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 187 毫秒
1.
SARS-CoV S蛋白受体结合区的重组表达及免疫原性分析   总被引:2,自引:0,他引:2  
为了表达SARS-CoV的S蛋白的受体结合区并对其免疫原性进行分析,用PCR方法扩增S蛋白的受体结合区基因片段,克隆至原核表达质粒pET-32a+并在大肠杆菌中表达,应用Western-blot鉴定表达的目的蛋白,而后以该蛋白作为诊断抗原包被酶联板来检测20份SARS病人血清和28份健康人血清,结果原核表达的S蛋白能够和所用的SARS病人血清反应.这提示表达的S重组蛋白具有良好的抗原性.将变性纯化的重组蛋白和复性蛋白分别皮下免疫小鼠,第三次免疫一周后收集抗血清,用ELISA测定抗体和同时测定中和抗体活性.用变性的抗原免疫的小鼠血清均无中和活性;而用复性的蛋白免疫的小鼠产生了中和抗体.实验表明,S蛋白受体结合区无线性中和表位,中和抗体的产生是由构象表位诱导的.提示该蛋白有可能应用于亚单位疫苗的研究.  相似文献   

2.
目的:利用昆虫杆状病毒表达系统重组表达中东呼吸综合征冠状病毒(MERS-Co V)S1蛋白,并对其免疫效果进行评价。方法:构建含有MERS-Co V S1基因的重组杆状病毒质粒,转染Sf9细胞包装杆状病毒;重组病毒传代3次获得种子病毒,感染Sf9细胞,收获感染上清,通过镍离子亲和层析纯化获得S1重组蛋白;用纯化的S1蛋白免疫BALB/c小鼠,采用ELISA检测免疫小鼠血清抗原特异性的抗体水平;采用假病毒中和试验检测血清中抗体的中和活性。结果:获得了表达MERS-Co V S1蛋白的重组病毒株,在昆虫细胞中表达并纯化了S1重组蛋白;利用重组表达的S1蛋白免疫小鼠3次,血清S1特异性Ig G抗体滴度可达1∶102 400,免疫小鼠血清稀释至1/5120后中和百分比仍达50%以上。结论:利用昆虫细胞重组表达的MERS-Co V S1蛋白具有良好的免疫原性,并能有效诱导产生高滴度中和抗体,为发展MERS-Co V重组蛋白疫苗奠定了基础。  相似文献   

3.
为探讨SARS-CoV的M蛋白的免疫学特性以及M蛋白作为SARS-CoV病毒疫苗组分的可行性和必要性.分别用pET-15b和pET-22b在大肠杆菌中表达SARS-CoV的M蛋白,亲和层析纯化后作为抗原应用.同时,将M蛋白的编码基因克隆进分泌型真核表达载体pSecTagB中得到重组质粒pSecM作为DNA疫苗,免疫BALB/c小白鼠、制备SARS-CoV M蛋白的抗血清.并用纯化后的M蛋白建立的SARS-CoV M抗体ELISA检测技术研究所构建的M-DNA疫苗的免疫效果.结果表明:两种重组M蛋白在大肠杆菌中均以可溶性形式得到高效表达,经与华大产的用灭活SARS全病毒制备的SARS-CoV抗体ELISA检测试剂盒比较实验,证明该原核表达的重组M蛋白能与SARS确诊病人血清以及M-DNA免疫鼠血清发生特异性抗原抗体反应.这两种重组M蛋白有可能作为抗原组分用于临床SARS-CoV检测中;所构建的SARS-CoV的M基因核酸疫苗能在小鼠体内产生特异性抗体,提示M蛋白在SARS-CoV疫苗尤其是组分疫苗的研制中应加以考虑,为DNA疫苗的开发提供了依据.  相似文献   

4.
为探讨SARS-CoV的M蛋白的免疫学特性以及M蛋白作为SARS-CoV病毒疫苗组分的可行性和必要性.分别用pET-15b和pET-22b在大肠杆菌中表达SARS-CoV的M蛋白,亲和层析纯化后作为抗原应用.同时,将M蛋白的编码基因克隆进分泌型真核表达载体pSecTagB中得到重组质粒pSecM作为DNA疫苗,免疫BALB/c小白鼠、制备SARS-CoV M蛋白的抗血清.并用纯化后的M蛋白建立的SARS-CoV M抗体ELISA检测技术研究所构建的M-DNA疫苗的免疫效果.结果表明两种重组M蛋白在大肠杆菌中均以可溶性形式得到高效表达,经与华大产的用灭活SARS全病毒制备的SARS-CoV抗体ELISA检测试剂盒比较实验,证明该原核表达的重组M蛋白能与SARS确诊病人血清以及M-DNA免疫鼠血清发生特异性抗原抗体反应.这两种重组M蛋白有可能作为抗原组分用于临床SARS-CoV检测中;所构建的SARS-CoV的M基因核酸疫苗能在小鼠体内产生特异性抗体,提示M蛋白在SARS-CoV疫苗尤其是组分疫苗的研制中应加以考虑,为DNA疫苗的开发提供了依据.  相似文献   

5.
目的:利用Bac-to-Bac1杆状病毒系统,在sf9昆虫细胞中表达严重急性呼吸综合征(SARS)冠状病毒(SARS-CoV)的S受体结合区蛋白片段,并对其免疫原性进行研究。方法:将S蛋白的受体结合区基因片段定向克隆至转座载体pFast-Bac1,转化大肠杆菌DH10Bac感受态细胞,用抗生素平板筛选重组杆粒。脂质体介导重组杆粒转染sf9昆虫细胞,待细胞形态明显改变后收获细胞和培养上清液。利用SARS病人恢复期抗血清做ELISA和Western印迹,分析重组蛋白的抗原性。结果:ELISA和Western印迹表明,在sf9昆虫细胞中表达的SARS-CoVS受体结合区重组蛋白可与SARS病人恢复期抗血清发生特异反应。结论:获得了在昆虫细胞内表达的SARS-CoVS受体结合区重组蛋白,并证明该蛋白有可能用于SARS感染的抗体检测,为SARS-CoV免疫机制及其疫苗的进一步研究奠定了基础。  相似文献   

6.
PCR扩增呼吸道合胞病毒(respiratory syncytial virus,RSV)M2 蛋白的CD8+T细胞表位F/M2:81-95和RSV-G蛋白的B细胞表位片段G:125~225(简称G1),以一个Linker连接,插入质粒pET-DsbA中构建原核表达重组质粒, 转染E.coli BL21(DE3)后成功表达了融合蛋白DsbA-G1-Linker-F/M2:81-95(简称D-G1LF/M2),Western-blot结果表明该融合蛋白是RSV特异性的,采用Ni+螯合亲和层析法纯化变性的包涵体溶液,经梯度透析法复性,用该蛋白免疫BALB/c小鼠,结果表明被免疫小鼠肺部及血清中产生了高滴度的抗D-G1LF/M2及抗RSV IgG抗体和中和抗体,同时还诱导产生了RSV特异性的CTL应答;IgG的亚型IgG1/IgG2a的比值为2.66;用RSV攻击免疫后的小鼠,病毒滴定法检测肺部RSV滴度,结果表明D-G1LF/M2对小鼠肺部具有保护作用。  相似文献   

7.
构建SARS冠状病毒M蛋白膜内区(Mc蛋白)基因(Mc区)融合表达载体,表达并复性该蛋白。克隆Mc区,构建融合表达载体pET-32a(+)/Mc,融合表达Mc蛋白。纯化表达蛋白,采用逐步透析、降低蛋白浓度、加入氧化还原剂复性融合蛋白。用复性蛋白免疫兔产生抗血清,复性蛋白与SARS患者血清和健康人血清反应鉴定复性结果。结果显示,复性蛋白免疫家兔后产生抗血清,该复性蛋白与SARS患者血清特异结合,表明已成功复性该表达蛋白。为SARS疫苗和诊断试剂的研制奠定基础。  相似文献   

8.
制备含破伤风毒素肽(tetanus toxin,TT)、促吞噬肽(tuftsin)和新型冠状病毒刺突蛋白(spike,S蛋白)受体结合域(receptor-binding domain,RBD)的融合蛋白,探讨分子内佐剂对RBD蛋白体液免疫和细胞免疫效果的影响。将破伤风毒素肽、促吞噬肽与S蛋白RBD区域通过柔性多肽串联,密码子优化后构建重组载体,原核表达纯化制备重组S-TT-tuftsin蛋白,与铝佐剂混合后免疫BALB/c小鼠,对其体液及细胞免疫效果进行评价。重组S-TT-tuftsin蛋白以包涵体形式表达,离子交换层析纯化后采用梯度透析进行复性,复性蛋白经Dot blotting鉴定,可与新冠亚单位疫苗(安徽智飞公司)免疫后人血清发生反应。小鼠免疫实验结果表明,免疫35 d时抗体水平到达平台期,含分子内佐剂重组蛋白(铝佐剂)免疫小鼠后血清ELISA抗体效价高达1︰66240,显著高于S-RBD蛋白(铝佐剂)免疫小鼠抗体效价(P<0.05)。同时,含分子内佐剂重组蛋白刺激小鼠产生更强的淋巴细胞增殖能力,刺激指数可达4.71±0.15,相较于S-RBD蛋白的刺激指数1.83±0.09具有显著性差异(P<0.0001)。分子内佐剂破伤风毒素肽和促吞噬肽可显著增强新冠S蛋白RBD域的体液免疫和细胞免疫效果,可为新冠亚单位疫苗和其他病毒亚单位疫苗的研制提供理论基础和参考。  相似文献   

9.
尼帕病毒融合蛋白和受体结合蛋白基因DNA免疫的研究   总被引:1,自引:0,他引:1  
构建了表达哺乳动物密码子优化的NiV囊膜蛋白F和G基因的真核表达质粒pCAGG-NiV-F和pCAGG-NiV-G.细胞融合试验表明,重组NiV融合蛋白F和受体结合蛋白G在pCAGG-NiV-F、pCAGG-NiV-G共转染BHK细胞中获得表达,并具有良好生物学活性.真核表达质粒pCAGG-NiV-F、pCAGG-NiV-G和pCAGG-NiV-F pCAGG-NiV-G DNA分别按100μg/只的剂量肌肉注射免疫6周龄BALB/c小鼠,间隔4周加强免疫,第二次加强免疫3周后采血,分离血清备用.分别以重组杆状病毒感染Sf9细胞表达的重组NiV融合蛋白(rNF)和受体结合蛋白(rNG)为包被抗原,应用间接ELISA检测上述质粒DNA免疫血清中的特异性抗体,具有较高的敏感性和特异性.另外,中和试验结果表明,DNA免疫小鼠产生的特异抗体可有效中和NiV囊膜蛋白F和G介导的伪型VSV重组病毒侵入NiV易感宿主细胞的感染性,并且受体结合蛋白G基因DNA诱导中和抗体的滴度高于融合蛋白F基因DNA.结果表明,DNA疫苗具有防制尼帕病毒性脑炎的潜力.  相似文献   

10.
目的:构建蜱传脑炎病毒(TBEV)结构蛋白E的原核表达载体,表达重组蛋白。方法:PCR扩增E蛋白全长基因片段,插入原核表达载体pET-32a并转化大肠杆菌进行诱导表达,镍柱纯化、复性。结果:E蛋白在大肠杆菌中获得表达,表达量达60 mg/L;重组E蛋白能与人抗TBEV多克隆抗体产生特异反应;用重组E抗原免疫家兔后,能检测到较高的抗体水平。结论:原核表达的E抗原蛋白具有抗体结合活性,可用于制备ELISA诊断试剂。  相似文献   

11.
SARS 冠状病毒 S 蛋白受体结合结构域的表达及其表位作图   总被引:1,自引:1,他引:0  
严重急性呼吸综合征 (SARS) 是一种新出现的人类传染病,该病的病原是 SARS 冠状病毒 (SARS-CoV). S 蛋白是 SARS 冠状病毒的一种主要结构蛋白,它在病毒与宿主细胞受体结合以及诱导机体产生中和抗体中起重要作用 . 研究表明 S 蛋白与受体结合的核心区域为第 318 ~ 510 氨基酸残基的片段 . 首先克隆并用 pGEX-6p-1 载体融合表达了该受体结合结构域,并且通过蛋白质印迹分析表明,该受体结合结构域融合蛋白能被 SARS 康复患者血清和 S 蛋白特异的单克隆抗体所识别 . 为了对这一区域进行抗原表位作图,进一步设计了一套 23 个覆盖受体结合结构域的长 16 个氨基酸残基的部分重叠短肽,并进行了 GST 融合表达 . 用免疫动物血清和单克隆抗体 D3D1 对 23 个融合蛋白进行蛋白质印迹和 ELISA 免疫反应性分析,结果鉴定出两个抗原表位 SRBD3(F334PSVYAWERKKISNCV349) 和表位 D3D1 (K447LRPFERDI455). 其结果对进一步分析 S 蛋白结构与功能以及诊断试剂和基因工程疫苗的研究有一定意义 .  相似文献   

12.
Severe acute respiratory syndrome (SARS) is an emerging infectious disease associated with a novel coronavirus and causing worldwide outbreaks. SARS coronavirus (SARS-CoV) is an enveloped RNA virus, which contains several structural proteins. Among these proteins, spike (S) protein is responsible for binding to specific cellular receptors and is a major antigenic determinant, which induces neutralizing antibody. In order to analyze the antigenicity and receptor-binding ability of SARS-CoV S protein, we expressed the S protein in Escherichia coli using a pET expression vector. After the isopropyl-beta-D-thiogalactoside induction, S protein was expressed in the soluble form and purified by nickel-affinity chromatography to homogeneity. The amount of S protein recovered was 0.2-0.3mg/100ml bacterial culture. The S protein was recognized by sera from SARS patients by ELISA and Western blot, which indicated that recombinant S protein retained its antigenicity. By biotinylated ELISA and Western blot using biotin-labeled S protein as the probe, we identified 130-kDa and 140-kDa proteins in Vero cells that might be the cellular receptors responsible for SARS-CoV infection. Taken together, these results suggested that recombinant S protein exhibited the antigenicity and receptor-binding ability, and it could be a good candidate for further developing SARS vaccine and anti-SARS therapy.  相似文献   

13.
Chen Z  Zhang L  Qin C  Ba L  Yi CE  Zhang F  Wei Q  He T  Yu W  Yu J  Gao H  Tu X  Gettie A  Farzan M  Yuen KY  Ho DD 《Journal of virology》2005,79(5):2678-2688
Immunization with a killed or inactivated viral vaccine provides significant protection in animals against challenge with certain corresponding pathogenic coronaviruses (CoVs). However, the promise of this approach in humans is hampered by serious concerns over the risk of leaking live severe acute respiratory syndrome (SARS) viruses. In this study, we generated a SARS vaccine candidate by using the live-attenuated modified vaccinia virus Ankara (MVA) as a vector. The full-length SARS-CoV envelope Spike (S) glycoprotein gene was introduced into the deletion III region of the MVA genome. The newly generated recombinant MVA, ADS-MVA, is replication incompetent in mammalian cells and highly immunogenic in terms of inducing potent neutralizing antibodies in mice, rabbits, and monkeys. After two intramuscular vaccinations with ADS-MVA alone, the 50% inhibitory concentration in serum was achieved with reciprocal sera dilutions of more than 1,000- to 10,000-fold in these animals. Using fragmented S genes as immunogens, we also mapped a neutralizing epitope in the region of N-terminal 400 to 600 amino acids of the S glycoprotein (S400-600), which overlaps with the angiotensin-converting enzyme 2 (ACE2) receptor-binding region (RBR; S318-510). Moreover, using a recombinant soluble RBR-Fc protein, we were able to absorb and remove the majority of the neutralizing antibodies despite observing that the full S protein tends to induce a broader spectrum of neutralizing activities in comparison with fragmented S proteins. Our data suggest that a major mechanism for neutralizing SARS-CoV likely occurs through blocking the interaction between virus and the cellular receptor ACE2. In addition, ADS-MVA induced potent immune responses which very likely protected Chinese rhesus monkeys from pathogenic SARS-CoV challenge.  相似文献   

14.
The receptor-binding domain (RBD) of SARS coronavirus (SARS-CoV) spike (S) protein contains multiple conformation-dependent epitopes that induce neutralizing antibody responses. Here we used CHO-K1 cells to establish a cell line for stable expression of a 193-mer (residues 318-510) RBD (RBD193-CHO) and determined its antigenicity and immunogenicity. We found that RBD193-CHO reacted strongly with a panel of six monoclonal antibodies recognizing various conformational and linear epitopes in RBD, suggesting that this recombinant protein maintains intact conformation and good antigenicity. Immunization of mice with RBD193-CHO resulted in induction of high titers of RBD-specific neutralizing antibodies and potent IL-4-expressing T cell responses. RBD193-CHO induced immunity that protected a majority of the vaccinated mice from SARS-CoV challenge. These results suggest that the recombinant RBD produced in an established stable cell line maintains strong immunogenicity with high potential for use as an effective and economic subunit SARS vaccine.  相似文献   

15.
A novel coronavirus, the severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV), was identified as the causative agent of SARS. The profile of specific antibodies to individual proteins of the virus is critical to the development of vaccine and diagnostic tools. In this study, 13 recombinant proteins associated with four structural proteins (S, E, M and N) and five putative uncharacterized proteins (3a, 3b, 6, 7a and 9b) of the SARS-CoV were prepared and used for screening and monitoring their specific IgG antibodies in SARS patient sera by protein microarray. Antibodies to proteins S, 3a, N and 9b were detected in the sera from convalescent-phase SARS patients, whereas those to proteins E, M, 3b, 6 and 7a were undetected. In the detectable specific antibodies, anti-S and anti-N were dominant and could persist in the sera of SARS patients until week 30. Among the rabbit antisera to recombinant proteins S3, N, 3a and 9b, only anti-S3 serum showed significant neutralizing activity to the SARS-CoV infection in Vero E6 cells. The results suggest (1) that anti-S and anti-N antibodies are diagnostic markers and in particular that S3 is immunogenic and therefore is a good candidate as a subunit vaccine antigen; and (2) that, from a virus structure viewpoint, the presence in some human sera of antibodies reacting with two recombinant polypeptides, 3a and 9b, supports the hypothesis that they are synthesized during the virus cycle.  相似文献   

16.
The spike (S) glycoprotein is one of the major structure proteins of SARS-associated coronavirus (CoV). Fragment 450-650 (S450-650) of the S protein contains receptor-binding domain and neutralizing epitopes. In this study, S450-650 was expressed with a histidine tag in Escherichia coli BL21. Bacterial inclusion bodies containing the recombinant S450-650 were solubilized with 8 M urea and then applied onto a Ni-nitrilotriacetic acid column. On-column refolding and purification was performed. Reduced glutathione and oxidized glutathione were included in the refolding buffer. In the wash and elution buffers, glycerol and glucose were necessary additives to prevent protein aggregation during purification. This refolding and purification procedure allowed production of S450-650 at up to 500 microg/ml in soluble form, which maintained appropriate antigenicity and immunogenicity. It was able to induce strong IgG responses in BALB/c mice. In Western blot assays, the recombinant S450-650 was recognized by monoclonal Ab against the His-tag and also sera from a convalescent SARS patient. S450-650-based ELISA system was able to detect anti-SARS-CoV IgG Abs in patient sera.  相似文献   

17.
He Y  Li J  Heck S  Lustigman S  Jiang S 《Journal of virology》2006,80(12):5757-5767
The spike (S) glycoprotein of severe acute respiratory syndrome coronavirus (SARS-CoV) mediates the receptor interaction and immune recognition and is considered a major target for vaccine design. However, its antigenic and immunogenic properties remain to be elucidated. In this study, we immunized mice with full-length S protein (FL-S) or its extracellular domain (EC-S) expressed by recombinant baculoviruses in insect cells. We found that the immunized mice developed high titers of anti-S antibodies with potent neutralizing activities against SARS pseudoviruses constructed with the S proteins of Tor2, GD03T13, and SZ3, the representative strains of 2002 to 2003 and 2003 to 2004 human SARS-CoV and palm civet SARS-CoV, respectively. These data suggest that the recombinant baculovirus-expressed S protein vaccines possess excellent immunogenicity, thereby inducing highly potent neutralizing responses against human and animal SARS-CoV variants. The antigenic structure of the S protein was characterized by a panel of 38 monoclonal antibodies (MAbs) isolated from the immunized mice. The epitopes of most anti-S MAbs (32 of 38) were localized within the S1 domain, and those of the remaining 6 MAbs were mapped to the S2 domain. Among the anti-S1 MAbs, 17 MAbs targeted the N-terminal region (amino acids [aa] 12 to 327), 9 MAbs recognized the receptor-binding domain (RBD; aa 318 to 510), and 6 MAbs reacted with the C-terminal region of S1 domain that contains the major immunodominant site (aa 528 to 635). Strikingly, all of the RBD-specific MAbs had potent neutralizing activity, 6 of which efficiently blocked the receptor binding, confirming that the RBD contains the main neutralizing epitopes and that blockage of the receptor association is the major mechanism of SARS-CoV neutralization. Five MAbs specific for the S1 N-terminal region exhibited moderate neutralizing activity, but none of the MAbs reacting with the S2 domain and the major immunodominant site in S1 showed neutralizing activity. All of the neutralizing MAbs recognize conformational epitopes. These data provide important information for understanding the antigenicity and immunogenicity of S protein and for designing SARS vaccines. This panel of anti-S MAbs can be used as tools for studying the structure and function of the SARS-CoV S protein.  相似文献   

18.
The spike (S) protein of severe acute respiratory syndrome coronavirus (SARS-CoV) has two major functions: interacting with the receptor to mediate virus entry and inducing protective immunity. Coincidently, the receptor-binding domain (RBD, residues 318-510) of SAR-CoV S protein is a major antigenic site to induce neutralizing antibodies. Here, we used RBD-Fc, a fusion protein containing the RBD and human IgG1 Fc, as a model in the studies and found that a single amino acid substitution in the RBD (R441A) could abolish the immunogenicity of RBD to induce neutralizing antibodies in immunized mice and rabbits. With a panel of anti-RBD mAbs as probes, we observed that R441A substitution was able to disrupt the majority of neutralizing epitopes in the RBD, suggesting that this residue is critical for the antigenic structure responsible for inducing protective immune responses. We also demonstrated that the RBD-Fc bearing R441A mutation could not bind to soluble and cell-associated angiotensin-converting enzyme 2 (ACE2), the functional receptor for SARS-CoV and failed to block S protein-mediated pseudovirus entry, indicating that this point mutation also disrupted the receptor-binding motif (RBM) in the RBD. Taken together, these data provide direct evidence to show that a single amino acid residue at key position in the RBD can determine the major function of SARS-CoV S protein and imply for designing SARS vaccines and therapeutics.  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