We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Identification of hub genes and discovery of promising compounds in gastric cancer based on bioinformatics analysis

    Jiani Huang‡

    Nanjing University of Chinese Medicine, Nanjing210029, Jiangsu Province, China

    College of Traditional ChineseMedicine, College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Fang Wen‡

    Nanjing University of Chinese Medicine, Nanjing210029, Jiangsu Province, China

    Department of Oncology, Affiliated Hospital ofNanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    Department of Oncology, Jiangsu Province Hospitalof Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Wenjie Huang

    Nanjing University of Chinese Medicine, Nanjing210029, Jiangsu Province, China

    Department of Oncology, Affiliated Hospital ofNanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    Department of Oncology, Jiangsu Province Hospitalof Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    ,
    Yingfeng Bai

    Nanjing University of Chinese Medicine, Nanjing210029, Jiangsu Province, China

    College of Traditional ChineseMedicine, College of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China

    ,
    Xiaona Lu

    Nanjing University of Chinese Medicine, Nanjing210029, Jiangsu Province, China

    Department of Oncology, Affiliated Hospital ofNanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    Department of Oncology, Jiangsu Province Hospitalof Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    &
    Peng Shu

    *Author for correspondence:

    E-mail Address: pengshu@ngcc.cn

    Nanjing University of Chinese Medicine, Nanjing210029, Jiangsu Province, China

    Department of Oncology, Affiliated Hospital ofNanjing University of Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    Department of Oncology, Jiangsu Province Hospitalof Chinese Medicine, Nanjing 210029, Jiangsu Province, China

    Published Online:https://doi.org/10.2217/bmm-2019-0608

    Aim: To explore the mechanism of gastric carcinogenesis by mining potential hub genes and to search for promising small-molecular compounds for gastric cancer (GC). Materials & methods: The microarray datasets were downloaded from Gene Expression Omnibus database and the genes and compounds were analyzed by bioinformatics-related tools and software. Results: Six hub genes (MKI67, PLK1, COL1A1, TPX2, COL1A2 and SPP1) related to the prognosis of GC were confirmed to be upregulated in GC and their high expression was correlated with poor overall survival rate in GC patients. In addition, eight candidate compounds with potential anti-GC activity were identified, among which resveratrol was closely correlated with six hub genes. Conclusion: Six hub genes identified in the present study may contribute to a more comprehensive understanding of the mechanism of gastric carcinogenesis and the predicted potential of resveratrol may provide valuable clues for the future development of targeted anti-GC inhibitors.

    References

    • 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68(6), 394–424 (2018).
    • 2. Verdecchia A, Francisci S, Brenner H et al. Recent cancer survival in Europe: a 2000–02 period analysis of EUROCARE-4 data. Lancet Oncol. 8(9), 784–796 (2007).
    • 3. Ajani JA, Lee J, Sano T, Janjigian YY, Fan D, Song S. Gastric adenocarcinoma. Nat. Rev. Dis. Primers 3, 17036 (2017).
    • 4. Chiurillo MA. Role of the Wnt/beta-catenin pathway in gastric cancer: an in-depth literature review. World J. Exp. Med. 5(2), 84–102 (2015).
    • 5. Maeda M, Moro H, Ushijima T. Mechanisms for the induction of gastric cancer by Helicobacter pylori infection: aberrant DNA methylation pathway. Gastric Cancer 20(Suppl. 1), 8–15 (2017).
    • 6. Sadjadi A, Derakhshan MH, Yazdanbod A et al. Neglected role of hookah and opium in gastric carcinogenesis: a cohort study on risk factors and attributable fractions. Int. J. Cancer 134(1), 181–188 (2014).
    • 7. Smyth EC, Turner NC, Pearson A et al. Phase II study of AZD4547 in FGFR amplified tumours: gastroesophageal cancer (GC) cohort pharmacodynamic and biomarker results. J. Clin. Oncol. 34(4), (2016).
    • 8. Lee J, Bendell JC, Rha SY et al. Antitumor activity and safety of FPA144, an ADCC-enhanced, FGFR2b isoform-selective monoclonal antibody, in patients with FGFR2b+gastric cancer and advanced solid tumors. J. Clin. Oncol. 34(15), 2502 (2016).
    • 9. Jin Y, Yang Y. Identification and analysis of genes associated with head and neck squamous cell carcinoma by integrated bioinformatics methods. Mol. Genet. Genomic Med. 7(8), e857 (2019).
    • 10. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science 339(6127), 1546–1558 (2013).
    • 11. Li H, Liu L, Zhuang J et al. Identification of key candidate targets and pathways for the targeted treatment of leukemia stem cells of chronic myelogenous leukemia using bioinformatics analysis. Mol. Genet. Genomic Med. 7(9), e851 (2019).
    • 12. Yang F, Zhai Z, Luo X et al. Bioinformatics identification of key candidate genes and pathways associated with systemic lupus erythematosus. Clin. Rheumatol. doi:10.1007/s10067-019-04751-7 (2019) (Epub ahead of print).
    • 13. Barrett T, Wilhite SE, Ledoux P et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 41(Database issue), D991–995 (2013).
    • 14. Huang Da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4(1), 44–57 (2009).
    • 15. Szklarczyk D, Gable AL, Lyon D et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1), D607–D613 (2019).
    • 16. Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45(W1), W98–W102 (2017).
    • 17. Uhlen M, Fagerberg L, Hallstrom BM et al. Proteomics. Tissue-based map of the human proteome. Science 347(6220), 1260419 (2015).
    • 18. Lamb J, Crawford ED, Peck D et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes and disease. Science 313(5795), 1929–1935 (2006).
    • 19. Lamb J. The Connectivity Map: a new tool for biomedical research. Nat. Rev. Cancer 7(1), 54–60 (2007).
    • 20. Davis AP, Grondin CJ, Johnson RJ et al. The comparative toxicogenomics database: update 2019. Nucleic Acids Res. 47(D1), D948–D954 (2019).
    • 21. Perkel JM. How bioinformatics tools are bringing genetic analysis to the masses. Nature 543(7643), 137–138 (2017).
    • 22. Jiang P, Liu XS. Big data mining yields novel insights on cancer. Nat. Genet. 47(2), 103–104 (2015).
    • 23. Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv. Drug Deliv. Rev. 97, 4–27 (2016).
    • 24. Bonnans C, Chou J, Werb Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 15(12), 786–801 (2014).
    • 25. Walker C, Mojares E, Del Rio Hernandez A. Role of extracellular matrix in development and cancer progression. Int. J. Mol. Sci. 19(10), (2018).
    • 26. Shang C, Sun L, Zhang J et al. Silence of cancer susceptibility candidate 9 inhibits gastric cancer and reverses chemoresistance. Oncotarget 8(9), 15393–15398 (2017).
    • 27. Tian L, Zhao Z, Xie L, Zhu J. MiR-361-5p suppresses chemoresistance of gastric cancer cells by targeting FOXM1 via the PI3K/Akt/mTOR pathway. Oncotarget 9(4), 4886–4896 (2018).
    • 28. Mayer IA, Arteaga CL. The PI3K/AKT pathway as a target for cancer treatment. Annu. Rev. Med. 67, 11–28 (2016).
    • 29. Gan L, Meng J, Xu M et al. Extracellular matrix protein 1 promotes cell metastasis and glucose metabolism by inducing integrin beta4/FAK/SOX2/HIF-1alpha signaling pathway in gastric cancer. Oncogene 37(6), 744–755 (2018).
    • 30. Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J. Cell. Physiol. 182(3), 311–322 (2000).
    • 31. Li LT, Jiang G, Chen Q, Zheng JN. Ki67 is a promising molecular target in the diagnosis of cancer (review). Mol. Med. Rep. 11(3), 1566–1572 (2015).
    • 32. Sugianto J, Sarode V, Peng Y. Ki-67 expression is increased in p16-expressing triple-negative breast carcinoma and correlates with p16 only in p53-negative tumors. Hum. Pathol. 45(4), 802–809 (2014).
    • 33. Li P, Xiao ZT, Braciak TA, Ou QJ, Chen G, Oduncu FS. Association between Ki67 index and clinicopathological features in colorectal cancer. Oncol. Res. Treat. 39(11), 696–702 (2016).
    • 34. Tang J, Gui C, Qiu S, Wang M. The clinicopathological significance of Ki67 in papillary thyroid carcinoma: a suitable indicator? World J. Surg. Oncol. 16(1), 100 (2018).
    • 35. Luo G, Hu Y, Zhang Z et al. Clinicopathologic significance and prognostic value of Ki-67 expression in patients with gastric cancer: a meta-analysis. Oncotarget 8(30), 50273–50283 (2017).
    • 36. Huang G, Chen S, Wang D et al. High Ki67 expression has prognostic value in surgically-resected T3 gastric adenocarcinoma. Clin. Lab. 62(1–2), 141–153 (2016).
    • 37. Ko GH, Go SI, Lee WS et al. Prognostic impact of Ki-67 in patients with gastric cancer-the importance of depth of invasion and histologic differentiation. Medicine (Baltimore) 96(25), e7181 (2017).
    • 38. Lens SM, Voest EE, Medema RH. Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nat. Rev. Cancer 10(12), 825–841 (2010).
    • 39. Combes G, Alharbi I, Braga LG, Elowe S. Playing polo during mitosis: PLK1 takes the lead. Oncogene 36(34), 4819–4827 (2017).
    • 40. Raab M, Sanhaji M, Matthess Y et al. PLK1 has tumor-suppressive potential in APC-truncated colon cancer cells. Nat. Commun. 9(1), 1106 (2018).
    • 41. Ren Y, Bi C, Zhao X et al. PLK1 stabilizes a MYC-dependent kinase network in aggressive B cell lymphomas. J. Clin. Invest. 128(12), 5517–5530 (2018).
    • 42. Hassan QN 2nd, Alinari L, Byrd JC. PLK1: a promising and previously unexplored target in double-hit lymphoma. J. Clin. Invest. 128(12), 5206–5208 (2018).
    • 43. Gutteridge RE, Ndiaye MA, Liu X, Ahmad N. Plk1 inhibitors in cancer therapy: from laboratory to clinics. Mol. Cancer Ther. 15(7), 1427–1435 (2016).
    • 44. Lian G, Li L, Shi Y et al. BI2536, a potent and selective inhibitor of polo-like kinase 1, in combination with cisplatin exerts synergistic effects on gastric cancer cells. Int. J. Oncol. 52(3), 804–814 (2018).
    • 45. Dang SC, Fan YY, Cui L, Chen JX, Qu JG, Gu M. PLK1 as a potential prognostic marker of gastric cancer through MEK-ERK pathway on PDTX models. Onco Targets Ther. 11, 6239–6247 (2018).
    • 46. Cai XP, Chen LD, Song HB, Zhang CX, Yuan ZW, Xiang ZX. PLK1 promotes epithelial-mesenchymal transition and metastasis of gastric carcinoma cells. Am. J. Transl. Res. 8(10), 4172–4183 (2016).
    • 47. Otsu H, Iimori M ando K et al. Gastric cancer patients with high PLK1 expression and DNA aneuploidy correlate with poor prognosis. Oncology 91(1), 31–40 (2016).
    • 48. Exposito JY, Valcourt U, Cluzel C, Lethias C. The fibrillar collagen family. Int. J. Mol. Sci. 11(2), 407–426 (2010).
    • 49. Ma HP, Chang HL, Bamodu OA et al. Collagen 1A1 (COL1A1) is a reliable biomarker and putative therapeutic target for hepatocellular carcinogenesis and metastasis. Cancers (Basel) 11(6), 786 (2019).
    • 50. Wang Q, Yu JH. MiR-129-5p suppresses gastric cancer cell invasion and proliferation by inhibiting COL1A1. Biochem. Cell Biol. 96(1), 19–25 (2018).
    • 51. Shi Y, Wang JH, Xin ZY, Duan ZP, Wang GQ, Li F. Transcription factors and microrna-co-regulated genes in gastric cancer invasion in ex vivo. PLoS ONE 10(4), e0122882 (2015).
    • 52. Li J, Ding YM, Li AQ. Identification of COL1A1 and COL1A2 as candidate prognostic factors in gastric cancer. World J. Surg. Oncol. 14(1), 297 (2016).
    • 53. Shi Y, Duan ZP, Zhang X, Zhang XT, Wang GQ, Li F. downregulation of the let-7i facilitates gastric cancer invasion and metastasis by targeting COL1A1. Protein Cell 10(2), 143–148 (2019).
    • 54. Ao R, Guan L, Wang Y, Wang JN. Silencing of COL1A2, COL6A3 and THBS2 inhibits gastric cancer cell proliferation, migration and invasion while promoting apoptosis through the PI3k-Akt signaling pathway. J. Cell. Biochem. 119(6), 4420–4434 (2018).
    • 55. Wittmann T, Wilm M, Karsenti E, Vernos I. TPX2, A novel xenopus MAP involved in spindle pole organization. J. Cell Biol. 149(7), 1405–1418 (2000).
    • 56. Ma S, Rong X, Gao F, Yang Y, Wei L. TPX2 promotes cell proliferation and migration via PLK1 in OC. Cancer Biomark. 22(3), 443–451 (2018).
    • 57. Yan L, Li Q, Yang J, Qiao B. TPX2-p53-GLIPR1 regulatory circuitry in cell proliferation, invasion and tumor growth of bladder cancer. J. Cell. Biochem. 119(2), 1791–1803 (2018).
    • 58. Liang B, Zheng W, Fang L et al. Overexpressed targeting protein for Xklp2 (TPX2) serves as a promising prognostic marker and therapeutic target for gastric cancer. Cancer Biol. Ther. 17(8), 824–832 (2016).
    • 59. Tomii C, Inokuchi M, Takagi Y et al. TPX2 expression is associated with poor survival in gastric cancer. World J Surg Oncol. 15(1), 14 (2017).
    • 60. Zeng B, Zhou M, Wu H, Xiong Z. SPP1 promotes ovarian cancer progression via Integrin beta1/FAK/AKT signaling pathway. Onco Targets Ther. 11, 1333–1343 (2018).
    • 61. Song SZ, Lin S, Liu JN et al. Targeting of SPP1 by microRNA-340 inhibits gastric cancer cell epithelial-mesenchymal transition through inhibition of the PI3K/AKT signaling pathway. J. Cell. Physiol. 234(10), 18587–18601 (2019).
    • 62. Junnila S, Kokkola A, Mizuguchi T et al. Gene expression analysis identifies over-expression of CXCL1, SPARC, SPP1 and SULF1 in gastric cancer. Genes Chromosomes Cancer 49(1), 28–39 (2010).
    • 63. Deng R, Zhang P, Liu W et al. HDAC is indispensable for IFN-gamma-induced B7-H1 expression in gastric cancer. Clin. Epigenetics 10(1), 153 (2018).
    • 64. Claerhout S, Lim JY, Choi W et al. Gene expression signature analysis identifies vorinostat as a candidate therapy for gastric cancer. PLoS ONE 6(9), e24662 (2011).
    • 65. Goossens JF, Bailly C. Ursodeoxycholic acid and cancer: from chemoprevention to chemotherapy. Pharmacol. Ther. 203, 107396 (2019).
    • 66. Wu YC, Chiu CF, Hsueh CT, Hsueh CT. The role of bile acids in cellular invasiveness of gastric cancer. Cancer Cell Int. 18, 75 (2018).
    • 67. Rauf A, Imran M, Butt MS, Nadeem M, Peters DG, Mubarak MS. Resveratrol as an anti-cancer agent: a review. Crit. Rev. Food Sci. Nutr. 58(9), 1428–1447 (2018).
    • 68. Xu J, Liu D, Niu H et al. Resveratrol reverses Doxorubicin resistance by inhibiting epithelial-mesenchymal transition (EMT) through modulating PTEN/Akt signaling pathway in gastric cancer. J. Exp. Clin. Cancer Res. 36(1), 19 (2017).