
Citation: Kewei Ni, Gaozhong Sun. The identification of key biomarkers in patients with lung adenocarcinoma based on bioinformatics[J]. Mathematical Biosciences and Engineering, 2019, 16(6): 7671-7687. doi: 10.3934/mbe.2019384
[1] | Yong Ding, Jian-Hong Liu . The signature lncRNAs associated with the lung adenocarcinoma patients prognosis. Mathematical Biosciences and Engineering, 2020, 17(2): 1593-1603. doi: 10.3934/mbe.2020083 |
[2] | Gaozhong Sun, Tongwei Zhao . Lung adenocarcinoma pathology stages related gene identification. Mathematical Biosciences and Engineering, 2020, 17(1): 737-746. doi: 10.3934/mbe.2020038 |
[3] | Kunpeng Li, Zepeng Wang, Yu Zhou, Sihai Li . Lung adenocarcinoma identification based on hybrid feature selections and attentional convolutional neural networks. Mathematical Biosciences and Engineering, 2024, 21(2): 2991-3015. doi: 10.3934/mbe.2024133 |
[4] | Jian Huang, Zheng-Fu Xie . Identification of SSBP1 as a prognostic marker in human lung adenocarcinoma using bioinformatics approaches. Mathematical Biosciences and Engineering, 2022, 19(3): 3022-3035. doi: 10.3934/mbe.2022139 |
[5] | Pingping Song, Jing Chen, Xu Zhang, Xiaofeng Yin . Construction of competitive endogenous RNA network related to circular RNA and prognostic nomogram model in lung adenocarcinoma. Mathematical Biosciences and Engineering, 2021, 18(6): 9806-9821. doi: 10.3934/mbe.2021481 |
[6] | Linbo Zhang, Mei Xin, Peng Wang . Identification of a novel snoRNA expression signature associated with overall survival in patients with lung adenocarcinoma: A comprehensive analysis based on RNA sequencing dataset. Mathematical Biosciences and Engineering, 2021, 18(6): 7837-7860. doi: 10.3934/mbe.2021389 |
[7] | Fang Niu, Zongwei Liu, Peidong Liu, Hongrui Pan, Jiaxue Bi, Peng Li, Guangze Luo, Yonghui Chen, Xiaoxing Zhang, Xiangchen Dai . Identification of novel genetic biomarkers and treatment targets for arteriosclerosis-related abdominal aortic aneurysm using bioinformatic tools. Mathematical Biosciences and Engineering, 2021, 18(6): 9761-9774. doi: 10.3934/mbe.2021478 |
[8] | Yanping Xie, Zhaohui Dong, Junhua Du, Xiaoliang Zang, Huihui Guo, Min Liu, Shengwen Shao . The relationship between mouse lung adenocarcinoma at different stages and the expression level of exosomes in serum. Mathematical Biosciences and Engineering, 2020, 17(2): 1548-1557. doi: 10.3934/mbe.2020080 |
[9] | Mengyang Han, Xiaoli Wang, Yaqi Li, Jianjun Tan, Chunhua Li, Wang Sheng . Identification of coagulation-associated subtypes of lung adenocarcinoma and establishment of prognostic models. Mathematical Biosciences and Engineering, 2023, 20(6): 10626-10658. doi: 10.3934/mbe.2023470 |
[10] | Ahmed Hammad, Mohamed Elshaer, Xiuwen Tang . Identification of potential biomarkers with colorectal cancer based on bioinformatics analysis and machine learning. Mathematical Biosciences and Engineering, 2021, 18(6): 8997-9015. doi: 10.3934/mbe.2021443 |
Lung cancer, the most common cause of cancer-related mortality around the world, giving rise to over a million deaths each year [1]. Lung adenocarcinoma (LUAD) is one of the most common histological types of lung cancer [2]. It is well acknowledged that smoking is the major risk factor and cause of LUAD. Unexpectedly, more LUAD cases occur proportionally in people without smoking history (defined as less than 100 cigarettes in a lifetime) following the decreased smoking rates [3]. The pathogenesis of LUAD is a complicated process implicated with the progressive accumulation of gene alterations that pinpoint various molecular and cellular events involving autophagy, endoplasmic reticulum stress, oxidative stress, and abnormal cell cycle [4,5,6,7,8].
In recent years, apart from traditional surgery, chemotherapy and radiotherapy, targeted therapy has also greatly improved treatment for patients whose tumours harbour somatically activated oncogenes including mutant EGFR1, ERBB2 and BRAF or translocated RET, ALK or ROS1, however, the likelihood of a complete cure for the patients with LUAD is very slim [9,10]. Hence, the detection of early-stage biomarkers and identification of core therapeutic target appears significant to decrease LUAD-related deaths.
The recent high-throughput gene microarray has been widely employed to screen the differentially expressed genes (DEGs) between normal samples and tumor samples in human beings and animal models, which makes it accessible for us to further explore the entire molecular alterations of tumors at multiple levels involving DNA, RNA, proteins, epigenetic alterations, and metabolism [11]. However, there still exist obstacles to put these microarrays in application in clinic for the reason that the number of DEGs identified by gene profiling were huge and the statistical analyses were also too complicated [12,13]. Therefore, it is urgent to verify a proper number of genes and develop a suitable approach which can be operated by routine assay in clinic.
In this study, we downloaded the GSE10072 from Gene Expression Omnibus (GEO) and applied bioinformatics analysis to screen the DEGs between LUAD and normal control. Moreover, we carried out the functional analysis of these DEGs, including biological process (BP), molecular function (MF), cellular component (CC) and KEGG pathways. We chose top5 up-regulated and top5 down-regulated DEGs to make the overall survival (OS) analysis and stage plot. Finally, we used STRING to construct the protein-protein interaction (PPI) network to identify the hub genes with top15 degree of connectivity in LUAD. These genes will assist us to screen and identify significant biomarkers and therapeutic target of LUAD in the near future.
The gene expression profile of GSE10072 was downloaded from the GEO database, which was a free and publicly available database. 58 LUAD tumor tissues and 49 non-tumor tissues from 20 never smokers, 26 former smokers, and 28 current smokers in this dataset were detected by GPL96 [HG-U133A] Affymetrix Human Genome U133A Array by Landi MT. We also downloaded the Series Matrix File of GSE10072 from the GEO database.
In our study, the online software GEO2R was employed to analyze the tissue samples from GSE10072 dataset. GEO2R is an online software by which users can divide the samples into two and more groups and select out the DEGs. We used the Benjamini and Hochberg methods by default to discover false rate and used the adjust P value to reduce the errors of false positive. The choice criterion contains the adjust P value < 0.05 and |logFC| ≥ 1.
Gene ontology (GO) analysis is a common framework annotating genes and gene products including functions of cellular components, biological pathways and molecular function [14]. Kyoto Encyclopedia of Genes and Genomes (KEGG) contains a set of genomes and biological pathways related with disease and drugs online database, which essentially is a resource for systematic understanding of biological system and certain high-level genome functional information [15]. The Database for Annotation, Visualization and Integrated Discovery (DAVID, http://david.ncifcrf.gov) is an online bioinformatics database [16]. It has widely covered a great many biological data and relevant analysis tools, then provided tools for the biological function annotation information for plenty of genes or proteins. P < 0.05 was considered as the cut-off criterion with significant difference. We could visualize the key biological processes, molecular functions, cellular components and pathways of DEGs by using DAVID online database. And further the scatter plot was performed by ImageGP according to the results of GO and KEGG pathway.
GEPIA (http://gepia.cancer-pku.cn/index.html), designed by Chenwei Li, Zefang Tang, and Boxi Kang of Zhang Lab, Peking University, is a newly developed interactive web server aiming at analyzing the RNA sequencing expression data of 9736 tumors and 8587 normal samples from the GTEx and TCGA projects in a standard processing pipeline [17]. In our study, we employed the boxplot to visualize the mRNA expression of top5 upregulated and top5 downregulated DEGs in LUAD tissues and and normal colorectum tissues.
Similarly, we used the GEPIA database to get the overall survival (OS) and stage plot information of these DEGs. The logrank P value and hazard ratio (HR) with 95% confidence intervals were showed on the plot. P < 0 05 was statistically significant.
Search Tool for the Retrieval of Interacting Genes (STRING) is an online tool for assessment and integration of the protein-protein interaction (PPI) information, containing physical and functional associations. It covered 9,643,763 proteins from 2031 organisms in STRING version 10.0. We drew DEGs using STRING to evaluate the interactional associations among them, thereby utilized the Cytoscape sofrware to build a PPI network. We set maximum number of interactors = 0, confidence score ≥ 0.4 as the cut off criterion.
In our study, 58 tumor tissues from patients with LUAD and 49 non-tumor tissues from normal individuals were included and analyzed. We applied the GEO2R online analysis tool with default parameters to screen the DEGs, using adjusted P value < 0.05 and logFC ≤ −1 or logFC ≥ 1 as the cut-off criteria. We captured 856 DEGs were captured, including 559 up-regulated genes and 297 down-regulated genes. Whereafter, the DEGs were presented in the form of a volcano plot (Figure 1). Among the 856 DEGs, the top5 up-regulated genes involved AGER, SFTPC, FABP4, CYP4B1 and WIF1 while the top5 down-regulated genes were GREM1, SPINK1, MMP1, COL11A1 and SPP1. The gene tiles and biological functions of top5 upregulated and top5 down regulated genes were displayed in Table 1.
DEGs | Gene title | Gene symbol | LogFC | Biological function |
Up-regulated | advanced glycosylation end-product specific receptor | AGER | 4.4174695 | A member of the immunoglobulin superfamily of cell surface receptors |
surfactant protein C | SFTPC | 3.9898216 | hydrophobic surfactant protein essential for lung function and homeostasis | |
fatty acid binding protein 4 | FABP4 | 3.8385413 | fatty acid uptake, transport, and metabolism | |
cytochrome P450 family 4 subfamily B member 1 | CYP4B1 | 3.7097964 | Metabolizing certain carcinogens | |
WNT inhibitory factor 1 | WIF1 | 3.6867095 | inhibit WNT proteins | |
Down-regulated | gremlin 1, DAN family BMP antagonist | GREM1 | −2.5483627 | cell growth and differentiation factor |
serine peptidase inhibitor, Kazal type 1 | SPINK1 | −2.7583995 | trypsin inhibitor | |
matrix metallopeptidase 1 | MMP1 | −2.8620356 | embryonic development, reproduction, and tissue remodeling | |
collagen type XI alpha 1 chain | COL11A1 | −3.061522 | extracellular matrix | |
secreted phosphoprotein 1 | SPP1 | −4.3644151 | attachment of osteoclasts to the mineralized bone matrix |
To ensure the credibility of the microarray of GSE10072 and proceed further credible analysis, we validated the top5 up-regulated genes and top5 down-regulated genes based on TCGA database via GEPIA. The results based on GEPIA demonstrated that the mRNA expression levels of GREM1, SPINK1, MMP1, COL11A1 and SPP1 were significantly lower in carcinoma group compared to non-tumor group while the mRNA expression level of AGER, SFTPC, FABP4, CYP4B1 and WIF1 in carcinoma group were statistically higher than the non-tumor group (P < 0.05) (Figure 2A-J).
Furthermore, we analyzed the potential association between the expression levels of top5 up-regulated genes as well as top5 down-regulated genes and the OS of patients with LUDA (Figure 3A-J). The Kaplan-Meier showed that AGER, SFTPC, CYP4B1, COL11A1 and SPP1 displayed significantly correlation with the OS of patients with LUAD. In detail, the high level of AGER and CYP4B1 may contribute to poorer prognosis of LUAD while the high level of SFTPC, COL11A1 and SPP1 may contribute to better prognosis (P < 0.05).
Meanwhile, we analyzed the correlation between DEGs expression and tumor stage in LUAD patients. The results showed that the expression level of AGER, SFTPC, CYP4B1, WIF1, GREM1, MMP1 and COL11A1 displayed strong correlation with the tumor stage in patients with LUAD while the FABP4, SPINK1 and SPINK1 groups did not significantly differ (Figure 4A-J).
The results (Table 2 & Figure 5A-C) from GO term enrichment analysis varied from expression levels and GO classification of the DEGs. By analyzing GO enrichment of these up-regulated and down-regulated DEGs via DAVID, we found that the up-regulated DEGs in BP were mainly enriched in positive regulation of transcription from RNA polymerase Ⅱ promoter, signal transduction, negative regulation of transcription from RNA polymerase Ⅱ promoter, cell adhesion and positive regulation of GTPase activity while the up-regulated DEGs in BP mainly focused on cell division, apoptotic process, mitotic nuclear division, negative regulation of apoptotic process and cell adhesion. As for CC, the up-regulated DEGs were principally enriched in plasma membrane, integral component of membrane, extracellular exosome, extracellular region and extracellular space while the down-regulated DEGs were enriched in cytoplasm, nucleus, extracellular exosome, cytosol and extracellular space. MF analysis uncovered that the up-regulated DEGs were mainly enriched in protein binding, protein homodimerization activity, calcium ion binding, transcription factor activity, sequence-specific DNA binding and identical protein binding. By contrast, the down-regulated DEGs were enriched in extracellular matrix structural constituent, serine-type endopeptidase activity, identical protein binding, protein binding and platelet-derived growth factor binding.
Expression | Category | Term | Count | % | P-Value | FDR |
Up-regulated | GOTERM_BP_DIRECT | GO:0045944~positive regulation of transcription from RNA polymerase Ⅱ promoter | 51 | 0.080578904 | 1.60E-07 | 2.81E-04 |
GOTERM_BP_DIRECT | GO:0007165~signal transduction | 45 | 0.071099033 | 0.001027981 | 1.794107407 | |
GOTERM_BP_DIRECT | GO:0000122~negative regulation of transcription from RNA polymerase Ⅱ promoter | 41 | 0.064779119 | 3.51E-07 | 6.18E-04 | |
GOTERM_BP_DIRECT | GO:0007155~cell adhesion | 37 | 0.058459205 | 2.09E-10 | 3.69E-07 | |
GOTERM_BP_DIRECT | GO:0043547~positive regulation of GTPase activity | 23 | 0.036578904 | 1.60E-07 | 1.58217909 | |
GOTERM_CC_DIRECT | GO:0005886~plasma membrane | 147 | 0.232256841 | 2.66E-10 | 0.006648866 | |
GOTERM_CC_DIRECT | GO:0016021~integral component of membrane | 132 | 0.208557164 | 0.035243685 | 0.027678442 | |
GOTERM_CC_DIRECT | GO:0070062~extracellular exosome | 114 | 0.18011755 | 4.84E-11 | 0.106773084 | |
GOTERM_CC_DIRECT | GO:0005576~extracellular region | 75 | 0.118498388 | 9.67E-10 | 21.20178557 | |
GOTERM_CC_DIRECT | GO:0005615~extracellular space | 74 | 0.11691841 | 6.08E-13 | 69.41801659 | |
GOTERM_MF_DIRECT | GO:0005515~protein binding | 240 | 0.379194843 | 4.47E-06 | 0.006648866 | |
GOTERM_MF_DIRECT | GO:0042803~protein homodimerization activity | 36 | 0.056879226 | 1.86E-05 | 0.027678442 | |
GOTERM_MF_DIRECT | GO:0005509~calcium ion binding | 34 | 0.053719269 | 7.18E-05 | 0.106773084 | |
GOTERM_MF_DIRECT | GO:0003700~transcription factor activity, sequence-specific DNA binding | 33 | 0.052139291 | 0.015885233 | 21.20178557 | |
GOTERM_MF_DIRECT | GO:0042802~identical protein binding | 24 | 0.037919484 | 0.076530545 | 69.41801659 | |
Down-regulated | GOTERM_BP_DIRECT | GO:0051301~cell division | 20 | 0.06355057 | 3.48E-08 | 5.71E-05 |
GOTERM_BP_DIRECT | GO:0006915~apoptotic process | 17 | 0.054017985 | 0.00114447 | 1.862881966 | |
GOTERM_BP_DIRECT | GO:0007067~mitotic nuclear division | 16 | 0.050840456 | 2.59E-07 | 4.25E-04 | |
GOTERM_BP_DIRECT | GO:0043066~negative regulation of apoptotic process | 16 | 0.050840456 | 3.35E-04 | 0.548772672 | |
GOTERM_BP_DIRECT | GO:0007155~cell adhesion | 16 | 0.050840456 | 3.68E-04 | 0.603031926 | |
GOTERM_CC_DIRECT | GO:0005737~cytoplasm | 75 | 0.238314639 | 0.007215425 | 8.951333631 | |
GOTERM_CC_DIRECT | GO:0005634~nucleus | 74 | 0.23513711 | 0.025200114 | 28.14465235 | |
GOTERM_CC_DIRECT | GO:0070062~extracellular exosome | 71 | 0.225604525 | 7.48E-12 | 9.69E-09 | |
GOTERM_CC_DIRECT | GO:0005829~cytosol | 56 | 0.177941597 | 8.29E-04 | 1.068609771 | |
GOTERM_CC_DIRECT | GO:0005615~extracellular space | 48 | 0.152521369 | 8.69E-13 | 1.13E-09 | |
GOTERM_MF_DIRECT | GO:0005201~extracellular matrix structural constituent | 11 | 0.034952814 | 4.94E-09 | 6.87E-06 | |
GOTERM_MF_DIRECT | GO:0004252~serine-type endopeptidase activity | 14 | 0.044485399 | 9.14E-06 | 0.012713971 | |
GOTERM_MF_DIRECT | GO:0042802~identical protein binding | 23 | 0.073083156 | 6.07E-05 | 0.084464967 | |
GOTERM_MF_DIRECT | GO:0005515~protein binding | 129 | 0.409901179 | 1.08E-04 | 0.150356421 | |
GOTERM_MF_DIRECT | GO:0048407~platelet-derived growth factor binding | 4 | 0.012710114 | 2.37E-04 | 0.329852854 | |
GO: Gene Ontology; FDR: False Discovery Rate. |
To acquire a more comprehensive information regarding to the critical pathways of those selected DEGs, KEGG pathways analysis were also carried out via DAVID. The results in Table 3 and Figure 5D unveiled the most vital KEGG pathways of the down-regulated and up-regulated DEGs. The up-regulated DEGs were mainly enriched in Pathways in cancer, PI3K-Akt signaling pathway, Endocytosis, MAPK signaling pathway and Complement and coagulation cascades while the down-regulated DEGs were mainly responsible for Cell cycle, PI3K-Akt signaling pathway, ECM-receptor interaction, Focal adhesion and p53 signaling pathway.
Category | Term | Count | % | P-Value | Genes | FDR |
Up-regulated DEGs | hsa05200: Pathways in cancer | 21 | 0.033179549 | 0.012311429 | FGFR2, COL4A3, IL6, BMP2, EPAS1, PTGER4, TGFBR2, GNG11, ZBTB16, MECOM, CXCL12, COL4A5, EDNRA, AGTR1, FOS, EDNRB, LAMA4, ADCY9, PTCH1, AKT3, PIK3R1 | 14.56872647 |
hsa04151: PI3K-Akt signaling pathway | 16 | 0.025279656 | 0.087340443 | FGFR2, COL4A3, FGFR4, IL6, NR4A1, GNG11, IL7R, COL4A5, VWF, LAMA4, ITGA8, TEK, ANGPT1, AKT3, PIK3R1, GHR | 68.70335639 | |
hsa04144: Endocytosis | 14 | 0.022119699 | 0.02683695 | FGFR2, CAV2, FGFR4, CAV1, LDLR, TGFBR2, PIP5K1B, SNX1, HLA-E, ARRB1, FOLR1, NEDD4L, GRK5, RAB11FIP1 | 29.23295979 | |
hsa04010: MAPK signaling pathway | 14 | 0.022119699 | 0.037175664 | FGFR2, FGFR4, TGFBR2, NR4A1, MECOM, CACNA2D2, FOS, DUSP1, ARRB1, RPS6KA2, NTRK2, RRAS, GADD45B, AKT3 | 38.21659244 | |
hsa04610: Complement and coagulation cascades | 13 | 0.020539721 | 6.68E-07 | C7, A2M, C5AR1, F8, SERPING1, C4BPA, C1QB, VWF, CD55, THBD, CFD, CPB2, PROS1 | 8.49E-04 | |
Down-regulated DEGs | hsa04110: Cell cycle | 11 | 0.034952814 | 1.44E-05 | CCNB1, CDK1, CDKN2A, MAD2L1, CCNB2, BUB1, TTK, BUB1B, CDC20, SFN, MCM4 | 0.017175633 |
hsa04151: PI3K-Akt signaling pathway | 11 | 0.034952814 | 0.033131219 | COMP, TNC, COL3A1, COL1A2, EFNA4, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 33.06243008 | |
hsa04512: ECM-receptor interaction | 10 | 0.031775285 | 5.18E-06 | COMP, TNC, COL3A1, COL1A2, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 0.00617031 | |
hsa04510: Focal adhesion | 10 | 0.031775285 | 0.003577108 | COMP, TNC, COL3A1, COL1A2, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 4.179525016 | |
hsa04115: p53 signaling pathway | 9 | 0.028597757 | 6.04E-06 | CCNB1, CDK1, CDKN2A, CCNB2, RRM2, PMAIP1, SFN, PERP, IGFBP3 | 0.00719634 | |
KEGG: Kyoto Encyclopedia of Genes and Genomes; FDR: False Discovery Rate. |
Applying the STRING online tool, 452 nodes with 311 PPI relationships were found, accounting for about 82.8% of these selected DEGs. According to the degree of connectivity of these DEGs, we constructed the PPI network and selected the top 15 hub genes (Table 4). The top 15 hub genes, possessing high degree of connectivity in LUAD are as follows, IL6, MMP9, EDN1, FOS, CDK1, CDH1, BIRC5, VWF, UBE2C, CDKN3, CDKN2A, CD34, AURKA, CCNB2 and EGR1. Among these 15 hub genes: IL6, EDN1, FOS, CDK1, VWF, CD34 and EGR1 were significantly up-regulated while MMP9, CDH1, BIRC5, UBE2C, CDKN3, CDKN2A, AURKA and CCNB2 significantly down-regulated (P < 0.05). The 15 hub genes could interact with 381 genes directly, and IL6 acted as the most intensive gene which could interact with 82 up-regulated genes and 44 down-regulated genes. Additionally, among these hub genes, there also displayed very strong interactions (Figure 6).
Gene | Degree of connectivity | Adjusted P value |
IL6 | 84 | 1.09E-05 |
MMP9 | 55 | 2.11E-13 |
EDN1 | 53 | 6.19E-12 |
FOS | 47 | 9.42E-06 |
CDK1 | 44 | 1.99E-16 |
CDH1 | 44 | 4.41E-10 |
BIRC5 | 43 | 1.44E-05 |
VWF | 42 | 7.41E-36 |
UBE2C | 41 | 1.83E-14 |
CDKN3 | 36 | 1.10E-15 |
CDKN2A | 36 | 1.03E-09 |
CD34 | 36 | 3.04E-27 |
AURKA | 33 | 2.33E-14 |
CCNB2 | 33 | 3.01E-17 |
EGR1 | 33 | 2.58E-07 |
Although cigarette smoking is one of dominating causes of lung cancer, surprisingly, among various major histological types of lung cancer, LUAD displayed the weakliest association with smoking, which often occurs in females and people without smoking history [18,19,20]. The somatic gene aberrations in LUAD have been most extensively explored. LUAD screening has been demonstrated to greatly decrease the morbidity and the mortality in a great many longstanding or newly economically developed countries [21,22]. However, at present, there is no an efficient and specific diagnostic methodology and treatment strategy for LUAD, which is mainly attributed to the intricate pathogenesis, and its symptoms that are difficult to diagnose in the first several years [23,24]. In the other one hand, the oncogenic pathway of LUAD is incompletely understood. In this study, 58 tumor tissues from patients with LUAD and 49 non-tumor tissues from normal individuals were analyzed. 856 DEGs including 559 up-regulated genes and 297 down-regulated genes were screened. To obtain a comprehensive understanding of these DEGs, we performed GO function and KEGG pathway analysis. Additionally, we analyzed the relationship between the 10 mosth significant DEGs and the overall survival as well as tumor stage.
Our analysis selected out 856 DEGs with 2-fold change between carcinoma tissues and normal tissues. According to the rank of the fold change of these DGEs, we picked up the top5 up-regulated DEGs and top5 down-regulated DEGs. From our perspective, these DEGs would be possible candidates for the diagnosis of LUAD in near future. At present, some of these DEGs, in fact, have been already disclosed to be novel indicators of LUAD. For instance, Tang Z et al. [25] found that the elevated expression of fatty acid binding proteins 4 (FABP4) in non-small cell lung cancer was not only significantly correlated with advanced tumor node metastasis (TNM) stage, but also exhibited a negative effect on the overall survival. WIF1, a vital component in the Wnt signaling pathway, was found to be down-regulated in multiple cancers, including breast, prostate, bladder, and lung cancer [26]. WIF-1 promoter region hypermethylation contributes to aberrant activation of Wnt signaling pathway in NSCLC. Meanwhile, WIF-1 promoter region hypermethylation is also a novel diagnostic marker for LUAD-related malignant pleural effusions [27]. An interesting finding in our study was that WIF-1 is highly expressed in LUAD tissue while its level in patients with LUAD is not associated with overall survival, which hints that WIF-1 is a switch of LUAD but could not promote the progression of LUAD. However, the role of other DEGs has not been explored in LUAD. The receptor for advanced glycation end products (AGER) as an oncogenic transmembranous receptor, was up-regulated in many human cancers. Elevated AGER may promote cervical cancer cell migration, proliferation, and inhibited its apoptosis [28]. Hence, the role of AGER in LUAD needs to be further investigated. Our study screened the DEGs of LUAD from the angle of bioinformatics for the first time, which not only verified the reported genes, but also prompted new biomarkers in LUAD.
At the same time, we picked out 15 hub genes imlpicated with LUAD, all of which were located in the core nodes in PPI network, meaning that these genes could be critical therapeutic targets to protect against LUAD. It is reported that IL-6 may serve as a mediator of many reactions involving an inflammatory response in patients with lung cancer [29,30,31]. Autocrine IL-6-induced Stat3 activation could result in the occurrence of LUAD and the production of malignant pleural effusion [32,33,34]. In our study, we disclosed that IL6 is the hub genes with the highest connective degree, suggesting that IL6 plays a core role in the occurrence and development of LUAD. CDH1, encoded by the CDH1 gene in humans, was also a hub gene with high connective degree in our study. Study showed that CDH1 methylation is closely correlated with an elevated risk of lung cancer, the hypermethylation of which could inactive CDH1, thereby influencing proliferation, invasion, and metastasis of lung cancer cells [35]. Hence, these hub genes are potential therapeutic targets in LUAD.
Abnormal uncontrolled cell growth and cell cycle-mediated cell transformation are the basical biological features of LUAD. Our study unveiled that the DEGs are mainly enriched in the events and pathways associated with cell proliferation and apoptosis. Furthermore, the GO analysis in CC and KEGG pathway analysis also proved that extracellular matrix (ECM) exerts an essential role in LUAD. Indeed, ECM is the direct tumor microenvironment, consisting of proteoglycans, non-proteinaceous glycosaminoglycans, and collagens. It is recognized that ECM molecules could activate autocrine or paracrine cell signaling directly, form a biomechanical scaffold for adherent cells, eventually remodelling tissue architecture during inflammation. Laura E. Stevens et al. [36] found that up-regulation of the hyaluronan receptor HMMR in mice with LUAD was correlated with a significant inflammatory molecular signature as well as poor prognosis. Finally, our study proved the critical roles of PI3K/Akt signaling pathway and MAPK pathway from the angle of bioinformatics, which further solid the previous experimental researches.
In conclusion, we provided a comprehensive and novel analysis of gene expression profiles patients with LUAD. Particularly, the top5 up-regulated genes including AGER, SFTPC, FABP4, CYP4B1 and WIF1 and the top5 down-regulated genes including GREM1, SPINK1, MMP1, COL11A1 and SPP1, which are expected to sensitive biomarkers in diagnosis of LUAD. Meanwhile, we also screened the top 15 hub genes involving IL6, MMP9, EDN1, FOS, CDK1, CDH1, BIRC5, VWF, UBE2C, CDKN3, CDKN2A, CD34, AURKA, CCNB2 and EGR1, which could be promising therapeutic targets of LUAD. Additionally, genes and pathways involved in ECM were also significantly altered in patients with LUAD. Anyway, this analysis may offer the powerful evidence and clues for the future genomic individualized treatment of LUAD.
The authors declare that they have no conflict of interest.
[1] | T. Akhurst, Staging of non-small-cell lung cancer, PET. Clin., 13 (2018), 1–10. |
[2] | Y. Lavin, S. Kobayashi, A. Leader, et al., Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses, Cell, 169 (2017), 750–765.e717. |
[3] | L. Han, C. K. Lee, H. Pang, et al., Genetic predisposition to lung adenocarcinoma among never-smoking Chinese with different epidermal growth factor receptor mutation status, Lung Cancer, 114 (2017), 79–89. |
[4] | E. Tsiambas, A. Y. Lefas, S. N. Georgiannos, et al., EGFR gene deregulation mechanisms in lung adenocarcinoma: A molecular review, Pathol. Res. Pract., 212 (2016), 672–677. |
[5] | J. F. Fahrmann, D. Grapov, B. S. Phinney, et al., Proteomic profiling of lung adenocarcinoma indicates heightened DNA repair, antioxidant mechanisms and identifies LASP1 as a potential negative predictor of survival, Clin. Proteom., 13 (2016), 31. |
[6] | T. Yamaoka, T. Ohmori, M. Ohba, et al., Acquired resistance mechanisms to combination Met-TKI/EGFR-TKI exposure in met-amplified EGFR-TKI-resistant lung adenocarcinoma harboring an activating EGFR mutation, Mol. Cancer. Ther., 15 (2016), 3040–3054. |
[7] | E. B. Choi, A. Y. Yang, S. C. Kim, et al., PARP1 enhances lung adenocarcinoma metastasis by novel mechanisms independent of DNA repair, Oncogene, 35 (2016), 4569–4579. |
[8] | D. Yang, D. Cheng, Q. Tu, et al., HUWE1 controls the development of non-small cell lung cancer through down-regulation of p53, Theranostics, 8 (2018), 3517–3529. |
[9] | I. Mender, R. LaRanger, K. Luitel, et al., Telomerase-Mediated Strategy for Overcoming Non-Small Cell Lung Cancer Targeted Therapy and Chemotherapy Resistance, Neoplasia, 20 (2018), 826–837. |
[10] | Comprehensive molecular profiling of lung adenocarcinoma, Nature, 511 (2014), 543–550. |
[11] | N. Li, L. Li and Y. Chen, The identification of core gene expression signature in hepatocellular carcinoma, Oxid. Med. Cell. Longev., 2018 (2018), 3478305. |
[12] | T. Braunschweig, J. Y. Chung and S. M. Hewitt, Tissue microarrays: bridging the gap between research and the clinic, Expert Rev. Proteom., 2 (2005), 325–336. |
[13] | L. Li, N. Li, C. He, et al., Proteomic analysis of differentially expressed proteins in kidneys of brain dead rabbits, Mol. Med. Rep., 16 (2017), 215–223. |
[14] | R. P. Huntley, T. Sawford, P. Mutowo-Meullenet, et al., The GOA database: Gene ontology annotation updates for 2015, Nucleic Acids Res., 43 (2015), D1057–1063. |
[15] | F. Fabris and A. A. Freitas, New KEGG pathway-based interpretable features for classifying ageing-related mouse proteins, Bioinformatics, 32 (2016), 2988–2995. |
[16] | G. Dennis, Jr., B. T. Sherman, D. A. Hosack, et al., DAVID: Database for annotation, visualization, and integrated discovery, Genome Biol., 4 (2003), P3. |
[17] | Z. Tang, C. Li, B. Kang, et al., GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses, Nucleic Acids Res., 45 (2017), W98–W102. |
[18] | Z. Yin, Z. Cui, Y. Ren, et al., Association between polymorphisms in pre-miRNA genes and risk of lung cancer in a Chinese non-smoking female population, Lung Cancer, 94 (2016), 15–21. |
[19] | T. Vu, L. Jin and P. K. Datta, Effect of cigarette smoking on epithelial to mesenchymal transition (EMT) in lung cancer, J. Clin. Med., 5 (2016). |
[20] | Y. Zhang, M. Elgizouli, B. Schottker, et al., Smoking-associated DNA methylation markers predict lung cancer incidence, Clin. Epigenet., 8 (2016), 127. |
[21] | F. Ciompi, K. Chung, S. J. van Riel, et al., Towards automatic pulmonary nodule management in lung cancer screening with deep learning, Sci. Rep., 7 (2017), 46479. |
[22] | U. Yousaf-Khan, C. van der Aalst, P. A. de Jong, et al., Risk stratification based on screening history: the NELSON lung cancer screening study, Thorax, 72 (2017), 819–824. |
[23] | Y. Zhang, J. Sui, X. Shen, et al., Differential expression profiles of microRNAs as potential biomarkers for the early diagnosis of lung cancer, Oncol. Rep., 37 (2017), 3543–3553. |
[24] | M. Reck and K. F. Rabe, Precision diagnosis and treatment for advanced non-small-cell lung cancer, N. Engl. J. Med., 377 (2017), 849–861. |
[25] | Z. Tang, Q. Shen, H. Xie, et al., Elevated expression of FABP3 and FABP4 cooperatively correlates with poor prognosis in non-small cell lung cancer (NSCLC), Oncotarget, 7 (2016), 46253–46262. |
[26] | T. M. Yang, S. W. Leu, J. M. Li, et al., WIF-1 promoter region hypermethylation as an adjuvant diagnostic marker for non-small cell lung cancer-related malignant pleural effusions, J. Cancer Res. Clin. Oncol., 135 (2009), 919–924. |
[27] | C. Wissmann, P. J. Wild, S. Kaiser, et al., WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer, J. Pathol., 201 (2003), 204–212. |
[28] | X. Zhu, L. Zhou, R. Li, et al., AGER promotes proliferation and migration in cervical cancer, Biosci. Rep., 38 (2018). |
[29] | H. Yanagawa, S. Sone, Y. Takahashi, et al., Serum levels of interleukin 6 in patients with lung cancer, Br. J. Cancer, 71 (1995), 1095–1098. |
[30] | Y. P. Chen, Y. Zhang, J. W. Lv, et al., Genomic analysis of tumor microenvironment immune types across 14 solid cancer types: Immunotherapeutic implications, Theranostics, 7 (2017), 3585–3594. |
[31] | C. Xie, J. Zhu, Y. Jiang, et al., Sulforaphane inhibits the acquisition of tobacco smoke-induced lung cancer stem cell-like properties via the IL-6/DeltaNp63alpha/Notch axis, Theranostics, 9 (2019), 4827–4840. |
[32] | H. H. Yeh, W. T. Chang, K. C. Lu, et al., Upregulation of tissue factor by activated Stat3 contributes to malignant pleural effusion generation via enhancing tumor metastasis and vascular permeability in lung adenocarcinoma, PLoS One, 8 (2013), e75287. |
[33] | L. Li, J. Xu, G. Qiu, et al., Epigenomic characterization of a p53-regulated 3p22.2 tumor suppressor that inhibits STAT3 phosphorylation via protein docking and is frequently methylated in esophageal and other carcinomas, Theranostics, 8 (2018), 61–77. |
[34] | L. Jiang, R. Wang, L. Fang, et al., HCP5 is a SMAD3-responsive long non-coding RNA that promotes lung adenocarcinoma metastasis via miR-203/SNAI axis, Theranostics, 9 (2019), 2460–2474. |
[35] | Q. Yu, Q. Guo, L. Chen, et al., Clinicopathological significance and potential drug targeting of CDH1 in lung cancer: A meta-analysis and literature review, Drug Des. Devel. Ther., 9 (2015), 2171–2178. |
[36] | L. E. Stevens, W. K. C. Cheung, S. J. Adua, et al., Extracellular matrix receptor expression in subtypes of lung adenocarcinoma potentiates outgrowth of micrometastases, Cancer Res., 77 (2017), 1905–1917. |
1. | Yan Wang, Liwei Qiu, Yu Chen, Xia Zhang, Peng Yang, Feng Xu, Screening and Identification of Four Prognostic Genes Related to Immune Infiltration and G-Protein Coupled Receptors Pathway in Lung Adenocarcinoma, 2021, 10, 2234-943X, 10.3389/fonc.2020.622251 | |
2. | Zhou Jiawei, Mu Min, Xing Yingru, Zhang Xin, Li Danting, Liu Yafeng, Xie Jun, Hu Wangfa, Zhang Lijun, Wu Jing, Hu Dong, Identification of Key Genes in Lung Adenocarcinoma and Establishment of Prognostic Mode, 2020, 7, 2296-889X, 10.3389/fmolb.2020.561456 | |
3. | Jingwei Zhang, Lin Jiao, Hao Bai, Qian Wu, Tao Wu, Tangyuheng Liu, Xuejiao Hu, Jiajia Song, Binwu Ying, A Notch4 missense mutation is associated with susceptibility to tuberculosis in Chinese population, 2020, 78, 15671348, 104145, 10.1016/j.meegid.2019.104145 | |
4. | Shanshan Zhao, Mingxin Yu, Identification ofMMP1as a Potential Prognostic Biomarker and Correlating with Immune Infiltrates in Cervical Squamous Cell Carcinoma, 2020, 39, 1044-5498, 255, 10.1089/dna.2019.5129 | |
5. |
Xiyong Wang, Huaiqing Xiao, Dongqiang Wu, Dongliang Zhang, Zhihao Zhang,
miR-335-5p Regulates Cell Cycle and Metastasis in Lung Adenocarcinoma by Targeting CCNB2 , 2020, Volume 13, 1178-6930, 6255, 10.2147/OTT.S245136 |
|
6. | Vilde D. Haakensen, Anand Khadse, Vandana Sandhu, Ann Rita Halvorsen, Steinar K. Solberg, Lars H. Jørgensen, Odd Terje Brustugun, Elin H. Kure, Åslaug Helland, Molecular characterisation of TP53 mutated squamous cell carcinomas of the lung to identify putative targets for therapy , 2020, 147, 0020-7136, 2957, 10.1002/ijc.33121 | |
7. | Fushi Han, Shusong Yang, Wei Wang, Xinghong Huang, Dongdong Huang, Shuzhen Chen, Silencing of lncRNA LINC00857 Enhances BIRC5-Dependent Radio-Sensitivity of Lung Adenocarcinoma Cells by Recruiting NF-κB1, 2020, 22, 21622531, 981, 10.1016/j.omtn.2020.09.020 | |
8. | Xiulin Jiang, Yulin Shi, Xi Chen, Haitao Xu, Xiaobin Huang, Lihua Li, Jun Pu, The N6-Methylandenosine-Related Gene BIRC5 as a Prognostic Biomarker Correlated With Cell Migration and Immune Cell Infiltrates in Low Grade Glioma, 2022, 9, 2296-889X, 10.3389/fmolb.2022.773662 | |
9. | Annika Röder, Saskia Hüsken, Michael C. Hutter, Allan E. Rettie, Helmut Hanenberg, Constanze Wiek, Marco Girhard, Spotlight on CYP4B1, 2023, 24, 1422-0067, 2038, 10.3390/ijms24032038 | |
10. | Junliang Shang, Jing Wang, Yan Sun, Feng Li, Jin-Xing Liu, Honghai Zhang, Jonathan Wren, Multiscale part mutual information for quantifying nonlinear direct associations in networks, 2021, 37, 1367-4803, 2920, 10.1093/bioinformatics/btab182 | |
11. | Kai Wang, Man Zhang, Jiao Wang, Pan Sun, Jizhuang Luo, Haizhen Jin, Rong Li, Changqing Pan, Liming Lu, A Systematic Analysis Identifies Key Regulators Involved in Cell Proliferation and Potential Drugs for the Treatment of Human Lung Adenocarcinoma, 2021, 11, 2234-943X, 10.3389/fonc.2021.737152 | |
12. | Yi Zheng, Hui You, Jingzhu Duan, Biyu Chen, Chenlin Wu, Peipei Chen, Meifang Wang, Centromere protein N promotes lung adenocarcinoma progression by activating PI3K/AKT signaling pathway, 2022, 44, 1976-9571, 1039, 10.1007/s13258-021-01215-8 | |
13. | Yue Wang, Xulong Huang, Bin Xian, Huajuan Jiang, Tao Zhou, Siyu Chen, Feiyan Wen, Jin Pei, Machine learning and bioinformatics-based insights into the potential targets of saponins in Paris polyphylla smith against non-small cell lung cancer, 2022, 13, 1664-8021, 10.3389/fgene.2022.1005896 | |
14. | Chongze Yuan, Haojie Chen, Shiqi Tu, Hsin-Yi Huang, Yunjian Pan, Xiuqi Gui, Muyu Kuang, Xuxia Shen, Qiang Zheng, Yang Zhang, Chao Cheng, Hui Hong, Xiaoting Tao, Yizhou Peng, Xingxin Yao, Feilong Meng, Hongbin Ji, Zhen Shao, Yihua Sun, A systematic dissection of the epigenomic heterogeneity of lung adenocarcinoma reveals two different subclasses with distinct prognosis and core regulatory networks, 2021, 22, 1474-760X, 10.1186/s13059-021-02376-1 | |
15. | Valerio Napolioni, Fortunato Bianconi, Rossella Potenza, Francesco M. Carpi, Vienna Ludovini, Matteo Picciolini, Francesca R. Tofanetti, Antonello Bufalari, Stefano Pallotti, Camilla Poggi, Marco Anile, Niccolò Daddi, Federico Venuta, Francesco Puma, Jacopo Vannucci, Genome-wide expression of the residual lung reacting to experimental Pneumonectomy, 2021, 22, 1471-2164, 10.1186/s12864-021-08171-3 | |
16. | Dengfeng Wang, Hongjiao Sun, Xiaohui Li, Gang Wang, Guizhong Yan, Haijun Ren, Boru Hou, CCNB2 is a novel prognostic factor and a potential therapeutic target in low-grade glioma, 2022, 42, 0144-8463, 10.1042/BSR20211939 | |
17. | Chuanlong Zhang, Qian Shen, Mengqi Gao, Junchen Li, Bo Pang, The role of Cyclin Dependent Kinase Inhibitor 3 (CDKN3) in promoting human tumors: Literature review and pan-cancer analysis, 2024, 10, 24058440, e26061, 10.1016/j.heliyon.2024.e26061 | |
18. | Abanti Bhattacharjya, Md Manowarul Islam, Md Ashraf Uddin, Md Alamin Talukder, AKM Azad, Sunil Aryal, Bikash Kumar Paul, Wahia Tasnim, Muhammad Ali Abdulllah Almoyad, Mohammad Ali Moni, Exploring gene regulatory interaction networks and predicting therapeutic molecules for hypopharyngeal cancer and EGFR‐mutated lung adenocarcinoma, 2024, 14, 2211-5463, 1166, 10.1002/2211-5463.13807 | |
19. | Qiongyin Zhang, Ning An, Yang Liu, Ying Zhu, Wuliang Pan, Peiling Gu, Jinzhu Zhao, Qiang Pu, Wen Zhu, Alveolar type 2 cells marker gene SFTPC inhibits epithelial-to-mesenchymal transition by upregulating SOX7 and suppressing WNT/β-catenin pathway in non-small cell lung cancer, 2024, 14, 2234-943X, 10.3389/fonc.2024.1448379 | |
20. | Md. Al Mehedi Hasan, Md. Maniruzzaman, Jungpil Shin, Gene Expression and Metadata Based Identification of Key Genes for Hepatocellular Carcinoma Using Machine Learning and Statistical Models, 2023, 20, 1545-5963, 3786, 10.1109/TCBB.2023.3322753 | |
21. | Ming Wang, Bone morphogenetic protein 1: a prognostic indicator and potential biomarker in three cancer types, 2024, 16, 19438141, 400, 10.62347/PWZA6242 |
DEGs | Gene title | Gene symbol | LogFC | Biological function |
Up-regulated | advanced glycosylation end-product specific receptor | AGER | 4.4174695 | A member of the immunoglobulin superfamily of cell surface receptors |
surfactant protein C | SFTPC | 3.9898216 | hydrophobic surfactant protein essential for lung function and homeostasis | |
fatty acid binding protein 4 | FABP4 | 3.8385413 | fatty acid uptake, transport, and metabolism | |
cytochrome P450 family 4 subfamily B member 1 | CYP4B1 | 3.7097964 | Metabolizing certain carcinogens | |
WNT inhibitory factor 1 | WIF1 | 3.6867095 | inhibit WNT proteins | |
Down-regulated | gremlin 1, DAN family BMP antagonist | GREM1 | −2.5483627 | cell growth and differentiation factor |
serine peptidase inhibitor, Kazal type 1 | SPINK1 | −2.7583995 | trypsin inhibitor | |
matrix metallopeptidase 1 | MMP1 | −2.8620356 | embryonic development, reproduction, and tissue remodeling | |
collagen type XI alpha 1 chain | COL11A1 | −3.061522 | extracellular matrix | |
secreted phosphoprotein 1 | SPP1 | −4.3644151 | attachment of osteoclasts to the mineralized bone matrix |
Expression | Category | Term | Count | % | P-Value | FDR |
Up-regulated | GOTERM_BP_DIRECT | GO:0045944~positive regulation of transcription from RNA polymerase Ⅱ promoter | 51 | 0.080578904 | 1.60E-07 | 2.81E-04 |
GOTERM_BP_DIRECT | GO:0007165~signal transduction | 45 | 0.071099033 | 0.001027981 | 1.794107407 | |
GOTERM_BP_DIRECT | GO:0000122~negative regulation of transcription from RNA polymerase Ⅱ promoter | 41 | 0.064779119 | 3.51E-07 | 6.18E-04 | |
GOTERM_BP_DIRECT | GO:0007155~cell adhesion | 37 | 0.058459205 | 2.09E-10 | 3.69E-07 | |
GOTERM_BP_DIRECT | GO:0043547~positive regulation of GTPase activity | 23 | 0.036578904 | 1.60E-07 | 1.58217909 | |
GOTERM_CC_DIRECT | GO:0005886~plasma membrane | 147 | 0.232256841 | 2.66E-10 | 0.006648866 | |
GOTERM_CC_DIRECT | GO:0016021~integral component of membrane | 132 | 0.208557164 | 0.035243685 | 0.027678442 | |
GOTERM_CC_DIRECT | GO:0070062~extracellular exosome | 114 | 0.18011755 | 4.84E-11 | 0.106773084 | |
GOTERM_CC_DIRECT | GO:0005576~extracellular region | 75 | 0.118498388 | 9.67E-10 | 21.20178557 | |
GOTERM_CC_DIRECT | GO:0005615~extracellular space | 74 | 0.11691841 | 6.08E-13 | 69.41801659 | |
GOTERM_MF_DIRECT | GO:0005515~protein binding | 240 | 0.379194843 | 4.47E-06 | 0.006648866 | |
GOTERM_MF_DIRECT | GO:0042803~protein homodimerization activity | 36 | 0.056879226 | 1.86E-05 | 0.027678442 | |
GOTERM_MF_DIRECT | GO:0005509~calcium ion binding | 34 | 0.053719269 | 7.18E-05 | 0.106773084 | |
GOTERM_MF_DIRECT | GO:0003700~transcription factor activity, sequence-specific DNA binding | 33 | 0.052139291 | 0.015885233 | 21.20178557 | |
GOTERM_MF_DIRECT | GO:0042802~identical protein binding | 24 | 0.037919484 | 0.076530545 | 69.41801659 | |
Down-regulated | GOTERM_BP_DIRECT | GO:0051301~cell division | 20 | 0.06355057 | 3.48E-08 | 5.71E-05 |
GOTERM_BP_DIRECT | GO:0006915~apoptotic process | 17 | 0.054017985 | 0.00114447 | 1.862881966 | |
GOTERM_BP_DIRECT | GO:0007067~mitotic nuclear division | 16 | 0.050840456 | 2.59E-07 | 4.25E-04 | |
GOTERM_BP_DIRECT | GO:0043066~negative regulation of apoptotic process | 16 | 0.050840456 | 3.35E-04 | 0.548772672 | |
GOTERM_BP_DIRECT | GO:0007155~cell adhesion | 16 | 0.050840456 | 3.68E-04 | 0.603031926 | |
GOTERM_CC_DIRECT | GO:0005737~cytoplasm | 75 | 0.238314639 | 0.007215425 | 8.951333631 | |
GOTERM_CC_DIRECT | GO:0005634~nucleus | 74 | 0.23513711 | 0.025200114 | 28.14465235 | |
GOTERM_CC_DIRECT | GO:0070062~extracellular exosome | 71 | 0.225604525 | 7.48E-12 | 9.69E-09 | |
GOTERM_CC_DIRECT | GO:0005829~cytosol | 56 | 0.177941597 | 8.29E-04 | 1.068609771 | |
GOTERM_CC_DIRECT | GO:0005615~extracellular space | 48 | 0.152521369 | 8.69E-13 | 1.13E-09 | |
GOTERM_MF_DIRECT | GO:0005201~extracellular matrix structural constituent | 11 | 0.034952814 | 4.94E-09 | 6.87E-06 | |
GOTERM_MF_DIRECT | GO:0004252~serine-type endopeptidase activity | 14 | 0.044485399 | 9.14E-06 | 0.012713971 | |
GOTERM_MF_DIRECT | GO:0042802~identical protein binding | 23 | 0.073083156 | 6.07E-05 | 0.084464967 | |
GOTERM_MF_DIRECT | GO:0005515~protein binding | 129 | 0.409901179 | 1.08E-04 | 0.150356421 | |
GOTERM_MF_DIRECT | GO:0048407~platelet-derived growth factor binding | 4 | 0.012710114 | 2.37E-04 | 0.329852854 | |
GO: Gene Ontology; FDR: False Discovery Rate. |
Category | Term | Count | % | P-Value | Genes | FDR |
Up-regulated DEGs | hsa05200: Pathways in cancer | 21 | 0.033179549 | 0.012311429 | FGFR2, COL4A3, IL6, BMP2, EPAS1, PTGER4, TGFBR2, GNG11, ZBTB16, MECOM, CXCL12, COL4A5, EDNRA, AGTR1, FOS, EDNRB, LAMA4, ADCY9, PTCH1, AKT3, PIK3R1 | 14.56872647 |
hsa04151: PI3K-Akt signaling pathway | 16 | 0.025279656 | 0.087340443 | FGFR2, COL4A3, FGFR4, IL6, NR4A1, GNG11, IL7R, COL4A5, VWF, LAMA4, ITGA8, TEK, ANGPT1, AKT3, PIK3R1, GHR | 68.70335639 | |
hsa04144: Endocytosis | 14 | 0.022119699 | 0.02683695 | FGFR2, CAV2, FGFR4, CAV1, LDLR, TGFBR2, PIP5K1B, SNX1, HLA-E, ARRB1, FOLR1, NEDD4L, GRK5, RAB11FIP1 | 29.23295979 | |
hsa04010: MAPK signaling pathway | 14 | 0.022119699 | 0.037175664 | FGFR2, FGFR4, TGFBR2, NR4A1, MECOM, CACNA2D2, FOS, DUSP1, ARRB1, RPS6KA2, NTRK2, RRAS, GADD45B, AKT3 | 38.21659244 | |
hsa04610: Complement and coagulation cascades | 13 | 0.020539721 | 6.68E-07 | C7, A2M, C5AR1, F8, SERPING1, C4BPA, C1QB, VWF, CD55, THBD, CFD, CPB2, PROS1 | 8.49E-04 | |
Down-regulated DEGs | hsa04110: Cell cycle | 11 | 0.034952814 | 1.44E-05 | CCNB1, CDK1, CDKN2A, MAD2L1, CCNB2, BUB1, TTK, BUB1B, CDC20, SFN, MCM4 | 0.017175633 |
hsa04151: PI3K-Akt signaling pathway | 11 | 0.034952814 | 0.033131219 | COMP, TNC, COL3A1, COL1A2, EFNA4, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 33.06243008 | |
hsa04512: ECM-receptor interaction | 10 | 0.031775285 | 5.18E-06 | COMP, TNC, COL3A1, COL1A2, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 0.00617031 | |
hsa04510: Focal adhesion | 10 | 0.031775285 | 0.003577108 | COMP, TNC, COL3A1, COL1A2, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 4.179525016 | |
hsa04115: p53 signaling pathway | 9 | 0.028597757 | 6.04E-06 | CCNB1, CDK1, CDKN2A, CCNB2, RRM2, PMAIP1, SFN, PERP, IGFBP3 | 0.00719634 | |
KEGG: Kyoto Encyclopedia of Genes and Genomes; FDR: False Discovery Rate. |
Gene | Degree of connectivity | Adjusted P value |
IL6 | 84 | 1.09E-05 |
MMP9 | 55 | 2.11E-13 |
EDN1 | 53 | 6.19E-12 |
FOS | 47 | 9.42E-06 |
CDK1 | 44 | 1.99E-16 |
CDH1 | 44 | 4.41E-10 |
BIRC5 | 43 | 1.44E-05 |
VWF | 42 | 7.41E-36 |
UBE2C | 41 | 1.83E-14 |
CDKN3 | 36 | 1.10E-15 |
CDKN2A | 36 | 1.03E-09 |
CD34 | 36 | 3.04E-27 |
AURKA | 33 | 2.33E-14 |
CCNB2 | 33 | 3.01E-17 |
EGR1 | 33 | 2.58E-07 |
DEGs | Gene title | Gene symbol | LogFC | Biological function |
Up-regulated | advanced glycosylation end-product specific receptor | AGER | 4.4174695 | A member of the immunoglobulin superfamily of cell surface receptors |
surfactant protein C | SFTPC | 3.9898216 | hydrophobic surfactant protein essential for lung function and homeostasis | |
fatty acid binding protein 4 | FABP4 | 3.8385413 | fatty acid uptake, transport, and metabolism | |
cytochrome P450 family 4 subfamily B member 1 | CYP4B1 | 3.7097964 | Metabolizing certain carcinogens | |
WNT inhibitory factor 1 | WIF1 | 3.6867095 | inhibit WNT proteins | |
Down-regulated | gremlin 1, DAN family BMP antagonist | GREM1 | −2.5483627 | cell growth and differentiation factor |
serine peptidase inhibitor, Kazal type 1 | SPINK1 | −2.7583995 | trypsin inhibitor | |
matrix metallopeptidase 1 | MMP1 | −2.8620356 | embryonic development, reproduction, and tissue remodeling | |
collagen type XI alpha 1 chain | COL11A1 | −3.061522 | extracellular matrix | |
secreted phosphoprotein 1 | SPP1 | −4.3644151 | attachment of osteoclasts to the mineralized bone matrix |
Expression | Category | Term | Count | % | P-Value | FDR |
Up-regulated | GOTERM_BP_DIRECT | GO:0045944~positive regulation of transcription from RNA polymerase Ⅱ promoter | 51 | 0.080578904 | 1.60E-07 | 2.81E-04 |
GOTERM_BP_DIRECT | GO:0007165~signal transduction | 45 | 0.071099033 | 0.001027981 | 1.794107407 | |
GOTERM_BP_DIRECT | GO:0000122~negative regulation of transcription from RNA polymerase Ⅱ promoter | 41 | 0.064779119 | 3.51E-07 | 6.18E-04 | |
GOTERM_BP_DIRECT | GO:0007155~cell adhesion | 37 | 0.058459205 | 2.09E-10 | 3.69E-07 | |
GOTERM_BP_DIRECT | GO:0043547~positive regulation of GTPase activity | 23 | 0.036578904 | 1.60E-07 | 1.58217909 | |
GOTERM_CC_DIRECT | GO:0005886~plasma membrane | 147 | 0.232256841 | 2.66E-10 | 0.006648866 | |
GOTERM_CC_DIRECT | GO:0016021~integral component of membrane | 132 | 0.208557164 | 0.035243685 | 0.027678442 | |
GOTERM_CC_DIRECT | GO:0070062~extracellular exosome | 114 | 0.18011755 | 4.84E-11 | 0.106773084 | |
GOTERM_CC_DIRECT | GO:0005576~extracellular region | 75 | 0.118498388 | 9.67E-10 | 21.20178557 | |
GOTERM_CC_DIRECT | GO:0005615~extracellular space | 74 | 0.11691841 | 6.08E-13 | 69.41801659 | |
GOTERM_MF_DIRECT | GO:0005515~protein binding | 240 | 0.379194843 | 4.47E-06 | 0.006648866 | |
GOTERM_MF_DIRECT | GO:0042803~protein homodimerization activity | 36 | 0.056879226 | 1.86E-05 | 0.027678442 | |
GOTERM_MF_DIRECT | GO:0005509~calcium ion binding | 34 | 0.053719269 | 7.18E-05 | 0.106773084 | |
GOTERM_MF_DIRECT | GO:0003700~transcription factor activity, sequence-specific DNA binding | 33 | 0.052139291 | 0.015885233 | 21.20178557 | |
GOTERM_MF_DIRECT | GO:0042802~identical protein binding | 24 | 0.037919484 | 0.076530545 | 69.41801659 | |
Down-regulated | GOTERM_BP_DIRECT | GO:0051301~cell division | 20 | 0.06355057 | 3.48E-08 | 5.71E-05 |
GOTERM_BP_DIRECT | GO:0006915~apoptotic process | 17 | 0.054017985 | 0.00114447 | 1.862881966 | |
GOTERM_BP_DIRECT | GO:0007067~mitotic nuclear division | 16 | 0.050840456 | 2.59E-07 | 4.25E-04 | |
GOTERM_BP_DIRECT | GO:0043066~negative regulation of apoptotic process | 16 | 0.050840456 | 3.35E-04 | 0.548772672 | |
GOTERM_BP_DIRECT | GO:0007155~cell adhesion | 16 | 0.050840456 | 3.68E-04 | 0.603031926 | |
GOTERM_CC_DIRECT | GO:0005737~cytoplasm | 75 | 0.238314639 | 0.007215425 | 8.951333631 | |
GOTERM_CC_DIRECT | GO:0005634~nucleus | 74 | 0.23513711 | 0.025200114 | 28.14465235 | |
GOTERM_CC_DIRECT | GO:0070062~extracellular exosome | 71 | 0.225604525 | 7.48E-12 | 9.69E-09 | |
GOTERM_CC_DIRECT | GO:0005829~cytosol | 56 | 0.177941597 | 8.29E-04 | 1.068609771 | |
GOTERM_CC_DIRECT | GO:0005615~extracellular space | 48 | 0.152521369 | 8.69E-13 | 1.13E-09 | |
GOTERM_MF_DIRECT | GO:0005201~extracellular matrix structural constituent | 11 | 0.034952814 | 4.94E-09 | 6.87E-06 | |
GOTERM_MF_DIRECT | GO:0004252~serine-type endopeptidase activity | 14 | 0.044485399 | 9.14E-06 | 0.012713971 | |
GOTERM_MF_DIRECT | GO:0042802~identical protein binding | 23 | 0.073083156 | 6.07E-05 | 0.084464967 | |
GOTERM_MF_DIRECT | GO:0005515~protein binding | 129 | 0.409901179 | 1.08E-04 | 0.150356421 | |
GOTERM_MF_DIRECT | GO:0048407~platelet-derived growth factor binding | 4 | 0.012710114 | 2.37E-04 | 0.329852854 | |
GO: Gene Ontology; FDR: False Discovery Rate. |
Category | Term | Count | % | P-Value | Genes | FDR |
Up-regulated DEGs | hsa05200: Pathways in cancer | 21 | 0.033179549 | 0.012311429 | FGFR2, COL4A3, IL6, BMP2, EPAS1, PTGER4, TGFBR2, GNG11, ZBTB16, MECOM, CXCL12, COL4A5, EDNRA, AGTR1, FOS, EDNRB, LAMA4, ADCY9, PTCH1, AKT3, PIK3R1 | 14.56872647 |
hsa04151: PI3K-Akt signaling pathway | 16 | 0.025279656 | 0.087340443 | FGFR2, COL4A3, FGFR4, IL6, NR4A1, GNG11, IL7R, COL4A5, VWF, LAMA4, ITGA8, TEK, ANGPT1, AKT3, PIK3R1, GHR | 68.70335639 | |
hsa04144: Endocytosis | 14 | 0.022119699 | 0.02683695 | FGFR2, CAV2, FGFR4, CAV1, LDLR, TGFBR2, PIP5K1B, SNX1, HLA-E, ARRB1, FOLR1, NEDD4L, GRK5, RAB11FIP1 | 29.23295979 | |
hsa04010: MAPK signaling pathway | 14 | 0.022119699 | 0.037175664 | FGFR2, FGFR4, TGFBR2, NR4A1, MECOM, CACNA2D2, FOS, DUSP1, ARRB1, RPS6KA2, NTRK2, RRAS, GADD45B, AKT3 | 38.21659244 | |
hsa04610: Complement and coagulation cascades | 13 | 0.020539721 | 6.68E-07 | C7, A2M, C5AR1, F8, SERPING1, C4BPA, C1QB, VWF, CD55, THBD, CFD, CPB2, PROS1 | 8.49E-04 | |
Down-regulated DEGs | hsa04110: Cell cycle | 11 | 0.034952814 | 1.44E-05 | CCNB1, CDK1, CDKN2A, MAD2L1, CCNB2, BUB1, TTK, BUB1B, CDC20, SFN, MCM4 | 0.017175633 |
hsa04151: PI3K-Akt signaling pathway | 11 | 0.034952814 | 0.033131219 | COMP, TNC, COL3A1, COL1A2, EFNA4, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 33.06243008 | |
hsa04512: ECM-receptor interaction | 10 | 0.031775285 | 5.18E-06 | COMP, TNC, COL3A1, COL1A2, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 0.00617031 | |
hsa04510: Focal adhesion | 10 | 0.031775285 | 0.003577108 | COMP, TNC, COL3A1, COL1A2, COL1A1, COL11A1, THBS2, COL5A2, COL5A1, SPP1 | 4.179525016 | |
hsa04115: p53 signaling pathway | 9 | 0.028597757 | 6.04E-06 | CCNB1, CDK1, CDKN2A, CCNB2, RRM2, PMAIP1, SFN, PERP, IGFBP3 | 0.00719634 | |
KEGG: Kyoto Encyclopedia of Genes and Genomes; FDR: False Discovery Rate. |
Gene | Degree of connectivity | Adjusted P value |
IL6 | 84 | 1.09E-05 |
MMP9 | 55 | 2.11E-13 |
EDN1 | 53 | 6.19E-12 |
FOS | 47 | 9.42E-06 |
CDK1 | 44 | 1.99E-16 |
CDH1 | 44 | 4.41E-10 |
BIRC5 | 43 | 1.44E-05 |
VWF | 42 | 7.41E-36 |
UBE2C | 41 | 1.83E-14 |
CDKN3 | 36 | 1.10E-15 |
CDKN2A | 36 | 1.03E-09 |
CD34 | 36 | 3.04E-27 |
AURKA | 33 | 2.33E-14 |
CCNB2 | 33 | 3.01E-17 |
EGR1 | 33 | 2.58E-07 |