Interferons

干扰素
  • 文章类型: Journal Article
    胶质瘤,一种脑肿瘤,由于其异质性和有限的治疗选择,提出了重大挑战。干扰素相关基因(IRGs)已成为神经胶质瘤发病机制的潜在参与者,然而,它们的表达模式和临床意义仍有待完全阐明。我们进行了全面分析,以研究IRGs在胶质瘤中的表达模式和功能富集。这涉及构建蛋白质-蛋白质相互作用网络,热图分析,生存曲线绘图,诊断和预后评估,神经胶质瘤亚组差异表达分析,GSVA,免疫浸润分析,和药物敏感性分析。我们的分析揭示了神经胶质瘤中IRGs的不同表达模式和功能富集。值得注意的是,与正常组织相比,胶质瘤组织中IFNW1和IFNA21显著下调,更高的表达水平与总生存率和疾病特异性生存率的提高相关。此外,这些基因在区分神经胶质瘤组织和正常组织方面显示出诊断能力,并且在更高级别和更侵袭性的神经胶质瘤中显著下调.神经胶质瘤亚组的差异表达分析强调了IFNW1和IFNA21表达与关键通路和生物学过程的关联。包括代谢重编程和免疫调节。免疫浸润分析揭示了它们对肿瘤微环境中免疫细胞组成的影响。此外,表达水平升高与化疗药物耐药性增加相关.我们的发现强调了IFNW1和IFNA21作为神经胶质瘤诊断生物标志物和预后指标的潜力。它们在调节神经胶质瘤进展中的作用,免疫反应,药物敏感性凸显了它们作为潜在治疗靶点的重要性。这些结果有助于更深入地了解神经胶质瘤生物学,并可能为神经胶质瘤患者制定个性化治疗策略提供信息。
    Glioma, a type of brain tumor, poses significant challenges due to its heterogeneous nature and limited treatment options. Interferon-related genes (IRGs) have emerged as potential players in glioma pathogenesis, yet their expression patterns and clinical implications remain to be fully elucidated. We conducted a comprehensive analysis to investigate the expression patterns and functional enrichment of IRGs in glioma. This involved constructing protein-protein interaction networks, heatmap analysis, survival curve plotting, diagnostic and prognostic assessments, differential expression analysis across glioma subgroups, GSVA, immune infiltration analysis, and drug sensitivity analysis. Our analysis revealed distinct expression patterns and functional enrichment of IRGs in glioma. Notably, IFNW1 and IFNA21 were markedly downregulated in glioma tissues compared to normal tissues, and higher expression levels were associated with improved overall survival and disease-specific survival. Furthermore, these genes showed diagnostic capabilities in distinguishing glioma tissues from normal tissues and were significantly downregulated in higher-grade and more aggressive gliomas. Differential expression analysis across glioma subgroups highlighted the association of IFNW1 and IFNA21 expression with key pathways and biological processes, including metabolic reprogramming and immune regulation. Immune infiltration analysis revealed their influence on immune cell composition in the tumor microenvironment. Additionally, elevated expression levels were associated with increased resistance to chemotherapeutic agents. Our findings underscore the potential of IFNW1 and IFNA21 as diagnostic biomarkers and prognostic indicators in glioma. Their roles in modulating glioma progression, immune response, and drug sensitivity highlight their significance as potential therapeutic targets. These results contribute to a deeper understanding of glioma biology and may inform the development of personalized treatment strategies for glioma patients.
    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
    Pulmonary aspergillosis is a serious pulmonary fungal infectious disease. It is difficult to manage and has limited treatment options. Existing anti-aspergillus medications have high rates of treatment failure and increased drug resistance, making it difficult to meet the clinical requirements. Therefore, the development of new, effective treatment programs is critical. According to research, interferons play an important role in the body\'s immune response to bacterial and viral infectious diseases. Inadequate interferon expression or dysfunction can put the body at risk for certain infectious diseases. Interferon has been used in clinical trials to prevent or treat infectious diseases. In recent years, researchers have focused on the immunological role of interferon in Aspergillus infections and its potential for clinical application. This review summarized the most recent advances in the immunoregulatory mechanisms of interferon and its clinical application in Aspergillus infections.
    肺曲霉病是一种严重的肺部真菌感染性疾病,临床诊治困难,现有抗曲霉药物存在治疗失败率高、药物耐药率增加等问题,无法满足临床需求。因此开发新的治疗方案是非常必要的。研究表明,干扰素广泛参与细菌和病毒等感染性疾病的免疫反应。干扰素表达不足或功能缺陷常常导致机体易发生感染性疾病。已有临床研究探索了干扰素在感染性疾病中作为预防或辅助治疗的潜在疗效。近年来,研究者们逐渐开始关注干扰素在曲霉感染中的免疫作用及其在临床应用中的潜力。本综述将总结曲霉感染中干扰素免疫作用及其临床应用的最新研究进展。.
    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
    STING中的功能增益突变导致STING相关的血管病变,在婴儿期(SAVI)发病,其特征是早发性全身性炎症,皮肤血管病变,和间质性肺病.这里,我们报告并描述了在SAVI患者中鉴定的新型STING变体(F269S)。患者骨髓的单细胞转录组学揭示了跨细胞类型的干扰素(IFN)和炎症途径的自发激活,并且观察到循环幼稚T细胞的惊人流行。诱导型STINGF269S表达通过蛋白质的配体非依赖性转运至高尔基体,保护细胞免受病毒感染,但阻止其有效的免疫启动。此外,SAVI免疫细胞分泌细胞因子可促进并进一步加剧内皮细胞活化,导致炎症和内皮损伤。我们的研究发现STINGF269S突变是导致SAVI的一种新的致病变异,强调内皮细胞和免疫细胞之间的串扰在肺部疾病中的重要性,并有助于更好地理解异常STING激活如何引起病理。
    Gain-of-function mutations in STING cause STING-associated vasculopathy with onset in infancy (SAVI) characterized by early-onset systemic inflammation, skin vasculopathy, and interstitial lung disease. Here, we report and characterize a novel STING variant (F269S) identified in a SAVI patient. Single-cell transcriptomics of patient bone marrow revealed spontaneous activation of interferon (IFN) and inflammatory pathways across cell types and a striking prevalence of circulating naïve T cells was observed. Inducible STING F269S expression conferred enhanced signaling through ligand-independent translocation of the protein to the Golgi, protecting cells from viral infections but preventing their efficient immune priming. Additionally, endothelial cell activation was promoted and further exacerbated by cytokine secretion by SAVI immune cells, resulting in inflammation and endothelial damage. Our findings identify STING F269S mutation as a novel pathogenic variant causing SAVI, highlight the importance of the crosstalk between endothelial and immune cells in the context of lung disease, and contribute to a better understanding of how aberrant STING activation can cause pathology.
    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
    候鸟是病毒传播的重要媒介,候鸟如何识别病毒和病毒在鸟类中持续存在仍然是个谜。作为候鸟中水禽的动物模型,研究和解剖鸭细胞中的抗病毒免疫和病毒逃避可能为破译这些难题铺平道路。这里,我们研究了鸭STING介导的DEF细胞抗病毒自噬的机制。结果发现,鸭STING可以显着提高LC3B-II/I的营业额,LC3B-EGFP斑点形成,和mCherry/EGFP比率,说明鸭STING可以诱导自噬。鸭STING诱导的自噬不受shRNA敲低ATG5表达的影响,删除STING的C末端尾部,或TBK1抑制剂BX795治疗,说明鸭STING激活的非经典选择性自噬与TBK1、TBK1磷酸化无关,和干扰素(IFN)信号。STINGR235A突变体和Sar1A/B激酶突变体消除了鸭STING诱导的自噬,提示与cGAMP和COPII复合物介导的转运结合是关键的先决条件。鸭STING通过LIR基序与LC3B相互作用诱导自噬,鸭STING的LIR4/7基序突变体消除了与LC3B的相互作用,既不激活自噬也不表达IFN,表明鸭STING与LC3B定向自噬相关,并决定先天免疫激活。最后,我们发现鸭STING介导的自噬通过广泛降解的病毒蛋白显着抑制鸭瘟病毒(DPV)的感染。我们的研究可能会揭示有关控制和逃避候鸟传播的疾病的一种情况。
    Migratory birds are important vectors for virus transmission, how migratory birds recognize viruses and viruses are sustained in birds is still enigmatic. As an animal model for waterfowl among migratory birds, studying and dissecting the antiviral immunity and viral evasion in duck cells may pave a path to deciphering these puzzles. Here, we studied the mechanism of antiviral autophagy mediated by duck STING in DEF cells. The results collaborated that duck STING could significantly enhance LC3B-II/I turnover, LC3B-EGFP puncta formation, and mCherry/EGFP ratio, indicating that duck STING could induce autophagy. The autophagy induced by duck STING is not affected by shRNA knockdown of ATG5 expression, deletion of the C-terminal tail of STING, or TBK1 inhibitor BX795 treatment, indicating that duck STING activated non-classical selective autophagy is independent of interaction with TBK1, TBK1 phosphorylation, and interferon (IFN) signaling. The STING R235A mutant and Sar1A/B kinase mutant abolished duck STING induced autophagy, suggesting binding with cGAMP and COPII complex mediated transport are the critical prerequisite. Duck STING interacted with LC3B through LIR motifs to induce autophagy, the LIR 4/7 motif mutants of duck STING abolished the interaction with LC3B, and neither activated autophagy nor IFN expression, indicating that duck STING associates with LC3B directed autophagy and dictated innate immunity activation. Finally, we found that duck STING mediated autophagy significantly inhibited duck plague virus (DPV) infection via ubiquitously degraded viral proteins. Our study may shed light on one scenario about the control and evasion of diseases transmitted by migratory birds.
    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
    SARS-CoV-2诱导I/III型干扰素产生延迟,让它逃避早期的先天免疫反应。Thedelayhasbeenattributedtoadeficiencyintheabilityofcellstosensevirusreplacationuponinfection,这反过来又阻碍了旁观者细胞中抗病毒状态的激活。这里,我们引入了一个细胞自动机模型来研究病毒感染的时空传播与病毒和宿主相关参数的关系.该模型表明,SARS-CoV-2感染中相当大的人与人之间的异质性是对接近临界阈值的生物学参数的轻微变化的高度敏感性的结果。它进一步表明,宿主内病毒增殖可以通过存在很少的用于IFN产生的细胞来减少。因此,观察到的细胞防御准备状态的异质性反映了一种成本效益高的保护策略。
    SARS-CoV-2 induces delayed type-I/III interferon production, allowing it to escape the early innate immune response. The delay has been attributed to a deficiency in the ability of cells to sense viral replication upon infection, which in turn hampers activation of the antiviral state in bystander cells. Here, we introduce a cellular automaton model to investigate the spatiotemporal spreading of viral infection as a function of virus and host-dependent parameters. The model suggests that the considerable person-to-person heterogeneity in SARS-CoV-2 infections is a consequence of high sensitivity to slight variations in biological parameters near a critical threshold. It further suggests that within-host viral proliferation can be curtailed by the presence of remarkably few cells that are primed for IFN production. Thus, the observed heterogeneity in defense readiness of cells reflects a remarkably cost-efficient strategy for protection.
    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
    III型干扰素信号传导有助于气道中重要的人类病原体金黄色葡萄球菌的发病。对这种反应中重要的细胞因子知之甚少。使用Ifnl2-绿色荧光蛋白报告小鼠结合流式细胞术和细胞耗竭策略,我们证明,肺泡巨噬细胞是响应呼吸道金黄色葡萄球菌的干扰素λ(IFN-λ)的主要生产者。骨髓嵌合体显示IFN-λ受体(IFNLR1)缺陷受体小鼠的细菌负荷降低,表明非造血细胞对发病机制很重要,除了显著减少肺部炎症。通过使用气道上皮特异性IFNLR敲除小鼠证实了这些观察结果。我们的数据表明,进入气道后,金黄色葡萄球菌激活肺泡巨噬细胞以产生III型IFN,其随后被气道上皮感知。未来的步骤将确定来自上皮的信号如何对细菌清除产生影响。这些结果突出了重要的,但有时是有害的,III型IFN信号传导在感染过程中的作用以及气道上皮在宿主-病原体相互作用过程中的影响。重要意义III型干扰素信号对控制细菌感染的贡献在很大程度上是未知的。我们以前已经证明,它有助于急性金黄色葡萄球菌呼吸道感染的发病机理。在这份报告中,我们记录了支持这种发病机制的两种细胞类型的重要性。我们证明肺泡巨噬细胞是负责产生III型干扰素的细胞,并且该分子被气道上皮细胞感知,影响细菌清除和炎症的诱导。这项工作揭示了这一重要致病级联的前两个方面。
    Type III interferon signaling contributes to the pathogenesis of the important human pathogen Staphylococcus aureus in the airway. Little is known of the cellular factors important in this response. Using Ifnl2-green fluorescent protein reporter mice combined with flow cytometry and cellular depletion strategies, we demonstrate that the alveolar macrophage is the primary producer of interferon lambda (IFN-λ) in response to S. aureus in the airway. Bone marrow chimeras showed reduced bacterial burden in IFN-λ receptor (IFNLR1)-deficient recipient mice, indicative that non-hematopoietic cells were important for pathogenesis, in addition to significant reductions in pulmonary inflammation. These observations were confirmed through the use of an airway epithelial-specific IFNLR knockout mouse. Our data suggest that upon entry to the airway, S. aureus activates alveolar macrophages to produce type III IFN that is subsequently sensed by the airway epithelium. Future steps will determine how signaling from the epithelium then exerts its influence on bacterial clearance. These results highlight the important, yet sometimes detrimental, role of type III IFN signaling during infection and the impact the airway epithelium plays during host-pathogen interactions.IMPORTANCEThe contribution of type III interferon signaling to the control of bacterial infections is largely unknown. We have previously demonstrated that it contributes to the pathogenesis of acute Staphylococcus aureus respiratory infection. In this report, we document the importance of two cell types that underpin this pathogenesis. We demonstrate that the alveolar macrophage is the cell that is responsible for the production of type III interferon and that this molecule is sensed by airway epithelial cells, which impacts both bacterial clearance and induction of inflammation. This work sheds light on the first two aspects of this important pathogenic cascade.
    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:三方基序(TRIM)家族包括80多个不同的人类基因。它们的功能与调节重要的细胞过程有关,包括细胞内信号,转录,自噬,和先天免疫。在病毒感染期间,巨噬细胞是产生干扰素(IFN)和IL27的先天免疫的关键成分。我们最近发表了IL27和IFN诱导各种基因的转录变化,包括那些参与JAK-STAT信号。此外,IL27和IFN在单核细胞衍生的巨噬细胞(MDM)中共享促炎和抗病毒途径,导致炎症因子和编码抗病毒蛋白的IFN刺激基因(ISGs)的共同和独特表达。有趣的是,近年来许多TRIM蛋白被认为是ISGs。尽管已经很好地描述了TRIM表达是由IFN诱导的,目前尚不完全清楚TRIM基因是否在巨噬细胞中被IL27诱导。因此,在这项研究中,我们检查了IL27和I型刺激的效果,II,和IIIIFNs对MDMs中TRIM基因mRNA表达谱的影响。
    方法:我们使用大量RNA-seq来检查用IFNs或IL27处理的MDMs的TRIM表达谱。最初,我们使用热图表征了不同TRIM亚家族的表达模式。随后,火山图用于鉴定常见差异表达的TRIM基因。此外,我们用ClueGO进行了基因本体论分析,以探索受调控的TRIMs的生物学过程,使用GeneMANIA创建了一个基因-基因相互作用网络,并使用STRING数据库检查蛋白质-蛋白质相互作用。最后,使用RT-qPCR验证RNA-seq数据。此外,还评估了IL27对Mayaro病毒复制的影响.
    结果:我们发现IL27与IFNs相似,上调几种TRIM基因在人巨噬细胞中的表达。具体来说,我们确定了由IL27和所有类型的人IFN诱导的三种常见TRIM基因(TRIM19,21和22).此外,我们首次报道了TRIM19,21,22和69基因响应IL27的转录调控.TRIM涉及广泛的生物过程,包括对病毒的防御反应,病毒生命周期调节,和病毒过程的负调控。此外,我们观察到之前接受IL27治疗的MDM中Mayaro病毒复制减少.
    结论:我们的结果表明,IL27与IFNs一样,调节参与诱导先天免疫和抗病毒反应的不同TRIM家族成员的转录表达。此外,功能分析表明,像IFN一样,IL27减少了Mayaro病毒在MDM中的复制。这意味着IL27和IFN在功能水平上具有许多相似性。此外,确定不同的TRIM组及其响应IL27的差异表达为人类巨噬细胞抗病毒反应的调节机制提供了新的见解。
    BACKGROUND: The Tripartite motif (TRIM) family includes more than 80 distinct human genes. Their function has been implicated in regulating important cellular processes, including intracellular signaling, transcription, autophagy, and innate immunity. During viral infections, macrophages are key components of innate immunity that produce interferons (IFNs) and IL27. We recently published that IL27 and IFNs induce transcriptional changes in various genes, including those involved in JAK-STAT signaling. Furthermore, IL27 and IFNs share proinflammatory and antiviral pathways in monocyte-derived macrophages (MDMs), resulting in both common and unique expression of inflammatory factors and IFN-stimulated genes (ISGs) encoding antiviral proteins. Interestingly, many TRIM proteins have been recognized as ISGs in recent years. Although it is already very well described that TRIM expression is induced by IFNs, it is not fully understood whether TRIM genes are induced in macrophages by IL27. Therefore, in this study, we examined the effect of stimulation with IL27 and type I, II, and III IFNs on the mRNA expression profiles of TRIM genes in MDMs.
    METHODS: We used bulk RNA-seq to examine the TRIM expression profile of MDMs treated with IFNs or IL27. Initially, we characterized the expression patterns of different TRIM subfamilies using a heatmap. Subsequently, a volcano plot was employed to identify commonly differentially expressed TRIM genes. Additionally, we conducted gene ontology analysis with ClueGO to explore the biological processes of the regulated TRIMs, created a gene-gene interaction network using GeneMANIA, and examined protein-protein interactions with the STRING database. Finally, RNA-seq data was validated using RT-qPCR. Furthermore, the effect of IL27 on Mayaro virus replication was also evaluated.
    RESULTS: We found that IL27, similar to IFNs, upregulates several TRIM genes\' expression in human macrophages. Specifically, we identified three common TRIM genes (TRIM19, 21, and 22) induced by IL27 and all types of human IFNs. Additionally, we performed the first report of transcriptional regulation of TRIM19, 21, 22, and 69 genes in response to IL27. The TRIMs involved a broad range of biological processes, including defense response to viruses, viral life cycle regulation, and negative regulation of viral processes. In addition, we observed a decrease in Mayaro virus replication in MDMs previously treated with IL27.
    CONCLUSIONS: Our results show that IL27, like IFNs, modulates the transcriptional expression of different TRIM-family members involved in the induction of innate immunity and an antiviral response. In addition, the functional analysis demonstrated that, like IFN, IL27 reduced Mayaro virus replication in MDMs. This implies that IL27 and IFNs share many similarities at a functional level. Moreover, identifying distinct TRIM groups and their differential expressions in response to IL27 provides new insights into the regulatory mechanisms underlying the antiviral response in human macrophages.
    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
    猪血凝性脑脊髓炎病毒(PHEV)在猪的上呼吸道和扁桃体中复制。使用气-液界面猪呼吸道上皮细胞(ALI-PRECs)培养系统,我们证明了PHEV通过损害纤毛功能和诱导抗病毒来破坏呼吸道上皮稳态,促炎细胞因子,和趋化因子反应。本研究通过宿主转录组分析探讨了PHEV感染期间驱动早期先天免疫反应的机制。在接种后24、36和48小时(hpi)从ALI-PREC收集总RNA。使用IlluminaHiseq600进行RNA-seq分析以产生100bp的双端读段。使用DeSeq2分析差异基因表达。PHEV在ALI-PREC中积极复制,引起细胞病变和进行性粘膜纤毛破坏。转录组分析显示纤毛相关基因如CILK1、DNAH11、LRRC-23、-49和-51以及酸性唾液酸粘蛋白CD164L2的下调。PHEV还激活抗病毒信号通路,显着增加干扰素刺激基因的表达(RSAD2,MX1,IFIT,和ISG15)和趋化因子基因(CCL5和CXCL10),强调炎症调节。这项研究有助于阐明气道上皮对PHEV感染的先天性免疫应答的分子机制。强调粘膜纤毛的关键作用,干扰素,和趋化因子反应。
    Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.
    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-Is)通过引发IFN刺激基因(ISGs)的表达,在人类免疫缺陷病毒I(HIV-1)的先天免疫中至关重要。其中包括有效的宿主限制因子。虽然ISGs通过靶向病毒生命周期的各个阶段来限制宿主细胞内的病毒复制,鲜为人知的IFN抑制基因(IRepGs),包括RNA结合蛋白(RBPs),通过改变对有效的HIV-1基因表达至关重要的宿主依赖性因子的表达来影响病毒复制。宿主限制和依赖性因素决定了病毒复制效率;然而,目前,对与HIV-1感染有关的IRepGs的了解仍然非常有限.这篇综述提供了关于RNA结合蛋白家族影响的当前理解的全面概述。特别是剪接相关蛋白SRSF和hnRNP的两个家族,HIV-1基因表达和病毒复制。由于最近的发现特别表明SRSF1和hnRNPA0在各种细胞系和原代细胞中受到IFN-I的调节,包括肠固有层单核细胞(LPMC)和外周血单核细胞(PBMC),我们特别讨论了它们在先天免疫影响HIV-1复制的背景下的作用.
    Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.
    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
    先天免疫系统,特别是干扰素(IFN)系统,构成抵御病毒感染的第一道防线。IFN信号诱导干扰素刺激基因(ISGs)的表达,和他们的产品经常限制病毒感染。逆转录病毒如人免疫缺陷病毒和人嗜T淋巴细胞病毒会引起严重的人类疾病,并被ISG编码的蛋白质靶向。这里,我们讨论了抑制逆转录病毒mRNA翻译的ISGs,从而抑制逆转录病毒繁殖。Schlafen蛋白降解翻译所需的细胞tRNA和rRNA。锌指抗病毒蛋白和RNA激活蛋白激酶抑制翻译起始因子,和Shiftless抑制逆转录病毒酶表达所必需的翻译重新编码。我们概述了多功能ISGs抗病毒活性的共同机制,并根据这些ISGs的作用方式讨论了潜在的抗逆转录病毒治疗方法。
    The innate immune system, particularly the interferon (IFN) system, constitutes the initial line of defense against viral infections. IFN signaling induces the expression of interferon-stimulated genes (ISGs), and their products frequently restrict viral infection. Retroviruses like the human immunodeficiency viruses and the human T-lymphotropic viruses cause severe human diseases and are targeted by ISG-encoded proteins. Here, we discuss ISGs that inhibit the translation of retroviral mRNAs and thereby retrovirus propagation. The Schlafen proteins degrade cellular tRNAs and rRNAs needed for translation. Zinc Finger Antiviral Protein and RNA-activated protein kinase inhibit translation initiation factors, and Shiftless suppresses translation recoding essential for the expression of retroviral enzymes. We outline common mechanisms that underlie the antiviral activity of multifunctional ISGs and discuss potential antiretroviral therapeutic approaches based on the mode of action of these ISGs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号