T cell biology

  • 文章类型: Journal Article
    粘膜相关的不变T(MAIT)细胞是一组非常规T细胞,可识别MHC-I相关蛋白1(MR1)呈递的小分子代谢物,通过αβT细胞受体(TCR)。MAITTCR具有基本上不变的TCRα链,这在哺乳动物之间是高度保守的。同样,MR1是最高度保守的MHC-I样分子。这种极端的保护,包括MAITTCR和MR1之间的相互作用模式,已显示允许T细胞生物学中独特的物种错配反应性,从而允许在比较免疫学研究中使用选定的物种错配MR1抗原(MR1-Ag)四聚体。然而,物种错配MR1-Ag四聚体在鉴定不同物种MAIT细胞时的交叉反应模式尚未得到正式评估.我们开发了新的牛和猪MR1-Ag四聚体,并利用这些与先前开发的人类,小鼠和猪尾猕猴MR1-Ag四聚体表征跨物种四聚体反应性。来自每个物种的MR1-Ag四聚体以与物种匹配的MR1-Ag四聚体相当的特异性鉴定了远亲物种中的T细胞群体。然而,染色特征存在细微差异,对MAIT细胞的准确鉴定具有实际意义。猪MR1在物种中充分保守,猪MR1-Ag四聚体鉴定了来自其他物种的MAIT细胞。然而,猪的MAIT细胞处于表型检测的极限。在没有绵羊MR1-Ag四聚体的情况下,通过表型鉴定了绵羊血液中的MAIT细胞群,利用物种错配的MR1-Ag四聚体。总的来说,我们的结果验证了物种错配MR1-Ag四聚体在比较免疫学研究中的用途和局限性.
    Mucosal-associated invariant T (MAIT) cells are a subset of unconventional T cells that recognize small molecule metabolites presented by major histocompatibility complex class I related protein 1 (MR1), via an αβ T cell receptor (TCR). MAIT TCRs feature an essentially invariant TCR α-chain, which is highly conserved between mammals. Similarly, MR1 is the most highly conserved major histocompatibility complex-I-like molecule. This extreme conservation, including the mode of interaction between the MAIT TCR and MR1, has been shown to allow for species-mismatched reactivities unique in T cell biology, thereby allowing the use of selected species-mismatched MR1-antigen (MR1-Ag) tetramers in comparative immunology studies. However, the pattern of cross-reactivity of species-mismatched MR1-Ag tetramers in identifying MAIT cells in diverse species has not been formally assessed. We developed novel cattle and pig MR1-Ag tetramers and utilized these alongside previously developed human, mouse, and pig-tailed macaque MR1-Ag tetramers to characterize cross-species tetramer reactivities. MR1-Ag tetramers from each species identified T cell populations in distantly related species with specificity that was comparable to species-matched MR1-Ag tetramers. However, there were subtle differences in staining characteristics with practical implications for the accurate identification of MAIT cells. Pig MR1 is sufficiently conserved across species that pig MR1-Ag tetramers identified MAIT cells from the other species. However, MAIT cells in pigs were at the limits of phenotypic detection. In the absence of sheep MR1-Ag tetramers, a MAIT cell population in sheep blood was identified phenotypically, utilizing species-mismatched MR1-Ag tetramers. Collectively, our results validate the use and define the limitations of species-mismatched MR1-Ag tetramers in comparative immunology studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Published Erratum
    [这更正了文章DOI:10.3389/fimmu.2023.1067352。].
    [This corrects the article DOI: 10.3389/fimmu.2023.1067352.].
    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
    肝胆胆(HPB)恶性肿瘤难以治疗,并且继续具有高死亡率和对标准疗法的显著治疗抗性。免疫肿瘤学(IO)疗法在几种实体恶性肿瘤中与化疗联合使用时已显示出疗效。而胰腺导管腺癌(PDA)的反应率很低。虽然在肝细胞癌(HCC)和胆管癌(CCA)中很有前途,充分利用IO疗法治疗HPB肿瘤的需求仍未满足.因此,我们使用一种新的方法定义了HPB患者肿瘤微环境中的T细胞亚群,多参数流式细胞术和生物信息学分析。我们的发现量化了与检查点受体表达相关的T细胞表型状态。我们证明了CD103+组织驻留记忆T细胞(TRM)的存在,CCR7+中央记忆T细胞,所有HPB癌症的CD57+终末分化效应细胞,而抗肿瘤功能被多个共抑制性检查点受体的表达所抑制。缺乏共刺激受体的耗尽的T细胞在PDA中更为普遍,而表达共抑制和共刺激受体的部分耗尽的T细胞在HCC中最普遍,尤其是在早期阶段。HCC患者具有明显较高的TRM,其表型可以赋予免疫检查点疗法的恢复激活。Further,我们发现,在PDA患者中,化疗后T细胞激活状态或检查点表达缺乏显著改变.这些结果支持HCC患者可能从联合检查点疗法中受益最大,而细胞毒性化疗以外的其他努力可能需要增加CCA和PDA中的整体T细胞活化,以便将来进行临床开发。
    Hepato-pancreatico-biliary (HPB) malignancies are difficult-to-treat and continue to to have a high mortality and significant therapeutic resistance to standard therapies. Immune oncology (IO) therapies have demonstrated efficacy in several solid malignancies when combined with chemotherapy, whereas response rates in pancreatic ductal adenocarcinoma (PDA) are poor. While promising in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), there remains an unmet need to fully leverage IO therapies to treat HPB tumors. We therefore defined T cell subsets in the tumor microenvironment of HPB patients utilizing a novel, multiparameter flow cytometry and bioinformatics analysis. Our findings quantify the T cell phenotypic states in relation to checkpoint receptor expression. We demonstrate the presence of CD103+ tissue resident memory T cells (TRM), CCR7+ central memory T cells, and CD57+ terminally differentiated effector cells across all HPB cancers, while the anti-tumor function was dampened by expression of multiple co-inhibitory checkpoint receptors. Terminally exhausted T cells lacking co-stimulatory receptors were more prevalent in PDA, whereas partially exhausted T cells expressing both co-inhibitory and co-stimulatory receptors were most prevalent in HCC, especially in early stage. HCC patients had significantly higher TRM with a phenotype that could confer restored activation in response to immune checkpoint therapies. Further, we found a lack of robust alteration in T cell activation state or checkpoint expression in response to chemotherapy in PDA patients. These results support that HCC patients might benefit most from combined checkpoint therapies, whereas efforts other than cytotoxic chemotherapy will likely be necessary to increase overall T cell activation in CCA and PDA for future clinical development.
    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
    肾移植中基于Belatacept的免疫抑制比钙调磷酸酶抑制剂赋予更少的脱靶毒性,但其代价是急性排斥反应的发生率和严重程度增加。可能是由于其对Foxp3+调节性T细胞(Tregs)的数量和功能的有害作用。TIGIT是在包括Tregs的免疫细胞的几个亚群上表达的CD28家族共抑制受体。我们假设通过TIGIT信号传导的共抑制可以改善共刺激阻断抵抗的排斥反应。结果表明,用激动性抗TIGIT抗体治疗,当与CTLA-4Ig的共刺激阻断相结合时,与单独使用CTLA-4Ig相比,可以延长小鼠皮肤移植模型中的同种异体移植物存活时间。Further,移植物存活的这种延长伴随着移植物浸润Tregs的频率和数量的增加以及移植物中CD8+T细胞数量的减少。通过使用Treg特异性TIGIT条件敲除动物,我们证明TIGIT介导的移植物浸润CD8+T细胞应答的减少依赖于TIGIT在Foxp3+Tregs上的信号传导.我们的结果强调了TIGIT在CTLA-4被阻断的条件下对Foxp3+Tregs的关键功能作用,以及TIGIT激动作用优化基于共刺激阻断的免疫抑制的治疗潜力。
    Belatacept-based immunosuppression in kidney transplantation confers fewer off-target toxicities than calcineurin inhibitors but comes at a cost of increased incidence and severity of acute rejection, potentially due to its deleterious effect on both the number and function of Foxp3+ regulatory T cells (Tregs). TIGIT is a CD28 family coinhibitory receptor expressed on several subsets of immune cells including Tregs. We hypothesized that coinhibition through TIGIT signaling could function to ameliorate costimulation blockade-resistant rejection. The results demonstrate that treatment with an agonistic anti-TIGIT antibody, when combined with costimulation blockade by CTLA-4Ig, can prolong allograft survival in a murine skin graft model compared with CTLA-4Ig alone. Further, this prolongation of graft survival is accompanied by an increase in the frequency and number of graft-infiltrating Tregs and a concomitant reduction in the number of CD8+ T cells in the graft. Through the use of Treg-specific TIGIT conditional knockout animals, we demonstrated that the TIGIT-mediated reduction in the graft-infiltrating CD8+ T cell response is dependent on signaling of TIGIT on Foxp3+ Tregs. Our results highlight both the key functional role of TIGIT on Foxp3+ Tregs under conditions in which CTLA-4 is blocked and the therapeutic potential of TIGIT agonism to optimize costimulation blockade-based immunosuppression.
    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
    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
    涉及T淋巴细胞的免疫发病机制,在防御病毒感染方面发挥着关键作用,可能有助于COVID-19疾病谱,并提供治疗途径。为了解决这个问题,我们对中英文临床观察性研究和尸检数据进行了回顾,并检索了注册的临床试验.与非严重疾病相比,影响CD4和CD8T细胞的外周淋巴细胞减少是严重COVID-19的一个显著特点。尸检数据显示T细胞渗入器官,尤其是肺。正在进行的74项临床试验可能针对T细胞相关的发病机制,特别是IL-6途径。SARS-CoV-2感染中断重症COVID-19患者的T细胞循环。这可能是由于T细胞重新分布到受感染的器官中,激活诱导的衰竭,凋亡,或焦亡。在COVID-19期间测量T细胞动力学将为住院患者的临床风险分层提供信息,并可以确定那些将从靶向T细胞的治疗中受益最大的患者。
    Immunopathogenesis involving T lymphocytes, which play a key role in defence against viral infection, could contribute to the spectrum of COVID-19 disease and provide an avenue for treatment. To address this question, a review of clinical observational studies and autopsy data in English and Chinese languages was conducted with a search of registered clinical trials. Peripheral lymphopenia affecting CD4 and CD8 T cells was a striking feature of severe COVID-19 compared with non-severe disease. Autopsy data demonstrated infiltration of T cells into organs, particularly the lung. Seventy-four clinical trials are on-going that could target T cell-related pathogenesis, particularly IL-6 pathways. SARS-CoV-2 infection interrupts T cell circulation in patients with severe COVID-19. This could be due to redistribution of T cells into infected organs, activation induced exhaustion, apoptosis, or pyroptosis. Measuring T cell dynamics during COVID-19 will inform clinical risk-stratification of hospitalised patients and could identify those who would benefit most from treatments that target T 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
    口服抗原暴露是一个强大的,非侵入性途径诱导对饮食抗原的免疫耐受,在啮齿动物移植模型中,在延长移植物存活方面取得了适度的成功。利用口腔耐受机制促进长期移植接受,我们开发了一种小鼠模型,其中在用表达OVA的皮肤移植物移植之前口服引入抗原卵清蛋白(OVA)。移植前口服OVA治疗可促进永久性移植物接受,并与表达与其他抗原2W融合的OVA的皮肤移植物的耐受性相关。耐受性是供体特异性的,作为次要捐赠者匹配,但不是自发接受第三方同种异体移植。口服OVA治疗促进移植前OVA反应性CD4+和CD8+常规T细胞(Tconvs)和扩增的OVA反应性Tregs的无能表型。然而,口服OVA后接受皮肤移植物抵抗了Tregs的部分消耗和PD-L1的阻断。机械上,我们揭示了近端肠道引流淋巴结(gdLNs)在介导这种作用中的作用,作为排入近端gdLNs的肠道感染阻止了耐受性诱导。我们的研究扩展了将口服抗原暴露于移植的先前工作,并证明了支持口服耐受性的全身免疫机制足以促进长期移植物接受。
    Oral antigen exposure is a powerful, non-invasive route to induce immune tolerance to dietary antigens, and has been modestly successful at prolonging graft survival in rodent models of transplantation. To harness the mechanisms of oral tolerance for promoting long-term graft acceptance, we developed a mouse model where the antigen ovalbumin (OVA) was introduced orally prior to transplantation with skin grafts expressing OVA. Oral OVA treatment pre-transplantation promoted permanent graft acceptance and linked tolerance to skin grafts expressing OVA fused to the additional antigen 2W. Tolerance was donor-specific, as secondary donor-matched, but not third-party allografts were spontaneously accepted. Oral OVA treatment promoted an anergic phenotype in OVA-reactive CD4+ and CD8+ conventional T cells (Tconvs) and expanded OVA-reactive Tregs pre-transplantation. However, skin graft acceptance following oral OVA resisted partial depletion of Tregs and blockade of PD-L1. Mechanistically, we revealed a role for the proximal gut draining lymph nodes (gdLNs) in mediating this effect, as an intestinal infection that drains to the proximal gdLNs prevented tolerance induction. Our study extends previous work applying oral antigen exposure to transplantation and serves as proof of concept that the systemic immune mechanisms supporting oral tolerance are sufficient to promote long-term graft acceptance.
    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
    同种异体抗原特异性调节性T细胞(Treg)治疗是抑制同种免疫应答和减少实体器官移植后免疫抑制的有希望的方法。靶向供体同种异体抗原的嵌合抗原受体(CAR)可以将供体反应性赋予Treg。然而,尚未在血管化移植或多MHC错配模型中评估CARTreg疗法。这里,我们评估了靶向HLA-A2(A2-CAR)的CAR-Tregs延长小鼠异位心脏移植存活的能力.验证体外激活后,扩散,在A2-抗原存在下增强A2-CAR-Tregs的抑制功能,我们分析了接受表达A2的同种异体心脏移植物的C57BL/6(B6)小鼠中Tregs的体内功能。A2-CAR-Treg输注增加了来自B6的移植物的中位存活率。HLA-A2转基因供体从23到99天,而多克隆Treg输注的中位生存期为35天.在来自BALB/cxB6的单声道错配心脏的更严格模型中。HLA-A2F1供体,A2-CARTregs从11到14天略微增加了移植物的中位存活率,当与9天的雷帕霉素疗程联合时,进一步延长至>100天。这些发现证明了CARTregs的功效,单独或与免疫抑制剂联合使用,在完全免疫能力受体中保护血管化移植物。
    Alloantigen-specific regulatory T cell (Treg) therapy is a promising approach for suppressing alloimmune responses and minimizing immunosuppression after solid organ transplantation. Chimeric antigen receptor (CAR) targeting donor alloantigens can confer donor reactivity to Tregs. However, CAR Treg therapy has not been evaluated in vascularized transplant or multi-MHC mismatched models. Here, we evaluated the ability of CAR Tregs targeting HLA-A2 (A2-CAR) to prolong the survival of heterotopic heart transplants in mice. After verifying the in vitro activation, proliferation, and enhanced suppressive function of A2-CAR Tregs in the presence of A2-antigen, we analyzed the in vivo function of Tregs in C57BL/6 (B6) mice receiving A2-expressing heart allografts. A2-CAR Treg infusion increased the median survival of grafts from B6.HLA-A2 transgenic donors from 23 to 99 days, whereas median survival with polyclonal Treg infusion was 35 days. In a more stringent model of haplo-mismatched hearts from BALB/cxB6.HLA-A2 F1 donors, A2-CAR Tregs slightly increased median graft survival from 11 to 14 days, which was further extended to >100 days when combined with a 9-day course of rapamycin treatment. These findings demonstrate the efficacy of CAR Tregs, alone or in combination with immunosuppressive agents, toward protecting vascularized grafts in fully immunocompetent recipients.
    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
    CCR5KO肾移植(KTx)接受者是非常高的同种抗体生产者,并发展出模拟人抗体介导的排斥(AMR)的病理学。C57BL/6和CCR5KO小鼠(H-2b)用A/J肾(H-2a)移植;选择的组群在KTx后第5天接受过继细胞疗法(ACT),其用同种异体CXCR5+CD8+T细胞(或对照细胞)。通过测量移植后同种抗体评估ACT疗效,病理学,和同种异体移植存活。评估受体的CXCR5+CD8+T细胞的数量和CD8介导的对同种异体IgG+B细胞的细胞毒性。CCR5KO受体的同种抗体滴度比C57BL/6受体高4倍。KTx后7天外周血中同种异体CXCR5+CD8+T细胞的比例,淋巴结,与C57BL/6受体相比,CCR5KO的脾脏明显更低。在CCR5KO受体中,对同种异体IgG+B细胞的体内细胞毒性也降低了六倍。使用同种异体CXCR5+CD8+T细胞(但不使用同种异体CXCR5-CD8+或第三方引发的CXCR5+CD8+T细胞)显著降低同种抗体滴度,改善AMR病理学,和延长同种异体移植物存活。这些结果表明,在CCR5KO受体小鼠中,同种异体CXCR5CD8T细胞的数量和功能缺乏有助于高同种抗体和AMR,可以用ACT营救。
    CCR5 KO kidney transplant (KTx) recipients are extraordinarily high alloantibody producers and develop pathology that mimics human antibody-mediated rejection (AMR). C57BL/6 and CCR5 KO mice (H-2b ) were transplanted with A/J kidneys (H-2a ); select cohorts received adoptive cell therapy (ACT) with alloprimed CXCR5+ CD8+ T cells (or control cells) on day 5 after KTx. ACT efficacy was evaluated by measuring posttransplant alloantibody, pathology, and allograft survival. Recipients were assessed for the quantity of CXCR5+ CD8+ T cells and CD8-mediated cytotoxicity to alloprimed IgG+ B cells. Alloantibody titer in CCR5 KO recipients was four-fold higher than in C57BL/6 recipients. The proportion of alloprimed CXCR5+ CD8+ T cells 7 days after KTx in peripheral blood, lymph node, and spleen was substantially lower in CCR5 KO compared to C57BL/6 recipients. In vivo cytotoxicity towards alloprimed IgG+ B cells was also reduced six-fold in CCR5 KO recipients. ACT with alloprimed CXCR5+ CD8+ T cells (but not alloprimed CXCR5- CD8+ or third-party primed CXCR5+ CD8+ T cells) substantially reduced alloantibody titer, ameliorated AMR pathology, and prolonged allograft survival. These results indicate that a deficiency in quantity and function of alloprimed CXCR5+ CD8+ T cells contributes to high alloantibody and AMR in CCR5 KO recipient mice, which can be rescued with ACT.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号