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 234 毫秒
1.
高致病性H5N1亚型禽流感病毒 (AIV) 严重威胁到人类健康,因此研制高效、安全的禽流感疫苗具有重要意义。以我国分离的首株人H5N1亚型禽流感病毒 (A/Anhui/1/2005) 作为研究对象,PCR扩增基质蛋白2 (M2) 和血凝素 (HA) 基因全长开放阅读框片段,构建共表达H5N1亚型AIV膜蛋白基因 M2和HA的重组质粒pStar-M2/HA。此外,还通过同源重组以293细胞包装出表达M2基因的重组腺病毒Ad-M2以及表达HA基因的重组腺病毒Ad-HA。用间接免疫荧光 (IFA) 方法检测到了各载体上插入基因的表达。按初免-加强程序分别用重组质粒pStar-M2/HA和重组腺病毒Ad-HA+Ad-M2免疫BALB/c小鼠,共免疫4次,每次间隔14 d。第1、3次用DNA疫苗,第2、4次用重组腺病毒载体疫苗,每次免疫前及末次免疫后14 d采集血清用于检测体液免疫应答,末次免疫后14 d采集脾淋巴细胞用于检测细胞免疫应答。血凝抑制 (HI) 实验检测到免疫后小鼠血清中的HI活性。ELISA实验检测到免疫后小鼠血清中抗H5N1亚型流感病毒表面蛋白的IgG抗体。ELISPOT实验检测到免疫后小鼠针对M2蛋白和HA蛋白的特异性细胞免疫应答。流感病毒M2与HA双基因共免疫的研究,为研究开发新型重组流感疫苗奠定了基础。  相似文献   

2.
Wang W  Chen H  Deng Y  Yang Y  Yin X  Wang M  Zhou JF  Shu YL  Ruan L  Tan WJ 《病毒学报》2011,27(6):594-598
本研究旨在研发经济、高效的人高致病性禽流感病毒H5N1实验疫苗并优化免疫方案。利用本实验室前期构建的含有H5N1(安徽株)结构基因的多个单顺反子(HAop和NAop)和双顺反子(HAop/M2和NAop/M1)DNA疫苗及重组痘苗病毒(天坛株)疫苗,采用不同方案(单独或联合)免疫BALB/c小鼠,初步分析了抗原特异性体液免疫(HA血凝抑制抗体,NA特异性抗体,中和抗体及M1与M2特异性抗体)和细胞免疫应答(IFN-γELIS-pot)的特点。结果表明:DNA疫苗与重组痘苗病毒(天坛株)疫苗联合免疫可以激发较强的多个抗原特异的免疫应答,尤其是体液免疫应答,明显优于DNA疫苗或重组痘苗病毒(天坛株)疫苗单独免疫;联合免疫方案中DNA疫苗初免所激发的HA与NA特异的体液免疫应答强于重组痘苗病毒(天坛株)疫苗初免,然而M1与M2特异的体液免疫应答则反之。本研究为新型H5N1疫苗的研发及免疫方案的优化奠定了基础。  相似文献   

3.
由于流感病毒容易突变,流感通用疫苗的研究势在必行.流感病毒血凝素(HA)柄部和基质蛋白2的胞外域(M2e)都是流感病毒通用疫苗的重要候选靶点.通过重叠PCR的方法用A/PR/8/34(H1N1)(简称PR8)流感病毒的M2e或者4个甘氨酸取代HA的头部,分别获得HAM2e和HA4G,然后将两种重组基因插入真核表达载体pCAGGS-P7中,制得pHAM2e和pHA4G两种DNA疫苗.通过电击免疫的方法对小鼠分别免疫pHAM2e和pHA4G,免疫3次,每次免疫间隔2周.第3次免疫2周后用5LD50的同源流感病毒感染小鼠.结果表明HAM2e组和HA4G组的小鼠均产生了特异性抗体,HAM2e组比HA4G组具有更好的抗PR8流感病毒的能力,这提示可以用M2e替换HA头部用于疫苗研发.  相似文献   

4.
通过RT-PCR扩增流行性感冒(流感)病毒HA基因,克隆至腺病毒穿梭载体pAd Track-MV,该重组质粒与腺病毒DNA共转化E.coli BJ5183,通过细菌内同源重组获得重组腺病毒DNA,将其转染293细胞获得重组腺病毒。PCR证实HA基因已整合至腺病毒基因组中,Western blot结果检测到重组病毒感染293细胞中HA的表达。重组病毒经滴鼻和灌胃两种途径免疫小鼠,结果2次免疫后滴鼻组和灌胃组均产生明显的免疫应答,血清IgG抗体滴度分别为1:10000和1:1000。除血清IgG外,还在肺灌洗液中检测到分泌型IgA。滴鼻组的免疫效果强于灌胃组。经小剂量攻毒实验显示,重组腺病毒保护率为100%。该文成功构建了表达流感病毒HA基因的非复制型重组腺病毒,重组病毒免疫小鼠可产生较好的免疫效果。  相似文献   

5.
本研究旨在研发经济、高效的人高致病性禽流感病毒H5N1实验疫苗并优化免疫方案。首先优化并合成H5N1(安徽株)的血凝素基因(HAop)和神经氨酸酶基因(NAop)并证实其正确表达,再构建含有H5N1(安徽株)结构基因的多个单顺反子(HAwt、HAop和NAop)和双顺反子(HAop/M2和NAop/M1)DNA疫苗质粒。随后,采用不同途径的体内电转法在第0周和第3周2次免疫Balb/C小鼠,初步分析了抗原特异性体液免疫(HA血凝抑制抗体,NA特异性抗体,中和抗体)和细胞免疫应答(IFN-γELISpot)的特点。结果表明:含有优化血凝素基因(HAop)和优化神经氨酸酶基因(NAop)的DNA疫苗可以快速激发较强的免疫应答,尤其是细胞免疫应答;皮内电转所激发的体液免疫应答强于肌肉电转。本研究为新型H5N1疫苗的研发以及免疫方案的优化奠定了基础。  相似文献   

6.
根据GenBank中已发表的H5亚型禽流感病毒HA基因序列,设计一对引物,通过RTPCR扩增鹅源H5亚型高致病力禽流感病毒HA基因,测序确认后,将其克隆入真核表达载体pVAX1和asdpVAX1得到重组表达载体pVAX1HA和asdpVAX1HA。将重组质粒转染P815细胞,经间接免疫荧光试验证实,HA基因在细胞内得到了瞬时表达。进一步将重组质粒转化减毒鼠伤寒沙门氏菌X4550得到两种运送DNA疫苗的重组沙门氏菌X4550(pVAX1HA)和X4550(asdpVAX1HA),以1×109CFU/只的剂量两次口服免疫BALB/c小鼠,免疫小鼠不仅可以检测到HA特异性的血清抗体应答,而且还能抵抗稳定表达H5亚型禽流感病毒HA基因的P815肥大细胞瘤的攻击,说明该运送DNA疫苗的减毒沙门氏菌系统在体内能够成功释放所携带的质粒,并且能够刺激机体产生保护性免疫应答。  相似文献   

7.
以我国分离的首株人H5N1亚型禽流感病毒(A/Anhui/1/2005)作为研究对象,通过引入内部核糖体进入位点序列(IRES),构建共表达H5N1亚型禽流感病毒膜蛋白基因M1和HA的重组腺病毒。PCR扩增禽流感病毒H5N1亚型M1和HA基因的全长可读框片段,先后亚克隆入pStar载体,然后扩增M1-IRES-HA片段并将其插入穿梭载体pShuttle-CMV,再与pAd-Easy载体在BJ5183菌中通过同源重组产生重组腺病毒载体,转化293细胞,包装出重组腺病毒Ad-M1/HA。将Ad-M1/HA感染293细胞,可观察到明显细胞病变效应,用免疫荧光及Western-blot方法均检测到M1和HA基因的表达。共表达M1和HA双基因的重组腺病毒的成功构建为开发新型重组腺病毒流感疫苗奠定了基础。  相似文献   

8.
以A/Swine/Guangdong/LM/2004(H1N1)猪流感病毒HA基因为模板,通过RT-PCR技术扩增出HA基因,并将其克隆到pCI-neo真核表达载体中,成功构建重组表达质粒pCI-HA,瞬时转染vero E6和293T细胞,通过免疫过氧化物酶单层细胞试验(Immunoperoxidase monolayer assay ,IPMA)、间接免疫荧光试验(indirect immunofluorescence assay, iIFA)和蛋白免疫印迹(Western blot,WB)实验证明,HA基因能够在哺乳动物细胞中有效表达并具有良好的生物学活性。将重组质粒三次免疫8w雌性Balb/c小鼠后,ELISA试验和中和试验结果表明该重组质粒能够诱导小鼠产生较高的抗体滴度,并具有良好的中和活性。因此为H1亚型猪流感DNA疫苗的研究奠定了理论基础。  相似文献   

9.
利用PCR扩增出猪繁殖与呼吸综合征病毒的M基因,按正确的读码框与GP5基因串联,成功构建穿梭载体pShuttle-CMV-M-GP5,经PCR、测序鉴定正确。PmeⅠ线性化后在BJ5183大肠杆菌内与腺病毒骨架载体pAdEasy-1同源重组,产生重组腺病毒DNA。重组腺病毒DNA经PacⅠ线性化后用脂质体转染HEK-293A细胞,在细胞内包装成完整的腺病毒,通过IFA可以检测到M与GP5串联的重组腺病毒构建成功,可以正确地表达目的蛋白。将构建好的重组腺病毒免疫小鼠,结果表明可以诱导产生较强的体液免疫应答(ELISA抗体和中和抗体)和细胞免疫应答(淋巴细胞增殖和CTL反应)。证明该重组腺病毒具有较好的免疫原性,为下一步猪体免疫试验奠定了基础。  相似文献   

10.
乙肝病毒DNA疫苗的构建及其诱导小鼠的免疫应答   总被引:7,自引:1,他引:6  
构建含adr亚型HBV表面抗原基因的核酸疫苗 ,考察人白细胞介素II基因及重组白细胞介素II的免疫佐剂作用。用含有人白细胞介素II基因的真核表达质粒及基因重组白细胞介素II蛋白作为佐剂 ,将编码乙型肝炎病毒表面抗原的重组真核表达质粒 pVAX/HBS免疫BALB/C小鼠 (试验组 ) ,同时设置注射质粒pVAX的阴性对照组 ,并分别于第 2 ,4周后加强免疫各 1次。试验组在第 4周时开始有HBsAb产生 ,阴性对照组未测到HbsAb ,试验组和对照组均未检测到HBsAg。乙肝病毒DNA疫苗能引起小鼠特异性体液免疫应答 ,白细胞介素II的真核表达质粒的佐剂作用不明显 ,基因重组白细胞介素II蛋白具有提高小鼠对乙肝病毒核酸疫苗免疫应答水平的佐剂活性。  相似文献   

11.
为了研究 H5N1 DNA 疫苗对小鼠和鸡的保护效率,用 H5N1 禽流感病毒 HA DNA 疫苗免疫 BALB/c 小鼠和 SPF 鸡 . 小鼠和鸡分别经电穿孔和肌肉注射免疫两次,间隔为 3 周 . 二次免疫后,用致死量的同源病毒进行攻毒实验 . 空白对照组在攻毒后全部死亡,而经电穿孔免疫的小鼠和鸡均获得了完全的保护,并能有效地抑制病毒在小鼠肺脏和鸡泄殖腔的繁殖 . 同时,电穿孔免疫的小鼠和鸡均产生了高水平的特异性抗体 . 经电穿孔免疫的小鼠攻毒后 CTL 反应明显加强 . 这些结果表明, HA DNA 疫苗能有效地保护小鼠和鸡对禽流感病毒的感染,同时也表明电穿孔免疫是 DNA 疫苗免疫的有效途径之一 .  相似文献   

12.
Conventional influenza vaccines need to be designed and manufactured yearly. However, they occasionally provide poor protection owing to antigenic mismatch. Hence, there is an urgent need to develop universal vaccines against influenza virus. Using nucleoprotein(NP) and extracellular domain of matrix protein 2(M2e) genes from the influenza A virus A/Beijing/30/95(H3N2), we constructed four recombinant vaccinia virus-based influenza vaccines carrying NP fused with one or four copies of M2e genes in different orders. The recombinant vaccinia viruses were used to immunize BALB/C mice. Humoral and cellular responses were measured, and then the immunized mice were challenged with the influenza A virus A/Puerto Rico/8/34(PR8). NP-specific humoral response was elicited in mice immunized with recombinant vaccinia viruses carrying full-length NP, while robust M2e-specific humoral response was elicited only in the mice immunized with recombinant vaccinia viruses carrying multiple copies of M2e. All recombinant viruses elicited NP-and M2e-specific cellular immune responses in mice. Only immunization with RVJ-4M2eNP induced remarkably higher levels of IL-2 and IL-10 cytokines specific to M2e. Furthermore, RVJ-4M2eNP immunization provided the highest cross-protection in mice challenged with 20 MLD_(50) of PR8. Therefore, the cross-protection potentially correlates with both NP and M2e-specific humoral and cellular immune responses induced by RVJ-4M2eNP, which expresses a fusion antigen of full-length NP preceded by four M2e repeats. These results suggest that the rational fusion of NP and multiple M2e antigens is critical toward inducing protective immune responses, and the 4M2eNP fusion antigen may be employed to develop a universal influenza vaccine.  相似文献   

13.
Du L  Leung VH  Zhang X  Zhou J  Chen M  He W  Zhang HY  Chan CC  Poon VK  Zhao G  Sun S  Cai L  Zhou Y  Zheng BJ  Jiang S 《PloS one》2011,6(1):e16555
Development of effective vaccines to prevent influenza, particularly highly pathogenic avian influenza (HPAI) caused by influenza A virus (IAV) subtype H5N1, is a challenging goal. In this study, we designed and constructed two recombinant influenza vaccine candidates by fusing hemagglutinin 1 (HA1) fragment of A/Anhui/1/2005(H5N1) to either Fc of human IgG (HA1-Fc) or foldon plus Fc (HA1-Fdc), and evaluated their immune responses and cross-protection against divergent strains of H5N1 virus. Results showed that these two recombinant vaccines induced strong immune responses in the vaccinated mice, which specifically reacted with HA1 proteins and an inactivated heterologous H5N1 virus. Both proteins were able to cross-neutralize infections by one homologous strain (clade 2.3) and four heterologous strains belonging to clades 0, 1, and 2.2 of H5N1 pseudoviruses as well as three heterologous strains (clades 0, 1, and 2.3.4) of H5N1 live virus. Importantly, immunization with these two vaccine candidates, especially HA1-Fdc, provided complete cross-clade protection against high-dose lethal challenge of different strains of H5N1 virus covering clade 0, 1, and 2.3.4 in the tested mouse model. This study suggests that the recombinant fusion proteins, particularly HA1-Fdc, could be developed into an efficacious universal H5N1 influenza vaccine, providing cross-protection against infections by divergent strains of highly pathogenic H5N1 virus.  相似文献   

14.
将我国分离的首株人H5N1亚型禽流感病毒A/Anhui/1/2005作为研究对象,扩增其HA和HA1基因片段并克隆至真核表达载体pStar,构建成真核表达质粒。通过Western blot和间接免疫荧光检测方法确认,构建的重组质粒在真核细胞中成功地表达了目的蛋白HA和HA1。将重组质粒免疫BALB/c小鼠,检测免疫后外周血中HA/HA1特异性抗体的效价,并比较HA和HA1的免疫原性。结果表明,重组质粒免疫后成功地诱导了体液免疫反应,且二者的血清抗体效价无显著性差异。  相似文献   

15.
Recurrent outbreaks of highly pathogenic avian influenza virus pose the threat of pandemic spread of lethal disease and make it a priority to develop safe and effective vaccines. Influenza virus-like particles (VLPs) have been suggested to be a promising vaccine approach. However, VLP-induced immune responses, and their roles in inducing memory immune responses and cross-protective immunity have not been investigated. In this study, we developed VLPs containing influenza virus A/PR8/34 (H1N1) hemagglutinin (HA) and matrix (M1) proteins and investigated their immunogenicity, long-term cross-protective efficacy, and effects on lung proinflammatory cytokines in mice. Intranasal immunization with VLPs containing HA induced high serum and mucosal antibody titers and neutralizing activity against PR8 and A/WSN/33 (H1N1) viruses. Mice immunized with VLPs containing HA showed little or no proinflammatory lung cytokines and were protected from a lethal challenge with mouse-adapted PR8 or WSN viruses even 5 months postimmunization. Influenza VLPs induced mucosal immunoglobulin G and cellular immune responses, which were reactivated rapidly upon virus challenge. Long-lived antibody-secreting cells were detected in the bone marrow of immunized mice. Immune sera administered intranasally were able to confer 100% protection from a lethal challenge with PR8 or WSN, which provides further evidence that anti-HA antibodies are primarily responsible for preventing infection. Taken together, these results indicate that nonreplicating influenza VLPs represent a promising strategy for the development of a safe and effective vaccine to control the spread of lethal influenza viruses.  相似文献   

16.
Despite the advantages of DNA vaccines, overcoming their lower efficacy relative to that of conventional vaccines remains a challenge. Here, we constructed a human endogenous retrovirus (HERV) envelope-coated, nonreplicable, baculovirus-based HA vaccine against swine influenza A/California/04/2009(H1N1) hemagglutin (HA) (AcHERV-sH1N1-HA) as an alternative to conventional vaccines and evaluated its efficacy in two strains of mice, BALB/c and C57BL/6. A commercially available, killed virus vaccine was used as a positive control. Mice were intramuscularly administered AcHERV-sH1N1-HA or the commercial vaccine and subsequently given two booster injections. Compared with the commercial vaccine, AcHERV-sH1N1-HA induced significantly higher levels of cellular immune responses in both BALB/c and C57BL/6 mice. Unlike cellular immune responses, humoral immune responses depended on the strain of mice. Following immunization with AcHERV-sH1N1-HA, C57BL/6 mice showed HA-specific IgG titers 10- to 100-fold lower than those of BALB/c mice. In line with the different levels of humoral immune responses, the survival of immunized mice after intranasal challenge with sH1N1 virus (A/California/04/2009) depended on the strain. After challenge with 10-times the median lethal dose (MLD50) of sH1N1 virus, 100% of BALB/c mice immunized with the commercial vaccine or AcHERV-sH1N1-HA survived. In contrast, C57BL/6 mice immunized with AcHERV-sH1N1-HA or the commercial vaccine showed 60% and 70% survival respectively, after challenge with sH1N1 virus. In all mice, virus titers and results of histological analyses of lung tissues were consistent with the survival data. Our results indicate the importance of humoral immune response as a major defense system against influenza viral infection. Moreover, the complete survival of BALB/c mice immunized with AcHERV-sH1N1-HA after challenge with sH1N1 virus suggests the potential of baculoviral vector-based vaccines to achieve an efficacy comparable to that of killed virus vaccines.  相似文献   

17.

Background

Vaccination is a cost-effective counter-measure to the threat of seasonal or pandemic outbreaks of influenza. To address the need for improved influenza vaccines and alternatives to egg-based manufacturing, we have engineered an influenza virus-like particle (VLP) as a new generation of non-egg or non-mammalian cell culture-based candidate vaccine.

Methodology/Principal Findings

We generated from a baculovirus expression system using insect cells, a non-infectious recombinant VLP vaccine from both influenza A H5N1 clade 1 and clade 2 isolates with pandemic potential. VLPs were administered to mice in either a one-dose or two-dose regimen and the immune responses were compared to those induced by recombinant hemagglutinin (rHA). Both humoral and cellular responses were analyzed. Mice vaccinated with VLPs were protected against challenge with lethal reassortant viruses expressing the H5N1 HA and NA, regardless if the H5N1 clade was homologous or heterologous to the vaccine. However, rHA-vaccinated mice showed considerable weight loss and death following challenge with the heterovariant clade virus. Protection against death induced by VLPs was independent of the pre-challenge HAI titer or cell-mediated responses to HA or M1 since vaccinated mice, with low to undetectable cross-clade HAI antibodies or cellular responses to influenza antigens, were still protected from a lethal viral challenge. However, an apparent association rate of antibody binding to HA correlated with protection and was enhanced using VLPs, particularly when delivered intranasally, compared to rHA vaccines.

Conclusion/Significance

This is the first report describing the use of an H5N1 VLP vaccine created from a clade 2 isolate. The results show that a non-replicating virus-like particle is effective at eliciting a broadened, cross-clade protective immune response to proteins from emerging H5N1 influenza isolates giving rise to a potential pandemic influenza vaccine candidate for humans that can be stockpiled for use in the event of an outbreak of H5N1 influenza.  相似文献   

18.

Background

Influenza virus remains a significant health and social concern in part because of newly emerging strains, such as avian H5N1 virus. We have developed a prototype H5N1 vaccine using a recombinant, replication-competent Adenovirus serotype 4 (Ad4) vector, derived from the U.S. military Ad4 vaccine strain, to express the hemagglutinin (HA) gene from A/Vietnam/1194/2004 influenza virus (Ad4-H5-Vtn). Our hypothesis is that a mucosally-delivered replicating Ad4-H5-Vtn recombinant vector will be safe and induce protective immunity against H5N1 influenza virus infection and disease pathogenesis.

Methodology/Principal Findings

The Ad4-H5-Vtn vaccine was designed with a partial deletion of the E3 region of Ad4 to accommodate the influenza HA gene. Replication and growth kinetics of the vaccine virus in multiple human cell lines indicated that the vaccine virus is attenuated relative to the wild type virus. Expression of the HA transgene in infected cells was documented by flow cytometry, western blot analysis and induction of HA-specific antibody and cellular immune responses in mice. Of particular note, mice immunized intranasally with the Ad4-H5-Vtn vaccine were protected against lethal H5N1 reassortant viral challenge even in the presence of pre-existing immunity to the Ad4 wild type virus.

Conclusions/Significance

Several non-clinical attributes of this vaccine including safety, induction of HA-specific humoral and cellular immunity, and efficacy were demonstrated using an animal model to support Phase 1 clinical trial evaluation of this new vaccine.  相似文献   

19.
Influenza poses a persistent worldwide threat to the human population. As evidenced by the 2009 H1N1 pandemic, current vaccine technologies are unable to respond rapidly to this constantly diverging pathogen. We tested the utility of adenovirus (Ad) vaccines expressing centralized consensus influenza antigens. Ad vaccines were produced within 2 months and protected against influenza in mice within 3 days of vaccination. Ad vaccines were able to protect at doses as low as 10(7) virus particles/kg indicating that approximately 1,000 human doses could be rapidly generated from standard Ad preparations. To generate broadly cross-reactive immune responses, centralized consensus antigens were constructed against H1 influenza and against H1 through H5 influenza. Twenty full-length H1 HA sequences representing the main branches of the H1 HA phylogenetic tree were used to create a synthetic centralized gene, HA1-con. HA1-con minimizes the degree of sequence dissimilarity between the vaccine and existing circulating viruses. The centralized H1 gene, HA1-con, induced stronger immune responses and better protection against mismatched virus challenges as compared to two wildtype H1 genes. HA1-con protected against three genetically diverse lethal influenza challenges. When mice were challenged with 1934 influenza A/PR/8/34, HA1-con protected 100% of mice while vaccine generated from 2009 A/TX/05/09 only protected 40%. Vaccination with 1934 A/PR/8/34 and 2009 A/TX/05/09 protected 60% and 20% against 1947 influenza A/FM/1/47, respectively, whereas 80% of mice vaccinated with HA1-con were protected. Notably, 80% of mice challenged with 2009 swine flu isolate A/California/4/09 were protected by HA1-con vaccination. These data show that HA1-con in Ad has potential as a rapid and universal vaccine for H1N1 influenza viruses.  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