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 31 毫秒
1.
We assessed the prediction that access of the viral NS1 protein to cellular PDZ domain protein networks enhances the virulence of highly pathogenic avian influenza A viruses. The NS1 proteins of most avian influenza viruses bear the C-terminal ligand sequence Glu-Ser-Glu-Val (ESEV) for PDZ domains present in multiple host proteins, whereas no such motif is found in the NS1 homologues of seasonal human virus strains. Previous analysis showed that a C-terminal ESEV motif increases viral virulence when introduced into the NS1 protein of mouse-adapted H1N1 influenza virus. To examine the role of the PDZ domain ligand motif in avian influenza virus virulence, we generated three recombinants, derived from the prototypic H5N1 influenza A/Vietnam/1203/04 virus, expressing NS1 proteins that either have the C-terminal ESEV motif or the human influenza virus RSKV consensus or bear a natural truncation of this motif, respectively. Cell biological analyses showed strong control of NS1 nuclear migration in infected mammalian and avian cells, with only minor differences between the three variants. The ESEV sequence attenuated viral replication on cultured human, murine, and duck cells but not on chicken fibroblasts. However, all three viruses caused highly lethal infections in mice and chickens, with little difference in viral titers in organs, mean lethal dose, or intravenous pathogenicity index. These findings demonstrate that a PDZ domain ligand sequence in NS1 contributes little to the virulence of H5N1 viruses in these hosts, and they indicate that this motif modulates viral replication in a strain- and host-dependent manner.The transmission of highly pathogenic avian influenza A viruses (HPAIV) of the H5N1 subtype to humans since the year 1997 has caused a high mortality rate of almost 60% (62). Patients infected with H5N1 influenza virus developed mainly severe respiratory disease, characterized by fever, cough, shortness of breath, and pneumonia, that frequently progressed to acute respiratory distress syndrome (ARDS) and multiorgan failure (28, 68, 69). In fatal cases, the median time from onset to death was 9 to 10 days (1). Systemic spread (18) and hypercytokinemia (11) have been described as possible disease-aggravating factors of HPAIV-H5N1 viruses, but the reasons for their high virulence in humans are incompletely understood.Due to the potential pandemic threat presented by H5N1 viruses, there is great interest in the identification of viral virulence determinants and their mode of action. This is critical not only for a better understanding of the pathogenic mechanisms induced by these viruses but also for the development of new drugs to treat the infections. The high virulence of HPAIV-H5N1 isolates in the avian host correlates with the presence of a polybasic cleavage site in the hemagglutinin (HA), facilitating its intracellular cleavage by furin-like proteases (27, 50). Further, amino acid substitutions in the PA protein (T515A) (30) and in the NS1 protein (V149A) (40) have been reported to regulate the virulence of corresponding HPAIV-H5N1 isolates in ducks and chickens. The known molecular determinants of virulence in mammalian hosts also include the polybasic cleavage site in the HA (23) and several polymorphisms in the PB2 polymerase subunit and the proapoptotic PB1-F2 protein. Thus, a serine residue at position 66 in the PB1-F2 protein increased viral replication and decreased survival in the mouse model (9). Also, specific amino acid polymorphisms within PB2 (E627K or D701N) can increase virulence in mice (23, 39) and viral replication in mammalian cells (7, 57, 58). Furthermore, the nonstructural NS1 protein, which has a major function in the inhibition of type I interferon (IFN) (17, 19) and in the limitation of the antiviral effects of IFN-induced proteins, including PKR (4, 22), OAS/RNase L (45), and RIG-I (16, 48, 63, 64), contributes to virulence in mammals (34, 55).The domain structure of the NS1 protein is well characterized; it includes an N-terminal RNA binding and dimerization domain and a nuclear localization signal (NLS) at positions 34 to 38 (summarized in reference 19). The NS1 proteins of most human strains circulating between 1950 and 1986 also contain a second NLS at positions 219 to 227 (NLS-2), which includes four conserved basic amino acids (K219, R220, R224, R227) (44). A large-scale sequence analysis showed that the NS1 proteins of avian and human influenza viruses differ in their C-terminal sequences, indicating possible differences in the associated activity (46). Among most high- and low-pathogenicity avian influenza viruses, the last four NS1 amino acids consist of the conserved sequence ESEV (3,007 of 3,692 isolates described in the NCBI database [3]), while for the majority of seasonal human influenza viruses, the motif RSKV is typical (1,911 of 2,713 isolates). Significantly, only the NS1 protein carrying the “avian” ESEV motif interacted in vitro with 24 cellular factors carrying a PDZ (postsynaptic density protein 95, Drosophila disc large tumor suppressor, and zonula occludens 1 protein) domain. The human genome encodes at least 214 proteins containing one or more of these protein interaction modules that recognize short peptide motifs, which are most often present at the C termini of their targets (36, 38). Many PDZ domain proteins have been shown to mediate the formation and localization of higher-order complexes and to participate in various cellular signaling events regulating, for instance, cell polarity and neuronal function (31). Therefore, it was hypothesized that the abundant expression of “avian” NS1 protein capable of interacting with human PDZ domains could possibly disturb their function and aggravate disease severity in H5N1 infections (46). However, there is only limited experimental support for the universal validity of this hypothesis. The grafting of the “avian” ESEV sequence into the C terminus of NS1 protein expressed by mouse-adapted influenza A/WSN/33 virus (H1N1) decreased the mean lethal dose by about 1 order of magnitude (32). Still, it is not clear to what extent this motif contributes to the virulence of HPAIV-H5N1 and other natural influenza A viruses in avian and mammalian hosts.The goal of the present study was to elucidate the role of the C-terminal NS1 motif in viral replication and disease caused by the prototypic influenza A/Vietnam/1203/04 (VN/1203) virus, isolated in a fatal human case (60). This virus expresses an NS1 protein that is very similar or identical at positions 1 to 215 to homologues expressed by other HPAIV-H5N1 strains but naturally lacks the 10 C-terminal amino acids (aa), including the terminal ESEV motif, due to a premature stop codon (Fig. (Fig.1).1). We used reverse genetics to produce a recombinant VN/1203 wild-type (WT) virus and two variants with reconstituted NS1 C termini ending either with the “avian” ESEV or with the “human” RSKV sequence. Experimental infections of mice and chickens revealed that all three viruses caused highly lethal infections in both species, with only moderate differences in viral titers in the organs of the mice. Thus, we show that the C-terminal ESEV motif of the NS1 protein contributes little to the virulence of H5N1 viruses in mice and chickens, and we suggest that this motif modulates viral virulence in a strain- and host-dependent manner.Open in a separate windowFIG. 1.Growth kinetics of recombinant VN/1203 viruses expressing WT or elongated NS1 proteins in human, murine, and avian cells. (A) Scheme of the viral VN/1203-NS1 protein with the RNA binding domain and the nuclear localization signals (NLS) at positions 34 to 38 and 214 to 225 indicated. Amino acids involved in NLS2 function are underlined. The C-terminal sequences of the WT and elongated mutant NS1 proteins are given, and the PL motif is shown in boldface. (B to E) Human A549 alveolar cells, murine NIH 3T3 fibroblasts, chicken embryo fibroblasts (CEFs), or EFB-R1 duck embryo fibroblasts (DEFs) were infected with recombinant VN/1203-WT, -ESEV, or -RSKV viruses at an MOI of 0.001. Aliquots of supernatants were harvested at the indicated time points, and samples were titrated by plaque assays in MDCK cells. (F) Human A549 cells were infected at an MOI of 2, and virus titers in supernatants taken at the indicated time points were determined by plaque assays. Results are averages for at least two independent experiments with biological duplicates. Error bars indicate standard deviations.  相似文献   

2.
NS1 Protein of Influenza A Virus Down-Regulates Apoptosis   总被引:21,自引:0,他引:21       下载免费PDF全文
Wild-type (WT) influenza A/PR/8/34 virus and its variant lacking the NS1 gene (delNS1) have been compared for their ability to mediate apoptosis in cultured cells and chicken embryos. Cell morphology, fragmentation of chromatin DNA, and caspase-dependent cleavage of the viral NP protein have been used as markers for apoptosis. Another marker was caspase cleavage of the viral M2 protein, which was also found to occur in an apoptosis-specific manner. In interferon (IFN)-competent host systems, such as MDCK cells, chicken fibroblasts, and 7-day-old chicken embryos, delNS1 virus induced apoptosis more rapidly and more efficiently than WT virus. As a consequence, delNS1 virus was also more lethal for chicken embryos than WT virus. In IFN-deficient Vero cells, however, apoptosis was delayed and developed with similar intensity after infection with both viruses. Taken together, these data indicate that the IFN antagonistic NS1 protein of influenza A viruses has IFN-dependent antiapoptotic potential.  相似文献   

3.
Domestic poultry serve as intermediates for transmission of influenza A virus from the wild aquatic bird reservoir to humans, resulting in influenza outbreaks in poultry and potential epidemics/pandemics among human beings. To combat emerging avian influenza virus, an inexpensive, heat-stable, and orally administered influenza vaccine would be useful to vaccinate large commercial poultry flocks and even migratory birds. Our hypothesized vaccine is a recombinant attenuated bacterial strain able to mediate production of attenuated influenza virus in vivo to induce protective immunity against influenza. Here we report the feasibility and technical limitations toward such an ideal vaccine based on our exploratory study. Five 8-unit plasmids carrying a chloramphenicol resistance gene or free of an antibiotic resistance marker were constructed. Influenza virus was successfully generated in avian cells transfected by each of the plasmids. The Salmonella carrier was engineered to allow stable maintenance and conditional release of the 8-unit plasmid into the avian cells for recovery of influenza virus. Influenza A virus up to 107 50% tissue culture infective doses (TCID50)/ml were recovered from 11 out of 26 co-cultures of chicken embryonic fibroblasts (CEF) and Madin-Darby canine kidney (MDCK) cells upon infection by the recombinant Salmonella carrying the 8-unit plasmid. Our data prove that a bacterial carrier can mediate generation of influenza virus by delivering its DNA cargoes into permissive host cells. Although we have made progress in developing this Salmonella influenza virus vaccine delivery system, further improvements are necessary to achieve efficient virus production, especially in vivo.  相似文献   

4.
We used the yeast interaction trap system to identify a novel human 70-kDa protein, termed NS1-binding protein (NS1-BP), which interacts with the nonstructural NS1 protein of the influenza A virus. The genetic interaction was confirmed by the specific coprecipitation of the NS1 protein from solution by a glutathione S-transferase–NS1-BP fusion protein and glutathione-Sepharose. NS1-BP contains an N-terminal BTB/POZ domain and five kelch-like tandem repeat elements of ~50 amino acids. In noninfected cells, affinity-purified antibodies localized NS1-BP in nuclear regions enriched with the spliceosome assembly factor SC35, suggesting an association of NS1-BP with the cellular splicing apparatus. In influenza A virus-infected cells, NS1-BP relocalized throughout the nucleoplasm and appeared distinct from the SC35 domains, which suggests that NS1-BP function may be disturbed or altered. The addition of a truncated NS1-BP mutant protein to a HeLa cell nuclear extract efficiently inhibited pre-mRNA splicing but not spliceosome assembly. This result could be explained by a possible dominant-negative effect of the NS1-BP mutant protein and suggests a role of the wild-type NS1-BP in promoting pre-mRNA splicing. These data suggest that the inhibition of splicing by the NS1 protein may be mediated by binding to NS1-BP.  相似文献   

5.
NS1蛋白(non—structural protein1)是A型流感病毒重要的非结构蛋白,作为流感病毒的致病因子,NS1通过多种方式增强病毒的致病性和毒力。就H5N1禽流感病毒NS1蛋白的结构与功能进行了综述。  相似文献   

6.
7.
目的:对2013年3月发生的感染人的新型H7N9亚型禽流感病毒的非结构蛋白1(NS1)基因序列进行同源性分析,构建NS1重组质粒并表达。方法:从GenBank获得2006~2013年不同来源的H7N9亚型病毒NS1序列,并进行同源性比较;利用PCR方法从H7N9亚型禽流感病毒株A/Shanghai/4664T/2013(H7N9)基因组cDNA中扩增得到全长NS1基因,并将该片段定向克隆到原核表达载体pET28a上,构建重组质粒pET28a-NS1,经酶切鉴定,将重组质粒转化大肠杆菌BL21(DE3)感受态细胞后,IPTG诱导表达,且进行Western印迹分析。结果:经序列分析,2013年暴发的H7N9型禽流感病毒的NS1基因核苷酸序列同源性为95%~100%,与之前暴发的H7N9型流感病毒NS1基因序列的同源性为86.4%~90.7%,表明2次暴发的该型流感分离株属于不同的进化分支;PCR扩增得到约680 bp的NS1基因序列,所克隆的NS1基因在原核细胞中的表达产物主要以包涵体形式存在,SDS-PAGE检测结果表明重组蛋白相对分子质量为25×103,Western印迹分析证实表达产物为H7N9禽流感病毒NS1蛋白。结论:为进一步研究H7N9亚型流感病毒NS1蛋白功能及基于NS1蛋白的抗病毒药物奠定了基础。  相似文献   

8.
9.
H5N1亚型禽流感病毒NS1基因在昆虫细胞中的表达   总被引:5,自引:0,他引:5  
将H5N1亚型禽流感病毒(AIV)NS1基因插入到杆状病毒转移载体pFastBac1中,获得重组转移载体pFastBac1- NS1。将pFastBac1- NS1转化到DH10Bac感受态细胞中,筛选到重组转座子rBacmid-NS1。在脂质体转染试剂介导下将rBacmid-NS1转染对数生长期的Sf9昆虫细胞获得重组杆状病毒rBV-NS1。rBV-NS1感染Sf9细胞后,通过SDS-PAGE、Western blot和ELISA分析表明:获得了分子量为26ku的特异性NS1蛋白;并且该蛋白可与H5N1 AIV攻毒鸭的血清发生特异性免疫反应,而不能与H5N1AIV灭活疫苗免疫鸭的血清发生反应。试验结果表明:NS1在Sf9昆虫细胞中获得了高效表达,具有与天然蛋白相似的免疫活性,并可以作为区分免疫及自然感染个体的鉴别诊断抗原。本实验为建立禽流感病毒自然感染家禽与禽流感灭活苗免疫家禽的鉴别诊断方法奠定基础。  相似文献   

10.
A型流感病毒NS1蛋白羧基端4个氨基酸可以与PDZ结构域(the domain of PSD95,Dig and ZO-1)相结合,称为PL结构域(PDZ ligand domain).对不同亚型或毒株的流感病毒而言,其NS1蛋白PL结构域的组成存在比较大的差异.有研究发现这种差异能够影响NS1与宿主细胞蛋白的相互作用进而影响病毒的致病力.为进一步探讨PL结构域对NS1蛋白生物学特性的影响,首先构建出4种不同亚型流感病毒(H1N1、H3N2、H5N1、H9N2)来源的NS1绿色荧光蛋白表达质粒.在此基础上,对野生型H3N2病毒NS1表达质粒进行人工改造,将其PL结构域缺失或者替换为其他亚型流感病毒的PL结构域,制备出4种重组NS1蛋白表达质粒.通过比较上述不同NS1蛋白在HeLa细胞中的定位情况发现,只有野生型H3N2病毒的NS1蛋白可以定位于核仁当中,而野生型H1N1、H5N1、H9N2病毒的NS1蛋白以及PL结构域缺失或替代的H3N2病毒NS1蛋白都不能定位于核仁.而通过比较上述NS1蛋白在流感病毒易感的MDCK细胞中的定位,进一步发现所有这些蛋白均不定位于核仁.上述结果表明:PL结构域的不同可以明显影响NS1蛋白在HeLa细胞核内的定位和分布,这有可能造成其生物学功能的差异.同时,NS1蛋白在细胞核内的定位还与宿主细胞的来源有着密切关系.  相似文献   

11.
禽流感病毒NS1蛋白对细胞的影响   总被引:1,自引:0,他引:1  
NS1蛋白为流感病毒非结构蛋白,只在病毒侵入宿主细胞后产生.目前NS1蛋白对细胞整体水平上的作用仍不清楚,为了解NS1蛋白在病毒感染细胞中的作用,构建了重组质粒pCMV-myc-NS1并将其转染A549细胞,利用双向电泳技术检测了受NS1蛋白调控的宿主蛋白,以期从蛋白质组水平上研究禽流感病毒与宿主细胞间的相互作用.同时,还检测了转染NS1对细胞增殖和细胞周期的影响.结果显示,NS1在细胞中的表达,能够明显引起宿主细胞代谢的变化,并通过阻滞细胞周期的正常进行而减缓细胞的增殖.  相似文献   

12.
A short model genome RNA and also the genome RNA of influenza A virus bearing both 5′- and 3′-terminal common sequences activated the interferon-induced double-stranded-RNA-dependent protein kinase, PKR, by stimulating autophosphorylation in vitro. The activated PKR catalyzed phosphorylation of the alpha subunit of eucaryotic translation initiation factor 2 (eIF2α). The NS1 protein efficiently eliminated the PKR-activating activity of these RNAs by binding to them. Two mutant NS1 proteins, each harboring a single amino acid substitution at different regions, exhibited temperature sensitivity in their RNA binding activity in the mutant virus-infected cell lysates as well as when they were prepared as fusion proteins expressed in bacteria. The virus strains carrying these mutant NS1 proteins exhibited temperature sensitivity in virus protein synthesis at the translational level, as reported previously, and could not repress the autophosphorylation of PKR developing during the virus growth, which is normally suppressed by a viral function(s). As a result, the level of eIF2α phosphorylation was elevated 2.5- to 3-fold. The defect in virus protein synthesis was well correlated with the level of phosphorylation of PKR and eIF2α.  相似文献   

13.
通过RT-PCR的方法克隆H5N1亚型禽流感病毒NS1基因,并构建了真核表达载体pCMV-Myc/NS1。将此真核表达质粒转染肺腺癌细胞A549,48 h后,经Western印迹检测,NS1基因能在细胞中正确表达。经荧光显微镜、透射电镜观察和流式细胞仪检测,发现该株流感病毒的NS1蛋白可诱导肺腺癌细胞A549凋亡。  相似文献   

14.
Influenza A viruses can adapt to new host species, leading to the emergence of novel pathogenic strains. There is evidence that highly pathogenic viruses encode for non-structural 1 (NS1) proteins that are more efficient in suppressing the host immune response. The NS1 protein inhibits type-I interferon (IFN) production partly by blocking the TRIM25 ubiquitin E3 ligase-mediated Lys63-linked ubiquitination of the viral RNA sensor RIG-I, required for its optimal downstream signaling. In order to understand possible mechanisms of viral adaptation and host tropism, we examined the ability of NS1 encoded by human (Cal04), avian (HK156), swine (SwTx98) and mouse-adapted (PR8) influenza viruses to interact with TRIM25 orthologues from mammalian and avian species. Using co-immunoprecipitation assays we show that human TRIM25 binds to all tested NS1 proteins, whereas the chicken TRIM25 ortholog binds preferentially to the NS1 from the avian virus. Strikingly, none of the NS1 proteins were able to bind mouse TRIM25. Since NS1 can inhibit IFN production in mouse, we tested the impact of TRIM25 and NS1 on RIG-I ubiquitination in mouse cells. While NS1 efficiently suppressed human TRIM25-dependent ubiquitination of RIG-I 2CARD, NS1 inhibited the ubiquitination of full-length mouse RIG-I in a mouse TRIM25-independent manner. Therefore, we tested if the ubiquitin E3 ligase Riplet, which has also been shown to ubiquitinate RIG-I, interacts with NS1. We found that NS1 binds mouse Riplet and inhibits its activity to induce IFN-β in murine cells. Furthermore, NS1 proteins of human but not swine or avian viruses were able to interact with human Riplet, thereby suppressing RIG-I ubiquitination. In conclusion, our results indicate that influenza NS1 protein targets TRIM25 and Riplet ubiquitin E3 ligases in a species-specific manner for the inhibition of RIG-I ubiquitination and antiviral IFN production.  相似文献   

15.
应用DNAStar软件,参照Genbank中注册的AIV H5亚型毒株NS1基因序列,设计了一对引物,用RTPCR方法成功地扩增出带双酶切位点的H5亚型AIV的NS1基因,通过BamH Ⅰ和EcoR Ⅰ双酶切位点将H5NS1基因插入转移质粒裁体pFastbac HTa中,获得重组转移载体pFastbac HTa-H5NS1并将其转化DH10 Bac细胞,与Bacmid发生位点特异性转座作用,得到重组穿梭载体Bacmid-H5NS1,再将其转染昆虫细胞High Five,PCR鉴定证实该基因正确地插入到病毒基因组的多角体蛋白基因启动子下游,经过SDS-PAGE和Western Blot检测,NS1基因在High Five细胞中得到了表达,H5NS1大小约为28kD,而且表达的产物具有特异免疫学反应性.  相似文献   

16.
The NS1A protein of influenza A virus binds the cellular CPSF30 protein, thereby inhibiting the 3′-end processing of all cellular pre-mRNAs, including beta interferon pre-mRNA. X-ray crystallography identified the CPSF30-binding pocket on the influenza virus A/Udorn/72 (Ud) NS1A protein and the critical role of two hydrophobic NS1A amino acids outside the pocket, F103 and M106, in stabilizing the CPSF30-NS1A complex. Although the NS1A protein of the 1997 H5N1 influenza A/Hong Kong/483/97 (HK97) virus contains L (not F) at position 103 and I (not M) at position 106, it binds CPSF30 in vivo to a significant extent because cognate (HK97) internal proteins stabilize the CPSF30-NS1A complex in infected cells. Here we show that the cognate HK97 polymerase complex, containing the viral polymerase proteins (PB1, PB2, and PA) and the nucleocapsid protein (NP), is responsible for this stabilization. The noncognate Ud polymerase complex cannot carry out this stabilization, but it can stabilize CPSF30 binding to a mutated (F103L M106I) cognate Ud NS1A protein. These results suggested that the viral polymerase complex is an integral component of the CPSF30-NS1A protein complex in infected cells even when the cognate NS1A protein contains F103 and M106, and we show that this is indeed the case. Finally, we show that cognate PA protein and NP, but not cognate PB1 and PB2 proteins, are required for stabilizing the CPSF30-NS1A complex, indicating that the NS1A protein interacts primarily with its cognate PA protein and NP in a complex that includes the cellular CPSF30 protein.  相似文献   

17.
18.
19.
研制了基因工程靶向融合蛋白XE-TNFαm2.其中,XE为HIV/SIV辅助受体CXCR4的第二胞外域;TNFαm2是经突变改型的TNFα,其毒副作用已降低18倍,已用于临床治疗恶性肿瘤.已有的研究表明XE-TNFαm2的功能之一是杀灭受HIV/SIV感染的细胞、但不杀伤未受HIV/SIV感染的正常细胞.探讨XE-TNFαm2可否引起细胞的凋亡,以阐明其杀伤细胞作用的可能机制.结果表明,XE-TNFαm2只能引起受HIV/SIV感染细胞的凋亡,但不能引起未受HIV/SIV感染的正常细胞的凋亡.这一结果表明XE-TNFαm2是一种可特异杀灭受HIV/SIV感染细胞的准确靶向融合蛋白,其毒副作用己最小化.  相似文献   

20.
The NS1 protein of influenza A virus is known to downregulate apoptosis early in infection in order to support virus replication (O. P. Zhirnov, T. E. Konakova, T. Wolff, and H. D. Klenk, J. Virol. 76:1617–1625, 2002). In the present study, we analyzed the development of autophagy, another mechanism to protect cells from degradation that depends on NS1 expression. To this end, we compared autophagy in cells infected with wild-type (WT) influenza virus and virus lacking the NS1 gene (delNS1 virus). The results show that in WT-infected cells but not in delNS1 virus-infected cells, synthesis of the autophagy marker LC3-II, the lipidated form of microtubule light chain-associated protein LC3, is stimulated and that LC3-II accumulates in a perinuclear zone enriched with double-layered membrane vesicles characteristic of autophagosomes. Transfection experiments revealed that NS1 expressed alone was unable to upregulate autophagy, whereas hemagglutinin (HA) and M2 were. Proteolytic cleavage of HA increased autophagy. Taken together, these observations indicate that NS1 stimulates autophagy indirectly by upregulating the synthesis of HA and M2. Thus, it appears that NS1, besides downregulating apoptosis, is involved in upregulation of autophagy and that it supports the survival of infected cells by both mechanisms.  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