missing self

  • 文章类型: Journal Article
    抗体介导的排斥(ABMR)是移植物失败的主要原因。新出现的证据表明自然杀伤(NK)细胞对微血管炎症(MVI)的重要贡献。我们研究了遗传决定的NK细胞功能对ABMR发育和活性的影响。该研究包括86例接受供体特异性抗体检测触发的系统活检的肾移植受者。我们进行了杀手免疫球蛋白样受体分型,以预测缺失的自身和基因型多态性决定NK细胞功能(FCGR3AV/F158[rs396991],KLRC2wt/del,KLRK1HNK/LNK[rs1049174],rs9916629-C/T)。50例患者的ABMR具有相当大的MVI和升高的NK细胞转录物。自我缺失与MVI无关。只有KLRC2wt/wt显示出相关性(在KLRC2wt/del接受者中,MVI评分:2[中位数;四分位数间距:0-3]vs0[0-1];P=.001),并且在比例赔率多变量模型中仍然显着(赔率比,7.84;95%置信区间,2.37-30.47;P=.001)。包含所有多态性和缺失自我的总和得分并没有超过仅包括KLRC2和FCGR3A变体的得分,在单变量分析中具有预测性。NK细胞遗传学不影响移植物功能下降和存活。总之,功能性KLRC2多态性成为ABMR活性的独立决定因素,没有相当大的贡献缺失自身和其他NK细胞基因多态性。
    Antibody-mediated rejection (ABMR) is a leading cause of graft failure. Emerging evidence suggests a significant contribution of natural killer (NK) cells to microvascular inflammation (MVI). We investigated the influence of genetically determined NK cell functionality on ABMR development and activity. The study included 86 kidney transplant recipients subjected to systematic biopsies triggered by donor-specific antibody detection. We performed killer immunoglobulin-like receptor typing to predict missing self and genotyped polymorphisms determining NK cell functionality (FCGR3AV/F158 [rs396991], KLRC2wt/del, KLRK1HNK/LNK [rs1049174], rs9916629-C/T). Fifty patients had ABMR with considerable MVI and elevated NK cell transcripts. Missing self was not related to MVI. Only KLRC2wt/wt showed an association (MVI score: 2 [median; interquartile range: 0-3] vs 0 [0-1] in KLRC2wt/del recipients; P = .001) and remained significant in a proportional odds multivariable model (odds ratio, 7.84; 95% confidence interval, 2.37-30.47; P = .001). A sum score incorporating all polymorphisms and missing self did not outperform a score including only KLRC2 and FCGR3A variants, which were predictive in univariable analysis. NK cell genetics did not affect graft functional decline and survival. In conclusion, a functional KLRC2 polymorphism emerged as an independent determinant of ABMR activity, without a considerable contribution of missing self and other NK cell gene polymorphisms.
    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
    自然杀伤(NK)细胞是先天免疫细胞,有助于宿主防御病毒感染。NK细胞在体外对严重急性呼吸综合征冠状病毒2(SARS-CoV-2)有反应,并在2019年急性冠状病毒病(COVID-19)患者中被激活。然而,NK细胞检测SARS-CoV-2感染细胞的机制在很大程度上仍然未知。这里,我们显示SARS-CoV-2的非结构蛋白13编码由人白细胞抗原E(HLA-E)呈递的肽。与自身肽相反,病毒肽阻止HLA-E与抑制性受体NKG2A的结合,从而使靶细胞对NK细胞攻击敏感。根据这些观察,表达NKG2A的NK细胞在COVID-19患者中特别活化,并在体外熟练地限制SARS-CoV-2在感染的肺上皮细胞中的复制。因此,这些数据表明,由HLA-E提供的病毒肽消除了对NKG2ANK细胞的抑制,导致自我识别缺失。
    Natural killer (NK) cells are innate immune cells that contribute to host defense against virus infections. NK cells respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro and are activated in patients with acute coronavirus disease 2019 (COVID-19). However, by which mechanisms NK cells detect SARS-CoV-2-infected cells remains largely unknown. Here, we show that the Non-structural protein 13 of SARS-CoV-2 encodes for a peptide that is presented by human leukocyte antigen E (HLA-E). In contrast with self-peptides, the viral peptide prevents binding of HLA-E to the inhibitory receptor NKG2A, thereby rendering target cells susceptible to NK cell attack. In line with these observations, NKG2A-expressing NK cells are particularly activated in patients with COVID-19 and proficiently limit SARS-CoV-2 replication in infected lung epithelial cells in vitro. Thus, these data suggest that a viral peptide presented by HLA-E abrogates inhibition of NKG2A+ NK cells, resulting in missing self-recognition.
    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
    Detection of mismatched human leukocyte antigens by adaptive immune cells is considered as the main cause of transplant rejection, leading to either T-cell mediated rejection or antibody-mediated rejection. This canonical view guided the successful development of immunosuppressive therapies and shaped the diagnostic Banff classification for kidney transplant rejection that is used in clinics worldwide. However, several observations have recently emerged that question this dichotomization between T-cell mediated rejection and antibody-mediated rejection, related to heterogeneity in the serology, histology, and prognosis of the rejection phenotypes. In parallel, novel insights were obtained concerning the dynamics of donor-specific anti-human leukocyte antigen antibodies, the immunogenicity of donor-recipient non-human leukocyte antigen mismatches, and the autoreactivity against self-antigens. Moreover, the potential of innate allorecognition was uncovered, as exemplified by natural killer cell-mediated microvascular inflammation through missing self, and by the emerging evidence on monocyte-driven allorecognition. In this review, we highlight the gaps in the current classification of rejection, provide an overview of the expanding insights into the mechanisms of allorecognition, and critically appraise how these could improve our understanding and clinical approach to kidney transplant rejection. We argue that consideration of the complex interplay of various allorecognition mechanisms can foster a more integrated view of kidney transplant rejection and can lead to improved risk stratification, targeted therapies, and better outcome after kidney transplantation.
    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
    在患有微血管炎症(MVI)的肾移植受者中,循环抗HLA供体特异性抗体(HLA-DSA)通常不存在。想念自己,供体内皮细胞无法向受体自然杀伤细胞上的抑制性杀伤细胞Ig样受体(KIRs)提供HLAI介导的信号,会在体外引起内皮损伤,并与HLA-DSA阴性MVI相关。然而,缺失自我作为同种异体移植排斥反应的非体液触发因素的临床重要性尚不清楚。
    在2004年3月至2013年2月期间924例连续肾移植的基于人群的研究中,我们进行了高分辨率供体和受体HLA分型和受体KIR基因分型。自我缺失定义为缺乏A3/A11,Bw4,C1或C2供体基因型,存在相应的受教育者抑制性KIR基因。
    我们在924次移植中的399次中发现了自我缺失。同时出现的缺失自我类型在增加MVI风险方面具有累加效应,具有两种并发类型的阈值(危险比[HR],1.78;95%置信区间[95%CI],1.26to2.53),独立于HLA-DSA(HR,5.65;95%CI,4.01至7.96)。自我缺失和细胞排斥反应的损伤没有相关性。在222名患有MVI的接受者中,有146名未检测到HLA-DSA;146人中有28名至少有两种缺失的自身类型。MVI后与移植肾小球病相关的自我缺失(HR,2.51;95%CI,1.12至5.62),尽管同种异体移植存活率优于HLA-DSA相关MVI。
    自我特异性缺失和累积性缺失会增加肾移植后的MVI风险,独立于HLA-DSA。对缺失自我的系统评估可以提高对HLA-DSA阴性MVI的理解,并且可能与改进的诊断分类和患者风险分层有关。
    Circulating anti-HLA donor-specific antibodies (HLA-DSA) are often absent in kidney transplant recipients with microvascular inflammation (MVI). Missing self, the inability of donor endothelial cells to provide HLA I-mediated signals to inhibitory killer cell Ig-like receptors (KIRs) on recipient natural killer cells, can cause endothelial damage in vitro, and has been associated with HLA-DSA-negative MVI. However, missing self\'s clinical importance as a nonhumoral trigger of allograft rejection remains unclear.
    In a population-based study of 924 consecutive kidney transplantations between March 2004 and February 2013, we performed high-resolution donor and recipient HLA typing and recipient KIR genotyping. Missing self was defined as the absence of A3/A11, Bw4, C1, or C2 donor genotype, with the presence of the corresponding educated recipient inhibitory KIR gene.
    We identified missing self in 399 of 924 transplantations. Co-occurrence of missing self types had an additive effect in increasing MVI risk, with a threshold at two concurrent types (hazard ratio [HR], 1.78; 95% confidence interval [95% CI], 1.26 to 2.53), independent of HLA-DSA (HR, 5.65; 95% CI, 4.01 to 7.96). Missing self and lesions of cellular rejection were not associated. No HLA-DSAs were detectable in 146 of 222 recipients with MVI; 28 of the 146 had at least two missing self types. Missing self associated with transplant glomerulopathy after MVI (HR, 2.51; 95% CI, 1.12 to 5.62), although allograft survival was better than with HLA-DSA-associated MVI.
    Missing self specifically and cumulatively increases MVI risk after kidney transplantation, independent of HLA-DSA. Systematic evaluation of missing self improves understanding of HLA-DSA-negative MVI and might be relevant for improved diagnostic classification and patient risk stratification.
    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
    目前的移植免疫学教条认为同种异体移植排斥是由受体的适应性免疫系统引发的。在这个流行的模型中,一般的先天免疫细胞,特别是自然杀伤(NK)细胞,仅被认为是下游效应子,仅在适应性效应子募集后才参与移植物的破坏:同种反应性T细胞或供体特异性抗体(DSA)。挑战这个愿景,最近的数据表明,受体NK细胞能够进行同种异体识别,因为它们可以感觉到移植物内皮细胞表面缺乏自身HLAI类分子。缺失自身触发mTORC1依赖性NK细胞活化,这反过来促进移植物微血管炎症的发展,并对移植物存活产生不利影响。一些患者在没有DSA或遗传预测的自我缺失的情况下发生慢性血管排斥反应,这一事实表明其他分子机制可能会低估NK细胞的同种异体识别。这篇综述概述了这些已证实和推定的分子机制,并讨论了器官移植免疫学这一新兴领域的未来研究方向。
    The current transplant immunology dogma defends that allograft rejection is initiated by recipient\'s adaptive immune system. In this prevalent model, innate immune cells in general, and natural killer (NK) cells in particular, are merely considered as downstream effectors which participate in the destruction of the graft only upon recruitment by adaptive effectors: alloreactive T cells or donor-specific antibodies (DSA). Challenging this vision, recent data demonstrated that recipients\' NK cells are capable of a form of allorecognition because they can sense the absence of self HLA class I molecules on the surface of graft endothelial cells. Missing-self triggers mTORC1-dependent activation of NK cells, which in turn promote the development of graft microvascular inflammation and detrimentally impact graft survival. The fact that some patients develop chronic vascular rejection in absence of DSA or genetically-predicted missing self suggests that other molecular mechanisms could underly NK cell allorecognition. This review provides an overview of these proven and putative molecular mechanisms and discusses future research directions in this emerging field in organ transplant immunology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    供体特异性抗体(DSA)与不激活经典补体级联反应的肾脏同种异体移植内皮细胞的结合可以触发先天免疫效应物的募集,包括NK细胞。活化的NK细胞有助于微血管炎症,导致慢性抗体介导的排斥(AMR)。受体NK细胞还可以通过感知同种异体移植内皮细胞上自身HLAI类分子的缺失(“缺失自身”)来触发抗体非依赖性微血管炎症。这项翻译研究调查了自身缺失的状况是否会放大DSA依赖性NK细胞活化以恶化慢性AMR。在2004年至2017年间在里昂大学医院接受同种异体移植活检的1682名肾移植受者中,有135人符合AMR的诊断标准,并被纳入研究。通过阳性C3d结合测定鉴定的补体固定DSA患者(n=73,54%)具有较高的移植失败风险(P=0.002)。在其余的补体非依赖性慢性AMR患者中(n=62,46%),通过供体和受体基因分型鉴定出自我缺失者的同种异体移植存活率较差(P=0.02).在多变量分析中,仅蛋白尿(HR:7.24;P=0.01)和自身缺失(HR:3.57;P=0.04)是诊断为慢性AMR后移植失败的独立预测因子。人NK细胞和内皮细胞的共培养证实,DSA诱导的NK细胞活化增加了内皮损伤。
    在诊断慢性AMR时对自身缺失的评估可确定肾移植失败风险较高的患者。
    Binding of donor-specific antibodies (DSAs) to kidney allograft endothelial cells that does not activate the classic complement cascade can trigger the recruitment of innate immune effectors, including NK cells. Activated NK cells contribute to microvascular inflammation leading to chronic antibody-mediated rejection (AMR). Recipient NK cells can also trigger antibody-independent microvascular inflammation by sensing the absence of self HLA class I molecules (\"missing self\") on allograft endothelial cells. This translational study investigated whether the condition of missing self amplifies DSA-dependent NK cell activation to worsen chronic AMR.
    Among 1682 kidney transplant recipients who underwent an allograft biopsy at Lyon University Hospital between 2004 and 2017, 135 fulfilled the diagnostic criteria for AMR and were enrolled in the study. Patients with complement-fixing DSAs identified by a positive C3d binding assay (n=73, 54%) had a higher risk of transplant failure (P=0.002). Among the remaining patients with complement-independent chronic AMR (n=62, 46%), those in whom missing self was identified through donor and recipient genotyping exhibited worse allograft survival (P=0.02). In multivariable analysis, only proteinuria (HR: 7.24; P=0.01) and the presence of missing self (HR: 3.57; P=0.04) were independent predictors for transplant failure following diagnosis of chronic AMR. Cocultures of human NK cells and endothelial cells confirmed that addition of missing self to DSA-induced NK cell activation increased endothelial damage.
    The assessment of missing self at the time of diagnosis of chronic AMR identifies patients at higher risk for kidney transplant failure.
    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
    The contribution of natural killer (NK) cells to immunosurveillance of human cancer remains debatable. Here, we discuss advances in several areas of human NK cell research, many of which support the ability of NK cells to prevent cancer development and avoid relapse following adoptive immunotherapy. We describe the molecular basis for NK cell recognition of human tumor cells and provide evidence for NK cell-mediated killing of human primary tumor cells ex vivo. Subsequently, we highlight studies demonstrating the ability of NK cells to migrate to, and reside in, the human tumor microenvironment where selection of tumor escape variants from NK cells can occur. Indirect evidence for NK cell immunosurveillance against human malignancies is provided by the reduced incidence of cancer in individuals with high levels of NK cell cytotoxicity, and the significant clinical responses observed following infusion of human NK cells into cancer patients. Finally, we describe studies showing enhanced tumor progression, or increased cancer incidence, in patients with inherited and acquired defects in cellular cytotoxicity. All these observations have in common that they, either indirectly or directly, suggest a role for NK cells in mediating immunosurveillance against human cancer. This opens up for exciting possibilities with respect to further exploring NK cells in settings of adoptive immunotherapy in human cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The ability of NK cells to specifically recognize cells lacking expression of self-MHC class I molecules was discovered over 30 years ago. It provided the foundation for the \"missing self\" hypothesis. Research in the two past decades has contributed to a detailed understanding of the molecular mechanisms that determine the specificity and strength of NK cell-mediated \"missing self\" responses to tumor cells. However, in light of the recent remarkable breakthroughs in clinical cancer immunotherapy, the cytolytic potential of NK cells still remains largely untapped in clinical settings. There is abundant evidence demonstrating partial or complete loss of HLA class I expression in a wide spectrum of human tumor types. Such loss may result from immune selection of escape variants by tumor-specific CD8 T cells and has more recently also been linked to acquired resistance to checkpoint inhibition therapy. In the present review, we discuss the early predictions of the \"missing self\" hypothesis, its molecular basis and outline the potential for NK cell-based adoptive immunotherapy to convert checkpoint inhibitor therapy-resistant patients into clinical responders.
    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
    Missing self recognition makes cancer sensitive to natural killer cell (NKc) reactivity. However, this model disregards the NKc licensing effect, which highly increases NKc reactivity through interactions of inhibitory killer cell immunoglobulin-like receptors (iKIR) with their cognate HLA-I ligands. The influence of iKIR/HLA-ligand (HLA-C1/C2) licensing interactions on the susceptibility to and progression of plasma cell (PC) dyscrasias was evaluated in 164 Caucasian patients and 286 controls. Compared to controls, myeloma accumulates KIR2DL1-L2+L3- genotypes (2.8% vs. 13.2%, p < 0.01, OR = 5.29) and less diverse peripheral repertoires of NKc clones. Less diverse and weaker-affinity repertoires of iKIR2D/HLA-C licensing interactions increased myeloma susceptibility. Thus, the complete absence of conventional iKIR2D/HLA-C licensing interactions (KIR2DL1-L2+L3-/C2C2, 2.56% vs. 0.35%; p < 0.05; OR = 15.014), single-KIR2DL3+/C1+ (20.51% vs. 10.84%; p < 0.05; OR = 2.795) and single-KIR2DL2+/C1+ (12.82% vs. 4.9%; p < 0.01; OR = 5.18) interactions were over-represented in myeloma, compared to controls. Additionally, KIR2DL1-L2+L3- (20% vs. 83%, p < 0.00001) as well as KIR3DL1- (23% vs. 82%, p < 0.00001) genotypes had a dramatic negative impact on the 3-y progression-free survival (PFS), particularly in patients with low-tumor burden. Remarkably, myeloma-PCs, compared to K562 and other hematological cancers, showed substantial over-expression of HLA-I (\"increasing-self\" instead of missing-self), including HLA-C, and mild expression of ligands for NKc activating receptors (aRec) CD112, CD155, ULBP-1 and MICA/B, which apparently renders myeloma-PCs susceptible to lysis mainly by licensed NKc. KIR2DL1-L2+L3-/C2C2 patients (with no conventional iKIR2D/HLA-C licensing interactions) lyse K562 but barely lyse myeloma-PCs (4% vs. 15%; p < 0.05, compared to controls). These results support a model where immunosurveillance of no-missing-self cancers, e.g., myeloma, mainly depends on NKc licensing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    自然杀伤(NK)细胞代表对抗病原体和肿瘤细胞的第一道防线。NK细胞的活化通过源自在其表面上表达的活化和抑制性受体的信号的整合来调节。然而,不同的NK细胞对相同的刺激有不同的反应,它是使激活受体交联的靶细胞或试剂。决定NK细胞反应性水平的过程统称为NK细胞教育。NK细胞教育在稳态条件下发挥着重要作用,潜在的自身反应性NK细胞对周围环境具有耐受性。根据“调谐”的概念,定量调整每个NK细胞的反应性以确保自身耐受性,同时确保对潜在威胁的有用反应性。NK细胞显示的MHC特异性抑制性受体在调节NK细胞反应性中发挥主要作用。但是最近的研究表明,来自激活受体的信号也很重要,表明关键的决定因素是来自两种类型受体的整合信号。一个重要且仍未解决的问题是NK细胞教育是否涉及与环境中特定细胞群的相互作用。造血和/或非造血细胞是否发挥作用仍在争论中。最近的结果表明,NK细胞调节在稳态条件下表现出可塑性,这意味着如果MHC环境发生变化,它可以重新设置。其他证据表明,然而,伴随感染的炎症可能有利于高反应性,这表明炎症因子可以克服NK细胞适应稳态环境的自然趋势。这些发现提出了许多问题,例如病毒和肿瘤细胞是否操纵NK细胞反应以逃避免疫识别。随着对基础过程的了解的增长,为治疗目的调节NK细胞反应性的可能性变得越来越有吸引力,目前正在进行临床研究。
    Natural killer (NK) cells represent a first line of defense against pathogens and tumor cells. The activation of NK cells is regulated by the integration of signals deriving from activating and inhibitory receptors expressed on their surface. However, different NK cells respond differently to the same stimulus, be it target cells or agents that crosslink activating receptors. The processes that determine the level of NK cell responsiveness have been referred to collectively as NK cell education. NK cell education plays an important role in steady state conditions, where potentially auto-reactive NK cells are rendered tolerant to the surrounding environment. According to the \"tuning\" concept, the responsiveness of each NK cell is quantitatively adjusted to ensure self tolerance while at the same time ensuring useful reactivity against potential threats. MHC-specific inhibitory receptors displayed by NK cells play a major role in tuning NK cell responsiveness, but recent studies indicate that signaling from activating receptors is also important, suggesting that the critical determinant is an integrated signal from both types of receptors. An important and still unresolved question is whether NK cell education involves interactions with a specific cell population in the environment. Whether hematopoietic and/or non-hematopoietic cells play a role is still under debate. Recent results demonstrated that NK cell tuning exhibits plasticity in steady state conditions, meaning that it can be re-set if the MHC environment changes. Other evidence suggests, however, that inflammatory conditions accompanying infections may favor high responsiveness, indicating that inflammatory agents can over-ride the natural tendency of NK cells to adjust to the steady state environment. These findings raise many questions such as whether viruses and tumor cells manipulate NK cell responsiveness to evade immune-recognition. As knowledge of the underlying processes grows, the possibility of modulating NK cell responsiveness for therapeutic purposes is becoming increasingly attractive, and is now under serious investigation in clinical studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号