Immunologic Memory

免疫记忆
  • 文章类型: Journal Article
    靶向细胞内抑制蛋白已被揭示为提高CD8+T细胞抗肿瘤功效的有希望的策略。这里,我们专注于对TCR信号特异性的细胞内抑制蛋白:在T细胞中表达的DOK1和DOK2。我们假设细胞内抑制检查点DOK1和DOK2的消耗可以改善基于CD8+T细胞的癌症治疗。评估DOK1和DOK2耗竭在CD8+T淋巴细胞的生理和效应子功能以及在癌症进展中的作用,我们在WT和Dok1/Dok2DKO(双KO)小鼠中建立了对黑色素瘤抗原hgp100(pmel-1TCRTg)特异性的转基因T细胞受体小鼠模型。我们表明,体外预刺激后,CD8+T细胞中的DOK1和DOK2耗竭诱导了更高百分比的效应记忆T细胞,以及CD3mAb诱导的TCR信号传导case的上调,包括pAKT和pERK水平的增加,参与T细胞功能的两种主要磷蛋白。有趣的是,在初始CD8+T细胞中未观察到这种改善的TCR信号传导。尽管这种增强的TCR信号基本上显示在通过CD3mAb刺激后,预先刺激的Dok1/Dok2DKOCD8+T细胞在体外对表达hgp100的黑素瘤细胞系的活化或细胞毒性能力没有任何增加。总之,我们在这里证明了CD8+T细胞中DOK1和DOK2蛋白负调节的新方面。的确,我们的结果使我们能够得出结论,DOK1和DOK2在长期T细胞刺激后具有抑制作用.
    Targeting intracellular inhibiting proteins has been revealed to be a promising strategy to improve CD8+ T cell anti-tumor efficacy. Here, we are focusing on intracellular inhibiting proteins specific to TCR signaling: DOK1 and DOK2 expressed in T cells. We hypothesized that depletion of intracellular inhibition checkpoint DOK1 and DOK2 could improve CD8+ T-cell based cancer therapies. To evaluate the role of DOK1 and DOK2 depletion in physiology and effector function of CD8+ T lymphocytes and in cancer progression, we established a transgenic T cell receptor mouse model specific to melanoma antigen hgp100 (pmel-1 TCR Tg) in WT and Dok1/Dok2 DKO (double KO) mice. We showed that both DOK1 and DOK2 depletion in CD8+ T cells after an in vitro pre-stimulation induced a higher percentage of effector memory T cells as well as an up regulation of TCR signaling cascade- induced by CD3 mAbs, including the increased levels of pAKT and pERK, two major phosphoproteins involved in T cell functions. Interestingly, this improved TCR signaling was not observed in naïve CD8+ T cells. Despite this enhanced TCR signaling essentially shown upon stimulation via CD3 mAbs, pre-stimulated Dok1/Dok2 DKO CD8+ T cells did not show any increase in their activation or cytotoxic capacities against melanoma cell line expressing hgp100 in vitro. Altogether we demonstrate here a novel aspect of the negative regulation by DOK1 and DOK2 proteins in CD8+ T cells. Indeed, our results allow us to conclude that DOK1 and DOK2 have an inhibitory role following long term T cell stimulations.
    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:组织内记忆CD103+CD8+T细胞(CD103+CD8+TRMs)是抗肿瘤免疫的重要组成部分。然而,CD103+CD8+TRMs在结直肠癌(CRC)中的意义及其优势尚不清楚.
    方法:使用临床数据和标本评估CD103+CD8+TRMs在CRC中的意义。通过小鼠皮下肿瘤发生模型和集落形成实验来评估CD103CD8TRMs的抗肿瘤作用。最后,使用流式细胞术评估肿瘤中CD103CD8TRMs的浸润密度和功能。
    结果:在这项研究中,我们发现,在CRC患者中,高度浸润的CD103+CD8+TRMs与早期临床分期和VEGF阴性表达相关,并预测CRC/CRC肝转移患者的预后良好.有趣的是,我们还发现,CD103+CD8+TRMs可能对CRC中是否发生肝转移具有预测潜力.此外,我们发现CRC中α-SMA+血管数与α-SMA+和CD31+血管数之和呈正相关,和CD103+CD8+TRMs的浸润水平。此外,抗血管生成治疗促进CD103+CD8+TRMs的浸润并增强其分泌干扰素(IFN)-γ的能力,从而进一步提高抗肿瘤效果。此外,体内实验表明,与外周血CD8+T细胞相比,CD103+CD8+TRMs输注回体内还可进一步促进CD8+T细胞浸润肿瘤,它们分泌IFN-γ的能力更强,从而产生更好的抗肿瘤效果。
    结论:我们证明了CD103+CD8+TRMs具有临床应用的潜力,并为联合抗肿瘤治疗策略提供了新思路。如抗肿瘤血管生成治疗和CAR-T联合免疫疗法。
    BACKGROUND: Tissue-resident memory CD103+CD8+ T cells (CD103+CD8+ TRMs) are important components of anti-tumor immunity. However, the significance of CD103+CD8+ TRMs in colorectal cancer (CRC) and their advantages remain unclear.
    METHODS: Clinical data and specimens were used to evaluate the significance of CD103+CD8+ TRMs in CRC. A mouse subcutaneous tumorigenesis model and colony-formation assay were conducted to evaluate the anti-tumor effects of CD103+CD8+ TRMs. Finally, the infiltration density and function of CD103+CD8+ TRMs in the tumors were evaluated using flow cytometry.
    RESULTS: In this study, we showed that highly infiltrated CD103+CD8+ TRMs were associated with earlier clinical stage and negative VEGF expression in CRC patients and predicted a favorable prognosis for CRC/CRC liver metastases patients. Interestingly, we also found that CD103+CD8+ TRMs may have predictive potential for whether CRC develops liver metastasis in CRC. In addition, we found a positive correlation between the ratio of the number of α-SMA+ vessels to the sum of the number of α-SMA+ and CD31+ vessels in CRC, and the infiltration level of CD103+CD8+ TRMs. In addition, anti-angiogenic therapy promoted infiltration of CD103+CD8+ TRMs and enhanced their ability to secrete interferon (IFN)-γ, thus further improving the anti-tumor effect. Moreover, in vivo experiments showed that compared with peripheral blood CD8+ T cells, CD103+CD8+ TRMs infused back into the body could also further promote CD8+ T cells to infiltrate the tumor, and they had a stronger ability to secrete IFN-γ, which resulted in better anti-tumor effects.
    CONCLUSIONS: We demonstrated that CD103+CD8+ TRMs have the potential for clinical applications and provide new ideas for combined anti-tumor therapeutic strategies, such as anti-tumor angiogenesis therapy and CAR-T combined immunotherapy.
    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
    滤泡辅助性T细胞对于在原发性病毒感染期间帮助B细胞的成熟和中和抗体(NAb)的产生是必不可少的。然而,他们在召回响应中的作用尚不清楚。这里,我们使用人类丙型肝炎病毒(HCV)再感染作为模型来研究循环CD4T滤泡辅助细胞(cTfh)和记忆B细胞(MBC)之间的回忆协作相互作用,从而导致NAb的产生.
    我们纵向评估了在随后的再感染事件中自发解决原发性HCV感染的受试者的这种相互作用,该事件导致另一次自发解决(SR/SR,n=14)或慢性感染(SR/CI,n=8)。
    两组都表现出病毒特异性记忆T细胞在再感染时扩增。然而,激活的cTfh(CD4+CXCR5+PD-1+ICOS+FoxP3-)的早期扩增仅发生在SR/SR中。cTfh的激活频率与感染后的时间呈负相关。同时,NAb和HCV特异性MBCs(CD19CD27IgM-E2-Tet)在SR/SR的急性期早期达到峰值,但在SR/CI中未达到峰值。最后,激活的cTfh1(CXCR3CCR6-)子集的频率与NAb的中和宽度和效力相关。
    这些结果强调了早期激活cTfh1细胞在帮助抗原特异性B细胞产生介导HCV再感染清除的NAb中的关键作用。
    UNASSIGNED: Follicular helper T cells are essential for helping in the maturation of B cells and the production of neutralizing antibodies (NAbs) during primary viral infections. However, their role during recall responses is unclear. Here, we used hepatitis C virus (HCV) reinfection in humans as a model to study the recall collaborative interaction between circulating CD4 T follicular helper cells (cTfh) and memory B cells (MBCs) leading to the generation of NAbs.
    UNASSIGNED: We evaluated this interaction longitudinally in subjects who have spontaneously resolved primary HCV infection during a subsequent reinfection episode that resulted in either another spontaneous resolution (SR/SR, n = 14) or chronic infection (SR/CI, n = 8).
    UNASSIGNED: Both groups exhibited virus-specific memory T cells that expanded upon reinfection. However, early expansion of activated cTfh (CD4+CXCR5+PD-1+ICOS+FoxP3-) occurred in SR/SR only. The frequency of activated cTfh negatively correlated with time post-infection. Concomitantly, NAbs and HCV-specific MBCs (CD19+CD27+IgM-E2-Tet+) peaked during the early acute phase in SR/SR but not in SR/CI. Finally, the frequency of the activated cTfh1 (CXCR3+CCR6-) subset correlated with the neutralization breadth and potency of NAbs.
    UNASSIGNED: These results underscore a key role for early activation of cTfh1 cells in helping antigen-specific B cells to produce NAbs that mediate the clearance of HCV reinfection.
    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
    疟疾流行地区的儿童可以在短时间内反复感染疟原虫。再感染对多个共存的CD4+T细胞亚群的影响仍未解决。这里,我们检查了在小鼠再感染过程中经历抗原的CD4+T细胞,使用scRNA-seq/TCR-seq和空间转录组学。TCR转基因TEM细胞在增殖之前启动快速的Th1/Tr1回忆反应,虽然GCTfh同行是耐火的,TCM/Tfh样细胞表现出适度的非增殖反应。Th1召回是主要Th1响应的部分传真,没有上调的效应子相关基因是唯一的回忆。多克隆,TCR多样化,CD4+T细胞表现出相似的回忆动态,单个克隆产生多种效应物,包括高度增殖的Th1/Tr1细胞,以及缺乏增殖能力的GCTfh和Tfh样细胞。因此,我们显示了由脾脏中多个共存的CD4+T细胞亚群安装的回忆反应的实质性多样性,并提供图形用户界面,用于研究再感染期间的基因表达动力学和克隆关系。
    Children in malaria-endemic regions can experience repeated Plasmodium infections over short periods of time. Effects of re-infection on multiple co-existing CD4+ T cell subsets remain unresolved. Here, we examine antigen-experienced CD4+ T cells during re-infection in mice, using scRNA-seq/TCR-seq and spatial transcriptomics. TCR transgenic TEM cells initiate rapid Th1/Tr1 recall responses prior to proliferating, while GC Tfh counterparts are refractory, with TCM/Tfh-like cells exhibiting modest non-proliferative responses. Th1-recall is a partial facsimile of primary Th1-responses, with no upregulated effector-associated genes being unique to recall. Polyclonal, TCR-diverse, CD4+ T cells exhibit similar recall dynamics, with individual clones giving rise to multiple effectors including highly proliferative Th1/Tr1 cells, as well as GC Tfh and Tfh-like cells lacking proliferative capacity. Thus, we show substantial diversity in recall responses mounted by multiple co-existing CD4+ T cell subsets in the spleen, and present graphical user interfaces for studying gene expression dynamics and clonal relationships during re-infection.
    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
    区域免疫监视依赖于不同记忆T细胞群体的共同努力。其中,组织驻留记忆T(TRM)细胞被策略性地定位在屏障组织中,在那里他们能够有效的前线防御感染和癌症。然而,这些细胞的长期持久性与多种免疫介导的病理有关。因此,调节TRM细胞群体代表了针对基于组织的疾病的新型疫苗接种和治疗性干预的有吸引力的策略。这里,我们提供了跨组织和疾病状态的TRM细胞异质性和功能的最新概述.我们讨论了TRM细胞介导的免疫保护机制及其对自身免疫性疾病的潜在贡献。最后,我们研究了TRM细胞反应如何持久增强或抑制治疗增益。
    Regionalized immune surveillance relies on the concerted efforts of diverse memory T cell populations. Of these, tissue-resident memory T (TRM) cells are strategically positioned in barrier tissues, where they enable efficient frontline defense against infections and cancer. However, the long-term persistence of these cells has been implicated in a variety of immune-mediated pathologies. Consequently, modulating TRM cell populations represents an attractive strategy for novel vaccination and therapeutic interventions against tissue-based diseases. Here, we provide an updated overview of TRM cell heterogeneity and function across tissues and disease states. We discuss mechanisms of TRM cell-mediated immune protection and their potential contributions to autoimmune disorders. Finally, we examine how TRM cell responses might be durably boosted or dampened for therapeutic gain.
    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
    细胞因子白细胞介素12(IL-12)激活自然杀伤(NK)细胞,IL-15和IL-18诱导它们分化成记忆样(ML)NK细胞;然而,潜在的表观遗传和转录机制尚不清楚.通过结合ATAC-seq,CITE-seq,和功能分析,我们发现IL-12/15/18激活导致两种主要的人类NK细胞:重编程为富集的记忆样(eML)NK细胞或引发效应常规NK(effcNK)细胞。EMLNK细胞具有不同的转录和表观遗传谱,并且功能增强,而effcNK细胞类似于细胞因子引发的cNK细胞。还鉴定了两个转录上离散的eMLNK细胞亚群,eML-1和eML-2,主要来自CD56bright或CD56dim成熟NK细胞亚群,分别。此外,这些eML亚群在IL-12/15/18激活的NK细胞转移至癌症患者后数周出现.我们的发现表明,IL-12/15/18的NK细胞活化导致以前未被重视的不同细胞命运,并确定了增强NK治疗的新策略。
    Activation of natural killer (NK) cells with the cytokines interleukin-12 (IL-12), IL-15, and IL-18 induces their differentiation into memory-like (ML) NK cells; however, the underlying epigenetic and transcriptional mechanisms are unclear. By combining ATAC-seq, CITE-seq, and functional analyses, we discovered that IL-12/15/18 activation results in two main human NK fates: reprogramming into enriched memory-like (eML) NK cells or priming into effector conventional NK (effcNK) cells. eML NK cells had distinct transcriptional and epigenetic profiles and enhanced function, whereas effcNK cells resembled cytokine-primed cNK cells. Two transcriptionally discrete subsets of eML NK cells were also identified, eML-1 and eML-2, primarily arising from CD56bright or CD56dim mature NK cell subsets, respectively. Furthermore, these eML subsets were evident weeks after transfer of IL-12/15/18-activated NK cells into patients with cancer. Our findings demonstrate that NK cell activation with IL-12/15/18 results in previously unappreciated diverse cellular fates and identifies new strategies to enhance NK therapies.
    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
    KLRG1+CD8T细胞在急性感染清除后持续数月,并维持高水平的效应分子,提供针对全身病原体的保护性免疫。继发感染时,这些长寿效应细胞(LLEC)不能形成其他循环的KLRG1-记忆亚群,如中枢和效应记忆T细胞.因此,KLRG1+记忆T细胞通常被称为寿命相对较短的终末分化群体。这里,我们证明在病毒感染小鼠后,来自LLEC的效应细胞迅速进入非淋巴组织并减少病原体负担,但在很大程度上依赖于从血管内皮细胞接收抗原信号。单细胞RNA测序显示,由KLRG1或KLRG1-记忆前体产生的非淋巴组织中的次级记忆细胞会产生类似的常驻记忆转录签名。因此,虽然LLECs不能分化为其他循环记忆群体,他们仍然保留进入组织和建立居住权的灵活性。
    KLRG1+ CD8 T cells persist for months after clearance of acute infections and maintain high levels of effector molecules, contributing protective immunity against systemic pathogens. Upon secondary infection, these long-lived effector cells (LLECs) are incapable of forming other circulating KLRG1- memory subsets such as central and effector memory T cells. Thus, KLRG1+ memory T cells are frequently referred to as a terminally differentiated population that is relatively short lived. Here, we show that after viral infection of mice, effector cells derived from LLECs rapidly enter nonlymphoid tissues and reduce pathogen burden but are largely dependent on receiving antigen cues from vascular endothelial cells. Single-cell RNA sequencing reveals that secondary memory cells in nonlymphoid tissues arising from either KLRG1+ or KLRG1- memory precursors develop a similar resident memory transcriptional signature. Thus, although LLECs cannot differentiate into other circulating memory populations, they still retain the flexibility to enter tissues and establish residency.
    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
    基于先前遇到抗原的经验,昆虫的先天免疫系统可以更迅速和有效地响应病原体。对控制免疫启动的分子机制的理解,昆虫的一种免疫记忆,包括它的跨代遗传,仍然难以捉摸。目前尚不清楚在引发的昆虫中观察到的免疫基因的增强表达是否可以持续存在,并通过DNA或组蛋白的表观遗传修饰通过染色质结构的变化来调节。反映哺乳动物的观察结果。越来越多的实验证据表明,DNA/RNA甲基化和组蛋白乙酰化水平的表观遗传变化可以调节昆虫对病原体暴露的免疫反应的激活。此外,模式昆虫宿主中某些表观遗传修饰的跨代遗传可以影响预编程免疫应答向后代的传递,导致进化抗性的发展。在模型昆虫宿主中的表观遗传研究正处于对先天免疫中染色质重塑的机理理解的重大进展的边缘。特别是免疫启动和表观遗传改变之间的直接关系。在这次审查中,我们讨论了DNA甲基化和组蛋白乙酰化参与发育的最新发现,维护,和昆虫免疫记忆的遗传,最终导致对病原体的抗性进化。
    The innate immune system of insects can respond more swiftly and efficiently to pathogens based on previous experience of encountering antigens. The understanding of molecular mechanisms governing immune priming, a form of immune memory in insects, including its transgenerational inheritance, remains elusive. It is still unclear if the enhanced expression of immune genes observed in primed insects can persist and be regulated through changes in chromatin structure via epigenetic modifications of DNA or histones, mirroring observations in mammals. Increasing experimental evidence suggests that epigenetic changes at the level of DNA/RNA methylation and histone acetylation can modulate the activation of insects\' immune responses to pathogen exposure. Moreover, transgenerational inheritance of certain epigenetic modifications in model insect hosts can influence the transmission of pre-programmed immune responses to the offspring, leading to the development of evolved resistance. Epigenetic research in model insect hosts is on the brink of significant progress in the mechanistic understanding of chromatin remodeling within innate immunity, particularly the direct relationships between immunological priming and epigenetic alterations. In this review, we discuss the latest discoveries concerning the involvement of DNA methylation and histone acetylation in shaping the development, maintenance, and inheritance of immune memory in insects, culminating in the evolution of resistance against 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: 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
    记忆CD4T细胞对人体免疫至关重要,然而,目前尚不清楚记忆形成过程中的病毒性炎症是否具有长期后果。这里,我们比较了Spike(S)特异性记忆CD4T细胞的转录和表观遗传特征,这些个体首次暴露于S是通过SARS-CoV-2感染或mRNA疫苗接种.记忆形成近2年后,通过感染建立的S特异性CD4T细胞仍然富集了与细胞毒性相关的转录本和干扰素刺激的基因,可能是因为炎症改变了染色质可及性景观。此外,与疫苗引发的细胞相比,感染引发的S特异性CD4T细胞在体外的增殖能力降低。此外,S特异性记忆CD4T细胞的转录状态在加强免疫和/或突破性感染时发生了最小程度的改变.因此,感染相关的炎症持久印记CD4T细胞记忆,这会影响这些细胞的功能,并可能对长期免疫产生影响。
    Memory CD4 T cells are critical to human immunity, yet it is unclear whether viral inflammation during memory formation has long-term consequences. Here, we compared transcriptional and epigenetic landscapes of Spike (S)-specific memory CD4 T cells in 24 individuals whose first exposure to S was via SARS-CoV-2 infection or mRNA vaccination. Nearly 2 years after memory formation, S-specific CD4 T cells established by infection remained enriched for transcripts related to cytotoxicity and for interferon-stimulated genes, likely because of a chromatin accessibility landscape altered by inflammation. Moreover, S-specific CD4 T cells primed by infection had reduced proliferative capacity in vitro relative to vaccine-primed cells. Furthermore, the transcriptional state of S-specific memory CD4 T cells was minimally altered by booster immunization and/or breakthrough infection. Thus, infection-associated inflammation durably imprints CD4 T cell memory, which affects the function of these cells and may have consequences for long-term immunity.
    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