CD8 T cells

CD8 T 细胞
  • 文章类型: Journal Article
    在先前感染或疫苗接种的背景下,免疫引发引发针对随后的结核分枝杆菌(Mtb)感染的保护性应答。然而,原发性Mtb感染后肺细胞环境中发生的变化及其对再感染时的保护作用仍知之甚少.在非人灵长类动物再感染模型中使用临床和微生物学终点,我们证明,之前的Mtb感染引发了针对随后的Mtb暴露的持久保护性反应,并且是CD4+T细胞依赖性的.通过分析原发感染的数据,再感染,和再感染-CD4+T细胞耗尽肉芽肿,我们发现,CD4+T细胞在再感染过程中的存在导致炎症较少的肺部环境,其特征是重新编程的CD8+T细胞,嗜中性粒细胞减少,和髓样细胞中的1型免疫信号减弱。这些结果为开发不仅靶向淋巴细胞而且调节先天免疫细胞以限制结核病(TB)疾病的疫苗和治疗剂开辟了途径。
    Immunological priming-in the context of either prior infection or vaccination-elicits protective responses against subsequent Mycobacterium tuberculosis (Mtb) infection. However, the changes that occur in the lung cellular milieu post-primary Mtb infection and their contributions to protection upon reinfection remain poorly understood. Using clinical and microbiological endpoints in a non-human primate reinfection model, we demonstrated that prior Mtb infection elicited a long-lasting protective response against subsequent Mtb exposure and was CD4+ T cell dependent. By analyzing data from primary infection, reinfection, and reinfection-CD4+ T cell-depleted granulomas, we found that the presence of CD4+ T cells during reinfection resulted in a less inflammatory lung milieu characterized by reprogrammed CD8+ T cells, reduced neutrophilia, and blunted type 1 immune signaling among myeloid cells. These results open avenues for developing vaccines and therapeutics that not only target lymphocytes but also modulate innate immune cells to limit tuberculosis (TB) disease.
    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
    CD8+T细胞在SARS-CoV-2发病机制或mRNA-LNP疫苗诱导的对致死性COVID-19的保护中的作用尚不清楚。在C57BL/6小鼠中使用小鼠适应的SARS-CoV-2病毒(MA30),我们表明CD8+T细胞对于雌性小鼠的固有抗性或雄性小鼠对致死性SARS-CoV-2感染的易感性是不必要的。此外,用二脯氨酸融合前稳定的全长SARS-CoV-2Spike(S-2P)mRNA-LNP疫苗免疫的小鼠,它诱导Spike特异性抗体和CD8+T细胞特异性针对Spike衍生的VNFNFNGL肽,免受SARS-CoV-2感染引起的致死性和体重减轻,而用仅编码VNFNFNGL的mRNA-LNP接种的小鼠被保护免受致死性但不体重减轻。CD8+T细胞耗竭在VNFNFNGL中消除保护,但在S-2PmRNA-LNP疫苗接种的小鼠中不消除。因此,当存在保护性抗体时,mRNA-LNP疫苗诱导的CD8+T细胞是可有可无的,但在它们不存在时对于存活是必需的。因此,疫苗诱导的CD8+T细胞可能是保护SARS-CoV-2变体的关键,这些变体突变了保护性抗体靶向的表位.
    The role of CD8+ T cells in SARS-CoV-2 pathogenesis or mRNA-LNP vaccine-induced protection from lethal COVID-19 is unclear. Using mouse-adapted SARS-CoV-2 virus (MA30) in C57BL/6 mice, we show that CD8+ T cells are unnecessary for the intrinsic resistance of female or the susceptibility of male mice to lethal SARS-CoV-2 infection. Also, mice immunized with a di-proline prefusion-stabilized full-length SARS-CoV-2 Spike (S-2P) mRNA-LNP vaccine, which induces Spike-specific antibodies and CD8+ T cells specific for the Spike-derived VNFNFNGL peptide, are protected from SARS-CoV-2 infection-induced lethality and weight loss, while mice vaccinated with mRNA-LNPs encoding only VNFNFNGL are protected from lethality but not weight loss. CD8+ T cell depletion ablates protection in VNFNFNGL but not in S-2P mRNA-LNP-vaccinated mice. Therefore, mRNA-LNP vaccine-induced CD8+ T cells are dispensable when protective antibodies are present but essential for survival in their absence. Hence, vaccine-induced CD8+ T cells may be critical to protect against SARS-CoV-2 variants that mutate epitopes targeted by protective antibodies.
    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
    CD8+T细胞是癌症免疫的末端效应子。大多数形式的有效癌症免疫疗法涉及CD8+T细胞效应子功能。这里,我们回顾了目前对癌症中T细胞功能的理解,重点关注关键CD8+T细胞亚型和状态。我们通过结合经典三信号模型和第四信号代谢的框架,讨论了影响CD8+T细胞分化和癌症功能的因素,并从T细胞角度考虑了肿瘤微环境的影响。我们认为免疫疗法的概念是“前药”,可以增强或调节T细胞,最终作为体内药物,以及整体免疫健康在癌症治疗和预防中的重要性。了解癌症中T细胞功能的进展已经并将继续改善免疫系统在更广泛的肿瘤类型中的利用,以使更多患者受益。
    CD8+ T cells are end effectors of cancer immunity. Most forms of effective cancer immunotherapy involve CD8+ T cell effector function. Here, we review the current understanding of T cell function in cancer, focusing on key CD8+ T cell subtypes and states. We discuss factors that influence CD8+ T cell differentiation and function in cancer through a framework that incorporates the classic three-signal model and a fourth signal-metabolism-and also consider the impact of the tumor microenvironment from a T cell perspective. We argue for the notion of immunotherapies as \"pro-drugs\" that act to augment or modulate T cells, which ultimately serve as the drug in vivo, and for the importance of overall immune health in cancer treatment and prevention. The progress in understanding T cell function in cancer has and will continue to improve harnessing of the immune system across broader tumor types to benefit more patients.
    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
    在开始抗逆转录病毒治疗(ART)之前,HIV特异性CD8+T细胞功能失调且寿命短。为了更好地理解CD4+T细胞中的HIV储库与HIV特异性CD8+T细胞的大小和分化状态之间的关系,我们调查了这些细胞来自急性和慢性HIV感染个体,经过2年的ART治疗.尽管在ART治疗2年后,HIV储库和CD8+T细胞反应均显著下降,持续的HIV特异性CD8+T细胞应答与整合的HIV前病毒的减少有关.然而,HIVGag特异性CD8+T细胞的大小,波尔,Nef,和Vif蛋白与ART期间的活性储库大小呈正相关,以细胞相关RNA测量。重要的是,高HIVDNA水平与短期HIV特异性CD8+T细胞的维持密切相关,无论ART启动时间如何。我们的数据表明,活跃的储库保持了HIV特异性CD8T细胞的数量,但阻止了它们分化为功能细胞。
    Before initiation of antiretroviral therapy (ART), HIV-specific CD8+ T cells are dysfunctional and short lived. To better understand the relationship between the HIV reservoir in CD4+ T cells and the magnitude and differentiation status of HIV-specific CD8+ T cells, we investigated these cells from acute and chronic HIV-infected individuals after 2 years of ART. Although both the HIV reservoir and the CD8+ T cell responses declined significantly after 2 years of ART, sustained HIV-specific CD8+ T cell responses correlated with a greater reduction of integrated HIV provirus. However, the magnitude of CD8+ T cells specific for HIV Gag, Pol, Nef, and Vif proteins positively associated with the active reservoir size during ART, measured as cell-associated RNA. Importantly, high HIV DNA levels strongly associate with maintenance of short-lived HIV-specific CD8+ T cells, regardless of ART initiation time. Our data suggest that the active reservoir maintains HIV-specific CD8+ T cell magnitude but prevents their differentiation into functional cells.
    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
    尽管放疗(RT)在非小细胞肺癌(NSCLC)的治疗中取得了巨大的成功,即使与免疫检查点阻断剂(ICBs)联合使用,局部复发仍然会发生,而且也很少见到外镜效应.这里,我们使用单细胞转录组和T细胞受体测序,在治疗耐药的小鼠肿瘤模型中描述了RT后肿瘤浸润免疫细胞的动态变化.在早期阶段,先天和适应性免疫系统被激活。在后期阶段,然而,肿瘤免疫微环境(TIME)转变为免疫抑制特性。我们的研究表明,抑制CD39结合RT优先降低耗尽的CD8+T细胞的百分比。此外,我们发现,T细胞激活的V域免疫球蛋白抑制因子(VISTA)阻断和RT的组合协同减少免疫抑制性骨髓细胞。临床上,VISTA高表达与NSCLC患者预后不良相关.总之,我们的数据提供了从免疫角度对获得性RT抗性的深入见解,并提出了合理的组合策略.
    Although radiotherapy (RT) has achieved great success in the treatment of non-small cell lung cancer (NSCLC), local relapses still occur and abscopal effects are rarely seen even when it is combined with immune checkpoint blockers (ICBs). Here, we characterize the dynamic changes of tumor-infiltrating immune cells after RT in a therapy-resistant murine tumor model using single-cell transcriptomes and T cell receptor sequencing. At the early stage, the innate and adaptive immune systems are activated. At the late stage, however, the tumor immune microenvironment (TIME) shifts into immunosuppressive properties. Our study reveals that inhibition of CD39 combined with RT preferentially decreases the percentage of exhausted CD8+ T cells. Moreover, we find that the combination of V-domain immunoglobulin suppressor of T cell activation (VISTA) blockade and RT synergistically reduces immunosuppressive myeloid cells. Clinically, high VISTA expression is associated with poor prognosis in patients with NSCLC. Altogether, our data provide deep insight into acquired resistance to RT from an immune perspective and present rational combination strategies.
    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
    HIV感染在抗逆转录病毒治疗(ART)期间持续存在,这是由于潜伏感染的细胞具有复制能力的病毒并逃避免疫力。先前的离体研究表明,HIV感染者的CD8+T细胞可能通过非细胞溶解机制抑制HIV表达,但造成这种影响的机制尚不清楚.这里,我们使用了基于原代细胞的体外潜伏期模型,并证明了自体活化CD8+T细胞与HIV感染的记忆CD4+T细胞的共培养促进了代谢和/或信号通路的特定变化,从而增加了CD4+T细胞的存活率。静止,和干劲。总的来说,这些途径负调控HIV的表达,并最终促进潜伏期的建立.如前面所示,我们观察到巨噬细胞,但不是B细胞,促进CD4+T细胞的潜伏期。CD8特异性机制的前潜伏期活性的鉴定可能有利于消除HIV感染者病毒库的方法的发展。
    HIV infection persists during antiretroviral therapy (ART) due to a reservoir of latently infected cells that harbor replication-competent virus and evade immunity. Previous ex vivo studies suggested that CD8+ T cells from people with HIV may suppress HIV expression via non-cytolytic mechanisms, but the mechanisms responsible for this effect remain unclear. Here, we used a primary cell-based in vitro latency model and demonstrated that co-culture of autologous activated CD8+ T cells with HIV-infected memory CD4+ T cells promoted specific changes in metabolic and/or signaling pathways resulting in increased CD4+ T cell survival, quiescence, and stemness. Collectively, these pathways negatively regulated HIV expression and ultimately promoted the establishment of latency. As shown previously, we observed that macrophages, but not B cells, promoted latency in CD4+ T cells. The identification of CD8-specific mechanisms of pro-latency activity may favor the development of approaches to eliminate the viral reservoir in people with HIV.
    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
    代谢状态代表了有效的过继性T细胞疗法(ACT)的主要障碍。的确,特异性脂质可损害CD8+T细胞(CTL)线粒体完整性,导致有缺陷的抗肿瘤反应。然而,脂质可影响CTL功能和命运的程度仍未研究。这里,我们表明,亚油酸(LA)是CTL活性的主要正调节剂,通过改善代谢适应性,防止精疲力竭,并刺激具有优越效应子功能的记忆样表型。我们报道LA治疗增强ER-线粒体接触(MERC)的形成,反过来促进钙(Ca2+)信号,线粒体能量学,和CTL效应子功能。作为一个直接的后果,LA指导的CD8T细胞在体外和体内的抗肿瘤效力均较高。因此,我们建议将LA治疗作为肿瘤治疗中的ACT增效剂。
    The metabolic state represents a major hurdle for an effective adoptive T cell therapy (ACT). Indeed, specific lipids can harm CD8+ T cell (CTL) mitochondrial integrity, leading to defective antitumor responses. However, the extent to which lipids can affect the CTL functions and fate remains unexplored. Here, we show that linoleic acid (LA) is a major positive regulator of CTL activity by improving metabolic fitness, preventing exhaustion, and stimulating a memory-like phenotype with superior effector functions. We report that LA treatment enhances the formation of ER-mitochondria contacts (MERC), which in turn promotes calcium (Ca2+) signaling, mitochondrial energetics, and CTL effector functions. As a direct consequence, the antitumor potency of LA-instructed CD8 T cells is superior in vitro and in vivo. We thus propose LA treatment as an ACT potentiator in tumor 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
    肿瘤免疫疗法的改进取决于对抗肿瘤T细胞应答的更好理解。通过研究人类肿瘤引流淋巴结(TDLN),我们发现TDLN中激活的CD8+T细胞共享功能,转录,肿瘤中具有TCF1+干细胞样细胞的表观遗传性状。表型和TCR重叠表明这些TDLN细胞是肿瘤驻留的干细胞样CD8+T细胞的前体。小鼠肿瘤模型显示,肿瘤特异性CD8+T细胞在TDLN中被激活,但缺乏效应子表型。这些干细胞样细胞迁移到肿瘤中,其中来自抗原呈递细胞的额外共刺激驱动效应子分化。这种响应于癌症的CD8+T细胞活化的模型不同于典型的CD8+T细胞对急性病毒的活化,它提出了肿瘤特异性CD8+T细胞激活的两个阶段:TDLN中的初始激活和额外共刺激后肿瘤内的后续效应程序采集。
    Improvements in tumor immunotherapies depend on better understanding of the anti-tumor T cell response. By studying human tumor-draining lymph nodes (TDLNs), we found that activated CD8+ T cells in TDLNs shared functional, transcriptional, and epigenetic traits with TCF1+ stem-like cells in the tumor. The phenotype and TCR overlap suggested that these TDLN cells were precursors to tumor-resident stem-like CD8+ T cells. Murine tumor models revealed that tumor-specific CD8+ T cells were activated in TDLNs but lacked an effector phenotype. These stem-like cells migrated into the tumor, where additional co-stimulation from antigen-presenting cells drove effector differentiation. This model of CD8+ T cell activation in response to cancer is different from that of canonical CD8+ T cell activation to acute viruses, and it proposes two stages of tumor-specific CD8+ T cell activation: initial activation in TDLNs and subsequent effector program acquisition within the tumor after additional co-stimulation.
    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
    CD4T细胞的帮助对于维持慢性感染期间的效应CD8T细胞反应至关重要,特别是通过T滤泡辅助(Tfh)衍生的白介素-21(IL-21)。相反,CD4耗竭导致严重的CD8T细胞功能障碍和终生病毒血症,尽管CD4T细胞在短暂耗竭后重新出现。这些观察结果表明,重新填充CD4亚群在功能或数字上不足以协调强大的CD8反应。我们利用空间转录组学和单细胞RNA测序(scRNA-seq)来研究CD4充足和耗尽条件下的CD4T细胞异质性,并探索慢性感染期间的细胞相互作用。尽管产生IL-21的Tfh细胞在短暂的CD4耗竭后重新繁殖,免疫调节性CD4T细胞的数量超过了它们。此外,脾结构似乎受到干扰,随着白浆区域的减少,与生发中心损失相吻合。脾结构的这些破坏与Tfh和祖细胞CD8T细胞共定位减少有关,在“无帮助”条件下,为受损的祖细胞-效应CD8T细胞分化提供了潜在机制。
    CD4 T cell help is critical to sustain effector CD8 T cell responses during chronic infection, notably via T follicular helper (Tfh)-derived interleukin-21 (IL-21). Conversely, CD4 depletion results in severe CD8 T cell dysfunction and lifelong viremia despite CD4 T cell reemergence following transient depletion. These observations suggest that repopulating CD4 subsets are functionally or numerically insufficient to orchestrate a robust CD8 response. We utilize spatial transcriptomics and single-cell RNA sequencing (scRNA-seq) to investigate CD4 T cell heterogeneity under CD4-replete and -deplete conditions and explore cellular interactions during chronic infection. Although IL-21-producing Tfh cells repopulate following transient CD4 depletion, they are outnumbered by immunomodulatory CD4 T cells. Moreover, the splenic architecture appears perturbed, with decreases in white pulp regions, coinciding with germinal center losses. These disruptions in splenic architecture are associated with diminished Tfh and progenitor CD8 T cell colocalization, providing a potential mechanism for impaired progenitor-to-effector CD8 T cell differentiation during \"un-helped\" conditions.
    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
    程序性细胞死亡蛋白1(PD-1)/程序性死亡配体1(PD-L1)阻断免疫疗法在治疗膀胱癌中具有有限的功效。这里,我们表明,在CD8+T细胞丰度高的膀胱肿瘤中,NKG2A与PD-L1阻断免疫疗法的生存率和反应性提高相关.在膀胱肿瘤中,NKG2A在CD8+T细胞上获得的时间晚于PD-1以及其他建立良好的免疫检查点。NKG2A+PD-1+CD8+T细胞通过其使用T细胞受体(TCR)独立的先天样机制与人类白细胞抗原(HLA)I类缺陷型肿瘤反应的能力而与经典定义的耗尽的T细胞不同。随着疾病进展,膀胱肿瘤的HLA-ABC表达逐渐减少,阐明靶向不依赖TCR的抗肿瘤功能的重要性。值得注意的是,当肿瘤表达HLA-E时,NKG2A+CD8+T细胞受到抑制,并且在NKG2A阻断时以HLA-E依赖性方式部分恢复。总的来说,我们的研究为随后的临床试验提供了一个框架,将NKG2A阻断与其他T细胞靶向免疫疗法相结合,其中肿瘤表达较高水平的HLA-E。
    Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1)-blockade immunotherapies have limited efficacy in the treatment of bladder cancer. Here, we show that NKG2A associates with improved survival and responsiveness to PD-L1 blockade immunotherapy in bladder tumors that have high abundance of CD8+ T cells. In bladder tumors, NKG2A is acquired on CD8+ T cells later than PD-1 as well as other well-established immune checkpoints. NKG2A+ PD-1+ CD8+ T cells diverge from classically defined exhausted T cells through their ability to react to human leukocyte antigen (HLA) class I-deficient tumors using T cell receptor (TCR)-independent innate-like mechanisms. HLA-ABC expression by bladder tumors is progressively diminished as disease progresses, framing the importance of targeting TCR-independent anti-tumor functions. Notably, NKG2A+ CD8+ T cells are inhibited when HLA-E is expressed by tumors and partly restored upon NKG2A blockade in an HLA-E-dependent manner. Overall, our study provides a framework for subsequent clinical trials combining NKG2A blockade with other T cell-targeted immunotherapies, where tumors express higher levels of HLA-E.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号