首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 328 毫秒
1.
黑色素瘤分化相关基因5(melanoma differentiation-associated gene-5,MDA5)是胞浆内核酸受体,与病原相关分子模式(pathogen-associated molecular patterns,PAMPs)相结合,特异性地识别较长的双链RNA,功能与视黄酸诱导表达基因Ⅰ(retinoic acid-inducude gene Ⅰ,RIG-Ⅰ)相似,通过自身级联激活和招募结构域(CARD)与接头蛋白CARD同源相互作用之后,与接头蛋白线粒体连接蛋白(MAVS)(也叫VISA、Cardif或IPS-1)结合,相互作用后会导致RIG-Ⅰ样受体(RLR)在内膜上重新定位,一边招募来TRAF2/TRAF6活化IKK激酶复合物,从而激活转录因子NF-κB;另一边招募来TRAF3和TBK1,从而促进IRF3的磷酸化激活,活化后的转录因子NF-κB及IRF会进入细胞核,共同协作促进Ⅰ型干扰素基因的表达.  相似文献   

2.
轮状病毒是导致许多哺乳动物严重腹泻的主要病原,它的入侵会导致宿主细胞抗病毒状态的建立,如病毒核酸与RIG-Ⅰ、MDA5、LGP2、dsRNA依赖的蛋白激酶及NLR炎症小体作用,导致IFN的分泌和细胞焦亡等;非结构蛋白NSP4会激活NF-κB信号通路。但轮状病毒也进化出多种策略来对抗宿主的天然免疫,其中以非结构蛋白1(NSP1)研究较多,NSP1可抑制STAT-Y701磷酸化,降解干扰素调节因子、MAVS和β-TrCP等,进而阻止IFN-β和NF-κB信号通路的激活;VP3降解MAVS,拮抗OAS/RNase L通路激活阻止宿主细胞对病毒RNA的应答;VP2、NSP2、NSP3也发挥了一定拮抗天然免疫的作用。论文主要对轮状病毒各蛋白调控宿主天然免疫的机制进行综述,以期为轮状病毒感染防控、新型疫苗研制等提供参考。  相似文献   

3.
口蹄疫病毒前导蛋白为一种木瓜蛋白半胱氨酸酶,用一个半胱氨酸区作为亲和试剂.由于结构上的特殊性,使其能够进行自身切割,从正合成的多聚蛋白中游离自己,并通过切割宿主蛋白eIF的两种相似因子4GⅠ和4GⅡ减弱宿主细胞转录自身mRNA的能力,尤其使α/β干扰素的翻译受到抑制,从而为病毒的复制及感染创造有利条件.文章从结构特点、功能特性方面对其进行阐述.  相似文献   

4.
家禽的天然免疫应答在抵抗病毒感染的过程中起着关键性作用,视黄酸诱导基因-Ⅰ(retinoic acid inducible gene-Ⅰ,RIG-Ⅰ)作为细胞质内一类识别病毒双链RNA的模式识别受体,与天然免疫应答密切相关。它可通过RNA配体结合病原相关分子模式监测细胞质中的病毒RNA,此过程激活了RIG-Ⅰ及下游线粒体抗病毒信号蛋白(MAVS),最终导致干扰素调节因子(IRF3/7)和核因子κB(NF-κB)活化,诱导产生Ⅰ型干扰素等免疫细胞因子,进而使细胞做出相应的抗病毒天然免疫反应。但由于鸡体内缺乏RIG-Ⅰ基因,目前大多将鸭源或鹅源RIG-Ⅰ基因转染鸡成纤维母细胞(DF-1)研究RIG-Ⅰ基因在鸡感染禽类病毒时是否具有免疫功能。文章介绍了RIG-Ⅰ在家禽体内的表达及其介导的抗病毒天然免疫信号通路,并简述了RIG-Ⅰ在家禽体内抗病毒作用的研究概况,为抑制家禽病毒的感染和免疫系统研究,以及研制新型抗病毒疫苗或免疫佐剂等提供参考。  相似文献   

5.
A型流感病毒NS1蛋白的结构与功能   总被引:2,自引:1,他引:1  
A型流感病毒片段8编码的NS1蛋白是病毒复制和传播的重要调节蛋白。它含有3个功能区,即RNA结合区,eIF4GI结合区和效应区。NS1蛋白在A型流感病毒感染细胞中高水平表达,它的主要功能是抑制NF-κB(nuclear factor-κB)活化和IFN(inter-feron)-α/β介导的抗病毒作用,抑制Jun N末端激酶(jun N-terminal kinase,JNK)和AP-1(activating posttranslation)转录因子活化;下调病毒感染细胞凋亡;促进病毒基因表达,抑制宿主mRNA加工和转运。NS1蛋白的深入研究,为今后预防和治疗A型流感病毒寻找突破口。  相似文献   

6.
天然免疫系统在病毒感染早期识别和诱发抗病毒反应中发挥重要作用,宿主模式识别受体(PRRs,如RIG-I、Toll和NOD样等受体)识别病原微生物结构上保守组分病原相关分子模式(PAMPs),进而激活下游级联信号通路,诱导干扰素、细胞因子和促炎性因子等产生,激发抗病毒天然免疫。而病毒通过降解天然免疫信号通路分子或抑制其激活,从而抑制抗病毒应答。口蹄疫病毒(FMDV)通过多种蛋白抑制天然免疫,肖少波团队和杜以军团队鉴定了非结构蛋白Lpro和3Cpro,作为水解酶蛋白,抑制RIG-I通路分子的激活,并阐明其抑制天然免疫的机制;郑海学团队鉴定了结构蛋白VP3和非结构蛋白2B和3A等抑制干扰素产生。  相似文献   

7.
视黄酸诱导基因Ⅰ(RIG-Ⅰ)为RLRs受体家族的成员,是比较关键的细胞质内病原体识别受体,可识别细胞内的单链、双链等RNA病毒成分,被激活的RIG-Ⅰ受体及其CARD在TRIM25的作用下连接泛素链使其寡聚化,通过与线粒体抗病毒信号蛋白(MAVS)相互作用,激活MAVS及下游转录因子IRF3和NF-κB,从而诱导Ⅰ型干扰素和炎性因子的表达,最终介导宿主的抗病毒免疫应答。鉴于RIG-Ⅰ持续激活可导致炎性因子对自身细胞的损伤,因此RIG-Ⅰ样受体信号通路受到宿主严格的调控。而某些病毒为逃避宿主细胞的免疫应答,进化出多种机制靶向调节RIG-Ⅰ及MAVS,从而阻断信号通路。论文从RIG-Ⅰ识别病毒机制、激活下游信号传导、宿主细胞对信号传导途径的调控以及病毒逃避机制等方面重点阐述RIG-Ⅰ所介导的天然免疫反应。  相似文献   

8.
口蹄疫是由口蹄疫病毒(FMDV)引起的急性、热性、高度接触性传染病。口蹄疫病毒感染宿主引起一系列严重的炎症反应,而TLR3通路是介导细胞炎性反应的主要途径之一。为研究口蹄疫病毒蛋白对TLR3通路的影响,本研究首先用双荧光素酶报告系统筛选影响TLR3通路的FMDV蛋白;接着用Q-PCR试验验证筛出来的候选蛋白对TLR3通路下游基因表达水平的影响;并用免疫共沉淀试验验证与候选蛋白有相互作用的TLR3通路蛋白;最后用Western blot试验检测候选蛋白对TLR3通路下游分子磷酸化水平的影响。双荧光素酶报告系统结果显示,口蹄疫病毒3D蛋白促进TLR3通路介导的Ⅰ型干扰素的产生并呈剂量依赖性,Q-PCR试验表明,3D能够促进TLR3通路下游基因表达水平;免疫共沉淀试验表明,FMDV 3D与TLR3有相互作用;Western blot试验进一步显示,过表达3D能够促进TLR3下游分子的磷酸化水平。综上,口蹄疫病毒3D蛋白能促进TLR3介导的Ⅰ型干扰素的产生,从而调控天然免疫反应。  相似文献   

9.
《中国兽医学报》2016,(11):1916-1922
为探讨氧化应激对猪繁殖与呼吸征病毒(PRRSV)体外感染猪肺泡巨噬细胞(PAMs)后诱导TLR3/NF-kB信号转导中相关蛋白的影响,从PRRSV阴性的健康仔猪肺脏中分离和培养PAMs,随后分为正常对照组、PRRSV各试验组、NAC+PRRSV组以及H_2O_2+PRRSV组,于感染后6、12、24、48、72h收集细胞,Western blot检测各组不同时间段PAMs中TLR3、TRIF、TRAF6以及NF-kB蛋白表达水平的动态变化。结果显示,在PRRSV各试验组,TLR3、TRIF、TRAF6及NF-κB蛋白水平表达与正常对照组相比均升高(P0.05),而且其表达量与感染之间有时间相关性;在NAC+PRRSV组中,TLR3、TRIF、TRAF6及NF-κB的蛋白表达虽有升高,但表达量都比正常接毒组明显下降;在H_2O_2+PRRSV组中,TLR3、TRIF、TRAF6及NF-κB的蛋白表达比正常接毒组表达量明显升高(P0.05)。结果表明,抗氧化剂和促氧化剂可引起PAMs氧化应激状态的改变,从而导致PRRSV感染PAMs后TLR3/NF-κB信号传导通路的相关蛋白水平发生了变化。  相似文献   

10.
本研究旨在研究壳聚糖(CTS)对无乳链球菌(S. agalactiae)引起的奶牛乳腺上皮细胞(bMECs)炎性反应的抑制作用和分子机制。试验用含不同浓度(0、15.625、31.250、62.500、125.000、250.000、500.000和1 000.000 mg/mL) CTS的脑心浸出液(BHI)培养基培养S. agalactiae,通过测定600 nm吸光度(OD)检测细菌活性;试验用含不同浓度(0、31.25、125.00和250.00 mg/mL)CTS的细胞培养基培养bMECs 24 h后,用S. agalactiae刺激细胞6 h,使用荧光定量PCR的方法检测白细胞介素-6 (IL-6)、白细胞介素-1β(IL-1β)、肿瘤坏死因子-α(TNF-α)、白细胞介素-8(IL-8)、Toll样受体2(TLR2)、髓样分化因子88(MyD88)、白细胞介素-1受体相关激酶4(IRAK4)、肿瘤坏死因子受体相关因子6(TRAF6)和转化生长因子激酶1(TAK1)的mRNA表达量,使用Western blot的方法检测核因子-κB抑制蛋白-α(IκB-α)、磷酸化核因子-κB-蛋白65(p-NF-κB-p65)、磷酸化蛋白38(p-p38)、磷酸化的细胞外信号调节激酶1/2(p-ERK1/2)和磷酸化c-Jun氨基末端激酶(p-JNK)的蛋白表达量。结果表明:1) CTS呈浓度依赖性抑制S. agalactiae的活性。2)S. agalactiae极显著提高了bMECs的IL-6、IL-1β、TNF-α和IL-8 mRNA表达量(P0.01);31.25、125.00和250.00 mg/mL的CTS显著或极显著降低了S. agalactiae诱导的bMECs的IL-6、IL-1β、TNF-α和IL-8 mRNA表达量(P0.05或P0.01)。3)S. agalactiae极显著提高了bMECs的TLR2、MyD88、IRAK4、TRAF6和TAK1 mRNA表达量(P0.01);31.25、125.00和250.00 mg/mL的CTS显著或极显著降低了S. agalactiae诱导的bMECs的TLR2、MyD88、IRAK4、TRAF6和TAK1 mRNA表达量(P0.05或P0.01)。4)S. agalactiae显著提高了bMECs的IκB-α、 p-NF-κB-p65、 p-p38和p-JNK蛋白表达量(P 0. 05);31.25、125.00和250.00 mg/mL的CTS显著或极显著降低了S. agalactiae诱导的bMECs的IκB-α、p-NF-κB-p65、p-p38、p-ERK1/2和p-JNK蛋白表达量(P0.05或P0.01)。综上所述,CTS可以呈浓度依赖性抑制S. agalactiae活性。CTS通过抑制核因子-κB(NF-κB)和丝裂原活化蛋白激酶(MAPK)信号通路的转导,减少了炎性细胞因子IL-6、IL-1β、TNF-α和IL-8 mRNA表达,从而减弱S. agalactiae诱导的bMECs的炎性损伤作用。  相似文献   

11.
旨在探究宿主蛋白程序性细胞死亡因子10(programmed cell death factor 10,PDCD10)通过抑制Ⅰ型干扰素表达进而促进口蹄疫病毒(foot-and-mouth disease virus,FMDV)的复制。首先,本研究验证了过表达和沉默PDCD10对FMDV复制的影响,接着利用双荧光素酶报告系统探究PDCD10对Ⅰ型干扰素信号通路活化的影响,最后,利用实时荧光定量PCR探究PDCD10对Ⅰ型干扰素通路下游刺激基因(IFN-stimulated genes,ISGs)转录的影响。结果表明,过表达PDCD10显著促进FMDV的复制,沉默PDCD10显著抑制FMDV的复制。与对照相比,过表达PDCD10后感染仙台病毒(Sendai virus,SeV)的细胞培养液上清液显著促进FMDV复制,进一步,PDCD10显著抑制SeV诱导的IFN-β启动子以及NF-κB的激活且呈剂量依赖性,并且PDCD10负调控Ⅰ型干扰素通路信号分子转录,最后还发现PDCD10负调控Ⅰ型干扰素下游ISGs转录。本研究结果为深入探究PDCD10在抗病毒天然免疫中的作用积累了资料。  相似文献   

12.
Natural killer (NK) cells play a role in innate antiviral immunity by directly lysing virus-infected cells and producing antiviral cytokines such as interferon gamma (IFN-γ). We developed a system for characterizing the bovine NK response to foot-and-mouth disease virus (FMDV), which causes a disease of cloven-hoofed animals and remains a threat to livestock industries throughout the world. IL-2 stimulation of PBMC resulted in poor killing of human K562 cells, which are often used as NK target cells, while lysis of the bovine BL3.1 cell line was readily detected. Depletion of NKp46-expressing cells revealed that 80% of the killing induced by IL-2 could be attributed to NKp46+ cells. In order to characterize the response of NK cells against FMDV in vivo, we infected groups of cattle with three different strains of the virus (A24 Cruzeiro, O1 Manisa, O Hong Kong) and evaluated the cytolytic ability of NK cells through the course of infection. We consistently observed a transient increase in cytolysis, although there was variation in magnitude and kinetics. This increase in cytolysis remained when CD3+ cells were removed from the preparation of lymphocytes, indicating that cytolysis was independent of MHC-T cell receptor interaction or γδ T cell activation. In contrast, animals monitored following vaccination against FMDV did not exhibit any increase in NK killing. These data suggest that NK cells play a role in the host immune response of cattle against FMDV, and contrast with the suppression of NK activity previously observed in swine infected with FMDV.  相似文献   

13.
The Exaltation of Newcastle disease virus (END) phenomenon is induced by the inhibition of type I interferon in pestivirus-infected cells in vitro, via proteasomal degradation of cellular interferon regulatory factor (IRF)-3 with the property of the viral autoprotease protein Npro. Reportedly, the amino acid residues in the zinc-binding TRASH motif of Npro determine the difference in characteristics between END-phenomenon-positive (END+) and END-phenomenon-negative (END) classical swine fever viruses (CSFVs). However, the basic mechanism underlying this function in bovine viral diarrhea virus (BVDV) has not been elucidated from the genomic differences between END+ and END viruses using reverse genetics till date. In the present study, comparison of complete genome sequences of a pair of END+ and END viruses isolated from the same virus stock revealed that there were only four amino acid substitutions (D136G, I2623V, D3148G and D3502Y) between two viruses. Based on these differences, viruses with and without mutations at these positions were generated using reverse genetics. The END assay, measurements of induced type I interferon and IRF-3 detection in cells infected with these viruses revealed that the aspartic acid at position 136 in the zinc-binding TRASH motif of Npro was required to inhibit the production of type I interferon via the degradation of cellular IRF-3, consistently with CSFV.  相似文献   

14.
Bungowannah virus is the most divergent atypical pestivirus that had been detected up to now, and does not fit into any of the four approved species: Bovine viral diarrhea virus type 1 (BVDV-1) and type 2 (BVDV-2), Classical swine fever virus (CSFV) and Border disease virus (BDV). However, the presence of Npro and Erns coding regions, which are unique to pestiviruses, provides clear evidence of a pestivirus. Nevertheless, the amino acid identity of Bungowannah virus Npro and BVDV-1 Npro (strain CP7) is only 51.5%. By using a BVDV-1 backbone, a novel chimeric construct was generated, in which the genomic region encoding the non-structural protein Npro was replaced by that of Bungowannah virus (CP7_Npro-Bungo). In vitro studies of CP7_Npro-Bungo revealed autonomous replication with the same efficacy as the BVDV backbone CP7 and infectious high-titer virus could be collected. In order to compare the ability of interferon (IFN) suppression, two reporter gene assays, specific for type-I IFN, were carried out. In virus-infected cells, no significant difference in blocking of IFN expression between the parental virus CP7, Bungowannah virus and the chimeric construct CP7_Npro-Bungo could be detected. In contrast, an Npro deletion mutant showed an impaired replication in bovine cells and a marked type-I IFN response.Taken together, our findings reveal the compatibility of non-structural protein Npro of atypical Bungowannah virus with a BVDV type 1 backbone and its characteristic feature as an inhibitor of type-I IFN induction with an inhibitor-activity comparable to other pestiviruses.  相似文献   

15.
口蹄疫病毒(FMDV)3C蛋白酶是FMDV基因组编码中具有酶学活性的病毒产物之一,在FMDV编码蛋白的成熟和子代病毒在宿主细胞体内大量扩增中发挥着重要作用。3C蛋白酶能剪切多聚蛋白,降解特定的蛋白质,是宿主细胞中重要的毒力因子。3C蛋白酶能调控蛋白的转录和翻译,使宿主细胞内的干扰素等多种抗病毒基因低水平表达,使FMDV逃避宿主的天然免疫。论文主要综述了FMDV 3C蛋白酶的结构、生物学功能,并介绍了其在研制新型疫苗中的应用,以期为今后FMDV 3C蛋白酶的研究、新型疫苗的研发提供参考。  相似文献   

16.
Classical swine fever (CSF) caused by CSF virus (CSFV) is a highly contagious disease of pigs. The viral protein Npro of CSFV interferes with alpha- and beta-interferon (IFN-α/β) induction by promoting the degradation of interferon regulatory factor 3 (IRF3). During the establishment of the live attenuated CSF vaccine strain GPE-, Npro acquired a mutation that abolished its capacity to bind and degrade IRF3, rendering it unable to prevent IFN-α/β induction. In a previous study, we showed that the GPE- vaccine virus became pathogenic after forced serial passages in pigs, which was attributed to the amino acid substitutions T830A in the viral proteins E2 and V2475A and A2563V in NS4B. Interestingly, during the re-adaptation of the GPE- vaccine virus in pigs, the IRF3-degrading function of Npro was not recovered. Therefore, we examined whether restoring the ability of Npro to block IFN-α/β induction of both the avirulent and moderately virulent GPE--derived virus would enhance pathogenicity in pigs. Viruses carrying the N136D substitution in Npro regained the ability to degrade IRF3 and suppress IFN-α/β induction in vitro. In pigs, functional Npro significantly reduced the local IFN-α mRNA expression in lymphoid organs while it increased quantities of IFN-α/β in the circulation, and enhanced pathogenicity of the moderately virulent virus. In conclusion, the present study demonstrates that functional Npro influences the innate immune response at local sites of virus replication in pigs and contributes to pathogenicity of CSFV in synergy with viral replication.  相似文献   

17.
本试验旨在建立一种用SYBR GreenⅠ荧光染料检测HeLa细胞Ⅰ型干扰素效应因子ISG15、ISG56、Mx1、OAS和PKR mRNA表达水平的实时荧光定量RT-PCR检测方法,并在口蹄疫病毒(FMDV)L蛋白抑制Ⅰ型IFN发挥效应的信号通路中进行初步应用。利用TRIzol法提取总RNA,经Oligo d(T)15进行反转录,利用PCR扩增各段目的基因,并克隆至pMD18-T载体,转化大肠杆菌DH5α,经鉴定为阳性的重组质粒作为标准品模板建立SYBR GreenⅠ荧光定量RT-PCR标准曲线和熔解曲线,并进行灵敏性、特异性和重复性试验。根据建立的实时荧光定量RT-PCR方法,检测FMDV L蛋白对Ⅰ型IFN效应因子的抑制效果。HeLa细胞在转染FMDV L蛋白真核表达质粒,并受到Ⅰ型IFN刺激后ISG15、ISG56、Mx1、OAS和PKR的相对表达量较转染空载体或表达GST的真核表达质粒明显降低。本试验建立了HeLa细胞Ⅰ型IFN效应因子的实时荧光定量RT-PCR检测方法,为在mRNA水平上对HeLa细胞Ⅰ型IFN效应因子的定量分析奠定了基础,并成功地初步应用于FMDV L蛋白抑制Ⅰ型IFN发挥效应的信号通路的研究中。  相似文献   

18.
通过构建蓝舌病毒(BTV)NS4基因真核表达载体pcDNA3.1-NS4-eGFP,转染HEK-293T细胞,利用Western blot及荧光显微镜分析NS4蛋白的表达与亚细胞定位特征;pcDNA3.1-NS4-eGFP转染的HEK-293T细胞添加20 HAU/mL仙台病毒(SeV)刺激后,qRT-PCR法分析NS4基因表达对SeV诱导的上游识别基因RIG-Ⅰ、MDA5、VISA、TBK1、IKKε、IRF3、TRAF3、TRAF6、IRF9、干扰素基因(IFN-α、IFN-β)以及干扰素刺激基因ISG15和USP18的mRNA表达水平的影响。在HEK-293T细胞内转染pcDNA3.1-NS4-eGFP质粒24 h后,分别添加20 HAU/mL SeV刺激24,48 h,qRT-PCR结果表明,细胞内表达NS4-EGFP后,RIG-Ⅰ、MDA5、TRAF6、IRF9、ISG15及IFN-β基因mRNA表达极显著下降,随着SeV诱导时间的延长,VISA、TBK1、IKKε、USP18基因mRNA表达差异呈不显著趋势。本研究成功构建BTV NS4基因真核表达载体pcDNA3.1-NS4-eGFP,NS4-EGFP融合蛋白在HEK-293T细胞中主要分布于细胞核周围及细胞核内。BTV NS4基因在HEK-293T细胞内的表达显著下调SeV诱导的IFN信号通路相关基因RIG-Ⅰ、MDA5、TRAF6、IRF9、ISG15和IFN-β的表达,为进一步探究NS4基因在BTV拮抗宿主细胞免疫应答中的机制奠定基础。  相似文献   

19.
The nucleotide sequence of the VP1 (1D) and partial 3D polymerase (3Dpol) coding regions of the foot and mouth disease virus (FMDV) vaccine strain A/Iran87, a highly passaged isolate (~150 passages), was determined and aligned with previously published FMDV serotype A sequences. Overall analysis of the amino acid substitutions revealed that the partial 3Dpol coding region contained four amino acid alterations. Amino acid sequence comparison of the VP1 coding region of the field isolates revealed deletions in the highly passaged Iranian isolate (A/Iran87). The prominent G-H loop of the FMDV VP1 protein contains the conserved arginine-glycine-aspartic acid (RGD) tripeptide, which is a well-known ligand for a specific cell surface integrin. Despite losing the RGD sequence of the VP1 protein and an Asp26→Glu substitution in a beta sheet located within a small groove of the 3Dpol protein, the virus grew in BHK 21 suspension cell cultures. Since this strain has been used as a vaccine strain, it may be inferred that the RGD deletion has no critical role in virus attachment to the cell during the initiation of infection. It is probable that this FMDV subtype can utilize other pathways for cell attachment.  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