首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 140 毫秒
1.
线粒体抗病毒信号蛋白(MAVS)作为一种接头蛋白在调节宿主天然免疫信号通路过程中扮演重要角色.Toll样受体(TLR)和RIG-Ⅰ样受体(RLR)等细胞模式识别受体识别入侵的病原体并将信号传递给MAVS,MAVS通过刺激下游的TBK1复合体和IKK复合体分别活化NF-κB和IRF3等信号通路,进而激活干扰素α/β表达,诱发细胞内抗感染天然免疫反应.MAVS除定位线粒体外,也可定位于过氧化物酶体上.MAVS在细胞内的不同定位决定了其在早期快速和持续性抗病毒天然免疫中的不同调节机制.MAVS只有同时定位在过氧化物酶体和线粒体上才可诱导干扰素刺激基因(ISG)快速且稳定地表达.本文通过对MAVS的发现、结构、细胞定位及其在天然免疫信号通路中的调控机制等最新进展进行综述,以期揭示MAVS蛋白在细胞内天然免疫信号通路中的重要调节作用,为研究病毒逃逸宿主天然免疫的机制和研究新型抗病毒免疫治疗策略提供新思路.  相似文献   

2.
天然免疫是宿主防御病原微生物入侵的第一道防线,其活化主要通过天然免疫细胞上的模式识别受体(pattern recognition receptors, PRRs)识别病原微生物上相对保守的相关分子模式(pathogen-associated molecular patterns, PAMPs).病毒相关的核酸成分可以被机体Toll样受体(Toll-like receptors, TLRs)、维甲酸诱导基因Ⅰ受体(RIG-I-like receptors, RLRs)以及胞浆DNA受体(cytoplasmic DNA sensors)等识别,通过一系列复杂的细胞信号通路诱导Ⅰ型干扰素(typeⅠinterferon)及炎症因子的表达,从而激发机体抗病毒反应.泛素化修饰是细胞内广泛存在的蛋白质翻译后修饰方式,在宿主防御病原微生物感染的动态调控过程中发挥着重要的作用.已有大量文献报道,天然免疫抗病毒信号通路中的多个关键接头分子可发生泛素化修饰,进而调控机体抗病毒免疫应答反应.本文综述了泛素化修饰在抗病毒天然免疫中的作用及其调控机制.  相似文献   

3.
RIG-I样受体与RNA病毒识别   总被引:2,自引:0,他引:2  
秦成峰  秦鄂德 《微生物学报》2008,48(10):1418-1423
RIG-I样受体(RIG-I like receptors,RLR)是一类新发现的模式识别受体,能够识别细胞质中的病毒RNA,通过RLR级联信号诱导干扰素和促炎症细胞因子的产生,对抗病毒天然免疫的建立起着非常重要的作用.RLR信号通路既受宿主的严格调控,也能够作为病毒逃避宿主干扰素反应的靶点.本文重点讨论了RLR及其在RNA病毒识别和抗病毒天然免疫中的作用.  相似文献   

4.
流感病毒(influenza virus)轻症感染可由机体免疫系统清除,但重症感染则诱发肺脏免疫损伤。流感病毒的病原体相关分子模式(pathogen-associated molecular patterns,PAMPs)可被位于细胞膜、细胞器膜及胞质内的重要模式识别受体(pattern recognition receptors,PRRs)介导识别,活化一系列激酶及转录因子,诱导促炎细胞因子和趋化因子的表达、成熟和分泌,进一步激活天然免疫及获得性免疫应答细胞,介导炎症反应和诱导免疫病理损伤。PRRs是研究天然免疫应答启动机制及抑制重症感染诱导免疫病理损伤的重要靶点。现就Toll样受体(toll-like receptors,TLRs)中的TLR3、TLR7/8、TLR4、RIG-I样受体(RIG-I like receptors,RLRs)和NOD样受体(NOD-like receptor,NLR)在流感病毒感染中的识别及下游信号通路在免疫病理损伤中的作用机制作一综述。  相似文献   

5.
RLR[retinoic acid-inducible gene Ⅰ(RIG-Ⅰ)-like Receptors]是一类表达在胞浆中的模式识别受体, 在识别细胞质中经病毒复制产生的病毒RNA后, 启动一系列信号级联反应, 以诱导机体Ⅰ型干扰素及干扰素诱导的抗病毒基因的表达, 最后达到清除机体病毒感染的目的。由于在病毒感染时机体干扰素反应必须迅速启动, 当病毒清除后干扰素反应又需要立即恢复到正常本底水平, 因此RLR激活的信号转导途径受到了严格的调控, 其中就包括由E3泛素连接酶参与的泛素化修饰调控和由去泛素化酶参与的去泛素化修饰调控。自2003年成功鉴定出鱼类干扰素基因以来, 鱼类也被发现具有保守的RLR信号转导途径诱导干扰素抗病毒免疫反应, 该信号途径同样受到泛素化修饰的调控。文章总结了近年来泛素化修饰在哺乳类和鱼类RLR介导的抗病毒免疫应答通路中的调节机制。  相似文献   

6.
STING在宿主天然免疫信号通路中的调节作用   总被引:1,自引:0,他引:1       下载免费PDF全文
STING(stimulator of interferon genes)是天然免疫信号通路中一种新发现的蛋白质,在防御病毒及胞内细菌感染、介导Ⅰ型IFN产生过程中发挥重要功能.来自病原体的B型DNA与5′-3p dsRNA暴露在宿主细胞中后被相应的模式识别受体识别,通过不同的通路传递信号给STING.STING随后通过相似的机制招募TBK1激活IRF3,诱导干扰素表达.对细菌中的环二核苷酸c-di-GMP和c-di-AMP,STING则可以直接作为模式识别受体引发Ⅰ型干扰素反应.此外STING还能激活STAT6诱导特异趋化因子产生,吸引各种免疫细胞抵抗病毒感染.本文通过对STING的发现、结构、定位、功能、机理以及调节机制进行综述,以期为揭示病毒逃逸天然免疫调节机制和抗病毒新型免疫调节剂提供新的思路.  相似文献   

7.
李倩  李向茸  冯若飞 《病毒学报》2019,35(2):313-317
RIG-I样受体(RIG-I like receptors,RLRs)是天然免疫系统中一类重要模式识别受体,在细胞天然免疫应答中发挥重要的作用,LGP2是RLRs家族成员之一,对RLRs信号转导有正向和负向调控的双重作用。LGP2依据感染病毒种类的不同发挥着不同的调控作用,但详细作用机制不明确。随着研究的深入,LGP2在天然免疫应答中的作用愈发重要。本文就近年来对LGP2参与RLRs介导的抗病毒天然免疫应答的调控及在不同病毒感染宿主中所起的作用作一综述,以期为LGP2深入研究提供参考。  相似文献   

8.
动物关键模式识别受体及其抗病毒天然免疫作用研究进展   总被引:1,自引:0,他引:1  
天然免疫系统是动物抵御病原入侵的第一道防线,在机体抗病毒感染过程中发挥重要作用,模式识别受体(pattern-recognition receptors,PRRs)是天然免疫系统的重要成分,动物机体的抗病毒免疫机制是由一系列PRRs对病原体的识别所启动的。近年来识别和感受病原体的一系列动物PRRs受到广泛关注,成为动物医学领域的研究热点,为揭示动物复杂的抗病毒天然免疫反应提供了新思路。该文简要介绍动物Toll样受体(Toll-like receptors,TLRs)和维甲酸诱导基因Ⅰ样受体(RIG-I like receptors,RLRs)的分子特征及其介导的抗病毒天然免疫作用研究进展。  相似文献   

9.
张其奥  王子路  李佩波  谢建平 《遗传》2023,(11):998-1006
干扰素诱导基因15 (interferon-stimulated gene 15,isg15)的表达受Ⅰ型干扰素诱导,该基因编码的蛋白ISG15可以分别通过E1、E2和E3酶的作用共价修饰靶蛋白,此过程被称为ISG化(ISGylation)。宿主蛋白的ISG化广泛参与天然免疫例如宿主的抗病毒过程。泛素特异性蛋白酶18 (ubiquitin-specific protease 18,USP18)作为一种去泛素化酶(deubiquitinase,DUB)可以去除靶蛋白偶联的ISG15,并通过抑制Ⅰ型干扰素信号通路来抑制宿主的免疫应答。ISG15介导的ISG化和USP18介导的去ISG化(deISGylation)建立的动态平衡对结核病的发生、发展和转归有重要影响。此外,同ISG15一样,USP18也广泛参与病毒感染和宿主细胞抗病毒反应,多种先天性免疫疾病和免疫信号通路都受到USP18的调节。本文综述了ISG15和USP18相关的研究进展,重点介绍了ISG15介导的ISGylation和USP18介导的去ISG化在结核病及其他重要疾病中的调控作用,以期为靶向宿主蛋白的结核病等重要疾病防治提供...  相似文献   

10.
弹状病毒(Rhabdovirus)是引起鱼类病毒性传染病的重要病原之一.近几年,鱼类弹状病毒感染斑马鱼(Danio rerio)诱导非特异性免疫反应的分子机制研究备受关注.作者通过鱼类弹状病毒感染斑马鱼进行转录组和蛋白质组学研究,结果表明,Toll样受体(TLRs)、RIG-Ⅰ样受体(RLRs)等介导的干扰素(IFNs)反应以及补体途径等被激活,并且丝裂原活化蛋白激酶(MAPKs)、髓样分化因子88(My D88)/核因子κB(NF-κB)炎症信号通路等参与抗病毒应答的调控过程.本文首次综述了4种鱼类弹状病毒(SVCV,VHSV,IHNV,SHRV)感染诱导斑马鱼免疫应答的分子调控及其研究进展,以期将来为鱼类弹状病毒性疾病研究与防治提供科学参考.  相似文献   

11.
Known therapies for influenza A virus infection are complicated by the frequent emergence of resistance. A therapeutic strategy that may escape viral resistance is targeting host cellular mechanisms involved in viral replication and pathogenesis. The endoplasmic reticulum (ER) stress response, also known as the unfolded protein response (UPR), is a primitive, evolutionary conserved molecular signaling cascade that has been implicated in multiple biological phenomena including innate immunity and the pathogenesis of certain viral infections. We investigated the effect of influenza A viral infection on ER stress pathways in lung epithelial cells. Influenza A virus induced ER stress in a pathway-specific manner. We showed that the virus activates the IRE1 pathway with little or no concomitant activation of the PERK and the ATF6 pathways. When we examined the effects of modulating the ER stress response on the virus, we found that the molecular chaperone tauroursodeoxycholic acid (TUDCA) significantly inhibits influenza A viral replication. In addition, a specific inhibitor of the IRE1 pathway also blocked viral replication. Our findings constitute the first evidence that ER stress plays a role in the pathogenesis of influenza A viral infection. Decreasing viral replication by modulating the host ER stress response is a novel strategy that has important therapeutic implications.  相似文献   

12.
Innate immunity to respiratory viruses   总被引:8,自引:0,他引:8  
Pattern recognition receptors are critically involved in the development of innate and adaptive antiviral immunity. Innate immune activation by viruses may occur via cell surface, intracellular and cytosolic pattern recognition receptors. These receptors sense viral components and may activate unique downstream pathways to generate antiviral immunity. In this article, we summarize the pattern recognition receptors that recognize major human respiratory viral pathogens, including influenza virus, respiratory syncytial virus and adenovirus. We also provide an overview of the current knowledge of regulation of type I interferons and inflammatory cytokines in viral infection.  相似文献   

13.
Influenza A viruses (IAV) have been the cause of several influenza pandemics in history and are a significant threat for the next global pandemic. Hospitalized influenza patients often have excess interferon production and a dysregulated immune response to the IAV infection. Obtaining a better understanding of the mechanisms of IAV infection that induce these harmful effects would help drug developers and health professionals create more effective treatments for IAV infection and improve patient outcomes. IAV stimulates viral sensors and receptors expressed by alveolar epithelial cells, like RIG-I and toll-like receptor 3 (TLR3). These two pathways coordinate with one another to induce expression of type III interferons to combat the infection. Presented here is a queuing theory-based model of these pathways that was designed to analyze the timing and amount of interferons produced in response to IAV single stranded RNA and double-stranded RNA detection. The model accurately represents biological data showing the necessary coordination of the RIG-I and TLR3 pathways for effective interferon production. This model can serve as the framework for future studies of IAV infection and identify new targets for potential treatments.  相似文献   

14.
15.
Influenza A viruses are important pathogens that cause acute respiratory diseases and annual epidemics in humans. Macrophages recognize influenza A virus infection with their pattern recognition receptors, and are involved in the activation of proper innate immune response. Here, we have used high-throughput subcellular proteomics combined with bioinformatics to provide a global view of host cellular events that are activated in response to influenza A virus infection in human primary macrophages. We show that viral infection regulates the expression and/or subcellular localization of more than one thousand host proteins at early phases of infection. Our data reveals that there are dramatic changes in mitochondrial and nuclear proteomes in response to infection. We show that a rapid cytoplasmic leakage of lysosomal proteins, including cathepsins, followed by their secretion, contributes to inflammasome activation and apoptosis seen in the infected macrophages. Also, our results demonstrate that P2X7 receptor and src tyrosine kinase activity are essential for inflammasome activation during influenza A virus infection. Finally, we show that influenza A virus infection is associated with robust secretion of different danger-associated molecular patterns (DAMPs) suggesting an important role for DAMPs in host response to influenza A virus infection. In conclusion, our high-throughput quantitative proteomics study provides important new insight into host-response against influenza A virus infection in human primary macrophages.  相似文献   

16.
A20 negatively regulates multiple inflammatory signalling pathways. We here addressed the role of A20 in club cells (also known as Clara cells) of the bronchial epithelium in their response to influenza A virus infection. Club cells provide a niche for influenza virus replication, but little is known about the functions of these cells in antiviral immunity. Using airway epithelial cell-specific A20 knockout (A20AEC-KO) mice, we show that A20 in club cells critically controls innate immune responses upon TNF or double stranded RNA stimulation. Surprisingly, A20AEC-KO mice are better protected against influenza A virus challenge than their wild type littermates. This phenotype is not due to decreased viral replication. Instead host innate and adaptive immune responses and lung damage are reduced in A20AEC-KO mice. These attenuated responses correlate with a dampened cytotoxic T cell (CTL) response at later stages during infection, indicating that A20AEC-KO mice are better equipped to tolerate Influenza A virus infection. Expression of the chemokine CCL2 (also named MCP-1) is particularly suppressed in the lungs of A20AEC-KO mice during later stages of infection. When A20AEC-KO mice were treated with recombinant CCL2 the protective effect was abrogated demonstrating the crucial contribution of this chemokine to the protection of A20AEC-KO mice to Influenza A virus infection. Taken together, we propose a mechanism of action by which A20 expression in club cells controls inflammation and antiviral CTL responses in response to influenza virus infection.  相似文献   

17.
Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity   总被引:11,自引:2,他引:9       下载免费PDF全文
Alpha/beta interferon immune defenses are essential for resistance to viruses and can be triggered through the actions of the cytoplasmic helicases retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). Signaling by each is initiated by the recognition of viral products such as RNA and occurs through downstream interaction with the IPS-1 adaptor protein. We directly compared the innate immune signaling requirements of representative viruses of the Flaviviridae, Orthomyxoviridae, Paramyxoviridae, and Reoviridae for RIG-I, MDA5, and interferon promoter-stimulating factor 1 (IPS-1). In cultured fibroblasts, IPS-1 was essential for innate immune signaling of downstream interferon regulatory factor 3 activation and interferon-stimulated gene expression, but the requirements for RIG-I and MDA5 were variable. Each was individually dispensable for signaling triggered by reovirus and dengue virus, whereas RIG-I was essential for signaling by influenza A virus, influenza B virus, and human respiratory syncytial virus. Functional genomics analyses identified cellular genes triggered during influenza A virus infection whose expression was strictly dependent on RIG-I and which are involved in processes of innate or adaptive immunity, apoptosis, cytokine signaling, and inflammation associated with the host response to contemporary and pandemic strains of influenza virus. These results define IPS-1-dependent signaling as an essential feature of host immunity to RNA virus infection. Our observations further demonstrate differential and redundant roles for RIG-I and MDA5 in pathogen recognition and innate immune signaling that may reflect unique and shared biologic properties of RNA viruses whose differential triggering and control of gene expression may impact pathogenesis and infection.  相似文献   

18.
19.
Influenza virus infections are major public health threats due to their high rates of morbidity and mortality. Upon influenza virus entry, host cells experience modifications of endomembranes, including those used for virus trafficking and replication. Here we report that influenza virus infection modifies mitochondrial morphodynamics by promoting mitochondria elongation and altering endoplasmic reticulum-mitochondria tethering in host cells. Expression of the viral RNA recapitulates these modifications inside cells. Virus induced mitochondria hyper-elongation was promoted by fission associated protein DRP1 relocalization to the cytosol, enhancing a pro-fusion status. We show that altering mitochondrial hyper-fusion with Mito-C, a novel pro-fission compound, not only restores mitochondrial morphodynamics and endoplasmic reticulum-mitochondria contact sites but also dramatically reduces influenza replication. Finally, we demonstrate that the observed Mito-C antiviral property is directly connected with the innate immunity signaling RIG-I complex at mitochondria. Our data highlight the importance of a functional interchange between mitochondrial morphodynamics and innate immunity machineries in the context of influenza viral infection.  相似文献   

20.
Toll-like receptors are key participants in innate immune responses   总被引:5,自引:0,他引:5  
During an infection, one of the principal challenges for the host is to detect the pathogen and activate a rapid defensive response. The Toll-like family of receptors (TLRs), among other pattern recognition receptors (PRR), performs this detection process in vertebrate and invertebrate organisms. These type I transmembrane receptors identify microbial conserved structures or pathogen-associated molecular patterns (PAMPs). Recognition of microbial components by TLRs initiates signaling transduction pathways that induce gene expression. These gene products regulate innate immune responses and further develop an antigen-specific acquired immunity. TLR signaling pathways are regulated by intracellular adaptor molecules, such as MyD88, TIRAP/Mal, between others that provide specificity of individual TLR- mediated signaling pathways. TLR-mediated activation of innate immunity is involved not only in host defense against pathogens but also in immune disorders. The involvement of TLR-mediated pathways in auto-immune and inflammatory diseases is described in this review article.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号