Adenoviral vector

腺病毒载体
  • 文章类型: Journal Article
    共价闭合环状DNA(cccDNA)作为肝细胞核中稳定的游离型微小染色体存在,并负责乙型肝炎病毒(HBV)的持久性。我们最近报道了一种通过位点特异性DNA重组涉及HBV重组cccDNA(rcccDNA)的技术。将漂浮的单体HBV基因组工程化到前体质粒(prcccDNA)中,该前体质粒通过Cre/loxP介导的DNA重组切除,形成带有loxP嵌合内含子的3.3kbrcccDNA。外源序列在RNA剪接过程中被有效去除,呈现功能无缝插入。我们表征了rcccDNA的形成,有效的病毒转录,和rcccDNA在体外和体内诱导的复制。此外,我们通过使用复制缺陷型重组腺病毒载体将rcccDNA传递给表达Cre重组酶的转基因小鼠来密切模拟慢性肝炎,这导致了突出的HBV持久性。这里,我们描述了一个详细的协议,如何构建和评估Cre/loxP为基础的重组HBVcccDNA系统在体外和体内。
    Covalently closed circular DNA (cccDNA) exists as a stable episomal minichromosome in the nucleus of hepatocytes and is responsible for hepatitis B virus (HBV) persistence. We recently reported a technique involving recombinant cccDNA (rcccDNA) of HBV by site-specific DNA recombination. A floxed monomeric HBV genome was engineered into a precursor plasmid (prcccDNA) which was excised via Cre/loxP-mediated DNA recombination to form a 3.3-kb rcccDNA bearing a loxP-chimeric intron. The foreign sequence was efficiently removed during RNA splicing, rendering a functionally seamless insertion. We characterized rcccDNA formation, effective viral transcription, and replication induced by rcccDNA both in vitro and in vivo. Furthermore, we closely simulated chronic hepatitis by using a replication-defective recombinant adenoviral vector to deliver rcccDNA to the transgenic mice expressing Cre recombinase, which led to prominent HBV persistence. Here, we describe a detailed protocol about how to construct and evaluate Cre/loxP-based recombinant HBV cccDNA system both in vitro and in vivo.
    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
    重链抗体(VHH)的可变结构域具有用于重定向腺病毒载体的细胞嗜性的潜力。这里,我们试图建立平台,以简化VHHs在整合到腺病毒纤维中时的特异性靶向功能的筛选.家禽腺病毒4(FAdV-4)和猿猴腺病毒1(SAdV-1)都有两种类型的纤维,其中之一对于病毒传播是不必要的,并且是VHH显示的适当位点。中间质粒,pMD-FAV4Fs,被构建为FAdV-4纤维2修饰的起始质粒。来自噬菌体T4的Foldon,三聚化的触发器,用于桥接纤维2和VHH的尾/轴域与人CD16A,自然杀伤(NK)细胞的关键膜标记。通过限制组装的一个步骤,修饰的纤维2被转移到腺病毒质粒,将其线性化并转染到包装细胞中。5种携带GFP基因的FAdV-4病毒最终被拯救并扩增,显示三个VHH。一种重组病毒,FAdV4FC21-EG,几乎不能转导人293或Jurkat细胞。相比之下,当它以每个细胞1000个病毒颗粒的感染复数使用时,表达外源CD16A的293或Jurkat细胞的转导效率达到51%或34%。将这种纤维修饰策略移植到SAdV-1载体上构建SAdV1FC28H-EG,适度转导原代人NK细胞,而亲本病毒没有转导。总的来说,我们改革了将VHH整合到纤维的策略,并建立了筛选VHH的新平台,以构建具有特定向性的腺病毒载体.
    The variable domain of a heavy-chain antibody (VHH) has the potential to be used to redirect the cell tropism of adenoviral vectors. Here, we attempted to establish platforms to simplify the screening of VHHs for their specific targeting function when being incorporated into the fiber of adenovirus. Both fowl adenovirus 4 (FAdV-4) and simian adenovirus 1 (SAdV-1) have two types of fiber, one of which is dispensable for virus propagation and is a proper site for VHH display. An intermediate plasmid, pMD-FAV4Fs, was constructed as the start plasmid for FAdV-4 fiber2 modification. Foldon from phage T4 fibritin, a trigger for trimerization, was employed to bridge the tail/shaft domain of fiber2 and VHHs against human CD16A, a key membrane marker of natural killer (NK) cells. Through one step of restriction-assembly, the modified fiber2 was transferred to the adenoviral plasmid, which was linearized and transfected to packaging cells. Five FAdV-4 viruses carrying the GFP gene were finally rescued and amplified, with three VHHs being displayed. One recombinant virus, FAdV4FC21-EG, could hardly transduce human 293 or Jurkat cells. In contrast, when it was used at a multiplicity of infection of 1000 viral particles per cell, the transduction efficiency reached 51% or 34% for 293 or Jurkat cells expressing exogenous CD16A. Such a strategy of fiber modification was transplanted to the SAdV-1 vector to construct SAdV1FC28H-EG, which moderately transduced primary human NK cells while the parental virus transduced none. Collectively, we reformed the strategy of integrating VHH to fiber and established novel platforms for screening VHHs to construct adenoviral vectors with a specific tropism.
    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
    目的:猪流行性腹泻(PED)是一种高度传染性和经济意义重大的胃肠道疾病,影响所有年龄的猪。预防和控制PED是通过用疫苗免疫母猪来实现的,通过初乳进行被动仔猪免疫。尽管使用了商业疫苗,G2b猪流行性腹泻病毒(PEDV)在中国的流行仍在继续,提出了有关当前疫苗功效和新型疫苗开发需求的问题。腺病毒血清型5(Ad5)有几个优点,包括高转导效率,广泛的宿主细胞,以及在不同阶段感染细胞的能力。在这项研究中,我们使用Ad5载体表达了spike(S)的免疫原性蛋白,并通过诱导显着的体液免疫产生了PED疫苗候选物。rAd5-PEDV-S可以预防PED引起的体重减轻,腹泻,和仔猪肠道损伤。这种新型疫苗候选株具有用于猪养殖业的潜力。
    OBJECTIVE: Porcine epidemic diarrhea (PED) is a highly infectious and economically significant gastrointestinal disorder that affects pigs of all ages. Preventing and controlling PED is achieved by immunizing sows with vaccines, enabling passive piglet immunization via colostrum. The prevalence of G2b porcine epidemic diarrhea virus (PEDV) continues in China despite the use of commercial vaccines, raising questions regarding current vaccine efficacy and the need for novel vaccine development. Adenovirus serotype 5 (Ad5) has several advantages, including high transduction efficiency, a wide range of host cells, and the ability to infect cells at various stages. In this study, we expressed the immunogenic proteins of spike (S) using an Ad5 vector and generated a PED vaccine candidate by inducing significant humoral immunity. The rAd5-PEDV-S prevented PED-induced weight loss, diarrhea, and intestinal damage in piglets. This novel vaccine candidate strain possesses the potential for use in the pig breeding industry.
    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
    摘要经典嵌合血凝素(cHA)被设计用于诱导针对HA的保守茎结构域的免疫应答。然而,尚不清楚将一个以上的HA头部结构域结合到一个茎结构域上是否具有免疫原性并进一步诱导针对流感病毒的免疫应答.这里,我们构建了许多新的cHA,它们包含来自不同亚型的两个或三个融合的头部结构域,嫁接到一个茎结构域上,命名为cH1-H3、cH1-H7、cH1-H3-H7和cH1-H7-H3。使用生物信息学模拟对这些新型cHA的三维结构进行建模。结构分析显示,完整的中和表位暴露于cH1-H7中,并预测为免疫原性的。使用黑猩猩腺病毒载体(AdC68)疫苗平台在小鼠中评估cHA构建体的免疫原性。结果表明,由AdC68表达的cH1-H7(AdC68-cH1-H7)诱导产生高水平的结合抗体,中和抗体,和血凝素抑制抗体,针对同源大流行H1N1,漂移季节性H1N1和H7N9病毒。此外,完全保护接种的小鼠免受上述流感病毒的致命攻击。因此,具有强大免疫原性的cH1-H7cHA可能是一种潜在的新型疫苗,可提供针对不同亚型流感病毒的保护作用。
    Classic chimeric hemagglutinin (cHA) was designed to induce immune responses against the conserved stalk domain of HA. However, it is unclear whether combining more than one HA head domain onto one stalk domain is immunogenic and further induce immune responses against influenza viruses. Here, we constructed numerous novel cHAs comprising two or three fuzed head domains from different subtypes grafted onto one stalk domain, designated as cH1-H3, cH1-H7, cH1-H3-H7, and cH1-H7-H3. The three-dimensional structures of these novel cHAs were modelled using bioinformatics simulations. Structural analysis showed that the intact neutralizing epitopes were exposed in cH1-H7 and were predicted to be immunogenic. The immunogenicity of the cHAs constructs was evaluated in mice using a chimpanzee adenoviral vector (AdC68) vaccine platform. The results demonstrated that cH1-H7 expressed by AdC68 (AdC68-cH1-H7) induced the production of high levels of binding antibodies, neutralizing antibodies, and hemagglutinin inhibition antibodies against homologous pandemic H1N1, drifted seasonal H1N1, and H7N9 virus. Moreover, vaccinated mice were fully protected from a lethal challenge with the aforementioned influenza viruses. Hence, cH1-H7 cHAs with potent immunogenicity might be a potential novel vaccine to provide protection against different subtypes of influenza virus.
    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疫苗未能预防SARS-CoV-2感染和传播,一个可能的关键原因是呼吸道缺乏保护性粘膜免疫。这里,我们通过鼻内(IN)滴注和肌内(IM)注射疫苗,对比Ad5-nCoV-S,评估了新型猿猴腺病毒载体COVID-19疫苗(Sad23L-nCoV-S)对小鼠黏膜和全身免疫的影响.和众所周知的Ad5-nCoV-S疫苗一样好,单剂量接种1×109PFUSad23L-nCoV-S疫苗可诱导血清中类似水平的IgGS结合抗体(S-BAb)和中和抗体(NAb)和更高的IgA,而IN途径在支气管肺泡灌洗(BAL)中显著升高IgG和IgAS-BAb和NAb,与IM途径相比,肺部特异性IFN-γ分泌T细胞反应,但脾脏中的T细胞反应较低。通过使用Sad23L-nCoV-S疫苗的IN和IM接种的不同组合的初免-加强疫苗接种方案,参与免疫的疫苗在BAL和肺中刺激了更高的保护性粘膜或局部免疫,而IM参与的免疫在血清和脾脏中诱导了更高的全身免疫。通过IN和IM途径的初免-加强疫苗接种方案,对SARS-CoV-2的长期持续的粘膜和全身性NAb和T细胞免疫在32周内维持在高水平。总之,用Sad23L-nCoV-S疫苗的IN接种引发或加强免疫可以诱导有效的粘膜免疫,并且IM途径的组合还可以实现全身免疫,为呼吸道病毒感染的疫苗接种方案提供了重要参考。疫苗接种的基本目标是产生有效和长期的疾病保护。几个因素,包括疫苗载体,交货路线,加强方案会影响初次加强免疫方法的结果。通过构建新型猿猴腺病毒载体的COVID-19疫苗,并采用鼻内和肌内接种的组合,可以在呼吸道中引发针对5种突变株的粘膜中和抗体,并具有较强的全身免疫力。免疫保护可以持续32周以上。疫苗的构建和免疫方案对呼吸道疾病的预防产生了积极的影响。
    OBJECTIVE: The essential goal of vaccination is to generate potent and long-term protection against diseases. Several factors including vaccine vector, delivery route, and boosting regimen influence the outcome of prime-boost immunization approaches. The immunization regimens by constructing a novel simian adenovirus-vectored COVID-19 vaccine and employing combination of intranasal and intramuscular inoculations could elicit mucosal neutralizing antibodies against five mutant strains in the respiratory tract and strong systemic immunity. Immune protection could last for more than 32 weeks. Vectored vaccine construction and immunization regimens have positively impacted respiratory disease prevention.
    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
    根据目前的情况,开发一种高效安全的2019冠状病毒病(COVID-19)疫苗是管理严重急性呼吸道疾病冠状病毒2(SARS-CoV-2)大流行的关键方法。在这项研究中,我们产生了优化的SARS-CoV-2刺突基因的缩短片段(2043bp,称为S1),能够编码截短的S1蛋白。测试该蛋白质以确定它是否可以在小鼠中引发针对SARS-CoV-2的有效免疫。用免疫荧光和蛋白质印迹证实S1蛋白的存在。将带有S1基因片段的腺病毒疫苗(Ad-S1)在4周内肌内给予小鼠4次。在所有免疫小鼠中均证明了SARS-CoV-2S1蛋白的体液免疫。免疫小鼠的血清在体外表现出优异的抗感染活性。在接种Ad-S1后,在小鼠中观察到针对SARS-CoV-2的强大体液免疫反应,这表明腺病毒疫苗可能有助于开发针对SARS-CoV-2和其他遗传上不同的病毒的疫苗。
    The development of an efficient and safe coronavirus disease 2019 (COVID-19) vaccine is a crucial approach for managing the severe acute respiratory disease coronavirus 2 (SARS-CoV-2) pandemic in light of current conditions. In this study, we produced a shortened segment of the optimized SARS-CoV-2 spike gene (2043 bp, termed S1) that was able to encode a truncated S1 protein. The protein was tested to determine if it could elicit efficient immunization in mice against SARS-CoV-2. The presence of the S1 protein was confirmed with immunofluorescence and Western blotting. An adenovirus vaccine bearing the S1 gene fragment (Ad-S1) was administered intramuscularly to mice four times over 4 weeks. SARS-CoV-2 S1 protein humoral immunity was demonstrated in all immunized mice. The serum from immunized mice demonstrated excellent anti-infection activity in vitro. A robust humoral immune response against SARS-CoV-2 was observed in the mice after vaccination with Ad-S1, suggesting that the adenovirus vaccine may aid the development of vaccines against SARS-CoV-2 and other genetically distinct 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
    基于抗体的免疫检查点阻断在患者中显示出巨大的临床成功,但是暂时的工作方式导致有限的治疗益处。在这里,在Hepa1-6肝癌细胞中,我们开发了一种基因工程腺病毒作为载体,用于递送CRISPR/Cas9(sgCas9-AdV),以实现永久性PD-L1基因编辑,效率高达78.7%.此外,将sgCas9-AdV加载到由丝纤维(SgCas9-AdV/Gel)制成的水凝胶中用于体内应用。丝胶不仅促进sgCas9-AdV在肿瘤组织中的局部保留,但也掩盖了它们对宿主免疫系统的影响,从而确保有效的基因转导超过9天。承担这些优点,sgCas9-AdV/Gel通过单剂量注射以100%应答率抑制Hepa1-6肿瘤生长,通过有效的PD-L1破坏引发T细胞介导的抗肿瘤反应。此外,sgCas9-AdV/Gel也成功地延伸到其他难治性肿瘤中。在以对抗PD-L1的不良反应为特征的CT26结肠肿瘤中,sgCas9-AdV/Gel被证明是抑制肿瘤进展的有能力和优异的抗PD-L1抗体。在高度侵袭性的原位4T1小鼠乳腺肿瘤中,这样的治疗范例显著抑制原发性肿瘤生长并诱导针对肿瘤复发/转移的持久免疫应答。因此,这项研究为免疫治疗提供了一种有吸引力的通用策略.
    Immune checkpoint blockade based on antibodies has shown great clinical success in patients, but the transitory working manner leads to restricted therapeutic benefits. Herein, a genetically engineered adenovirus is developed as the vector to deliver CRISPR/Cas9 (sgCas9-AdV) to achieve permanent PD-L1 gene editing with efficiency up to 78.7% exemplified in Hepa 1-6 liver cancer cells. Furthermore, the sgCas9-AdV is loaded into hydrogel made by silk fiber (SgCas9-AdV/Gel) for in vivo application. The silk-gel not only promotes local retention of sgCas9-AdV in tumor tissue, but also masks them from host immune system, thus ensuring effectively gene transduction over 9 days. Bearing these advantages, the sgCas9-AdV/Gel inhibits Hepa 1-6 tumor growth with 100% response rate by single-dose injection, through efficient PD-L1 disruption to elicit a T cell-mediated antitumor response. In addition, the sgCas9-AdV/Gel is also successfully extended into other refractory tumors. In CT26 colon tumor characterized by poor response to anti-PD-L1, sgCas9-AdV/Gel is demonstrated to competent and superior anti-PD-L1 antibody to suppress tumor progression. In highly aggressive orthotopic 4T1 mouse breast tumor, such a therapeutic paradigm significantly inhibits primary tumor growth and induces a durable immune response against tumor relapse/metastasis. Thus, this study provides an attractive and universal strategy for immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    新出现的SARS-CoV-2变体(VOCs)威胁到当前肌内注射的COVID-19疫苗的有效性,这些疫苗仅针对刺突蛋白。迫切需要开发下一代疫苗策略,以实现更广泛和持久的保护。使用人和黑猩猩来源的腺病毒载体(Ad),我们评估了表达spike-1,核衣壳,和小鼠模型中的RdRp抗原。我们显示单剂量鼻内免疫,特别是黑猩猩Ad-vectored疫苗,在诱导由局部和全身抗体反应组成的三方保护性免疫方面优于肌内免疫,粘膜组织驻留记忆T细胞和粘膜训练的先天免疫。我们进一步表明,鼻内免疫提供了对祖先SARS-CoV-2和两种VOC的保护,B.1.1.7和B.1.351.我们的发现表明,Ad-vectored多价疫苗的呼吸道粘膜递送代表了一种有效的下一代COVID-19疫苗策略,可诱导针对当前和未来VOC的全方位粘膜免疫。
    The emerging SARS-CoV-2 variants of concern (VOCs) threaten the effectiveness of current COVID-19 vaccines administered intramuscularly and designed to only target the spike protein. There is a pressing need to develop next-generation vaccine strategies for broader and long-lasting protection. Using adenoviral vectors (Ad) of human and chimpanzee origin, we evaluated Ad-vectored trivalent COVID-19 vaccines expressing spike-1, nucleocapsid, and RdRp antigens in murine models. We show that single-dose intranasal immunization, particularly with chimpanzee Ad-vectored vaccine, is superior to intramuscular immunization in induction of the tripartite protective immunity consisting of local and systemic antibody responses, mucosal tissue-resident memory T cells and mucosal trained innate immunity. We further show that intranasal immunization provides protection against both the ancestral SARS-CoV-2 and two VOC, B.1.1.7 and B.1.351. Our findings indicate that respiratory mucosal delivery of Ad-vectored multivalent vaccine represents an effective next-generation COVID-19 vaccine strategy to induce all-around mucosal immunity against current and future VOC.
    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
    目的:缺氧缺血性脑损伤(HIBD)是新生儿脑损伤的主要原因。骨髓间充质干细胞(BMSCs)显示出治疗HIBD的潜力,和基因修饰可以增强它们的神经保护作用。这项研究的目的是研究肝细胞生长因子(HGF)过表达的BMSCs(BMSCs-HGF)对HIBD的神经保护作用及其潜在机制。
    方法:使用腺病毒载体用HGF转染BMSCs。建立HIBD模型,然后通过脑室内注射将BMSCs移植到HIBD大鼠的脑内。2,3,5-氯化三苯基四唑(TTC)染色用于测量脑梗死体积。体外,将原代培养的皮质神经元与BMSCs在Transwell板系统中共培养。氧糖剥夺(OGD)用于模拟缺氧缺血性损伤,和PD98059加入到培养基中以阻断细胞外信号调节激酶(ERK)的磷酸化。使用TUNEL染色确定细胞凋亡。免疫荧光法检测HGF的表达,实时定量PCR(RT-qPCR),和西方印迹。通过蛋白质印迹测量磷酸化ERK(p-ERK)和B细胞淋巴瘤2(Bcl-2)的表达。
    结果:HGF基因转染促进BMSC增殖。此外,BMSCs-HGF减少了HIBD诱导的脑梗死体积,增强了BMSCs对HIBD的保护作用。BMSCs-HGF也增加了HGF的表达,p-ERK,和脑组织中的Bcl-2。体外,BMSC-HGF保护神经元免受OGD诱导的细胞凋亡。ERK磷酸化的抑制消除了BMSCs-HGF对OGD的神经保护作用。
    结论:BMSCs-HGF是治疗HIBD的潜在方法,ERK/Bcl-2通路参与潜在的神经保护机制。
    OBJECTIVE: Hypoxic-ischemic brain damage (HIBD) is a major cause of brain injury in neonates. Bone marrow mesenchymal stem cells (BMSCs) show therapeutic potential for HIBD, and genetic modification may enhance their neuroprotective effects. The goal of this study was to investigate the neuroprotective effects of hepatocyte growth factor (HGF)-overexpressing BMSCs (BMSCs-HGF) against HIBD and their underlying mechanisms.
    METHODS: BMSCs were transfected with HGF using adenoviral vectors. HIBD models were established and then BMSCs were transplanted into the brains of HIBD rats via intraventricular injection. 2,3,5-Triphenyltetrazolium chloride (TTC) staining was used to measure cerebral infarction volumes. In vitro, primary cultured cortical neurons were co-cultured with BMSCs in a Transwell plate system. Oxygen-glucose deprivation (OGD) was applied to imitate hypoxic-ischemic insult, and PD98059 was added to the culture medium to block the phosphorylation of extracellular signal-regulated kinase (ERK). Cell apoptosis was determined using TUNEL staining. The expression of HGF was measured by immunofluorescence, real-time quantitative PCR (RT-qPCR), and western blots. The expression of phosphorylated ERK (p-ERK) and B-cell lymphoma-2 (Bcl-2) was measured by western blots.
    RESULTS: HGF-gene transfection promoted BMSC proliferation. Moreover, BMSCs-HGF decreased HIBD-induced cerebral infarction volumes and enhanced the protective effects of the BMSCs against HIBD. BMSCs-HGF also increased expression of HGF, p-ERK, and Bcl-2 in brain tissues. In vitro, BMSC-HGF protected neurons against OGD-induced apoptosis. Inhibition of ERK phosphorylation abolished the neuroprotective effect of BMSCs-HGF against OGD.
    CONCLUSIONS: BMSCs-HGF is a potential treatment for HIBD and that the ERK/Bcl-2 pathway is involved in the underlying neuroprotective mechanism.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号