Mucosal immunization

粘膜免疫
  • 文章类型: Journal Article
    自从第一次使用疫苗告诉SARS-CoV-2在世界范围内传播引起的最后一次COVID-19大流行以来,先进生物技术的使用加速了不同类型和免疫方法的发展。上一次大流行表明,基于核酸的疫苗,尤其是mRNA,在开发时间方面具有优势;然而,它显示了一个非常关键的缺点,即,与其他策略相比,成本更高,以及它无法抵御新的变种。这表明需要更多的改进以达到更好的递送和功效。在这篇综述中,我们将描述不同的疫苗输送系统,包括,最常用的病毒载体,以及用于递送基于核酸的疫苗,特别是基于脂质的纳米颗粒制剂的可变策略,聚合物囊泡,电穿孔以及传递mRNA的新的强大工具,这是基于细胞穿透肽(CPPs)的使用。此外,我们还将讨论与每个系统相关的主要挑战。芬利,疫苗的有效性和安全性不仅取决于配方和递送系统,而且剂量和给药途径也是重要的参与者,因此,我们将看到疫苗给药的不同途径,包括传统途径(肌内,经皮,皮下),口腔吸入或通过鼻粘膜,并将描述每种给药途径的优缺点。
    Since the first use of vaccine tell the last COVID-19 pandemic caused by spread of SARS-CoV-2 worldwide, the use of advanced biotechnological techniques has accelerated the development of different types and methods for immunization. The last pandemic showed that the nucleic acid-based vaccine, especially mRNA, has an advantage in terms of development time; however, it showed a very critical drawback namely, the higher costs when compared to other strategies, and its inability to protect against new variants. This showed the need of more improvement to reach a better delivery and efficacy. In this review we will describe different vaccine delivery systems including, the most used viral vector, and also variable strategies for delivering of nucleic acid-based vaccines especially lipid-based nanoparticles formulation, polymersomes, electroporation and also the new powerful tools for the delivery of mRNA, which is based on the use of cell-penetrating peptides (CPPs). Additionally, we will also discuss the main challenges associated with each system. Finlay, the efficacy and safety of the vaccines depends not only on the formulations and delivery systems, but also the dosage and route of administration are also important players, therefore we will see the different routes for the vaccine administration including traditionally routes (intramuscular, Transdermal, subcutaneous), oral inhalation or via nasal mucosa, and will describe the advantages and disadvantage of each administration route.
    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
    口腔粘膜由于其可接近性和引发局部和全身免疫应答的能力而成为免疫的有吸引力的部位。然而,由于口腔粘膜的物理屏障和免疫复杂性,评估口腔粘膜免疫原性已被证明具有挑战性。微针可以克服这些物理障碍,但是以前的工作仅限于微针递送部位的范围,几何图形,和释放动力学,所有这些都会影响生理反应。这里,我们开发了集成的纤维微针装置,一种具有可调几何形状和材料构型的口服剂型,能够在口腔粘膜中爆发和持续释放到受控深度。施用至颊粘膜或舌下粘膜的整合纤维微针导致脾细胞中的血清转化和抗原特异性干扰素-γ分泌。所产生的免疫应答的动力学和量级可以通过调节微针释放动力学来调节。最佳微针几何形状是位点特异性的,较长的微针在颊粘膜中引起更大的免疫原性,和较短的微针在舌下粘膜中引起更大的免疫原性。所产生的免疫应答的Th1/Th2表型也依赖于整合的纤维微针长度。一起,这些结果建立了集成纤维微针作为口腔黏膜的多功能递送系统,并激发了使用可调递送系统的进一步探索,以更好地了解口腔黏膜免疫。
    The oral mucosa is an attractive site for immunization due to its accessibility and ability to elicit local and systemic immune responses. However, evaluating oral mucosal immunogenicity has proven challenging due to the physical barriers and immunological complexity of the oral mucosa. Microneedles can overcome these physical barriers, but previous work has been limited in the scope of microneedle delivery site, geometry, and release kinetics, all of which are expected to affect physiological responses. Here, we develop integrated fiber microneedle devices, an oral dosage form with tunable geometries and material configurations capable of both burst and sustained release to controlled depths in the oral mucosa. Integrated fiber microneedles administered to either the buccal or sublingual mucosa result in seroconversion and antigen-specific interferon-γ secretion in splenocytes. The dynamics and magnitude of the resulting immune response can be modulated by tuning microneedle release kinetics. Optimal microneedle geometry is site-specific, with longer microneedles eliciting greater immunogenicity in the buccal mucosa, and shorter microneedles eliciting greater immunogenicity in the sublingual mucosa. The Th1/Th2 phenotype of the resulting immune response is also dependent on integrated fiber microneedle length. Together, these results establish integrated fiber microneedles as a multifunctional delivery system for the oral mucosa and motivate further exploration using tunable delivery systems to better understand oral mucosal immunity.
    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
    粘膜疫苗通过刺激IgA抗体的产生来防止呼吸道病毒感染,所述IgA抗体防止病毒侵入粘膜上皮。在这项研究中,构建了一种新型蛋白质亚单位粘膜疫苗,用于预防β冠状病毒SARS-CoV-2的感染.通过将编码SARS-CoV-2病毒S1血管紧张素转换酶受体结合域(ACE-2-RBD)的基因连接到编码霍乱毒素B亚基(CTB)的DNA片段的下游来组装疫苗,一种已知刺激疫苗免疫原性的粘膜佐剂。通过丙烯酰胺凝胶电泳和通过抗CTB和抗ACE-2-RBD初级抗体的免疫印迹,在转化的大肠杆菌BL-21细胞的匀浆中鉴定了42kDa疫苗融合蛋白。通过镍亲和柱色谱法从澄清的细菌匀浆中部分纯化嵌合CTB-SARS-CoV-2-ACE-2-RBD疫苗融合蛋白。通过聚丙烯酰胺凝胶电泳和42kDa嵌合疫苗蛋白的电洗脱来完成进一步的疫苗纯化。通过口服评估疫苗对SARS-CoV-2感染的保护作用,鼻部,并用CTB-SARS-CoV-2-ACE-2-RBD蛋白对BALB/c小鼠进行肠胃外免疫。通过ELISA分析在免疫小鼠血清中定量疫苗诱导的SARS-CoV-2特异性抗体。免疫小鼠的血清中含有IgG和IgA抗体,可中和VeroE6细胞培养物中的SARS-CoV-2感染。与未免疫的小鼠相反,从免疫小鼠切除的肺组织中的细胞坏死的细胞学检查显示没有可检测到的细胞异常。疫苗免疫后的小鼠行为在整个实验期间保持正常。一起,我们的数据表明,在细菌中合成的CTB-佐剂刺激的CTB-SARS-CoV-2-ACE-2-RBD嵌合粘膜疫苗蛋白可以在小鼠中产生持久和持久的IgA抗体,从而中和SARS-CoV-2亚变体OmicronBA.1.1。
    Mucosal vaccines protect against respiratory virus infection by stimulating the production of IgA antibodies that protect against virus invasion of the mucosal epithelium. In this study, a novel protein subunit mucosal vaccine was constructed for protection against infection by the beta coronavirus SARS-CoV-2. The vaccine was assembled by linking a gene encoding the SARS-CoV-2 virus S1 angiotensin converting enzyme receptor binding domain (ACE-2-RBD) downstream from a DNA fragment encoding the cholera toxin B subunit (CTB), a mucosal adjuvant known to stimulate vaccine immunogenicity. A 42 kDa vaccine fusion protein was identified in homogenates of transformed E. coli BL-21 cells by acrylamide gel electrophoresis and by immunoblotting against anti-CTB and anti-ACE-2-RBD primary antibodies. The chimeric CTB-SARS-CoV-2-ACE-2-RBD vaccine fusion protein was partially purified from clarified bacterial homogenates by nickel affinity column chromatography. Further vaccine purification was accomplished by polyacrylamide gel electrophoresis and electro-elution of the 42 kDa chimeric vaccine protein. Vaccine protection against SARS-CoV-2 infection was assessed by oral, nasal, and parenteral immunization of BALB/c mice with the CTB-SARS-CoV-2-ACE-2-RBD protein. Vaccine-induced SARS-CoV-2 specific antibodies were quantified in immunized mouse serum by ELISA analysis. Serum from immunized mice contained IgG and IgA antibodies that neutralized SARS-CoV-2 infection in Vero E6 cell cultures. In contrast to unimmunized mice, cytological examination of cell necrosis in lung tissues excised from immunized mice revealed no detectable cellular abnormalities. Mouse behavior following vaccine immunization remained normal throughout the duration of the experiments. Together, our data show that a CTB-adjuvant-stimulated CTB-SARS-CoV-2-ACE-2-RBD chimeric mucosal vaccine protein synthesized in bacteria can produce durable and persistent IgA antibodies in mice that neutralize the SARS-CoV-2 subvariant Omicron BA.1.1.
    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
    非人腺病毒(AdV)基因递送平台由于其在大多数个体中逃避预先存在的AdV载体免疫的能力而具有显著的价值。以前,我们已经证明,用BAd-H5HA鼻内(IN)免疫小鼠,表达H5N1流感病毒血凝素(HA)的牛AdV3型(BAdV3)载体,导致增强的体液和细胞介导的免疫反应。与用HAd-H5HA类似免疫的小鼠组相比,用抗原性不同的H5N1病毒攻击后,Bad-H5HAIN免疫导致完全保护。表达HA的人AdV5型(HAdV5)载体。
    这里,我们试图确定与HAd-H5HA接种组相比,鼻内接种BAd-H5HA的小鼠肺中先天免疫应答的激活。
    肺组织的RNA-Seq分析揭示了在用BAd-H5HA免疫的动物中参与先天和适应性免疫的基因的差异表达(DE)。通过RT-PCR验证前10个增强基因。始终如一,细胞因子(IL-1α,IL-1β,IL-5,TNF-α,LIF,IL-17,G-CSF,MIP-1β,与HAdV载体组相比,接种BAdV载体组的肺中的MCP-1,MIP-2和GM-CSF)和toll样受体。
    这些结果表明,与HAdV载体相比,BAdV载体在小鼠中诱导增强的先天和适应性免疫相关因子。因此,BAdV载体平台可能是重组疫苗和癌症免疫治疗的优良基因传递系统。
    Nonhuman adenoviral (AdV) gene delivery platforms have significant value due to their ability to elude preexisting AdV vector immunity in most individuals. Previously, we have demonstrated that intranasal (IN) immunization of mice with BAd-H5HA, a bovine AdV type 3 (BAdV3) vector expressing H5N1 influenza virus hemagglutinin (HA), resulted in enhanced humoral and cell-mediated immune responses. The BAd-H5HA IN immunization resulted in complete protection following the challenge with an antigenically distinct H5N1 virus compared to the mouse group similarly immunized with HAd-H5HA, a human AdV type 5 (HAdV5) vector expressing HA.
    Here, we attempted to determine the activation of innate immune responses in the lungs of mice inoculated intranasally with BAd-H5HA compared to the HAd-H5HA-inoculated group.
    RNA-Seq analyses of the lung tissues revealed differential expression (DE) of genes involved in innate and adaptive immunity in animals immunized with BAd-H5HA. The top ten enhanced genes were verified by RT-PCR. Consistently, there were transient increases in the levels of cytokines (IL-1α, IL-1β, IL-5, TNF- α, LIF, IL-17, G-CSF, MIP-1β, MCP-1, MIP-2, and GM-CSF) and toll-like receptors in the lungs of the group inoculated with BAdV vectors compared to that of the HAdV vector group.
    These results demonstrate that the BAdV vectors induce enhanced innate and adaptive immunity-related factors compared to HAdV vectors in mice. Thus, the BAdV vector platform could be an excellent gene delivery system for recombinant vaccines and cancer 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: Journal Article
    结核病是全球对人类健康的主要威胁。由于广泛使用的BCG疫苗在成人中效果不佳,需要开发一种新型的加强结核病疫苗。我们设计了一种新型鼻内结核候选疫苗,TB/FLU-04L,它基于编码两种分枝杆菌抗原的减毒甲型流感病毒载体,Ag85A和ESAT-6。由于结核病是一种空气传播疾病,诱导粘膜免疫的能力是流感载体的潜在优势之一。将ESAT-6和Ag85A抗原的序列插入甲型流感病毒的NS1开放阅读框以替换NS1蛋白的缺失的羧基部分。表达嵌合NS1蛋白的载体在小鼠和非人灵长类动物中似乎是遗传稳定且复制缺陷的。用TB/FLU-04L候选疫苗鼻内免疫C57BL/6小鼠或食蟹猴诱导Mtb特异性Th1免疫应答。与BCG相比,小鼠的单次TB/FLU-04L免疫显示出相当的保护水平,并且在“初免”方案中应用时,BCG的保护作用显着增加。我们的研究结果表明,用TB/FLU-04L疫苗鼻内免疫,携带两种分枝杆菌抗原,是安全的,并诱导针对毒力结核分枝杆菌的保护性免疫应答。
    Tuberculosis is a major global threat to human health. Since the widely used BCG vaccine is poorly effective in adults, there is a demand for the development of a new type of boost tuberculosis vaccine. We designed a novel intranasal tuberculosis vaccine candidate, TB/FLU-04L, which is based on an attenuated influenza A virus vector encoding two mycobacterium antigens, Ag85A and ESAT-6. As tuberculosis is an airborne disease, the ability to induce mucosal immunity is one of the potential advantages of influenza vectors. Sequences of ESAT-6 and Ag85A antigens were inserted into the NS1 open reading frame of the influenza A virus to replace the deleted carboxyl part of the NS1 protein. The vector expressing chimeric NS1 protein appeared to be genetically stable and replication-deficient in mice and non-human primates. Intranasal immunization of C57BL/6 mice or cynomolgus macaques with the TB/FLU-04L vaccine candidate induced Mtb-specific Th1 immune response. Single TB/FLU-04L immunization in mice showed commensurate levels of protection in comparison to BCG and significantly increased the protective effect of BCG when applied in a \"prime-boost\" scheme. Our findings show that intranasal immunization with the TB/FLU-04L vaccine, which carries two mycobacterium antigens, is safe, and induces a protective immune response against virulent M. tuberculosis.
    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
    新生儿免疫系统的不成熟导致对传染病的易感性增加和疫苗应答差。因此,需要更好的早期疫苗接种策略。佐剂可以增强免疫应答的幅度和持续时间。在这项研究中,我们评估了佐剂dmLT和mmCT和不同免疫途径的作用,皮下(s.c.)和鼻内(i.n.),新生儿对肺炎球菌结合疫苗Pn1-CRM197的免疫反应。单独用Pn1-CRM197免疫的新生小鼠的Pn1特异性抗体(Ab)水平较低。佐剂在免疫后8周增强IgGAb应答,s.c.免疫后比i.n.免疫后更多。相反,i.n.与s.c.免疫相比,用任一佐剂免疫增强血清和唾液IgA水平。此外,dmLT和mmCT都增强了生发中心的形成,因此,dmLT和mmCT增强了脾和骨髓(BM)中Pn1特异性IgGAb分泌细胞(ASCs)的诱导和持久性,无论免疫途径如何。此外,i.n.免疫增强BM中的Pn1特异性IgA+ASCs比s.c.免疫接种2022.1078904n。然而,需要较高i.n.剂量的Pn1-CRM197来实现与用任一佐剂进行s.c.免疫所引发的IgG应答相当的IgG应答。我们得出的结论是,在粘膜或肠胃外免疫后,dmLT和mmCT增强了新生儿对疫苗Pn1-CRM197的免疫反应的诱导和持久性。这表明dmLT和mmCT是开发安全有效的生命早期疫苗接种策略的有前途的佐剂。
    Immaturity of the neonatal immune system contributes to increased susceptibility to infectious diseases and poor vaccine responses. Therefore, better strategies for early life vaccination are needed. Adjuvants can enhance the magnitude and duration of immune responses. In this study we assessed the effects of the adjuvants dmLT and mmCT and different immunization routes, subcutaneous (s.c.) and intranasal (i.n.), on neonatal immune response to a pneumococcal conjugate vaccine Pn1-CRM197. Pn1-specific antibody (Ab) levels of neonatal mice immunized with Pn1-CRM197 alone were low. The adjuvants enhanced IgG Ab responses up to 8 weeks after immunization, more after s.c. than i.n. immunization. On the contrary, i.n. immunization with either adjuvant enhanced serum and salivary IgA levels more than s.c. immunization. In addition, both dmLT and mmCT enhanced germinal center formation and accordingly, dmLT and mmCT enhanced the induction and persistence of Pn1-specific IgG+ Ab-secreting cells (ASCs) in spleen and bone marrow (BM), irrespective of the immunization route. Furthermore, i.n. immunization enhanced Pn1-specific IgA+ ASCs in BM more than s.c. immunizatiofimmu.2022.1078904n. However, a higher i.n. dose of the Pn1-CRM197 was needed to achieve IgG response comparable to that elicited by s.c. immunization with either adjuvant. We conclude that dmLT and mmCT enhance both induction and persistence of the neonatal immune response to the vaccine Pn1-CRM197, following mucosal or parenteral immunization. This indicates that dmLT and mmCT are promising adjuvants for developing safe and effective early life vaccination strategies.
    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
    简介:卡介苗(卡介苗)的疗效有限,迫切需要新的有效疫苗接种方法来控制结核病。聚乳酸-羟基乙酸共聚物(PLGA)是一种普遍的药物递送系统。然而,基于PLGA的纳米颗粒(NPs)对结核病诱导粘膜免疫应答的作用尚未完全阐明.在这项研究中,我们假设用培养物滤液蛋白10(CFP10)负载的PLGANP(CFP10-NP)鼻内免疫可以增强BCG对小鼠牛分枝杆菌的保护性免疫力。方法:用PLGANPs包裹重组蛋白CFP10,通过经典的水-油-水溶剂蒸发法制备CFP10-NPs。然后,研究了CFP10-NP对体外巨噬细胞和体内BCG免疫小鼠的免疫调节作用。结果:我们使用具有带负电荷的表面(ζ电位-28.5±1.7mV)的球形CFP10-NP,其粒径为281.7±28.5nm。值得注意的是,CFP10-NPs显著增强J774A.1巨噬细胞中肿瘤坏死因子α(TNF-α)和白细胞介素(IL)-1β的分泌。此外,粘膜免疫CFP10-NP显著增加血清中TNF-α和IL-1β的产生,支气管肺泡灌洗液(BALF)中免疫球蛋白A(IgA)的分泌,并促进小鼠脾细胞中CFP10特异性干扰素-γ(IFN-γ)的分泌。此外,CFP10-NP免疫在M后3周显着降低了肺组织中的炎症面积和细菌负荷。Bovis挑战赛.结论:CFP10-NPs能显著提高卡介苗的免疫原性和保护作用。我们的发现探索了基于PLGANP的气道粘膜疫苗作为靶向肺部递送的载体的潜力。
    Introduction: The limited efficacy of BCG (bacillus Calmette-Guérin) urgently requires new effective vaccination approaches for the control of tuberculosis. Poly lactic-co-glycolic acid (PLGA) is a prevalent drug delivery system. However, the effect of PLGA-based nanoparticles (NPs) against tuberculosis for the induction of mucosal immune response is no fully elucidated. In this study, we hypothesized that intranasal immunization with culture filtrate protein-10 (CFP10)-loaded PLGA NPs (CFP10-NPs) could boost the protective immunity of BCG against Mycobacterium bovis in mice. Methods: The recombinant protein CFP10 was encapsulated with PLGA NPs to prepare CFP10-NPs by the classical water-oil-water solvent-evaporation method. Then, the immunoregulatory effects of CFP10-NPs on macrophages in vitro and on BCG-immunized mice in vivo were investigated. Results: We used spherical CFP10-NPs with a negatively charged surface (zeta-potential -28.5 ± 1.7 mV) having a particle size of 281.7 ± 28.5 nm in diameter. Notably, CFP10-NPs significantly enhanced the secretion of tumor necrosis factor α (TNF-α) and interleukin (IL)-1β in J774A.1 macrophages. Moreover, mucosal immunization with CFP10-NPs significantly increased TNF-α and IL-1β production in serum, and immunoglobulin A (IgA) secretion in bronchoalveolar lavage fluid (BALF), and promoted the secretion of CFP10-specific interferon-γ (IFN-γ) in splenocytes of mice. Furthermore, CFP10-NPs immunization significantly reduced the inflammatory area and bacterial load in lung tissues at 3-week post-M. bovis challenge. Conclusion: CFP10-NPs markedly improve the immunogenicity and protective efficacy of BCG. Our findings explore the potential of the airway mucosal vaccine based on PLGA NPs as a vehicle for targeted lung delivery.
    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
    粘膜组织构成了人体与外界环境之间最大的界面,调节病原体的进入,粒子,和分子。粘膜免疫是引发保护性粘膜免疫反应的最有效方法。然而,大多数目前许可的疫苗建议通过肌肉注射给药,有明显的缺点,例如高生产成本,患者依从性低,缺乏粘膜免疫反应。引发粘膜和全身免疫反应的策略正在开发中,包括适当的疫苗佐剂,输送系统,和细菌或病毒载体。由于其固有的免疫佐剂特性和保护抗原不被降解的能力,生物可降解的粘膜粘附纳米颗粒(NPs)是疫苗递送系统最有前途的候选物。持续释放加载的抗原,增加抗原在给药部位的停留时间。本综述概述了粘膜的复杂结构,NPs与粘膜相互作用的机制,影响NPs粘膜粘附的因素,以及基于粘膜粘附NPs的给药系统在疫苗领域的应用。此外,这篇综述表明,基于可生物降解和粘膜粘附NP的递送系统具有用于疫苗粘膜给药的潜力。
    Mucosal tissue constitutes the largest interface between the body and the external environment, regulating the entry of pathogens, particles, and molecules. Mucosal immunization is the most effective way to trigger a protective mucosal immune response. However, the majority of the currently licensed vaccines are recommended to be administered by intramuscular injection, which has obvious shortcomings, such as high production costs, low patient compliance, and lack of mucosal immune response. Strategies for eliciting mucosal and systemic immune responses are being developed, including appropriate vaccine adjuvant, delivery system, and bacterial or viral vectors. Biodegradable mucoadhesive nanoparticles (NPs) are the most promising candidate for vaccine delivery systems due to their inherent immune adjuvant property and the ability to protect the antigen from degradation, sustain the release of loaded antigen, and increase the residence time of antigen at the administration site. The current review outlined the complex structure of mucosa, the mechanism of interaction between NPs and mucosa, factors affecting the mucoadhesion of NPs, and the application of the delivery system based on mucoadhesive NPs in the field of vaccines. Moreover, this review demonstrated that the biodegradable and mucoadhesive NP-based delivery system has the potential for mucosal administration of 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
    新的疫苗设计方法,平台,和免疫策略可能促进抗病毒粘膜效应和记忆反应,以减少接种疫苗个体的无症状感染和传播。这里,我们研究了一种肠外和粘膜免疫联合方案,以诱导局部和血清抗体反应,采用基于表位的抗原3BT和NG19m。这些抗原分别靶向重要的新出现和重新出现的病毒PRRSV-2和SARS-CoV-2。我们评估了两种版本的3BT蛋白,其含有来自PRRSV-2的GP5包膜蛋白的保守表位:可溶并由重组杆状病毒BacDual-3BT表达。另一方面,NG19m,包含SARS-CoV-2的S蛋白的受体结合基序,仅被评估为可溶性重组蛋白。越南小型猪采用不同的接种途径进行免疫接种:皮下,鼻内,或两者的组合(s.c.-i.n.)。动物在血清和粘膜液中产生抗原结合和中性粒细胞抗体,不同的浓度和活动模式,取决于抗原和免疫计划。可溶性3BT是在血清中引发结合和中和抗体的有效免疫原,鼻粘液,还有阴道拭子.载体免疫原BacDual-3BT诱导血清和粘膜中的结合抗体,但PRRSV-2中和活性仅在鼻腔粘液中发现。NG19m促进血清和粘膜结合抗体,表现出不同的中和活性。只有皮下免疫动物的血清样品抑制RBD-ACE2相互作用,而小型猪鼻内接种或通过联合s.c.i.n.方案在上下呼吸道粘膜产生微妙的中和体液反应。我们的结果表明,鼻内免疫,单独或联合皮下递送基于表位的抗原,产生局部和全身结合和中和抗体。需要进一步的研究来评估诱导反应预防感染和减少传播的能力。
    New vaccine design approaches, platforms, and immunization strategies might foster antiviral mucosal effector and memory responses to reduce asymptomatic infection and transmission in vaccinated individuals. Here, we investigated a combined parenteral and mucosal immunization scheme to induce local and serum antibody responses, employing the epitope-based antigens 3BT and NG19m. These antigens target the important emerging and re-emerging viruses PRRSV-2 and SARS-CoV-2, respectively. We assessed two versions of the 3BT protein, which contains conserved epitopes from the GP5 envelope protein of PRRSV-2: soluble and expressed by the recombinant baculovirus BacDual-3BT. On the other hand, NG19m, comprising the receptor-binding motif of the S protein of SARS-CoV-2, was evaluated as a soluble recombinant protein only. Vietnamese mini-pigs were immunized employing different inoculation routes: subcutaneous, intranasal, or a combination of both (s.c.-i.n.). Animals produced antigen-binding and neut1ralizing antibodies in serum and mucosal fluids, with varying patterns of concentration and activity, depending on the antigen and the immunization schedule. Soluble 3BT was a potent immunogen to elicit binding and neutralizing antibodies in serum, nasal mucus, and vaginal swabs. The vectored immunogen BacDual-3BT induced binding antibodies in serum and mucosae, but PRRSV-2 neutralizing activity was found in nasal mucus exclusively when administered intranasally. NG19m promoted serum and mucosal binding antibodies, which showed differing neutralizing activity. Only serum samples from subcutaneously immunized animals inhibited RBD-ACE2 interaction, while mini-pigs inoculated intranasally or via the combined s.c.-i.n. scheme produced subtle neutralizing humoral responses in the upper and lower respiratory mucosae. Our results show that intranasal immunization, alone or combined with subcutaneous delivery of epitope-based antigens, generates local and systemic binding and neutralizing antibodies. Further investigation is needed to evaluate the capability of the induced responses to prevent infection and reduce transmission.
    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
    BCG is the only licensed vaccine against Mycobacterium tuberculosis (M.tb) infection. Due to its intramuscular administration route, BCG is unable to induce a local protective immune response in the respiratory system. Moreover, BCG has a diminished ability to induce long-lived memory T-cells which are indispensable for antituberculosis protection. Recently we described the protective efficacy of new mucosal TB vaccine candidate based on recombinant attenuated influenza vector (Flu/THSP) co-expressing TB10.4 and HspX proteins of M.tb within an NS1 influenza protein open reading frame. In the present work, the innate and adaptive immune response to immunization with the Flu/THSP and the immunological properties of vaccine candidate in the BCG-prime → Flu/THSP vector boost vaccination scheme are studied in mice. It was shown that the mucosal administration of Flu/THSP induces the incoming of interstitial macrophages in the lung tissue and stimulates the expression of co-stimulatory CD86 and CD83 molecules on antigen-presenting cells. The T-cellular immune response to Flu/THSP vector was mediated predominantly by the IFNγ-producing CD8+ lymphocytes. BCG-prime → Flu/THSP vector boost immunization scheme was shown to protect mice from severe lung injury caused by M.tb infection due to the enhanced T-cellular immune response, mediated by antigen-specific effector and central memory CD4+ and CD8+ T-lymphocytes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号