Research article Special Issues

The identification of key biomarkers in patients with lung adenocarcinoma based on bioinformatics

  • Received: 20 February 2019 Accepted: 10 July 2019 Published: 21 August 2019
  • Lung adenocarcinoma (LUAD) is one of the leading causes of cancer death globally. This study aims to investigate the underlying mechanisms implicated with LUAD and identify the key biomarkers. LUAD-associated gene expression dataset (GSE10072) was obtained from GEO database. Based on the GEO2R tool, we screened the differentially expressed genes (DEGs) between the patients with LUAD and normal individuals. Subsequently, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to find out the enriched pathways of these DEGs. Meanwhile, a protein-protein interaction (PPI) network was also employed to construct to visualize the interactions of these DEGs. Finally, the survival analysis of the top5 up-regulated and top5 down-regulated genes were conducted via GEPIA, aiming to figure out their potential effects on LUAD. In our study, a total of 856 DEGs were captured, including 559 up-regulated genes and 297 down-regulated genes. Among these DEGs, the top5 up-regulated genes were AGER, SFTPC, FABP4, CYP4B1 and WIF1 while the top5 down-regulated genes were GREM1, SPINK1, MMP1, COL11A1 and SPP1. GO analysis disclosed that these DEGs were mainly enriched in DNA synthesis, cell adhesion, signal transduction and cell apoptosis. KEGG analysis unveiled that the enriched pathway included pathways in cancer, PI3K/Akt signaling pathway, MAPK signaling pathway and cell cycle. Survival analysis showed that the expression level of ZG16 may correlate with the prognosis of LUAD patients. Furthermore, according to the connectivity degree of these DEGs, we selected the top15 hub genes, namely IL6, MMP9, EDN1, FOS, CDK1, CDH1, BIRC5, VWF, UBE2C, CDKN3, CDKN2A, CD34, AURKA, CCNB2 and EGR1, which were expected to be promising therapeutic target in LUAD. In conclusion, our study disclosed potential biomarkers and candidate targets in LUAD, which could be helpful to the diagnosis and treatment of LUAD.

    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

    Related Papers:

  • Lung adenocarcinoma (LUAD) is one of the leading causes of cancer death globally. This study aims to investigate the underlying mechanisms implicated with LUAD and identify the key biomarkers. LUAD-associated gene expression dataset (GSE10072) was obtained from GEO database. Based on the GEO2R tool, we screened the differentially expressed genes (DEGs) between the patients with LUAD and normal individuals. Subsequently, Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were employed to find out the enriched pathways of these DEGs. Meanwhile, a protein-protein interaction (PPI) network was also employed to construct to visualize the interactions of these DEGs. Finally, the survival analysis of the top5 up-regulated and top5 down-regulated genes were conducted via GEPIA, aiming to figure out their potential effects on LUAD. In our study, a total of 856 DEGs were captured, including 559 up-regulated genes and 297 down-regulated genes. Among these DEGs, the top5 up-regulated genes were AGER, SFTPC, FABP4, CYP4B1 and WIF1 while the top5 down-regulated genes were GREM1, SPINK1, MMP1, COL11A1 and SPP1. GO analysis disclosed that these DEGs were mainly enriched in DNA synthesis, cell adhesion, signal transduction and cell apoptosis. KEGG analysis unveiled that the enriched pathway included pathways in cancer, PI3K/Akt signaling pathway, MAPK signaling pathway and cell cycle. Survival analysis showed that the expression level of ZG16 may correlate with the prognosis of LUAD patients. Furthermore, according to the connectivity degree of these DEGs, we selected the top15 hub genes, namely IL6, MMP9, EDN1, FOS, CDK1, CDH1, BIRC5, VWF, UBE2C, CDKN3, CDKN2A, CD34, AURKA, CCNB2 and EGR1, which were expected to be promising therapeutic target in LUAD. In conclusion, our study disclosed potential biomarkers and candidate targets in LUAD, which could be helpful to the diagnosis and treatment of LUAD.


    加载中


    [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.
  • Reader Comments
  • © 2019 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(5290) PDF downloads(921) Cited by(16)

Article outline

Figures and Tables

Figures(6)  /  Tables(4)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog