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 187 毫秒
1.
在本实验室研制出的多株针对H5N1亚型禽流感血凝素单抗中,13D4单抗对所有H5亚型病毒均有血凝抑制和中和活性,具有特异性高、反应性强和识别广的特点,且在小鼠实验中显示了对各种代表株禽流感的感染和发病均具有良好的治疗效果。在此研究基础上,本实验通过基因工程构建含有13D4单链抗体(scFv)基因的毕赤酵母表达载体,实现目的蛋白的分泌性表达和纯化。经过竞争法和血凝抑制检测其活性,表明获得的单链抗体具有与原始鼠源抗体相近的反应活性和相同的识别表位。H5N1广谱中和单抗13D4的单链抗体的成功构建,为进一步研制针对H5N1禽流感病毒的治疗性抗体奠定了基础。  相似文献   

2.
利用杆状病毒-昆虫细胞表达H5N1型禽流感病毒的血凝素蛋白(HA),纯化后的重组蛋白HA免疫小鼠并制备杂交瘤单克隆抗体,用H5N1型禽流感病毒的全病毒进行筛选,成功地获得了抗H5N1型禽流感病毒血凝素蛋白HA的单抗.MDCK细胞微量中和试验表明,单抗8G10D7可对clade2和clade9的H5N1型禽流感病毒起中和作用.Western-blot及血凝抑制实验进一步证明了该单抗的结合位点位于HA蛋白的HA1亚基上.鸡胚感染病毒预防试验结果表明,8G10D7对禽来源的和人来源的H5N1型禽流感病毒均可达到100%的保护率;在治疗试验组中,8G10D7对禽来源的病毒感染具有较高的保护率,可达100%,对人来源的H5N1型禽流感病毒最高也可达87.5%的保护率.该抗体的获得不仅为H5N1型高致病性禽流感病毒的预防和治疗带来了希望,同时其中和位点的发现也为以后亚单位疫苗的研制提供新的思路.  相似文献   

3.
目的从人源化噬菌体抗体库(human single fold scFv libraries I+J)中筛选到能高亲和性、特异结合人禽流感病毒H5N1的单链抗体,为建立H5N1快速筛查试剂和人源化治疗单抗奠定基础。方法以H5N1病毒的血凝素(hemagglutitin,HA)蛋白和核蛋白(nucleoprotein,NP)为目的蛋白,对上述单抗噬菌体文库以亲和性为原理进行筛选,经过3轮筛选富集后,随机挑选了96个噬菌体克隆扩增培养,ELISA法挑选能特异性、高亲和性结合目的蛋白的噬菌体克隆,并换用HB2151宿主菌对阳性单链抗体克隆进行可溶性表达,ELISA法鉴定可溶性单链抗体的结合活性,PCR扩增阳性克隆的轻、重链基因片段,并对阳性单链抗体分子测序和序列分析。结果经过3轮筛选,分别从96个噬菌体克隆中挑选到了两株能特异结合NP蛋白、3株能特异结合HA蛋白的单链抗体,PCR扩增都得到了长为300、302和935bp的轻链、重链和轻链-连接片段-重链的基因片段,测序结果分析发现上述5条单链抗体片段在轻链的47、49、50、51、53、54、56、96、97、98和99位的氨基酸组成不同,而特异结合NP蛋白的单链在重链区域氨基酸组成完全相同,而特异结合HA蛋白的单链在重链的44、47、85、86、87、88和89位氨基酸组成不同。结论从噬菌体抗体库中筛选到的特异结合HA和NP蛋白的单链抗体片段,可为进一步研发H5N1快速筛选试剂和人源性治疗抗体奠定基础,也可为鉴定HA和NP蛋白中的抗原决定簇提供结构信息。  相似文献   

4.
研究黄芩苷抑制H5N1禽流感假病毒的活性及其作用机理。采用H5N1假病毒检测体系,观察黄芩苷对不同H5N1禽流感假病毒株进入的抑制作用;并结合VSVG假病毒为阴性对照,判定黄芩苷是特异性的作用于H5N1禽流感病毒的进入环节;采用血凝抑制实验、ELISA实验分析其作用机理,采用MTT法测定黄芩苷毒性。黄芩苷能特异性的抑制A/Anhui/1/2005,A/Xinjiang/1/2006,A/Hong Kong/156/1997,A/Qinghai/59/2005,A/Thailand/Kan353/2004,A/Viet Nam/1194/2004 H5N1禽流感假病毒株的进入,IC50分别为65.76±5.61μM、54.98±4.38μM、46.81±5.12μM、32.88±4.18μM、63.30±1.59μM、43.23±3.43μM;黄芩苷对血凝素HA2亚基有抑制作用,IC50为35.86±3.09μM。黄芩苷在体外具有抑制H5N1禽流感进入的作用,其作用机制为抑制血凝素HA2亚基。  相似文献   

5.
对深圳首例疑似人禽流感病人的标本,进行了RT-PCR、Real-time PCR检测及病毒分离培养、血清中和试验、抗原比检测及发病早期不同病程多份标本的病毒载量分析;对分离物进行了HA基因、NA基因及M基因的核酸检测.结果表明:患者气管吸出物的H5N1亚型和A型流感病毒的特异核酸均呈阳性,并通过细胞培养分离到禽流感病毒A/Guangdong/2/06(H5N1)株.气管吸取物病毒载量随着病程延长逐渐减少,而血清中和抗体水平逐渐上升达到1∶160之后又缓缓下降.A/Guangdong/2/06株8个片段的核苷酸序列显示,其与2005~2006年中国南部的禽流感分离株高度同源,与越南、泰国、印度尼西亚等分离到的禽流感分离株存在明显的差异.  相似文献   

6.
从具有高滴度狂犬病毒抗体的多位疫苗注射者采集外周血淋巴细胞,构建人源抗狂犬病毒Fab基因工程抗体文库。用纯化的狂犬aG和CTN株病毒颗粒富集筛选所得Fab噬菌体抗体文库,利用ELISA和间接免疫荧光法IFA鉴定所得人源单克隆抗体Fab段基因的功能特性,并通过序列测定确定所得抗体的轻链和重链的型别,成功获得11株抗狂犬病毒糖蛋白的人源单克隆Fab抗体。将其中5株人源单克隆Fab抗体的轻链和重链分别克隆入全抗体表达载体pAC-L-Fc后转染昆虫Sf9细胞,利用杆状病毒系统实现全抗体的分泌型表达。5株全抗体在体外与狂犬病毒CVS-11株的中和反应中均显示具有狂犬病毒中和活性。人源中和性抗狂犬病毒基因工程全抗体的获得为我国自行生产抗狂犬病单克隆抗体鸡尾酒奠定了物质基础。  相似文献   

7.
人源抗狂犬病毒单克隆抗体Fab段基因的获得和表达   总被引:4,自引:2,他引:2  
运用噬菌体表面呈现(phage display)技术获得了人源抗狂犬病毒糖蛋白基因工程单克隆抗体Fab段基因及其表达。从狂犬病毒PM株Vero细胞疫苗免疫的人抗凝血中分离获得外周淋巴细胞,提取细胞总RNA,通过RTPCR方法,用一组人IgG Fab基因4特异性引物,从合成的cDNA中扩增了一组轻链和重链Fab段基因,将轻链和重链Fab段基因,将轻链和重链先后克隆入噬菌体载体pComb3,成功地建立了抗狂犬病毒抗原的方法,对此抗体库进行富积筛选表达,成功地获得了抗狂犬病毒的人源单抗Fab段基因及其在大肠杆菌中的有效表达,对其中一株单抗G10进行了较为系统的分析,发现它与一株鼠源中和性狂犬病毒糖蛋白特异性单抗存在竞争,证实该单抗能识别狂犬病毒糖蛋白,其序列资料分析表明,该单抗为一株新的抗狂犬病毒人源基因工程抗体。  相似文献   

8.
利用反向遗传学技术构建H5亚型禽流感高产疫苗株   总被引:13,自引:0,他引:13  
采用RT-PCR技术分别扩增了鹅源高产禽流感病毒的6条内部基因片段,近期分离的H5N1亚型禽流感病毒的血凝素基因以及N3亚型参考毒株的神经氨酸酶基因,分别构建了8个基因的转录与表达载体,利用反向遗传学技术拯救出了全部基因都源于禽源的重组流感病毒疫苗株rH5N3。通过对血凝素蛋白HA1和HA2连接肽处的5个碱性氨基酸(R-R-R-K-K)基因缺失与修饰,从而消除了病毒基因的毒力相关序列,拯救的rH5N3疫苗株对鸡和鸡胚均无致病性,病毒在鸡胚尿囊液和细胞培养上清的HA效价得到极大提高,分别为12048和1512。制备的禽流感疫苗免疫动物后4~5周即可诱导产生高效价的HI抗体,鸡免疫后18周依然保持高水平的HI抗体。重组疫苗不论是对于国内早期分离的禽流感病毒A/Goose/Guangdong/1/96还是近期分离的A/Goose/HLJ/QFY/04都能够产生完全的免疫保护作用,免疫鸡攻毒后不发病、不排毒、不死亡。带有N3鉴别诊断标记禽流感疫苗株的研制为H5N1高致病性禽流感的防治提供了新的技术保障。  相似文献   

9.
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   

10.
在本实验室研制出的多株针对H5N1血凝素的鼠单抗中,10F7对34株H5N1病毒株都有血凝抑制和中和活性,具有特异性高、反应性强、识别谱广的特点。通过基因工程构建10F7单链抗体(scFv)表达重组质粒,在大肠杆菌中表达并纯化scFv,经血凝抑制实验及中和实验检测其活性。结果在针对3株病毒的血凝抑制实验中,10F7scFv蛋白对其中2株H5N1病毒均显示出结合活性,而对H9毒株没有反应。在针对7株H5N1病毒的中和实验中,10F7scFv对5株病毒具有较好的中和能力。H5N1广谱中和抗体10F7的单链抗体构建,为进一步研制针对H5N1禽流感病毒的治疗性抗体奠定了基础。  相似文献   

11.

Background

The development of new therapeutic targets and strategies to control highly pathogenic avian influenza (HPAI) H5N1 virus infection in humans is urgently needed. Broadly cross-neutralizing recombinant human antibodies obtained from the survivors of H5N1 avian influenza provide an important role in immunotherapy for human H5N1 virus infection and definition of the critical epitopes for vaccine development.

Methodology/Principal Findings

We have characterized two recombinant baculovirus-expressed human antibodies (rhAbs), AVFluIgG01 and AVFluIgG03, generated by screening a Fab antibody phage library derived from a patient recovered from infection with a highly pathogenic avian influenza A H5N1 clade 2.3 virus. AVFluIgG01 cross-neutralized the most of clade 0, clade 1, and clade 2 viruses tested, in contrast, AVFluIgG03 only neutralized clade 2 viruses. Passive immunization of mice with either AVFluIgG01 or AVFluIgG03 antibody resulted in protection from a lethal H5N1 clade 2.3 virus infection. Furthermore, through epitope mapping, we identify two distinct epitopes on H5 HA molecule recognized by these rhAbs and demonstrate their potential to protect against a lethal H5N1 virus infection in a mouse model.

Conclusions/Significance

Importantly, localization of the epitopes recognized by these two neutralizing and protective antibodies has provided, for the first time, insight into the human antibody responses to H5N1 viruses which contribute to the H5 immunity in the recovered patient. These results highlight the potential of a rhAbs treatment strategy for human H5N1 virus infection and provide new insight for the development of effective H5N1 pandemic vaccines.  相似文献   

12.
Due to the high mortality associated with recent, widely circulating strains of H5N1 influenza virus in poultry, the recurring introduction of H5N1 viruses from birds to humans, and the difficulties in H5N1 eradication by elimination of affected flocks, an effective vaccine against HPAI (highly pathogenic avian influenza) is highly desirable. Using reverse genetics, a set of experimental live attenuated vaccine strains based on recombinant H5N1 influenza virus A/Viet Nam/1203/04 was generated. Each virus was attenuated through expression of a hemagglutinin protein in which the polybasic cleavage site had been removed. Viruses were generated which possessed a full-length NS1 or a C-terminally truncated NS1 protein of 73, 99, or 126 amino acids. Viruses with each NS genotype were combined with a PB2 polymerase gene which carried either a lysine or a glutamic acid at position 627. We predicted that glutamic acid at position 627 of PB2 would attenuate the virus in mammalian hosts, thus increasing the safety of the vaccine. All recombinant viruses grew to high titers in 10-day-old embryonated chicken eggs but were attenuated in mammalian cell culture. Induction of high levels of beta interferon by all viruses possessing truncations in the NS1 protein was demonstrated by interferon bioassay. The viruses were each found to be highly attenuated in a mouse model. Vaccination with a single dose of any virus conferred complete protection from death upon challenge with a mouse lethal virus expressing H5N1 hemagglutinin and neuraminidase proteins. In a chicken model, vaccination with a single dose of a selected virus encoding the NS1 1-99 protein completely protected chickens from lethal challenge with homologous HPAI virus A/Viet Nam/1203/04 (H5N1) and provided a high level of protection from a heterologous virus, A/egret/Egypt/01/06 (H5N1). Thus, recombinant influenza A/Viet Nam/1203/04 viruses attenuated through the introduction of mutations in the hemagglutinin, NS1, and PB2 coding regions display characteristics desirable for live attenuated vaccines and hold potential as vaccine candidates in poultry as well as in mammalian hosts.  相似文献   

13.
It is commonly perceived that the human immune system is naive to the newly emerged H5N1 virus. In contrast, most adults have been exposed to influenza A H1N1 and H3N2 viruses through vaccination or infection. Adults born before 1968 have likely been exposed to H2N2 viruses. We hypothesized that CD4(+) T cells generated in response to H1N1, H3N2, and H2N2 influenza A viruses also recognize H5N1 epitopes. Tetramer-guided epitope mapping and Ag-specific class II tetramers were used to identify H5N1-specific T cell epitopes and detect H5N1-specific T cell responses. Fifteen of 15 healthy subjects tested had robust CD4(+) T cell responses against matrix protein, nucleoprotein, and neuraminidase of the influenza A/Viet Nam/1203/2004 (H5N1) virus. These results are not surprising, because the matrix protein and nucleoprotein of influenza A viruses are conserved while the neuraminidase of the H5N1 virus is of the same subtype as that of the circulating H1N1 influenza strain. However, H5N1 hemagglutinin-reactive CD4(+) T cells were also detected in 14 of 14 subjects examined despite the fact that hemagglutinin is less conserved. Most were cross-reactive to H1, H2, or H3 hemagglutinin epitopes. H5N1-reactive T cells were also detected ex vivo, exhibited a memory phenotype, and were capable of secreting IFN-gamma, TNF-alpha, IL-5, and IL-13. These data demonstrate the presence of H5N1 cross-reactive T cells in healthy Caucasian subjects, implying that exposure to influenza A H1N1, H3N2, or H2N2 viruses through either vaccination or infection may provide partial immunity to the H5N1 virus.  相似文献   

14.
Highly pathogenic avian influenza (HPAI) H5N1 viruses continue to cause sporadic human infections with a high fatality rate. Respiratory failure due to acute respiratory distress syndrome (ARDS) is a complication among hospitalized patients. Since progressive pulmonary endothelial damage is the hallmark of ARDS, we investigated host responses following HPAI virus infection of human pulmonary microvascular endothelial cells. Evaluation of these cells for the presence of receptors preferred by influenza virus demonstrated that avian-like (α2-3-linked) receptors were more abundant than human-like (α2-6-linked) receptors. To test the permissiveness of pulmonary endothelial cells to virus infection, we compared the replication of selected seasonal, pandemic (2009 H1N1 and 1918), and potentially pandemic (H5N1) influenza virus strains. We observed that these cells support productive replication only of HPAI H5N1 viruses, which preferentially enter through and are released from the apical surface of polarized human endothelial monolayers. Furthermore, A/Thailand/16/2004 and A/Vietnam/1203/2004 (VN/1203) H5N1 viruses, which exhibit heightened virulence in mammalian models, replicated to higher titers than less virulent H5N1 strains. VN/1203 infection caused a significant decrease in endothelial cell proliferation compared to other subtype viruses. VN/1203 virus was also found to be a potent inducer of cytokines and adhesion molecules known to regulate inflammation during acute lung injury. Deletion of the H5 hemagglutinin (HA) multibasic cleavage site did not affect virus infectivity but resulted in decreased virus replication in endothelial cells. Our results highlight remarkable tropism and infectivity of the H5N1 viruses for human pulmonary endothelial cells, resulting in the potent induction of host inflammatory responses.  相似文献   

15.
Yang P  Duan Y  Zhang P  Li Z  Wang C  Dong M  Tang C  Xing L  Gu H  Zhao Z  Liu X  Zhang S  Wang X 《PloS one》2012,7(1):e30252

Background

The increase in recent outbreaks and unpredictable changes of highly pathogenic avian influenza (HPAI) H5N1 in birds and humans highlights the urgent need to develop a cross-protective H5N1 vaccine. We here report our development of a multiple-clade H5N1 influenza vaccine tested for immunogenicity and efficacy to confer cross-protection in an animal model.

Methodology/Principal Findings

Mice received two doses of influenza split vaccine with oil-in-water emulsion adjuvant SP01 by intranasal administration separated by two weeks. Single vaccines (3 µg HA per dose) included rg-A/Vietnam/1203/2004(Clade 1), rg-A/Indonesia/05/2005(Clade 2.1), and rg-A/Anhui/1/2005(Clade 2.3.4). The trivalent vaccine contained 1 µg HA per dose of each single vaccine. Importantly, complete cross-protection was observed in mice immunized using trivalent vaccine with oil-in-water emulsion adjuvant SP01 that was subsequently challenged with the lethal A/OT/SZ/097/03 influenza strain (Clade 0), whereas only the survival rate was up to 60% in single A/Anhui/1/2005 vaccine group.

Conclusion/Significance

Our findings demonstrated that the multiple-clade H5N1 influenza vaccine was able to elicit a cross-protective immune response to heterologous HPAI H5N1 virus, thus giving rise to a broadly cross-reactive vaccine to potential prevention use ahead of the strain-specific pandemic influenza vaccine in the event of an HPAI H5N1 influenza outbreak. Also, the multiple-clade adjuvanted vaccine could be useful in allowing timely initiation of vaccination against unknown pandemic virus.  相似文献   

16.
The global spread of highly pathogenic avian influenza A H5N1 viruses raises concerns about more widespread infection in the human population. Pre-pandemic vaccine for H5N1 clade 1 influenza viruses has been produced from the A/Viet Nam/1194/2004 strain (VN1194), but recent prevalent avian H5N1 viruses have been categorized into the clade 2 strains, which are antigenically distinct from the pre-pandemic vaccine. To understand the antigenicity of H5N1 hemagglutinin (HA), we produced a neutralizing monoclonal antibody (mAb12-1G6) using the pre-pandemic vaccine. Analysis with chimeric and point mutant HAs revealed that mAb12-1G6 bound to the loop (amino acid positions 140-145) corresponding to an antigenic site A in the H3 HA. mAb12-1G6 failed to bind to the mutant VN1194 HA when only 3 residues were substituted with the corresponding residues of the clade 2.1.3.2 A/Indonesia/5/05 strain (amino acid substitutions at positions Q142L, K144S, and S145P), suggesting that these amino acids are critical for binding of mAb12-1G6. Escape mutants of VN1194 selected with mAb12-1G6 carried a S145P mutation. Interestingly, mAb12-1G6 cross-neutralized clade 1 and clade 2.2.1 but not clade 2.1.3.2 or clade 2.3.4 of the H5N1 virus. We discuss the cross-reactivity, based on the amino acid sequence of the epitope.  相似文献   

17.
Suguitan AL  Cheng X  Wang W  Wang S  Jin H  Lu S 《PloS one》2011,6(7):e21942
Priming immunization plays a key role in protecting individuals or populations to influenza viruses that are novel to humans. To identify the most promising vaccine priming strategy, we have evaluated different prime-boost regimens using inactivated, DNA and live attenuated vaccines in ferrets. Live attenuated influenza A/Vietnam/1203/2004 (H5N1) candidate vaccine (LAIV, VN04 ca) primed ferrets efficiently while inactivated H5N1 vaccine could not prime the immune response in seronegative ferrets unless an adjuvant was used. However, the H5 HA DNA vaccine alone was as successful as an adjuvanted inactivated VN04 vaccine in priming the immune response to VN04 ca virus. The serum antibody titers of ferrets primed with H5 HA DNA followed by intranasal vaccination of VN04 ca virus were comparable to that induced by two doses of VN04 ca virus. Both LAIV-LAIV and DNA-LAIV vaccine regimens could induce antibody responses that cross-neutralized antigenically distinct H5N1 virus isolates including A/HongKong/213/2003 (HK03) and prevented nasal infection of HK03 vaccine virus. Thus, H5 HA DNA vaccination may offer an alternative option for pandemic preparedness.  相似文献   

18.
The impending influenza virus pandemic requires global vaccination to prevent large-scale mortality and morbidity, but traditional influenza virus vaccine production is too slow for rapid responses. We have developed bacterial systems for expression and purification of properly folded functional hemagglutinin as a rapid response to emerging pandemic strains. A recombinant H5N1 (A/Vietnam/1203/2004) hemagglutinin globular domain (HA1) was produced in Escherichia coli under controlled redox refolding conditions. Importantly, the properly folded HA1(1-320), i.e., HA1 lacking amino acids 321 to 330, contained ≥75% functional oligomers without addition of foreign oligomerization sequence. Site-directed mutagenesis mapped the oligomerization signal to the HA1 N-terminal Ile-Cys-Ile residues at positions 3 to 5. The purified HA1 oligomers (but not monomers) bound fetuin and agglutinated red blood cells. Upon immunization of rabbits, the oligomeric HA1(1-320) elicited potent neutralizing antibodies against homologous and heterologous H5N1 viruses more rapidly than HA1(28-320) containing only monomers. Ferrets vaccinated with oligomeric HA1 (but not monomeric HA1 with the N terminus deleted) at 15 and 3 μg/dose were fully protected from lethality and weight loss after challenge with homologous H5N1 (A/Vietnam/1203/2004, clade 1) virus, as well as heterologous clade 2.2 H5N1 (A/WooperSwan/Mongolia/244/2005) virus. Protection was associated with a significant reduction in viral loads in the nasal washes of homologous and heterologous virus challenged ferrets. This is the first study that describes the presence of an N-terminal oligomerization sequence in the globular domain of influenza virus hemagglutinin. Our findings suggest that functional oligomeric rHA1-based vaccines can be produced efficiently in bacterial systems and can be easily upscaled in response to a pandemic influenza virus threat.  相似文献   

19.
We mapped the hemagglutinin (HA) antigenic epitopes of a highly pathogenic H5N1 influenza virus on the three-dimensional HA structure by characterizing escape mutants of a recombinant virus containing A/Vietnam/1203/04 (H5N1) ΔHA and neuraminidase genes in the genetic background of A/Puerto Rico/8/34 (H1N1) virus. The mutants were selected with a panel of eight anti-HA monoclonal antibodies (MAbs), seven to A/Vietnam/1203/04 (H5N1) virus and one to A/Chicken/Pennsylvania/8125/83 (H5N2) virus, and the mutants’ HA genes were sequenced. The amino acid changes suggested three MAb groups: four MAbs reacted with the complex epitope comprising parts of the antigenic site B of H3 HA and site Sa of H1 HA, two MAbs reacted with the epitope corresponding to the antigenic site A in H3 HA, and two MAbs displayed unusual behavior: each recognized amino acid changes at two widely separate antigenic sites. Five changes were detected in amino acid residues not previously reported as changed in H5 escape mutants, and four others had substitutions not previously described. The HA antigenic structure differs substantially between A/Vietnam/1203/04 (H5N1) virus and the low-pathogenic A/Mallard/Pennsylvania/10218/84 (H5N2) virus we previously characterized (N. V. Kaverin et al., J. Gen. Virol. 83:2497-2505, 2002). The hemagglutination inhibition reactions of the MAbs with recent highly pathogenic H5N1 viruses were consistent with the antigenic-site amino acid changes but not with clades and subclades based on H5 phylogenetic analysis. These results provide information on the recognition sites of the MAbs widely used to study H5N1 viruses and demonstrate the involvement of the HA antigenic sites in the evolution of highly pathogenic H5N1 viruses, findings that can be critical for characterizing pathogenesis and vaccine design.  相似文献   

20.
Highly pathogenic avian influenza A viruses (HPAIV) of the H5N1 subtype occasionally transmit from birds to humans and can cause severe systemic infections in both hosts. PB1-F2 is an alternative translation product of the viral PB1 segment that was initially characterized as a pro-apoptotic mitochondrial viral pathogenicity factor. A full-length PB1-F2 has been present in all human influenza pandemic virus isolates of the 20(th) century, but appears to be lost evolutionarily over time as the new virus establishes itself and circulates in the human host. In contrast, the open reading frame (ORF) for PB1-F2 is exceptionally well-conserved in avian influenza virus isolates. Here we perform a comparative study to show for the first time that PB1-F2 is a pathogenicity determinant for HPAIV (A/Viet Nam/1203/2004, VN1203 (H5N1)) in both mammals and birds. In a mammalian host, the rare N66S polymorphism in PB1-F2 that was previously described to be associated with high lethality of the 1918 influenza A virus showed increased replication and virulence of a recombinant VN1203 H5N1 virus, while deletion of the entire PB1-F2 ORF had negligible effects. Interestingly, the N66S substituted virus efficiently invades the CNS and replicates in the brain of Mx+/+ mice. In ducks deletion of PB1-F2 clearly resulted in delayed onset of clinical symptoms and systemic spreading of virus, while variations at position 66 played only a minor role in pathogenesis. These data implicate PB1-F2 as an important pathogenicity factor in ducks independent of sequence variations at position 66. Our data could explain why PB1-F2 is conserved in avian influenza virus isolates and only impacts pathogenicity in mammals when containing certain amino acid motifs such as the rare N66S polymorphism.  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