NLRC5

NLRC5
  • 文章类型: Journal Article
    NLRC5,核苷酸结合和寡聚化结构域(NOD)样受体(NLR)家族的最大成员,已报道参与免疫功能的调节,并与慢性炎症性疾病有关。然而,NLRC5在肝细胞癌(HCC)中的生物学功能尚未得到充分证实。这项研究的目的是评估NLRC5在接受手术治疗的HCC患者肿瘤组织中的表达,评估其预后价值,并探讨其与肿瘤微环境中关键免疫相关分子的关系。共有100例接受手术治疗的乙型肝炎病毒相关HCC患者参加了研究。通过对细胞染色的强度和组织切片中阳性细胞的百分比进行评分来获得免疫组织化学结果。采用卡方检验方法分析NLRC5表达水平与主要临床病理因素的相关性。采用COX回归模型和Kaplan-Meier生存曲线分析预后价值。进行受试者工作特征(ROC)曲线分析以评估NLRC5在HCC术后患者中的预测性能。IHC显示在67%的HCC组织样品中观察到NLRC5的高表达。卡方检验显示NLRC5是与肿瘤数量相关的危险因素,卫星结节,和信封入侵。Kaplan-Meier生存曲线和COX生存分析显示,NLRC5的高表达与HCC患者的总生存期(OS)降低显著相关(HR=1.79,95%CI1.03-3.12,p=.041)。然而,单因素logistic回归分析显示,NLRC5与GZMB和CD8α呈正相关,提示其在HCC的免疫逃逸中发挥作用。ROC曲线分析显示,肿瘤数目、信封入侵,和NLRC5表达(曲线下面积=0.824,灵敏度=77.30%,特异性=82.4%)与仅肿瘤数量和包膜侵犯的组合(曲线下面积=0.690,敏感性=43.9%,特异性=94.1%)。NLRC5在HCC的进展中起着至关重要的作用,可以被认为是潜在的预后和预测生物标志物。靶向NLRC5可能为HCC提供有吸引力的治疗方法。
    NLRC5, the largest member of the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family, has been reported to participate in the regulation of immune function and is associated with chronic inflammatory diseases. However, the biological function of NLRC5 in hepatocellular carcinoma (HCC) has not been fully demonstrated. The aim of this study is to evaluate NLRC5 expression in the tumor tissues of HCC patients undergoing surgical treatment, assess its prognostic value, and explore its relationship with critical immune-related molecules within the tumor microenvironment. A total of 100 patients with hepatitis B virus-associated HCC receiving surgical treatment were enrolled in the study. Immunohistochemical results were obtained by scoring the intensity of cellular staining and the percentage of positive cells in the tissue sections. The association between NLRC5 expression levels and the main clinicopathological factors was analyzed by Chi-square test method. The prognostic values were analyzed by COX regression model and the Kaplan-Meier survival curve. Receiver operating characteristic (ROC) curve analysis was performed to assess the predictive performance of NLRC5 in postoperative patients with HCC. IHC showed that high expression of NLRC5 was observed in 67% of HCC tissue samples. Chi-square test showed that NLRC5 was a risk factor associated with tumor number, satellite nodule, and envelope invasion. Kaplan-Meier survival curves and COX survival analysis showed that high expression of NLRC5 was significantly associated with decreased overall survival (OS) in HCC patients (HR = 1.79, 95% CI 1.03-3.12, p = .041). However, univariate logistic regression analysis revealed that NLRC5 showed positive relationship with GZMB and CD8α suggesting its role in immune escape of HCC. ROC curve analysis showed that the combination of tumor number, envelope invasion, and NLRC5 expression (area under the curve = 0.824, sensitivity = 77.30%, specificity = 82.4%) can more accurately evaluate the prognosis of HCC patients compared to the combination of only tumor number and envelope invasion (area under the curve = 0.690, sensitivity = 43.9%, specificity = 94.1%).NLRC5 plays a crucial role in progression of HCC and can be considered as a potential prognostic and predictive biomarker. Targeting NLRC5 may provide an attractive therapeutic approach for HCC.
    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:颞下颌关节骨关节炎(TMJOA)是一种退行性软骨疾病。17β-雌二醇(E2)加重TMJOA的病理过程;然而,其作用机制尚未阐明。因此,我们研究了E2对滑膜细胞生物学行为的影响及其分子机制。
    方法:用TNF-α处理大鼠原代成纤维样滑膜细胞,建立细胞模型,和表型使用细胞计数试剂盒-8,EdU,坦斯韦尔,酶联免疫吸附测定,和定量实时PCR(qPCR)。E2,FTO介导的NLRC5m6A甲基化的潜在机制,使用微阵列评估,甲基化RNA免疫沉淀,qPCR,和westernblot.此外,通过关节内注射碘乙酸钠(MIA)建立TMJOA样大鼠模型,采用显微CT和H&E染色评估骨形态和病理。
    结果:结果表明E2促进了增殖,迁移,入侵,和TNF-α处理的FLS的炎症。FTO表达在TMJOA中下调,在FLS中被E2降低。FTO的敲低促进了NLRC5的m6A甲基化并通过IGF2BP1识别增强了NLRC5的稳定性。此外,E2促进TMJ病理和髁突重塑,骨矿物质密度和骨小梁体积分数增加,通过NLRC5击倒获救。
    结论:E2促进了TMJOA的进展。
    BACKGROUND: Temporomandibular joint osteoarthritis (TMJOA) is a degenerative cartilage disease. 17β-estradiol (E2) aggravates the pathological process of TMJOA; however, the mechanisms of its action have not been elucidated. Thus, we investigate the influence of E2 on the cellular biological behaviors of synoviocytes and the molecular mechanisms.
    METHODS: Primary fibroblast-like synoviocytes (FLSs) isolated from rats were treated with TNF-α to establish cell model, and phenotypes were evaluated using cell counting kit-8, EdU, Tanswell, enzyme-linked immunosorbent assay, and quantitative real-time PCR (qPCR). The underlying mechanism of E2, FTO-mediated NLRC5 m6A methylation, was assessed using microarray, methylated RNA immunoprecipitation, qPCR, and western blot. Moreover, TMJOA-like rat model was established by intra-articular injection of monosodium iodoacetate (MIA), and bone morphology and pathology were assessed using micro-CT and H&E staining.
    RESULTS: The results illustrated that E2 facilitated the proliferation, migration, invasion, and inflammation of TNF-α-treated FLSs. FTO expression was downregulated in TMJOA and was reduced by E2 in FLSs. Knockdown of FTO promoted m6A methylation of NLRC5 and enhanced NLRC5 stability by IGF2BP1 recognition. Moreover, E2 promoted TMJ pathology and condyle remodeling, and increased bone mineral density and trabecular bone volume fraction, which was rescued by NLRC5 knockdown.
    CONCLUSIONS: E2 promoted the progression of TMJOA.
    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
    核苷酸结合和寡聚化结构域样受体(NLR)NLR家族含有CARD结构域的蛋白5(NLRC5)和II类主要组织相容性复合物反式激活因子(CIITA)是主要组织相容性复合物(MHC)I类和II类基因的转录调节因子,分别。MHC分子是我们免疫系统的核心角色,允许检测危险的“非自身”抗原,因此,从生物体中识别和消除感染或转化的细胞。最近,CIITA和NLRC5已成为感兴趣地位于延伸的MHC基因座中的嗜丁基酶(BTN)家族的选定基因的调节因子。BTN是与B7家族共调节分子表现出结构相似性的跨膜蛋白。家族成员BTN2A2确实有助于控制T细胞活化,被发现受CIITA转录调控。相反,NLRC5作为BTN3A1、BTN3A2和BTN3A3基因的重要调节因子出现。与BTN2A1一起,BTN3As调节由微生物来源或在血液癌细胞中积累的选定代谢物触发的非常规Vγ9Vδ2T细胞反应。即使内源性代谢物符合“自我”的规范定义,代谢异常的细胞可能对生物体构成危险,应该由免疫系统细胞识别和控制。总的来说,关于NLRC5在BTN3As表达中的作用的新数据将调节典型的“非自身”呈现的机制与那些标记具有异常代谢构型的细胞进行免疫识别的机制联系起来,我们在这篇透视综述中讨论的进化平行。
    The nucleotide-binding and oligomerization domain-like receptors (NLRs) NLR family CARD domain-containing protein 5 (NLRC5) and Class II Major Histocompatibility Complex Transactivator (CIITA) are transcriptional regulators of major histocompatibility complex (MHC) class I and class II genes, respectively. MHC molecules are central players in our immune system, allowing the detection of hazardous \'non-self\' antigens and, thus, the recognition and elimination of infected or transformed cells from the organism. Recently, CIITA and NLRC5 have emerged as regulators of selected genes of the butyrophilin (BTN) family that interestingly are located in the extended MHC locus. BTNs are transmembrane proteins exhibiting structural similarities to B7 family co-modulatory molecules. The family member BTN2A2, which indeed contributes to the control of T cell activation, was found to be transcriptionally regulated by CIITA. NLRC5 emerged instead as an important regulator of the BTN3A1, BTN3A2, and BTN3A3 genes. Together with BTN2A1, BTN3As regulate non-conventional Vγ9Vδ2 T cell responses triggered by selected metabolites of microbial origin or accumulating in hematologic cancer cells. Even if endogenous metabolites conform to the canonical definition of \'self\', metabolically abnormal cells can represent a danger for the organism and should be recognized and controlled by immune system cells. Collectively, new data on the role of NLRC5 in the expression of BTN3As link the mechanisms regulating canonical \'non-self\' presentation and those marking cells with abnormal metabolic configurations for immune recognition, an evolutionary parallel that we discuss in this perspective review.
    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
    宫颈癌(CC)是全球女性中第四大最常见的癌症。含NLR家族CARD结构域5(NLRC5)在肿瘤发生中起重要作用。然而,其在CC中的作用和机制尚不清楚。在这项研究中,我们旨在研究NLRC5在CC中的功能。与正常宫颈组织相比,发现NLRC5在CC组织中下调。然而,NLRC5表达较高的患者预后较好,年龄较高的患者,HPV感染,淋巴结转移,复发和组织学分级预后较差。单变量和多变量分析显示NLRC5是CC的潜在预后指标。Pearson相关分析显示NLRC5可能通过自噬相关蛋白在CC中发挥其功能,尤其是LC3.体外实验表明,NLRC5抑制LC3水平,促进细胞增殖,迁移,通过激活PI3K/AKT信号通路实现CC细胞的侵袭。用LY294002处理逆转了上述表型。一起来看,我们的发现提示,NLRC5通过调节PI3K/AKT信号通路参与宫颈肿瘤发生和进展.此外,NLRC5和LC3组合为CC中的可能预测因子。
    Cervical cancer (CC) is the fourth most common cancer among women worldwide. NLR Family CARD Domain Containing 5 (NLRC5) plays an important role in tumorigenesis. However, its effect and mechanism in CC remains unclear. In this study, we aimed to investigate the function of NLRC5 in CC. NLRC5 was found to be down-regulated in CC tissues compared with normal cervical tissues. However, patients with higher NLRC5 expression had better prognosis, patients with higher age, HPV infection, lymph node metastasis, recurrence and histological grade had worse prognosis. Univariate and multivariate analyses showed NLRC5 to be a potential prognostic indicator for CC. Pearson correlation analysis showed that NLRC5 might exert its function in CC through autophagy related proteins, especially LC3. In vitro experiments demonstrated that NLRC5 inhibited LC3 levels and promoted the proliferation, migration, and invasion of CC cells by activating the PI3K/AKT signaling pathway. Treatment with LY294002 reversed the above phenotype. Taken together, our finding suggested that NLRC5 would participate in cervical tumorigenesis and progression by regulating PI3K/AKT signaling pathway. In addition, NLRC5 and LC3 combined as possible predictors in CC.
    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
    NLR构成了一个很大的,对健康和疾病至关重要的高度保守的胞浆模式识别受体家族,使它们成为关键的治疗靶点。NLRC5是一种神秘的NLR,具有与炎症和传染病相关的突变,但对其作为先天免疫传感器和细胞死亡调节因子的功能知之甚少。因此,我们筛查了NLRC5在感染反应中的作用,PAMPs,DAMPs,和细胞因子。我们发现NLRC5作为驱动炎性细胞死亡的先天免疫传感器,全角下垂,响应特定的配体,包括PAMP/血红素和血红素/细胞因子组合。NLRC5与NLRP12和PANphosome组件相互作用形成细胞死亡复合物,这表明NLR网络的形式与植物中的类似。机械上,TLR信号和NAD+水平调节NLRC5表达和ROS产生以控制细胞死亡。此外,NLRC5缺陷小鼠在溶血和炎症模型中受到保护,表明NLRC5可能是一个潜在的治疗靶点。
    NLRs constitute a large, highly conserved family of cytosolic pattern recognition receptors that are central to health and disease, making them key therapeutic targets. NLRC5 is an enigmatic NLR with mutations associated with inflammatory and infectious diseases, but little is known about its function as an innate immune sensor and cell death regulator. Therefore, we screened for NLRC5\'s role in response to infections, PAMPs, DAMPs, and cytokines. We identified that NLRC5 acts as an innate immune sensor to drive inflammatory cell death, PANoptosis, in response to specific ligands, including PAMP/heme and heme/cytokine combinations. NLRC5 interacted with NLRP12 and PANoptosome components to form a cell death complex, suggesting an NLR network forms similar to those in plants. Mechanistically, TLR signaling and NAD+ levels regulated NLRC5 expression and ROS production to control cell death. Furthermore, NLRC5-deficient mice were protected in hemolytic and inflammatory models, suggesting that NLRC5 could be a potential therapeutic target.
    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
    MHC(主要组织相容性复合体)I类分子通过将抗原呈递至CD8T细胞而在调节适应性免疫系统中起重要作用。CITA(MHCI类反式激活因子),也称为NLRC5(NLR家族,包含CARD域的5),调节MHCI类和参与MHCI类抗原呈递途径的必需成分的表达。虽然NLRC5的核分布在其反式激活活性中的关键作用是已知的,确定NLRC5核定位的调节机制仍然知之甚少。在这项研究中,对NLRC5所有领域的综合分析表明,NLRC5核进出口的监管机制并存,相互抵消。此外,GCN5(一般对照非阻遏5蛋白),HAT(组蛋白乙酰转移酶)的成员,被发现是将NLRC5保留在细胞核中的关键角色,从而有助于I类MHC的表达。因此,NLRC5的进口和出口之间的平衡已经成为MHCI类反式激活的额外调节机制,这将是治疗癌症和病毒感染疾病的潜在治疗靶点。
    Major histocompatibility complex (MHC) class I molecules play an essential role in regulating the adaptive immune system by presenting antigens to CD8 T cells. CITA (MHC class I transactivator), also known as NLRC5 (NLR family, CARD domain-containing 5), regulates the expression of MHC class I and essential components involved in the MHC class I antigen presentation pathway. While the critical role of the nuclear distribution of NLRC5 in its transactivation activity has been known, the regulatory mechanism to determine the nuclear localization of NLRC5 remains poorly understood. In this study, a comprehensive analysis of all domains in NLRC5 revealed that the regulatory mechanisms for nuclear import and export of NLRC5 coexist and counterbalance each other. Moreover, GCN5 (general control non-repressed 5 protein), a member of HATs (histone acetyltransferases), was found to be a key player to retain NLRC5 in the nucleus, thereby contributing to the expression of MHC class I. Therefore, the balance between import and export of NLRC5 has emerged as an additional regulatory mechanism for MHC class I transactivation, which would be a potential therapeutic target for the treatment of cancer and virus-infected diseases.
    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:内皮-间充质转化(EndMT)在动脉粥样硬化的病理进展过程中在内皮功能障碍中起关键作用;然而,其详细机制尚不清楚。在这里,我们探讨了EndMT中上游刺激因子1(USF1)在动脉粥样硬化过程中的生物学功能和机制.
    方法:在高脂饮食喂养的ApoE-/-小鼠和ox-LDL暴露的人脐静脉内皮细胞(HUVECs)中建立体内和体外动脉粥样硬化模型。斑块的形成,胶原蛋白和脂质沉积,并通过苏木精和伊红(HE)评估主动脉组织的形态学变化,Masson,油红O和Verhoeff-VanGieson(EVG)染色,分别。通过EndMT相关蛋白的表达水平测定EndMT。通过RT-qPCR和Western印迹检测靶分子表达。通过ELISA测量促炎细胞因子的释放。通过transwell和划痕测定检测HUVEC的迁移。通过双荧光素酶报告实验研究了分子机制,ChIP,和Co-IP测定。
    结果:USF1在动脉粥样硬化患者中表达上调。USF1敲低通过上调CD31和VE-Cadherin抑制EndMT,同时下调α-SMA和波形蛋白,从而抑制炎症,和在暴露于ox-LDL的HUVECs中的迁移。此外,USF1转录激活泛素特异性蛋白酶14(USP14),这促进了NLR家族CARD结构域含5(NLRC5)的去泛素化和上调,以及随后的Smad2/3途径激活。sh-USF1或sh-USP14对EndMT的抑制作用被USP14或NLRC5过表达部分逆转。最后,USF1敲低通过抑制小鼠EndMT延迟动脉粥样硬化进展。
    结论:我们的发现表明USF1/USP14/NLRC5轴通过促进EndMT对动脉粥样硬化发展的贡献,提供有效的治疗靶点。
    BACKGROUND: Endothelial-to-Mesenchymal Transformation (EndMT) plays key roles in endothelial dysfunction during the pathological progression of atherosclerosis; however, its detailed mechanism remains unclear. Herein, we explored the biological function and mechanisms of upstream stimulating factor 1 (USF1) in EndMT during atherosclerosis.
    METHODS: The in vivo and in vitro atherosclerotic models were established in high fat diet-fed ApoE-/- mice and ox-LDL-exposed human umbilical vein endothelial cells (HUVECs). The plaque formation, collagen and lipid deposition, and morphological changes in the aortic tissues were evaluated by hematoxylin and eosin (HE), Masson, Oil red O and Verhoeff-Van Gieson (EVG) staining, respectively. EndMT was determined by expression levels of EndMT-related proteins. Target molecule expression was detected by RT-qPCR and Western blotting. The release of pro-inflammatory cytokines was measured by ELISA. Migration of HUVECs was detected by transwell and scratch assays. Molecular mechanism was investigated by dual-luciferase reporter assay, ChIP, and Co-IP assays.
    RESULTS: USF1 was up-regulated in atherosclerosis patients. USF1 knockdown inhibited EndMT by up-regulating CD31 and VE-Cadherin, while down-regulating α-SMA and vimentin, thereby repressing inflammation, and migration in ox-LDL-exposed HUVECs. In addition, USF1 transcriptionally activated ubiquitin-specific protease 14 (USP14), which promoted de-ubiquitination and up-regulation of NLR Family CARD Domain Containing 5 (NLRC5) and subsequent Smad2/3 pathway activation. The inhibitory effect of sh-USF1 or sh-USP14 on EndMT was partly reversed by USP14 or NLRC5 overexpression. Finally, USF1 knockdown delayed atherosclerosis progression via inhibiting EndMT in mice.
    CONCLUSIONS: Our findings indicate the contribution of the USF1/USP14/NLRC5 axis to atherosclerosis development via promoting EndMT, which provide effective therapeutic targets.
    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
    上皮性卵巢癌(OC)是最致命的妇科恶性肿瘤之一,迫切需要新的治疗策略。大约60%的卵巢肿瘤显示主要组织相容性复合体I类(MHCI)的表达降低,强化免疫逃避机制,使免疫疗法无效。含有NOD样受体CARD结构域的5(NLRC5)转录调节MHCI基因和许多抗原呈递机制组分。因此,我们探索了NLRC5在OC中的治疗潜力。
    我们产生了过表达NLRC5的OC细胞以挽救MHCI表达和抗原呈递,然后评估了它们对PD-L1阻断和感染的细胞疫苗的反应能力。
    对微阵列数据集的分析显示,在OC患者中,NLRC5表达升高与生存期延长之间存在相关性;然而,在OC肿瘤微环境中几乎检测不到NLRC5。过表达NLRC5的OC细胞表现出较慢的肿瘤生长,并导致TME中白细胞的募集增加,CD4/CD8T细胞比率降低,T细胞的活化增加。来自外周血的免疫细胞,脾,脾当暴露于自体肿瘤相关抗原时,这些小鼠的腹水显示出增强的激活和干扰素-γ产生。最后,作为概念的证明,当用亲本肿瘤攻击时,与对照组相比,受感染的细胞疫苗平台内的NLRC5过表达增强应答并延长存活。
    这些发现为在“冷”肿瘤模型中利用NLRC5过表达通过增强内源性肿瘤抗原的呈递来增强肿瘤对T细胞识别和消除的易感性提供了令人信服的理由。这种方法有望改善OC中的抗肿瘤免疫反应。
    Epithelial ovarian cancer (OC) stands as one of the deadliest gynecologic malignancies, urgently necessitating novel therapeutic strategies. Approximately 60% of ovarian tumors exhibit reduced expression of major histocompatibility complex class I (MHC I), intensifying immune evasion mechanisms and rendering immunotherapies ineffective. NOD-like receptor CARD domain containing 5 (NLRC5) transcriptionally regulates MHC I genes and many antigen presentation machinery components. We therefore explored the therapeutic potential of NLRC5 in OC.
    We generated OC cells overexpressing NLRC5 to rescue MHC I expression and antigen presentation and then assessed their capability to respond to PD-L1 blockade and an infected cell vaccine.
    Analysis of microarray datasets revealed a correlation between elevated NLRC5 expression and extended survival in OC patients; however, NLRC5 was scarcely detected in the OC tumor microenvironment. OC cells overexpressing NLRC5 exhibited slower tumor growth and resulted in higher recruitment of leukocytes in the TME with lower CD4/CD8 T-cell ratios and increased activation of T cells. Immune cells from peripheral blood, spleen, and ascites from these mice displayed heightened activation and interferon-gamma production when exposed to autologous tumor-associated antigens. Finally, as a proof of concept, NLRC5 overexpression within an infected cell vaccine platform enhanced responses and prolonged survival in comparison with control groups when challenged with parental tumors.
    These findings provide a compelling rationale for utilizing NLRC5 overexpression in \"cold\" tumor models to enhance tumor susceptibility to T-cell recognition and elimination by boosting the presentation of endogenous tumor antigens. This approach holds promise for improving antitumoral immune responses in OC.
    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
    肿瘤细胞向T细胞呈递抗原所需的抗原加工机制(APM)成分在实体瘤中以低水平表达,构成免疫逃逸的重要机制。比大多数其他实体瘤多,头颈部鳞状细胞癌(HNSCC)细胞往往具有低APM表达,使他们对免疫检查点阻断和大多数其他形式的免疫疗法不敏感。在HNSCC,这种APM缺乏主要是由高水平的EGFR和SHP2引起的,导致STAT1的低表达和激活;然而,最近的研究表明,经常在HNSCCs中突变的p53,也可以发挥作用。在目前的研究中,我们旨在研究STAT1和p53在HNSCC细胞中各自调节APM组分表达的程度。我们发现,在缺乏功能性p53的细胞中,只要STAT1表达保持完整,干扰素-γ或DNA损伤化疗(顺铂)仍然可以诱导APM表达;当两个转录因子被敲低时,APM组分表达被废除。当我们通过挽救NLRC5的表达绕过这些缺陷途径时,APM表达也恢复了。这些结果表明,功能性STAT1和p53的双重丧失可能使HNSCC细胞无法加工和呈递抗原,但拯救下游NLRC5表达可能是恢复T细胞免疫治疗敏感性的有吸引力的策略.
    The antigen processing machinery (APM) components needed for a tumor cell to present an antigen to a T cell are expressed at low levels in solid tumors, constituting an important mechanism of immune escape. More than most other solid tumors, head and neck squamous cell carcinoma (HNSCC) cells tend to have low APM expression, rendering them insensitive to immune checkpoint blockade and most other forms of immunotherapy. In HNSCC, this APM deficiency is largely driven by high levels of EGFR and SHP2, leading to low expression and activation of STAT1; however, recent studies suggest that p53, which is often mutated in HNSCCs, may also play a role. In the current study, we aimed to investigate the extent to which STAT1 and p53 individually regulate APM component expression in HNSCC cells. We found that in cells lacking functional p53, APM expression could still be induced by interferon-gamma or DNA-damaging chemotherapy (cisplatin) as long as STAT1 expression remained intact; when both transcription factors were knocked down, APM component expression was abolished. When we bypassed these deficient pathways by rescuing the expression of NLRC5, APM expression was also restored. These results suggest that dual loss of functional STAT1 and p53 may render HNSCC cells incapable of processing and presenting antigens, but rescue of downstream NLRC5 expression may be an attractive strategy for restoring sensitivity to T cell-based immunotherapy.
    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
    Eribulin抑制微管聚合并改善复发转移性乳腺癌患者的总体生存率。亚组分析显示,低中性粒细胞与淋巴细胞比率(NLR)(<3)是eribulin治疗的预后因素。因此,我们假设eribulin可能与乳腺癌细胞的免疫反应有关,并分析了eribulin对免疫系统的影响。免疫组织化学染色显示,艾瑞布林治疗后,临床样品中人类白细胞抗原(HLA)I类表达增加。体外分析显示,eribulin治疗可增加乳腺癌细胞系细胞中HLAI类的表达。RNA测序表明,eribulin处理增加了NOD样家族CARD结构域-含5(NLRC5)的表达,HLAI类表达的主要调节因子。Eribulin治疗增加了纽约食管鳞状细胞癌1(NY-ESO-1)过表达乳腺癌细胞的NY-ESO-1特异性T细胞受体(TCR)转导的T(TCR-T)细胞反应。eribulin和TCR-T联合治疗模型显示eribulin和使用TCR-T细胞的免疫治疗具有协同作用。总之,eribulin通过HLA1类转录因子NLRC5增加HLA1类的表达,eribulin与免疫疗法联合可有效治疗乳腺癌。
    Eribulin inhibits microtubule polymerization and improves the overall survival of patients with recurrent metastatic breast cancer. A subgroup analysis revealed a low neutrophil to lymphocyte ratio (NLR) (<3) to be a prognostic factor of eribulin treatment. We thus hypothesized that eribulin might be related to the immune response for breast cancer cells and we analyzed the effects of eribulin on the immune system. Immunohistochemical staining revealed that human leukocyte antigen (HLA) class I expression was increased in clinical samples after eribulin treatment. In vitro assays revealed that eribulin treatment increased HLA class I expression in breast cancer line cells. RNA-sequencing demonstrated that eribulin treatment increased the expression of the NOD-like family CARD domain-containing 5 (NLRC5), a master regulator of HLA class I expression. Eribulin treatment increased the NY-ESO-1-specific T-cell receptor (TCR) transduced T (TCR-T) cell response for New York oesophageal squamous cell carcinoma 1 (NY-ESO-1) overexpressed breast cancer cells. The eribulin and TCR-T combined therapy model revealed that eribulin and immunotherapy using TCR-T cells has a synergistic effect. In summary, eribulin increases the expression of HLA class 1 via HLA class 1 transactivatior NLRC5 and eribulin combination with immunotherapy can be effective for the treatment of breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号