IFN-λ

IFN - λ
  • 文章类型: Journal Article
    概述人类甲疱疹病毒1(HSV-1)是一种非常成功的嗜神经病原体,主要感染口腔粘膜衬里的上皮细胞。在口服的初级裂解复制后,眼,和鼻粘膜上皮细胞,HSV-1在三叉神经节内的神经元中建立了终生潜伏期。免疫系统受损的患者从潜伏期开始经历HSV-1的频繁再激活,导致病毒进入感觉神经元,然后在神经支配的粘膜上皮表面进行顺行运输和溶解复制。尽管角膜粘膜表面的反复感染很少见,它可以导致慢性免疫炎症状态称为疱疹性基质角膜炎(HSK)。HSK导致逐渐的视力丧失,并且在严重的未经治疗的病例中可导致永久性失明。目前,没有治愈或成功的疫苗来预防潜伏或复发性HSV-1感染,对管理HSK和预防视力丧失构成重大临床挑战。HSK的常规临床管理主要依靠抗病毒药物来抑制HSV-1的复制,抗炎药(如皮质类固醇),以缓解疼痛和炎症,和手术干预在更严重的情况下,以取代受损的角膜。然而,每种临床治疗策略都有局限性,如局部和全身药物毒性和抗病毒耐药HSV-1株的出现。在这次审查中,我们总结了HSK发病机制中涉及的因素和免疫细胞,并重点介绍了成功临床治疗HSK的替代治疗策略.我们还讨论了免疫调节细胞因子和免疫代谢调节剂作为针对新兴的抗病毒抗性HSV-1株的有希望的HSK疗法的治疗潜力。
    SUMMARYHuman alphaherpesvirus 1 (HSV-1) is a highly successful neurotropic pathogen that primarily infects the epithelial cells lining the orofacial mucosa. After primary lytic replication in the oral, ocular, and nasal mucosal epithelial cells, HSV-1 establishes life-long latency in neurons within the trigeminal ganglion. Patients with compromised immune systems experience frequent reactivation of HSV-1 from latency, leading to virus entry in the sensory neurons, followed by anterograde transport and lytic replication at the innervated mucosal epithelial surface. Although recurrent infection of the corneal mucosal surface is rare, it can result in a chronic immuno-inflammatory condition called herpetic stromal keratitis (HSK). HSK leads to gradual vision loss and can cause permanent blindness in severe untreated cases. Currently, there is no cure or successful vaccine to prevent latent or recurrent HSV-1 infections, posing a significant clinical challenge to managing HSK and preventing vision loss. The conventional clinical management of HSK primarily relies on anti-virals to suppress HSV-1 replication, anti-inflammatory drugs (such as corticosteroids) to provide symptomatic relief from pain and inflammation, and surgical interventions in more severe cases to replace damaged cornea. However, each clinical treatment strategy has limitations, such as local and systemic drug toxicities and the emergence of anti-viral-resistant HSV-1 strains. In this review, we summarize the factors and immune cells involved in HSK pathogenesis and highlight alternate therapeutic strategies for successful clinical management of HSK. We also discuss the therapeutic potential of immunoregulatory cytokines and immunometabolism modulators as promising HSK therapies against emerging anti-viral-resistant HSV-1 strains.
    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
    PGAP3是位于染色体17q12-21中的糖基磷脂酰肌醇(GPI)磷脂酶基因,该区域与哮喘高度相关。尽管人们对其他在支气管上皮中表达水平升高的染色体17q12-21基因如ORMDL3和GSDMB的功能了解很多,关于哮喘患者支气管上皮中PGAP3表达增加的功能知之甚少。因此,这项研究的目的是通过利用RNA测序和生物信息学分析来确定人支气管上皮细胞中PGAP3表达的增加是否调节了对哮喘发病机理重要的mRNA途径的表达。我们对用PGAP3转染24和48小时的正常人支气管上皮细胞进行RNA测序。PGAP3调节基因与哮喘和呼吸道病毒(甲型流感,鼻病毒,呼吸道合胞病毒)参考数据集,以鉴定PGAP3靶基因和途径。在哮喘参考数据集中发现了大约9%的上调的PGAP3诱导基因。在鼻病毒参考数据集中有41%,在甲型流感参考数据集中有33%,在呼吸道合胞病毒参考数据集中为3%。PGAP3显著上调与先天免疫应答相关的几种基因的表达和与哮喘恶化相关的呼吸道病毒的病毒特征。PGAP3诱导表达量最高的两个基因是RSAD2、OASL、和IFN-λ,与哮喘相关的抗病毒基因。PGAP3还上调抗病毒基因BST2,其与PGAP3一样是GPI锚定蛋白。我们得出结论,人支气管上皮细胞中的PGAP3表达调节已知与哮喘相关的基因的表达,并且还调节与呼吸道病毒引发的哮喘加重的发病机制有关的基因的支气管上皮表达。
    PGAP3 is a glycosylphosphatidylinositol (GPI) phospholipase gene localized within chromosome 17q12-21, a region highly linked to asthma. Although much is known about the function of other chromosome 17q12-21 genes expressed at increased levels in bronchial epithelium such as ORMDL3 and GSDMB, little is known about the function of increased PGAP3 expression in bronchial epithelium in the context of asthma. The aim of this study was therefore to determine whether increased PGAP3 expression in human bronchial epithelial cells regulated expression of mRNA pathways important to the pathogenesis of asthma by utilizing RNA-sequencing and bioinformatic analysis. We performed RNA-sequencing on normal human bronchial epithelial cells transfected with PGAP3 for 24 and 48 hours. PGAP3 regulated genes were compared to asthma and respiratory virus (influenza A, rhinovirus, respiratory syncytial virus) reference data sets to identify PGAP3 target genes and pathways. Approximately 9% of the upregulated PGAP3-induced genes were found in an asthma reference data set, 41% in a rhinovirus reference data set, 33% in an influenza A reference data set, and 3% in a respiratory syncytial virus reference data set. PGAP3 significantly upregulated the expression of several genes associated with the innate immune response and viral signatures of respiratory viruses associated with asthma exacerbations. Two of the highest expressed genes induced by PGAP3 are RSAD2, OASL, and IFN-λ, which are anti-viral genes associated with asthma. PGAP3 also upregulated the antiviral gene BST2, which like PGAP3 is a GPI-anchored protein. We conclude that PGAP3 expression in human bronchial epithelial cells regulates expression of genes known to be linked to asthma, and also regulates the bronchial epithelial expression of genes pertinent to the pathogenesis of respiratory viral triggered asthma exacerbations.
    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
    猪急性腹泻综合征冠状病毒(SADS-CoV)是一种新发现的具有潜在跨种传播风险的猪冠状病毒。尽管SADS-CoV诱导的宿主细胞凋亡和先天免疫拮抗作用已被揭示,潜在的信号通路仍然不清楚。这里,我们证明SADS-CoV感染在体内和体外诱导细胞凋亡,病毒蛋白NS7a主要负责SADS-CoV诱导的宿主细胞凋亡。此外,我们发现在SADS-CoV感染的细胞中,NS7a与凋亡诱导因子线粒体相关因子1(AIFM1)相互作用,通过caspase-6激活caspase-3,和增强SADS-CoV复制。重要的是,NS7a通过激活caspase-3裂解干扰素调节因子3(IRF3)来抑制poly(I:C)诱导的III型干扰素(IFN-λ)表达,和caspase-3抑制剂在体内保护仔猪免受SADS-CoV感染。这些发现揭示了SADS-CoV如何诱导细胞凋亡以抑制先天免疫,并为开发有效的药物为临床控制SADS-CoV感染提供了有价值的线索。重要性在过去的20年里,多种动物来源的冠状病毒,包括严重急性呼吸道综合症冠状病毒(SARS-CoV),中东呼吸综合征冠状病毒(MERS-CoV),和SARS-CoV-2,已经造成数百万人死亡,严重危害人类健康,阻碍了社会发展,这表明研究具有跨物种传播潜力的动物源性冠状病毒尤为重要。蝙蝠起源的猪急性腹泻综合征冠状病毒(SADS-CoV),在2017年发现,不仅会导致仔猪致命的腹泻,还会感染多种人类细胞,具有跨物种传播的潜在风险,但其发病机制尚不清楚。在这项研究中,我们证明了SADS-CoV的NS7a通过凋亡诱导因子线粒体相关因子1(AIFM1)-caspase-6-caspase-3-干扰素调节因子3(IRF3)途径抑制IFN-λ的产生,caspase-3抑制剂(Z-DEVD-FMK)能有效抑制SADS-CoV复制,保护感染仔猪。我们在这项研究中的发现有助于更好地理解SADS-CoV-宿主相互作用作为冠状病毒发病机理的一部分,并将凋亡抑制剂用作预防和控制SADS-CoV感染的潜在治疗方法。
    Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a newly discovered swine coronavirus with potential cross-species transmission risk. Although SADS-CoV-induced host cell apoptosis and innate immunity antagonization has been revealed, underlying signaling pathways remain obscure. Here, we demonstrated that infection of SADS-CoV induced apoptosis in vivo and in vitro, and that viral protein NS7a is mainly responsible for SADS-CoV-induced apoptosis in host cells. Furthermore, we found that NS7a interacted with apoptosis-inducing factor mitochondria associated 1 (AIFM1) to activate caspase-3 via caspase-6 in SADS-CoV-infected cells, and enhanced SADS-CoV replication. Importantly, NS7a suppressed poly(I:C)-induced expression of type III interferon (IFN-λ) via activating caspase-3 to cleave interferon regulatory factor 3 (IRF3), and caspase-3 inhibitor protects piglets against SADS-CoV infection in vivo. These findings reveal how SADS-CoV induced apoptosis to inhibit innate immunity and provide a valuable clue to the development of effective drugs for the clinical control of SADS-CoV infection.IMPORTANCEOver the last 20 years, multiple animal-originated coronaviruses, including severe acute respiratory syndrome coronavirus (SARS-CoV), middle east respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2, have caused millions of deaths, seriously jeopardized human health, and hindered social development, indicating that the study of animal-originated coronaviruses with potential for cross-species transmission is particularly important. Bat-originated swine acute diarrhea syndrome coronavirus (SADS-CoV), discovered in 2017, can not only cause fatal diarrhea in piglets, but also infect multiple human cells, with a potential risk of cross-species transmission, but its pathogenesis is unclear. In this study, we demonstrated that NS7a of SADS-CoV suppresses IFN-λ production via apoptosis-inducing factor mitochondria associated 1 (AIFM1)-caspase-6-caspase-3-interferon regulatory factor 3 (IRF3) pathway, and caspase-3 inhibitor (Z-DEVD-FMK) can effectively inhibit SADS-CoV replication and protect infected piglets. Our findings in this study contribute to a better understanding of SADS-CoV-host interactions as a part of the coronaviruses pathogenesis and using apoptosis-inhibitor as a drug as potential therapeutic approaches for prevention and control of SADS-CoV 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
    猪急性腹泻综合征冠状病毒(SADS-CoV)是一种引起急性水样腹泻的新型猪肠道冠状病毒,呕吐,新生仔猪脱水。III型干扰素(IFN-λ)应答充当对抗在肠上皮细胞中复制的病毒的主要防御。然而,目前没有关于SADS-CoV如何调节IFN-λ产生的信息。在这项研究中,我们利用IPI-FX细胞(猪回肠上皮细胞系)作为体外模型来研究SADS-CoV针对IFN-λ应答的潜在免疫逃避策略.我们的结果表明,SADS-CoV感染抑制了聚(I:C)诱导的IFN-λ1的产生。通过筛选SADS-CoV编码的蛋白质,nsp1,nsp5,nsp10,nsp12,nsp16,E,S1和S2被鉴定为IFN-λ1产生的拮抗剂。具体来说,SADS-CoVnsp1阻碍了MAVS介导的IFN-λ1启动子的激活,TBK1,IKKε,IRF1SADS-CoV和nsp1均阻碍了聚(I:C)诱导的IRF1核易位。此外,SADS-CoVnsp1通过泛素介导的蛋白酶体途径降解IRF1而不与之相互作用。总的来说,我们的研究提供了SADS-CoV抑制III型IFN应答的第一个证据,揭示SADS-CoV逃避宿主免疫反应的分子机制。
    Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel porcine enteric coronavirus that causes acute watery diarrhea, vomiting, and dehydration in newborn piglets. The type III interferon (IFN-λ) response serves as the primary defense against viruses that replicate in intestinal epithelial cells. However, there is currently no information available on how SADS-CoV modulates the production of IFN-λ. In this study, we utilized IPI-FX cells (a cell line of porcine ileum epithelium) as an in vitro model to investigate the potential immune evasion strategies employed by SADS-CoV against the IFN-λ response. Our results showed that SADS-CoV infection suppressed the production of IFN-λ1 induced by poly(I:C). Through screening SADS-CoV-encoded proteins, nsp1, nsp5, nsp10, nsp12, nsp16, E, S1, and S2 were identified as antagonists of IFN-λ1 production. Specifically, SADS-CoV nsp1 impeded the activation of the IFN-λ1 promoter mediated by MAVS, TBK1, IKKε, and IRF1. Both SADS-CoV and nsp1 obstructed poly(I:C)-induced nuclear translocation of IRF1. Moreover, SADS-CoV nsp1 degraded IRF1 via the ubiquitin-mediated proteasome pathway without interacting with it. Overall, our study provides the first evidence that SADS-CoV inhibits the type III IFN response, shedding light on the molecular mechanisms employed by SADS-CoV to evade the host immune response.
    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
    肠道病毒D68(EV-D68)主要通过呼吸道传播,并在儿童和急性弛缓性脊髓炎(AFM)中引起呼吸道症状。III型干扰素(IFN)在抑制呼吸道上皮细胞中的病毒生长中起关键作用。然而,EV-D68诱导III型IFN产生的机制尚不完全清楚。在这项研究中,我们显示EV-D68感染刺激Calu-3细胞分泌IFN-λ。EV-D68病毒RNA(vRNA)的转染通过MDA5刺激IFN-λ。此外,我们的研究结果提供了证据,即EV-D68感染也诱导MDA5-IRF3/IRF7介导的IFN-λ。此外,我们发现EV-D68感染下调MDA5的表达。敲除MDA5增加了Calu-3细胞中的EV-D68复制。最后,我们证明了IFN-λ1和IFN-λ2/3蛋白有效抑制呼吸道上皮细胞中的EV-D68感染。总之,我们的研究表明,EV-D68通过激活的MDA5-IRF3/IRF7途径诱导III型IFN的产生,而III型IFN抑制Calu-3细胞中EV-D68的复制。
    Enterovirus D68 (EV-D68) primarily spreads through the respiratory tract and causes respiratory symptoms in children and acute flaccid myelitis (AFM). Type III interferons (IFNs) play a critical role in inhibiting viral growth in respiratory epithelial cells. However, the mechanism by which EV-D68 induces type III IFN production is not yet fully understood. In this study, we show that EV-D68 infection stimulates Calu-3 cells to secrete IFN-λ. The transfection of EV-D68 viral RNA (vRNA) stimulated IFN-λ via MDA5. Furthermore, our findings provide evidence that EV-D68 infection also induces MDA5-IRF3/IRF7-mediated IFN-λ. In addition, we discovered that EV-D68 infection downregulated MDA5 expression. Knockdown of MDA5 increased EV-D68 replication in Calu-3 cells. Finally, we demonstrated that the IFN-λ1 and IFN-λ2/3 proteins effectively inhibit EV-D68 infection in respiratory epithelial cells. In summary, our study shows that EV-D68 induces type III IFN production via the activated MDA5-IRF3/IRF7 pathway and that type III IFNs inhibit EV-D68 replication in Calu-3 cells.
    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
    乳杆菌是人和动物肠道中的主要微生物之一,乳杆菌对猪场仔猪病毒性腹泻有有效的应用。然而,在后生生物的抗感染研究中,乳酸菌活性成分的功能和具体的细胞单一途径尚不清楚。这里,我们比较了从干酪乳杆菌(L.casei)和使用CRISPER-Cas9技术对干酪乳杆菌进行基因编辑,具有抗氧化和抗炎的能力,EPS还可以抑制猪小肠上皮细胞-J2(IPEC-J2)内的ROS产生。有趣的是,我们发现EPS和基因组编辑干酪乳杆菌都可以特异性靶向IFN-λ在IPEC-J2中的表达,这有利于通过qRT-PCR和间接免疫荧光方法在病毒复制和生产中预防PEDV感染。最后,用EPS刺激STAT3细胞单通路转录IFN-λ,以阐明激活IL-10R2的III型IFN信号受体的详细机制,在PEDV感染中发挥抗炎和抗病毒的功能。一起来看,我们的研究将EPS的博士后与PEDV的抗病毒感染联系起来,这表明乳杆菌本身仍然显示出潜在的免疫调节活性,并强调了产生EPS的微生物的免疫调节潜力。
    Lacticaseibacillus is one of the predominant microorganisms in gut from human and animal, and the lacticaseibacillus have effective applications against the viral diarrhea of piglets in the farm. However, the function and the concrete cell single pathways of the active ingredient from lacticaseibacillus was not clear within anti-infection in the postbiotics research. Here, we compared the biological function of extracellular polysaccharides (EPS) purified from lacticaseibacillus casei (L. casei) and gene editing lacticaseibacillus casei with the CRISPER-Cas9 technology, which were with the ability of antioxidation and anti-inflammation, and the EPS could also inhibit the ROS production within the Porcine Small Intestinal Epithelial Cells-J2 (IPEC-J2). Interestingly, we found that both of EPS and genome editing lacticaseibacillus casei could specifically target the IFN-λ expression in the IPEC-J2, which was beneficial against the PEDV infection in the virus replication and production with the qRT-PCR and indirect immunofluorescence methods. Finally, the STAT3 cell single pathway was stimulated to transcribe IFN-λ with the EPS to elucidate the detailed mechanism of activating type III IFN signals receptor of IL-10R2, which play the function between anti-inflammation and anti-virus in the PEDV infection. Taken together, our research linked a postbiotics of EPS with the antiviral infection of PEDV, which suggest that the lacticaseibacillus itself still have displayed the potential immunomodulatory activities, and highlight the immunomodulatory potential of EPS-producing microbes.
    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
    干扰素(IFN)是分泌的细胞因子,能够激活IFN刺激的基因的表达,从而增加细胞对病毒感染的抵抗力。激活的转录因子与染色质重塑剂联合诱导重新编程IFN应答的表观遗传变化。出乎意料的是,8-氧鸟嘌呤DNA糖基化酶1(Ogg1)敲除小鼠显示增强的刺激驱动的IFN表达,其赋予对病毒和细菌感染和过敏原挑战的增加的抗性。这里,我们测试了DNA修复蛋白OGG1在调节IFN表达的启动子中识别8-氧鸟嘌呤(8-oxoGua)的假设。我们发现功能抑制,遗传消融,通过翻译后修饰OGG1而失活可显着增加人呼吸道合胞病毒(RSV)感染的上皮细胞中IFN-λ的表达。机械上,OGG1与干扰素反应元件附近的8-oxoGua结合,抑制IRF3/IRF7和NF-κB/RelADNA占据,同时促进抑制因子NF-κB1/p50-p50同二聚体与IFN-λ2/3启动子的结合。在RSV感染诱导的细支气管炎小鼠模型中,小分子抑制剂(TH5487)对OGG1的功能消融可增强IFN-λ的产生,降低免疫病理,嗜中性粒细胞增多症,并赋予抗病毒保护。这些发现表明,ROS通过其阅读器OGG1产生的表观遗传标记8-oxoGua在IFN-λ表达中充当稳态阈值因子。OGG1活性的药物靶向在调节抗病毒应答中可能具有临床效用。
    Interferons (IFNs) are secreted cytokines with the ability to activate expression of IFN stimulated genes that increase resistance of cells to virus infections. Activated transcription factors in conjunction with chromatin remodelers induce epigenetic changes that reprogram IFN responses. Unexpectedly, 8-oxoguanine DNA glycosylase1 (Ogg1) knockout mice show enhanced stimuli-driven IFN expression that confers increased resistance to viral and bacterial infections and allergen challenges. Here, we tested the hypothesis that the DNA repair protein OGG1 recognizes 8-oxoguanine (8-oxoGua) in promoters modulating IFN expression. We found that functional inhibition, genetic ablation, and inactivation by post-translational modification of OGG1 significantly augment IFN-λ expression in epithelial cells infected by human respiratory syncytial virus (RSV). Mechanistically, OGG1 bound to 8-oxoGua in proximity to interferon response elements, which inhibits the IRF3/IRF7 and NF-κB/RelA DNA occupancy, while promoting the suppressor NF-κB1/p50-p50 homodimer binding to the IFN-λ2/3 promoter. In a mouse model of bronchiolitis induced by RSV infection, functional ablation of OGG1 by a small molecule inhibitor (TH5487) enhances IFN-λ production, decreases immunopathology, neutrophilia, and confers antiviral protection. These findings suggest that the ROS-generated epigenetic mark 8-oxoGua via its reader OGG1 serves as a homeostatic thresholding factor in IFN-λ expression. Pharmaceutical targeting of OGG1 activity may have clinical utility in modulating antiviral response.
    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
    向全球人群接种疫苗剂量导致COVID-19发病率下降。然而,由于即使在接种疫苗的个体中,病例的严重程度也存在很大差异,因此受感染个体形成的临床表现仍然非常令人担忧。病理的临床进展以各种影响因素为特征,例如性别,年龄组,合并症,和个体的遗传学。对病毒感染的免疫反应可能受到个体遗传学的强烈影响;在涉及先天和适应性免疫反应的结构中的核苷酸变异称为单核苷酸多态性(SNP),例如干扰素(IFN)-λ。人类白细胞抗原(HLA),白细胞介素(IL)-6经常与病理进展有关。在这项研究中,我们对与COVID-19严重程度相关的这些结构的主要SNP进行了回顾.在国家生物技术和信息中心(NCBI)的一些平台上进行了搜索,根据纳入标准选择了102项研究进行全面阅读.IFN显示与抗病毒功能的强关联,具体来说,IFN-λ3(IL-28B)证明了在各种病理中通常与临床进展相关的遗传变体。对于COVID-19,rs12979860和rs1298275经常描述丙型肝炎和肝细胞癌病理状况的不利基因型。HLA的高遗传变异性在研究中被报道为与晚期免疫反应相关的关键因素,主要是由于其识别抗原的能力,HLA-B*46:01SNP与COVID-19易感性相关。对于IL-6,rs1554606与COVID-19的临床进展密切相关。此外,当与这种感染相关时,rs2069837被确定具有可能的宿主保护关系。
    The administration of vaccination doses to the global population has led to a decrease in the incidence of COVID-19. However, the clinical picture developed by infected individuals remains extremely concerning due to the great variability in the severity of cases even in vaccinated individuals. The clinical progression of the pathology is characterized by various influential factors such as sex, age group, comorbidities, and the genetics of the individual. The immune response to viral infections can be strongly influenced by the genetics of individuals; nucleotide variations called single-nucleotide polymorphisms (SNPs) in structures involved in the innate and adaptive immune response such as interferon (IFN)-λ, human leukocyte antigen (HLA), and interleukin (IL)-6 are frequently associated with pathological progression. In this study, we conducted a review of the main SNPs of these structures that are associated with severity in COVID-19. Searches were conducted on some platforms of the National Center for Biotechnology and Information (NCBI), and 102 studies were selected for full reading according to the inclusion criteria. IFNs showed a strong association with antiviral function, specifically, IFN-λ3 (IL-28B) demonstrated genetic variants commonly related to clinical progression in various pathologies. For COVID-19, rs12979860 and rs1298275 presented frequently described unfavorable genotypes for pathological conditions of hepatitis C and hepatocellular carcinoma. The high genetic variability of HLA was reported in the studies as a crucial factor relevant to the late immune response, mainly due to its ability to recognize antigens, with the HLA-B*46:01 SNP being associated with susceptibility to COVID-19. For IL-6, rs1554606 showed a strong relationship with the clinical progression of COVID-19. In addition, rs2069837 was identified with possible host protection relationships when linked to this 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
    先天免疫系统通过诱导各种干扰素(IFN)刺激的基因(ISG)来提供针对入侵病原体的防御。我们最近报道了三方基序蛋白25(TRIM25),一个重要的ISG,感染1型鸭病毒性甲型肝炎病毒(DHAV-1)后,鸭胚肝细胞(DEF)高度上调。然而,TRIM25上调的机制尚不清楚。这里我们报道了白细胞介素-22(IL-22),DHAV-1感染后,其在1日龄鸭的DEF和各种器官中的表达高度促进,高度增强IFN-λ诱导的TRIM25的产生。用IL-22中和抗体处理或IL-22过表达高度抑制或促进TRIM25表达,分别。信号转导和转录激活因子3(STAT3)的磷酸化对于IL-22增强IFN-λ诱导的TRIM25产生的过程至关重要,被一种新的STAT3磷酸化抑制剂WP1066抑制。TRIM25在DEF中的过表达导致高产量的IFN和减少的DHAV-1复制,而在RNAi组中观察到IFN的表达减弱和DHAV-1的复制促进,这意味着TRIM25通过诱导IFN的产生来保护生物体免受DHAV-1的繁殖。总之,我们报道,IL-22激活STAT3的磷酸化,增强IFN-λ介导的TRIM25表达,并通过诱导IFN产生提供对DHAV-1的防御.
    The innate immune system provides a defense against invading pathogens by inducing various interferon (IFN)-stimulated genes (ISGs). We recently reported that tripartite motif protein 25 (TRIM25), an important ISG, was highly upregulated in duck embryo hepatocyte cells (DEFs) after infection with duck viral hepatitis A virus type 1 (DHAV-1). However, the mechanism of upregulation of TRIM25 remains unknown. Here we reported that interleukin-22 (IL-22), whose expression was highly facilitated in DEFs and various organs of 1-day-old ducklings after DHAV-1 infection, highly enhanced the IFN-λ-induced production of TRIM25. The treatment with IL-22 neutralizing antibody or the overexpression of IL-22 highly suppressed or facilitated TRIM25 expression, respectively. The phosphorylation of signal transducer and activator of transcription 3 (STAT3) was crucial for the process of IL-22 enhancing IFN-λ-induced TRIM25 production, which was suppressed by WP1066, a novel inhibitor of STAT3 phosphorylation. The overexpression of TRIM25 in DEFs resulted in a high production of IFNs and reduced DHAV-1 replication, whereas the attenuated expression of IFNs and facilitated replication of DHAV-1 were observed in the RNAi group, implying that TRIM25 defended the organism against DHAV-1 propagation by inducing the production of IFNs. In summary, we reported that IL-22 activated the phosphorylation of STAT3 to enhance the IFN-λ-mediated TRIM25 expression and provide a defense against DHAV-1 by inducing IFN 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
    目的:干扰素-λ(IFN-λ)是一种新型的非冗余调节因子,参与胎儿-母体免疫相互作用,包括免疫调节,子宫容受性,细胞迁移和粘附,和子宫内膜凋亡。然而,IFN-λ子宫内膜信号的确切转录基础尚未完全了解,关于IFN-λ在体内植入失败的研究是有限的。
    方法:使用RNA测序分析用IFN-λ或IFN-α(100ng/mL)处理6小时的人子宫内膜Ishikawa细胞系的基因表达谱。实时qPCR,西方印迹,和酶联免疫吸附测定(ELISA)测试用于验证这些测序数据。进行体内IFN-λ敲低小鼠妊娠模型,并对子宫样本进行表型分析和宫内生物标志物检测。
    结果:先前与子宫内膜容受性相关的基因检测到高水平的信使RNA(mRNA),包括LIF,AXL,CRYAB,IFN-λ处理后的EPHB2、CCL5和DDX58。此外,数据表明,与IFN-α相比,IFN-λ降低了促炎基因活性,包括ISG成员,TNF,SP100和白细胞介素基因。体内小鼠妊娠模型表明,抑制宫内IFN-λ会导致上皮表型异常,并显着降低胚胎植入率并破坏正常的子宫容受性。
    结论:这些发现证明了IFNs在子宫内膜细胞中的拮抗和激动作用,提示IFN-λ在子宫内膜容受性和免疫耐受调节中的选择性作用。此外,这些研究结果为了解与子宫内膜容受性相关的潜在生物标志物提供了有价值的见解,并有助于了解在不孕症治疗和避孕使用过程中观察到的分子变化.
    Interferon-λ (IFN-λ) is a novel non-redundant regulator that participates in the fetal-maternal immune interaction, including immune regulation, uterine receptivity, cell migration and adhesion, and endometrium apoptosis. However, the exact transcriptional foundation for endometrial signaling of IFN-λ is not completely understood, and studies regarding IFN-λ to implantation failure in vivo are limited.
    The gene expression profile of human endometrial Ishikawa cell line treated with IFN-λ or IFN-α (100 ng/mL) for 6 h was analyzed using RNA-sequencing. Real-time qPCR, western blotting, and enzyme-linked immunosorbent assay (ELISA) tests were used to validate these sequencing data. An in vivo IFN-λ knock-down mouse pregnancy model was performed, and the phenotype analysis and the intrauterine biomarkers detection were applied with the uterus samples.
    High levels of messenger RNA (mRNA) were detected for genes previously associated with endometrial receptivity, including LIF, AXL, CRYAB, EPHB2, CCL5, and DDX58, following IFN-λ treatment. Moreover, the data indicated IFN-λ reduced pro-inflammatory gene activity compared with IFN-α, including members of the ISG, TNF, SP100 and interleukin genes. The in vivo mouse pregnancy model showed that inhibition of intrauterine IFN-λ results in aberrant epithelial phenotype and significantly decreases the embryo implantation rates and derails normal uterine receptivity.
    These findings demonstrate the antagonistic and agonistic roles of IFNs in the endometrial cell, suggesting a selective role of IFN-λ in endometrial receptivity and immunological tolerance regulation. Moreover, the findings provide valuable insight into potential biomarkers related to endometrial receptivity and facilitate an understanding of the molecular changes observed during infertility treatment and contraception usage.
    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