baculovirus expression system

杆状病毒表达系统
  • 文章类型: Journal Article
    禽流感对家禽生产的巨大影响以及对人类可能的健康风险已引起人们对这种疾病的极大关注。H9N2亚型禽流感病毒在禽类中广泛流行,通过感染或通过向各种人畜共患禽流感毒株提供内部基因对人类构成直接威胁。尽管广泛使用H9N2亚型疫苗,由于流感病毒的快速抗原漂移和转移,病毒的爆发持续存在。因此,开发更广泛的H9N2亚型禽流感疫苗并评估其有效性至关重要。在这项研究中,通过生物信息学分析和杆状病毒表达系统(BES)获得了表达广谱HA蛋白的重组杆状病毒。这种重组血凝素(HA)蛋白对几种分支H9亚型AIV的阳性血清表现出交叉反应性。然后使用佐剂和纯化的HA蛋白来产生rHA疫苗候选物。对疫苗的评估表明,用rHA候选疫苗对颈部进行皮下免疫刺激了强烈的免疫反应,针对各种H9N2病毒挑战提供完整的临床保护。此外,rHA比商业疫苗更有效地抑制病毒脱落。因此,我们的发现说明了rHA候选疫苗在保护鸡免受H9N2病毒攻击方面的功效,强调其作为传统疫苗替代品的潜力。
    The enormous effects of avian influenza on poultry production and the possible health risks to humans have drawn much attention to this disease. The H9N2 subtype of avian influenza virus is widely prevalent among poultry, posing a direct threat to humans through infection or by contributing internal genes to various zoonotic strains of avian influenza. Despite the widespread use of H9N2 subtype vaccines, outbreaks of the virus persist due to the rapid antigenic drift and shifts in the influenza virus. As a result, it is critical to develop a broader spectrum of H9N2 subtype avian influenza vaccines and evaluate their effectiveness. In this study, a recombinant baculovirus expressing the broad-spectrum HA protein was obtained via bioinformatics analysis and a baculovirus expression system (BES). This recombinant hemagglutinin (HA) protein displayed cross-reactivity to positive sera against several subbranch H9 subtype AIVs. An adjuvant and purified HA protein were then used to create an rHA vaccine candidate. Evaluation of the vaccine demonstrated that subcutaneous immunization of the neck with the rHA vaccine candidate stimulated a robust immune response, providing complete clinical protection against various H9N2 virus challenges. Additionally, virus shedding was more effectively inhibited by rHA than by the commercial vaccine. Thus, our findings illustrate the efficacy of the rHA vaccine candidate in shielding chickens against the H9N2 virus challenge, underscoring its potential as an alternative to conventional vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    作为一种高致病性禽流感病毒,H5流感对牲畜构成严重威胁,家禽业,和公共卫生安全。血凝素(HA)是流感中和抗体的优势表位和主要靶标。这里,我们设计了一种纳米血凝素流感疫苗来提高流感疫苗的免疫原性.在这项研究中,以HA5亚型流感病毒为候选抗原,与人工设计的双分支支架蛋白I53_dn5A和B结合,利用昆虫杆状病毒表达系统通过分泌纯化获得结构正确、具有生物活性的三聚体HA5-I53_dn5B/Y98F;利用原核表达系统纯化获得I53_dn5A。HA5-I53_dn5B/Y98F和I53_dn5A在体外自组装成球形纳米颗粒(HA5-I53_dn5),直径约为45nm。免疫和血清检测结果表明,HA5-I53_dn5B/Y98F和HA5-I53_dn5均能诱导HA5特异性抗体;HA5-I53_dn5的免疫原性优于HA5-I53_dn5B/Y98F。用HA5-I53_dn5B和HA5-I53_dn5纳米颗粒处理的组产生的IgG抗体滴度与含有纳米颗粒的佐剂组的IgG抗体滴度没有统计学差异。这种利用杆状病毒表达的三聚HA5-I53_dn5B和HA5-I53_dn5纳米粒的生产为开发新型,安全,和有效的流感疫苗。
    As a highly pathogenic avian virus, H5 influenza poses a serious threat to livestock, the poultry industry, and public health security. Hemagglutinin (HA) is both the dominant epitope and the main target of influenza-neutralizing antibodies. Here, we designed a nanoparticle hemagglutinin influenza vaccine to improve the immunogenicity of the influenza vaccine. In this study, HA5 subtype influenza virus was used as the candidate antigen and was combined with the artificially designed double-branch scaffold protein I53_dn5 A and B. A structurally correct and bioactive trimer HA5-I53_dn5B/Y98F was obtained through secretion and purification using an insect baculovirus expression system; I53_dn5A was obtained by purification using a prokaryotic expression system. HA5-I53_dn5B/Y98F and I53_dn5A self-assembled into spherical nanoparticles (HA5-I53_dn5) in vitro with a diameter of about 45 nm. Immunization and serum test results showed that both HA5-I53_dn5B/Y98F and HA5-I53_dn5 could induce HA5-specific antibodies; however, the immunogenicity of HA5-I53_dn5 was better than that of HA5-I53_dn5B/Y98F. Groups treated with HA5-I53_dn5B and HA5-I53_dn5 nanoparticles produced IgG antibody titers that were not statistically different from those of the nanoparticle-containing adjuvant group. This production of trimerized HA5-I53_dn5B and HA5-I53_dn5 nanoparticles using baculovirus expression provides a reference for the development of novel, safe, and efficient influenza vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    流感病毒可引起高度传染性的呼吸道疾病,构成值得注意的流行病和大流行威胁。疫苗接种是预防流感及其并发症的最具成本效益的干预措施。然而,依赖胚胎鸡蛋进行商业流感疫苗生产存在潜在风险,包括由于HA基因突变导致的疗效降低和由于可扩展性挑战导致的供应延迟。因此,迫切需要替代平台来替代基于鸡蛋的方法,并有效满足日益增长的疫苗需求。在这项研究中,我们使用杆状病毒表达载体系统来设计HA,NA,和M1基因来自季节性流感毒株A/H1N1,A/H3N2,B/Yamagata,B/维多利亚,产生病毒样颗粒(VLP)疫苗抗原,H1N1-VLP,H3N2-VLP,Yamagata-VLP,维多利亚-VLP然后我们评估了它们的功能和抗原特性,包括血凝试验,蛋白质组成,形态学,稳定性,和免疫原性。我们发现重组VLP表现出功能活性,在形态和大小上类似流感病毒粒子,同时保持结构完整性。在小鼠中的比较免疫原性评估显示,与商业重组HA和基于卵的疫苗(Vaxigrip)相比,我们的四价VLP在诱导针对同源病毒的血凝抑制和中和抗体滴度方面是一致的。研究结果强调了基于昆虫细胞的VLP疫苗是四价季节性流感疫苗的有希望的候选疫苗。值得进一步研究。
    Influenza viruses can cause highly infectious respiratory diseases, posing noteworthy epidemic and pandemic threats. Vaccination is the most cost-effective intervention to prevent influenza and its complications. However, reliance on embryonic chicken eggs for commercial influenza vaccine production presents potential risks, including reductions in efficacy due to HA gene mutations and supply delays due to scalability challenges. Thus, alternative platforms are needed urgently to replace egg-based methods and efficiently meet the increasing demand for vaccines. In this study, we employed a baculovirus expression vector system to engineer HA, NA, and M1 genes from seasonal influenza strains A/H1N1, A/H3N2, B/Yamagata, and B/Victoria, generating virus-like particle (VLP) vaccine antigens, H1N1-VLP, H3N2-VLP, Yamagata-VLP, and Victoria-VLP. We then assessed their functional and antigenic characteristics, including hemagglutination assay, protein composition, morphology, stability, and immunogenicity. We found that recombinant VLPs displayed functional activity, resembling influenza virions in morphology and size while maintaining structural integrity. Comparative immunogenicity assessments in mice showed that our quadrivalent VLPs were consistent in inducing hemagglutination inhibition and neutralizing antibody titers against homologous viruses compared to both commercial recombinant HA and egg-based vaccines (Vaxigrip). The findings highlight insect cell-based VLP vaccines as promising candidates for quadrivalent seasonal influenza vaccines. Further studies are worth conducting.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: The COVID-19 pandemic caused by SARS-CoV-2 has created serious health problems worldwide. The most effective way to prevent the occurrence of new epidemic outbreaks is vaccination. One of the modern and effective approaches to vaccine development is the use of virus-like particles (VLPs). The aim of the study is to develop a technology for production of VLP based on recombinant SARS-CoV-2 proteins (E, M, N and S) in insect cells.
    METHODS: Synthetic genes encoding coronavirus proteins E, M, N and S were used. VLP with various surface proteins of strains similar to the Wuhan virus, Delta, Alpha and Omicron were developed and cloned into the pFastBac plasmid. The proteins were synthesized in the baculovirus expression system and assembled into VLP in the portable Trichoplusia ni cell. The presence of insertion in the baculovirus genome was determined by PCR. ELISA and immunoblotting were used to study the antigenic activity of VLP. VLP purification was performed by ultracentrifugation using 20% sucrose. Morphology was assessed using electron microscopy and dynamic light scattering.
    RESULTS: VLPs consisting of recombinant SARS-CoV-2 proteins (S, M, E and N) were obtained and characterized. The specific binding of antigenic determinants in synthesized VLPs with antibodies to SARS-CoV-2 proteins has been demonstrated. The immunogenic properties of VLPs have been studied.
    CONCLUSIONS: The production and purification of recombinant VLPs consisting of full-length SARS-CoV-2 proteins with a universal set of surface antigens have been developed and optimized. Self-assembling particles that mimic the coronavirus virion induce a specific immune response against SARS-CoV-2.
    Введение. Пандемия COVID-19, вызванная коронавирусом SARS-CoV-2, породила серьезные проблемы в здравоохранении по всему миру. Ученым в кратчайшие сроки пришлось решать задачи по разработке методов лечения и профилактики этого заболевания. Наиболее эффективным способом прерывания развивающихся новых эпидемических вспышек является вакцинация. Одним из современных и эффективных подходов при разработке вакцин является использование вирусоподобных частиц (Virus like particles, VLP). Цель исследования – разработать технологию получения VLP на основе рекомбинантных белков SARS-CoV-2 (E, M, N и S), продуцируемых в клетках насекомых, и дать их комплексную характеристику. Материалы и методы. Источником вирусных белков послужили синтетические гены, кодирующие белки коронавируса E, M, N и S. Были разработаны VLP с разными поверхностными S-белками 4 штаммов коронавируса: подобный вирусу Ухань, Delta, Alpha и Omicron, клонированные в плазмиду pFastBac. Белки были синтезированы в бакуловирусной системе экспрессии и собраны в VLP в перевиваемой линии клеток Trichoplusia ni (T.ni). Синтез генов, клонирование в трансферные плазмиды и получение рекомбинантных бакуловирусов проводили стандартными методами. Наличие вставки в геноме бакуловируса определяли методом полимеразной цепной реакции. Для исследования антигенной активности VLP применяли иммуноферментный анализ, иммуноблоттинг. Очистку VLP проводили ультрацентрифугированием через 20% сахарозу. Оценку морфологии выполняли с помощью электронной микроскопии и методом динамического светорассеяния. Результаты. Получены и охарактеризованы VLP, состоящие из рекомбинантных белков S, M, E и N, на основе консенсусных последовательностей, циркулирующих в мире геновариантов SARS-CoV-2. Показана специфичность антигенных детерминант синтезированных VLP антителам, формирующимся к белкам SARS-CoV-2, изучены иммуногенные свойства VLP. Заключение. Разработаны способы получения и очистки VLP с универсальным набором поверхностных антигенов, способных к самосборке и индуцирующих специфический иммунитет против SARS-CoV-2.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    H7N9亚型禽流感病毒(AIV)对养禽业提出了伟大的挑衅。病毒样颗粒(VLP)是传统的基于鸡蛋的流感疫苗的潜在替代品。N-连接糖基化(NLG)调节流感疫苗的功效,而NLG修饰对流感VLP疫苗效力的影响尚不清楚.这里,H7N9VLP通过与表达NLG修饰的血凝素(HA)的杆状病毒共感染在昆虫细胞中组装,神经氨酸酶和基质蛋白,并在鸡和小鼠中评估VLP疫苗。NLG修饰显着增强小鼠的血凝抑制和病毒中和抗体反应,而不是鸡,因为在这些动物模型中使用了不同的免疫策略。双NLG在残基133和158处的存在显著提高了鸡和小鼠中的HA结合IgG滴度。VLP疫苗在鸡和小鼠中用H7N9病毒攻击后赋予完全保护并显著抑制病毒复制和肺病理学。VLP免疫激活了T细胞免疫相关的细胞因子反应并抑制了小鼠肺中的炎性细胞因子反应。值得注意的是,双NLG在残基133和158的存在优化了VLP疫苗刺激白细胞介素-4表达的能力,抑制病毒脱落或减轻鸡或小鼠的肺部病理。有趣的是,在第133位残基添加NLG的VLP疫苗在鸡和小鼠中提供了针对H5Nx亚型AIV的部分交叉保护。总之,HA中残基133和158处的双NLG可潜在地用于增强H7N9VLP疫苗在鸡和哺乳动物中的功效。
    H7N9 subtype avian influenza virus (AIV) poses a great challenge to poultry industry. Virus-like particle (VLP) is a prospective alternative for the traditional egg-based influenza vaccines. N-linked glycosylation (NLG) regulates the efficacy of influenza vaccines, whereas the impact of NLG modifications on the efficacy of influenza VLP vaccines remains unclear. Here, H7N9 VLPs were assembled in insect cells through co-infection with the baculoviruses expressing the NLG-modified hemagglutinin (HA), neuraminidase and matrix proteins, and the VLP vaccines were assessed in chickens and mice. NLG modifications significantly enhanced hemagglutination-inhibition and virus neutralization antibody responses in mice, rather than in chickens, because different immunization strategies were used in these animal models. The presence of dual NLG at residues 133 and 158 significantly elevated HA-binding IgG titers in chickens and mice. The VLP vaccines conferred complete protection and significantly suppressed virus replication and lung pathology post challenge with H7N9 viruses in chickens and mice. VLP immunization activated T cell immunity-related cytokine response and inhibited inflammatory cytokine response in mouse lung. Of note, the presence of dual NLG at residues 133 and 158 optimized the capacity of the VLP vaccine to stimulate interleukin-4 expression, inhibit virus shedding or alleviate lung pathology in chickens or mice. Intriguingly, the VLP vaccine with NLG addition at residue 133 provided partial cross-protection against the H5Nx subtype AIVs in chickens and mice. In conclusion, dual NLG at residues 133 and 158 in HA can be potentially used to enhance the efficacy of H7N9 VLP vaccines in chickens and mammals.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    许多蛋白质表达系统主要用于产生单一的,特异性重组蛋白。相比之下,大多数生物过程,如病毒组装依赖于几个相互作用的蛋白质的复合物,而不是单一蛋白质的活性。杆状病毒基因组的高度复杂性,加上包含不同转录步骤的多相复制周期,使其成为操纵单一高水平表达的理想系统,或多重共表达,单个细胞内的外来蛋白质。我们已经开发并利用了一系列重组杆状病毒系统来解开复杂的无包膜模型病毒的顺序组装过程,蓝舌病毒(BTV)。杆状病毒系统表达的高蛋白质产量不仅促进了每种病毒蛋白的结构功能分析,而且有利于晶体学研究,并支持重组病毒蛋白的第一个原子级分辨率。主要的BTV衣壳蛋白。Further,重组双壳病毒样颗粒(VLP)的形成提供了对BTV四种主要结构蛋白之间结构-功能关系的见解,同时也代表了病毒疫苗的潜在候选者.杆状病毒多基因表达系统促进了结构复杂病毒(无包膜和包膜病毒)的研究,并预示着新一代的病毒疫苗。
    Many protein expression systems are primarily utilised to produce a single, specific recombinant protein. In contrast, most biological processes such as virus assembly rely upon a complex of several interacting proteins rather than the activity of a sole protein. The high complexity of the baculovirus genome, coupled with a multiphase replication cycle incorporating distinct transcriptional steps, made it the ideal system to manipulate for high-level expression of a single, or co-expression of multiple, foreign proteins within a single cell. We have developed and utilised a series of recombinant baculovirus systems to unravel the sequential assembly process of a complex non-enveloped model virus, bluetongue virus (BTV). The high protein yields expressed by the baculovirus system not only facilitated structure-function analysis of each viral protein but were also advantageous to crystallography studies and supported the first atomic-level resolution of a recombinant viral protein, the major BTV capsid protein. Further, the formation of recombinant double-shelled virus-like particles (VLPs) provided insights into the structure-function relationships among the four major structural proteins of the BTV whilst also representing a potential candidate for a viral vaccine. The baculovirus multi-gene expression system facilitated the study of structurally complex viruses (both non-enveloped and enveloped viruses) and heralded a new generation of viral vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    马流感(EI)是一种传染性病毒性呼吸道疾病的equidae家族。两种病毒,H7N7和H3N8引起EI;然而,H7N7已经有几十年没有被发现了。H3N8已经传播并分为欧亚和美国血统。后者随后多元化进入肯塔基州,南美洲,和佛罗里达亚血统。同时,佛罗里达进化枝1(FC1)和佛罗里达进化枝2(FC2)菌株是唯一的流行EI病毒(EIV)。免疫被认为是预防和控制EI感染的主要手段。使用不同的技术和平台,已经开发并商业化了几种疫苗。根据世界动物卫生组织(WOAH)的建议,所有商业疫苗应包含FC1和FC2菌株的代表。不幸的是,大多数市售疫苗未更新为纳入FC2菌株的代表.
    这项研究的目的是开发一种新的EI疫苗候选物,其中掺入了来自当前循环的FC2的血凝素(HA)抗原。
    在这项研究中,我们报道了FC2的全长重组HA基因在杆状病毒表达系统中的表达。
    HA重组蛋白已通过血液吸附(HAD)和血凝试验被证明可以保持其生物学特性。此外,使用参考特异性血清,HA的特异性已通过免疫过氧化物酶和Western免疫印迹测定得到证实。
    总而言之,我们报道了FC2的特定生物活性重组HA的表达,这将成为产生更新的EI亚基或病毒载体疫苗候选物的基础。
    UNASSIGNED: Equine influenza (EI) is a transmissible viral respiratory sickness of the Equidae family. Two viruses, H7N7 and H3N8 caused EI; however, H7N7 has not been detected for decades. H3N8 has circulated and bifurcated into Eurasian and American lineages. The latter subsequently diversified into Kentucky, South America, and Florida sub-lineages. Florida clade 1 (FC1) and Florida clade 2 (FC2) strains are the only circulating EI viruses (EIVs) in the meantime. Immunization is considered the major means for the prevention and control of EI infection. Using disparate technologies and platforms, several vaccines have been developed and commercialized. According to the recommendations of the World Organization for Animal Health (WOAH), all commercial vaccines shall comprise representatives of both FC1 and FC2 strains. Unfortunately, most of the commercially available vaccines were not updated to incorporate a representative of FC2 strains.
    UNASSIGNED: The purpose of this research was to develop a new EI vaccine candidate that incorporates the hemagglutinin (HA) antigen from the currently circulating FC2.
    UNASSIGNED: In this study, we report the expression of the full-length recombinant HA gene of FC2 in the baculovirus expression system.
    UNASSIGNED: The HA recombinant protein has been proven to maintain its biological characteristics by hemadsorption (HAD) and hemagglutination tests. Moreover, using a reference-specific serum, the specificity of the HA has been confirmed through the implementation of immunoperoxidase and western immunoblotting assays.
    UNASSIGNED: In conclusion, we report the expression of specific biologically active recombinant HA of FC2, which would act as a foundation for the generation of an updated EI subunit or virus vector vaccine candidates.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    COVID-19在世界各地猖獗,给人类生活造成了极大的损害,严重阻碍了全球经济的发展。针对SARS-CoV-2的治疗,我们提出了一种基于铁蛋白(Fe)自组装技术的新型非诺抗体策略。
    与末端修饰的人铁蛋白的C末端融合的SARS-CoV-2的中和纳米抗体H11-D4在大肠杆菌和家蚕杆状病毒表达系统中表达。大量的纳米粒子在蚕体内成功自组装,虽然相对较少的纳米颗粒可以在处理过的产品从大肠杆菌通过电子显微镜观察。随后,然后评估非诺体的表达水平和中和活性。
    结果表明,在大肠杆菌中表达的H11-D4和非诺体Fe-H11-D4的IC50分别为171.1nmolL-1和20.87nmolL-1。然而,家蚕中表达的Fe-HD11-D4的IC50为1.46nmolL-1,显示出更好的中和活性。
    因此,在家蚕杆状病毒表达系统中,铁蛋白自组装技术可以有效提高纳米抗体的中和活性。
    UNASSIGNED: COVID-19 is rampant throughout the world, which has caused great damage to human lives and seriously hindered the development of the global economy. Aiming at the treatment of SARS-CoV-2, in this study, we proposed a novel fenobody strategy based on ferritin (Fe) self-assembly technology.
    UNASSIGNED: The neutralizing nanobody H11-D4 of SARS-CoV-2 fused to the C-terminus of end-modified human ferritin was expressed in E. coli and silkworm baculovirus expression systems. A large number of nanoparticles were successfully self-assembled in silkworms, while relatively few nanoparticles can be observed in the treated products from E. coli by electron microscopy. Subsequently, the fenobody\'s expression level and neutralizing activity were then evaluated.
    UNASSIGNED: The results showed that the IC50 of H11-D4 and fenobody Fe-H11-D4 expressed in E. coli were 171.1 nmol L-1 and 20.87 nmol L-1, respectively. However, the IC50 of Fe-HD11-D4 expressed in silkworms was 1.46 nmol L-1 showing better neutralization activity.
    UNASSIGNED: Therefore, fenobodies can be well self-assembled in silkworm baculovirus expression system, and ferritin self-assembly technology can effectively improve nanobody neutralization activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    H7N9禽流感病毒(AIV)给家禽业造成巨大损失,影响人类公共卫生安全,仍然构成潜在威胁。目前,禽流感的免疫预防和控制依赖于传统的灭活疫苗;然而,它们有一些局限性,基因工程禽流感亚单位疫苗可能是潜在的候选疫苗。在这项研究中,使用杆状病毒表达系统(BVES)在源自H7N9AIV的HA蛋白中产生T169A突变A/鸡/广东/16876(H7N9-16876)。结果表明,与野生型HA蛋白(HA-WT)相比,突变体(HAm)的热稳定性显着提高。重要的是,用HAm结合ISA71VG免疫鸡引起更高的交叉反应性血凝抑制(HI)抗体反应和细胞因子(IFN-γ和IL-4)分泌。在用异源H7N9AIV进行致命攻击后,该疫苗赋予鸡100%(10/10)的临床保护作用,有效抑制病毒脱落,90%(9/10)的鸡没有病毒脱落。HAm的热稳定性可以代表在实际疫苗生产和应用中的优势。总的来说,本研究中产生的HAm代表了预防和控制H7N9禽流感的有希望的亚单位疫苗候选物。
    H7N9 avian influenza virus (AIV) has caused huge losses in the poultry industry and impacted human public health security, and still poses a potential threat. Currently, immune prevention and control of avian influenza relies on traditional inactivated vaccines; however, they have some limitations and genetically engineered avian influenza subunit vaccines may be potential candidate vaccines. In this study, a T169A mutation in the HA protein derived from H7N9 AIV A/Chicken/Guangdong/16876 (H7N9-16876) was generated using the baculovirus expression system (BVES). The results showed that the mutant (HAm) had significantly increased thermostability compared with the wild-type HA protein (HA-WT). Importantly, immunizing chickens with HAm combined with ISA 71VG elicited higher cross-reactive hemagglutination inhibition (HI) antibody responses and cytokine (IFN-γ and IL-4) secretion. After a lethal challenge with heterologous H7N9 AIV, the vaccine conferred chickens with 100% (10/10) clinical protection and effectively inhibited viral shedding, with 90% (9/10) of the chickens showing no virus shedding. The thermostability of HAm may represent an advantage in practical vaccine manufacture and application. In general, the HAm generated in this study represents a promising subunit vaccine candidate for the prevention and control of H7N9 avian influenza.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    克里米亚-刚果出血热(CCHF)是由克里米亚-刚果出血热病毒(CCHF)引起的人畜共患疾病,这可能会导致严重的临床疾病,甚至导致人类死亡。近年来,这种疾病已经蔓延到更广泛的地区,对中国和中东构成重大公共卫生威胁,欧洲和非洲,没有安全有效的疫苗来预防这种疾病。最近,研究表明,使用Zera融合靶向蛋白可以增强免疫原性并提高开发病毒疫苗的潜力。基于这一发现,在这项研究中,两种候选疫苗,Zera-Gn和Zera-Np,使用昆虫杆状病毒系统制备,该系统表达与Zera标签融合的CCHFV糖蛋白(Gn)和核衣壳蛋白(Np),并评价其在BALB/c小鼠中的免疫原性。结果表明,Zera-Gn和Zera-Np重组纳米粒都成功表达,Zera-Gn对小鼠体液免疫和细胞免疫具有良好的诱导作用,其免疫原性显著高于Zera-Np。结果表明,通过将Zera标签与CCHFV刺入蛋白Gn融合制备的Zera-Gn自组装纳米颗粒有可能成为CCHF的候选疫苗,本研究为CCHFZera自组装纳米粒子疫苗的研制提供了参考。
    Crimean-Congo hemorrhagic fever (CCHF) is a zoonotic disease caused by Crimean-Congo hemorrhagic fever virus (CCHFV), which can cause severe clinical disease and even death in humans. In recent years, the disease has spread to a wider area, posing a major public health threat to China as well as the Middle East, Europe and Africa, and there is no safe and effective vaccine to prevent the disease. Recently, it has been shown that using the Zera fusion to target proteins can enhance immunogenicity and improve the potential for developing viral vaccines. Based on this finding, in this study, two vaccine candidates, Zera-Gn and Zera-Np, were prepared using an insect baculovirus system expressing CCHFV glycoprotein (Gn) and nucleocapsid protein (Np) fused with Zera tags, and evaluated for immunogenicity in BALB/c mice. The obtainedresults showed that both Zera-Gn and Zera-Np recombinant nanoparticles were successfully expressed, and Zera-Gn had good induction of humoral and cellular immunity in mice, and its immunogenicity was significantly higher than that of Zera-Np. The results indicated that Zera-Gn self-assembled nanoparticles prepared by fusing Zera tags with CCHFV spike-in protein Gn have the potential to be a candidate vaccine for CCHF, and this study provides a reference for the development of Zera self-assembled nanoparticle vaccine for CCHF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号