peptide vaccine

肽疫苗
  • 文章类型: Journal Article
    对用鸟分枝杆菌亚种疫苗接种的牛的离体回忆反应进行分析。副结核(Map)rel缺失突变体显示免疫应答针对Map的35kD主要膜蛋白(MMP)。用MMP引发的抗原呈递细胞(APC)引起CD8细胞毒性记忆T细胞(CTL)的扩增,具有杀死细胞内细菌的能力。CTL的发育是MHC限制的。基因MAP2121c,编码MMP,被修饰用于在哺乳动物细胞系中表达MMP(tPA-MMP-2mut)以探索开发MMP作为疫苗的潜力。PBMC的离体刺激,从地图免费牛,用tPA-MMP-2mut表达的p35引发的APC引起的主要CD8CTL反应与用Maprel缺失突变体或MMP疫苗接种的牛的PBMC引起的回忆反应相当。在本研究中,改良的MMP基因,现在被称为p35NN,将其置于牛疱疹病毒-4(BoHV4)载体中以确定BoHV-4AΔTK-p35NN作为基于肽的疫苗的潜在用途。用BoHV-4AΔTK-p35NN对健康牛进行皮下注射引起CTL回忆反应,离体检测。结果表明,使用病毒载体是递送作为疫苗的MMP的有效方式。当被修饰用于在哺乳动物细胞中表达时,MMP的免疫原性活性没有丧失。下一步是进行田间试验,以确定是否存在对MMP的免疫反应阻止Map建立感染。
    Analysis of the recall response ex vivo in cattle vaccinated with a Mycobacterium avium subsp. paratuberculosis (Map) rel deletion mutant revealed the immune response was directed toward a 35 kD major membrane protein (MMP) of Map. Antigen presenting cells (APC) primed with MMP elicited expansion of CD8 cytotoxic memory T cells (CTL) with ability to kill intracellular bacteria. Development of CTL was MHC-restricted. The gene MAP2121c, encoding MMP, was modified for expression of MMP (tPA-MMP-2mut) in a mammalian cell line to explore the potential of developing MMP as a vaccine. Ex vivo stimulation of PBMC, from Map free cattle, with APC primed with tPA-MMP-2mut expressed p35 elicited a primary CD8 CTL response comparable to the recall response elicited with PBMC from cattle vaccinated with either the Maprel deletion mutant or MMP. In the present study, the modified gene for MMP, now referred to as p35NN, was placed into a bovine herpes virus-4 (BoHV4) vector to determine the potential use of BoHV-4AΔTK-p35NN as a peptide-based vaccine. Subcutaneous vaccination of healthy cattle with BoHV-4AΔTK-p35NN elicited a CTL recall response, as detected ex vivo. The results show use of a virus vector is an effective way for delivery of MMP as a vaccine. The immunogenic activity of MMP was not lost when modified for expression in mammalian cells. The next step is to conduct a field trial to determine if presence of an immune response to MMP prevents Map from establishing an infection.
    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
    中东已经见证了传染性登革热病毒的更大传播,血清型2(DENV-2)是最普遍的形式。通过这项工作,通过免疫信息学方法产生了针对DENV-2的靶向E和非结构(NS1)蛋白的多表位肽疫苗.预测了NS1和包膜E蛋白序列中的MHCI类和II类和LBL表位及其抗原性,毒性,和变应原性进行了调查。对人群覆盖率的研究表明,在DENV-2流行的不同国家中,NS1和包膜E表位的患病率很高。Further,从NS1表位检索的CTL和HTL表位与其他DENV血清型(1、3和4)均表现出高保守百分比。产生三种疫苗构建体,并使用C-IMMSIM和HADDOCK(针对TLR2,3,4,5和7)估计构建体的预期免疫应答。具有TLR4的疫苗构建体2的分子动力学模拟表明构建体与受体的高结合亲和力和稳定性,这可能预示有利的体内相互作用和免疫应答。
    The Middle East has witnessed a greater spread of infectious Dengue viruses, with serotype 2 (DENV-2) being the most prevalent form. Through this work, multi-epitope peptide vaccines against DENV-2 that target E and nonstructural (NS1) proteins were generated through an immunoinformatic approach. MHC class I and II and LBL epitopes among NS1 and envelope E proteins sequences were predicted and their antigenicity, toxicity, and allergenicity were investigated. Studies of the population coverage denoted the high prevalence of NS1 and envelope-E epitopes among different countries where DENV-2 endemic. Further, both the CTL and HTL epitopes retrieved from NS1 epitopes exhibited high conservancies\' percentages with other DENV serotypes (1, 3, and 4). Three vaccine constructs were created and the expected immune responses for the constructs were estimated using C-IMMSIM and HADDOCK (against TLR 2,3,4,5, and 7). Molecular dynamics simulation for vaccine construct 2 with TLR4 denoted high binding affinity and stability of the construct with the receptor which might foretell favorable in vivo interaction and 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
    在这项研究中,我们使用免疫信息学方法来预测寨卡病毒(ZIKV)蛋白的抗原表位,以帮助设计针对ZIKV的疫苗抗原。我们进行了CD8+T淋巴细胞和ZIKV蛋白的抗原B细胞表位的预测。评估了T细胞表位与主要组织相容性复合物I类(MHC-I)蛋白的结合相互作用。我们选择了抗原性,保守的,无毒,和免疫原性表位,这表明与人类白细胞抗原(HLA-A和HLA-B)等位基因存在显着的相互作用,全球人口覆盖率为76.35%。预测的表位在接头和佐剂的帮助下连接。然后通过与TLR3和TLR8的分子对接分析疫苗抗原,并将其在pVAX1载体中进行计算机克隆以用作DNA疫苗并设计为mRNA疫苗。
    In this study, we used an immunoinformatics approach to predict antigenic epitopes of Zika virus (ZIKV) proteins to assist in designing a vaccine antigen against ZIKV. We performed the prediction of CD8+ T-lymphocyte and antigenic B-cell epitopes of ZIKV proteins. The binding interactions of T-cell epitopes with major histocompatibility complex class I (MHC-I) proteins were assessed. We selected the antigenic, conserved, nontoxic, and immunogenic epitopes, which indicated significant interactions with the human leucocyte antigen (HLA-A and HLA-B) alleles and worldwide population coverage of 76.35%. The predicted epitopes were joined with the help of linkers and an adjuvant. The vaccine antigen was then analyzed through molecular docking with TLR3 and TLR8, and it was in silico cloned in the pVAX1 vector to be used as a DNA vaccine and designed as a mRNA vaccine.
    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
    在许多国家,糖尿病肾病仍然是终末期肾病的主要原因,需要额外的治疗靶点来防止其发展和进展。一些血管生成因子参与了糖尿病肾病的发病机制。Vasohibin-2(VASH2)是一种新型的促血管生成因子,我们先前的研究表明,糖尿病Vash2纯合基因敲除小鼠的肾小球损伤受到抑制。因此,我们建立了VASH2靶向肽疫苗,作为糖尿病肾病抗VASH2治疗的工具.在这项研究中,在链脲佐菌素(STZ)诱导的糖尿病小鼠模型中,研究了VASH2靶向肽疫苗对肾小球损伤的预防作用.以间隔2周的两个剂量对小鼠皮下注射疫苗,然后连续5天腹膜内注射50mg/kgSTZ。在第一次接种疫苗后20周评估肾小球损伤。用VASH2靶向肽疫苗治疗成功地诱导循环抗VASH2抗体,而在主要器官中没有炎症。虽然接种疫苗不会影响血糖水平,它显著阻止了高血糖引起的尿白蛋白排泄和肾小球体积增加.疫苗接种不影响增加的VASH2表达,但显著抑制糖尿病小鼠中的肾血管生成素-2(Angpt2)表达。此外,它显著防止肾小球巨噬细胞浸润。在db/db小鼠中也证实了疫苗接种对肾小球损伤的预防作用。一起来看,这项研究的结果表明,VASH2靶向肽疫苗可能通过抑制Angpt2介导的微炎症来预防小鼠糖尿病性肾小球损伤.
    Diabetic nephropathy remains the leading cause of end-stage kidney disease in many countries, and additional therapeutic targets are needed to prevent its development and progression. Some angiogenic factors are involved in the pathogenesis of diabetic nephropathy. Vasohibin-2 (VASH2) is a novel proangiogenic factor, and our previous study showed that glomerular damage is inhibited in diabetic Vash2 homozygous knockout mice. Therefore, we established a VASH2-targeting peptide vaccine as a tool for anti-VASH2 therapy in diabetic nephropathy. In this study, the preventive effects of the VASH2-targeting peptide vaccine against glomerular injury were examined in a streptozotocin (STZ)-induced diabetic mouse model. The mice were subcutaneously injected with the vaccine at two doses 2 wk apart and then intraperitoneally injected with 50 mg/kg STZ for 5 consecutive days. Glomerular injury was evaluated 20 wk after the first vaccination. Treatment with the VASH2-targeting peptide vaccine successfully induced circulating anti-VASH2 antibody without inflammation in major organs. Although the vaccination did not affect blood glucose levels, it significantly prevented hyperglycemia-induced increases in urinary albumin excretion and glomerular volume. The vaccination did not affect increased VASH2 expression but significantly inhibited renal angiopoietin-2 (Angpt2) expression in the diabetic mice. Furthermore, it significantly prevented glomerular macrophage infiltration. The preventive effects of vaccination on glomerular injury were also confirmed in db/db mice. Taken together, the results of this study suggest that the VASH2-targeting peptide vaccine may prevent diabetic glomerular injury in mice by inhibiting Angpt2-mediated microinflammation.NEW & NOTEWORTHY This study demonstrated preventive effects of VASH2-targeting peptide vaccine therapy on albuminuria and glomerular microinflammation in STZ-induced diabetic mouse model by inhibiting renal Angpt2 expression. The vaccination was also effective in db/db mice. The results highlight the importance of VASH2 in the pathogenesis of early-stage diabetic nephropathy and the practicability of anti-VASH2 strategy as a vaccine therapy.
    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
    多重耐药(MDR)鲍曼不动杆菌是与医院获得性感染相关的机会致病菌。由于其环境持久性,毒力,和有限的治疗选择,这种病菌导致患者死亡率增加和医疗费用增加.因此,预防性疫苗接种可能是干预易感人群中MDR不动杆菌感染的理想方法.在这项研究中,我们利用免疫信息学鉴定了含有与鲍曼不动杆菌发病机制相关的蛋白质中推定的B细胞和T细胞表位的肽.使用鲍曼不动杆菌硫氧还蛋白A(TrxA)前导蛋白序列和五种鉴定的肽抗原,构建了一种新型不动杆菌多表位疫苗(AMEV2)。来自鲍曼不动杆菌感染的小鼠的抗血清证明了对rAMEV2的反应性,并且用rAMEV2对小鼠的皮下免疫产生了针对构建体以及肽组分的高抗体效价。免疫导致分泌IL-4的脾细胞的频率增加,指示Th2应答。AMEV2免疫的小鼠被保护免受鲍曼不动杆菌高毒力菌株的鼻内攻击,并在48小时时显示出减少的细菌负荷。所有模拟接种疫苗的小鼠在3天内死于感染。本文的结果提供了对基于免疫信息学的疫苗设计的有效性及其作为对抗MDR病原体上升的有效策略的潜力的见解。
    Multi-drug-resistant (MDR) Acinetobacter baumannii is an opportunistic pathogen associated with hospital-acquired infections. Due to its environmental persistence, virulence, and limited treatment options, this organism causes both increased patient mortality and incurred healthcare costs. Thus, prophylactic vaccination could be ideal for intervention against MDR Acinetobacter infection in susceptible populations. In this study, we employed immunoinformatics to identify peptides containing both putative B- and T-cell epitopes from proteins associated with A. baumannii pathogenesis. A novel Acinetobacter Multi-Epitope Vaccine (AMEV2) was constructed using an A. baumannii thioredoxin A (TrxA) leading protein sequence followed by five identified peptide antigens. Antisera from A. baumannii infected mice demonstrated reactivity to rAMEV2, and subcutaneous immunization of mice with rAMEV2 produced high antibody titer against the construct as well as peptide components. Immunization results in increased frequency of IL-4-secreting splenocytes indicative of a Th2 response. AMEV2-immunized mice were protected against intranasal challenge with a hypervirulent strain of A. baumannii and demonstrated reduced bacterial burden at 48 h. In contrast, all mock vaccinated mice succumbed to infection within 3 days. Results presented here provide insight into the effectiveness of immunoinformatic-based vaccine design and its potential as an effective strategy to combat the rise of MDR pathogens.
    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
    人类中的啮齿动物传播的Arenavirus导致了区域性流行情况的出现,并已深深成为引起大流行的病毒。Arenavirus有一个双egmentedAmbisenseRNA,产生四种蛋白质:糖蛋白,核衣壳,RdRp和Z蛋白。基于肽的疫苗靶向免疫系统遇到的病毒的糖蛋白。B细胞和T细胞表位的筛选是基于它们的免疫学特性,如抗原性,变应原性,进行毒性和抗炎特性。然后将选择的表位聚类,并使用接头序列将表位缝合。检查了疫苗构建体的免疫学和物理化学性质,并通过2步MD模拟验证了模型结构。检查疫苗的热稳定性,然后进行免疫模拟,以测试疫苗在接下来的100天的过程中引入体内后的免疫原性,并进行密码子优化。最后,设计了443个氨基酸长的肽疫苗,其可以在世界范围内的多种群体中提供针对哺乳动物病毒家族的若干成员的保护,如表位保护和群体覆盖率分析所指示的。设计靶向哺乳动物病毒糖蛋白的肽疫苗的这项研究可能有助于在不久的将来开发新的疗法。
    The rodent-borne Arenavirus in humans has led to the emergence of regional endemic situations and has deeply emerged into pandemic-causing viruses. Arenavirus have a bisegmented ambisense RNA that produces four proteins: glycoprotein, nucleocapsid, RdRp and Z protein. The peptide-based vaccine targets the glycoprotein of the virus encountered by the immune system. Screening of B-Cell and T-Cell epitopes was done based on their immunological properties like antigenicity, allergenicity, toxicity and anti-inflammatory properties were performed. Selected epitopes were then clustered and epitopes were stitched using linker sequences. The immunological and physico-chemical properties of the vaccine construct was checked and modelled structure was validated by a 2-step MD simulation. The thermostability of the vaccine was checked followed by the immune simulation to test the immunogenicity of the vaccine upon introduction into the body over the course of the next 100 days and codon optimization was performed. Finally a 443 amino acid long peptide vaccine was designed which could provide protection against several members of the mammarenavirus family in a variety of population worldwide as denoted by the epitope conservancy and population coverage analysis. This study of designing a peptide vaccine targeting the glycoprotein of mammarenavirues may help develop novel therapeutics in near future.
    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
    疫苗已经大大减少了许多致命的病毒感染的影响。然而,鉴于反复发生的大流行和流行病,越来越需要加快疫苗的开发。此外,由于各种因素,识别针对某些病毒的疫苗具有挑战性,特别是不能在细胞培养物中培养某些病毒,以及人类MHC谱的广泛多样性。幸运的是,反向疫苗(RV)有效地克服了这些限制,并简化了整个蛋白质组抗原蛋白表位的鉴定,简化疫苗开发过程。此外,它使得能够产生多表位疫苗,所述多表位疫苗能够有效地解释人类群体中MHC谱的变化。RV方法在开发针对病毒病原体的精确和有效的疫苗方面提供了许多优势,包括广泛的蛋白质组覆盖,准确的表位鉴定,交叉保护能力,MHC相容性。随着RV的推出,研究人员越来越重视创建基于多表位的疫苗,旨在刺激宿主对多种血清型的免疫反应,与单组分单价替代品相反。不管基于RV的候选疫苗看起来多么有前途,它们必须经过实验验证,以探索其在实际应用中的保护功效。时间,努力,现在可以将分配给费力的表位鉴定过程的资源重定向到通过RV方法鉴定的验证疫苗候选物。然而,为了克服基于RV的方法中的失败,必须努力纳入免疫学原理,并考虑靶向涉及疾病发病机理的表位区域,免疫反应,和中和抗体成熟。在RV中整合多组学并整合人工智能和基于机器学习的工具和技术将增加开发有效疫苗的机会。这篇评论彻底解释了RV方法,理想的基于RV的疫苗构建组件,基于RV的疫苗旨在对抗病毒病原体,其挑战,和未来的前景。
    Vaccines have significantly reduced the impact of numerous deadly viral infections. However, there is an increasing need to expedite vaccine development in light of the recurrent pandemics and epidemics. Also, identifying vaccines against certain viruses is challenging due to various factors, notably the inability to culture certain viruses in cell cultures and the wide-ranging diversity of MHC profiles in humans. Fortunately, reverse vaccinology (RV) efficiently overcomes these limitations and has simplified the identification of epitopes from antigenic proteins across the entire proteome, streamlining the vaccine development process. Furthermore, it enables the creation of multiepitope vaccines that can effectively account for the variations in MHC profiles within the human population. The RV approach offers numerous advantages in developing precise and effective vaccines against viral pathogens, including extensive proteome coverage, accurate epitope identification, cross-protection capabilities, and MHC compatibility. With the introduction of RV, there is a growing emphasis among researchers on creating multiepitope-based vaccines aiming to stimulate the host\'s immune responses against multiple serotypes, as opposed to single-component monovalent alternatives. Regardless of how promising the RV-based vaccine candidates may appear, they must undergo experimental validation to probe their protection efficacy for real-world applications. The time, effort, and resources allocated to the laborious epitope identification process can now be redirected toward validating vaccine candidates identified through the RV approach. However, to overcome failures in the RV-based approach, efforts must be made to incorporate immunological principles and consider targeting the epitope regions involved in disease pathogenesis, immune responses, and neutralizing antibody maturation. Integrating multi-omics and incorporating artificial intelligence and machine learning-based tools and techniques in RV would increase the chances of developing an effective vaccine. This review thoroughly explains the RV approach, ideal RV-based vaccine construct components, RV-based vaccines designed to combat viral pathogens, its challenges, and future perspectives.
    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
    类风湿性关节炎(RA)被认为是由引发和加重炎症状况的自身(或修饰的自身或微生物模拟物)抗原的T细胞应答驱动的自身免疫性关节疾病。RA的较新治疗采用针对细胞因子或免疫细胞受体的单克隆抗体或重组受体以及小分子Janus激酶(JAK)抑制剂来全身消除助长炎症的细胞因子或细胞应答。与这些治疗方法不同,治疗性疫苗,例如CEL-4000,通过促进抗原特异性调节而不是炎症反应来帮助平衡适应性免疫稳态,从而调节RA的免疫病理过程。在这次审查中,我们讨论了目前和拟议的RA治疗产品,重点是抗原特异性治疗性疫苗方法来治疗该疾病。作为一个例子,我们描述了CEL-4000疫苗对RA动物模型有益作用的已发表结果.我们还建议为这些最新的治疗方法设计适当的临床研究,以CEL-4000疫苗为例。与产生或增强新免疫反应的疫苗不同,用于RA的免疫调节治疗性疫苗的临床成功在于其能够重新定向自身反应性促炎记忆T细胞,以重新平衡表征该疾病的“失控”免疫/炎症反应.这种疗法的人体试验将需要临床试验设计和实施中的替代方法来确定安全性,毒性,和功效。这些方法包括自适应设计(如贝叶斯最优设计(BOIN),目前用于肿瘤临床研究),以及使用疾病相关的生物标志物作为治疗成功的指标。
    Rheumatoid arthritis (RA) is recognized as an autoimmune joint disease driven by T cell responses to self (or modified self or microbial mimic) antigens that trigger and aggravate the inflammatory condition. Newer treatments of RA employ monoclonal antibodies or recombinant receptors against cytokines or immune cell receptors as well as small-molecule Janus kinase (JAK) inhibitors to systemically ablate the cytokine or cellular responses that fuel inflammation. Unlike these treatments, a therapeutic vaccine, such as CEL-4000, helps balance adaptive immune homeostasis by promoting antigen-specific regulatory rather than inflammatory responses, and hence modulates the immunopathological course of RA. In this review, we discuss the current and proposed therapeutic products for RA, with an emphasis on antigen-specific therapeutic vaccine approaches to the treatment of the disease. As an example, we describe published results of the beneficial effects of CEL-4000 vaccine on animal models of RA. We also make a recommendation for the design of appropriate clinical studies for these newest therapeutic approaches, using the CEL-4000 vaccine as an example. Unlike vaccines that create or boost a new immune response, the clinical success of an immunomodulatory therapeutic vaccine for RA lies in its ability to redirect autoreactive pro-inflammatory memory T cells towards rebalancing the \"runaway\" immune/inflammatory responses that characterize the disease. Human trials of such a therapy will require alternative approaches in clinical trial design and implementation for determining safety, toxicity, and efficacy. These approaches include adaptive design (such as the Bayesian optimal design (BOIN), currently employed in oncological clinical studies), and the use of disease-related biomarkers as indicators of treatment success.
    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
    背景:来自肿瘤相关抗原(TAA)的表位肽的鉴定为开发肿瘤特异性免疫疗法提供了信息。然而,在头颈癌(HNSCC)的小鼠TAAs中仅检测到少数表位。
    方法:通过基于计算机的算法预测新型小鼠c-Met衍生的T细胞表位。用c-Met肽疫苗处理携带小鼠HNSCC细胞系的小鼠。CD8和/或CD4T细胞耗竭的影响,并评估了疫苗与免疫检查点抑制剂(ICIs)的组合。进行肿瘤再接种以评估T细胞记忆。
    结果:我们鉴定了c-Met衍生的短表位和长表位,这些表位引起c-Met反应性抗肿瘤CD8和/或CD4T细胞反应。使用这些肽的疫苗通过T细胞显示出显著的抗肿瘤应答,其中不需要ICI。接种c-Met肽的小鼠排斥重新接种的肿瘤。
    结论:我们证明了新型c-Met肽疫苗可以诱导抗肿瘤T细胞反应,在同基因小鼠HNSCC模型中可能是一种有效的免疫疗法。
    BACKGROUND: The identification of epitope peptides from tumor-associated antigens (TAAs) is informative for developing tumor-specific immunotherapy. However, only a few epitopes have been detected in mouse TAAs of head and neck cancer (HNSCC).
    METHODS: Novel mouse c-Met-derived T-cell epitopes were predicted by computer-based algorithms. Mouse HNSCC cell line-bearing mice were treated with a c-Met peptide vaccine. The effects of CD8 and/or CD4 T-cell depletion, and vaccine combination with immune checkpoint inhibitors (ICIs) were evaluated. Tumor re-inoculation was performed to assess T-cell memory.
    RESULTS: We identified c-Met-derived short and long epitopes that elicited c-Met-reactive antitumor CD8 and/or CD4 T-cell responses. Vaccination using these peptides showed remarkable antitumor responses via T cells in which ICIs were not required. The c-Met peptide-vaccinated mice rejected the re-inoculated tumors.
    CONCLUSIONS: We demonstrated that novel c-Met peptide vaccines can induce antitumor T-cell response, and could be a potent immunotherapy in a syngeneic mouse HNSCC model.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号