Mucosal immunity

粘膜免疫
  • 文章类型: Journal Article
    背景:单纯疱疹病毒1型(HSV-1)是病毒性脑炎的主要原因,生殖器粘膜感染,和新生儿感染。乳酸乳球菌(L.乳酸)已被证明是递送蛋白质抗原并刺激粘膜和全身免疫反应的有效载体。在这项研究中,我们构建了表达HSV-1保护性抗原糖蛋白D(gD)的重组乳酸乳球菌系统。
    结果:为了提高局部粘膜抗原刺激的稳定性和持久性,我们将免疫佐剂白细胞介素(IL)-2和IgG的Fc片段插入表达系统,构建了名为NZ3900-gD-IL-2-Fc的重组乳酸乳球菌。通过利用这种重组乳酸乳球菌菌株在小鼠中引发免疫反应并评估其保护作用,重组乳酸乳球菌疫苗诱导特异性中和抗体显著增加,IgG,IgA,干扰素-γ,和小鼠血清中的IL-4水平。此外,与对照组的小鼠相比,该疫苗还增强了响应gD的淋巴细胞的增殖水平。此外,表达gD的重组乳酸乳球菌通过激活免疫相关基因显著增强小鼠的非特异性免疫反应。此外,在鼠肺粘膜的HSV-1攻击后,接种实验疫苗的小鼠比对照小鼠表现出更少的肺损伤。
    结论:我们的研究提出了一种使用食品级,非致病性,和非商业细菌乳酸乳球菌。研究结果表明,该重组疫苗有望预防小鼠HSV-1感染。
    BACKGROUND: Herpes simplex virus type 1 (HSV-1) is a major cause of viral encephalitis, genital mucosal infections, and neonatal infections. Lactococcus lactis (L. lactis) has been proven to be an effective vehicle for delivering protein antigens and stimulating both mucosal and systemic immune responses. In this study, we constructed a recombinant L. lactis system expressing the protective antigen glycoprotein D (gD) of HSV-1.
    RESULTS: To improve the stability and persistence of antigen stimulation of the local mucosa, we inserted the immunologic adjuvant interleukin (IL)-2 and the Fc fragment of IgG into the expression system, and a recombinant L. lactis named NZ3900-gD-IL-2-Fc was constructed. By utilizing this recombinant L. lactis strain to elicit an immune response and evaluate the protective effect in mice, the recombinant L. lactis vaccine induced a significant increase in specific neutralizing antibodies, IgG, IgA, interferon-γ, and IL-4 levels in the serum of mice. Furthermore, in comparison to the mice in the control group, the vaccine also enhanced the proliferation levels of lymphocytes in response to gD. Moreover, recombinant L. lactis expressing gD significantly boosted nonspecific immune reactions in mice through the activation of immune-related genes. Furthermore, following the HSV-1 challenge of the murine lung mucosa, mice inoculated with the experimental vaccine exhibited less lung damage than control mice.
    CONCLUSIONS: Our study presents a novel method for constructing a recombinant vaccine using the food-grade, non-pathogenic, and non-commercial bacterium L. lactis. The findings indicate that this recombinant vaccine shows promise in preventing HSV-1 infection in mice.
    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
    口服疫苗通常被认为是安全的,易于管理,并有可能诱导全身和粘膜免疫反应。然而,鉴于恶劣的胃肠环境和粘液屏障带来的挑战,口服疫苗的开发需要使用安全有效的递送系统。近年来,基于纳米颗粒的递送已被证明是用于制造口服疫苗的理想递送载体。因此,考虑到上述情况,硫糖铝酸化(SA)包封N-2-羟丙基三甲基氯化铵壳聚糖(N-2-HACC)/N,制备了O-羧甲基壳聚糖(CMCS)纳米颗粒(SA@N-2-HACC/CMCSNPs),并将BSA作为模型抗原来研究免疫应答。SA@N-2-HACC/CMCSNPs具有227±7.0nm的粒度和8.43±2.62mV的ζ电位。NPs在模拟胃液和肠液中表现出缓慢和持续的释放和高稳定性。RAW264.7巨噬细胞样细胞系证明了SA@N-2-HACC/CMCS/BSA纳米颗粒的摄取增强。通过口服给药的疫苗显着增加了BSA在肠中的停留时间>12小时,并引起IgG和sIgA的产生。此处开发的用于口服给药的SA@N-2-HACC/CMCSNP是用于递送抗原的出色技术,并提供了粘膜疫苗研究的途径。
    Oral vaccines are generally perceived to be safe, easy to administer, and have the potential to induce both systemic and mucosal immune responses. However, given the challenges posed by the harsh gastrointestinal environment and mucus barriers, the development of oral vaccines necessitates the employment of a safe and efficient delivery system. In recent years, nanoparticle-based delivery has proven to be an ideal delivery vector for the manufacture of oral vaccines. Hence, considering the above, the sucralfate acidified (SA) encapsulated N-2-Hydroxypropyl trimethyl ammonium chloride chitosan (N-2-HACC)/N,O-carboxymethyl chitosan (CMCS) nanoparticles (SA@N-2-HACC/CMCS NPs) were prepared, and the BSA was used as a model antigen to investigated the immune responses. The SA@N-2-HACC/CMCS NPs had a particle size of 227 ± 7.0 nm and a zeta potential of 8.43 ± 2.62 mV. The NPs displayed slow and sustained release and high stability in simulated gastric juice and intestinal fluid. RAW 264.7 macrophage-like cell line demonstrated enhanced uptake of the SA@N-2-HACC/CMCS/BSA nanoparticles. The vaccine via oral administration markedly enhanced the residence time of BSA in the intestine for >12 h and elicited the production of IgG and sIgA. The SA@N-2-HACC/CMCS NPs developed here for oral administration is an excellent technique for delivering antigens and provides a path of mucosal vaccine research.
    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
    微生物组和粘膜免疫之间复杂的相互作用的重要性,特别是在呼吸道内,由于其对肺部疾病的严重程度和进展的潜在影响,已获得了广泛的关注。因此,这篇综述总结了呼吸道特异性微生物组影响粘膜免疫并最终影响呼吸道健康的特定相互作用。此外,我们讨论了微生物组如何影响粘膜免疫,考虑到组织特异性变异,以及它在包含哮喘的呼吸系统疾病中的能力,慢性阻塞性肺疾病,还有肺癌.此外,我们研究了影响呼吸道微生物组与粘膜免疫反应关系的外部因素。通过探索这些复杂的相互作用,这篇综述为基于微生物组的干预措施在调节粘膜免疫和减轻呼吸系统疾病严重程度方面的潜力提供了有价值的见解.
    The importance of the complex interplay between the microbiome and mucosal immunity, particularly within the respiratory tract, has gained significant attention due to its potential implications for the severity and progression of lung diseases. Therefore, this review summarizes the specific interactions through which the respiratory tract-specific microbiome influences mucosal immunity and ultimately impacts respiratory health. Furthermore, we discuss how the microbiome affects mucosal immunity, considering tissue-specific variations, and its capacity in respiratory diseases containing asthma, chronic obstructive pulmonary disease, and lung cancer. Additionally, we investigate the external factors which affect the relationship between respiratory microbiome and mucosal immune responses. By exploring these intricate interactions, this review provides valuable insights into the potential for microbiome-based interventions to modulate mucosal immunity and alleviate the severity of respiratory diseases.
    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
    Naegleriafowleri(N.fowleri)通过上呼吸道感染会导致致命的CNS疾病,称为原发性阿米巴脑膜脑炎(PAM)。对禽N.fowleri感染的强烈的体内免疫应答是表征该疾病的免疫病理学的基础。然而,对这种病原体逃避免疫控制的原因知之甚少。感染发生在看似健康的个体中,缺乏有效的临床选择,因此死亡率接近98%。尚不清楚宿主因素如何或是否可能导致易感性或疾病恶化。然而,缺乏工具阻碍了体内免疫反应和疾病进展的机理研究。在这项研究中,我们已经产生了单克隆抗体,以N.fowleri表面抗原,并证明他们是很好的工具,用于研究体内免疫应答。我们还鉴定了一个单克隆,2B6,在体外具有有效的固有抗变形虫活性。这种抗体还能够在体内治疗上延长宿主的存活时间,具有更能够指导免疫效应物活性的同种型的重组抗体在治疗性给予时进一步提高了存活率。因此,我们报道了一种新的单克隆抗体的产生,该抗体可以增强有益的免疫功能。即使在疾病期间给予治疗。我们相信这为PAM中治疗性抗体治疗的潜力提供了证据。IMPORTANCENaegleriafowleri(N.fowleri)是一种自由生活的变形虫,在温暖的淡水中无处不在。虽然人类接触很常见,它很少导致发病机制。然而,当N.Fowleri进入上呼吸道时,特别是嗅觉粘膜,感染导致称为原发性阿米巴脑膜脑炎(PAM)的致命疾病。作为一个自由生活的变形虫,N.fowleri不需要哺乳动物宿主;事实上,它可以准确地描述为偶然的机会病原体。虽然大多数机会性感染发生在免疫功能低下的人类中,没有报道的免疫功能障碍与鸡传染性猪瘟感染相关。因此,N.Fowleri机会主义的基础尚不清楚,以及为什么有些人开发PAM而另一些人不开发PAM的原因根本没有得到很好的理解。有理由推测局部或急性免疫衰竭,甚至可能缺乏先前的适应性免疫,与疾病易感性有关。在哺乳动物宿主中仔细的免疫分析和体内对福氏N.fowleri免疫反应的表征是迫切需要的,以了解哪些宿主因素对防御至关重要。以及这些反应如何以导致致命感染的方式受损。为了确定基因和途径,提供抵抗体内鸡的感染,我们产生了表面反应性单克隆抗体(Abs),可在体内快速检测和定量变形虫。有趣的是,在人和动物的血清和唾液中很容易检测到N.fowleri结合抗体,这表明非致死性暴露会引起针对变形虫的体液免疫反应。然而,目前尚不清楚Abs如何在体内与Naegleria相互作用或有助于预防致死性感染.在这项研究中,我们已经产生并表征了单克隆抗体(Ab),克隆2B6,其识别存在于体外培养的家禽猪链球菌以及小鼠传代的家禽猪链球菌中的糖基化表面抗原。当克隆2B6与家禽N.Fowleri结合时,抑制阿米巴的运动性和摄食行为,导致强烈的生长抑制作用。用Ab全身和脑内处理的小鼠表现出延迟的疾病发作和延长的存活。此外,我们发现,通过抗体同种型增强免疫定向效应子活性可以进一步增强生存率,而没有明显的免疫致病性副作用.这些发现显示了抗体治疗作为目前在PAM中使用的那些的额外治疗剂的潜力。
    Naegleria fowleri (N. fowleri) infection via the upper respiratory tract causes a fatal CNS disease known as primary amoebic meningoencephalitis (PAM). The robust in vivo immune response to N. fowleri infection underlies the immunopathology that characterizes the disease. However, little is known about why this pathogen evades immune control. Infections occur in seemingly healthy individuals and effective clinical options are lacking, thus a nearly 98% fatality rate. It is unclear how or if host factors may contribute to susceptibility or disease exacerbation, yet mechanistic studies of the in vivo immune response and disease progression are hampered by a lack of tools. In this study, we have generated monoclonal antibodies to N. fowleri surface antigens and shown them to be excellent tools for studying the in vivo immune response. We also identified one monoclonal, 2B6, with potent inherent anti-amoebastatic activity in vitro. This antibody is also able to therapeutically prolong host survival in vivo and furthermore, recombinant antibodies with an isotype more capable of directing immune effector activity further improved survival when given therapeutically. Thus, we report the generation of a novel monoclonal antibody to N. fowleri that can enhance beneficial immune functions, even when given therapeutically during disease. We believe this provides evidence for the potential of therapeutic antibody treatments in PAM.IMPORTANCENaegleria fowleri (N. fowleri) is a free-living amoeba that is found ubiquitously in warm freshwater. While human exposure is common, it rarely results in pathogenesis. However, when N. fowleri gains access to the upper airway, specifically the olfactory mucosa, infection leads to a lethal disease known as primary amoebic meningoencephalitis (PAM). As a free-living amoeba, N. fowleri does not need a mammalian host; indeed, it can be accurately described as an accidental opportunistic pathogen. While most opportunistic infections occur in humans who are immunocompromised, there are no reported immune dysfunctions associated with N. fowleri infection. Therefore, the basis for N. fowleri opportunism is not known, and the reasons why some humans develop PAM while others do not are simply not well understood. It is reasonable to speculate that local or acute immune failures, potentially even a lack of prior adaptive immunity, are related to disease susceptibility. Careful immune profiling and characterization of the in vivo immune response to N. fowleri in a mammalian host are desperately needed to understand which host factors are critical to defense, and how these responses might be compromised in a way that results in lethal infection. To identify genes and pathways that provide resistance against in vivo N. fowleri infection, we generated surface reactive monoclonal antibodies (Abs) that provide rapid amoeba detection and quantification in vivo. Interestingly, N. fowleri binding Abs have been readily detected in the serum and saliva of humans and animals suggesting that non-lethal exposure drives a humoral immune response against the amoeba. Yet, how Abs might interact with Naegleria in vivo or contribute to preventing lethal infection is not well understood. In this study, we have generated and characterized a monoclonal antibody (Ab), Clone 2B6, that recognizes a glycosylated surface antigen present in cultured in vitro N. fowleri as well as mouse passaged N. fowleri. When clone 2B6 binds to N. fowleri, it inhibits amoeba motility and feeding behavior, leading to strong growth inhibition. Mice treated systemically and intracerebrally with Ab displayed a delayed disease onset and prolonged survival. In addition, we found that enhancing immune-directed effector activity via antibody isotype could further enhance survival without obvious immunopathogenic side effects. These findings show the potential for antibody treatment as an additional therapeutic to those used currently in PAM.
    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
    这篇透视文章探讨了阴阳理论-中国古代哲学基石-与复杂的免疫学领域的新颖整合。鉴于免疫学固有的复杂概念,许多学生发现很难理解控制免疫平衡和调节的微妙机制。鉴于中国学生对阴阳理论的根深蒂固的理解,我们提倡一种教育策略,将免疫平衡的概念置于阴阳的框架内,从而提供更直观和引人入胜的学习体验。这种方法不仅利用了阴阳的文化意义,但也符合其平衡与和谐的原则,从而反映了免疫反应的稳态本质。本文严格评估了这种技术增强中国学生免疫理解能力的能力,同时也考虑到它的局限性。尽管有这些限制,这些看似不同的领域的融合为增强免疫学教育带来了巨大的希望,促进批判性思维,推进跨文化学术话语。古老的哲学见解与现代科学探索的融合促使人们重新评估免疫学中的教育方法,强调一种新颖的教学方法,将传统智慧与当代科学教育联系起来。
    This perspective article delves into a novel integration of Yin-Yang theory-an ancient Chinese philosophical cornerstone-with the sophisticated realm of immunology. Given the intricate concepts inherent in immunology, many students find it challenging to comprehend the delicate mechanisms governing immune equilibrium and regulation. Given the deep-rooted understanding of Yin-Yang theory among Chinese students, we advocate for an educational strategy that contextualizes the concept of immune equilibrium within the framework of Yin-Yang, thereby offering a more intuitive and engaging learning experience. This method not only capitalizes on the cultural significance of Yin-Yang, but also corresponds to its principles of equilibrium and harmony, thus mirroring the homeostatic essence of immune responses. This article critically assesses this technique\'s capacity to bolster immune comprehension amongst Chinese students, while also considering its limitations. Despite these limitations, the fusion of these seemingly divergent fields holds substantial promise for augmenting immunology education, promoting critical thinking, and advancing cross-cultural academic discourse. The amalgamation of age-old philosophical insights with modern scientific exploration prompts a reassessment of educational methodologies within immunology, underscoring a novel pedagogical approach that bridges traditional wisdom with contemporary scientific education.
    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
    抗球虫疫苗包含艾美耳球虫的活卵囊,艾美球虫,最大艾美球虫,和艾美耳球虫被用来控制球虫病。本研究探讨了IL-1β作为分子佐剂增强坏死艾美耳球虫免疫原性和粘膜免疫的潜力。我们设计了E.necatrix以表达功能性chIL-1β(EnIL-1β),并用野生型(EnWT)和转基因(EnIL-1β)菌株的卵囊免疫鸡,分别。然后用EnWT卵囊攻击鸡,以检查chIL-1β的免疫原性增强潜力。不出所料,与使用EnWT免疫的鸡相比,EnIL-1β免疫的鸡的卵囊输出显着降低。EnIL-1β和EnWT组的体重增加和病变评分没有差异。小肠和盲肠中的寄生虫负荷表明,EnIL-1β的侵袭和复制不受影响。然而,免疫原性和粘膜屏障的标志物,在EnIL-1β感染的鸡中,Claudin-1和禽β-防御素-1升高。因此,chIL-1β在E.necatrix中的异位表达似乎可以改善其免疫原性和粘膜免疫,不增加致病性。我们的发现支持chIL-1β作为开发有效的基于活卵囊的抗球虫疫苗的候选者。
    Anticoccidial vaccines comprising living oocysts of Eimeria tenella, Eimeria necatrix, Eimeria maxima, and Eimeria acervulina are used to control coccidiosis. This study explored the potential of IL-1β to act as a molecular adjuvant for enhancing the immunogenicity of Eimeria necatrix and mucosal immunity. We engineered E. necatrix to express a functional chIL-1β (EnIL-1β) and immunized chickens with oocysts of the wild type (EnWT) and tranegenic (EnIL-1β) strains, respectively. The chickens were then challenged with EnWT oocysts to examine the immunogenicity-enhancing potential of chIL-1β. As expected, the oocyst output of EnIL-1β-immunized chickens was significantly reduced compared to those immunized using EnWT. No difference in body weight gain and lesion scores of EnIL-1β and EnWT groups was observed. The parasite load in the small intestine and caeca showed that the invasion and replication of EnIL-1β was not affected. However, the markers of immunogenicity and mucosal barrier, Claudin-1 and avian β-defensin-1, were elevated in EnIL-1β-infected chickens. Ectopic expression of chIL-1β in E. necatrix thus appears to improve its immunogenicity and mucosal immunity, without increasing pathogenicity. Our findings support chIL-1β as a candidate for development of effective live-oocyst-based anticoccidial 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: Case Reports
    自2022年以来,欧洲疾病预防和控制中心报告了19例新生儿严重的回声病毒感染,其中9例是致命的。我们报告了一个新的致命新生儿病例,该病例发生在一个男性双胞胎中,我们评估了呼吸道和肠道粘膜先天免疫反应。
    The European Center for Disease Prevention and Control has reported 19 cases of severe echovirus 11 infections in neonates since 2022, nine of which were fatal. We report a new fatal neonatal case that occurred in a male twin for which we evaluated the respiratory and intestinal mucosal innate immune response.
    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
    针对严重急性呼吸综合征(SARS)-CoV-2的分泌性(S)IgA抗体通过自然感染在唾液和上呼吸道(URT)分泌物中诱导,并且可能对确定初始感染的结果至关重要。分泌型IgA1(SIgA1)是这些分泌物中抗体的主要同种型。SARS-CoV-2的中和最有效地通过聚合抗体如SIgA来完成。我们假设,通过独特的细菌IgA1蛋白酶将针对SARS-CoV-2的SIgA1抗体裂解为具有降低的病毒中和活性的单价Fabα抗体片段,将促进病毒下降到肺部以引起严重疾病,并增强其通过空气传播给其他人。最近对SARS-CoV-2感染患者的鼻咽微生物群的研究表明,与健康受试者相比,产生IgA1蛋白酶的细菌的比例显着增加。类似的考虑也可能适用于其他呼吸道病毒感染,包括流感,可能解释流感最初归因于流感嗜血杆菌,产生IgA1蛋白酶。
    Secretory (S) IgA antibodies against severe acute respiratory syndrome (SARS)-CoV-2 are induced in saliva and upper respiratory tract (URT) secretions by natural infection and may be critical in determining the outcome of initial infection. Secretory IgA1 (SIgA1) is the predominant isotype of antibodies in these secretions. Neutralization of SARS-CoV-2 is most effectively accomplished by polymeric antibodies such as SIgA. We hypothesize that cleavage of SIgA1 antibodies against SARS-CoV-2 by unique bacterial IgA1 proteases to univalent Fabα antibody fragments with diminished virus neutralizing activity would facilitate the descent of the virus into the lungs to cause serious disease and also enhance its airborne transmission to others. Recent studies of the nasopharyngeal microbiota of patients with SARS-CoV-2 infection have revealed significant increases in the proportions of IgA1 protease-producing bacteria in comparison with healthy subjects. Similar considerations might apply also to other respiratory viral infections including influenza, possibly explaining the original attribution of influenza to Haemophilus influenzae, which produces IgA1 protease.
    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
    黏膜免疫在防御冠状病毒中起着至关重要的作用,特别是在呼吸部位,作为抵御病毒入侵和复制的第一道防线。冠状病毒已经在粘膜免疫系统中开发了各种免疫逃避策略,阻碍被感染细胞的识别和逃避抗体反应。了解免疫机制和反应对于开发针对冠状病毒的有效疫苗和疗法至关重要。粘膜免疫在COVID-19中的作用是显著的,影响对病毒的局部和全身免疫反应。尽管大多数临床研究集中在外周血中的抗体和细胞免疫,呼吸道粘膜免疫反应在病毒复制的早期限制和SARS-CoV-2的清除中起关键作用。与病毒清除相关的粘膜生物标志物的鉴定将允许监测感染诱导的免疫。正在对粘膜递送疫苗和临床试验中的疫苗进行比较和对比,以了解它们在诱导针对冠状病毒的粘膜免疫方面的有效性。更深入地了解基于肺组织的免疫可能导致改进的诊断和预后程序以及旨在减少社区获得性肺炎疾病负担的新治疗策略。避免感染的全身表现和过高的发病率和死亡率。这篇全面的综述文章概述了目前关于粘膜免疫反应在清除SARS-CoV-2感染中的作用的证据,以及针对(再)感染的潜在粘膜保护机制。它还提出,在COVID-19中,粘膜免疫以及分泌和循环IgA抗体都有重要作用,并且重要的是要阐明这一点,以了解特别是无症状和轻度感染状态,这似乎占了大多数病例。此外,粘膜免疫可能被用于有益的诊断,治疗性的,或预防目的。最近对COVID-19粘膜免疫的研究结果可用于开发有效的疫苗和治疗方法,可以有效地靶向粘膜和全身免疫反应。
    Mucosal immunity plays a crucial role in defending against coronaviruses, particularly at respiratory sites, serving as the first line of defense against viral invasion and replication. Coronaviruses have developed various immune evasion strategies at the mucosal immune system, hindering the recognition of infected cells and evading antibody responses. Understanding the immune mechanisms and responses is crucial for developing effective vaccines and therapeutics against coronaviruses. The role of mucosal immunity in COVID-19 is significant, influencing both local and systemic immune responses to the virus. Although most clinical studies focus on antibodies and cellular immunity in peripheral blood, mucosal immune responses in the respiratory tract play a key role in the early restriction of viral replication and the clearance of SARS-CoV-2. Identification of mucosal biomarkers associated with viral clearance will allow monitoring of infection-induced immunity. Mucosally delivered vaccines and those under clinical trials are being compared and contrasted to understand their effectiveness in inducing mucosal immunity against coronaviruses. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community-acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality. This comprehensive review article outlines the current evidence about the role of mucosal immune responses in the clearance of SARS-CoV-2 infection, as well as potential mucosal mechanisms of protection against (re-)infection. It also proposes that there is a significant role for mucosal immunity and for secretory as well as circulating IgA antibodies in COVID-19, and that it is important to elucidate this in order to comprehend especially the asymptomatic and mild states of the infection, which appear to account for the majority of cases. Moreover, it is possible that mucosal immunity can be exploited for beneficial diagnostic, therapeutic, or prophylactic purposes. The findings from recent studies on mucosal immunity in COVID-19 can be used to develop effective vaccines and treatments that can effectively target both mucosal and systemic immune responses.
    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
    流产衣原体(C.abortus)是一种重要的人畜共患病原体,严重危害畜牧业的发展。接种疫苗是预防流产梭菌感染的最有效方法。我们先前报道了一种基于重组大肠杆菌幽灵(rECG)的C.abortus疫苗,该疫苗表现出出色的保护功效。在这项研究中,我们进一步尝试融合霍乱毒素B亚基(CTB),一种被广泛研究的强效粘膜免疫佐剂,与巨噬细胞感染增效剂(MIP),流产梭菌的候选抗原,并探讨其对流产梭菌感染的保护作用。MIP融合蛋白在rECGs中高表达,和CTB修饰的rECGs在体外显着诱导小鼠骨髓来源的树突状细胞的激活。用rECGs鼻内免疫诱导Th1偏向的细胞免疫应答。与没有CTB的rECG相比,CTB修饰的rECGs诱导血清和阴道洗涤液中IgA的浓度较高.此外,在小鼠感染模型中,CTB修饰的rECGs可显著提高流产梭菌的清除效率,减轻子宫的病理损伤。该研究表明,将CTB掺入rECG显著增强rECG疫苗的免疫原性潜力,并且可以显著增强其针对流产梭菌攻击的保护功效。
    Chlamydia abortus (C. abortus) is an important zoonotic pathogen that seriously endangers the development of animal husbandry. Vaccination is the most effective approach to preventing C. abortus infection. We previously reported a recombinant Escherichia coli ghost (rECG)-based C. abortus vaccine that demonstrated outstanding protective efficacy. In this study, we further attempted to fuse the cholera toxin B subunit (CTB), a widely studied potent mucosal immune adjuvant, with macrophage infectivity potentiator (MIP), a candidate antigen of C. abortus, on the surface of the rECG and explore its protective effect against C. abortus infection. The MIP fusion protein was highly expressed in the rECGs, and the CTB-modified rECGs significantly induced the activation of mouse bone marrow-derived dendritic cells in vitro. Intranasal immunization with rECGs induced a Th1-biased cellular immune response. Compared to the rECGs without CTB, the CTB-modified rECGs induced higher concentrations of IgA in the serum and vaginal wash solution. Moreover, in a mouse infection model, the CTB-modified rECGs significantly improved the clearance efficiency of C. abortus and reduced the pathological damage to the uterus. This study demonstrates that incorporating CTB into rECGs significantly enhances the immunogenic potential of the rECG vaccine and can significantly enhance its protective efficacy against a C. abortus challenge.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号