LUAD

LUAD
  • 文章类型: Journal Article
    M2肿瘤相关巨噬细胞(M2TAM),肿瘤微环境的重要组成部分,肺腺癌(LUAD)血管生成对肿瘤侵袭和转移有显著影响。在这项研究中,分析单细胞RNA和批量RNA测序数据,以鉴定12个M2TAM和血管生成相关基因(OLR1,CTSL,HLA-DPB1,NUPR1,ALOX5,DOCK4,CSF2RB,PTPN6,TNFSF12,HNRNPA2B1,NCL,和BIRC2)。这些基因被用来构建预后标签,随后使用外部队列进行了验证。此外,免疫谱分析表明,低危组表现出明显的免疫细胞浸润和相对活跃的状态.重要的是,预后特征与PD-1,CTLA4,肿瘤突变负荷,和抗癌药物敏感性。总之,这项研究提出了基于M2TAM和血管生成相关基因的LUAD患者的新预后特征.签名通过独立的方式预测LUAD的预后,揭示了参与肿瘤免疫相关功能的潜在分子机制,并为LUAD患者的治疗提供适当的临床策略。
    M2 tumor-associated macrophage (M2 TAM), a crucial component of the tumor microenvironment, has a significant impact on tumor invasion and metastasis in the form of angiogenesis for lung adenocarcinoma (LUAD). In this study, both single-cell RNA and bulk RNA sequencing data were analyzed to identify 12 M2 TAM and angiogenesis-related genes (OLR1, CTSL, HLA-DPB1, NUPR1, ALOX5, DOCK4, CSF2RB, PTPN6, TNFSF12, HNRNPA2B1, NCL, and BIRC2). These genes were used to construct a prognostic signature, which was subsequently validated using an external cohort. Moreover, the immune profile analysis indicated that the low-risk group exhibited a distinct immune cell infiltration and relatively active status. Importantly, the prognostic signature was closely associated with PD-1, CTLA4, tumor mutation burden, and anti-cancer drug sensitivity. In summary, this study proposes a new prognostic signature for patients with LUAD based on M2 TAM and angiogenesis-related genes. The signature forecasts the prognosis of LUAD by an independent manner, reveals the potential molecular mechanisms involved in tumor immune-related functions, and offers appropriate clinical strategies for the treatment of patients with LUAD.
    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
    转化生长因子-β(TGF-β)信号通路在诱导上皮间质转化(EMT)和促进癌症转移中至关重要。长链非编码RNA(lncRNAs)已经成为这些过程中的重要参与者,然而,他们的精确机制仍然难以捉摸。这里,我们证明TGF-β上调的lncRNA1(TBUR1)在肺腺癌(LUAD)细胞中通过Smad3/4信号传导被TGF-β显着激活。功能上,TBUR1触发EMT,在体外增强LUAD细胞的迁移和侵袭,并促进裸鼠的转移。机械上,TBUR1与异质核核糖核蛋白C(hnRNPC)相互作用,以m6A依赖性方式稳定GRB2mRNA。临床上,TBUR1在LUAD组织中上调,并与不良预后相关。强调其作为LUAD预后生物标志物和治疗靶点的潜力。一起来看,我们的发现强调了TBUR1在介导TGF-β诱导的EMT和LUAD转移中的关键作用,为未来的治疗干预提供见解。
    The transforming growth factor-β (TGF-β) signaling pathway is pivotal in inducing epithelial-mesenchymal transition (EMT) and promoting cancer metastasis. Long non-coding RNAs (lncRNAs) have emerged as significant players in these processes, yet their precise mechanisms remain elusive. Here, we demonstrate that TGF-β-upregulated lncRNA 1 (TBUR1) is significantly activated by TGF-β via Smad3/4 signaling in lung adenocarcinoma (LUAD) cells. Functionally, TBUR1 triggers EMT, enhances LUAD cell migration and invasion in vitro, and promotes metastasis in nude mice. Mechanistically, TBUR1 interacts with heterogeneous nuclear ribonucleoproteins C (hnRNPC) to stabilize GRB2 mRNA in an m6A-dependent manner. Clinically, TBUR1 is upregulated in LUAD tissues and correlates with poor prognosis, highlighting its potential as a prognostic biomarker and therapeutic target for LUAD. Taken together, our findings underscore the crucial role of TBUR1 in mediating TGF-β-induced EMT and metastasis in LUAD, providing insights for future therapeutic interventions.
    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
    目的:为了确定能够区分小细胞肺癌(SCLC)和非SCLC(NSCLC)的生物标志物,并探讨其与小细胞肺癌放化疗预后的关系。
    方法:GSE40275数据集用于鉴定SCLC中的潜在靶标。本研究队列1中有196例肺癌(LC)患者。在组群1中通过免疫组织化学测定测定组织中的MTHFD1水平。所有接受局部放化疗(CRT)的肺癌患者均纳入队列2,并在队列2中确定MTHFD1水平与CRT治疗结果的相关性。细胞实验用于确定MTHFD1对SCLC和NSCLC细胞的放射敏感性的功能。
    结果:LC组织中的MTHFD1水平升高,并且可以将SCLC与肺鳞癌(LUSC)和肺腺癌(LUAD)区分开。MTHFD1高表型的小细胞肺癌患者经CRT治疗后预后较差,而在LUSC和LUAD组中,MTHFD1水平与预后无显著相关性。细胞实验表明,MTHFD1的过表达增加了体外SCLC和NSCLC中的放射抗性。
    结论:MTHFD1表达可能是SCLC和CRT治疗结果的新型特异性预后生物标志物。
    OBJECTIVE: To identify biomarkers that can discriminated small cell lung cancer (SCLC) from non-SCLC (NSCLC), and explore their association with the prognosis of SCLC under chemoradiotherapy.
    METHODS: The GSE40275 dataset was used to identify potential targets in SCLC. There were 196 patients of lung cancer (LC) in cohort 1 of this study. MTHFD1 levels in tissues were determined by immunohistochemistry assay in cohort 1. Lung cancer patients who were all underwent local chemoradiotherapy (CRT) were included in cohort 2, and the association of MTHFD1 levels with CRT treatment outcome were determined in cohort 2. Cell experiments were used to determine the function of MTHFD1 on the radio-sensitivity of SCLC and NSCLC cells.
    RESULTS: The MTHFD1 levels in LC tissues were increased, and could discriminate SCLC from both lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). Small cell lung cancer patients with MTHFD1 high phenotype had a poorer prognosis after CRT treatment, whereas no significant correlation was found between MTHFD1 levels and prognosis in LUSC and LUAD group. Cell experiments demonstrated that overexpression of MTHFD1 increases radio-resistance in both SCLC and NSCLC in vitro.
    CONCLUSIONS: MTHFD1 expressions might be a novel specifically prognostic biomarker for SCLC and the CRT treatment outcome.
    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:肺癌是一种常见的恶性肿瘤,具有很高的发病率和死亡率。氨基葡萄糖6-磷酸N-乙酰转移酶(GNPNAT1),它是己糖胺生物合成途径(HBP)中的关键酶,已被鉴定为转移相关基因,并在肺腺癌(LUAD)中上调。然而,GNPNAT1在LUAD转移中的作用及相关机制尚不清楚。
    方法:我们分析了GNPNAT1在公共数据库中的表达,并通过免疫组织化学(IHC)证实了结果。基于癌症基因组图谱(TCGA)研究了GNPNAT1在LUAD中的生物学功能。通过使用表达数据(ESTIMATE)估计恶性肿瘤组织中的基质细胞和免疫细胞和通过估计RNA转录物的相对子集(CIBERSORT)R包来进行细胞类型鉴定,分析GNPNAT1与癌症免疫特征之间的相关性。GNPNAT1表达改变对LUAD细胞肿瘤发生的潜在机制,扩散,迁移,入侵,和转移进行了体外和体内探索。
    结果:我们证明GNPNAT1表达在LUAD中显著增加,并且与患者的总生存期(OS)呈负相关。hsa-miR-1-3p和hsa-miR-26a-5p被鉴定为GNPNAT1的上游miRNA靶标。GNPNAT1与CD8T细胞的浸润水平有关,记忆激活的CD4T细胞,NK细胞静息,巨噬细胞M0,巨噬细胞M1,中性粒细胞,γδT细胞,和嗜酸性粒细胞,虽然它与记忆静息CD4T细胞呈负相关,调节性T细胞(Tregs),静息NK细胞,单核细胞,静息树突状细胞,和静止的肥大细胞。GNPNAT1敲低显著抑制增殖,迁移,入侵,上皮-间质转化(EMT)过程,和LUAD细胞的转移,而GNPNAT1的过表达显示出相反的作用。拯救实验显示Snai2敲低可逆转GNPNAT1诱导的LUAD细胞迁移,入侵,EMT。机械上,GNPNAT1通过抑制LUAD中Snai2的泛素化降解促进癌细胞转移。
    结论:综合来看,这些数据表明GNPNAT1是LUAD患者的预后生物标志物.此外,GNPNAT1对于促进LUAD细胞的肿瘤发生和转移至关重要,并且可能是预防LUAD转移的潜在治疗靶标。
    BACKGROUND: Lung cancer is a common malignant tumor with high morbidity and mortality rate. Glucosamine 6-phosphate N-acetyltransferase (GNPNAT1), which serves as a critical enzyme in hexosamine biosynthetic pathway (HBP), has been identified as a metastasis-associated gene and is upregulated in lung adenocarcinoma (LUAD). However, the exact role and related mechanism of GNPNAT1 in LUAD metastasis remain unknown.
    METHODS: We analyzed the expression of GNPNAT1 in the public databases and confirmed the results by immunohistochemistry (IHC). The biological functions of GNPNAT1 in LUAD were investigated based on The Cancer Genome Atlas (TCGA). Correlations between GNPNAT1 and cancer immune characteristics were analyzed via the Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE) and Cell-type Identification by Estimating Relative Subsets of RNA Transcript (CIBERSORT) R package. The underlying mechanisms of altered GNPNAT1 expression on LUAD cell tumorigenesis, proliferation, migration, invasion, and metastasis were explored in vitro and in vivo.
    RESULTS: We demonstrated that GNPNAT1 expression was significantly increased in LUAD and negatively associated with the overall survival (OS) of patients. hsa-miR-1-3p and hsa-miR-26a-5p were identified as upstream miRNA targets of GNPNAT1. GNPNAT1 was associated with the infiltration levels of CD8 T cells, memory-activated CD4 T cells, NK cells resting, macrophages M0, macrophages M1, neutrophils, gamma delta T cells, and eosinophils, while it was negatively correlated with memory-resting CD4 T cells, regulatory T cells (Tregs), resting NK cells, monocytes, resting dendritic cells, and resting mast cells. GNPNAT1 knockdown significantly inhibited proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) process, and metastasis of LUAD cells, while overexpression of GNPNAT1 revealed the opposite effects. Rescue assay showed that Snai2 knockdown reversed GNPNAT1-induced LUAD cells migration, invasion, and EMT. Mechanistically, GNPNAT1 promoted cancer cell metastasis via repressing ubiquitination degradation of Snai2 in LUAD.
    CONCLUSIONS: Taken together, these data indicate that GNPNAT1 serves as a prognostic biomarker for LUAD patient. Additionally, GNPNAT1 is critical for promoting tumorigenesis and metastasis of LUAD cells and may be a potential therapeutic target for preventing LUAD metastasis.
    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
    聚嘧啶束结合蛋白1(PTBP1)在剪接和转录后调控中起重要作用。此外,PTBP1已被认为是肿瘤发生的原因。然而,PTBP1参与细胞衰老,衰老和癌症抑制的关键生物学过程,还有待澄清。这里,结果表明,PTBP1与肺腺癌(LUAD)的肿瘤生长和预后有关。PTBP1在包括LUAD在内的各种癌症类型中表现出显著增加的表达,并且在多种细胞衰老模型中表现出一致降低的表达。抑制PTBP1诱导LUAD细胞中的细胞衰老。从分子机制来看,PTBP1的沉默增强了F-box蛋白5(FBXO5)外显子3的跳跃,导致产生不太稳定的RNA剪接变体,FBXO5-S,这随后降低了FBXO5的整体表达。此外,发现FBXO5的下调可诱导LUAD衰老。总的来说,这些发现说明PTBP1通过抑制衰老在LUAD中具有致癌功能,靶向PTBP1介导的异常剪接在癌症治疗中具有治疗潜力。
    Polypyrimidine tract-binding protein 1 (PTBP1) plays an essential role in splicing and post-transcriptional regulation. Moreover, PTBP1 has been implicated as a causal factor in tumorigenesis. However, the involvement of PTBP1 in cellular senescence, a key biological process in aging and cancer suppression, remains to be clarified. Here, it is shown that PTBP1 is associated with the facilitation of tumor growth and the prognosis in lung adenocarcinoma (LUAD). PTBP1 exhibited significantly increased expression in various cancer types including LUAD and showed consistently decreased expression in multiple cellular senescence models. Suppression of PTBP1 induced cellular senescence in LUAD cells. In terms of molecular mechanisms, the silencing of PTBP1 enhanced the skipping of exon 3 in F-box protein 5 (FBXO5), resulting in the generation of a less stable RNA splice variant, FBXO5-S, which subsequently reduces the overall FBXO5 expression. Additionally, downregulation of FBXO5 was found to induce senescence in LUAD. Collectively, these findings illustrate that PTBP1 possesses an oncogenic function in LUAD through inhibiting senescence, and that targeting aberrant splicing mediated by PTBP1 has therapeutic potential in cancer treatment.
    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
    肺腺癌(LUAD)是全球癌症死亡的主要原因,发病率高,生存率低。烟碱乙酰胆碱受体在LUAD的进展中起重要作用。在这项研究中,对17种烟碱乙酰胆碱受体变构剂的筛选显示,多杀菌素有效抑制了LUAD细胞的增殖。实验证明,多杀菌素诱导细胞周期阻滞在G1期,并刺激细胞凋亡,从而阻碍LUAD的生长,并在体外和体内增强对吉非替尼的反应性。通过转录组测序获得的机制见解,共同IP,和蛋白质免疫印迹表明多杀菌素破坏了CHRNA5和EGFR之间的相互作用,从而抑制下游复合物的形成和EGFR信号通路的激活。补充外源性乙酰胆碱可以减轻多杀菌素对LUAD细胞增殖的抑制作用。本研究阐明了多杀菌素在LUAD中的治疗作用和机制。为新型LUAD治疗提供了理论和实验基础。
    Lung adenocarcinoma (LUAD) is the leading cause of cancer death worldwide, with high incidence and low survival rates. Nicotinic acetylcholine receptors play an important role in the progression of LUAD. In this study, a screening of 17 nicotinic acetylcholine receptor allosteric agents revealed that spinosad effectively suppressed the proliferation of LUAD cells. The experiments demonstrated that spinosad induced cell cycle arrest in the G1 phase and stimulated apoptosis, thereby impeding the growth of LUAD and enhancing the responsiveness to gefitinib in vitro and vivo. Mechanistic insights obtained through transcriptome sequencing, Co-IP, and protein immunoblots indicated that spinosad disrupted the interaction between CHRNA5 and EGFR, thereby inhibiting the formation of downstream complexes and activation of the EGFR signaling pathway. The supplementation of exogenous acetylcholine showed to mitigate the inhibition of LUAD cell proliferation induced by spinosad. This study elucidates the therapeutic effects and mechanisms of spinosad in LUAD, and offers a theoretical and experimental foundation for novel LUAD treatments.
    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:肺腺癌(LUAD)的复杂生物学机制,以缺乏独特的生物标志物为特征,仍然难以捉摸。长非编码RNA(lncRNA)的存在已被确定为在癌发生中起作用。然而,lncRNACYTOR在LUAD中的调节作用和机制尚未阐明。
    方法:在本研究中,采用RT-qPCR和Westernblot检测基因mRNA和蛋白表达,分别。通过CCK-8测定评价细胞增殖。进行Transwell以测定细胞迁移和侵袭。通过异种移植动物模型研究体内CYTOR的功能。
    结果:我们观察到LUAD中CYTOR的明显上调。沉默CYTOR显著减少增殖,迁移,和LUAD细胞的侵袭能力。机制分析表明CYTOR靶向miR-503-5p/PCSK9轴。此外,miR-503-5p的抑制部分逆转了CYTOR沉默对LUAD细胞恶性进展的抑制作用。动物实验表明CYTOR/miR-503-5p/PCSK9抑制了裸鼠体内肿瘤的形成。
    结论:这些研究结果表明,lncRNACYTOR在LUAD中起着癌基因的作用,通过miR-503-5p/PCSK9轴调节肿瘤恶性进展。这项研究揭示了LUAD进展的新调控机制,为LUAD提供潜在的治疗靶点。
    BACKGROUND: The intricate biological mechanism underlying lung adenocarcinoma (LUAD), characterized by a deficiency of distinctive biomarkers, remain elusive. The presence of Long non-coding RNAs (lncRNAs) have been established to play a role in carcinogenesis. Nevertheless, the regulatory effects and mechanisms of lncRNA CYTOR in LUAD have yet to be elucidated.
    METHODS: In this study, RT-qPCR and Western blot were adopted to examine gene mRNA and protein expression, respectively. Cell proliferation was evaluated by CCK-8 assays. Transwell was performed to assay cell migration and invasion. The function of CYTOR in vivo was investigated through a xenograft animal model.
    RESULTS: We observed an apparent upregulation of CYTOR in LUAD. Silencing CYTOR significantly reduced proliferation, migration, and invasion capabilities of LUAD cells. Mechanism analysis indicated that CYTOR targeted the miR-503-5p/PCSK9 axis. Additionally, inhibiting of miR-503-5p partially reversed the inhibitory effects of CYTOR silencing on the malignant progression of LUAD cells. Animal experiments revealed that CYTOR/miR-503-5p/PCSK9 curbed tumor formation of nude mice in vivo.
    CONCLUSIONS: These findings demonstrated that lncRNA CYTOR acted as an oncogene in LUAD, regulating tumor malignant progression through the miR-503-5p/PCSK9 axis. This study unveiled a new regulation mechanism of LUAD progression, offering potential therapeutic targets for LUAD.
    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:在肿瘤微环境(TME)中,缺氧和乳酸代谢之间存在双向关系,每个组件对另一个组件施加相互影响,形成了不可分割的联系。本研究的目的是通过合并与缺氧和乳酸代谢相关的基因来建立预后模型。该模型旨在作为预测患者预后的工具,包括生存率,免疫微环境的状态,以及肺腺癌(LUAD)患者对治疗的反应性。
    方法:从癌症基因组图谱(TCGA)和基因表达综合(GEO)的综合存储库中获得对LUAD特异的转录组测序数据和患者临床信息。从一系列可访问的数据集组装了与缺氧和乳酸代谢有关的基因汇编。采用单变量和多变量Cox回归分析。额外的调查程序,包括肿瘤突变负荷(TMB),微卫星不稳定性(MSI),功能富集评估和估计,CIBERSORT,和TIDE算法,用于评估药物敏感性和预测免疫疗法的疗效。
    结果:包含5个乳酸和缺氧相关基因(LHRGs)的新型预后标记,PKFP,SLC2A1,BCAN,CDKN3和ANLN,已建立。该模型表明,LHRG相关风险评分升高的LUAD患者的生存率显着降低。单变量和多变量Cox分析均证实,风险评分是总体生存率的可靠预后指标。免疫表型显示记忆CD4+T细胞浸润增加,与低风险患者相比,树突状细胞和NK细胞被归类为高风险类别。在高危人群中,肺腺癌驱动基因突变的可能性更高,MSI与风险评分相关.功能富集分析表明,在高风险组中,细胞周期相关途径占优势。而代谢途径在低危组更为普遍.此外,药物敏感性分析显示,高危人群对各种药物的敏感性增加,特别是PI3K-AKT的抑制剂,EGFR,和ELK通路。
    结论:该预后模型整合了乳酸代谢和缺氧参数,提供关于生存的预测性见解,免疫细胞浸润和功能,以及LUAD患者的治疗反应性。该模型可以促进个性化治疗策略,根据每个患者疾病的独特分子特征定制干预措施。
    BACKGROUND: In the tumor microenvironment (TME), a bidirectional relationship exists between hypoxia and lactate metabolism, with each component exerting a reciprocal influence on the other, forming an inextricable link. The aim of the present investigation was to develop a prognostic model by amalgamating genes associated with hypoxia and lactate metabolism. This model is intended to serve as a tool for predicting patient outcomes, including survival rates, the status of the immune microenvironment, and responsiveness to therapy in patients with lung adenocarcinoma (LUAD).
    METHODS: Transcriptomic sequencing data and patient clinical information specific to LUAD were obtained from comprehensive repositories of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). A compendium of genes implicated in hypoxia and lactate metabolism was assembled from an array of accessible datasets. Univariate and multivariate Cox regression analyses were employed. Additional investigative procedures, including tumor mutational load (TMB), microsatellite instability (MSI), functional enrichment assessments and the ESTIMATE, CIBERSORT, and TIDE algorithms, were used to evaluate drug sensitivity and predict the efficacy of immune-based therapies.
    RESULTS: A novel prognostic signature comprising five lactate and hypoxia-related genes (LHRGs), PKFP, SLC2A1, BCAN, CDKN3, and ANLN, was established. This model demonstrated that LUAD patients with elevated LHRG-related risk scores exhibited significantly reduced survival rates. Both univariate and multivariate Cox analyses confirmed that the risk score was a robust prognostic indicator of overall survival. Immunophenotyping revealed increased infiltration of memory CD4 + T cells, dendritic cells and NK cells in patients classified within the high-risk category compared to their low-risk counterparts. Higher probability of mutations in lung adenocarcinoma driver genes in high-risk groups, and the MSI was associated with the risk-score. Functional enrichment analyses indicated a predominance of cell cycle-related pathways in the high-risk group, whereas metabolic pathways were more prevalent in the low-risk group. Moreover, drug sensitivity analyses revealed increased sensitivity to a variety of drugs in the high-risk group, especially inhibitors of the PI3K-AKT, EGFR, and ELK pathways.
    CONCLUSIONS: This prognostic model integrates lactate metabolism and hypoxia parameters, offering predictive insights regarding survival, immune cell infiltration and functionality, as well as therapeutic responsiveness in LUAD patients. This model may facilitate personalized treatment strategies, tailoring interventions to the unique molecular profile of each patient\'s 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
    背景:在肺腺癌(LUAD)和严重急性呼吸综合征(SARS)中,可以在肺组织中检测到不受控制的炎症。SARS-CoV-1E蛋白中的PDZ结合基序(PBM)已被证明是诱导细胞因子风暴的毒力因子。
    方法:为了确定PBM诱导的基因表达波动,分析了感染野生型(SARS-CoV-1-E-wt)或重组病毒(SARS-CoV-1-E-mutPBM)的肺组织的微阵列测序数据,其次是功能富集分析。为了了解所筛选的基因在LUAD中的作用,计算总生存期和免疫相关性.
    结果:共有12个基因可能通过表达变异和突变参与LUAD的初始和发育阶段。此外,共有12个基因失调可能导致预后较差.此外,PBM下调MAMDC2和ITGA8也可能影响患者预后.尽管保守的PBM(-D-L-L-V-)可以在冠状病毒的多个E蛋白的羧基末端发现,每种蛋白质的特定功能取决于整个氨基酸序列。
    结论:总之,含有SARS-CoV-1E蛋白的PBM通过调节重要基因表达谱并随后影响免疫反应和总体预后来促进LUAD的致癌作用。
    BACKGROUND: In both lung adenocarcinoma (LUAD) and severe acute respiratory syndrome (SARS), uncontrolled inflammation can be detected in lung tissue. The PDZ-binding motif (PBM) in the SARS-CoV-1 E protein has been demonstrated to be a virulence factor that induces a cytokine storm.
    METHODS: To identify gene expression fluctuations induced by PBM, microarray sequencing data of lung tissue infected with wild-type (SARS-CoV-1-E-wt) or recombinant virus (SARS-CoV-1-E-mutPBM) were analyzed, followed by functional enrichment analysis. To understand the role of the screened genes in LUAD, overall survival and immune correlation were calculated.
    RESULTS: A total of 12 genes might participate in the initial and developmental stages of LUAD through expression variation and mutation. Moreover, dysregulation of a total of 12 genes could lead to a poorer prognosis. In addition, the downregulation of MAMDC2 and ITGA8 by PBM could also affect patient prognosis. Although the conserved PBM (-D-L-L-V-) can be found at the end of the carboxyl terminus in multiple E proteins of coronaviruses, the specific function of each protein depends on the entire amino acid sequence.
    CONCLUSIONS: In summary, PBM containing the SARS-CoV-1 E protein promoted the carcinogenesis of LUAD by dysregulating important gene expression profiles and subsequently influencing the immune response and overall prognosis.
    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:根据2022年全球癌症统计数据,肺癌是全球癌症相关死亡的主要原因。肺腺癌(LUAD),非小细胞肺癌(NSCLC)的组织学亚型,占原发性肺癌的40%。因此,迫切需要鉴定新的预后标志物作为LUAD的临床预测标志物.
    目的:本研究旨在探讨角蛋白80(KRT80)在LUAD预后中的作用及其机制。
    方法:使用从癌症基因组图谱(TCGA)数据库检索的数据进行生物信息学分析。基因本体论(GO)和京都基因和基因组百科全书(KEGG)数据库被用来预测所涉及的生物过程和信号通路,分别。LinkedOmics数据库用于鉴定与KRT80相关的差异表达基因(DEGs)。构建列线图和Kaplan-Meier图以评估诊断为LUAD的患者的生存结果。此外,采用TIMER对KRT80表达与免疫细胞浸润进行相关性分析,揭示了LUAD中KRT80与肿瘤微环境之间复杂的相互作用。确定KRT80在LUAD和邻近正常组织中的RNA和蛋白表达水平,采用逆转录定量聚合酶链反应(RT-qPCR)和免疫组织化学技术,分别。
    结果:对TCGA数据集的审查显示,整个泛癌组织中的KRT80上调,与健康肺组织相比,LUAD显著升高。这一发现在我们的临床样本中得到了验证,其中Kaplan-Meier存活曲线表明LUAD中KRT80高表达的存活率较差。LUAD样本中KRT80的转录水平与临床参数呈正相关。如淋巴结转移分期,远处转移,和病理阶段。生存,逻辑回归,Cox回归分析强调了LUAD中KRT80高表达的临床预后意义。列线图结果强调了KRT80对LUAD患者生存的强大预测潜力。基因功能富集分析主要将KRT80与细胞因子-细胞因子受体相互作用相关,细胞周期,凋亡,和趋化因子信号通路。根据免疫浸润分析的结果,可以发现KRT80的表达与LUAD患者的免疫细胞亚群和生存率有关。
    结论:我们的研究揭示了LUAD中KRT80的显著上调,升高的KRT80表达与不良预后相关。这项研究代表了对LUAD中KRT80表达的全面和系统的评估,包括其预后和诊断意义,以及潜在的机制。我们的研究结果表明,KRT80可能成为LUAD中一种新的预后和预测性生物标志物。
    BACKGROUND: According to the 2022 Global Cancer Statistics, lung cancer is the leading cause of cancer-related mortality worldwide. Lung adenocarcinoma (LUAD), which is a histological subtype of Non- Small Cell Lung Cancer (NSCLC), accounts for 40% of primary lung cancer. Therefore, there is an urgent need to identify new prognostic markers as clinical predictive markers for LUAD.
    OBJECTIVE: This study aimed to investigate the role of Keratin 80 (KRT80) in the prognosis of LUAD and its underlying mechanisms.
    METHODS: Bioinformatics analysis was conducted using data retrieved from The Cancer Genome Atlas (TCGA) databases. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases were employed to predict the involved biological processes and signaling pathways, respectively. The LinkedOmics database was utilized to identify differentially expressed genes (DEGs) correlated with KRT80. Nomograms and Kaplan-Meier plots were constructed to evaluate the survival outcomes of patients diagnosed with LUAD. Moreover, TIMER was employed to conduct correlation analyses between KRT80 expression and immune cell infiltration, shedding light on the intricate interplay between KRT80 and the tumor microenvironment in LUAD. To ascertain the RNA and protein expression levels of KRT80 in LUAD and adjacent normal tissues, Reverse Transcription-quantitative Polymerase Chain Reaction (RT-qPCR) and immunohistochemistry techniques were employed, respectively.
    RESULTS: Scrutiny of the TCGA dataset revealed KRT80 up-regulation across pan-cancer tissues, notably elevated in LUAD compared to healthy lung tissues. This finding was validated in our clinical samples, where Kaplan-Meier survival curves indicated poorer survival rates for high KRT80 expression in LUAD. A positive correlation was found between the transcription level of KRT80 in LUAD samples and clinical parameters, such as lymph node metastasis stage, distant metastasis, and pathological stage. Survival, logistic regression, and Cox regression analyses emphasized the clinical prognostic significance of high KRT80 expression in LUAD. Nomogram results underscored the robust predictive potential of KRT80 for the survival of LUAD patients. Gene functional enrichment analyses mainly associated KRT80 with cytokine-cytokine receptor interactions, cell cycle, apoptosis, and chemokine signaling pathways. Based on the results of the immune infiltration analysis, it can be found that the expression of KRT80 is related to the immune cell subsets and survival rate of patients with LUAD.
    CONCLUSIONS: Our research revealed a significant upregulation of KRT80 in LUAD, with heightened KRT80 expression correlating with unfavorable prognosis. This study represents a comprehensive and systematic evaluation of KRT80 expression in LUAD, encompassing its prognostic and diagnostic significance, as well as underlying mechanisms. Our findings suggest that KRT80 may emerge as a novel prognostic and predictive biomarker in LUAD.
    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