首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 405 毫秒
1.
2.
在对华东地区家养水禽中流感病毒的带毒状况的流行病学监测过程中,从表观健康家鸭体内分离到一株H5N1亚型禽流感病毒A/duck/Shandong/009/2008(简称Dk/SD/009/08)。为了解该毒株的基因组构成,对该分离株进行全基因测序。测序结果显示:该毒株HA裂解位点处的氨基酸序列为PLRERRRK-R/GL,符合高致病性禽流感病毒的分子特征,且参照H5N1国际统一命名准则,Dk/SD/009/08的HA基因属于2.3.4进化支。BLAST结果显示,HA、NA、NP及NS基因均与H5N1亚型病毒的核苷酸一致性最高,而RNA聚合酶基因(PB2、PB1、PA)及M基因则与H9N2亚型病毒的亲缘关系最近,故推测该分离株可能是一株天然重组病毒;遗传进化分析进一步表明,流行于华南地区鹌鹑中的G1-like H9N2亚型病毒可能为该分离株提供部分的内部基因。  相似文献   

3.
目的利用A/H6N1亚型禽流感病毒的反向遗传平台,评估PB2 E627K对A/H6N1亚型禽流感病毒的致病性,探究A/H6N1流感病毒的致病性分子基础。方法通过A/H6N1亚型禽流感病毒A/Mallard/San-Jiang/275/2007株反向遗传操作系统和点突变技术拯救病毒rA/H6N1和PB2 E627K位点发生突变的rA/H6N1-627,两株拯救病毒分别以101EID50~106EID50的攻毒剂量人工感染BALB/c小鼠,通过体重变化、死亡率、病毒滴定等方面进行致病性分析。结果成功构建A/H6N1亚型禽流感病毒的反向遗传平台,rA/H6N1的8个基因片段完全源于A/H6N1的基因组,核苷酸序列及生物学特性与A/H6N1完全一致。rA/H6N1能够人工感染BALB/c小鼠,但不致死,对BALB/c小鼠呈现低致病性(MLD50>106.5EID50),病毒在小鼠体内的分布情况及各个脏器中的病毒滴度与A/H6N1保持一致;rA/H6N1-627能感染小鼠,引起小鼠体重下降,但不能引起所有106EID50组小鼠死亡,病毒能在小鼠的肺脏和脑部进行增殖。结论实验结果表明,在H5N1禽流感中发挥重要作用的PB2-E627K位点并非A/H6N1流感病毒的毒力决定因子。A/H6N1流感病毒致病性的分子基础还有待继续研究,该反向遗传操作系统和点突变技术的建立为研究该亚型流感病毒致病机制、传播机制及病毒基因功能奠定了基础,同时也为A/H6N1亚型禽流感病毒新型疫苗的研制开辟了新途径。  相似文献   

4.
设计带有BsmBI、BsaI或AarI酶切位点的引物,用RT-PCR扩增H9N2亚型禽流感病毒(AIV)的8个基因全长片段,克隆入双向转录/表达载体pHW2000,并在PB2、PB1和NA基因中共引入了3个沉默突变标签。将其2个表面基因(HA和NA基因)加上任意1个内部基因,而其它5个内部基因来自A/WSN/33,进行了6种3 5组合形式的基因重排,把相应组合的转录/表达质粒共转染COS-1细胞,均产生了预期组合、有感染性的H9N2亚型流感病毒,表明亲缘关系遥远的流感病毒可以互相获取基因片段产生重组病毒,提示表面结构基因和单个内部基因不足以限制H9N2AIV在哺乳动物细胞上的宿主范围,同时也验证了构建的8个转录/表达载体均能有效工作,为进一步研究H9N2亚型AIV基因结构与功能、AIV与宿主之间的关系打下了基础。  相似文献   

5.
设计带有BsmBI、BsaⅠ或AarⅠ酶切位点的引物,用RT PCR扩增H9N2亚型禽流感病毒(AIV)的8个基因全长片段,克隆入双向转录/表达载体pHW2000,并在PB2、PB1和NA基因中共引入了3个沉默突变标签.将其2个表面基因(HA和NA基因)加上任意1个内部基因,而其它5个内部基因来自A/WSN/33,进行了6种3+5组合形式的基因重排,把相应组合的转录/表达质粒共转染COS-1细胞,均产生了预期组合、有感染性的H9N2亚型流感病毒,表明亲缘关系遥远的流感病毒可以互相获取基因片段产生重组病毒,提示表面结构基因和单个内部基因不足以限制H9N2 AIV在哺乳动物细胞上的宿主范围,同时也验证了构建的8个转录/表达载体均能有效工作,为进一步研究H9N2亚型AIV基因结构与功能、AIV与宿主之间的关系打下了基础.  相似文献   

6.
浙江省首例人禽流感病例的病原学与分子生物学研究   总被引:3,自引:0,他引:3  
为确认浙江省首例疑似人禽流感病例,进行病原学分析,对患者气管吸出物进行核酸RT-PCR、荧光定量RT-PCR检测以及病毒分离,并对患者血清进行HI抗体测定.结果表明患者气管吸出物H5N1亚型和A型流感病毒特异核酸均呈阳性,分离到禽流感病毒A/Zhejiang/16/06(H5N1)株;双份血清中禽流感病毒(H5N1)HI抗体滴度分别为1320和1640,从病原学和血清学上证实为人禽流感病例.分离毒株测序结果显示,A/Zhejiang/16/06(H5N1)株在HA裂解位点为多个碱性氨基酸,符合高致病性禽流感病毒特征;该毒株的HA、NA、PB2、NP、M和NS基因序列均为禽源,与2005年我国福建、安徽等地禽流感病毒分离株高度同源,而与越南、泰国以及香港1997年分离到的禽流感病毒株之间存在明显差异.  相似文献   

7.
H5N6禽流感是重要的人兽共患病,给公共卫生带来严重威胁。为研究人感染H5N6禽流感病毒的基因特征,本文对广州市两株人感染H5N6禽流感病毒进行全基因组序列扩增,应用生物信息学软件分析分子变异和遗传进化特征。结果显示:两毒株各基因片段同源性存在差异,血凝素(Hemagglutinin,HA)基因同源性最高为98.3%,PB2基因同源性最低为85.2%。A/Guangzhou/41641/2014(H5N6)病毒的HA、神经氨酸酶(Neuraminidase,NA)、聚合酶碱性蛋白2(Polymerase basic protein 2,PB2)基因与猫源毒株A/feline/Guangdong/1/2015(H5N6)亲缘关系较近,推测可能起源于共同祖先。两株病毒均为禽源高致病性病毒,HA和NA表面蛋白受体结合位点、裂解位点和耐药位点未发生变异。内部基因重要位点均有不同程度的变异,其中以41641病毒变异较大,并发生PB2蛋白E627K突变。两株病毒均发生与不同亚型病毒之间的重组现象,41641病毒的内部基因分别与H5和H9N2/H7N9发生重组,其中PB2和PB1基因分别与2013年暴发的华南分支和华东分支H7N9禽流感病毒亲缘关系相近,A/Guangzhou/37845/2015(H5N6)病毒的内部基因与H5N1/H5N6病毒发生重组。因此,广州市两株人感染H5N6禽流感病毒进化起源不同,属于两种不同的基因型,本研究推测2013年暴发的H7N9禽流感病毒在新型H5N6重组病毒的进化过程中起到重要作用。  相似文献   

8.
为拯救出一株能够在动物传代细胞中高水平复制的H3N2亚型猪流感疫苗株,利用反向遗传操作技术,将A/Goose/Dalian/3/01(H9N2)毒株的PB1、PA、NP、M、NS基因和A/PR/8/34毒株的PB2基因作为内部基因与猪流感病毒A/Swine/Henan/S4/01(H3N2)毒株的HA、NA基因进行重组,成功拯救出了具有高度细胞适应性毒株rH3N2株,该毒株接毒MDCK细胞60h后,血凝价可以达到1∶512,表明该毒株具有高度适应细胞繁殖特性,为H3N2亚型猪流感病毒细胞培养型疫苗的研制奠定了基础。  相似文献   

9.
2012年7~9月从来源于青海湖地区活禽市场的环境样本中分离到5株H9N2亚型禽流感病毒,为了了解其基因遗传进化情况,本研究通过RT-PCR技术扩增分离毒株的8个基因片段,并进行全基因序列测定。对其分子特征及全基因序列进行遗传进化分析。结果显示5株病毒的HA基因片段的核苷酸相似度为93.2%~99.1%。NA基因核苷酸的相似度为94.5%~99.8%。A/environment/qinghai/017/2012的裂解位点为PSKSSRGLF,其它4个毒株的HA裂解位点均为PSRSSRGLF。5个病毒的HA基因第226位受体结合位点均为L。M1基因片段中发生了N30D和T215A替换。遗传进化分析表明5株病毒同2005年湖南分离的A/chicken/Hunan/5260/2005(H9N2)毒株类似,为一种重配基因型禽流感病毒。其中HA、NA、NS基因片段属于Y280-like支系,MP基因片段属于G1-like支系,NP、PB1、PB2、PA四个基因片段属于F98-like支系。  相似文献   

10.
为确认浙江省首例疑似人禽流感病例,进行病原学分析,对患者气管吸出物进行核酸RT-PCR、荧光定量RT-PCR检测以及病毒分离,并对患者血清进行HI抗体测定。结果表明:患者气管吸出物H5N1亚型和A型流感病毒特异核酸均呈阳性,分离到禽流感病毒A/Zhejiang/16/06(H5N1)株;双份血清中禽流感病毒(H5N1)HI抗体滴度分别为1:320和1:640,从病原学和血清学上证实为人禽流感病例。分离毒株测序结果显示,A/Zhejiang/16/06(H5N1)株在HA裂解位点为多个碱性氨基酸,符合高致病性禽流感病毒特征;该毒株的HA、NA、PB2、NP、M和NS基因序列均为禽源,与2005年我国福建、安徽等地禽流感病毒分离株高度同源,而与越南、泰国以及香港1997年分离到的禽流感病毒株之间存在明显差异。  相似文献   

11.
用8质粒病毒拯救系统产生H9N2/WSN重组A型流行性感冒病毒   总被引:9,自引:0,他引:9  
把禽流行性感冒(流感)病毒A/Chicken/Shanghai/F/98(H9N2)的血凝素(HA)和神经氨酸酶(NA)基因cDNA克隆至polⅠ-pol Ⅱ双向转录和表达载体pHW2000,用这两种质粒与8质粒病毒拯救系统中流感病毒A/WSN/33(H1N1)6个内部基因cDNA的质粒组合(6 2重排),共转染COS-1细胞,产生了能在鸡胚中高滴度增殖的H9N2/、WSN重组病毒。用A/WSN/33的8个基因cDNA质粒作对照,也产生了转染子病毒。经过EID50测定和MDCK感染实验,新基因型H9N2/WSN病毒感染鸡胚的能力强(EID50为10^-11/0.2m1),而且对鸡胚的毒力弱,在不加胰酶的情况下不使MDCK细胞产牛病变。经电镜观察,两个转染子病毒的形态与野生型流感病毒相似。反向遗传操作技术的建立,为对禽流感病毒基因功能和疫苗构建等方面的研究提供了新的手段。  相似文献   

12.
13.
Song H  Nieto GR  Perez DR 《Journal of virology》2007,81(17):9238-9248
In light of the recurrent outbreaks of low pathogenic avian influenza (LPAI) and highly pathogenic avian influenza (HPAI), there is a pressing need for the development of vaccines that allow rapid mass vaccination. In this study, we introduced by reverse genetics temperature-sensitive mutations in the PB1 and PB2 genes of an avian influenza virus, A/Guinea Fowl/Hong Kong/WF10/99 (H9N2) (WF10). Further genetic modifications were introduced into the PB1 gene to enhance the attenuated (att) phenotype of the virus in vivo. Using the att WF10 as a backbone, we substituted neuraminidase (NA) for hemagglutinin (HA) for vaccine purposes. In chickens, a vaccination scheme consisting of a single dose of an att H7N2 vaccine virus at 2 weeks of age and subsequent challenge with the wild-type H7N2 LPAI virus resulted in complete protection. We further extended our vaccination strategy against the HPAI H5N1. In this case, we reconstituted an att H5N1 vaccine virus, whose HA and NA genes were derived from an Asian H5N1 virus. A single-dose immunization in ovo with the att H5N1 vaccine virus in 18-day-old chicken embryos resulted in more than 60% protection for 4-week-old chickens and 100% protection for 9- to 12-week-old chickens. Boosting at 2 weeks posthatching provided 100% protection against challenge with the HPAI H5N1 virus for chickens as young as 4 weeks old, with undetectable virus shedding postchallenge. Our results highlight the potential of live att avian influenza vaccines for mass vaccination in poultry.  相似文献   

14.
J Wang  Y Sun  Q Xu  Y Tan  J Pu  H Yang  EG Brown  J Liu 《PloS one》2012,7(7):e40752
H9N2 influenza viruses have been circulating worldwide in multiple avian species and have repeatedly infected humans to cause typical disease. The continued avian-to-human interspecies transmission of H9N2 viruses raises concerns about the possibility of viral adaption with increased virulence for humans. To investigate the genetic basis of H9N2 influenza virus host range and pathogenicity in mammals, we generated a mouse-adapted H9N2 virus (SD16-MA) that possessed significantly higher virulence than wide-type virus (SD16). Increased virulence was detectable after 8 sequential lung passages in mice. Five amino acid substitutions were found in the genome of SD16-MA compared with SD16 virus: PB2 (M147L, V250G and E627K), HA (L226Q) and M1 (R210K). Assessments of replication in mice showed that all of the SD16-MA PB2, HA and M1 genome segments increased virus replication; however, only the mouse-adapted PB2 significantly increased virulence. Although the PB2 E627K amino acid substitution enhanced viral polymerase activity and replication, none of the single mutations of mouse adapted PB2 could confer increased virulence on the SD16 backbone. The combination of M147L and E627K significantly enhanced viral replication ability and virulence in mice. Thus, our results show that the combination of PB2 amino acids at position 147 and 627 is critical for the increased pathogenicity of H9N2 influenza virus in mammalian host.  相似文献   

15.
Chen LM  Davis CT  Zhou H  Cox NJ  Donis RO 《PLoS pathogens》2008,4(5):e1000072
The segmented structure of the influenza virus genome plays a pivotal role in its adaptation to new hosts and the emergence of pandemics. Despite concerns about the pandemic threat posed by highly pathogenic avian influenza H5N1 viruses, little is known about the biological properties of H5N1 viruses that may emerge following reassortment with contemporary human influenza viruses. In this study, we used reverse genetics to generate the 63 possible virus reassortants derived from H5N1 and H3N2 viruses, containing the H5N1 surface protein genes, and analyzed their viability, replication efficiency, and mouse virulence. Specific constellations of avian-human viral genes proved deleterious for viral replication in cell culture, possibly due to disruption of molecular interaction networks. In particular, striking phenotypes were noted with heterologous polymerase subunits, as well as NP and M, or NS. However, nearly one-half of the reassortants replicated with high efficiency in vitro, revealing a high degree of compatibility between avian and human virus genes. Thirteen reassortants displayed virulent phenotypes in mice and may pose the greatest threat for mammalian hosts. Interestingly, one of the most pathogenic reassortants contained avian PB1, resembling the 1957 and 1968 pandemic viruses. Our results reveal the broad spectrum of phenotypes associated with H5N1/H3N2 reassortment and a possible role for the avian PB1 in the emergence of pandemic influenza. These observations have important implications for risk assessment of H5N1 reassortant viruses detected in surveillance programs.  相似文献   

16.
A series of reassortants has been constructed by crossing of UV-inactivated avian influenza virus of H3N8 subtype and live human influenza virus of H1N1 subtype, adapted to growth in continuous canine kidney cell line (MDCK). The analysis of RNA duplexes has shown that the reassortants contain HA gene of avian influenza virus whereas the other genes belong to human parent virus. The reassortants were efficiently reproduced in MDCK cells at low temperature (limiting for the avian parent virus). The data suggest that the avian virus HA gene does not hamper the reproduction of reassortant viruses in mammalian cells under the conditions unfavorable for the multiplication of avian influenza subtype H3N8 viruses.  相似文献   

17.
Highly pathogenic avian influenza (HPAI) H5N1 viruses, which have emerged in poultry and other wildlife worldwide, contain a characteristic multi-basic cleavage site (CS) in the hemagglutinin protein (HA). Because this arginine-rich CS is unique among influenza virus subtypes, antibodies against this site have the potential to specifically diagnose pathogenic H5N1. By immunizing mice with the CS peptide and screening a phage display library, we isolated four antibody Fab fragment clones that specifically bind the antigen peptide and several HPAI H5N1 HA proteins in different clades. The soluble Fab fragments expressed in Escherichia coli bound the CS peptide and the H5N1 HA protein with nanomolar affinity. In an immunofluorescence assay, these Fab fragments stained cells infected with HPAI H5N1 but not those infected with a less virulent strain. Lastly, all the Fab clones could detect the CS peptide and H5N1 HA protein by open sandwich ELISA. Thus, these recombinant Fab fragments will be useful novel reagents for the rapid and specific detection of HPAI H5N1 virus.  相似文献   

18.
Reassortant viruses which possessed the hemagglutinin and neuraminidase genes of wild-type human influenza A viruses and the remaining six RNA segments (internal genes) of the avian A/Pintail/Alberta/119/79 (H4N6) virus were previously found to be attenuated in humans. To study the genetic basis of this attenuation, we isolated influenza A/Pintail/79 X A/Washington/897/80 reassortant viruses which contained human influenza virus H3N2 surface glycoprotein genes and various combinations of avian or human influenza virus internal genes. Twenty-four reassortant viruses were isolated and first evaluated for infectivity in avian (primary chick kidney [PCK]) and mammalian (Madin-Darby canine kidney [MDCK]) tissue culture lines. Reassortant viruses with two specific constellations of viral polymerase genes exhibited a significant host range restriction of replication in mammalian (MDCK) tissue culture compared with that in avian (PCK) tissue culture. The viral polymerase genotype PB2-avian (A) virus, PB1-A virus, and PA-human (H) virus was associated with a 900-fold restriction, while the viral polymerase genotype PB2-H, PB1-A, and PA-H was associated with an 80,000-fold restriction of replication in MDCK compared with that in PCK. Fifteen reassortant viruses were subsequently evaluated for their level of replication in the respiratory tract of squirrel monkeys, and two genetic determinants of attenuation were identified. First, reassortant viruses which possessed the avian influenza virus nucleoprotein gene were as restricted in replication as a virus which possessed all six internal genes of the avian influenza A virus parent, indicating that the nucleoprotein gene is the major determinant of attenuation of avian-human A/Pintail/79 reassortant viruses for monkeys. Second, reassortant viruses which possessed the viral polymerase gene constellation of PB2-H, PB1-A, and PA-H, which was associated with the greater degree of host range restriction in vitro, were highly restricted in replication in monkeys. Since the avian-human influenza reassortant viruses which expressed either mode of attenuation in monkeys replicated to high titer in eggs and in PCK tissue culture, their failure to replicate efficiently in the respiratory epithelium of primates must be due to the failure of viral factors to interact with primate host cell factors. The implications of these findings for the development of live-virus vaccines and for the evolution of influenza A viruses in nature are discussed.  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