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:本研究旨在探讨ataxin7(circleRNAcircuatXN7)通过microRNA(miR)-4319/NLR家族CARD结构域5(NLRC5)调控食管癌(EC)细胞增殖和侵袭的分子机制。
    方法:通过RNA荧光原位杂交(RNA-FISH)确定ciratXN7在EC细胞中的定位。通过逆转录-聚合酶链反应定量cirATXN7、miR-4319和NLRC5的mRNA水平。使用RNA结合蛋白免疫沉淀评估ciratXN7与miR-4319的结合活性。使用双荧光素酶报告基因集落形成测定来探索circatXN7是否通过miR-4319调节EC细胞的增殖。通过蛋白质印迹定量蛋白质水平。使用集落形成和Transwell测定检查NLRC5对EC细胞增殖和侵袭的影响。建立皮下移植瘤裸鼠模型,观察ciratXN7对EC细胞体内增殖的影响。
    结果:cirATXN7主要定位于细胞质。miR-4319的过表达或抑制显著调控EC细胞的增殖,而cirATXN7竞争性抑制miR-4319表达。miR-4319过表达显著抑制NLRC5表达,表明NLRC5是miR-4319的下游调控靶标。cirATXN7通过miR-4319影响NLRC5表达。裸鼠体内肿瘤形成实验表明,敲低ciratXN7通过miR-4319调节NLRC5的表达,并显著抑制EC细胞的增殖。
    结论:体外细胞和体内动物实验表明ciratXN7调节增殖,入侵,并通过miR-4319/NLRC5信号通路实现EC细胞的迁移。
    BACKGROUND: This study aimed to explore the molecular mechanism through which circular RNA of ataxin 7 (circATXN7) regulates the proliferation and invasion of esophageal cancer (EC) cells via microRNA (miR)-4319/NLR family CARD domain containing 5 (NLRC5).
    METHODS: The localization of circATXN7 in EC cells was determined by RNA fluorescent in situ hybridization (RNA-FISH). The mRNA levels of circATXN7, miR-4319, and NLRC5 were quantified by reverse transcription-polymerase chain reactions. The binding activity of circATXN7 to miR-4319 was assessed using RNA-binding protein immunoprecipitation. Whether circATXN7 regulates the proliferation of EC cells via miR-4319 was explored using dual-luciferase reporter gene colony formation assays. Protein levels were quantified by western blot. The effect of NLRC5 on the proliferation and invasion of EC cells was examined using colony formation and Transwell assays. A subcutaneous transplanted tumor nude mouse model was established to observe the effect of circATXN7 on the proliferation of EC cells in vivo.
    RESULTS: circATXN7 localized mainly to the cytoplasm. Overexpression or inhibition of miR-4319 significantly regulated the proliferation of EC cells, while circATXN7 competitively inhibited miR-4319 expression. Overexpression of miR-4319 significantly inhibited NLRC5 expression, indicating NLRC5 is a downstream regulatory target of miR-4319. circATXN7 influenced NLRC5 expression via miR-4319. In vivo tumor formation experiments in nude mice revealed that knocking down circATXN7 regulated NLRC5 expression via miR-4319 and significantly inhibited the proliferation of EC cells.
    CONCLUSIONS: In vitro cell and in vivo animal experiments showed that circATXN7 regulates the proliferation, invasion, and migration of EC cells through the miR-4319/NLRC5 signaling pathway.
    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
    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
    我国心肌梗死的发病率和死亡率呈逐年上升趋势。巨噬细胞向经典活化的巨噬细胞(M1)表型的极化在心肌梗塞后的炎性应激的进展中至关重要。聚(ADP-核糖)聚合酶1(PARP1)是PARP家族中普遍存在且特征最好的成员,据报道,这支持巨噬细胞向促炎表型的极化。然而,PARP1在心肌缺血损伤中的作用仍有待阐明。这里,我们证明,心肌梗死小鼠模型诱导了以心功能不全为特征的心脏损伤,并增加了心肌巨噬细胞中PARP1的表达.PJ34抑制剂对PARP1的抑制作用可有效缓解M1巨噬细胞极化,减少梗死面积,减少炎症并挽救小鼠MI后的心功能。机械上,抑制PARP1增加NLRC5基因表达,从而抑制NF-κB通路,从而减少炎性细胞因子如IL-1β和TNF-α的产生。NLRC5的抑制通过有效消除上述这种机制的影响来促进感染。有趣的是,抑制NLRC5可促进心脏巨噬细胞向M1表型极化,但对M2巨噬细胞无重大影响.我们的结果表明,抑制PARP1增加NLRC5基因表达,从而抑制M1极化,改善心脏功能,减少梗死面积和减轻炎症损伤。上述发现为心肌梗死后驱动巨噬细胞极化的促炎机制提供了新的见解,从而为受心肌梗死影响的个体的未来治疗干预引入了新的潜在目标。
    The incidence and mortality rate of myocardial infarction are increasing per year in China. The polarization of macrophages towards the classically activated macrophages (M1) phenotype is of utmost importance in the progression of inflammatory stress subsequent to myocardial infarction. Poly (ADP-ribose) polymerase 1(PARP1) is the ubiquitous and best characterized member of the PARP family, which has been reported to support macrophage polarization towards the pro-inflammatory phenotype. Yet, the role of PARP1 in myocardial ischemic injury remains to be elucidated. Here, we demonstrated that a myocardial infarction mouse model induced cardiac damage characterized by cardiac dysfunction and increased PARP1 expression in cardiac macrophages. Inhibition of PARP1 by the PJ34 inhibitors could effectively alleviate M1 macrophage polarization, reduce infarction size, decrease inflammation and rescue the cardiac function post-MI in mice. Mechanistically, the suppression of PARP1 increase NLRC5 gene expression, and thus inhibits the NF-κB pathway, thereby decreasing the production of inflammatory cytokines such as IL-1β and TNF-α. Inhibition of NLRC5 promote infection by effectively abolishing the influence of this mechanism discussed above. Interestingly, inhibition of NLRC5 promotes cardiac macrophage polarization toward an M1 phenotype but without having major effects on M2 macrophages. Our results demonstrate that inhibition of PARP1 increased NLRC5 gene expression, thereby suppressing M1 polarization, improving cardiac function, decreasing infarct area and attenuating inflammatory injury. The aforementioned findings provide new insights into the proinflammatory mechanisms that drive macrophage polarization following myocardial infarction, thereby introducing novel potential targets for future therapeutic interventions in individuals affected by myocardial infarction.
    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
    主要组织相容性复合物I类(MHCI类)介导的肿瘤抗原加工和呈递(APP)途径对于细胞毒性CD8T淋巴细胞(CD8CTL)的募集和激活至关重要。然而,这种通路在许多癌症中经常失调,从而导致免疫疗法的失败。这里,我们报道,在小鼠肿瘤和患者中,肿瘤固有Hippo通路的激活与MHCI类APP基因的表达和CD8+CTL的丰度正相关.阻断Hippo途径效应物Yes相关蛋白/转录增强相关结构域(YAP/TEAD)有效改善抗肿瘤免疫。机械上,YAP/TEAD复合物与核小体重塑和脱乙酰酶复合物协作以抑制NLRC5转录。通过YAP/TEAD消耗或药理学抑制对NLRC5的上调增加了MHCI类APP基因的表达并增强了CD8+CTL介导的对癌细胞的杀伤。总的来说,我们的结果表明,YAP的重要肿瘤促进功能依赖于NLRC5,从而损害MHCI类APP通路,并为在癌症免疫治疗中抑制YAP活性提供了理论基础.
    The major histocompatibility complex class I (MHC class I)-mediated tumor antigen processing and presentation (APP) pathway is essential for the recruitment and activation of cytotoxic CD8+ T lymphocytes (CD8+ CTLs). However, this pathway is frequently dysregulated in many cancers, thus leading to a failure of immunotherapy. Here, we report that activation of the tumor-intrinsic Hippo pathway positively correlates with the expression of MHC class I APP genes and the abundance of CD8+ CTLs in mouse tumors and patients. Blocking the Hippo pathway effector Yes-associated protein/transcriptional enhanced associate domain (YAP/TEAD) potently improves antitumor immunity. Mechanistically, the YAP/TEAD complex cooperates with the nucleosome remodeling and deacetylase complex to repress NLRC5 transcription. The upregulation of NLRC5 by YAP/TEAD depletion or pharmacological inhibition increases the expression of MHC class I APP genes and enhances CD8+ CTL-mediated killing of cancer cells. Collectively, our results suggest a crucial tumor-promoting function of YAP depending on NLRC5 to impair the MHC class I APP pathway and provide a rationale for inhibiting YAP activity in immunotherapy for cancer.
    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
    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: Review
    MHCI类分子向CD8+T细胞的抗原呈递对于宿主防御病毒感染是必需的。多种机制已经在多种病毒中进化以逃避免疫监视和防御以支持宿主细胞中的病毒增殖。通过体外SARS-CoV-2感染研究和COVID-19患者样本分析,我们发现SARS-CoV-2通过抑制MHCI类基因的主要转录调节因子NLRC5的表达和功能来抑制MHCI类途径的诱导。在这次审查中,我们讨论了抑制MHCI类通路的分子机制和对COVID-19的临床意义。
    Antigen presentation to CD8+ T cells by MHC class I molecules is essential for host defense against viral infections. Various mechanisms have evolved in multiple viruses to escape immune surveillance and defense to support viral proliferation in host cells. Through in vitro SARS-CoV-2 infection studies and analysis of COVID-19 patient samples, we found that SARS-CoV-2 suppresses the induction of the MHC class I pathway by inhibiting the expression and function of NLRC5, a major transcriptional regulator of MHC class I genes. In this review, we discuss the molecular mechanisms for suppression of the MHC class I pathway and clinical implications for COVID-19.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号