T cell response

T 细胞反应
  • 文章类型: Journal Article
    弓形虫是一种广泛分布的原生动物寄生虫,约感染世界人口的三分之一,可引起严重的公共卫生问题。在这项研究中,我们研究了减毒疫苗Pru:Δcdpk2对急性弓形虫病的保护作用,并探索了猪保护的潜在免疫机制。系统性T细胞和自然杀伤(NK)细胞反应进行了分析,包括动力学,表型,和多功能性(干扰素[IFN]-γ,肿瘤坏死因子[TNF]-α),并分析PBMC中的IFN-γ水平。我们的结果表明,弓形虫特异性抗体是由Pru:Δcdpk2诱导的。挑战RH后,在免疫组中,抗体能够快速反应,表达水平明显高于未免疫组。接种后IFN-γ的表达水平显著升高,和CD3+γδ-,NK,CD3+γδ+细胞亚群也显著增加。同时,功能分析表明,这些细胞向Th1表型极化,显示分泌IFN-γ和TNF-α的能力。在疫苗接种的早期,与CD4-CD8α和CD4CD8α细胞群体相比,CD4CD8α-T细胞群体表现出更高的IFN-γ产生细胞频率。我们的结果表明,减毒疫苗可以诱导NK细胞的表达,γδ,和猪的CD3αβ细胞,这些细胞分泌的IFN-γ和TNF-α对于抵抗弓形虫感染是重要的。
    Toxoplasma gondii is a widespread protozoan parasite approximately infecting one-third of the world population and can cause serious public health problems. In this study, we investigated the protective effect of the attenuated vaccine Pru:Δcdpk2 against acute toxoplasmosis and explored the underlying immune mechanisms of the protection in pigs. The systemic T-cell and natural killer (NK) cell responses were analyzed, including kinetics, phenotype, and multifunctionality (interferon [IFN]-γ, tumor necrosis factor [TNF]-α), and the IFN-γ levels were analyzed in PBMCs. Our results showed that T. gondii-specific antibodies were induced by Pru:Δcdpk2. After challenging with RH, the antibodies were able to respond quickly in the immunized group, and the expression level was significantly higher than that in the unimmunized group. The expression level of IFN-γ significantly increased after vaccination, and the CD3+ γδ-, NK, and CD3+ γδ+ cell subsets also significantly increased. At the same time, functional analysis indicated that these cells were polarized toward a Th1 phenotype, showing the ability to secrete IFN-γ and TNF-α. The CD4+CD8α-T cell population exhibited a higher frequency of IFN-γ+ producing cells compared with the CD4-CD8α+ and CD4+CD8α+ cell populations during the early days of vaccination. Our results indicated that the attenuated vaccine could induce the expression of NK, γδ, and CD3αβ cells in pigs, and IFN-γ and TNF-α secreted by these cells are important for resistance to T. gondii 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
    背景:精心选择合适的疫苗佐剂对于优化免疫反应至关重要。传统上,寻常型天疱疮(PV),一种自身免疫性疾病,已使用完全弗氏佐剂(CFA)建模。在这项研究中,我们的目的是辨别与CFA相比,Toll样受体(TLR)配体引起的免疫反应的潜在变化.
    方法:进行了全面调查,比较这些佐剂与卵清蛋白或桥粒糖蛋白-3的联合作用。流式细胞术用于分析不同的细胞亚群,而酶联免疫吸附试验定量抗原特异性抗体和细胞因子水平。进行收获的皮肤组织的组织学检查和皮肤损伤的转录组分析以鉴定差异表达的基因。
    结果:TLR配体显示出在野生型小鼠中诱导PV样症状的功效,与CFA相反。这强调了佐剂对自身抗原耐受性的实质性影响。此外,我们提出了一种通过过继转移建立光伏模型的增强方法,用TLR配体取代CFA。我们的结果表明,与CFA是最有效的免疫增强剂的观点相反,CFA促进调节性T细胞(Treg),滤泡调节性T细胞和产生IL-10的中性粒细胞,而TLR配体下调CCL17和IL-10。这表明对Treg亚群的募集和激活的潜在影响。虽然B细胞和CD8+T细胞反应表现出相似性,CFA在树突状细胞亚群中诱导较少的活化。本研究提供了一种新型PV小鼠模型和佐剂免疫刺激作用的系统比较。
    结论:CFA和TLR配体的系统比较揭示了这些佐剂的独特性质,为天疱疮的研究提供创新的小鼠模型。这项研究为辅助研究做出了重要贡献,并促进了我们对PV发病机理的理解。
    用桥粒蛋白3和Toll样受体(TLR)配体免疫可有效诱导野生型小鼠天疱疮症状,而完全弗氏佐剂(CFA)失败。TLR配体在过继转移天疱疮模型中提高了供体细胞的自身反应性。CFA促进调节性T细胞和产生IL-10的中性粒细胞,而TLR配体下调CCL17和IL-10,导致更有效的免疫反应。
    BACKGROUND: The meticulous selection of appropriate vaccine adjuvants is crucial for optimizing immune responses. Traditionally, pemphigus vulgaris (PV), an autoimmune disorder, has been modelled using complete Freund\'s adjuvant (CFA). In this study, we aimed to discern potential variations in immune responses elicited by Toll-like receptor (TLR) ligands as compared to CFA.
    METHODS: A comprehensive investigation was conducted, comparing the effects of these adjuvants in conjunction with ovalbumin or desmoglein-3. Flow cytometry was employed to analyse distinct cell subsets, while enzyme-linked immunosorbent assay quantified antigen-specific antibodies and cytokine levels. Histological examination of harvested skin tissues and transcriptome analysis of skin lesions were performed to identify differentially expressed genes.
    RESULTS: TLR ligands demonstrated efficacy in inducing PV-like symptoms in wild-type mice, in contrast to CFA. This underscored the substantial impact of the adjuvant on self-antigen tolerance. Furthermore, we proposed an enhanced method for establishing a PV model through adoptive transfer, substituting CFA with TLR ligands. Our results revealed that in contrast to the perception that CFA being the most potent immunopotentiator reported, CFA promoted regulatory T cells (Treg), follicular regulatory T cells and IL-10-producing neutrophils, whereas TLR ligands downregulated CCL17 and IL-10. This suggested potential implications for the recruitment and activation of Treg subsets. While B cell and CD8+ T cell responses exhibited similarity, CFA induced less activation in dendritic cell subsets. A novel mouse model of PV and systemic comparison of immunostimulatory effects of adjuvants were provided by this study.
    CONCLUSIONS: The systematic comparison of CFA and TLR ligands shed light on the distinctive properties of these adjuvants, presenting innovative mouse models for the investigation of pemphigus. This study significantly contributes to adjuvant research and advances our understanding of PV pathogenesis.
    UNASSIGNED: Immunization with desmoglein 3 and Toll-like receptor (TLR) ligands effectively induces pemphigus symptoms in wild-type mice, whereas complete Freund\'s adjuvant (CFA) fails. TLR ligands heightened the autoreactivity of donor cells in the adoptive transfer pemphigus model. CFA promoted regulatory T cells and IL-10-producing neutrophils, whereas TLR ligands downregulated CCL17 and IL-10, leading to more effective 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
    选择合适的佐剂对于开发有效的疫苗至关重要。然而,关于COVID-19灭活疫苗中不同佐剂引发的免疫反应的研究很少。在这里,我们评估了明矾的疗效,CpGHP021,明矾与CpGHP021(明矾/CpG)结合,或MF-59佐剂与COVID-19灭活疫苗在K18-hACE2小鼠中,并比较了K18-hACE2和BALB/c小鼠的不同免疫反应。在K18-hACE2小鼠中,与明矾组相比,明矾/CpG组抗受体结合域(RBD)IgG抗体滴度增加7.5倍,即使抗原量减少了三分之二,也产生了相当水平的抗体,可能是由于脾中央区域生发中心(GC)结构的显着激活。不同的佐剂诱导多种结合抗体同种型。CpGHP021和明矾/CpG偏向Th1/IgG2a,明矾和MF-59偏向于Th2/IgG1。细胞因子IFN-γ,在明矾/CpG组中特异性刺激的脾细胞的培养上清液中,IL-2和TNF-α显着增加。BALB/c小鼠的抗体反应与K18-hACE2小鼠相似,但中和抗体(NAb)水平较低。值得注意的是,Alum/CpG佐剂灭活疫苗诱导更多的T细胞分泌IFN-γ和IL-2,增加CD8+T细胞中效应记忆T(TEM)细胞的百分比,并有效保护K18-hACE2小鼠免受Delta变体攻击。我们的结果表明,明矾/CpG复合佐剂显着增强了对灭活的COVID-19抗原的免疫反应,并可以诱导持久的免疫反应。
    Selecting appropriate adjuvants is crucial for developing an effective vaccine. However, studies on the immune responses triggered by different adjuvants in COVID-19 inactivated vaccines are scarce. Herein, we evaluated the efficacy of Alum, CpG HP021, Alum combined with CpG HP021 (Alum/CpG), or MF-59 adjuvants with COVID-19 inactivated vaccines in K18-hACE2 mice, and compared the different immune responses between K18-hACE2 and BALB/c mice. In K18-hACE2 mice, the Alum/CpG group produced a 6.5-fold increase in anti-receptor-binding domain (RBD) IgG antibody titers compared to the Alum group, and generated a comparable level of antibodies even when the antigen amount was reduced by two-thirds, possibly due to the significant activation of germinal center (GC) structures in the central region of the spleen. Different adjuvants induced a variety of binding antibody isotypes. CpG HP021 and Alum/CpG were biased towards Th1/IgG2c, while Alum and MF-59 were biased toward Th2/IgG1. Cytokines IFN-γ, IL-2, and TNF-α were significantly increased in the culture supernatants of splenocytes specifically stimulated in the Alum/CpG group. The antibody responses in BALB/c mice were similar to those in K18-hACE2 mice, but with lower levels of neutralizing antibodies (NAbs). Notably, the Alum/CpG-adjuvanted inactivated vaccine induced a higher number of T cells secreting IFN-γ and IL-2, increased the percentage of effector memory T (TEM) cells among CD8+ T cells, and effectively protected K18-hACE2 mice from Delta variant challenge. Our results showed that Alum/CpG complex adjuvant significantly enhanced the immune response to inactivated COVID-19 antigens and could induce a long-lasting immune response.
    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: Video-Audio Media
    背景:肠道菌群是调节宿主健康的关键因素,但是宿主对病原体的抗性差异与肠道微生物相互作用之间的关系尚不清楚。在这里,我们使用单细胞转录组学研究了仔猪肠道菌群与其抗病性之间的潜在相关性,16S扩增子测序,宏基因组学,和非靶向代谢组学。
    结果:猪流行性腹泻病毒(PEDV)感染导致仔猪肠道菌群发生显著变化。值得注意的是,长白猪感染PEDV后很快失去抵抗力,但是将民猪的粪便微生物群移植到长白猪可以减轻感染状况。宏观基因组和动物保护模型确定肠微生物群中的罗伊氏乳杆菌和淀粉乳杆菌发挥抗感染作用。此外,次级胆汁酸(DCA)和石胆酸(LCA)的代谢组学筛选与罗伊氏乳杆菌和淀粉乳杆菌显着相关,但在动物模型中只有LCA发挥保护作用。此外,LCA补充改变了肠道T细胞群的分布,并导致显著富集的CD8+CTL,体内和体外实验表明,LCA通过FXR受体增加猪肠上皮细胞中SLA-I的表达,从而招募CD8+CTL发挥抗病毒作用。
    结论:总体而言,我们的研究结果表明,肠道微生物群的多样性影响疾病的发展,操纵罗伊乳杆菌和淀粉乳杆菌,以及LCA,代表了改善仔猪PEDV感染的有希望的策略。视频摘要。
    BACKGROUND: The gut microbiota is a critical factor in the regulation of host health, but the relationship between the differential resistance of hosts to pathogens and the interaction of gut microbes is not yet clear. Herein, we investigated the potential correlation between the gut microbiota of piglets and their disease resistance using single-cell transcriptomics, 16S amplicon sequencing, metagenomics, and untargeted metabolomics.
    RESULTS: Porcine epidemic diarrhea virus (PEDV) infection leads to significant changes in the gut microbiota of piglets. Notably, Landrace pigs lose their resistance quickly after being infected with PEDV, but transplanting the fecal microbiota of Min pigs to Landrace pigs alleviated the infection status. Macrogenomic and animal protection models identified Lactobacillus reuteri and Lactobacillus amylovorus in the gut microbiota as playing an anti-infective role. Moreover, metabolomic screening of the secondary bile acids\' deoxycholic acid (DCA) and lithocholic acid (LCA) correlated significantly with Lactobacillus reuteri and Lactobacillus amylovorus, but only LCA exerted a protective function in the animal model. In addition, LCA supplementation altered the distribution of intestinal T-cell populations and resulted in significantly enriched CD8+ CTLs, and in vivo and in vitro experiments showed that LCA increased SLA-I expression in porcine intestinal epithelial cells via FXR receptors, thereby recruiting CD8+ CTLs to exert antiviral effects.
    CONCLUSIONS: Overall, our findings indicate that the diversity of gut microbiota influences the development of the disease, and manipulating Lactobacillus reuteri and Lactobacillus amylovorus, as well as LCA, represents a promising strategy to improve PEDV infection in piglets. Video Abstract.
    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
    有效的免疫应答依赖于CD8+T细胞正确分化为效应细胞和记忆细胞。这里,我们显示了急性病毒感染时CD8+T细胞应答期间N6-甲基腺苷(m6A)甲基转移酶Mettl3的关键需求。CD8+T细胞中Mettl3的条件缺失以m6A依赖性方式损害效应子扩增和终末分化,随后影响记忆形成和CD8+T细胞的次级反应。我们的组合RNA-seq和m6A-miCLIP-seq分析显示Mettl3缺陷广泛影响细胞周期和转录调节因子的表达。值得注意的是,Mettl3与Tbx21转录物结合并使其稳定,促进CD8+T细胞的效应子分化。此外,在没有Mettl3的情况下,T-bet的异位表达部分恢复了CD8+T细胞分化的缺陷。因此,我们的研究强调了Mettl3在以m6A依赖性方式调节多个靶基因中的作用,并强调了在CD8+T细胞应答过程中m6A修饰的重要性.
    Efficient immune responses rely on the proper differentiation of CD8+ T cells into effector and memory cells. Here, we show a critical requirement of N6-Methyladenosine (m6A) methyltransferase Mettl3 during CD8+ T cell responses upon acute viral infection. Conditional deletion of Mettl3 in CD8+ T cells impairs effector expansion and terminal differentiation in an m6A-dependent manner, subsequently affecting memory formation and the secondary response of CD8+ T cells. Our combined RNA-seq and m6A-miCLIP-seq analyses reveal that Mettl3 deficiency broadly impacts the expression of cell cycle and transcriptional regulators. Remarkably, Mettl3 binds to the Tbx21 transcript and stabilizes it, promoting effector differentiation of CD8+ T cells. Moreover, ectopic expression of T-bet partially restores the defects in CD8+ T cell differentiation in the absence of Mettl3. Thus, our study highlights the role of Mettl3 in regulating multiple target genes in an m6A-dependent manner and underscores the importance of m6A modification during CD8+ T cell response.
    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
    在SARS-CoV-2灭活疫苗的所有成分中,核衣壳蛋白(N)是最丰富和高度保守的蛋白质。然而,N在这些疫苗中的功能,尤其是它对靶向刺突蛋白反应的影响,仍然未知。在这项研究中,用单独的N蛋白免疫小鼠被证明可以减少病毒载量,减轻SARS-CoV-2病毒攻击后的肺部病理损害。此外,发现用N的共免疫和预免疫诱导更高的S-特异性抗体滴度而不是损害它们。值得注意的是,当在全灭活病毒疫苗接种后N作为加强剂量施用时,也观察到相同的趋势.在所有组中都检测到N特异性IFN-γ分泌T细胞反应,并表现出与S特异性IgG抗体改善的一定关系。一起,这些数据表明,N在疫苗诱导的保护中具有独立的作用,并改善了对灭活疫苗的S特异性抗体应答,揭示了在对复杂病毒成分的免疫反应中可能存在相互作用机制。
    Of all of the components in SARS-CoV-2 inactivated vaccines, nucleocapsid protein (N) is the most abundant and highly conserved protein. However, the function of N in these vaccines, especially its influence on the targeted spike protein\'s response, remains unknown. In this study, the immunization of mice with the N protein alone was shown to reduce the viral load, alleviating pulmonary pathological lesions after challenge with the SARS-CoV-2 virus. In addition, co-immunization and pre-immunization with N were found to induce higher S-specific antibody titers rather than compromise them. Remarkably, the same trend was also observed when N was administered as the booster dose after whole inactivated virus vaccination. N-specific IFN-γ-secreting T cell response was detected in all groups and exhibited a certain relationship with S-specific IgG antibody improvements. Together, these data indicate that N has an independent role in vaccine-induced protection and improves the S-specific antibody response to inactivated vaccines, revealing that an interplay mechanism may exist in the immune responses to complex virus components.
    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:了解肠道菌群与中药对中枢神经系统(CNS)自身免疫益处之间的机制,我们研究了Heshouwu()对实验性自身免疫性脑炎(EAE)的神经保护作用和肠道微生物群的变化,多发性硬化症(MS)的动物模型。
    方法:小鼠随机分为四组:EAE小鼠(对照磷酸盐缓冲盐水组),50mg·kg·d合首乌()处理的EAE小鼠,100mg·kg·d合首乌()处理的EAE小鼠,和200mg·kg·d的Heshouwu()处理的EAE小鼠。脊髓用苏木精和伊红(HE)和luxol固蓝染色,以评估炎症浸润和脱髓鞘。粒细胞巨噬细胞集落刺激因子(GM-CSF)+CD4+的百分比,白细胞介素17(IL-17)+CD4+,Foxp3CD4+,通过流式细胞术分析确定腹股沟淋巴结(LN)和大脑中的干扰素-γ(IFN-γ)CD4T细胞。16SrRNA基因测序用于分析肠道微生物群的变化。
    结果:我们发现Heshouwu()减轻了通过临床和组织病理学评分评估的EAE的疾病严重程度和神经病理学。Heshouwu()增加了肠道微生物群的多样性和丰度,和降低/比率(F/B比率)。Heshouwu()还降低了IL-10和IL-21的浓度,并增加了GM-CSF的水平,IL-17A,EAE小鼠血清中的IL-17F和IL-22。此外,Heshouwu()通过抑制Th17细胞和恢复小肠淋巴组织和腹股沟淋巴结中的Treg细胞来调节T细胞反应。接受Heshouwu()处理的粪便微生物群移植的微生物群耗尽小鼠的疾病严重程度较低,与随意组相比,神经病理学评分和Th17/Treg失衡的缓解。
    结论:我们的发现表明,何首乌(TCM)在免疫调节作用中的重要神经保护作用部分是通过肠道微生物组的调节。
    To learn the mechanisms between gut microbiome and the autoimmunity benefits on Traditional Chinese Medicine (TCM) in central nervous system (CNS), we investigated the neuro-protection effects and gut mircobiota changes of Heshouwu () on experimental autoimmune encepha-lomyelitis (EAE), an animal model of multiple sclerosis (MS).
    Mice were randomly divided into four groups: EAE mice (control phosphate-buffered saline group), 50 mg·kg·d Heshouwu ()-treated EAE mice, 100 mg·kg·d Heshouwu ()-treated EAE mice, and 200 mg·kg·d Heshouwu ()-treated EAE mice. The spinal cords were stained with hematoxylin and eosin (HE) and luxol fast blue for evaluating inflammatory infiltration and demyelination. The percentages of granulocyte macrophage-colony stimulating factor (GM-CSF)+CD4+, interleukin 17 (IL-17)+CD4+, Foxp3 CD4+, and interferon-γ (IFN-γ)+CD4+ T cells in the inguinal lymph nodes (LNs) and brain were determined by flow cytometry analysis. 16S rRNA gene sequencing was employed to analyze the changes in gut microbiota.
    We found that Heshouwu () alleviated the disease severity and neuropathology of EAE as evaluated by clinical and histopathologyical scores. Heshouwu () increased the diversity and abundance of the gut microbiota, and decreased / ratio (F/B ratio). Heshouwu () also decreased the concentrations of IL-10, and IL-21 and increase the levels of GM-CSF, IL-17A, IL-17F and IL-22 in serum of EAE mice. Moreover, Heshouwu () modulated the T cell responses by inhibiting Th17 cells and restoring Treg cells in the small intestine lymphoid tissues and inguinal lymph nodes. Microbiota-depleted mice receiving Heshouwu ()-treated fecal microbiota trans-plantation had lower disease severity, neuropathology scores and alleviation of Th17/Treg imbalance compared to ad libitum group.
    Our findings suggested that the vital neuro-protection role of Heshouwu () (TCM) in immunomodulation effects partly by regulations of gut microbiome.
    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)大流行的一个关键问题是初次感染后针对严重急性呼吸道综合症冠状病毒2(SARS-CoV-2)的特定T细胞反应的持续时间,由于大规模的COVID-19疫苗接种和再次暴露于该病毒,这一问题很难解决。这里,我们对一组恢复期个体(CIs)的长期SARS-CoV-2特异性T细胞应答进行了分析,这些个体是全球首批感染的个体,此后没有任何可能的抗原再暴露.SARS-CoV-2特异性T细胞反应的大小和广度与疾病发作所经过的时间和这些CI的年龄成反比。SARS-CoV-2特异性CD4和CD8T细胞反应的平均幅度降低了约82%和76%,分别,在感染后十个月的时间内。因此,纵向分析还表明,在随访期间,在75%的CI中,SARS-CoV-2特异性T细胞应答显著减弱.总的来说,我们提供了CI中长期记忆T细胞反应的全面表征,提示初次感染后强大的SARS-CoV-2特异性T细胞免疫可能不如先前预期的持久。
    A key question in the coronavirus disease 2019 (COVID-19) pandemic is the duration of specific T cell responses against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) post primary infection, which is difficult to address due to the large-scale COVID-19 vaccination and re-exposure to the virus. Here, we conducted an analysis of the long-term SARS-CoV-2-specific T cell responses in a unique cohort of convalescent individuals (CIs) that were among the first to be infected worldwide and without any possible antigen re-exposure since then. The magnitude and breadth of SARS-CoV-2-specific T cell responses correlated inversely with the time that had elapsed from disease onset and the age of those CIs. The mean magnitude of SARS-CoV-2-specific CD4 and CD8 T cell responses decreased about 82% and 76%, respectively, over the time period of ten months after infection. Accordingly, the longitudinal analysis also demonstrated that SARS-CoV-2-specific T cell responses waned significantly in 75% of CIs during the follow-up. Collectively, we provide a comprehensive characterization of the long-term memory T cell response in CIs, suggesting that robust SARS-CoV-2-specific T cell immunity post primary infection may be less durable than previously expected.
    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
    在过去的几年里,由SARS-CoV-2引起的COVID-19的持续流行给公共卫生带来了巨大的负担。为了有效应对SARS-CoV-2新变种的出现,进一步增强完成第一代疫苗接种的个体的免疫反应变得有意义。为了了解使用基于不同变体序列的灭活疫苗的顺序给药是否可以诱导针对即将到来的变体的更好的免疫力,我们在小鼠模型中尝试了5种灭活疫苗组合,并比较了它们的免疫反应.我们的结果表明,通过在免疫的早期阶段诱导强大的抗原特异性T细胞免疫反应,序贯策略比同源免疫具有显着的优势。此外,我们研究中的三剂量疫苗接种策略引发了针对BA.2Omicron菌株的更好的中和抗体反应.这些数据为在现有疫苗平台内寻找产生针对多种变体(包括先前未暴露的菌株)的交叉免疫的最佳策略提供了科学线索。
    In the past few years, the continuous pandemic of COVID-19 caused by SARS-CoV-2 has placed a huge burden on public health. In order to effectively deal with the emergence of new SARS-CoV-2 variants, it becomes meaningful to further enhance the immune responses of individuals who have completed the first-generation vaccination. To understand whether sequential administration using different variant sequence-based inactivated vaccines could induce better immunity against the forthcoming variants, we tried five inactivated vaccine combinations in a mouse model and compared their immune responses. Our results showed that the sequential strategies have a significant advantage over homologous immunization by inducing robust antigen-specific T cell immune responses in the early stages of immunization. Furthermore, the three-dose vaccination strategies in our research elicited better neutralizing antibody responses against the BA.2 Omicron strain. These data provide scientific clues for finding the optimal strategy within the existing vaccine platform in generating cross-immunity against multiple variants including previously unexposed strains.
    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
    原理:对5-氟尿嘧啶(5-FU)化疗的耐药性仍然是结直肠癌(CRC)患者有效临床结局的主要障碍。需要更好地理解5-FU抗性的详细机制以增加存活率。白细胞介素(IL)-33是一种新发现的alarmin样分子,在各种癌症中发挥促和抗肿瘤作用。然而,IL-33在CRC进展中的确切作用,以及在5-FU抗性的发展中,尚不清楚。方法:对来自5-FU敏感和5-FU耐药CRC患者的匹配样本进行高质量RNA测序分析。IL-33的临床和生物学意义,包括其对T细胞和肿瘤细胞的影响,以及它与5-FU化疗活性的关系在离体检查,CRC的体外和体内模型。探索了这些过程的分子机制。结果:在5-FU敏感的CRC患者中,肿瘤细胞表达的IL-33是抗肿瘤免疫的主要介质。通过结合它的ST2受体,IL-33通过激活膜联蛋白A1下游信号传导级联来触发CD4+(Th1和Th2)和CD8+T细胞应答。机械上,IL-33仅在T细胞存在下增强CRC细胞对5-FU的敏感性,这导致肿瘤细胞固有的凋亡和免疫杀伤相关信号的激活,从而与5-FU协同诱导CRC细胞凋亡。此外,损伤的CRC细胞释放更多的IL-33和T细胞趋化因子CXCL10和CXCL13,形成正反馈回路以进一步增强T细胞应答。结论:我们的结果确定了IL-33与对5-FU的敏感性增强之间的先前未识别的联系。IL-33通过协调抗肿瘤T细胞反应创造了免疫活性肿瘤微环境。因此,IL-33是5-FU化学敏感性和良好预后的潜在预测生物标志物,并且具有作为有希望的辅助免疫疗法的潜力,以改善基于5-FU的疗法在CRC治疗中的临床益处。
    Rationale: Resistance to 5-fluorouracil (5-FU) chemotherapy remains the main barrier to effective clinical outcomes for patients with colorectal cancer (CRC). A better understanding of the detailed mechanisms underlying 5-FU resistance is needed to increase survival. Interleukin (IL)-33 is a newly discovered alarmin-like molecule that exerts pro- and anti-tumorigenic effects in various cancers. However, the precise role of IL-33 in CRC progression, as well as in the development of 5-FU resistance, remains unclear. Methods: High-quality RNA-sequencing analyses were performed on matched samples from patients with 5-FU-sensitive and 5-FU-resistant CRC. The clinical and biological significance of IL-33, including its effects on both T cells and tumor cells, as well as its relationship with 5-FU chemotherapeutic activity were examined in ex vivo, in vitro and in vivo models of CRC. The molecular mechanisms underlying these processes were explored. Results: IL-33 expressed by tumor cells was a dominant mediator of antitumoral immunity in 5-FU-sensitive patients with CRC. By binding to its ST2 receptor, IL-33 triggered CD4+ (Th1 and Th2) and CD8+ T cell responses by activating annexin A1 downstream signaling cascades. Mechanistically, IL-33 enhanced the sensitivity of CRC cells to 5-FU only in the presence of T cells, which led to the activation of both tumor cell-intrinsic apoptotic and immune killing-related signals, thereby synergizing with 5-FU to induce apoptosis of CRC cells. Moreover, injured CRC cells released more IL-33 and the T cell chemokines CXCL10 and CXCL13, forming a positive feedback loop to further augment T cell responses. Conclusions: Our results identified a previously unrecognized connection between IL-33 and enhanced sensitivity to 5-FU. IL-33 created an immune-active tumor microenvironment by orchestrating antitumoral T cell responses. Thus, IL-33 is a potential predictive biomarker for 5-FU chemosensitivity and favorable prognosis and has potential as a promising adjuvant immunotherapy to improve the clinical benefits of 5-FU-based therapies in the treatment of CRC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号