influenza A virus

甲型流感病毒
  • 文章类型: Journal Article
    最近,甲型流感病毒的高通量测序已成为常规检测。应当注意,甲型流感病毒的极高多样性使确定所有八个基因组区段的序列的任务复杂化。为了快速准确的分析,有必要为每个部分选择最合适的参考。同时,在解码测序结果的领域中没有标准化的方法允许用户更新通过病毒测序获得的读段与之比较的序列数据库。IAVCP(甲型流感病毒共识和系统发育)的开发目的是自动分析甲型流感病毒的高通量测序数据。其目标包括直接从配对的原始读段中提取共有基因组。此外,通过分析自动重建的系统发育树的拓扑结构,该管道能够识别感兴趣病毒进化史中的潜在重配事件。
    Recently, high-throughput sequencing of influenza A viruses has become a routine test. It should be noted that the extremely high diversity of the influenza A virus complicates the task of determining the sequences of all eight genome segments. For a fast and accurate analysis, it is necessary to select the most suitable reference for each segment. At the same time, there is no standardized method in the field of decoding sequencing results that allows the user to update the sequence databases to which the reads obtained by virus sequencing are compared. The IAVCP (influenza A virus consensus and phylogeny) was developed with the goal of automatically analyzing high-throughput sequencing data of influenza A viruses. Its goals include the extraction of a consensus genome directly from paired raw reads. In addition, the pipeline enables the identification of potential reassortment events in the evolutionary history of the virus of interest by analyzing the topological structure of phylogenetic trees that are automatically reconstructed.
    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
    属于H1亚型的甲型流感病毒(IAV-S)在全世界猪中流行。抗原漂移和抗原转移导致循环IAV-S菌株中大量的抗原多样性。因此,最常用的基于完整灭活病毒(WIV)的疫苗由于疫苗病毒株与循环病毒株不匹配,对不同H1株的保护作用较低.这里,在对从公共数据库获得的IAV-S分离株的序列进行比对后,在计算机模拟中产生H1亚型HA全长的共有编码序列,并使用Orf病毒(ORFV)载体平台将其递送至猪.在仔猪中针对不同的IAV-S毒株评估了所得ORFVΔ121conH1重组病毒的免疫原性和保护效力。通过实时RT-PCR和病毒滴定评估用两种IAV-S毒株鼻内/气管内攻击后的病毒脱落。免疫动物的鼻分泌物中的病毒基因组拷贝和感染性病毒载量减少。流式细胞仪分析表明,T辅助/记忆细胞的频率,以及细胞毒性T淋巴细胞(CTL),与未接种疫苗的动物相比,接种疫苗组的外周血单核细胞(PBMC)在接受IAVH1N1大流行菌株(CA/09)的攻击时明显更高。有趣的是,在接受来自γ进化枝的H1N1病毒(OH/07)攻击的组中,与未接种疫苗的动物相比,接种疫苗的动物的支气管肺泡灌洗液中T细胞的百分比更高.总之,副痘病毒ORFV载体从H1IAV-S亚型递送共有HA减少了感染性病毒的脱落和鼻分泌物中IAV-S的病毒载量,并诱导了猪针对不同流感病毒的细胞保护性免疫。
    Influenza A viruses (IAV-S) belonging to the H1 subtype are endemic in swine worldwide. Antigenic drift and antigenic shift lead to a substantial antigenic diversity in circulating IAV-S strains. As a result, the most commonly used vaccines based on whole inactivated viruses (WIVs) provide low protection against divergent H1 strains due to the mismatch between the vaccine virus strain and the circulating one. Here, a consensus coding sequence of the full-length of HA from H1 subtype was generated in silico after alignment of the sequences from IAV-S isolates obtained from public databases and was delivered to pigs using the Orf virus (ORFV) vector platform. The immunogenicity and protective efficacy of the resulting ORFVΔ121conH1 recombinant virus were evaluated against divergent IAV-S strains in piglets. Virus shedding after intranasal/intratracheal challenge with two IAV-S strains was assessed by real-time RT-PCR and virus titration. Viral genome copies and infectious virus load were reduced in nasal secretions of immunized animals. Flow cytometry analysis showed that the frequency of T helper/memory cells, as well as cytotoxic T lymphocytes (CTLs), were significantly higher in the peripheral blood mononuclear cells (PBMCs) of the vaccinated groups compared to unvaccinated animals when they were challenged with a pandemic strain of IAV H1N1 (CA/09). Interestingly, the percentage of T cells was higher in the bronchoalveolar lavage of vaccinated animals in relation to unvaccinated animals in the groups challenged with a H1N1 from the gamma clade (OH/07). In summary, delivery of the consensus HA from the H1 IAV-S subtype by the parapoxvirus ORFV vector decreased shedding of infectious virus and viral load of IAV-S in nasal secretions and induced cellular protective immunity against divergent influenza viruses in swine.
    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
    季节性流感,每年导致数百万人死亡,对人类健康构成严重威胁。目前可用的流感疫苗仅针对特定毒株或保守表位;然而,这些疫苗并不完全有效,因为流感病毒在循环过程中会发生突变,导致推荐菌株和循环菌株之间的抗原错配和免疫系统的逃避。因此,开发一种快速的流感疫苗,有效,广泛的保护已经变得至关重要,血凝素(HA)的组成部分仍然是疫苗开发的理想目标。这项研究开发了一种脂质纳米颗粒包裹的核苷修饰的mRNA疫苗(mRNA-LNP),编码共有全长HA序列(H1c),并通过体外和体内测定评估了其保护功效和免疫原性。在两次肌内免疫后(2µg,10µg,或20µg)在BALB/c小鼠中间隔3周,H1c-mRNA-LNP疫苗诱导强抗体,如血凝抑制试验所示,以及针对多种异源H1N1流感病毒的保护性中和抗体,如微中和试验所示。此外,Th1-和Th2-偏向的细胞免疫应答都被引发,Th1偏向的反应更强。两剂H1c-mRNA-LNP疫苗可以中和一组异源H1N1流感病毒,并可以在小鼠中提供保护。一起来看,这些研究结果表明,编码共有全长HA的H1c-mRNA-LNP疫苗是开发针对一组异源H1N1流感病毒交叉保护疫苗的可行策略.
    Seasonal influenza, causes hundreds of thousands of deaths annually, posing a severe threat to human health. Currently available influenza vaccines are targeted only at specific strains or conserved epitopes; however, these vaccines are not completely efficacious because influenza viruses can undergo mutation during circulation, leading to antigenic mismatch between recommended strains and circulating strains and elusion from the immune system. Therefore, developing an influenza vaccine that is quick, effective, and broadly protective has become crucial, and the integral part of hemagglutinin (HA) remains an ideal target for vaccine development. This study developed a lipid nanoparticle-encapsulated nucleoside-modified mRNA vaccine (mRNA-LNPs) encoding a consensus full-length HA sequence (H1c) and evaluated its protective efficacy and immunogenicity through in vitro and in vivo assays. Following two intramuscular immunizations (2, 10 µg, or 20 µg) at a 3-week interval in BALB/c mice, H1c-mRNA-LNP vaccine induced strong antibodies as shown in the hemagglutination-inhibition test and protective neutralizing antibodies against numerous heterologous H1N1 influenza viruses as shown in the microneutralization assay. Additionally, both Th1- and Th2-biased cellular immune responses were elicited, with the Th1-biased response being stronger. Two doses of the H1c-mRNA-LNP vaccine could neutralize a panel of heterologous H1N1 influenza viruses and could confer protection in mice. Taken together, these findings suggest that the H1c-mRNA-LNP vaccine encoding a consensus full-length HA is a feasible strategy for developing a cross-protective vaccine against a panel of heterologous H1N1 influenza viruses.
    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
    Continuous outbreaks of highly pathogenic avian influenza (HPAI) viruses in commercial poultry have caused devastating losses to domestic poultry with a raising public health concern. The outbreaks of HPAI viruses have increased worldwide, including the North America. Therefore, vaccination has been considered as an alternative strategy for an efficient control of HPAI viruses. In this study, we aimed to generate Newcastle disease virus (NDV) vectored H7 serotype-specific vaccines by expressing the consensus sequence of the HA protein. Conventional NDV strain LaSota vector and a chimeric NDV vector containing the avian paramyxovirus type-2 F and HN protein were able to express the consensus sequence of HA protein. The protective efficacy of vaccines was evaluated in broiler chickens and in turkeys. One-day-old poults were prime immunized with the chimeric vector expressing the HA protein followed by boost immunization with LaSota vector expressing the HA protein or co-expressing the HA and NA proteins. Our vaccine candidates provided complete protection of broiler chickens from mortality and shedding of H7N8 HPAI challenge virus. Turkeys were better protected by boosting with the LaSota vector co-expressing the HA and NA proteins than the LaSota vector expressing only the HA protein. Our study demonstrated a potential use of heterologous prime and boost vaccination strategy to protect poultry against H7 HPAI viruses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    H1N1, one of the most prevalent influenza A virus subtypes affecting the human population, can cause infections varying from mild respiratory syndrome to severe pneumonia. The current H1N1 vaccine needs to be updated annually and does not protect against future outbreaks. Here, we downloaded 2,656 HA protein sequences of human H1N1 viruses from the NCBI influenza database (up to the date of Aug. 2012) and constructed a phylogenetic tree of these H1 proteins via the neighbor-joining method using MEGA 5.0 software. A consensus H1 protein (CH1) was generated and was further modified with published conserved T-cell and B-cell epitopes. Interestingly, this CH1 protein is genetically similar to an H1 isolate obtained during the 1980s (A/Memphis/7/1980), indicating that a universal HA antigen may exist in nature. Vaccination with a DNA vaccine expressing CH1 elicited broadly reactive T-cell and B-cell responses to heterologous H1N1 viruses, though this vaccine did not successfully neutralize pdm09 H1N1 viruses. A combination of CH1 and pdm09 HA in a DNA vaccination neutralized pdm09 H1N1 viruses and protected mice from lethal infections by all representative H1N1 viruses. Moreover, a recombinant chimeric PR8-CH1 virus carrying HA sequence of the consensus H1 and all other seven genes from the PR8 strain was highly attenuated in mice, with a lethal dose (LD50) of more than 106 pfu. Vaccination with PR8-CH1 virus provided complete protection against infections by heterologous H1N1 strains. Taken together, a universal H1 antigen, CH1, was developed by constructing a consensus HA sequence, and the PR8-CH1 virus containing this consensus sequence elicited broadly protective immunity against heterologous H1N1 viruses.
    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
    H7N3和H7N7是高致病性禽流感(HPAI)病毒,不仅对家禽业而且对人类健康构成了巨大威胁。H7N9,一种低致病性禽流感(LPAI)病毒,对人类也有很高的致病性,人们非常担心这些H7亚型会获得在人类之间有效传播的能力,从而成为大流行威胁。涵盖所有三种亚型的候选疫苗必须,因此,是任何大流行防备计划的组成部分。为了满足这一需求,我们根据2012-2015年初NCBI提供的数据构建了H7N3,H7N7和H7N9的共有血凝素(HA)序列.然后将该人工序列优化用于蛋白质表达,然后转化为鼠伤寒沙门氏菌的减毒营养缺陷型突变体,JOL1863菌株。用JOL1863对鸡进行免疫,肌肉注射,经鼻或经口,引起有效的体液和细胞介导的免疫反应,独立于疫苗接种途径。我们的结果还表明,JOL1863向鸡单核细胞衍生的树突状细胞(MoDC)传递有效的成熟信号,其特征在于共刺激分子的上调和更高的细胞因子诱导。此外,用JOL1863在鸡中进行免疫接种赋予了针对异源LPAIH7N1病毒攻击的显着保护作用,如泄殖腔拭子中病毒脱落减少所示。我们得出结论,这种疫苗,基于共识HA,可以诱导更广泛的针对不同H7流感病毒的保护,因此值得进一步研究。
    H7N3 and H7N7 are highly pathogenic avian influenza (HPAI) viruses and have posed a great threat not only for the poultry industry but for the human health as well. H7N9, a low pathogenic avian influenza (LPAI) virus, is also highly pathogenic to humans, and there is a great concern that these H7 subtypes would acquire the ability to spread efficiently between humans, thereby becoming a pandemic threat. A vaccine candidate covering all the three subtypes must, therefore, be an integral part of any pandemic preparedness plan. To address this need, we constructed a consensus hemagglutinin (HA) sequence of H7N3, H7N7, and H7N9 based on the data available in the NCBI in early 2012-2015. This artificial sequence was then optimized for protein expression before being transformed into an attenuated auxotrophic mutant of Salmonella Typhimurium, JOL1863 strain. Immunizing chickens with JOL1863, delivered intramuscularly, nasally or orally, elicited efficient humoral and cell mediated immune responses, independently of the route of vaccination. Our results also showed that JOL1863 deliver efficient maturation signals to chicken monocyte derived dendritic cells (MoDCs) which were characterized by upregulation of costimulatory molecules and higher cytokine induction. Moreover, immunization with JOL1863 in chickens conferred a significant protection against the heterologous LPAI H7N1 virus challenge as indicated by reduced viral sheddings in the cloacal swabs. We conclude that this vaccine, based on a consensus HA, could induce broader spectrum of protection against divergent H7 influenza viruses and thus warrants further study.
    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
    甲型流感离子通道膜基质蛋白2(M2e)的胞外结构域被认为是开发通用甲型流感疫苗的潜在候选者。然而,M2e的低免疫原性存在显著的障碍。我们已经开发了包含与金纳米颗粒(AuNP)缀合的共有M2e肽和作为可溶性佐剂的CpG(AuNP-M2e+sCpG)的疫苗制剂。我们证明了AuNP-M2e+sCpG在小鼠中的鼻内递送诱导肺B细胞活化和稳健的血清抗M2e免疫球蛋白G(IgG)应答,同时刺激IgG1和IgG2a亚型。使用Madin-Darby犬肾(MDCK)细胞感染A/California/04/2009(H1N1pdm)大流行毒株,或A/Victoria/3/75(H3N2),或高致病性禽流感病毒A/Vietnam/1203/2004(H5N1)作为免疫吸附剂,我们进一步表明,产生的抗体也能够与感染细胞上表达的M2的同四聚体形式结合。用A/California/04/2009(H1N1N1pdm)大流行毒株接种疫苗的小鼠的致命攻击,A/Victoria/3/75(H3N2),高致病性禽流感病毒A/越南/1203/2004(H5N1)导致100%,92%,100%保护,分别。总的来说,这项研究有助于奠定潜在的通用甲型流感疫苗的基础。
    The extracellular domain of influenza A ion channel membrane matrix protein 2 (M2e) is considered to be a potential candidate to develop a universal influenza A vaccine. However poor immunogenicity of M2e presents a significant roadblock. We have developed a vaccine formulation comprising of the consensus M2e peptide conjugated to gold nanoparticles (AuNPs) with CpG as a soluble adjuvant (AuNP-M2e + sCpG). We demonstrate that intranasal delivery of AuNP-M2e + sCpG in mice induces lung B cell activation and robust serum anti-M2e immunoglobulin G (IgG) response, with stimulation of both IgG1 and IgG2a subtypes. Using Madin-Darby canine kidney (MDCK) cells infected with A/California/04/2009 (H1N1pdm) pandemic strain, or A/Victoria/3/75 (H3N2), or the highly pathogenic avian influenza virus A/Vietnam/1203/2004 (H5N1) as immunosorbants we further show that the antibodies generated are also capable of binding to the homotetrameric form of M2 expressed on infected cells. Lethal challenge of vaccinated mice with A/California/04/2009 (H1N1pdm) pandemic strain, A/Victoria/3/75 (H3N2), and the highly pathogenic avian influenza virus A/Vietnam/1203/2004 (H5N1) led to 100%, 92%, and 100% protection, respectively. Overall, this study helps to lay the foundation of a potential universal influenza A vaccine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    由流感病毒引起的季节性和新出现的流行病仍然是公共卫生问题和经济负担。血凝素上的保守表位诱导广泛的保护性免疫应答的弱免疫原性是开发通用疫苗的主要障碍。在本报告中,我们设计了跨亚型序贯疫苗接种策略,并通过基于假病毒的中和试验评估了其中和抗体(nAb)活性.结果清楚地表明,与传统疫苗策略相比,交叉亚型序贯免疫可以显著诱导小鼠的广泛的血清交叉反应性nAb反应以及对同源菌株,并提供保护免受异源病毒PR8(H1N1)攻击。此外,我们从依次免疫的小鼠中分离出两种单克隆抗体,对多种流感毒株具有有效的广泛中和活性。这些数据表明序贯免疫在通用流感疫苗开发中的可行性。
    Seasonal and emerging epidemics caused by influenza virus remain as a public health concern and an economic burden. The weak immunogenicity of conserved epitopes on hemagglutinin that induces broad protective immune responses is the main obstacle to the development of universal vaccines. In the present report, we designed the cross-subtypic sequential vaccination strategy and evaluated its neutralizing antibody (nAb) activity by pseudovirus-based neutralization assays. The results clearly indicated that compared with traditional vaccines strategy, the cross-subtypic sequential immunization could significantly induce a broad serum cross-reactive nAb response in mice as well as against homologous strains, and provide protection from heterologous virus PR8 (H1N1) challenge. Furthermore, we isolated two monoclonal antibodies from sequentially immunized mice, which had potent broadly neutralizing activity against multiple influenza strains. These data suggest the feasibility of sequential immunization in universal flu vaccine development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Comparative Study
    The assembly of influenza virus progeny virions requires machinery that exports viral genomic ribonucleoproteins from the cell nucleus. Currently, seven nuclear export signal (NES) consensus sequences have been identified in different viral proteins, including NS1, NS2, M1, and NP. The present study examined the roles of viral NES consensus sequences and their significance in terms of viral replication and nuclear export. Mutation of the NP-NES3 consensus sequence resulted in a failure to rescue viruses using a reverse genetics approach, whereas mutation of the NS2-NES1 and NS2-NES2 sequences led to a strong reduction in viral replication kinetics compared with the wild-type sequence. While the viral replication kinetics for other NES mutant viruses were also lower than those of the wild-type, the difference was not so marked. Immunofluorescence analysis after transient expression of NP-NES3, NS2-NES1, or NS2-NES2 proteins in host cells showed that they accumulated in the cell nucleus. These results suggest that the NP-NES3 consensus sequence is mostly required for viral replication. Therefore, each of the hydrophobic (Φ) residues within this NES consensus sequence (Φ1, Φ2, Φ3, or Φ4) was mutated, and its viral replication and nuclear export function were analyzed. No viruses harboring NP-NES3 Φ2 or Φ3 mutants could be rescued. Consistent with this, the NP-NES3 Φ2 and Φ3 mutants showed reduced binding affinity with CRM1 in a pull-down assay, and both accumulated in the cell nucleus. Indeed, a nuclear export assay revealed that these mutant proteins showed lower nuclear export activity than the wild-type protein. Moreover, the Φ2 and Φ3 residues (along with other Φ residues) within the NP-NES3 consensus were highly conserved among different influenza A viruses, including human, avian, and swine. Taken together, these results suggest that the Φ2 and Φ3 residues within the NP-NES3 protein are important for its nuclear export function during viral replication.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    To develop a safe and effective mucosal vaccine against pathogenic influenza viruses, we constructed recombinant Lactobacillus casei strains that express conserved matrix protein 2 with (pgsA-CTA1-sM2/L. casei) or without (pgsA-sM2/L. casei) cholera toxin subunit A1 (CTA1) on the surface. The surface localization of the fusion protein was verified by cellular fractionation analyses, flow cytometry and immunofluorescence microscopy. Oral and nasal inoculations of recombinant L. casei into mice resulted in high levels of serum immunoglobulin G (IgG) and mucosal IgA. However, the conjugation of cholera toxin subunit A1 induced more potent mucosal, humoral and cell-mediated immune responses. In a challenge test with 10 MLD50 of A/EM/Korea/W149/06(H5N1), A/Puerto Rico/8/34(H1N1), A/Aquatic bird /Korea/W81/2005(H5N2), A/Aquatic bird/Korea/W44/2005(H7N3), and A/Chicken/Korea/116/2004(H9N2) viruses, the recombinant pgsA-CTA1-sM2/L. casei provided better protection against lethal challenges than pgsA-sM2/L. casei, pgsA/L. casei and PBS in mice. These results indicate that mucosal immunization with recombinant L. casei expressing CTA1-conjugated sM2 protein on its surface is an effective means of eliciting protective immune responses against diverse influenza subtypes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号