type I IFNs

I 型 IFNs
  • 文章类型: Journal Article
    背景:放疗可以调节全身抗肿瘤免疫,虽然肿瘤微环境中的免疫状态也会影响放疗的疗效,但相关分子机制在肺腺癌(LUAD)中知之甚少。
    方法:在本研究中,我们创新性地提出了LUAD的放射治疗反应分类,并发现ESYT3作为肿瘤抑制因子和放射免疫应答增敏剂。在放射抗性和放射敏感性LUAD组织和细胞中测量ESYT3表达。然后研究ESYT3对放疗敏感性和耐药性的影响。ESYT3和STING之间的相互作用通过多重免疫荧光染色和免疫共沉淀进行评估,和下游分子进一步分析。构建体内模型以评估ESYT3过表达与放射疗法的组合治疗功效。
    结果:我们发现放射抗性亚型比放射敏感性亚型呈现免疫抑制状态和DNA损伤修复途径的激活。ESYT3表达在耐放射性LUAD组织和细胞中均显著减弱。临床上,低ESYT3表达与放射抗性有关。ESYT3的过表达能够减轻放射抗性,并使LUAD细胞对辐射诱导的DNA损伤敏感。机械上,ESYT3直接与STING交互,和激活的cGAS-STING信号,随后增加I型IFN以及下游趋化因子CCL5和CXCL10的产生,从而改善放射免疫应答。ESYT3过表达与放疗的联合治疗在体外和体内具有协同抗癌作用。
    结论:总之,低ESYT3表达赋予LUAD对放疗的抗性,其过度表达可通过激活cGAS-STING依赖性途径改善放射免疫反应,从而为LUAD患者提供了一种替代的联合治疗策略.
    BACKGROUND: Radiotherapy can modulate systemic antitumor immunity, while immune status in the tumor microenvironment also influences the efficacy of radiotherapy, but relevant molecular mechanisms are poorly understood in lung adenocarcinoma (LUAD).
    METHODS: In this study, we innovatively proposed a radiotherapy response classification for LUAD, and discovered ESYT3 served as a tumor suppressor and radioimmune response sensitizer. ESYT3 expression was measured both in radioresistant and radiosensitive LUAD tissues and cells. The influence of ESYT3 on radiotherapy sensitivity and resistance was then investigated. Interaction between ESYT3 and STING was evaluated through multiple immunofluorescent staining and coimmunoprecipitation, and downstream molecules were further analyzed. In vivo models were constructed to assess the combination treatment efficacy of ESYT3 overexpression with radiotherapy.
    RESULTS: We found that radioresistant subtype presented immunosuppressive state and activation of DNA damage repair pathways than radiosensitive subtype. ESYT3 expression was remarkably attenuated both in radioresistant LUAD tissues and cells. Clinically, low ESYT3 expression was linked with radioresistance. Overexpression of ESYT3 enabled to alleviate radioresistance, and sensitize LUAD cells to DNA damage induced by irradiation. Mechanically, ESYT3 directly interacted with STING, and activated cGAS-STING signaling, subsequently increasing the generation of type I IFNs as well as downstream chemokines CCL5 and CXCL10, thus improving radioimmune responses. The combination treatment of ESYT3 overexpression with radiotherapy had a synergistic anticancer effect in vitro and in vivo.
    CONCLUSIONS: In summary, low ESYT3 expression confers resistance to radiotherapy in LUAD, and its overexpression can improve radioimmune responses through activating cGAS-STING-dependent pathway, thus providing an alternative combination therapeutic strategy for LUAD patients.
    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IFN是主要通过JAK-STAT信号传导发挥其抗病毒作用的细胞因子的子集。免疫遗传学研究表明,鱼类具有IFN-JAK-STAT级联的关键成分,但是关于STAT1和STAT2对不同IFN的不同反应的信息在鱼类中相当有限。这里,我们从罗非鱼中鉴定并克隆了STAT1和STAT2基因(命名为On-STAT1和On-STAT2),尼罗罗非鱼。在所有检查的orangs/组织中检测到On-STAT1和On-STAT2基因,并在脾脏中迅速诱导,头肾,和在聚(I:C)刺激后的肝脏。此外,聚(I:C)的刺激,聚(A:T),不同亚组的重组IFN可以诱导TA-02细胞中On-STAT1和On-STAT2的表达,诱导水平不同。重要的是,On-STAT2被IFN的所有三个亚组快速磷酸化,但On-STAT1的磷酸化仅在IFNc和IFNh处理的TA-02细胞中观察到,反映了鱼类IFN的不同亚组对STAT的不同激活。因此,本结果有助于更好地理解鱼类中不同IFN亚组介导的JAK-STAT信号传导。
    Type I IFNs are a subset of cytokines exerting their antiviral effects mainly through the JAK-STAT signalling. Immunogenetic studies have shown that fish possess key components of IFN-JAK-STAT cascade, but the information about the distinct responses of STAT1 and STAT2 to different IFNs is rather limited in fish. Here, we identified and cloned STAT1 and STAT2 genes (named as On-STAT1 and On-STAT2) from tilapia, Oreochromis niloticus. On-STAT1 and On-STAT2 genes were detected in all orangs/tissues examined, and were rapidly induced in spleen, head kidney, and liver following the stimulation of poly(I:C). In addition, the stimulation of poly(I:C), poly(A:T), and different subgroups of recombinant IFNs could induce the expression of On-STAT1 and On-STAT2 in TA-02 cells with distinct induction levels. Importantly, On-STAT2 was rapidly phosphorylated by all three subgroups of IFNs, but the phosphorylation of On-STAT1 was only observed in IFNc- and IFNh-treated TA-02 cells, reflecting the distinct activation of STAT by different subgroups of fish IFNs. The present results thus contribute to better understanding of the JAK-STAT signalling mediated by different subgroups of IFNs in fish.
    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
    在病毒感染期间,先天性免疫系统利用各种特定的细胞内传感器来检测病毒衍生的核酸并激活一系列产生I型IFN和促炎细胞因子和趋化因子的细胞信号级联。卡波西肉瘤相关疱疹病毒(KSHV)是一种致癌双链DNA病毒,与多种人类恶性肿瘤有关。包括卡波西肉瘤,原发性渗出性淋巴瘤,和多中心Castleman病.KSHV感染激活了各种DNA传感器,包括CGAS,STING,IFI16和DExD/H-box解旋酶。这些DNA传感器的激活诱导先天性免疫应答以拮抗病毒。为了抵消这一点,KSHV已经开发了无数的策略来逃避或抑制DNA感应并促进其自身的感染。这篇综述总结了主要的DNA触发感应信号通路,并详细介绍了KSHV感染中涉及的DNA感应机制的最新知识。以及KSHV如何逃避抗病毒信号通路以成功建立潜伏感染并进行裂解剂再激活。
    During viral infection, the innate immune system utilizes a variety of specific intracellular sensors to detect virus-derived nucleic acids and activate a series of cellular signaling cascades that produce type I IFNs and proinflammatory cytokines and chemokines. Kaposi\'s sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus that has been associated with a variety of human malignancies, including Kaposi\'s sarcoma, primary effusion lymphoma, and multicentric Castleman disease. Infection with KSHV activates various DNA sensors, including cGAS, STING, IFI16, and DExD/H-box helicases. Activation of these DNA sensors induces the innate immune response to antagonize the virus. To counteract this, KSHV has developed countless strategies to evade or inhibit DNA sensing and facilitate its own infection. This review summarizes the major DNA-triggered sensing signaling pathways and details the current knowledge of DNA-sensing mechanisms involved in KSHV infection, as well as how KSHV evades antiviral signaling pathways to successfully establish latent infection and undergo lytic reactivation.
    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
    目标:已在危及生命的2019年冠状病毒病(COVID-19)患者中鉴定出I型干扰素(IFN)的自身抗体(auto-abs),提示自体腹肌的存在可能是疾病严重程度的危险因素。因此,我们研究了auto-abs诱导COVID-19恶化为I型IFN的潜在机制。
    方法:我们评估了123例COVID-19患者的血浆,以测量I型IFN的auto-abs。我们对auto-abs患者的外周血单核细胞进行了单细胞RNA测序(scRNA-seq),并对auto-abs进行了表位定位。
    结果:19例重症患者中的3例和42例重症COVID-19患者中的4例具有中和I型IFN的自体腹肌。患有I型IFNs的患者没有特征性临床特征。来自38例COVID-19患者的scRNA-seq显示,常规树突状细胞和经典单核细胞中的IFN信号传导减弱,自动腹肌患者的SARS-CoV-2特异性BCR谱减少。此外,COVID-19患者对IFN-α2的自体abs识别IFN-α2的特征性表位,IFN-α2与受体结合。
    结论:在COVID-19患者中发现的I型IFN的Auto-abs通过阻断I型IFN与其受体的结合来抑制树突状细胞和单核细胞中的IFN信号传导。未能正确诱导SARS-CoV-2抗体的产生可能是COVID-19严重程度的致病因素。
    OBJECTIVE: Auto-antibodies (auto-abs) to type I interferons (IFNs) have been identified in patients with life-threatening coronavirus disease 2019 (COVID-19), suggesting that the presence of auto-abs may be a risk factor for disease severity. We therefore investigated the mechanism underlying COVID-19 exacerbation induced by auto-abs to type I IFNs.
    METHODS: We evaluated plasma from 123 patients with COVID-19 to measure auto-abs to type I IFNs. We performed single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells from the patients with auto-abs and conducted epitope mapping of the auto-abs.
    RESULTS: Three of 19 severe and 4 of 42 critical COVID-19 patients had neutralizing auto-abs to type I IFNs. Patients with auto-abs to type I IFNs showed no characteristic clinical features. scRNA-seq from 38 patients with COVID-19 revealed that IFN signaling in conventional dendritic cells and canonical monocytes was attenuated, and SARS-CoV-2-specific BCR repertoires were decreased in patients with auto-abs. Furthermore, auto-abs to IFN-α2 from COVID-19 patients with auto-abs recognized characteristic epitopes of IFN-α2, which binds to the receptor.
    CONCLUSIONS: Auto-abs to type I IFN found in COVID-19 patients inhibited IFN signaling in dendritic cells and monocytes by blocking the binding of type I IFN to its receptor. The failure to properly induce production of an antibody to SARS-CoV-2 may be a causative factor of COVID-19 severity.
    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),在大多数病毒感染后立即触发,在直接抗病毒免疫中起关键作用,并充当先天和适应性免疫应答之间的桥梁。然而,许多病毒已经进化出针对IFN应答的逃避策略,促使人们探索病毒感染的治疗方法。在I型IFN家族中,存在12种IFNα亚型,它们都与相同的受体结合,但在它们的生物活性上表现出显著的差异。目前,慢性病毒感染的临床治疗主要依赖于单一的IFNα亚型(IFNα2a/b)。然而,这种治疗的疗效相对有限,特别是在人类免疫缺陷病毒(HIV)感染的情况下。最近的研究已经深入研究了替代的IFNα亚型,确定某些亚型是高度有效的,其抗病毒和免疫调节特性已被广泛表征。这篇综述巩固了有关单个IFNα亚型在HIV和猿猴免疫缺陷病毒(SIV)感染中的作用的最新发现。它包括它们在艾滋病毒/SIV感染的背景下的诱导,它们的抗逆转录病毒活性,以及不同的IFNα亚型对HIV免疫应答的不同调节。这些见解可能为HIV治愈或功能治愈研究的创新策略铺平道路。
    Type I interferons (IFN), immediately triggered following most viral infections, play a pivotal role in direct antiviral immunity and act as a bridge between innate and adaptive immune responses. However, numerous viruses have evolved evasion strategies against IFN responses, prompting the exploration of therapeutic alternatives for viral infections. Within the type I IFN family, 12 IFNα subtypes exist, all binding to the same receptor but displaying significant variations in their biological activities. Currently, clinical treatments for chronic virus infections predominantly rely on a single IFNα subtype (IFNα2a/b). However, the efficacy of this therapeutic treatment is relatively limited, particularly in the context of Human Immunodeficiency Virus (HIV) infection. Recent investigations have delved into alternative IFNα subtypes, identifying certain subtypes as highly potent, and their antiviral and immunomodulatory properties have been extensively characterized. This review consolidates recent findings on the roles of individual IFNα subtypes during HIV and Simian Immunodeficiency Virus (SIV) infections. It encompasses their induction in the context of HIV/SIV infection, their antiretroviral activity, and the diverse regulation of the immune response against HIV by distinct IFNα subtypes. These insights may pave the way for innovative strategies in HIV cure or functional cure studies.
    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:干扰素(IFN)的产生对于控制病毒感染至关重要,干扰素调节因子7(IRF7)被认为是I型IFN转录的重要调节因子。两栖动物似乎拥有高度扩展的I型IFN库,由在鱼类中观察到的含内含子的基因组成,和其他高等脊椎动物中的无内含子基因。然而,关于这两种类型的I型IFN基因的转录调控机制的知识在两栖动物中相当匮乏。
    结果:在藏蛙(Nanoranaparkeri)中鉴定出一种名为Np-IRF7的IRF7基因,和生物信息学分析表明,Np-IRF7的预测蛋白包含IRF7中已知的几个重要结构特征。表达分析表明,Np-IRF7基因在不同器官/组织中被poly(I:C)广泛表达并快速诱导。有趣的是,荧光素酶报告基因分析显示,在Np-IRF7转染的细胞中,无内含子IFN启动子比含内含子的IFN启动子更有效地激活。此外,Np-IRF7的过表达可以诱导ISGs的表达并抑制A6细胞中FV3的复制。
    结论:Np-IRF7确实是已知IRF7的直系同源物,并且IRF7在脊椎动物的不同谱系中在结构上是保守的。Np-IRF7在含内含子和无内含子I型IFN启动子的激活中起着不同的作用,从而诱导干扰素刺激的抗病毒效应物的表达,并提供对流感病毒感染的保护。因此,本研究有助于更好地了解IRF7在IFN介导的两栖动物的抗病毒反应中的调节功能。
    BACKGROUND: The production of interferons (IFNs) is essential for the control of viral infections, and interferon regulatory factor 7 (IRF7) is considered as a vital regulator for the transcription of type I IFNs. Amphibians appear to possess a highly expanded type I IFN repertoire, consisting of intron-containing genes as observed in fish, and intronless genes as in other higher vertebrates. However, the knowledge on transcriptional regulatory mechanism of these two types of type I IFN genes is rather scarce in amphibians.
    RESULTS: A IRF7 gene named as Np-IRF7 was identified in Tibetan frog (Nanorana parkeri), and bioinformatic analysis revealed that the predicted protein of Np-IRF7 contains several important structural features known in IRF7. Expression analysis showed that Np-IRF7 gene was widely expressed and rapidly induced by poly(I:C) in different organs/tissues. Interestingly, luciferase reporter assay revealed that intronless IFN promoters were more effectively activated than intron-containing IFN promoter in Np-IRF7-transfected cells. Moreover, the overexpression of Np-IRF7 could induce the expression of ISGs and suppress the replication of FV3 in A6 cells.
    CONCLUSIONS: Np-IRF7 is indeed the ortholog of known IRF7, and IRF7 is structurally conserved in different lineages of vertebrates. Np-IRF7 played distinct roles in the activation of intron-containing and intronless type I IFN promoters, thus inducing the expression of interferon-stimulated antiviral effectors and providing a protection against ranavirus infection. The present research thus contributes to a better understanding of regulatory function of IRF7 in the IFN-mediated antiviral response of anuran amphibians.
    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
    自然杀伤(NK)细胞是重要的抗病毒效应细胞,也参与肿瘤的清除。NK细胞表达IFNAR,使它们响应于类型IIFNs。为了评估I型IFN介导的NK细胞功能的调节,评估个体I型IFNs亚型激活NK细胞的能力。不同的I型IFN亚型在诱导和调节NK细胞活化和脱粒的能力方面表现出广泛的范围,通过响应于白血病细胞系K562的CD69和CD107a表达来测量。当在分析中包括生物性别作为变量时,用TLR7/8激动剂CL097刺激的NK细胞与雄性或雌性来源的PBMC或pDC的transwell共培养表明,NK细胞被来自雌性的CL097刺激的细胞更激活。这些性别特异性差异与来自女性的pDC产生更高的CL097诱导的IFNα有关,表明I型IFN对NK细胞功能的外在性别特异性作用。有趣的是,除了外在效应,我们还观察到NK细胞内在的性别差异,由于雌性NK细胞在IFNα刺激后以及与CL097刺激的pDCs共培养后显示出更高的活化水平,提示女性NK细胞中IFNα信号转导的更高激活。一起来看,这些研究的结果确定了I型IFN依赖性NK细胞功能的外在和内在性别特异性差异,有助于更好地理解先天免疫的性别差异。
    Natural killer (NK) cells are important antiviral effector cells and also involved in tumor clearance. NK cells express IFNAR, rendering them responsive to Type I IFNs. To evaluate Type I IFN-mediated modulation of NK cell functions, individual Type I IFNs subtypes were assessed for their ability to activate NK cells. Different Type I IFN subtypes displayed a broad range in the capacity to induce and modulate NK cell activation and degranulation, measured by CD69 and CD107a expression in response to leukemia cell line K562. When including biological sex as a variable in the analysis, transwell co-cultures of NK cells with either male- or female-derived PBMCs or pDCs stimulated with the TLR7/8 agonist CL097 showed that NK cells were more activated by CL097-stimulated cells derived from females. These sex-specific differences were linked to higher CL097-induced IFNα production by pDCs derived from females, indicating an extrinsic sex-specific effect of Type I IFNs on NK cell function. Interestingly, in addition to the extrinsic effect, we also observed NK cell-intrinsic sex differences, as female NK cells displayed higher activation levels after IFNα-stimulation and after co-culture with CL097-stimulated pDCs, suggesting higher activation of IFNα-signaling transduction in female NK cells. Taken together, the results from these studies identify both extrinsic and intrinsic sex-specific differences in Type I IFN-dependent NK cell functions, contributing to a better understanding of sex-specific differences in innate immunity.
    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合成酶(cGAS),被认为是细胞内的主要DNA传感器,具有鉴定外源DNA分子和游离DNA片段的能力。该鉴定过程通过干扰素基因的激活剂(STING)促进I型IFN的产生,所述激活剂诱导下游转录因子的磷酸化。该作用表征了cGAS-STING途径的最典型的生物学功能。当使用抗肿瘤药物治疗时,细胞经历DNA损伤,触发cGAS-STING途径的激活,最终导致I型IFN和相关下游干扰素刺激基因的表达。cGAS-STING是重要的先天免疫途径之一,I型IFN在先天免疫和T细胞抗肿瘤免疫之间的衔接中的作用。I型IFN促进肿瘤部位炎性细胞(包括NK细胞)的募集和活化。I型IFN还可以促进树突状细胞(DC)的激活和成熟,改善CD4+T淋巴细胞的抗原呈递,并增强CD8T淋巴细胞的交叉呈递以上调抗肿瘤反应。本文就cGAS-STING信号通路及其在传统肿瘤治疗和免疫治疗中的作用机制和生物学功能作一综述。
    Cyclic GMP-AMP synthetase (cGAS), recognized as the primary DNA sensor within cells, possesses the capability to identify foreign DNA molecules along with free DNA fragments. This identification process facilitates the production of type I IFNs through the activator of the interferon gene (STING) which induces the phosphorylation of downstream transcription factors. This action characterizes the most archetypal biological functionality of the cGAS-STING pathway. When treated with anti-tumor agents, cells experience DNA damage that triggers activation of the cGAS-STING pathway, culminating in the expression of type I IFNs and associated downstream interferon-stimulated genes. cGAS-STING is one of the important innate immune pathways,the role of type I IFNs in the articulation between innate immunity and T-cell antitumour immunity.type I IFNs promote the recruitment and activation of inflammatory cells (including NK cells) at the tumor site.Type I IFNs also can promote the activation and maturation of dendritic cel(DC), improve the antigen presentation of CD4+T lymphocytes, and enhance the cross-presentation of CD8+T lymphocytes to upregulating anti-tumor responses. This review discussed the cGAS-STING signaling and its mechanism and biological function in traditional tumor therapy and 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
    环磷酸鸟苷(GMP)-AMP(cGAMP)合酶(cGAS)是一种通用的双链DNA(dsDNA)传感器,可识别细胞质中的外源DNA和自身DNA并引发先天免疫反应,并涉及各种感染性和非感染性环境。cGAS与dsDNA的骨架结合并产生第二信使,cGAMP,激活干扰素基因(STING)的刺激物。这里,我们显示内源性多胺精胺和亚精胺减弱cGAS活性和先天免疫应答。机械上,精胺和亚精胺诱导B型DNA向Z型DNA(Z-DNA)的转变,从而降低其与cGAS的结合亲和力。亚精胺/精胺N1-乙酰转移酶1(SAT1),多胺分解代谢中的限速酶,可降低精胺和亚精胺的细胞浓度,通过抑制细胞Z-DNA积累增强cGAS激活;SAT1缺乏促进单纯疱疹病毒1(HSV-1)在体内复制。结果表明,精胺和亚精胺诱导dsDNA采用Z型构象,而SAT1介导的多胺代谢会协调cGAS活性。
    Cyclic guanosine monophosphate (GMP)-AMP (cGAMP) synthase (cGAS) is a universal double-stranded DNA (dsDNA) sensor that recognizes foreign and self-DNA in the cytoplasm and initiates innate immune responses and has been implicated in various infectious and non-infectious contexts. cGAS binds to the backbone of dsDNA and generates the second messenger, cGAMP, which activates the stimulator of interferon genes (STING). Here, we show that the endogenous polyamines spermine and spermidine attenuated cGAS activity and innate immune responses. Mechanistically, spermine and spermidine induced the transition of B-form DNA to Z-form DNA (Z-DNA), thereby decreasing its binding affinity with cGAS. Spermidine/spermine N1-acetyltransferase 1 (SAT1), the rate-limiting enzyme in polyamine catabolism that decreases the cellular concentrations of spermine and spermidine, enhanced cGAS activation by inhibiting cellular Z-DNA accumulation; SAT1 deficiency promoted herpes simplex virus 1 (HSV-1) replication in vivo. The results indicate that spermine and spermidine induce dsDNA to adopt the Z-form conformation and that SAT1-mediated polyamine metabolism orchestrates cGAS activity.
    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
    炎症性肠病(IBD)的发展是由包括TNF-α在内的促炎细胞因子的过度产生驱动的。IL-12和IL-23。这一观点得到了靶向这些细胞因子的生物制剂的显著临床成功的支持。在人IBD和实验性结肠炎模型中,通过Toll样受体(TLR)识别源自肠道细菌的细胞壁组分诱导巨噬细胞和树突状细胞产生这些促炎细胞因子。尽管通过内体TLRs感知细菌核酸,特别是TLR3、TLR7和TLR9导致I型IFN的稳健生产,TLR介导的I型IFN应答在IBD患者中是否具有致病性或保护性仍有争议.此外,最近的研究确定去泛素化酶A(DUBA)是TLR介导的I型IFN应答的新型负调节因子。鉴于这些观察结果及其潜在的应用,在这次审查中,我们总结了人类IBD和实验性结肠炎模型中I型IFN应答和DUBA介导的这些应答的负调控作用的最新发现。
    The development of Inflammatory bowel disease (IBD) is driven by excessive production of pro-inflammatory cytokines including TNF-α, IL-12, and IL-23. This notion is supported by the remarkable clinical success of biologics targeting these cytokines. Recognition of cell wall components derived from intestinal bacteria by Toll-like receptors (TLRs) induces the production of these pro-inflammatory cytokines by macrophages and dendritic cells in human IBD and experimental colitis model. Although sensing of bacterial nucleic acids by endosomal TLRs, specifically TLR3, TLR7, and TLR9 leads to robust production of type I IFNs, it remains debatable whether TLR-mediated type I IFN responses are pathogenic or protective in IBD patients. Additionally, recent studies identified deubiquitinating enzyme A (DUBA) as a novel negative regulator of TLR-mediated type I IFN responses. In light of these observations and their potential applications, in this review, we summarize recent findings on the roles of type I IFN responses and DUBA-mediated negative regulation of these responses in human IBD and experimental colitis model.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号