lung adenocarcinoma (LUAD)

肺腺癌 (LUAD)
  • 文章类型: Journal Article
    肺腺癌(LUAD)是肺癌最常见的组织学亚型,结果表现出异质性和对治疗的不同反应。CD8T细胞始终存在于肿瘤发展的所有阶段,并在肿瘤微环境(TME)中发挥关键作用。我们的目的是研究CD8T细胞标记基因的表达谱,基于这些基因在LUAD中建立预后风险模型,并探讨其与免疫治疗反应的关系。
    通过利用来自癌症基因组图谱(TCGA)和基因表达综合(GEO)队列的表达数据和临床记录,我们确定了23个共有的预后基因.采用十种机器学习算法,我们生成了101个组合,最终选择最佳算法来构建名为riskScore的人工智能衍生预后签名。该选择基于三个测试队列的平均一致性指数(C指数)。
    RiskScore成为总生存期(OS)的独立危险因素,无进展间隔(PFI),无病间隔(DFI),和LUAD的疾病特异性生存率(DSS)。值得注意的是,与传统的临床变量相比,riskScore表现出明显的预测准确性。此外,我们观察到高风险RiskScore组与促进肿瘤的生物学功能之间呈正相关,较低的肿瘤突变负担(TMB),降低新抗原(NEO)负荷,和较低的微卫星不稳定性(MSI)分数,以及减少的免疫细胞浸润和TME内免疫逃避的可能性增加。意义重大,免疫表型(IPS)评分在风险亚组之间显示出显着差异,和riskScore根据患者的生存结局,有效地对IMtiven210和GSE135222免疫治疗队列中的患者进行了分层.此外,我们确定了可以针对特定风险亚组的潜在药物.
    总之,riskScore证明了其作为指导LUAD患者的临床管理和定制个性化治疗的强大和有前景的工具的潜力。
    UNASSIGNED: Lung adenocarcinoma (LUAD) stands as the most prevalent histological subtype of lung cancer, exhibiting heterogeneity in outcomes and diverse responses to therapy. CD8 T cells are consistently present throughout all stages of tumor development and play a pivotal role within the tumor microenvironment (TME). Our objective was to investigate the expression profiles of CD8 T cell marker genes, establish a prognostic risk model based on these genes in LUAD, and explore its relationship with immunotherapy response.
    UNASSIGNED: By leveraging the expression data and clinical records from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) cohorts, we identified 23 consensus prognostic genes. Employing ten machine-learning algorithms, we generated 101 combinations, ultimately selecting the optimal algorithm to construct an artificial intelligence-derived prognostic signature named riskScore. This selection was based on the average concordance index (C-index) across three testing cohorts.
    UNASSIGNED: RiskScore emerged as an independent risk factor for overall survival (OS), progression-free interval (PFI), disease-free interval (DFI), and disease-specific survival (DSS) in LUAD. Notably, riskScore exhibited notably superior predictive accuracy compared to traditional clinical variables. Furthermore, we observed a positive correlation between the high-risk riskScore group and tumor-promoting biological functions, lower tumor mutational burden (TMB), lower neoantigen (NEO) load, and lower microsatellite instability (MSI) scores, as well as reduced immune cell infiltration and an increased probability of immune evasion within the TME. Of significance, the immunophenoscore (IPS) score displayed significant differences among risk subgroups, and riskScore effectively stratified patients in the IMvigor210 and GSE135222 immunotherapy cohort based on their survival outcomes. Additionally, we identified potential drugs that could target specific risk subgroups.
    UNASSIGNED: In summary, riskScore demonstrates its potential as a robust and promising tool for guiding clinical management and tailoring individualized treatments for LUAD patients.
    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
    简介:环状RNA(circularRNAs)已被确定为癌症发展和进展的重要贡献者。这项研究的目的是检查circRNAcirc_BBS9在肺腺癌(LUAD)中的表达和临床意义,以及它潜在的行动模式。
    通过利用微阵列分析在LUAD的组织和细胞系中检查Circ_BBS9的表达,定量实时聚合酶链反应(qRT-PCR),和蛋白质印迹分析。在这项研究中,我们评估了circ_BBS9对LUAD细胞增殖的影响,以及它对铁死亡和肿瘤形成的影响。为了分析这些影响,我们采用CCK-8测定和铁凋亡测定。通过利用RNA下拉和质谱技术实现与Circ_BBS9相互作用的蛋白质的鉴定。使用生物信息学研究建立了包含circ_BBS9、miR-7150和IFIT3的推定调控网络。该研究还包括检查IFIT3的表达与免疫细胞侵袭之间的相关性。
    Circ_BBS9在LUAD组织和细胞系中显著下调。低circ_BBS9表达与不良预后相关。功能实验表明,circ_BBS9过表达在体外抑制LUAD细胞增殖,促进铁凋亡,在体内抑制肿瘤生长。机械上,发现circ_BBS9直接与IFIT3相互作用,并通过充当miR-7150的海绵来调节其表达。此外,LUAD中IFIT3的表达与免疫浸润呈正相关。
    Circ_BBS9已被确定为肺腺癌(LUAD)的肿瘤抑制因子,并有望作为诊断生物标志物。潜在的作用机制涉及通过与IFIT3的直接相互作用和与miR-7150的竞争性结合来调节铁凋亡和免疫微环境。上述发现为LUAD的病理生理学提供了新的观点,并强调了circ_BBS9作为治疗干预的潜在有价值的目标。
    UNASSIGNED: Introduction: Circular RNAs (circRNAs) have been identified as significant contributors to the development and advancement of cancer. The objective of this study was to examine the expression and clinical implications of circRNA circ_BBS9 in lung adenocarcinoma (LUAD), as well as its potential modes of action.
    UNASSIGNED: The expression of Circ_BBS9 was examined in tissues and cell lines of LUAD through the utilization of microarray profiling, quantitative real-time polymerase chain reaction (qRT-PCR), and western blot analysis. In this study, we assessed the impact of circ_BBS9 on the proliferation of LUAD cells, as well as its influence on ferroptosis and tumor formation. To analyze these effects, we employed CCK-8 assays and ferroptosis assays. The identification of proteins that interact with Circ_BBS9 was achieved through the utilization of RNA pull-down and mass spectrometry techniques. A putative regulatory network comprising circ_BBS9, miR-7150, and IFIT3 was established using bioinformatics study. The investigation also encompassed the examination of the correlation between the expression of IFIT3 and the invasion of immune cells.
    UNASSIGNED: Circ_BBS9 was significantly downregulated in LUAD tissues and cell lines. Low circ_BBS9 expression correlated with poor prognosis. Functional experiments showed that circ_BBS9 overexpression inhibited LUAD cell proliferation and promoted ferroptosis in vitro and suppressed tumor growth in vivo. Mechanistically, circ_BBS9 was found to directly interact with IFIT3 and regulate its expression by acting as a sponge for miR-7150. Additionally, IFIT3 expression correlated positively with immune infiltration in LUAD.
    UNASSIGNED: Circ_BBS9 has been identified as a tumor suppressor in lung adenocarcinoma (LUAD) and holds promise as a diagnostic biomarker. The potential mechanism of action involves the modulation of ferroptosis and the immunological microenvironment through direct interaction with IFIT3 and competitive binding to miR-7150. The aforementioned findings offer new perspectives on the pathophysiology of LUAD and highlight circ_BBS9 as a potentially valuable target for therapeutic interventions.
    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(NK)细胞是先天免疫系统的重要组成部分,对于对抗感染和肿瘤生长至关重要,使它们在癌症预后和免疫疗法中至关重要。我们试图通过进行单细胞RNA测序分析来了解肺腺癌(LUAD)中NK细胞的多种特征。
    使用多原发性肺癌(MPLCs)的scRNA-seq数据集,我们检查了两个主要的NK细胞群,NK1和NK2,比较了422个差异表达的NK标记基因的表达谱。我们鉴定了8个基因(SPON2,PLEKHG3,CAMK2N1,RAB27B,CTBP2,EFHD2,GOLM1和PLOD1)可区分NK1和NK2细胞。预后特征,NK基因签名(NKGS)评分,通过LASSOCox回归建立。高NKGS评分与TCGA-LUAD患者总体生存率较差相关,并在其他数据集(GSE31210和GSE14814)中得到一致验证。
    功能分析显示,在高NKGS评分组中,与TGF-β信号通路相关的基因富集。此外,高NKGS评分与免疫逃避机制驱动的免疫抑制肿瘤微环境(TME)相关。我们还观察到高风险NKGS组中T细胞受体(TCR)的多样性减少,表明炎症和风险评分之间呈负相关。
    这项研究引入了创新的NKGS评分,从NK2细胞中分化出NK1。高NKGS评分与TGF-β途径相关,并提供了对LUAD预后和免疫活性的见解。
    UNASSIGNED: Natural Killer (NK) cells are vital components of the innate immune system, crucial for combating infections and tumor growth, making them pivotal in cancer prognosis and immunotherapy. We sought to understand the diverse characteristics of NK cells within lung adenocarcinoma (LUAD) by conducting single-cell RNA sequencing analyses.
    UNASSIGNED: Using the scRNA-seq dataset for multiple primary lung cancers (MPLCs), we examined two major NK cell groups, NK1 and NK2, comparing the expression profiles of 422 differentially expressed NK signature genes. We identified eight genes (SPON2, PLEKHG3, CAMK2N1, RAB27B, CTBP2, EFHD2, GOLM1, and PLOD1) that distinguish NK1 from NK2 cells. A prognostic signature, the NK gene signature (NKGS) score, was established through LASSO Cox regression. High NKGS scores were linked to poorer overall survival in TCGA-LUAD patients and consistently validated in other datasets (GSE31210 and GSE14814).
    UNASSIGNED: Functional analysis revealed an enrichment of genes related to the TGF-β signaling pathway in the high NKGS score group. Moreover, a high NKGS score correlated with an immunosuppressive tumor microenvironment (TME) driven by immune evasion mechanisms. We also observed reduced T-cell receptor (TCR) repertoire diversity in the high-risk NKGS group, indicating a negative association between inflammation and risk score.
    UNASSIGNED: This study introduced the innovative NKGS score, differentiating NK1 from NK2 cells. High NKGS scores were associated with the TGF-β pathway and provided insights into LUAD prognosis and immune activities.
    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
    肺腺癌是一种高死亡率和预后不良的恶性肿瘤。然而,白三烯C4合成酶(LTC4S)在肺癌中的作用仍未被研究.
    使用GEPIA在线数据库分析LTC4S在LUAD中的表达和预后价值。随后,通过功能获得实验检查LTC4S在肺癌细胞中的功能,使用测定法来评估肿瘤的恶性行为。体内皮下异种移植实验用于研究LTC4S的功能。然后,通过GSEA分析肿瘤标志通路。Western印迹分析用于验证LTC4S对mTORC1途径的影响。最后,用cBioPortal分析LTC4SmRNA与甲基化的相关性。qRT-PCR,使用ChIP-qPCR和ChIP-Atlas验证LUAD细胞中LTC4S低表达的调控因子。
    LTC4S在LUAD中表达显著降低,预后意义良好。LTC4S与LUAD的临床分期相关,随着TNM分期,其表达量逐渐降低,且显著降低。LTC4S共表达基因与Ras信号通路密切相关,和MAPK信号通路。LTC4S的过表达在体外和体内抑制癌症恶性表型和肿瘤生长。GSEA分析和Westernblot检测提示LTC4S激活的mTORC1信号通路在LUAD中的低表达。此外,与正常组织相比,LUAD组织中LTC4S的DNA甲基化水平显着升高。DNMT3A对LTC4S启动子的过度甲基化导致LUAD中LTC4S的表达降低。
    总而言之,LTC4S的低表达是一个不利的预后标志物,也是LTC4S控制LUAD进展的关键功能.这凸显了探索在LUAD靶向治疗中操纵LTC4S的临床益处的前景。
    UNASSIGNED: Lung adenocarcinoma is a malignancy characterized by high mortality rates and unfavorable prognosis. However, the role of Leukotriene C4 Synthase (LTC4S) in lung cancer remains uninvestigated.
    UNASSIGNED: The expression and prognostic value of LTC4S in LUAD were analyzed using the GEPIA online database. Subsequently, the function of LTC4S in lung cancer cells was examined through gain-of function experiments, using assays to evaluate tumor malignant behavior. Subcutaneous xenograft experiments in vivo was used for investigating the functions of LTC4S. Then, tumor hallmark pathways were analyzed by GSEA. Western blot assay was used to validate the impact of LTC4S on mTORC1 pathway. Finally, the correlation of mRNA and methylation of LTC4S were analyzed by cBioPortal. qRT-PCR, ChIP-qPCR and ChIP-Atlas were used to verify the regulation factors of LTC4S low expression in LUAD cells.
    UNASSIGNED: LTC4S presented significant decreased expression and favorable prognostic significance in LUAD. LTC4S was correlated with clinical stages in LUAD, which showed decreased expression gradually and significantly along with TNM stages. LTC4S-co-expressed genes were closely related to Ras signaling pathway, and MAPK signaling pathway. Overexpression of LTC4S inhibited cancer malignant phenotype and tumor growth in vitro and vivo. GSEA analysis and Western blot assay suggested low expression of LTC4S activated mTORC1 signaling pathway in LUAD. Moreover, the DNA methylation level of LTC4S in LUAD tissue was markedly elevated compared to normal tissue. The hypermethylation of the LTC4S promoter by DNMT3A leads to the decreased expression of LTC4S in LUAD.
    UNASSIGNED: In conclusion, low expression of LTC4S serves as an unfavorable prognostic marker and the critical function of LTC4S in controlling the progression of LUAD. This highlights the promise for exploring the clinical benefits of manipulating LTC4S in LUAD targeted therapies.
    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
    肺癌是人类最常见的癌症之一,肺腺癌(LUAD)已成为最常见的肺癌组织学类型。免疫逃逸促进LUAD从早期到转移晚期的进展,并且是改善靶向免疫检测点的免疫治疗的临床结果的主要障碍之一。本研究旨在探讨在肺腺癌中异常表达的免疫逃逸相关基因,为预测肺腺癌的预后提供帮助。
    LUAD患者的RNA数据和相关临床细节来自癌症基因组图谱(TCGA)数据库。通过加权基因共表达网络分析(WGCNA),筛选了3112个关键基因,并与从先前研究中获得的182个免疫逃逸基因相交,以鉴定免疫逃逸相关基因(IERG)。通过基因本体论(GO)和京都基因和基因组百科全书(KEGG)分析,系统地探讨了IERGs在LUAD中的作用,用于丰富IERGs的相关途径。最小绝对收缩和选择算子(LASSO)算法和多变量Cox回归分析用于识别关键的预后基因,并构建了预后风险模型。使用表达数据(ESTIMATE)和微环境细胞群(MCP)计数器方法(其可以准确地评估八个免疫细胞群和两个基质细胞群的量)估计恶性肿瘤组织中的基质和免疫细胞用于分析高和低风险亚组的肿瘤免疫状态。通过使用人蛋白质图谱(HPA)数据库测定肺癌样品中差异表达基因的蛋白质表达水平。构造了一个列线图,并通过基因表达综合(GEO)数据集GSE72094和GSE30219验证了预后风险模型。
    获得20个差异表达的IERG。对这20个IERG的GO分析显示,它们主要与免疫系统过程的调节有关,免疫反应,以及γ-干扰素在介导信号通路和凋亡信号通路中的富集;同时,KEGG分析显示IERG与坏死性凋亡有关,抗原加工和呈递,程序性细胞死亡配体1(PD-L1)在肿瘤中的表达和程序性细胞死亡1(PD-1)途径,细胞因子-细胞因子受体相互作用,辅助性T细胞1(Th1)和Th2分化,和肿瘤坏死因子信号通路。使用LASSO和Cox回归分析,我们构建了一个可以预测LUAD患者预后的四基因模型,模型通过验证队列进行验证.HPA数据库免疫组化成果显示,AHSA1和CEP55在正常肺组织中低表达,而在肺癌组织中高表达。
    我们构建了一个基于IERG的LUAD预后预测模型。在鉴定的基因中,CEP55和AHSA1可能是潜在的预后和治疗靶点,减少其表达可能是治疗LUAD的一种新方法。
    UNASSIGNED: Lung cancer is one of the most common cancers in humans, and lung adenocarcinoma (LUAD) has become the most common histological type of lung cancer. Immune escape promotes progression of LUAD from the early to metastatic late stages and is one of the main obstacles to improving clinical outcomes for immunotherapy targeting immune detection points. Our study aims to explore the immune escape related genes that are abnormally expressed in lung adenocarcinoma, providing assistance in predicting the prognosis of lung adenocarcinoma and targeted.
    UNASSIGNED: RNA data and related clinical details of patients with LUAD were obtained from The Cancer Genome Atlas (TCGA) database. Through weighted gene coexpression network analysis (WGCNA), 3112 key genes were screened and intersected with 182 immune escape genes obtained from a previous study to identify the immune escape-related genes (IERGs). The role of IERGs in LUAD was systematically explored through gene ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) analyses, which were used to enrich the relevant pathways of IERGs. The least absolute shrinkage and selection operator (LASSO) algorithm and multivariate Cox regression analysis were used to identify the key prognostic genes, and a prognostic risk model was constructed. Estimation of Stromal and Immune Cells in Malignant Tumor Tissues Using Expression Data (ESTIMATE) and microenvironment cell populations (MCP) counter methods (which can accurately assess the amount of eight immune cell populations and two stromal cell groups) were used to analyze the tumor immune status of the high and low risk subgroups. The protein expression level of the differentially expressed genes in lung cancer samples was determined by using the Human Protein Atlas (HPA) database. A nomogram was constructed, and the prognostic risk model was verified via the Gene Expression Omnibus (GEO) datasets GSE72094 and GSE30219.
    UNASSIGNED: Twenty differentially expressed IERGs were obtained. GO analysis of these 20 IERGs revealed that they were mainly associated with the regulation of immune system processes, immune responses, and interferon-γ enrichment in mediating signaling pathways and apoptotic signaling pathways; meanwhile, KEGG analysis revealed that IERGs were associated with necroptosis, antigen processing and presentation, programmed cell death ligand 1 (PD-L1) expression and programmed cell death 1 (PD-1) pathway in tumors, cytokine-cytokine receptor interactions, T helper cell 1 (Th1) and Th2 differentiation, and tumor necrosis factor signaling pathways. Using LASSO and Cox regression analysis, we constructed a four-gene model that could predict the prognosis of patients with LUAD, and the model was validated with a validation cohort. The immunohistochemical results of the HPA database showed that AHSA1 and CEP55 had low expression in normal lung tissue but high expression in lung cancer tissue.
    UNASSIGNED: We constructed an IERG-based model for predicting the prognosis of LUAD. Among the genes identified, CEP55 and AHSA1 may be potential prognostic and therapeutic targets, and reducing their expression may represent a novel approach in the treatment of 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
    肺癌是肺部最常见的原发性恶性肿瘤,作为对人口健康构成最大威胁的恶性肿瘤之一,近年来发病率一直很高。先前的研究表明,KLRB1在肺腺癌中转录抑制,并与肺腺癌的预后相关。本研究的目的是探讨KLRB1影响肺腺癌恶性表型如免疫浸润的内在机制,扩散,生长和转移。
    我们在公开数据库中评估了KLRB1的表达水平,并研究了其与临床和病理变量的关联。随后进行富集分析以研究可能的信号通路及其相关的生物学功能。统计分析,包括Spearman相关性和多基因预测模型的应用,用于评估KLRB1的表达与免疫细胞浸润之间的关系。使用Kaplan-Meier存活曲线评估KLRB1的诊断和预后价值,诊断受体工作特征(ROC)曲线,直方图模型,和Cox回归分析。收集肺腺癌(LUAD)患者的标本,通过蛋白质印迹分析检测KLRB1的表达水平,并通过实时定量逆转录聚合酶链反应(RT-qPCR)在mRNA水平检测KLRB1的表达水平。小干扰RNA(siRNA)用于沉默基因表达,还有Transwell,随后进行细胞计数试剂盒-8(CCK-8)和集落形成测定以分析KLRB1对LUAD细胞迁移的影响。侵袭和增殖。
    KLRB1在肺癌组织中的表达低于周围健康组织。发现在低和高KLRB1表达组中差异表达的基因在与免疫相关的途径中显著富集。KLRB1对MAPK/ERK信号通路有影响,从而调节LUAD细胞的生长和增殖。KLRB1表达与预后有关,免疫浸润,以及LUAD中的细胞迁移和增殖。
    证据表明,KLRB1与LUAD患者的预后和免疫浸润之间存在相关性。
    UNASSIGNED: Lung cancer is the most common primary malignant tumor of the lung, and as one of the malignant tumors that pose the greatest threat to the health of the population, the incidence rate has remained high in recent years. Previous studies have shown that KLRB1 is transcriptionally repressed in lung adenocarcinoma and correlates with lung adenocarcinoma prognosis. The objective of this study is to investigate the intrinsic mechanisms by which KLRB1 affects the malignant phenotypes of lung adenocarcinoma such as immune infiltration, proliferation, growth and metastasis.
    UNASSIGNED: We assessed the expression levels of KLRB1 in publicly available databases and investigated its associations with clinical and pathological variables. Enrichment analysis was subsequently conducted to investigate possible signaling pathways and their associated biological functions. Statistical analysis, including Spearman correlation and the application of multigene prediction models, was utilized to assess the relationship between the expression of KLRB1 and the infiltration of immune cells. The diagnostic and prognostic value of KLRB1 was evaluated using Kaplan-Meier survival curves, diagnostic receptor operating characteristic (ROC) curves, histogram models, and Cox regression analysis. Specimens from lung adenocarcinoma (LUAD) patients were collected, the expression level of KLRB1 was detected by protein blotting analysis, and the expression level of KLRB1 was detected at the mRNA level by real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR). Small interfering RNA (siRNA) was used to silence gene expression, and Transwell, Cell Counting Kit-8 (CCK-8) and colony formation assays were subsequently performed to analyze the effects of KLRB1 on LUAD cell migration, invasion and proliferation.
    UNASSIGNED: KLRB1 expression was lower in lung cancer tissue than in surrounding healthy tissue. Genes differentially expressed in the low and high KLRB1 expression groups were found to be significantly enriched in pathways related to immunity. KLRB1 exerted an impact on the MAPK/ERK signaling pathway, thereby modulating the growth and proliferation of LUAD cells. KLRB1 expression is linked to prognosis, immune infiltration, and cell migration and proliferation in LUAD.
    UNASSIGNED: The evidence revealed a correlation between KLRB1 and both prognosis and immune infiltration in LUAD patients.
    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
    驱动基因是靶向治疗功效的重要预测因子。检测肺腺癌(LUAD)患者的驱动基因突变可以帮助筛选靶向药物并提高患者的生存效益。本研究旨在探讨LUAD中驱动基因的突变特征及其与临床病理特征的相关性。
    在2019年7月至2022年9月期间,共从邵逸夫爵士医院选择了440名LUAD患者。使用下一代测序技术分析术后组织标本的基因突变,聚焦,包括表皮生长因子受体EGFR,ALK,ROS1,RET,KRAS,MET,BRAF,HER2、PIK3CA和NRAS。同时,收集并整理临床病理资料进行多维相关分析.
    440名LUAD患者,48例患者未检测到驱动基因突变.驱动基因突变的患者比例高达89.09%。前三个驱动基因突变是EGFR,KRAS,和MET。共检测到69种类型的EGFR突变,并分布在蛋白酪氨酸激酶催化域(56,81.16%),富弗林蛋白酶样半胱氨酸区(9,13.04%),受体结合域(3,4.35%),和EGFR跨膜结构域(1,1.45%)。343例LUAD患者发生单基因位点突变,但是突变基因类型涵盖了所有测试基因。我们的研究结果表明,EGFR突变更常见于非吸烟和女性患者(P<0.01)。KRAS突变在男性患者和吸烟者中更为普遍(P<0.01)。ROS1突变的肿瘤直径较大(P<0.01),RET突变在吸烟者中更为普遍(P<0.05)。
    LUAD患者表现出不同的基因突变,这可能同时发生。多种突变的综合分析对于疾病的准确诊断和有效治疗至关重要。使用NGS可以大大扩展我们对基因突变的理解,并促进对多个基因突变的综合分析,为有针对性的治疗方法提供关键证据。
    UNASSIGNED: Driver genes are essential predictors of targeted therapeutic efficacy. Detecting driver gene mutations in lung adenocarcinoma (LUAD) patients can help to screen for targeted drugs and improve patient survival benefits. This study aims to investigate the mutation characterization of driver genes and their correlation with clinicopathological features in LUAD.
    UNASSIGNED: A total of 440 LUAD patients were selected from Sir Run Run Shaw Hospital between July 2019 and September 2022. Postoperative tissue specimens were analyzed for gene mutations using next-generation sequencing technology, focusing, including epidermal growth factor receptor EGFR, ALK, ROS1, RET, KRAS, MET, BRAF, HER2, PIK3CA and NRAS. At the same time, clinicopathological data were collected and organized for multidimensional correlation analysis.
    UNASSIGNED: Of 440 LUAD patients, driver gene mutations were not detected in 48 patients. The proportion of patients with driver gene mutations was as high as 89.09%. The top three driver genetic mutations were EGFR, KRAS, and MET. Sixty-nine types of EGFR mutations were detected and distributed in the protein tyrosine kinase catalytic domain (56, 81.16%), Furin-like cysteine-rich region (9, 13.04%), receptor binding domain (3, 4.35%), and EGFR transmembrane domain (1, 1.45%). Single gene locus mutation occurred in 343 LUAD patients, but the mutation gene types covered all tested genes. Our findings showed that EGFR mutations were more commonly observed in non-smoking and female patients (P<0.01), KRAS mutations were more prevalent in male patients and smokers (P<0.01), ROS1 mutations had larger tumor diameters (P<0.01) and RET mutations were more prevalent in smokers (P<0.05).
    UNASSIGNED: LUAD patients exhibit diverse genetic mutations, which may co-occur simultaneously. Integrated analysis of multiple mutations is essential for accurate diagnosis and effective treatment of the disease. The use of NGS can significantly expand our understanding of gene mutations and facilitate integrated analysis of multiple gene mutations, providing critical evidence for targeted treatment methods.
    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
    免疫疗法已成为肺癌患者的一线治疗选择,但有些病人对免疫疗法反应不佳,尤其是肺腺癌(LUAD)患者。需要新的工具来筛查LUAD患者对免疫治疗的潜在反应者,从而更好地预测预后,指导临床决策。尽管已经做出了许多努力来预测LUAD患者的反应性,结果有限。在免疫疗法时代,本研究试图通过在具有不同免疫治疗反应的患者中利用差异表达基因(DEGs)构建一种新的LUAD预后模型.
    598例LUAD患者的转录组数据从癌症基因组图谱(TCGA)数据库下载,其中包括539个肿瘤样本和59个正常对照样本,平均随访时间为29.69个月(63.1%的患者在随访结束时仍存活).分析了其他数据源,包括来自基因表达综合(GEO)数据库的三个数据集,免疫疗法应答者和非应答者之间的DEG被鉴定和筛选。应用单变量Cox回归分析,TCGA队列作为训练集,GSE72094队列作为验证集,和最小绝对收缩和选择算子(LASSO)Cox回归被应用在预后相关基因中,这些基因满足过滤标准以建立预后公式,然后用时间相关的接收器工作特性(ROC)分析进行测试。用基因本体论(GO)和京都基因和基因组百科全书(KEGG)富集分析分析了预后相关基因的富集途径,和肿瘤免疫微环境(TIME),肿瘤突变负担,在R(R统计计算基金会)中使用适当的软件包完成药物敏感性测试。最后,建立了包含预后公式的列线图。
    总共确定了1,636个DEG,提取了1,163个与预后相关的DEGs,选择34个DEGs并将其纳入免疫治疗反应性相关风险评分(IRRS)公式.IRRS公式在预测LUAD患者的总体预后方面具有良好的性能,并且在所有LUAD亚组中都具有出色的预后预测性能。此外,IRRS公式可以预测LUAD患者的抗癌药物敏感性和免疫治疗反应性。机械上,两个IRRS组之间的免疫微环境差异很大;高IRRS组和低IRRS组之间最显著变化的途径是核糖核蛋白复合物生物发生,与TP53和TTN突变负荷密切相关。此外,我们建立了一个包含IRRS的列线图,年龄,性别,临床分期,T-stage,N级,和M阶段作为可以预测1年预后的预测因子,3年,LUAD患者的5年生存率,曲线下面积(AUC)分别为0.718、0.702和0.68。
    本研究建立的模型可以预测LUAD患者的预后,帮助识别对抗癌药物和免疫疗法反应良好的患者,并作为指导临床决策的宝贵工具。
    UNASSIGNED: Immune therapy has become first-line treatment option for patients with lung cancer, but some patients respond poorly to immune therapy, especially among patients with lung adenocarcinoma (LUAD). Novel tools are needed to screen potential responders to immune therapy in LUAD patients, to better predict the prognosis and guide clinical decision-making. Although many efforts have been made to predict the responsiveness of LUAD patients, the results were limited. During the era of immunotherapy, this study attempts to construct a novel prognostic model for LUAD by utilizing differentially expressed genes (DEGs) among patients with differential immune therapy responses.
    UNASSIGNED: Transcriptome data of 598 patients with LUAD were downloaded from The Cancer Genome Atlas (TCGA) database, which included 539 tumor samples and 59 normal control samples, with a mean follow-up time of 29.69 months (63.1% of patients remained alive by the end of follow-up). Other data sources including three datasets from the Gene Expression Omnibus (GEO) database were analyzed, and the DEGs between immunotherapy responders and nonresponders were identified and screened. Univariate Cox regression analysis was applied with the TCGA cohort as the training set and GSE72094 cohort as the validation set, and least absolute shrinkage and selection operator (LASSO) Cox regression were applied in the prognostic-related genes which fulfilled the filter criteria to establish a prognostic formula, which was then tested with time-dependent receiver operating characteristic (ROC) analysis. Enriched pathways of the prognostic-related genes were analyzed with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, and tumor immune microenvironment (TIME), tumor mutational burden, and drug sensitivity tests were completed with appropriate packages in R (The R Foundation of Statistical Computing). Finally, a nomogram incorporating the prognostic formula was established.
    UNASSIGNED: A total of 1,636 DEGs were identified, 1,163 prognostic-related DEGs were extracted, and 34 DEGs were selected and incorporated into the immunotherapy responsiveness-related risk score (IRRS) formula. The IRRS formula had good performance in predicting the overall prognoses in patients with LUAD and had excellent performance in prognosis prediction in all LUAD subgroups. Moreover, the IRRS formula could predict anticancer drug sensitivity and immunotherapy responsiveness in patients with LUAD. Mechanistically, immune microenvironments varied profoundly between the two IRRS groups; the most significantly varied pathway between the high-IRRS and low-IRRS groups was ribonucleoprotein complex biogenesis, which correlated closely with the TP53 and TTN mutation burdens. In addition, we established a nomogram incorporating the IRRS, age, sex, clinical stage, T-stage, N-stage, and M-stage as predictors that could predict the prognoses of 1-year, 3-year, and 5-year survival in patients with LUAD, with an area under curve (AUC) of 0.718, 0.702, and 0.68, respectively.
    UNASSIGNED: The model we established in the present study could predict the prognosis of LUAD patients, help to identify patients with good responses to anticancer drugs and immunotherapy, and serve as a valuable tool to guide clinical decision-making.
    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)是全球最常见的癌症类型之一。蛋白酶体激活剂亚基3(PSME3)是蛋白酶体激活剂的一个亚基,PSME3的变化可导致生物体内许多疾病的发展。然而,PSME3在LUAD中的具体机制尚未阐明。本研究初步揭示了PSME3促进肺腺癌进展的机制,这为临床治疗提供了潜在的分子靶标。
    通过生物信息学分析评估LUAD细胞和组织中PSME3的表达,免疫组织化学(IHC),蛋白质印迹(WB),和定量实时聚合酶链反应(qRT-PCR)。通过一系列功能实验来评估PSME3敲低和过表达对LUAD细胞增殖的影响,迁移,和凋亡。通过转录组测序和WB实验探索了PSME3的潜在机制。
    在这项研究中,我们的初步研究结果表明,PSME3在LUAD中表达异常高,并且与患者预后不良相关.Further,我们发现PSME3的下调显著抑制了LUAD细胞的增殖,通过裸鼠皮下肿瘤形成实验证实的效果。同样,下调PSME3后,LUAD细胞的侵袭和迁移速率显著降低。使用流式细胞术,我们发现PSME3的敲除导致细胞周期停滞在G1/S期。通过转录组测序,我们发现转化生长因子-β(TGF-β)/SMAD信号通路与LUAD密切相关,然后我们使用WB分析验证了该途径。
    我们证明PSME3在LUAD中异常高表达,与患者预后不良有关;因此,靶向PSME3治疗LUAD可能是一种新的治疗方法.
    UNASSIGNED: Lung adenocarcinoma (LUAD) is one of the most common types of cancer worldwide. Proteasome activator subunit 3 (PSME3) is a subunit of a proteasome activator, and changes in PSME3 can lead to the development of many diseases in organisms. However, the specific mechanism of PSME3 in LUAD has not yet been elucidated. This study initially revealed the mechanism of PSME3 promoting the progression of lung adenocarcinoma, which provided a potential molecular target for clinical treatment.
    UNASSIGNED: PSME3 expression in LUAD cells and tissues was assessed by bioinformatics analysis, immunohistochemistry (IHC), Western blotting (WB), and quantitative real time polymerase chain reaction (qRT-PCR). A series of functional experiments were used to evaluate the effects of PSME3 knockdown and overexpression on LUAD cell proliferation, migration, and apoptosis. The potential mechanism of PSME3 was explored by transcriptome sequencing and WB experiments.
    UNASSIGNED: In this study, our initial findings indicated that PSME3 expression was abnormally high in LUAD and was associated with poor patient prognosis. Further, we found that the downregulation of PSME3 significantly inhibited LUAD cell proliferation, an effect that was verified by subcutaneous tumor formation experiments in nude mice. Similarly, the rate of invasion and migration of LUAD cells significantly decreased after the downregulation of PSME3. Using flow cytometry, we found that the knockdown of PSME3 caused cell cycle arrest at the G1/S phase. Through transcriptome sequencing, we found that the transforming growth factor-beta (TGF-β)/SMAD signaling pathway was closely related to LUAD, and we then validated the pathway using WB assays.
    UNASSIGNED: We demonstrated that PSME3 was abnormally highly expressed in LUAD and related to poor patient prognosis; therefore, targeting PSME3 in the treatment of LUAD may represent a novel therapeutic approach.
    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)是全球最常见的恶性肿瘤之一,不利的治疗结果。已知肽基氨酰异构酶F(PPIF)通过调节癌细胞的生物能学和线粒体通透性来影响肿瘤进展的恶性特征;然而,其在LUAD中的作用尚不清楚。我们的研究旨在探讨临床意义,肿瘤增殖,以及PPIF在LUAD中的免疫调节功能。
    使用生物信息学分析评估了PPIF在LUAD组织和细胞中的表达,免疫组织化学(IHC),和西方印迹。进行生存曲线分析以检查PPIF表达与LUAD之间的预后关联。通过分析PPIF表达和免疫细胞浸润来评估PPIF在LUAD中的免疫调节作用。对PPIF进行了一系列功能增益和功能丧失实验,以研究其在LUAD中的体外和体内生物学功能。通过功能富集分析和Western印迹分析,描述了PPIF对LUAD影响的潜在机制。与正常对照相比,
    PPIF在LUAD组织中表现出过表达。生存曲线分析显示,表现出较高PPIF表达的LUAD患者表现出总体生存期降低和无进展间隔较短。PPIF与调节免疫细胞浸润有关,特别是在调节T辅助1-T辅助2细胞平衡。功能上,发现PPIF可促进肿瘤细胞增殖并促进细胞周期进程。此外,PPIF可能通过靶向FOXO3a/PINK1-Parkin信号通路阻止线粒体自噬。
    这项研究的结果表明,预后相关基因PPIF可能在调节LUAD细胞增殖中起重要作用,肿瘤相关免疫细胞浸润,和线粒体自噬,因此PPIF可能是LUAD的一个有希望的治疗靶点。
    UNASSIGNED: Lung adenocarcinoma (LUAD) is among the most prevalent malignancies worldwide, with unfavorable treatment outcomes. Peptidyl-prolyl isomerase F (PPIF) is known to influence the malignancy traits of tumor progression by modulating the bioenergetics and mitochondrial permeability in cancer cells; however, its role in LUAD remains unclear. Our study seeks to investigate the clinical significance, tumor proliferation, and immune regulatory functions of PPIF in LUAD.
    UNASSIGNED: The expression of PPIF in LUAD tissues and cells was assessed using bioinformatics analysis, immunohistochemistry (IHC), and Western blotting. Survival curve analysis was conducted to examine the prognostic association between PPIF expression and LUAD. The immunomodulatory role of PPIF in LUAD was assessed through the analysis of PPIF expression and immune cell infiltration. A series of gain- and loss-of-function experiments were conducted on PPIF to investigate its biological functions in LUAD both in vitro and in vivo. The mechanisms underlying PPIF\'s effects on LUAD were delineated through functional enrichment analysis and Western blotting assays.
    UNASSIGNED: PPIF exhibited overexpression in LUAD tissues compared to normal controls. Survival curve analysis revealed that patients with LUAD exhibiting higher PPIF expression demonstrated decreased overall survival and a shorter progression-free interval. PPIF was implicated in modulating immune cell infiltration, particularly in regulating the T helper 1-T helper 2 cell balance. Functionally, PPIF was discovered to promote tumor cell proliferation and advance cell-cycle progression. Furthermore, PPIF could impede mitophagy by targeting the FOXO3a/PINK1-Parkin signaling pathway.
    UNASSIGNED: The findings of this study indicate that the prognosis-related gene PPIF may have a significant role in the regulation of LUAD cell proliferation, tumor-associated immune cell infiltration, and mitophagy, and thus PPIF may be a promising therapeutic target of LUAD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号