Qa-1b

  • 文章类型: Journal Article
    慢性病毒感染和癌症中的耗尽的CD8T(Tex)细胞具有抑制受体(IR)的持续共表达。Tex细胞可以通过阻断IR来恢复活力,例如PD-1,但是通过共同靶向包括PD-1和LAG-3在内的多个IR,可以实现协同恢复和增强的疾病控制。为了剖析这些IR通路被破坏时固有的分子变化,我们研究了慢性感染期间PD-1和/或LAG-3丢失对Tex细胞的影响.这些分析揭示了PD-1和LAG-3在调节Tex细胞增殖和效应子功能中的不同作用。分别。此外,这些研究确定了LAG-3在维持TOX和Tex细胞耐久性方面的重要作用,以及LAG-3依赖性电路,该电路产生了Tex细胞的CD94/NKG2亚群,具有通过识别应激配体Qa-1b介导的增强的细胞毒性,在人类中也有类似的观察。这些分析解开了PD-1和LAG-3的非冗余机制及其在调节Tex细胞中的协同作用。
    Exhausted CD8 T (Tex) cells in chronic viral infection and cancer have sustained co-expression of inhibitory receptors (IRs). Tex cells can be reinvigorated by blocking IRs, such as PD-1, but synergistic reinvigoration and enhanced disease control can be achieved by co-targeting multiple IRs including PD-1 and LAG-3. To dissect the molecular changes intrinsic when these IR pathways are disrupted, we investigated the impact of loss of PD-1 and/or LAG-3 on Tex cells during chronic infection. These analyses revealed distinct roles of PD-1 and LAG-3 in regulating Tex cell proliferation and effector functions, respectively. Moreover, these studies identified an essential role for LAG-3 in sustaining TOX and Tex cell durability as well as a LAG-3-dependent circuit that generated a CD94/NKG2+ subset of Tex cells with enhanced cytotoxicity mediated by recognition of the stress ligand Qa-1b, with similar observations in humans. These analyses disentangle the non-redundant mechanisms of PD-1 and LAG-3 and their synergy in regulating Tex 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
    可以想象,抑制性受体NKG2A与非经典MHC-I分子HLA-E或小鼠Qa-1b的相互作用在肿瘤微环境中抑制了NK细胞对癌症的反应性。我们发现,只有在与抗NKG2A和抗Qa-1b阻断性单克隆抗体共同注射针对实体小鼠肿瘤模型时,NK细胞的瘤内递送才能达到显着的治疗效果。这种治疗活性取决于内源性CD8T细胞和1型常规树突细胞(cDC1)。此外,与全身性抗PD-1mAb治疗联合治疗后,抗肿瘤作用得到增强,并对远处未注射的肿瘤取得部分外移疗效.在携带表达HLA-E的人类癌细胞的异种移植小鼠中,肿瘤内共注射活化的同种异体人NK细胞和临床级抗NKG2AmAb(monalizumab)协同实现了治疗效果。总之,这些研究为基于肿瘤内NK细胞的免疫治疗的临床潜力提供了证据,这些免疫治疗通过引发内源性T细胞反应而发挥抗肿瘤功效.
    NK-cell reactivity against cancer is conceivably suppressed in the tumor microenvironment by the interaction of the inhibitory receptor NKG2A with the non-classical MHC-I molecules HLA-E in humans or Qa-1b in mice. We found that intratumoral delivery of NK cells attains significant therapeutic effects only if co-injected with anti-NKG2A and anti-Qa-1b blocking monoclonal antibodies against solid mouse tumor models. Such therapeutic activity was contingent on endogenous CD8 T cells and type-1 conventional dendritic cells (cDC1). Moreover, the anti-tumor effects were enhanced upon combination with systemic anti-PD-1 mAb treatment and achieved partial abscopal efficacy against distant non-injected tumors. In xenografted mice bearing HLA-E-expressing human cancer cells, intratumoral co-injection of activated allogeneic human NK cells and clinical-grade anti-NKG2A mAb (monalizumab) synergistically achieved therapeutic effects. In conclusion, these studies provide evidence for the clinical potential of intratumoral NK cell-based immunotherapies that exert their anti-tumor efficacy as a result of eliciting endogenous T-cell 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
    背景:恶性黑色素瘤是一种罕见的人类和动物癌症,不仅发生在皮肤中,也发生在鼻子的粘膜中,嘴,肛门,消化道,在眼睛的葡萄膜(脉络膜)。为了开发治疗方法,我们需要更好地了解基因景观和信号通路。大量研究强调了NKG2A-HLA-E在肿瘤免疫治疗中的重要性。但HLA-E在肿瘤细胞中的功能和机制很少单独研究。来自可公开获得的数据库Oncomine和癌症基因组图谱(TCGA)的统计分析显示,与未转化的对应物相比,HLA-E在黑素瘤中高度表达。此外,HLA-E高表达的黑色素瘤患者的OS率低于低表达的患者。这些数据表明HLA-E在人黑素瘤中的重要性。Qa-1b是小鼠HLA-E的同源物。
    目的:探讨Qa-1b在小鼠黑色素瘤中的作用及机制。
    方法:用小鼠黑色素瘤细胞株B16-F10和同种异体黑色素瘤模型研究Qa-1b在黑色素瘤中的作用及机制。
    结果:与正常小鼠表皮细胞相比,Qa-1b在B16-F10中高表达。Qa-1b敲低抑制B16-F10细胞的体外增殖和迁移以及体内同种异体黑色素瘤过程。此外,Qa-1b敲低通过Ras-Raf-MAPK信号通路促进G0/G1期细胞周期阻滞和细胞凋亡。
    结论:Qa-1b具有致癌因子的作用,可作为黑色素瘤新的治疗靶点。
    BACKGROUND: Malignant melanoma is one kind of rare cancer in human and animals, which occurs not only in skin but also in the mucous membranes of the nose, mouth, anus, digestive tract, and in the uvea (choroid) of the eye. In order to develop therapies, a better understanding of the genetic landscape and signal pathways are needed. Numerous studies highlighted the importance of NKG2A-HLA-E in tumor immunotherapy, but the function and mechanism of HLA-E in tumor cells have seldom been studied alone. The statistical analyses from publicly available database Oncomine and The Cancer Genome Atlas (TCGA) showed that HLA-E is highly expressed in melanoma compared to non-transformed counterparts. In addition, melanoma patients with HLA-E high expression had a worse OS rate than the patients with low expression. These data indicate the importance of HLA-E in human melanoma. Qa-1b is the homolog of HLA-E in mouse.
    OBJECTIVE: To investigate the function and mechanism of Qa-1b in mouse melanoma.
    METHODS: Mouse melanoma cell line B16-F10 and allogenic melanoma model were used to investigate the function and mechanism of Qa-1b in melanoma.
    RESULTS: Qa-1b was highly expressed in B16-F10 compared with normal mouse epidermal cells. Qa-1b knockdown inhibited B16-F10 cell proliferation and migration in vitro and allogenic melanoma process in vivo. Furthermore, Qa-1b knockdown promoted cell cycle arrest at G0/G1 phase and cell apoptosis through Ras-Raf-MAPK signal pathway.
    CONCLUSIONS: Qa-1b functions as an oncogenic factor and it can be used as a new therapeutic target in melanoma.
    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
    免疫共受体CD8分子(CD8)有两个亚基,CD8α和CD8β,可以组装成同型或异二聚体。非经典(Ib类)主要组织相容性复合物(MHC)分子(MHC-Ib)最近已被鉴定为CD8αα同二聚体的配体。观察到组织相容性2,Q区基因座10(H2-Q10)是CD8αα的高亲和力配体,也与MHC-Ib分子H2-TL结合。这表明MHC-Ib蛋白可能是CD8αα配体的扩展来源。H2-T3/TL和H2-Q10的表达局限于小肠和肝脏,分别,然而,含有CD8αα的淋巴细胞存在更广泛。因此,在这里,我们试图确定鼠CD8αα是否仅与组织特异性MHC-Ib分子结合,还是与广泛表达的MHC-Ib分子结合.使用重组蛋白和基于表面等离子体共振的结合测定,我们显示MHC-Ib家族为小鼠CD8αα提供了多个结合配偶体,包括H2-T22和CD94/NKG2-A/B激活NK受体(NKG2)配体Qa-1b我们还证明了MHC-Ib蛋白之间关于CD8αα结合的层次结构,其中Qa-1b>H2-Q10>TL。最后,我们提供证据证明Qa-1b是CD8αα的功能性配体,将其与其人类同源MHCI类抗原E(HLA-E)区分开。这些发现提供了关于如何在不同组织中控制CD8αα-表达细胞的额外线索。他们还强调了Qa-1b/HLA-E功能的意想不到的免疫学差异,这表明需要对鼠MHC-Ib蛋白作为人类疾病模型进行更强有力的研究。
    The immune co-receptor CD8 molecule (CD8) has two subunits, CD8α and CD8β, which can assemble into homo or heterodimers. Nonclassical (class-Ib) major histocompatibility complex (MHC) molecules (MHC-Ibs) have recently been identified as ligands for the CD8αα homodimer. This was demonstrated by the observation that histocompatibility 2, Q region locus 10 (H2-Q10) is a high-affinity ligand for CD8αα which also binds the MHC-Ib molecule H2-TL. This suggests that MHC-Ib proteins may be an extended source of CD8αα ligands. Expression of H2-T3/TL and H2-Q10 is restricted to the small intestine and liver, respectively, yet CD8αα-containing lymphocytes are present more broadly. Therefore, here we sought to determine whether murine CD8αα binds only to tissue-specific MHC-Ib molecules or also to ubiquitously expressed MHC-Ib molecules. Using recombinant proteins and surface plasmon resonance-based binding assays, we show that the MHC-Ib family furnishes multiple binding partners for murine CD8αα, including H2-T22 and the CD94/NKG2-A/B-activating NK receptor (NKG2) ligand Qa-1b We also demonstrate a hierarchy among MHC-Ib proteins with respect to CD8αα binding, in which Qa-1b > H2-Q10 > TL. Finally, we provide evidence that Qa-1b is a functional ligand for CD8αα, distinguishing it from its human homologue MHC class I antigen E (HLA-E). These findings provide additional clues as to how CD8αα-expressing cells are controlled in different tissues. They also highlight an unexpected immunological divergence of Qa-1b/HLA-E function, indicating the need for more robust studies of murine MHC-Ib proteins as models for human disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The HLA-E homolog in the mouse (Qa-1b) is a conserved MHC class Ib molecule presenting monomorphic peptides to germline-encoded natural killer receptor CD94/NKG2A. Previously, we demonstrated the replacement of this canonical peptide by a diverse peptidome upon deficiency of the TAP peptide transporter. Analysis of this Qa-1b-restricted T cell repertoire against these non-mutated neoantigens revealed characteristics of conventional hypervariable CD8+ T cells, but also of invariant T cell receptor (TCR)αβ T cells. A shared TCR Vα chain was used by this subset in combination with a variety of Vβ chains. The TCRs target peptide ligands that are conserved between mouse and man, like the identified peptide derived from the transcriptional cofactor Med15. The thymus selection was studied in a TCR-transgenic mouse and emerging naïve CD8+ T cells displayed a slightly activated phenotype, as witnessed by higher CD122 and Ly6C expression. Moreover, the Qa-1b protein was dispensable for thymus selection. Importantly, no self-reactivity was observed as reported for other MHC class Ib-restricted subsets. Naïve Qa-1b restricted T cells expanded, contracted, and formed memory cells in vivo upon peptide vaccination in a similar manner as conventional CD8+ T cells. Based on these data, the Qa-1b restricted T cell subset might be positioned closest to conventional CD8+ T cells of all MHC class Ib populations.
    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
    NK cells can reduce anti-viral T cell immunity during chronic viral infections, including infection with the lymphocytic choriomeningitis virus (LCMV). However, regulating factors that maintain the equilibrium between productive T cell and NK cell immunity are poorly understood. Here, we show that a large viral load resulted in inhibition of NK cell activation, which correlated with increased expression of Qa-1b, a ligand for inhibitory NK cell receptors. Qa-1b was predominantly upregulated on B cells following LCMV infection, and this upregulation was dependent on type I interferons. Absence of Qa-1b resulted in increased NK cell-mediated regulation of anti-viral T cells following viral infection. Consequently, anti-viral T cell immunity was reduced in Qa-1b- and NKG2A-deficient mice, resulting in increased viral replication and immunopathology. NK cell depletion restored anti-viral immunity and virus control in the absence of Qa-1b. Taken together, our findings indicate that lymphocytes limit NK cell activity during viral infection in order to promote anti-viral T cell immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号