intranasal vaccine

鼻内疫苗
  • 文章类型: Journal Article
    严重急性呼吸道综合征冠状病毒2(SARS-CoV-2),冠状病毒病(COVID-19)的病原体,2019年在全球范围内迅速传播。随着Omicron变体的出现,COVID-19进入流行阶段。鉴于秋季和冬季的病例预计会增加,实施COVID-19季节性加强疫苗的策略对于保护公众健康变得越来越有价值.这种做法已经存在用于季节性流感疫苗以对抗年度流感季节。我们的目标是研究一种易于修改的疫苗平台,用于季节性对抗SARS-CoV-2。在这项研究中,我们评估了转基因流感病毒ΔNA(RBD)作为COVID-19的鼻内疫苗候选物。对这种修饰的病毒进行工程改造,以将神经氨酸酶(NA)蛋白的编码序列替换为SARS-CoV-2的受体结合域(RBD)蛋白的膜锚定形式。我们设计了实验,使用致死(Delta)和非致死(Omicron)攻击模型评估K18-hACE2小鼠中ΔNA(RBD)的保护作用。COVID-19mRNA疫苗的对照和我们实验室先前描述的鼻内病毒样颗粒疫苗被用作比较。用表达祖先RBD的ΔNA(RBD)免疫在小鼠血清中引起高抗RBDIgG水平,肺组织中的高抗RBDIgA,并提高了Delta变异攻击后的生存率。修饰ΔNA(RBD)以表达Omicron变体RBD改变了Omicron变体攻击后小鼠肺中的变体特异性抗体应答和有限的病毒负荷。总的来说,该数据表明ΔNA(RBD)可能是产生针对SARS-CoV-2的粘膜和全身免疫的有效鼻内疫苗平台。
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease (COVID-19), rapidly spread across the globe in 2019. With the emergence of the Omicron variant, COVID-19 shifted into an endemic phase. Given the anticipated rise in cases during the fall and winter seasons, the strategy of implementing seasonal booster vaccines for COVID-19 is becoming increasingly valuable to protect public health. This practice already exists for seasonal influenza vaccines to combat annual influenza seasons. Our goal was to investigate an easily modifiable vaccine platform for seasonal use against SARS-CoV-2. In this study, we evaluated the genetically modified influenza virus ΔNA(RBD) as an intranasal vaccine candidate for COVID-19. This modified virus was engineered to replace the coding sequence for the neuraminidase (NA) protein with a membrane-anchored form of the receptor binding domain (RBD) protein of SARS-CoV-2. We designed experiments to assess the protection of ΔNA(RBD) in K18-hACE2 mice using lethal (Delta) and non-lethal (Omicron) challenge models. Controls of COVID-19 mRNA vaccine and our lab\'s previously described intranasal virus like particle vaccine were used as comparisons. Immunization with ΔNA(RBD) expressing ancestral RBD elicited high anti-RBD IgG levels in the serum of mice, high anti-RBD IgA in lung tissue, and improved survival after Delta variant challenge. Modifying ΔNA(RBD) to express Omicron variant RBD shifted variant-specific antibody responses and limited viral burden in the lungs of mice after Omicron variant challenge. Overall, this data suggests that ΔNA(RBD) could be an effective intranasal vaccine platform that generates mucosal and systemic immunity towards SARS-CoV-2.
    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
    2019年冠状病毒病(COVID-19),由严重急性呼吸综合征冠状病毒-2(SARS-CoV-2)引起,成为2019年的全球疫情,深刻影响人类健康和全球经济。为了克服这一挑战,开发了各种疫苗模式并将其商业化。包括灭活疫苗,mRNA疫苗,基于腺病毒载体的疫苗,和亚单位疫苗。虽然肌内疫苗诱导高IgG水平,它们通常不能刺激呼吸系统中显著的粘膜免疫。我们使用表达SARS-CoV-2Beta变体(rK148/beta-S)的刺突蛋白的新城疫病毒(NDV)载体,并评估了在K18-hACE2转基因小鼠中用rK148/β-S鼻内接种的功效。低剂量(106.0EID50)的鼻内疫苗接种在用SARS-CoV-2Beta变体攻击后可获得86%的存活率。高剂量(107.0EID50)的给药导致肺病毒载量降低,并且针对SARS-CoV-2Beta和Delta变体的存活率为100%。在SARS-CoV-2攻击后,在接种疫苗的小鼠肺中也诱导了高水平的SARS-CoV-2尖峰特异性IgA。我们的发现表明,rK148/beta-S有望作为一种鼻内疫苗候选物,可有效诱导针对SARS-CoV-2的粘膜免疫。
    Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), emerged as a global outbreak in 2019, profoundly affecting both human health and the global economy. Various vaccine modalities were developed and commercialized to overcome this challenge, including inactivated vaccines, mRNA vaccines, adenovirus vector-based vaccines, and subunit vaccines. While intramuscular vaccines induce high IgG levels, they often fail to stimulate significant mucosal immunity in the respiratory system. We employed the Newcastle disease virus (NDV) vector expressing the spike protein of the SARS-CoV-2 Beta variant (rK148/beta-S), and evaluated the efficacy of intranasal vaccination with rK148/beta-S in K18-hACE2 transgenic mice. Intranasal vaccination with a low dose (106.0 EID50) resulted in an 86% survival rate after challenge with the SARS-CoV-2 Beta variant. Administration at a high dose (107.0 EID50) led to a reduction in lung viral load and 100% survival against the SARS-CoV-2 Beta and Delta variants. A high level of the SARS-CoV-2 spike-specific IgA was also induced in vaccinated mice lungs following the SARS-CoV-2 challenge. Our findings suggest that rK148/beta-S holds promise as an intranasal vaccine candidate that effectively induces mucosal immunity against SARS-CoV-2.
    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
    这项研究评估了到达奥地利育肥农场时超声检查和体格检查的意义和预测价值。治疗频率和平均日增重(ADG)与物理和超声检查结果有关。此外,研究了鼻内接种疫苗对一半受检小牛的影响。在育肥开始和结束时记录600头小牛的临床和超声健康状况。一半的小牛接受鼻内接种(Bovalto®Respi鼻内接种)。总的来说,44.5%的人在到达农场时经胸超声检查(TUS)显示呼吸评分(RS)异常,56.0%的人显示呼吸道疾病的迹象。对于RS和TUS,对疾病和健康进行了分类。结果显示,与健康小牛(RS中位数:1.01kg/d;TUS中位数:1.01kg/d)相比,ILL小牛的ADG较低(RS中位数:0.93kg/d;TUS中位数:0.96kg/d)。与治疗和ILL小牛(RS中位数1.01kg/d;TUS:1.02kg/d)相比,未治疗和ILL小牛(RS中位数0.90kg/d;TUS:0.93kg/d)的中位ADG较低。疫苗接种不影响生长性能或ILL的发生,虽然VAC小牛的治疗频率较低(NVAC为17.0%;VAC为11.3%)。呼吸道疾病检查方案的实施可能会对生产参数产生积极影响(例如,治疗频率和ADG)。
    This study evaluated the significance and predictive value of ultrasonographic and physical examination on arrival at an Austrian fattening farm. Treatment frequency and average daily weight gain (ADG) were related to physical and ultrasonographic examination results. Additionally, the effect of an intranasal vaccination in half of the examined calves was studied. The clinical and ultrasonographic health status 600 calves was recorded at the beginning and end of fattening. Half of the calves received an intranasal vaccination (Bovalto® Respi Intranasal). Overall, 44.5% showed an abnormal respiratory scoring (RS) and 56.0% showed signs of respiratory diseases in transthoracic ultrasonography (TUS) at arrival on the farm. For both RS and TUS, a categorization between ILL and HEALTHY was conducted. Results showed lower ADG in ILL calves (RS median: 0.93 kg/d; TUS median: 0.96 kg/d) compared to HEALTHY calves (RS median: 1.01 kg/d; TUS median: 1.01 kg/d). The median ADG was lower in not treated and ILL calves (RS median 0.90 kg/d; TUS: 0.93 kg/d) compared to treated and ILL calves (RS median 1.01 kg/d; TUS: 1.02 kg/d). Vaccination did not affect growth performance or occurrence of ILL, though treatment frequency was lower in VAC calves (17.0% in NVAC; 11.3% in VAC). The implementation of examination protocols for respiratory diseases may have a positive impact on production parameters (e.g., treatment frequency and ADG).
    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
    流感是一种高度传染性的呼吸道疾病,估计每年有300万至500万例严重疾病病例。虽然大多数流感疫苗是通过注射肠胃外给药的,一个缺点是它们在感染部位不会产生强烈的免疫反应,这在大流行中可能变得很重要。鼻内疫苗可以产生局部和全身保护性免疫反应,可以降低成本,加强行政管理的便捷性。先前的研究表明,肠胃外施用的携带M2e蛋白(OMV-M2e)序列的外膜囊泡(OMV)可防止小鼠和雪貂的甲型/PR8流感攻击。在目前的研究中,我们测量了OMV-M2e疫苗鼻内途径对小鼠流感A/PR8株的有效性.我们在用OMV-M2e鼻内接种的小鼠中观察到攻击后的高抗M2eIgG和IgA滴度。此外,我们在接种疫苗的小鼠中观察到Th1/Tc1偏差,Th17/Tc17反应增加,两者均与A/PR8攻击后的生存率相关,并显着降低肺病毒滴度。我们得出的结论是,0MV-M2e疫苗的鼻内途径施用是产生针对甲型流感的保护的有希望的方法,因为它导致与病毒攻击的存活相关的促炎免疫应答增加。
    Influenza is a highly contagious respiratory disease, resulting in an estimated 3 to 5 million cases of severe illness annually. While most influenza vaccines are administered parenterally via injection, one shortcoming is that they do not generate a strong immune response at the site of infection, which can become important in a pandemic. Intranasal vaccines can generate both local and systemic protective immune responses, can reduce costs, and enhance ease of administration. Previous studies showed that parenterally administered outer membrane vesicles (OMVs) that carry sequences of the M2e protein (OMV-M2e) protect against influenza A/PR8 challenge in mice and ferrets. In the current study, we measured the effectiveness of the intranasal route of the OMV-M2e vaccine against the influenza A/PR8 strain in mice. We observed high anti-M2e IgG and IgA titers post-challenge in mice vaccinated intranasally with OMV-M2e. In addition, we observed a Th1/Tc1 bias in the vaccinated mice, and an increased Th17/Tc17 response, both of which correlated with survival to A/PR8 challenge and significantly lower lung viral titers. We conclude that the intranasal-route administration of the OMV-M2e vaccine is a promising approach toward generating protection against influenza A as it leads to an increased proinflammatory immune response correlating with survival to viral challenge.
    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
    嵌合蛋白,由SARS-CoV-2的保守核衣壳(N)和S2蛋白的两个片段形成,在大肠杆菌中作为重组构建体获得。N片段属于C末端结构域,而S2片段在刺突蛋白的融合后构象中跨越纤维结构。产生的蛋白质,名为S2NDH,能够形成10纳米的球形颗粒,其在与CpGODN-39M混合时形成聚集体。两种制剂均被阳性COVID-19人血清识别。通过鼻内途径施用的S2NDH+ODN-39M制剂在Balb/c小鼠中产生高度免疫原性。它在血清和支气管肺泡液中诱导交叉反应性抗N体液免疫,在Th1模式下。细胞介导的免疫(CMI)也很广泛,甚至对SARS-CoV-1的N蛋白也有阳性反应。然而,未获得针对S2区的中和抗体(NAb)或CMI。作为替代,RBD蛋白作为NAb的诱导物包含在制剂中。通过鼻内途径对小鼠进行评估后,S2NDH+ODN-39M制剂相对于RBD检测到明显的佐剂作用。针对SARS-CoV-2和SARS-CoV-1诱导高水平的NAb。双价配方S2NDH+ODN-39M+RBD,通过鼻内途径给药,构成了一个有吸引力的建议作为加强疫苗的sbecovirus范围。
    A chimeric protein, formed by two fragments of the conserved nucleocapsid (N) and S2 proteins from SARS-CoV-2, was obtained as a recombinant construct in Escherichia coli. The N fragment belongs to the C-terminal domain whereas the S2 fragment spans the fibre structure in the post-fusion conformation of the spike protein. The resultant protein, named S2NDH, was able to form spherical particles of 10 nm, which forms aggregates upon mixture with the CpG ODN-39M. Both preparations were recognized by positive COVID-19 human sera. The S2NDH + ODN-39M formulation administered by the intranasal route resulted highly immunogenic in Balb/c mice. It induced cross-reactive anti-N humoral immunity in both sera and bronchoalveolar fluids, under a Th1 pattern. The cell-mediated immunity (CMI) was also broad, with positive response even against the N protein of SARS-CoV-1. However, neither neutralizing antibodies (NAb) nor CMI against the S2 region were obtained. As alternative, the RBD protein was included in the formulation as inducer of NAb. Upon evaluation in mice by the intranasal route, a clear adjuvant effect was detected for the S2NDH + ODN-39M preparation over RBD. High levels of NAb were induced against SARS-CoV-2 and SARS-CoV-1. The bivalent formulation S2NDH + ODN-39M + RBD, administered by the intranasal route, constitutes an attractive proposal as booster vaccine of sarbecovirus scope.
    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
    人类副流感病毒3型(HPIV3)是一种主要的儿科呼吸道病原体,缺乏可用的疫苗或抗病毒药物。我们通过密码子对去优化(CPD)产生了活的减毒HPIV3疫苗候选物。HPIV3开放阅读框(ORFs)编码核蛋白(N),磷蛋白(P),矩阵(M),融合(F),血凝素-神经氨酸酶(HN),和聚合酶(L)被单独或组合修饰以产生12种病毒,命名为Min-N,Min-P,Min-M,Min-FHN,Min-L,Min-NP,Min-NPM,最小不良贷款,Min-PM,Min-PFHN,Min-MFHN,和Min-PMFHN。N或L的CPD严重降低了体外生长,没有进一步评估。P或M的CPD与体外干扰素(IFN)反应增加和减少有关,分别,但对病毒复制影响不大。在Vero细胞中,F和HN延迟病毒复制的CPD,但最终滴度与野生型(wt)HPIV3相当。在人肺上皮A549细胞中,CPDF和HN诱导更强的IFN应答,病毒滴度降低了100倍,F和HN蛋白的表达显着降低,而不影响N或P或蛋白质在病毒体中的相对包装。仓鼠鼻内感染后,对于携带CPDF和HN的病毒,鼻甲和肺中的复制倾向于减少最多,最大减少约10倍。尽管体内复制减少(体外CPDF和HN的表达降低),所有病毒均诱导与wt相似的血清HPIV3中和抗体滴度,并提供针对HPIV3攻击的完全保护。总之,HPIV3的CPD产生了适合进一步开发的有希望的疫苗候选物。
    Human parainfluenza virus type 3 (HPIV3) is a major pediatric respiratory pathogen lacking available vaccines or antiviral drugs. We generated live-attenuated HPIV3 vaccine candidates by codon-pair deoptimization (CPD). HPIV3 open reading frames (ORFs) encoding the nucleoprotein (N), phosphoprotein (P), matrix (M), fusion (F), hemagglutinin-neuraminidase (HN), and polymerase (L) were modified singly or in combination to generate 12 viruses designated Min-N, Min-P, Min-M, Min-FHN, Min-L, Min-NP, Min-NPM, Min-NPL, Min-PM, Min-PFHN, Min-MFHN, and Min-PMFHN. CPD of N or L severely reduced growth in vitro and was not further evaluated. CPD of P or M was associated with increased and decreased interferon (IFN) response in vitro, respectively, but had little effect on virus replication. In Vero cells, CPD of F and HN delayed virus replication, but final titers were comparable to wild-type (wt) HPIV3. In human lung epithelial A549 cells, CPD F and HN induced a stronger IFN response, viral titers were reduced 100-fold, and the expression of F and HN proteins was significantly reduced without affecting N or P or the relative packaging of proteins into virions. Following intranasal infection in hamsters, replication in the nasal turbinates and lungs tended to be the most reduced for viruses bearing CPD F and HN, with maximum reductions of approximately 10-fold. Despite decreased in vivo replication (and lower expression of CPD F and HN in vitro), all viruses induced titers of serum HPIV3-neutralizing antibodies similar to wt and provided complete protection against HPIV3 challenge. In summary, CPD of HPIV3 yielded promising vaccine candidates suitable for further development.
    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
    紧急研究创新的严重急性呼吸道冠状病毒-2(SARS-CoV-2)疫苗,可以成功地防止各种新出现的变种,特别是Omicron变体及其亚变体,是必要的。这里,我们设计了一种嵌合腺病毒载体疫苗,命名为Ad5-Beta/Delta.这种疫苗是通过整合来自Delta变体的受体结合结构域而产生的,有L452R和T478K突变,变成β变体的完整刺突蛋白。用Ad5-β/Deta疫苗进行的肌内(IM)和鼻内(IN)疫苗接种均诱导了针对包含OmicronBA.5的变体的强大的广谱中和。用Ad5-Beta/Delta疫苗进行的IN免疫表现出优异的粘膜免疫,表现为较高的分泌性IgA抗体和呼吸道中更多的组织驻留记忆T细胞(TRM)。Ad5-β/δ疫苗的IM和IN递送的组合能够协同地引发更强的全身和粘膜免疫应答。此外,在两个剂量的mRNA或亚单位重组蛋白疫苗后,Ad5-Beta/Delta疫苗接种显示出更有效的增强作用,表明其在异源疫苗接种中作为加强剂的利用能力。这些结果定量的Ad5-β/Delta疫苗作为有利的疫苗可以提供相对于关注的SARS-CoV-2pre-Omicron变体和包含BA.5的Omicron亚变体的保护性免疫。
    Urgent research into innovative severe acute respiratory coronavirus-2 (SARS-CoV-2) vaccines that may successfully prevent various emerging emerged variants, particularly the Omicron variant and its subvariants, is necessary. Here, we designed a chimeric adenovirus-vectored vaccine named Ad5-Beta/Delta. This vaccine was created by incorporating the receptor-binding domain from the Delta variant, which has the L452R and T478K mutations, into the complete spike protein of the Beta variant. Both intramuscular (IM) and intranasal (IN) vaccination with Ad5-Beta/Deta vaccine induced robust broad-spectrum neutralization against Omicron BA.5-included variants. IN immunization with Ad5-Beta/Delta vaccine exhibited superior mucosal immunity, manifested by higher secretory IgA antibodies and more tissue-resident memory T cells (TRM) in respiratory tract. The combination of IM and IN delivery of the Ad5-Beta/Delta vaccine was capable of synergically eliciting stronger systemic and mucosal immune responses. Furthermore, the Ad5-Beta/Delta vaccination demonstrated more effective boosting implications after two dosages of mRNA or subunit recombinant protein vaccine, indicating its capacity for utilization as a booster shot in the heterologous vaccination. These outcomes quantified Ad5-Beta/Delta vaccine as a favorable vaccine can provide protective immunity versus SARS-CoV-2 pre-Omicron variants of concern and BA.5-included Omicron subvariants.
    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
    用编码利什曼菌抗原LACK(LACKDNA)的无佐剂质粒DNA进行鼻内(i.n.)接种已显示可诱导针对啮齿动物皮肤和内脏利什曼病的保护性免疫。在目前的工作中,我们试图评估d的安全性和有效性,1-甘油醛交联壳聚糖微粒(CCM)作为LACKDNA非膨胀粘膜粘附递送系统。制备直径为5μm的CCM,并吸附了最大2.4%(w/w)的DNA,而没有体积变化。用LACKDNA/CCM滴注的小鼠鼻孔的组织学分析显示,微粒不仅具有粘膜粘附性,而且还具有粘膜减少性,不诱导局部炎症。观察到两次鼻腔滴注间隔一周没有改变支气管肺泡细胞或血液嗜酸性粒细胞的数量;没有改变ALT,AST和肌酐血清水平;并且没有引起皮肤超敏反应。当亚马逊利什曼原虫在脚垫上挑战时,与单独使用裸LACKDNA或CCM的动物相比,小鼠的寄生虫负荷显着降低。伴随着Th1偏倚反应的刺激增加,从较高的T-bet/GATA-3比率和IFN-γ水平可以看出。一起,这些结果表明,CCM是一种安全有效的减毒载体,可以提高i.n.LACKDNA疫苗对皮肤利什曼病的疗效。
    Intranasal (i.n.) vaccination with adjuvant-free plasmid DNA encoding the leishmanial antigen LACK (LACK DNA) has shown to induce protective immunity against both cutaneous and visceral leishmaniasis in rodents. In the present work, we sought to evaluate the safety and effectiveness of d,l-glyceraldehyde cross-linked chitosan microparticles (CCM) as a LACK DNA non-intumescent mucoadhesive delivery system. CCM with 5 μm of diameter was prepared and adsorbed with a maximum of 2.4 % (w/w) of DNA with no volume alteration. Histological analysis of mouse nostrils instilled with LACK DNA / CCM showed microparticles to be not only mucoadherent but also mucopenetrant, inducing no local inflammation. Systemic safeness was confirmed by the observation that two nasal instillations one week apart did not alter the numbers of bronchoalveolar cells or blood eosinophils; did not alter ALT, AST and creatinine serum levels; and did not induce cutaneous hypersensitivity. When challenged in the footpad with Leishmania amazonensis, mice developed significantly lower parasite loads as compared with animals given naked LACK DNA or CCM alone. That was accompanied by increased stimulation of Th1-biased responses, as seen by the higher T-bet / GATA-3 ratio and IFN-γ levels. Together, these results demonstrate that CCM is a safe and effective mucopenetrating carrier that can increase the efficacy of i.n. LACK DNA vaccination against cutaneous leishmaniasis.
    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
    鼻内减毒活疫苗(LAIV)用于在分离后刺激扁桃体单眼细胞(MNC)。几种流感病毒株的血凝素(HA)蛋白用于检测HA特异性IgG,IgM和IgA抗体使用ELISA。刺激后在细胞培养上清液中检测到显着的抗sH1N1HAIgGIgA和IgM抗体滴度(分别为平均值±SE:0.43±0.09,平均值±SE:0.23±0.04和平均值±SE:0.47±0.05,p<0.01)。LAIV刺激扁桃体MNCs诱导显著IgG,针对pH1N1HA的IgA和IgM抗体(平均值±SE:1.35±0.12),(平均值±SE:0.35±0.06)和(平均值±SE:0.58±0.10),p<0.01。令人惊讶的是,显示LAIV诱导与禽流感病毒(aH5N1)交叉反应性抗aH5N1HA抗体(平均值±SE:0.84±0.20,p<0.01)。在LAIV刺激后,在细胞培养上清液中也检测到显著水平的抗H2N2HAIgG抗体(平均值±SE:0.93±0.23,p<0.01)。LAIV刺激扁桃体MNCs后发现高水平的抗sH3N2HAIgG抗体,(平均值±SE:1.2±0.23p<0.01)。用于评估B细胞对LAIV的应答的人鼻相关淋巴组织(NALT)的当前模型显然是研究未来鼻内疫苗的成功模型。
    Intranasal live attenuated influenza vaccine (LAIV) was used to stimulate tonsillar monocular cells (MNCs) following isolation. Haemagglutinin (HA) proteins of several influenza strains were used for the detection of HA-specific IgG, IgM and IgA antibodies using ELISA. Significant anti-sH1N1 HA IgG IgA and IgM antibody titres were detected in cell culture supernatants after stimulation (mean ± SE: 0.43 ± 0.09, mean ± SE: 0.23 ± 0.04 and mean ± SE: 0.47 ± 0.05 respectively, p < 0.01). LAIV stimulation of tonsillar MNCs induced significant IgG, IgA and IgM antibodies to the pH1N1 HA (mean ± SE:1.35 ± 0.12), (mean ± SE: 0.35 ± 0.06) and (mean ± SE: 0.58 ± 0.10) respectively, p < 0.01. Surprisingly, LAIV was shown to induce cross-reactive anti-aH5N1 HA antibodies (mean ± SE: 0.84 ± 0.20, p < 0.01) to avian influenza virus (aH5N1). Anti-H2N2 HA IgG antibody was also detected in the cell culture supernatants in a significant level after LAIV stimulation (mean ± SE: 0.93 ± 0.23, p < 0.01). High levels of anti-sH3N2 HA IgG antibody was discovered after LAIV stimulation of tonsillar MNCs, (mean ± SE: 1.2 ± 0.23p < 0.01). The current model of human nasal-associated lymphoid tissue (NALT) to evaluate B cells responses to LAIV was evident that it is a successful model to study future intranasal 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
    引起粘膜免疫的鼻内疫苗被认为对呼吸道感染有效,例如严重急性呼吸道综合征冠状病毒2(SARS-CoV-2),但是它们诱导以免疫球蛋白A(IgA)和IgG产生为特征的体液免疫的能力很低。据报道,用粘性碱性羧基乙烯基聚合物(CVP)和病毒抗原的混合物接种疫苗可诱导强烈的全身和粘膜免疫应答。在这项研究中,我们通过使用CVP在抗原接种后立即诱导的空间转录组谱分析分析了鼻腔中免疫活性细胞随时间的行为.我们建立了一种在小鼠鼻腔中使用Visium系统进行空间转录组学的方法,并在鼻内接种疫苗后分析了鼻腔内的基因表达谱。糖蛋白2(Gp2)-,SRY-box转录因子8(Sox8)-,SARS-CoV-2刺突蛋白和CVP(S-CVP)接种后3-6小时,或表达Spi-B转录因子(Spib)的细胞在鼻通道(NP)区域增加。结果表明,微折(M)细胞在短时间内(3-6小时)被激活。随后鼻腔中细胞的聚类分析显示,在用S-CVP鼻内接种后3-6小时,簇9增加。我们发现簇9中的Il6在3-6小时在NP内具有最高的log2倍数值。搜索与Il6相似的基因表达模式显示,3-6小时后,鼻腔中Edn2,Ccl20和Hk2的log2倍数值也增加。结果表明,免疫细胞在鼻内接种疫苗后立即发生早期反应。在这项研究中,我们使用空间转录组学数据的时间序列分析,鉴定了在小鼠鼻内接种抗原-CVP后,导致M细胞和免疫活性细胞活化的基因表达变化.结果有助于鉴定在鼻黏膜免疫的初始诱导过程中被激活的细胞类型。
    Intranasal vaccines that elicit mucosal immunity are deemed effective against respiratory tract infections such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but their ability to induce humoral immunity characterized by immunoglobulin A (IgA) and IgG production is low. It has been reported that vaccination with a mixture of a viscous base carboxyvinyl polymer (CVP) and viral antigens induced robust systemic and mucosal immune responses. In this study, we analyzed the behavior of immunocompetent cells in the nasal cavity over time by spatial transcriptome profiling induced immediately after antigen vaccination using CVP. We established a method for performing spatial transcriptomics using the Visium system in the mouse nasal cavity and analyzed gene expression profiles within the nasal cavity after intranasal vaccination. Glycoprotein 2 (Gp2)-, SRY-box transcription factor 8 (Sox8)-, or Spi-B transcription factor (Spib)-expressing cells were increased in the nasal passage (NP) region at 3-6 hr after SARS-CoV-2 spike protein and CVP (S-CVP) vaccination. The results suggested that microfold (M) cells are activated within a short period of time (3-6 hr). Subsequent cluster analysis of cells in the nasal cavity showed an increase in Cluster 9 at 3-6 hr after intranasal vaccination with the S-CVP. We found that Il6 in Cluster 9 had the highest log2 fold values within the NP at 3-6 hr. A search for gene expression patterns similar to that of Il6 revealed that the log2 fold values of Edn2, Ccl20, and Hk2 also increased in the nasal cavity after 3-6 hr. The results showed that the early response of immune cells occurred immediately after intranasal vaccination. In this study, we identified changes in gene expression that contribute to the activation of M cells and immunocompetent cells after intranasal vaccination of mice with antigen-CVP using a time-series analysis of spatial transcriptomics data. The results facilitated the identification of the cell types that are activated during the initial induction of nasal mucosal immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号