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
    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
    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
    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
    长链脂肪酸(LCFAs)及其激活酶的免疫调节作用,酰基辅酶A(CoA)合成酶长链家族(ACSL),肿瘤微环境在很大程度上仍然未知。这里,我们发现ACSL5具有免疫依赖性肿瘤抑制因子的功能。ACSL5表达通过调节主要组织相容性复合物I类(MHC-I)介导的抗原呈递使肿瘤体内对PD-1阻断治疗敏感,并在体外使CD8T细胞介导的细胞毒性敏感。通过筛选ACSL5的潜在底物,我们进一步确定反油酸(EA),一种长期以来被认为对人体健康有害的反式LCFA,表型以增强MHC-I表达。EA补充可以抑制肿瘤生长并使PD-1阻断疗法敏感。临床上,ACSL5表达与肺癌患者生存率改善呈正相关,血浆EA水平也是免疫治疗效率的预测指标。我们的发现为通过饮食EA补充靶向ACSL5或抗原呈递的代谢重编程来增强免疫治疗提供了基础。
    Immunomodulatory effects of long-chain fatty acids (LCFAs) and their activating enzyme, acyl-coenzyme A (CoA) synthetase long-chain family (ACSL), in the tumor microenvironment remain largely unknown. Here, we find that ACSL5 functions as an immune-dependent tumor suppressor. ACSL5 expression sensitizes tumors to PD-1 blockade therapy in vivo and the cytotoxicity mediated by CD8+ T cells in vitro via regulation of major histocompatibility complex class I (MHC-I)-mediated antigen presentation. Through screening potential substrates for ACSL5, we further identify that elaidic acid (EA), a trans LCFA that has long been considered harmful to human health, phenocopies to enhance MHC-I expression. EA supplementation can suppress tumor growth and sensitize PD-1 blockade therapy. Clinically, ACSL5 expression is positively associated with improved survival in patients with lung cancer, and plasma EA level is also predictive for immunotherapy efficiency. Our findings provide a foundation for enhancing immunotherapy through either targeting ACSL5 or metabolic reprogramming of antigen presentation via dietary EA supplementation.
    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
    小胶质细胞被认为是中枢免疫微环境的关键免疫效应,它们的失调与神经炎症和情绪障碍有关。含核苷酸结合寡聚化结构域样受体家族caspase募集结构域五(NLRC5)是Nod样受体家族的新成员。最近,据报道NLRC5由小胶质细胞表达。尽管如此,NLRC5在小胶质细胞激活中的确切作用及其在抑郁症中的功能尚未研究。在这里,我们发现减少NLRC5减少了脂多糖(LPS)诱导的促炎细胞因子(IL-1β,IL-6和TNF-α)在原代培养的小胶质细胞和小胶质细胞系中,但在骨髓衍生的巨噬细胞(BMDM)中没有。更详细地说,减少NLRC5通过减弱IKKα/β磷酸化和抑制NF-κB信号来减少LPS诱导的细胞因子的分泌。此外,LPS或慢性不可预知轻度应激(CUMS)诱导的抑郁小鼠海马中Nlrc5的表达增加。根据体外研究结果,Nlrc5缺乏抑制了小鼠海马中的小胶质细胞激活,并改善了LPS或CUMS诱导的抑郁样行为。总之,我们证明了NLRC5在LPS诱导的小胶质细胞激活和LPS或CUMS诱导的抑郁小鼠模型中的关键作用。
    Microglia are believed to be the key immune effectors of the central immune microenvironment, and their dysregulation is associated with neuroinflammation and mood disorders. Nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain-containing five (NLRC5) is a new member of the Nod-like receptor family. Recently, NLRC5 has been reported to be expressed by microglia. Nonetheless, the exact roles of NLRC5 in microglial activation and its function in depression have not been investigated yet. Herein, we found that reducing NLRC5 decreased lipopolysaccharide (LPS)-induced secretion of pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in primary cultured microglia and microglial cell lines but not in bone marrow-derived macrophages (BMDMs). In more detail, reducing NLRC5 diminished the secretion of LPS-induced cytokines by attenuating IKKα/β phosphorylation and inhibiting NF-κB signaling. Moreover, the expression of Nlrc5 in the hippocampus of LPS- or chronic unpredictable mild stress (CUMS)-induced depressive mice was increased. In line with the in vitro findings, Nlrc5 deficiency inhibited microglial activation in the mouse hippocampus and improved LPS- or CUMS-induced depressive-like behaviors. In summary, we demonstrated the critical role of NLRC5 in LPS-induced microglial activation and LPS- or CUMS-induced depressive mouse models.
    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
    帕金森病(PD)的主要特征是黑质致密质(SNpc)中多巴胺能神经元的进行性变性和过度激活的小胶质细胞和星形胶质细胞介导的神经炎症。据报道,NLRC5(核苷酸结合寡聚化结构域样受体家族caspase募集结构域)参与各种免疫疾病,但其在神经退行性疾病中的作用尚不清楚。在目前的研究中,我们发现,NLRC5的表达在1-甲基-4-苯基-1,2,3,6-四氢吡啶盐酸盐(MPTP)诱导的PD小鼠的黑质纹状体轴中增加,以及在原代星形胶质细胞中,小胶质细胞和神经元暴露于不同的神经毒性刺激。在急性MPTP诱导的PD模型中,NLRC5缺乏可显着减少多巴胺能系统变性并改善运动缺陷和纹状体炎症。此外,我们发现NLRC5缺乏降低了促炎基因IL-1β的表达,用神经炎性刺激处理的原代小胶质细胞和原代星形胶质细胞中的IL-6,TNF-α和COX2,并减少了响应于LPS处理的混合神经胶质细胞中的炎症反应。此外,NLRC5缺陷抑制了NF-κB和MAPK信号通路的激活,并增强了混合胶质细胞中AKT-GSK-3β和AMPK信号的激活。此外,NLRC5缺陷增加了用MPP+或来自LPS刺激的混合神经胶质细胞的条件培养基处理的原代神经元的存活,并促进了NF-κB和AKT信号通路的激活。此外,与健康受试者相比,PD患者血液中NLRC5的mRNA表达降低。因此,我们建议NLRC5促进PD的神经炎症和多巴胺能变性,并可能作为神经胶质激活的标志.
    Parkinson\'s disease (PD) is mainly characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and neuroinflammation mediated by overactivated microglia and astrocytes. NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) has been reported to participate in various immune disorders, but its role in neurodegenerative diseases remains unclear. In the current study, we found that the expression of NLRC5 was increased in the nigrostriatal axis of mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced PD, as well as in primary astrocytes, microglia and neurons exposed to different neurotoxic stimuli. In an acute MPTP-induced PD model, NLRC5 deficiency significantly reduced dopaminergic system degeneration and ameliorated motor deficits and striatal inflammation. Furthermore, we found that NLRC5 deficiency decreased the expression of the proinflammatory genes IL-1β, IL-6, TNF-α and COX2 in primary microglia and primary astrocytes treated with neuroinflammatory stimuli and reduced the inflammatory response in mixed glial cells in response to LPS treatment. Moreover, NLRC5 deficiency suppressed activation of the NF-κB and MAPK signaling pathways and enhanced the activation of AKT-GSK-3β and AMPK signaling in mixed glial cells. Furthermore, NLRC5 deficiency increased the survival of primary neurons treated with MPP+ or conditioned medium from LPS-stimulated mixed glial cells and promoted activation of the NF-κB and AKT signaling pathways. Moreover, the mRNA expression of NLRC5 was decreased in the blood of PD patients compared to healthy subjects. Therefore, we suggest that NLRC5 promotes neuroinflammation and dopaminergic degeneration in PD and may serve as a marker of glial activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号