NK Cell Lectin-Like Receptor Subfamily K

NK 细胞凝集素样受体亚家族 K
  • 文章类型: Journal Article
    背景:激活和抑制自然杀伤(NK)细胞的受体,如NKp,NKG2或CLEC与冷肿瘤(包括成胶质细胞瘤(GBM))高度相关。这里,我们旨在表征这些受体在GBM中的表达,以深入了解它们作为肿瘤内微环境调节剂的潜在作用。
    方法:我们对几种NK受体进行了转录组学分析,重点是由KLRC2,NKG2C编码的激活受体,在批量和单细胞RNA测序GBM数据集之间。我们还评估了KLRC2过表达的GL261细胞在用或不用程序性细胞死亡蛋白-1(PD-1)单克隆抗体(mAb)处理的小鼠中的作用。最后,我们分析了两项评估PD-1mAb在GBM患者中作用的临床试验的样本,以确定NKG2C作为应答生物标志物的潜力.
    结果:我们观察到几种抑制性NK受体在GBM浸润NK和T细胞上的显著表达,这与KLRC2在肿瘤细胞上的强表达相反,主要在渗透边缘。肿瘤性KLRC2表达与骨髓来源的抑制细胞数量减少和肿瘤驻留淋巴细胞水平升高有关。在小鼠模型和GBM患者中,在表达NKG2Chigh的肿瘤中观察到PD-1mAb治疗后具有更强的抗肿瘤活性,而抑制性NK受体的表达显示出负相关。
    结论:本研究探讨了肿瘤NKG2C/KLRC2表达在塑造GBM免疫谱中的作用,并表明它是GBM患者对免疫检查点抑制剂治疗阳性反应的预测性生物标志物。未来的研究可以在前瞻性试验中进一步验证这一发现。
    BACKGROUND: Activating and inhibitory receptors of natural killer (NK) cells such as NKp, NKG2, or CLEC are highly relevant to cold tumors including glioblastoma (GBM). Here, we aimed to characterize the expression of these receptors in GBM to gain insight into their potential role as modulators of the intratumoral microenvironment.
    METHODS: We performed a transcriptomic analysis of several NK receptors with a focus on the activating receptor encoded by KLRC2, NKG2C, among bulk and single-cell RNA sequencing GBM data sets. We also evaluated the effects of KLRC2-overexpressing GL261 cells in mice treated with or without programmed cell death protein-1 (PD-1) monoclonal antibody (mAb). Finally, we analyzed samples from two clinical trials evaluating PD-1 mAb effects in patients with GBM to determine the potential of NKG2C to serve as a biomarker of response.
    RESULTS: We observed significant expression of several inhibitory NK receptors on GBM-infiltrating NK and T cells, which contrasts with the strong expression of KLRC2 on tumor cells, mainly at the infiltrative margin. Neoplastic KLRC2 expression was associated with a reduction in the number of myeloid-derived suppressor cells and with a higher level of tumor-resident lymphocytes. A stronger antitumor activity after PD-1 mAb treatment was observed in NKG2Chigh-expressing tumors both in mouse models and patients with GBM whereas the expression of inhibitory NK receptors showed an inverse association.
    CONCLUSIONS: This study explored the role of neoplastic NKG2C/KLRC2 expression in shaping the immune profile of GBM and suggests that it is a predictive biomarker for positive responses to immune checkpoint inhibitor treatment in patients with GBM. Future studies could further validate this finding in prospective trials.
    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
    免疫系统是监测和根除肿瘤细胞的组成部分。自然杀手组2成员D(NKG2D)受体及其配体(NKG2DL)之间的相互作用对于激活NKG2D受体阳性免疫细胞至关重要,比如自然杀伤细胞。这种激活使这些细胞能够识别和破坏呈NKG2DL的肿瘤细胞,这是肿瘤免疫的一个重要方面。然而,从肿瘤细胞表面脱落的可溶性NKG2DL(sNKG2DL)促进了肿瘤免疫逃逸。sNKG2DL的产生主要由金属蛋白酶[解整合素和金属蛋白酶(ADAM)和基质金属蛋白酶(MMP)家族]和外来体调节。sNKG2DL不仅减少了肿瘤细胞表面的免疫识别,而且抑制了免疫细胞的功能,如NK细胞,并降低NKG2D受体的表达。这个过程促进免疫逃避,programming,和肿瘤的转移。在这次审查中,对影响sNKG2DL产生的机制和因素及其对肿瘤微环境中免疫抑制的贡献进行了深入总结。此外,由于sNKG2DL与肿瘤进展和转移之间的显著联系,它们作为新型生物标志物具有巨大潜力。可通过液体活检发现,sNKG2DL可以评估肿瘤的恶性程度和预后,并作为免疫疗法的关键靶标。这可能导致新药的发现或现有治疗的增强。因此,探讨了sNKG2DLs在临床肿瘤学中的应用,为开发sNKG2DL的创新免疫治疗策略提供了大量的理论支持。
    The immune system is integral to the surveillance and eradication of tumor cells. Interactions between the natural killer group 2 member D (NKG2D) receptor and its ligands (NKG2DLs) are vital for activating NKG2D receptor‑positive immune cells, such as natural killer cells. This activation enables these cells to identify and destroy tumor cells presenting with NKG2DLs, which is an essential aspect of tumor immunity. However, tumor immune escape is facilitated by soluble NKG2DL (sNKG2DL) shed from the surface of tumor cells. The production of sNKG2DL is predominantly regulated by metalloproteinases [a disintegrin and metalloproteinases (ADAM) and matrix metalloproteinase (MMP) families] and exosomes. sNKG2DL not only diminish immune recognition on the tumor cell surface but also suppress the function of immune cells, such as NK cells, and reduce the expression of the NKG2D receptor. This process promotes immune evasion, progression, and metastasis of tumors. In this review, an in‑depth summary of the mechanisms and factors that influence sNKG2DL production and their contribution to immune suppression within the tumor microenvironment are provided. Furthermore, due to the significant link between sNKG2DLs and tumor progression and metastasis, they have great potential as novel biomarkers. Detectable via liquid biopsies, sNKG2DLs could assess tumor malignancy and prognosis, and act as pivotal targets for immunotherapy. This could lead to the discovery of new drugs or the enhancement of existing treatments. Thus, the application of sNKG2DLs in clinical oncology was explored, offering substantial theoretical support for the development of innovative immunotherapeutic strategies for sNKG2DLs.
    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:引发免疫细胞反应的癌症免疫治疗方法,包括T细胞和NK细胞,彻底改变了肿瘤学领域。然而,免疫抑制机制抑制实体瘤中的免疫细胞活化,因此需要其他策略来增强活性。
    方法:我们基于共刺激受体NKG2D在乳腺癌患者样本中的大部分CD8+肿瘤浸润淋巴细胞(TIL)上的表达,将其鉴定为靶标。结合NK和CD8+T细胞上的NKG2D以及乳腺癌细胞上的HER2(HER2-CRB)的人和鼠替代NKG2D共刺激受体双特异性(CRB)被开发为体外和体内靶向该信号传导轴的概念证明。
    结果:当与表达HER2的乳腺癌细胞系共培养时,HER2-CRB增强NK细胞活化和细胞因子产生。HER2-CRB与T细胞依赖性双特异性(TDB)抗体组合,该抗体通过将CD3与HER2(HER2-TDB)交联而合成激活T细胞,增强的T细胞毒性,细胞因子的产生和体内抗肿瘤活性。小鼠替代HER2-CRB(mHER2-CRB)改善了HER2-TDB的体内功效,并增强了NK和T细胞活化,细胞因子产生和效应CD8+T细胞分化。
    结论:我们证明了用双特异性抗体(BsAbs)靶向NKG2D是增强NK和CD8+T细胞抗肿瘤免疫应答的有效方法。鉴于正在进行的大量利用NK和T细胞进行癌症免疫治疗的临床试验,NKG2D-双特异性化合物具有广泛的组合潜力。
    BACKGROUND: Cancer immunotherapy approaches that elicit immune cell responses, including T and NK cells, have revolutionized the field of oncology. However, immunosuppressive mechanisms restrain immune cell activation within solid tumors so additional strategies to augment activity are required.
    METHODS: We identified the co-stimulatory receptor NKG2D as a target based on its expression on a large proportion of CD8+ tumor infiltrating lymphocytes (TILs) from breast cancer patient samples. Human and murine surrogate NKG2D co-stimulatory receptor-bispecifics (CRB) that bind NKG2D on NK and CD8+ T cells as well as HER2 on breast cancer cells (HER2-CRB) were developed as a proof of concept for targeting this signaling axis in vitro and in vivo.
    RESULTS: HER2-CRB enhanced NK cell activation and cytokine production when co-cultured with HER2 expressing breast cancer cell lines. HER2-CRB when combined with a T cell-dependent-bispecific (TDB) antibody that synthetically activates T cells by crosslinking CD3 to HER2 (HER2-TDB), enhanced T cell cytotoxicity, cytokine production and in vivo antitumor activity. A mouse surrogate HER2-CRB (mHER2-CRB) improved in vivo efficacy of HER2-TDB and augmented NK as well as T cell activation, cytokine production and effector CD8+ T cell differentiation.
    CONCLUSIONS: We demonstrate that targeting NKG2D with bispecific antibodies (BsAbs) is an effective approach to augment NK and CD8+ T cell antitumor immune responses. Given the large number of ongoing clinical trials leveraging NK and T cells for cancer immunotherapy, NKG2D-bispecifics have broad combinatorial potential.
    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
    自然杀伤细胞(NK)是肿瘤的“职业杀手”,在抗肿瘤免疫治疗中起着至关重要的作用。NK细胞脱敏是肿瘤免疫逃逸的一个症结机制。失调的NKG2D-NKG2DL信号传导是这种脱敏过程的主要驱动因素。然而,调节NK细胞脱敏的因素在很大程度上仍未表征。这里,我们首次报道了环状RNAcircARAP2(hsa_circ_0069396)参与NK细胞脱敏模型中可溶性MICA(sMICA)诱导的NKG2D内吞。在NK细胞脱敏过程中CircARAP2上调,CircARAP2的丢失减轻了NKG2D内吞和NK细胞脱敏。使用通过RNA纯化(ChIRP)和RNA下拉方法分离染色质,我们发现RAB5A,早期内体的分子标记,是它的下游目标。值得注意的是,转录因子CTCF是circARAP2的中间功能伴侣.机械上,我们发现circARAP2与CTCF相互作用并抑制CTCF-Polycomb阻遏复合物2(PRC2)向RAB5A启动子区的募集,从而消除组蛋白H3K27和H3K9甲基化抑制以增强RAB5A转录。这些数据表明,circARAP2的抑制有效缓解sMICA诱导的NKG2D内吞和NK细胞脱敏,为肿瘤免疫逃避的治疗干预提供了新的靶点。
    Natural killer cells (NK) are the \"professional killer\" of tumors and play a crucial role in anti-tumor immunotherapy. NK cell desensitization is a key mechanism of tumor immune escape. Dysregulated NKG2D-NKG2DL signaling is a primary driver of this desensitization process. However, the factors that regulate NK cell desensitization remain largely uncharacterized. Here, we present the first report that circular RNA circARAP2 (hsa_circ_0069396) is involved in the soluble MICA (sMICA)-induced NKG2D endocytosis in the NK cell desensitization model. CircARAP2 was upregulated during NK cell desensitization and the loss of circARAP2 alleviated NKG2D endocytosis and NK cell desensitization. Using Chromatin isolation by RNA purification (ChIRP) and RNA pull-down approaches, we identified that RAB5A, a molecular marker of early endosomes, was its downstream target. Notably, transcription factor CTCF was an intermediate functional partner of circARAP2. Mechanistically, we discovered that circARAP2 interacted with CTCF and inhibited the recruitment of CTCF-Polycomb Repressive Complex 2 (PRC2) to the promoter region of RAB5A, thereby erasing histone H3K27 and H3K9 methylation suppression to enhance RAB5A transcription. These data demonstrate that inhibition of circARAP2 effectively alleviates sMICA-induced NKG2D endocytosis and NK cell desensitization, providing a novel target for therapeutic intervention in tumor immune evasion.
    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
    子宫内膜异位症,影响了10%的女性,被定义为植入,生存,和子宫腔外子宫内膜样/子宫内膜异位组织的生长,引起炎症,不孕症,疼痛,和卵巢癌的易感性。尽管进行了广泛的研究,其病因和发病机制了解甚少,而且基本上不清楚。普遍的观点是,子宫内膜异位症患者的免疫系统无法从逆行月经中清除异位播散的子宫内膜。外来体是表现出免疫调节特性的小细胞外囊泡。我们研究了子宫内膜异位组织分泌的外泌体在子宫内膜异位症病理生理学中的作用。研究了已知会损害免疫反应的两种外泌体介导的机制:1)NKG2D介导的细胞毒性的下调,以及2)FasL和TRAIL诱导的活化免疫细胞凋亡。我们表明,从短期外植体培养上清液中分离出的分泌的子宫内膜异位症外泌体在其表面携带NKG2D配体MICA/B和ULBP1-3以及促凋亡分子FasL和TRAIL,即,外泌体介导的免疫抑制的特征分子。充当诱饵,这些外泌体下调NKG2D受体,削弱NKG2D介导的细胞毒性,并通过FasL-和TRAIL途径诱导活化的PBMC和Jurkat细胞凋亡。分泌的子宫内膜异位症外泌体在异位部位产生免疫抑制梯度,在子宫内膜异位病变周围形成“保护盾”。这种梯度保护子宫内膜异位病变免受细胞毒性攻击的清除,并通过诱导激活的免疫细胞凋亡而产生免疫特权。一起来看,我们的结果提供了一个合理的,基于外泌体的机制解释了子宫内膜异位症中的免疫功能障碍和受损的免疫监视,并为这种神秘疾病的发病机制提供了新的见解。
    Endometriosis, affecting 10% of women, is defined as implantation, survival, and growth of endometrium-like/endometriotic tissue outside the uterine cavity, causing inflammation, infertility, pain, and susceptibility to ovarian cancer. Despite extensive studies, its etiology and pathogenesis are poorly understood and largely unknown. The prevailing view is that the immune system of endometriosis patients fails to clear ectopically disseminated endometrium from retrograde menstruation. Exosomes are small extracellular vesicles that exhibit immunomodulatory properties. We studied the role of endometriotic tissue-secreted exosomes in the pathophysiology of endometriosis. Two exosome-mediated mechanisms known to impair the immune response were investigated: 1) downregulation of NKG2D-mediated cytotoxicity and 2) FasL- and TRAIL-induced apoptosis of activated immune cells. We showed that secreted endometriotic exosomes isolated from supernatants of short-term explant cultures carry the NKG2D ligands MICA/B and ULBP1-3 and the proapoptotic molecules FasL and TRAIL on their surface, i.e., signature molecules of exosome-mediated immune suppression. Acting as decoys, these exosomes downregulate the NKG2D receptor, impair NKG2D-mediated cytotoxicity, and induce apoptosis of activated PBMCs and Jurkat cells through the FasL- and TRAIL pathway. The secreted endometriotic exosomes create an immunosuppressive gradient at the ectopic site, forming a \"protective shield\" around the endometriotic lesions. This gradient guards the endometriotic lesions against clearance by a cytotoxic attack and creates immunologic privilege by induction of apoptosis in activated immune cells. Taken together, our results provide a plausible, exosome-based mechanistic explanation for the immune dysfunction and the compromised immune surveillance in endometriosis and contribute novel insights into the pathogenesis of this enigmatic disease.
    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
    人疱疹病毒6B(HHV-6B)通过下调I类MHC分子(MHC-I)来阻止宿主免疫反应,阻碍抗原呈递到CD8+T细胞。MHC-I的下调释放自然杀伤(NK)细胞上的抑制性受体,如果NK细胞活化受体如NKG2D已经参与在靶细胞上上调的应激配体,则导致靶细胞的活化和杀伤。以前的工作表明,HHV-6B下调三个MHC样应激配体MICB,ULBP1和ULBP3,由NKG2D识别。相关病毒HHV-6A的U20糖蛋白与ULBP1的下调有关,但确切的机制仍未确定。
    我们着手研究HHV-6BU20在调节NK细胞活性中的作用。我们使用HHV-6BU20表达为重组蛋白或转导到靶细胞中,以及HHV-6B感染,研究与NK细胞配体和受体的结合相互作用,并评估对NK细胞活化的影响。小角度X射线散射用于对齐从机器学习方法导出的分子模型。
    我们证明U20以亚微摩尔亲和力直接结合ULBP1。U20的转导降低了NKG2D在细胞表面与ULBP1的结合,但不降低ULBP1蛋白水平。在细胞表面或在toto中。HHV-6B感染和可溶性U20具有相同的作用。U20的转导阻断响应于细胞表面ULBP1的NK细胞活化。U20-ULBP1复合物的结构模型表明与m152-RAE1γ复合物有一些相似之处。
    UNASSIGNED: Human Herpesvirus 6B (HHV-6B) impedes host immune responses by downregulating class I MHC molecules (MHC-I), hindering antigen presentation to CD8+ T cells. Downregulation of MHC-I disengages inhibitory receptors on natural killer (NK) cells, resulting in activation and killing of the target cell if NK cell activating receptors such as NKG2D have engaged stress ligands upregulated on the target cells. Previous work has shown that HHV-6B downregulates three MHC-like stress ligands MICB, ULBP1, and ULBP3, which are recognized by NKG2D. The U20 glycoprotein of the related virus HHV-6A has been implicated in the downregulation of ULBP1, but the precise mechanism remains undetermined.
    UNASSIGNED: We set out to investigate the role of HHV-6B U20 in modulating NK cell activity. We used HHV-6B U20 expressed as a recombinant protein or transduced into target cells, as well as HHV-6B infection, to investigate binding interactions with NK cell ligands and receptors and to assess effects on NK cell activation. Small-angle X-ray scattering was used to align molecular models derived from machine-learning approaches.
    UNASSIGNED: We demonstrate that U20 binds directly to ULBP1 with sub-micromolar affinity. Transduction of U20 decreases NKG2D binding to ULBP1 at the cell surface but does not decrease ULBP1 protein levels, either at the cell surface or in toto. HHV-6B infection and soluble U20 have the same effect. Transduction of U20 blocks NK cell activation in response to cell-surface ULBP1. Structural modeling of the U20 - ULBP1 complex indicates some similarities to the m152-RAE1γ complex.
    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
    持续的人乳头瘤病毒感染与最终导致宫颈癌的癌前病变的发展有关。在这项研究中,我们评估了激活的表达(NKG2D,DNAM-1)和抑制性免疫检查点受体(PD-1,TIGIT,和Tim-3)在宫颈癌患者外周血NKT样(CD3CD56)淋巴细胞中(CC,n=19),高级别病变(HG,n=8),低度病变(LG,n=19)和健康捐赠者(HD,n=17)使用多参数流式细胞术。维度数据分析显示CD3+CD56+细胞内的四个簇具有不同的受体表达模式。我们观察到其中一组CC和HG患者中CD16的上调。在另一个,TIGIT被上调,而DNAM-1下调。在整个手动门控过程中,我们观察到表达激活受体的NKT样细胞也共表达抑制性受体(PD-1和TIGIT),会影响这些细胞的激活。对细胞功能状态的更深入表征可能有助于阐明它们在宫颈癌中的作用,将NKT样细胞表征为细胞毒性CD8+T细胞或I型或II型NKT细胞的成员。
    Persistent human papillomavirus infection is associated with the development of premalignant lesions that can eventually lead to cervical cancer. In this study, we evaluated the expression of activating (NKG2D, DNAM-1) and inhibitory immune checkpoints receptors (PD-1, TIGIT, and Tim-3) in peripheral blood NKT-like (CD3+CD56+) lymphocytes from patients with cervical carcinoma (CC, n = 19), high-grade lesions (HG, n = 8), low-grade lesions (LG, n = 19) and healthy donors (HD, n = 17) using multiparametric flow cytometry. Dimensional data analysis showed four clusters within the CD3+CD56+ cells with different patterns of receptor expression. We observed upregulation of CD16 in CC and HG patients in one of the clusters. In another, TIGIT was upregulated, while DNAM-1 was downregulated. Throughout manual gating, we observed that NKT-like cells expressing activating receptors also co-express inhibitory receptors (PD-1 and TIGIT), which can affect the activation of these cells. A deeper characterization of the functional state of the cells may help to clarify their role in cervical cancer, as will the characterization of the NKT-like cells as cytotoxic CD8+ T cells or members of type I or type II NKT cells.
    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)细胞的短命性质和异质性限制了基于NK细胞的疗法的发展。尽管它们被证明具有抗癌的安全性和有效性。这里,我们描述了生物学基础,长寿命抗肿瘤人NK细胞(CD56highCD16+)的详细表型和功能,在没有细胞分选或饲养细胞的情况下获得,在用卡介苗(BCG)引发外周血细胞后。Further,我们证明了细胞因子组合的存活剂量,不包括IL18,每周给予BCG引发的NK细胞,避免先天淋巴细胞耗尽,并导致先天细胞的特定长期增殖,对广泛的实体瘤发挥有效的细胞毒性功能,主要通过NKG2D。引人注目的是,在BCG和细胞因子刺激后,NKG2C+CD57-FcεRIγ+NK细胞群扩增,独立于HCMV血清学。这一策略被用来拯救抗肿瘤NK细胞,甚至来自癌症患者骨髓的抑制环境。证明BCG赋予NK细胞持久的抗肿瘤特征。
    The short-lived nature and heterogeneity of Natural Killer (NK) cells limit the development of NK cell-based therapies, despite their proven safety and efficacy against cancer. Here, we describe the biological basis, detailed phenotype and function of long-lived anti-tumour human NK cells (CD56highCD16+), obtained without cell sorting or feeder cells, after priming of peripheral blood cells with Bacillus Calmette-Guérin (BCG). Further, we demonstrate that survival doses of a cytokine combination, excluding IL18, administered just weekly to BCG-primed NK cells avoids innate lymphocyte exhaustion and leads to specific long-term proliferation of innate cells that exert potent cytotoxic function against a broad range of solid tumours, mainly through NKG2D. Strikingly, a NKG2C+CD57-FcεRIγ+ NK cell population expands after BCG and cytokine stimulation, independently of HCMV serology. This strategy was exploited to rescue anti-tumour NK cells even from the suppressor environment of cancer patients\' bone marrow, demonstrating that BCG confers durable anti-tumour features to NK 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
    目的:自然杀伤(NK)细胞是肿瘤微环境中对抗恶性肿瘤的坚定防线的组成部分。现有研究表明,miRNA可以通过负调控基因表达来影响NK细胞的发育。在这项研究中,我们旨在探讨miR-17-5p在肝细胞癌(HCC)外泌体中如何通过转录因子RNX1调节NK细胞对HCC细胞的杀伤功能.
    方法:从肝癌组织和细胞系中分离外泌体,然后进行第二代测序以比较差异miRNA。使用qRT-PCR和Western印迹方法进行验证。miR-17-5p与RUNX1之间以及RUNX1与NKG2D之间的相互作用,使用荧光素酶报告基因测定等技术进行认证,西方印迹,和染色质免疫沉淀(ChIP)。使用诸如RTCA和ELISPOT的方法测量NK细胞在体外对HCC细胞的细胞毒活性。斑马鱼异种移植用于评估NK细胞对HCC细胞的体内杀伤能力。
    结果:与癌旁组织相比,HCC组织外泌体中miR-17-5p的水平升高。我们证实RUNX1是miR-17-5p的靶标,并且RUNX1增强NKG2D的转录。发现MiR-17-5p下调RUNX1和NKG2D的表达,随后降低NK细胞对HCC细胞的体外和体内细胞毒性能力。
    结论:在HCC外泌体中发现的miR-17-5p可以靶向RUNX1,随后减弱NK细胞的细胞毒活性。
    OBJECTIVE: Natural killer (NK) cells are an integral part of the staunch defense line against malignant tumors within the tumor microenvironment. Existing research indicates that miRNAs can influence the development of NK cells by negatively modulating gene expression. In this study, we aim to explore how the miR-17-5p in Hepatocellular Carcinoma (HCC) exosomes regulates the killing function of NK cells towards HCC cells through the transcription factor RNX1.
    METHODS: The exosomes were isolated from HCC tissues and cell lines, followed by a second generation sequencing to compare differential miRNAs. Verification was performed using qRT-PCR and Western blot methods. The mutual interactions between miR-17-5p and RUNX1, as well as between RUNX1 and NKG2D, were authenticated using techniques like luciferase reporter gene assays, Western blotting, and Chromatin Immunoprecipitation (ChIP). The cytotoxic activity of NK cells towards HCC cells in vitro was measured using methods such as RTCA and ELISPOT. The zebrafish xenotransplantation was utilized to assess the in vivo killing capacity of NK cells against HCC cells.
    RESULTS: The level of miR-17-5p in exosomes from HCC tissue increased compared to adjacent tissues. We verified that RUNX1 was a target of miR-17-5p and that RUNX1 enhances the transcription of NKG2D. MiR-17-5p was found to downregulate the expression of RUNX1 and NKG2D, subsequently reducing the in vitro and in vivo cytotoxic capabilities of NK cells against HCC cells.
    CONCLUSIONS: The miR-17-5p found within HCC exosomes can target RUNX1, subsequently attenuating the cytotoxic activity of NK cells.
    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
    在我们之前的研究中,开发了一种新型低温疗法(CTT)以诱导全身长期抗肿瘤免疫。发现自然杀伤(NK)细胞在CTT后晚期CTT诱导的长期免疫介导的肿瘤控制中起重要作用,但潜在的机制尚不清楚。骨髓来源的抑制细胞(MDSC)是未成熟的骨髓细胞,对T细胞具有有效的免疫抑制作用,并削弱了免疫治疗的长期益处。因此,克服MDSC免疫抑制对于维持免疫治疗的长期疗效至关重要。在这项研究中,我们发现NK细胞在CTT后的晚期显著减少MDSC的积累,促进T细胞生产,增加T细胞活化,促进MDSC成熟,最终导致以Th1为主的CD4+T细胞分化并增强NK和CD8+T细胞的细胞毒性。此外,NK细胞通过NKG2D-配体相互作用激活MDSCs中的ERK信号,以增加肿瘤坏死因子(TNF)-α转化酶(TACE)裂解膜TNF-α的活性。此外,增加的TACE活性从MDSC释放更多的可溶性TNF-α以促进MDSC成熟。在我们的研究中,我们提出了一种新的机制,通过该机制NK细胞可以克服MDSC诱导的免疫抑制并维持CTT诱导的持续抗肿瘤免疫,提供了一个前瞻性的治疗选择,以提高癌症免疫治疗的性能。
    In our previous studies, a novel cryothermal therapy (CTT) was developed to induce systemic long-term anti-tumor immunity. Natural killer (NK) cells were found to play an important role in CTT-induced long-term immune-mediated tumor control at the late stage after CTT, but the underlying mechanism is unclear. Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that have potent immunosuppressive effects on T cells and weaken the long-term benefits of immunotherapy. Consequently, overcoming MDSC immunosuppression is essential for maintaining the long-term efficacy of immunotherapy. In this study, we revealed that NK cells considerably diminish MDSC accumulation at the late stage after CTT, boost T cell production, increase T cell activation, and promote MDSC maturation, culminating in Th1-dominant CD4+ T cell differentiation and enhancing NK and CD8+ T cell cytotoxicity. Additionally, NK cells activate ERK signaling in MDSCs through NKG2D-ligand interaction to increase the activity of tumor necrosis factor (TNF)-α converting enzyme (TACE)-cleaved membrane TNF-α. Furthermore, Increased TACE activity releases more soluble TNF-α from MDSCs to promote MDSC maturation. In our studies, we propose a novel mechanism by which NK cells can overcome MDSC-induced immunosuppression and maintain CTT-induced persistent anti-tumor immunity, providing a prospective therapeutic option to improve the performance of cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号