and proper interaction of different components of the immunologic and hematopoietic system. Although SAA is an extremely rare complication in this disorder, it gained a more widespread interest when it became clear that the underlying causative pathomechanism may, in a similar fashion, also be instrumental in at least some of the idiopathic SAA cases. Based on these premises, we present herein what is the historically most likely first cord blood-transplanted SAA case in a CMC family with a documented STAT1 GOF pathogenic variant. In addition, we recapitulate the characteristics of the six CMC SAA cases that have been reported so far and discuss the significance of STAT1 GOF pathogenic variants and other STAT1 signaling derangements in the context of these specific types of bone marrow failure syndromes. Because a constitutively activated STAT1 signaling, be it driven by STAT1 GOF germ line pathogenic variants or any other pathogenic variant-independent events, is apparently important for initiating and maintaining the SAA disease process, we propose to acknowledge that SAA is one of the definite disease manifestations in STAT1-mutated CMC cases. For the same reason, we deem it necessary to also incorporate molecular and functional analyses of STAT1 into the diagnostic work-up of SAA cases.
    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:缺血性卒中在大脑中引起复杂且持续的免疫应答。免疫调节治疗长期以来一直有望改善卒中结局,然而,没有一个在临床上成功。这种缺乏成功的主要原因是我们对免疫细胞如何应对中风的理解不完全。本研究的目的是剖析永久性中风对小胶质细胞的影响,大脑实质内的固有免疫细胞。
    方法:采用永久性大脑中动脉闭塞(pMCAO)模型在幼年雄性和雌性小鼠中诱发缺血性中风。pMCAO或假手术后,从荧光报告小鼠中分选小胶质细胞,然后进行单细胞RNA测序分析。各种方法,包括流式细胞术,RNA原位杂交,免疫组织化学,全脑成像,骨髓移植,还用于解剖小胶质细胞对中风的反应。通过梗死面积和行为测试评估卒中结果。
    结果:首先,我们显示了卒中后小胶质细胞的形态和空间变化。然后,我们对从两种性别的假手术和中风小鼠中分离的小胶质细胞进行了单细胞RNA测序分析。数据表明,对永久性中风的小胶质细胞反应中没有主要的性二态性。值得注意的是,我们确定了七个潜在的卒中相关小胶质细胞簇,包括以疾病相关的小胶质细胞样特征为特征的四个主要簇,一种高度增殖的状态,巨噬细胞样的轮廓,和干扰素(IFN)应答签名,分别。重要的是,我们提供的证据表明,巨噬细胞样簇可能代表长期寻找的卒中诱导的小胶质细胞亚群,CD45表达增加.最后,鉴于IFN反应性子集构成了中风大脑中最突出的小胶质细胞群体,我们使用氟达拉滨在药理学上靶向STAT1信号,发现氟达拉滨治疗可改善长期卒中结局.
    结论:我们的研究结果为小胶质细胞在卒中病理学中的异质性提供了新的启示,并强调了针对特定小胶质细胞群体进行有效卒中治疗的潜力。
    BACKGROUND: Ischemic stroke elicits a complex and sustained immune response in the brain. Immunomodulatory treatments have long held promise for improving stroke outcomes, yet none have succeeded in the clinical setting. This lack of success is largely due to our incomplete understanding of how immune cells respond to stroke. The objective of the current study was to dissect the effect of permanent stroke on microglia, the resident immune cells within the brain parenchyma.
    METHODS: A permanent middle cerebral artery occlusion (pMCAO) model was used to induce ischemic stroke in young male and female mice. Microglia were sorted from fluorescence reporter mice after pMCAO or sham surgery and then subjected to single-cell RNA sequencing analysis. Various methods, including flow cytometry, RNA in situ hybridization, immunohistochemistry, whole-brain imaging, and bone marrow transplantation, were also employed to dissect the microglial response to stroke. Stroke outcomes were evaluated by infarct size and behavioral tests.
    RESULTS: First, we showed the morphologic and spatial changes in microglia after stroke. We then performed single-cell RNA sequencing analysis on microglia isolated from sham and stroke mice of both sexes. The data indicate no major sexual dimorphism in the microglial response to permanent stroke. Notably, we identified seven potential stroke-associated microglial clusters, including four major clusters characterized by a disease-associated microglia-like signature, a highly proliferative state, a macrophage-like profile, and an interferon (IFN) response signature, respectively. Importantly, we provided evidence that the macrophage-like cluster may represent the long-sought stroke-induced microglia subpopulation with increased CD45 expression. Lastly, given that the IFN-responsive subset constitutes the most prominent microglial population in the stroke brain, we used fludarabine to pharmacologically target STAT1 signaling and found that fludarabine treatment improved long-term stroke outcome.
    CONCLUSIONS: Our findings shed new light on microglia heterogeneity in stroke pathology and underscore the potential of targeting specific microglial populations for effective stroke therapies.
    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
    射血分数保留的心力衰竭(HFpEF)是一种致命的临床综合征,没有有效的治疗方法。Empagliflozin(EMPA)改善HFpEF患者的心血管结局,但是潜在的机制仍然难以捉摸。这里,给小鼠喂食补充有L-NAME的高脂肪饮食(HFD)12周,随后腹膜内注射EMPA,再注射4周.进行4D-DIA蛋白质组学测定以检测衰竭心脏中的蛋白质变化。我们鉴定了310种差异表达蛋白(DEP)(ctrl与HFpEF组)和173DEP(HFpEF与EMPA组)。在所有组中都富集了免疫系统过程的调节,并且在HFpEF小鼠中干扰素反应基因(STAT1,Ifit1,Ifi35和Ifi47)上调,但在EMPA施用后下调。此外,EMPA治疗抑制了HFpEF心脏中衰老标志物(p16和p21)水平的增加。进一步的生物信息学分析证实STAT1是HFpEF小鼠病理变化过程中的hub转录因子。我们接下来用IFN-γ处理H9C2细胞,STAT1磷酸化的主要激动剂,调查EMPA是否通过阻断STAT1激活发挥有益作用。我们的结果表明,IFN-γ治疗引起心肌细胞衰老和STAT1激活,被EMPA给药抑制。值得注意的是,STAT1抑制可能通过调节STING表达显著降低细胞衰老。我们的发现表明,EMPA通过抑制STAT1激活减轻HFpEF小鼠的心脏炎症和衰老。STAT1-STING轴可能是HFpEF发病机制中的一个重要机制,尤其是在炎症和衰老条件下。
    Heart failure with preserved ejection fraction (HFpEF) is a mortal clinical syndrome without effective therapies. Empagliflozin (EMPA) improves cardiovascular outcomes in HFpEF patients, but the underlying mechanism remains elusive. Here, mice were fed a high-fat diet (HFD) supplemented with L-NAME for 12 weeks and subsequently intraperitoneally injected with EMPA for another 4 weeks. A 4D-DIA proteomic assay was performed to detect protein changes in the failing hearts. We identified 310 differentially expressed proteins (DEPs) (ctrl vs. HFpEF group) and 173 DEPs (HFpEF vs. EMPA group). The regulation of immune system processes was enriched in all groups and the interferon response genes (STAT1, Ifit1, Ifi35 and Ifi47) were upregulated in HFpEF mice but downregulated after EMPA administration. In addition, EMPA treatment suppressed the increase in the levels of aging markers (p16 and p21) in HFpEF hearts. Further bioinformatics analysis verified STAT1 as the hub transcription factor during pathological changes in HFpEF mice. We next treated H9C2 cells with IFN-γ, a primary agonist of STAT1 phosphorylation, to investigate whether EMPA plays a beneficial role by blocking STAT1 activation. Our results showed that IFN-γ treatment caused cardiomyocyte senescence and STAT1 activation, which were inhibited by EMPA administration. Notably, STAT1 inhibition significantly reduced cellular senescence possibly by regulating STING expression. Our findings revealed that EMPA mitigates cardiac inflammation and aging in HFpEF mice by inhibiting STAT1 activation. The STAT1-STING axis may act as a pivotal mechanism in the pathogenesis of HFpEF, especially under inflammatory and aging conditions.
    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
    肿瘤相关巨噬细胞在肿瘤微环境中起着至关重要的作用。三方主题59(TRIM59),三方主题(TRIM)家族的成员,已知与免疫性疾病和巨噬细胞活化有关。TRIM59通过巨噬细胞影响结直肠癌(CRC)发生发展的功能和分子机制尚不清楚。为了解决这个问题,我们产生了巨噬细胞特异性TRIM59条件性基因敲除小鼠,并利用这些小鼠建立结肠炎相关癌症和MC38移植CRC模型进行进一步研究.我们发现巨噬细胞中TRIM59的缺乏抑制小鼠结直肠肿瘤发生。这种肿瘤抑制作用是通过STAT1信号通路促进M1巨噬细胞的活化来实现的。进一步的机制研究表明,TRIM59可以通过泛素化和降解STAT1来调节巨噬细胞极化。这些发现提供了证据,表明TRIM59缺乏促进M1巨噬细胞活化,并通过STAT1信号通路抑制CRC,提示TRIM59/STAT1信号通路可能是CRC的一个有希望的靶点。
    Tumor-associated macrophages play a crucial role in the tumor microenvironment. Tripartite motif 59 (TRIM59), a member of the tripartite motif (TRIM) family, is known to be associated with immunological diseases and macrophage activation. The functional and molecular mechanisms by which TRIM59 affects the occurrence and development of colorectal cancer (CRC) through macrophages are still not well understood. To address this, we generated macrophage-specific TRIM59 conditional knockout mice and utilized these mice to establish colitis-associated cancer and MC38 transplanted CRC models for further investigation. We found that the deficiency of TRIM59 in macrophages inhibited colorectal tumorigenesis in mice. This tumor-suppressive effect was achieved by promoting the activation of M1 macrophages via STAT1 signaling pathway. Further mechanistic studies revealed that TRIM59 could regulate macrophage polarization by ubiquitinating and degrading STAT1. These findings provide evidence that TRIM59 deficiency promotes M1 macrophage activation and inhibits CRC through the STAT1 signaling pathway, suggesting that the TRIM59/STAT1 signaling pathway may be a promising target for CRC.
    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
    低度神经胶质瘤(LGG)是一种常见且致命的原发性脑恶性肿瘤,大多数患者死于复发和进展。信号转导和转录激活因子(STAT)家族长期以来一直与肿瘤的发生和发展有关。然而,对LGG中STAT基因的表达状态和整体功能的综合评估仍未报道。在这项研究中,我们研究了STAT家族基因的表达与LGG进展之间的关联。通过综合分析,结合生物信息学筛选和验证分析,我们确定STAT1,STAT3和STAT5A上调,并导致LGG的恶性进展.值得注意的是,我们的研究结果表明,STAT3是调节LGG进展的重要预后标志物.STAT3成为控制LGG进展的最重要的预后指标。此外,我们对STAT3结合蛋白和差异表达相关基因(DEGs)的研究发现,STAT3通过刺激STAT1,FOXO1和MYC的表达在LGG的进展中起关键作用.总之,我们最近的研究对STAT家族基因进行了全面分析,发现针对STAT3的治疗干预措施有可能作为治疗LGG患者的可行策略.
    Low-grade glioma (LGG) is a prevalent and lethal primary brain malignancy, with most patients succumbing to recurrence and progression. The signal transducer and activator of transcription (STAT) family has long been implicated in tumor initiation and progression. However, a comprehensive evaluation of the expression status and overall function of STAT genes in LGG remains largely unreported. In this study, we investigated the association between the expression of STAT family genes and the progression of LGG. Through a comprehensive analysis that combined bioinformatics screening and validation assays, we determined that STAT1, STAT3, and STAT5A were upregulated and contributed to the malignant progression of LGG. Notably, our findings suggest that STAT3 is a critical prognostic marker that regulates the progression of LGG. STAT3 emerged as the most significant prognostic indicator governing the advancement of LGG. Additionally, our inquiry into the STAT3-binding proteins and differentially expressed-correlated genes (DEGs) revealed that STAT3 played a pivotal role in the progression of LGG by stimulating the expression of STAT1, FOXO1, and MYC. In summary, our recent study conducted a thorough analysis of the STAT family genes and revealed that directing therapeutic interventions towards STAT3 holds potential as a viable strategy for treating patients with LGG.
    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
    招募到大脑和脊髓中的促炎性T淋巴细胞介导多发性硬化症(MS),目前尚无治愈MS的方法。产生IFN-γ的Th1细胞诱导脊髓上行性瘫痪,而产生IL-17的Th17细胞介导小脑共济失调。STAT1和STAT3是Th1和Th17开发所必需的,分别,因此,同时靶向STAT1和STAT3途径是抑制脊髓和脑部疾病的潜在治疗策略。然而,STAT1和STAT3的药理学靶向由于它们的细胞内定位而提出了重大挑战。我们已经开发了一种源自骆驼科动物的STAT特异性单结构域纳米抗体(SBT-100),其靶向STAT1和STAT3的Src同源物2(SH2)结构域中的保守残基。这项研究调查了SBT-100是否可以抑制实验性自身免疫性脑脊髓炎(EAE),MS的小鼠模型我们表明SBT-100通过抑制脑和脊髓中Th17和Th1细胞的扩增来改善脑脊髓炎。过继转移实验表明,SBT-100处理的EAE小鼠的淋巴细胞诱导EAE的能力降低,这表明免疫抑制作用来源于直接抑制产脑病T细胞。SBT-100的小尺寸使这种STAT特异性纳米抗体成为中枢神经系统自身免疫性疾病的有前途的免疫疗法,包括多发性硬化症.
    Proinflammatory T-lymphocytes recruited into the brain and spinal cord mediate multiple sclerosis (MS) and currently there is no cure for MS. IFN-γ-producing Th1 cells induce ascending paralysis in the spinal cord while IL-17-producing Th17 cells mediate cerebellar ataxia. STAT1 and STAT3 are required for Th1 and Th17 development, respectively, and the simultaneous targeting of STAT1 and STAT3 pathways is therefore a potential therapeutic strategy for suppressing disease in the spinal cord and brain. However, the pharmacological targeting of STAT1 and STAT3 presents significant challenges because of their intracellular localization. We have developed a STAT-specific single-domain nanobody (SBT-100) derived from camelids that targets conserved residues in Src homolog 2 (SH2) domains of STAT1 and STAT3. This study investigated whether SBT-100 could suppress experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show that SBT-100 ameliorates encephalomyelitis through suppressing the expansion of Th17 and Th1 cells in the brain and spinal cord. Adoptive transfer experiments revealed that lymphocytes from SBT-100-treated EAE mice have reduced capacity to induce EAE, indicating that the immunosuppressive effects derived from the direct suppression of encephalitogenic T-cells. The small size of SBT-100 makes this STAT-specific nanobody a promising immunotherapy for CNS autoimmune diseases, including multiple sclerosis.
    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
    癫痫的发病机制尚不清楚;然而,一个普遍的假设表明,主要的根本原因是神经元兴奋性和抑制之间的不平衡。葡萄糖-6-磷酸脱氢酶(G6PD)是戊糖磷酸途径中的关键酶,主要参与脱氧核酸合成和抗氧化防御机制,在癫痫慢性期表现出表达增加,主要与神经元共定位。G6PD过表达显着降低了自发性复发性癫痫发作的频率和持续时间。此外,G6PD过表达增强信号转导和转录激活因子1(STAT1)的表达,从而影响N-甲基-d-天冬氨酸受体的表达,并随后影响癫痫发作活动。重要的是,G6PD对STAT1的调节似乎主要通过活性氧信号通路介导.总的来说,我们的发现强调了G6PD在调节癫痫发生中的关键作用,并提示其作为癫痫治疗靶点的潜力。
    The pathogenesis of epilepsy remains unclear; however, a prevailing hypothesis suggests that the primary underlying cause is an imbalance between neuronal excitability and inhibition. Glucose-6-phosphate dehydrogenase (G6PD) is a key enzyme in the pentose phosphate pathway, which is primarily involved in deoxynucleic acid synthesis and antioxidant defense mechanisms and exhibits increased expression during the chronic phase of epilepsy, predominantly colocalizing with neurons. G6PD overexpression significantly reduces the frequency and duration of spontaneous recurrent seizures. Furthermore, G6PD overexpression enhances signal transducer and activator of transcription 1 (STAT1) expression, thus influencing N-methyl-d-aspartic acid receptors expression, and subsequently affecting seizure activity. Importantly, the regulation of STAT1 by G6PD appears to be mediated primarily through reactive oxygen species signaling pathways. Collectively, our findings highlight the pivotal role of G6PD in modulating epileptogenesis, and suggest its potential as a therapeutic target for epilepsy.
    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
    猪deltacoronavirus(PDCoV)是一种肠道致病性冠状病毒,可引起仔猪急性和严重的水样腹泻,并具有跨物种传播的能力,对养猪生产和公众健康造成极大威胁。干扰素(IFN)介导的信号转导是病毒与宿主相互作用的重要组成部分,在调节病毒感染中起着至关重要的作用。以前的研究表明,冠状病毒编码的多功能病毒蛋白通过各种方式拮抗IFN的产生。然而,这些病毒蛋白在调节IFN介导的信号通路中的功能在很大程度上是未知的。在这项研究中,我们证明PDCoV及其编码的核衣壳(N)蛋白拮抗I型IFN介导的JAK-STAT信号通路。我们确定PDCoV感染刺激但延迟了IFN刺激基因(ISG)的产生。此外,PDCoV通过靶向STAT1和ISGF3形成的核易位抑制JAK-STAT信号转导。进一步的证据表明,PDCoVN是通过经由其C末端结构域靶向STAT1核易位而参与抑制I型IFN信号传导的必需蛋白。机械上,PDCoVN通过与STAT1相互作用并随后抑制其核易位来靶向STAT1。此外,PDCoVN通过溶酶体途径特异性靶向KPNA2降解抑制STAT1核易位,从而抑制JAK-STAT信号通路下游传感器的激活。一起来看,我们的结果揭示了PDCoVN干扰宿主抗病毒反应的新机制。IMPORTANCEP猪deltacronovirus(PDCoV)是一种新型肠致病性冠状病毒,日益受到关注,严重威胁养猪业和公众健康。了解PDCoV在感染过程中逃避宿主防御的潜在机制对于开发针对PDCoV的靶向药物和有效疫苗至关重要。这项研究表明,PDCoV及其编码的核衣壳(N)蛋白通过靶向STAT1拮抗I型干扰素信号,STAT1是JAK-STAT信号通路中的关键信号传感器。进一步的实验表明,PDCoVN介导的STAT1核易位抑制涉及KPNA2的降解,溶酶体在KPNA2降解中起作用。这项研究为PDCoVN在JAK-STAT信号通路中的调控提供了新的见解,并揭示了PDCoV逃避宿主抗病毒反应的新机制。新发现可能指导我们发现新的治疗靶点并开发用于PDCoV感染的减毒活疫苗。
    Porcine deltacoronavirus (PDCoV) is an enteric pathogenic coronavirus that causes acute and severe watery diarrhea in piglets and has the ability of cross-species transmission, posing a great threat to swine production and public health. The interferon (IFN)-mediated signal transduction represents an important component of virus-host interactions and plays an essential role in regulating viral infection. Previous studies have suggested that multifunctional viral proteins encoded by coronaviruses antagonize the production of IFN via various means. However, the function of these viral proteins in regulating IFN-mediated signaling pathways is largely unknown. In this study, we demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I IFN-mediated JAK-STAT signaling pathway. We identified that PDCoV infection stimulated but delayed the production of IFN-stimulated genes (ISGs). In addition, PDCoV inhibited JAK-STAT signal transduction by targeting the nuclear translocation of STAT1 and ISGF3 formation. Further evidence showed that PDCoV N is the essential protein involved in the inhibition of type I IFN signaling by targeting STAT1 nuclear translocation via its C-terminal domain. Mechanistically, PDCoV N targets STAT1 by interacting with it and subsequently inhibiting its nuclear translocation. Furthermore, PDCoV N inhibits STAT1 nuclear translocation by specifically targeting KPNA2 degradation through the lysosomal pathway, thereby inhibiting the activation of downstream sensors in the JAK-STAT signaling pathway. Taken together, our results reveal a novel mechanism by which PDCoV N interferes with the host antiviral response.IMPORTANCEPorcine deltacoronavirus (PDCoV) is a novel enteropathogenic coronavirus that receives increased attention and seriously threatens the pig industry and public health. Understanding the underlying mechanism of PDCoV evading the host defense during infection is essential for developing targeted drugs and effective vaccines against PDCoV. This study demonstrated that PDCoV and its encoded nucleocapsid (N) protein antagonize type I interferon signaling by targeting STAT1, which is a crucial signal sensor in the JAK-STAT signaling pathway. Further experiments suggested that PDCoV N-mediated inhibition of the STAT1 nuclear translocation involves the degradation of KPNA2, and the lysosome plays a role in KPNA2 degradation. This study provides new insights into the regulation of PDCoV N in the JAK-STAT signaling pathway and reveals a novel mechanism by which PDCoV evades the host antiviral response. The novel findings may guide us to discover new therapeutic targets and develop live attenuated vaccines for PDCoV infection.
    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:肝内胆管癌(ICCA)是一类异质性恶性肿瘤,其特点是复发率高,预后差。异染色质蛋白1α(HP1α)是通过异染色质形成和结构维持参与转录沉默的最重要的非组蛋白染色体蛋白之一。HP1α对ICCA进展的影响尚不清楚。
    方法:通过在两种细胞系和两种ICCA小鼠模型中的实验来检测对ICCA增殖的影响。使用电喷雾电离质谱(ESI-MS)确定HP1α与组蛋白脱乙酰酶1(HDAC1)之间的相互作用,并使用免疫沉淀测定(co-IP)研究结合机制。通过RNA测序(RNA-seq)筛选出目标基因。通过生物信息学方法预测DNA结合蛋白和组蛋白修饰的占用,并通过靶标和标签下的裂解(CUT&Tag)和染色质免疫沉淀(ChIP)进行评估。
    结果:HP1α在肝内胆管癌(ICCA)组织中上调,并通过抑制干扰素途径,以信号转导和转录激活因子1(STAT1)依赖性方式调节ICCA细胞的增殖。机械上,STAT1由HP1α-HDAC1复合物直接和表观遗传地通过启动子结合和不同组蛋白修饰的变化进行转录调节,通过高通量测序验证。广谱HDAC抑制剂(HDACi)通过下调HP1α和靶向异源二聚体激活干扰素途径并抑制ICCA细胞的增殖。发现广谱HDACi加干扰素制备方案可改善体内和体外的抗增殖作用并延迟ICCA的发展,它利用了基础激活以及干扰素途径的直接激活。HP1α参与介导对两种试剂的细胞抗性。
    结论:HP1α-HDAC1复合物通过在转录水平上直接和表观调节STAT1影响干扰素途径的激活。广谱HDACi加干扰素制剂方案抑制ICCA发展,为ICCA治疗提供可行的策略。靶向HP1α-HDAC1-STAT1轴是治疗ICCA的可能策略,尤其是HP1α阳性病例。
    BACKGROUND: Intrahepatic cholangiocarcinoma (ICCA) is a heterogeneous group of malignant tumors characterized by high recurrence rate and poor prognosis. Heterochromatin Protein 1α (HP1α) is one of the most important nonhistone chromosomal proteins involved in transcriptional silencing via heterochromatin formation and structural maintenance. The effect of HP1α on the progression of ICCA remained unclear.
    METHODS: The effect on the proliferation of ICCA was detected by experiments in two cell lines and two ICCA mouse models. The interaction between HP1α and Histone Deacetylase 1 (HDAC1) was determined using Electrospray Ionization Mass Spectrometry (ESI-MS) and the binding mechanism was studied using immunoprecipitation assays (co-IP). The target gene was screened out by RNA sequencing (RNA-seq). The occupation of DNA binding proteins and histone modifications were predicted by bioinformatic methods and evaluated by Cleavage Under Targets and Tagmentation (CUT & Tag) and Chromatin immunoprecipitation (ChIP).
    RESULTS: HP1α was upregulated in intrahepatic cholangiocarcinoma (ICCA) tissues and regulated the proliferation of ICCA cells by inhibiting the interferon pathway in a Signal Transducer and Activator of Transcription 1 (STAT1)-dependent manner. Mechanistically, STAT1 is transcriptionally regulated by the HP1α-HDAC1 complex directly and epigenetically via promoter binding and changes in different histone modifications, as validated by high-throughput sequencing. Broad-spectrum HDAC inhibitor (HDACi) activates the interferon pathway and inhibits the proliferation of ICCA cells by downregulating HP1α and targeting the heterodimer. Broad-spectrum HDACi plus interferon preparation regimen was found to improve the antiproliferative effects and delay ICCA development in vivo and in vitro, which took advantage of basal activation as well as direct activation of the interferon pathway. HP1α participates in mediating the cellular resistance to both agents.
    CONCLUSIONS: HP1α-HDAC1 complex influences interferon pathway activation by directly and epigenetically regulating STAT1 in transcriptional level. The broad-spectrum HDACi plus interferon preparation regimen inhibits ICCA development, providing feasible strategies for ICCA treatment. Targeting the HP1α-HDAC1-STAT1 axis is a possible strategy for treating ICCA, especially HP1α-positive cases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号