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 0 毫秒
1.
Conserved tryptophan-187 facilitates homodimerization of the influenza A virus NS1 protein effector domain. We generated a mutant influenza virus strain expressing NS1-W187R to destabilize this self-interaction. NS1-W187R protein exhibited lower double-stranded RNA (dsRNA)-binding activity, showed a temporal redistribution during infection, and was minimally compromised for interferon antagonism. The mutant virus replicated similarly to the wild type in vitro, but it was slightly attenuated for replication in mice, causing notably reduced morbidity and mortality. These data suggest biological relevance for the W187-mediated homotypic interaction of NS1.  相似文献   

2.
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   

3.
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   

4.
Although an effective interferon antagonist in human and avian cells, the novel H7N9 influenza virus NS1 protein is defective at inhibiting CPSF30. An I106M substitution in H7N9 NS1 can restore CPSF30 binding together with the ability to block host gene expression. Furthermore, a recombinant virus expressing H7N9 NS1-I106M replicates to higher titers in vivo, and is subtly more virulent, than the parental virus. Natural polymorphisms in H7N9 NS1 that enhance CPSF30 binding may be cause for concern.  相似文献   

5.
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   

6.
7.
8.
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   

9.
10.
11.
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   

12.
A型流感病毒的NS1(Nonstructurol 1 protein,NS1)蛋白是病毒复制、毒力等的重要调节蛋白.运用RT-PCR方法扩增A/Beijing/501/2009(H1N1)流感病毒NS1基因,克隆至真核表达载体pCMV-HA,用Lipofectamine2000将线性化pCMV-HA-NS1与neo基因共同转染A549细胞,通过G418筛选获得阳性重组细胞,并采用PCR、RT-PCR、Western blot技术检测重组细胞中NS1蛋白的表达,通过免疫荧光技术观察NS1蛋白在细胞中的定位.PCR、RT-PCR检测显示NS1基因成功整合进入细胞基因组,并转录为mRNA;Western blot检测显示重组细胞系稳定表达NS1蛋白,免疫荧光显示NS1蛋白定位于细胞核内.表明通过G418筛选,成功构建稳定表达NS1蛋白的重组A549-HA-NS1细胞系,且NS1蛋白定位于细胞核内,为进一步研究NS1蛋白的生物学功能奠定基础.  相似文献   

13.
The influenza A virus NS1 protein, a virus-encoded alpha/beta interferon (IFN-alpha/beta) antagonist, appears to be a key regulator of protein expression in infected cells. We now show that NS1 protein expression results in enhancement of reporter gene activity from transfected plasmids. This effect appears to be mediated at the translational level, and it is reminiscent of the activity of the adenoviral virus-associated I (VAI) RNA, a known inhibitor of the antiviral, IFN-induced, PKR protein. To study the effects of the NS1 protein on viral and cellular protein synthesis during influenza A virus infection, we used recombinant influenza viruses lacking the NS1 gene (delNS1) or expressing truncated NS1 proteins. Our results demonstrate that the NS1 protein is required for efficient viral protein synthesis in COS-7 cells. This activity maps to the amino-terminal domain of the NS1 protein, since cells infected with wild-type virus or with a mutant virus expressing a truncated NS1 protein-lacking approximately half of its carboxy-terminal end-showed similar kinetics of viral and cellular protein expression. Interestingly, no major differences in host cell protein synthesis shutoff or in viral protein expression were found among NS1 mutant viruses in Vero cells. Thus, another viral component(s) different from the NS1 protein is responsible for the inhibition of host protein synthesis during viral infection. In contrast to the earlier proposal suggesting that the NS1 protein regulates the levels of spliced M2 mRNA, no effects on M2 protein accumulation were seen in Vero cells infected with delNS1 virus.  相似文献   

14.
Highly pathogenic H5N1 influenza A viruses have spread across Asia, Europe, and Africa. More than 500 cases of H5N1 virus infection in humans, with a high lethality rate, have been reported. To understand the molecular basis for the high virulence of H5N1 viruses in mammals, we tested the virulence in ferrets of several H5N1 viruses isolated from humans and found A/Vietnam/UT3062/04 (UT3062) to be the most virulent and A/Vietnam/UT3028/03 (UT3028) to be avirulent in this animal model. We then generated a series of reassortant viruses between the two viruses and assessed their virulence in ferrets. All of the viruses that possessed both the UT3062 hemagglutinin (HA) and nonstructural protein (NS) genes were highly virulent. By contrast, all those possessing the UT3028 HA or NS genes were attenuated in ferrets. These results demonstrate that the HA and NS genes are responsible for the difference in virulence in ferrets between the two viruses. Amino acid differences were identified at position 134 of HA, at positions 200 and 205 of NS1, and at positions 47 and 51 of NS2. We found that the residue at position 134 of HA alters the receptor-binding property of the virus, as measured by viral elution from erythrocytes. Further, both of the residues at positions 200 and 205 of NS1 contributed to enhanced type I interferon (IFN) antagonistic activity. These findings further our understanding of the determinants of pathogenicity of H5N1 viruses in mammals.  相似文献   

15.
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   

16.
17.
The infection of chickens with avian Hepatitis E virus (avian HEV) can be asymptomatic or induces clinical signs characterized by increased mortality and decreased egg production in adult birds. Due to the lack of an efficient cell culture system for avian HEV, the interaction between virus and host cells is still barely understood. In this study, four truncated avian HEV capsid proteins (ORF2-1 – ORF2-4) with an identical 338aa deletion at the N-terminus and gradual deletions from 0, 42, 99 and 136aa at the C-terminus, respectively, were expressed and used to map the possible binding site within avian HEV capsid protein. Results from the binding assay showed that three truncated capsid proteins attached to avian LMH cells, but did not penetrate into cells. However, the shortest construct, ORF2-4, lost the capability of binding to cells suggesting that the presence of amino acids 471 to 507 of the capsid protein is crucial for the attachment. The construct ORF2-3 (aa339-507) was used to study the potential binding of avian HEV capsid protein to human and other avian species. It could be demonstrated that ORF2-3 was capable of binding to QT-35 cells from Japanese quail and human HepG2 cells but failed to bind to P815 cells. Additionally, chicken serum raised against ORF2-3 successfully blocked the binding to LMH cells. Treatment with heparin sodium salt or sodium chlorate significantly reduced binding of ORF2-3 to LMH cells. However, heparinase II treatment of LMH cells had no effect on binding of the ORF2-3 construct, suggesting a possible distinct attachment mechanism of avian as compared to human HEV. For the first time, interactions between avian HEV capsid protein and host cells were investigated demonstrating that aa471 to 507 of the capsid protein are needed to facilitate interaction with different kind of cells from different species.  相似文献   

18.
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   

19.
华南流感病毒NS1基因特性研究   总被引:7,自引:0,他引:7  
为了解H9N2和H5N1亚型流行性感冒病毒株的NS1基因特性,采用RT-PCR方法测定了12株2000~2003年间在华南地区分离的禽流感病毒株的NS1基因核苷酸序列. 测序显示6株H9N2亚型流感病毒NS1基因开放阅读框(ORF)长654 bp,编码217个氨基酸. 6株H5N1亚型毒株NS1基因ORF长678 bp,编码225个氨基酸. 核苷酸和氨基酸同源性分析表明,同一亚型分离株之间有很高的同源性,而不同亚型的H9N2和H5N1毒株之间存在较大差异. BLAST分析表明,H5N1和H9N2亚型流感病毒分离株的NS1基因分别与近两年从香港特区和华南地区的鸭中分离的毒株A/Duck/Hong Kong/646.3/01 (H5N1)、A/Duck/Shantou/2143/01 (H9N2)有很高的亲缘关系. 该研究结果为进一步进行NS1功能研究奠定了基础.  相似文献   

20.
Influenza A virus NS2 protein, also called nuclear export protein (NEP), is crucial for the nuclear export of viral ribonucleoproteins. However, the molecular mechanisms of NEP mediation in this process remain incompletely understood. A leucine-rich nuclear export signal (NES2) in NEP, located at the predicted N2 helix of the N-terminal domain, was identified in the present study. NES2 was demonstrated to be a transferable NES, with its nuclear export activity depending on the nuclear export receptor chromosome region maintenance 1 (CRM1)-mediated pathway. The interaction between NEP and CRM1 is coordinately regulated by both the previously reported NES (NES1) and now the new NES2. Deletion of the NES1 enhances the interaction between NEP and CRM1, and deletion of the NES1 and NES2 motifs completely abolishes this interaction. Moreover, NES2 interacts with CRM1 in the mammalian two-hybrid system. Mutant viruses containing NES2 alterations generated by reversed genetics exhibit reduced viral growth and delay in the nuclear export of viral ribonucleoproteins (vRNPs). The NES2 motif is highly conserved in the influenza A and B viruses. The results demonstrate that leucine-rich NES2 is involved in the nuclear export of vRNPs and contributes to the understanding of nucleocytoplasmic transport of influenza virus vRNPs.  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