Skip to main content

Advertisement

Log in

Identification of candidate biomarkers and therapeutic drugs of colorectal cancer by integrated bioinformatics analysis

  • Original Paper
  • Published:
Medical Oncology Aims and scope Submit manuscript

Abstract

Most colorectal cancer (CRC) patients are diagnosed with advanced stages and low prognosis. We aimed to identify potential diagnostic and prognostic biomarkers, as well as active small molecules of CRC. Microarray data (GSE9348, GSE35279, and GSE106582) were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified by the GEO2R platform. Common DEGs were selected for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Cytoscape software was used to construct protein–protein interaction networks and identify hub genes. Hub genes were evaluated by Kaplan–Meier survival analysis in the GEPIA database and validated in two independent microarray data (GSE74602 and GSE83889). Common DEGs were used to select active small molecules by the connectivity map database. A total of 166 DEGs were identified as common DEGs. GO analysis demonstrated that common DEGs were significantly enriched in the apoptotic process, cell proliferation, and cell adhesion. KEGG analysis indicated that the most enriched pathways were the PI3K-Akt signaling pathway and extracellular matrix-receptor interaction. COL1A2, THBS2, TIMP1, and CXCL8 significantly upregulated in colorectal tumor. High expressions of COL1A2, THBS2, and TIMP1 were associated with poor survival, while high expressions of CXCL8 were associated with better survival. We selected 11 small molecules for CRC therapy. In conclusion, we found key dysregulated genes associated with CRC and potential small molecules to reverse them. COL1A2, THBS2, TIMP1, and CXCL8 may act as diagnostic and prognostic biomarkers of CRC.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Data availability

The datasets analyzed during the current study are available in the Gene Expression Omnibus repository (https://www.ncbi.nlm.nih.gov/geo/), GEPIA repository (https://gepia.cancer-pku.cn/detail.php), and connectivity map database (https://portals.broadinstitute.org/CMap/).

Abbreviations

CRC:

Colorectal cancer

GEO:

Gene Expression Omnibus

DEG:

Differentially expressed genes

GO:

Gene Ontology

KEGG:

Kyoto Encyclopedia of Genes and Genomes

PPI:

Protein–protein interaction

COL1A2:

Collagen type I alpha 2 chain

THBS2:

Thrombospondin 2

CXCL8:

C-X-C motif chemokine ligand 8

TIMP1:

TIMP metallopeptidase inhibitor 1

CMap:

Connectivity map

logFC:

Log fold change

DAVID:

Database for Annotation Visualization and Integrated Discovery

BP:

Biological processes

CC:

Cellular components

MF:

Molecular functions

MCC:

Maximum Correlation Criteria

ECM:

Extracellular matrix

References

  1. Ferlay J, et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 2019;144(8):1941–53.

    PubMed  CAS  Google Scholar 

  2. Rossi, M, et al. Colorectal cancer and alcohol consumption—populations to molecules. Cancers (Basel). 2018;10(2):38.

  3. Hofseth LJ, et al. Early-onset colorectal cancer: initial clues and current views. Nat Rev Gastroenterol Hepatol. 2020;17(6):352–64.

    PubMed  Google Scholar 

  4. Koncina E, et al. Prognostic and Predictive Molecular Biomarkers for Colorectal Cancer: Updates and Challenges. Cancers (Basel). 2020;12(2):319.

    CAS  Google Scholar 

  5. Carethers JM, Jung BH. Genetics and Genetic Biomarkers in Sporadic Colorectal Cancer. Gastroenterology. 2015;149(5):1177–90.

    PubMed  PubMed Central  CAS  Google Scholar 

  6. Liu YR, et al. Neurexophilin and PC-esterase domain family member 4 (NXPE4) and prostate androgen-regulated mucin-like protein 1 (PARM1) as prognostic biomarkers for colorectal cancer. J Cell Biochem. 2019;120(10):18041–52.

    PubMed  CAS  Google Scholar 

  7. Xie XJ, et al. The Generation and Validation of a 20-Genes Model Influencing the Prognosis of Colorectal Cancer. J Cell Biochem. 2017;118(11):3675–85.

    PubMed  CAS  Google Scholar 

  8. Sun G, et al. Identification of a five-gene signature with prognostic value in colorectal cancer. J Cell Physiol. 2019;234(4):3829–36.

    PubMed  CAS  Google Scholar 

  9. Wan L, et al. HOTAIRM1 as a potential biomarker for diagnosis of colorectal cancer functions the role in the tumour suppressor. J Cell Mol Med. 2016;20(11):2036–44.

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Wang W, et al. Gene selection for the discrimination of colorectal cancer. Curr Mol Med. 2020;20(6):415–28.

    PubMed  Google Scholar 

  11. Giopanou I, Pintzas A. RAS and BRAF in the foreground for non-small cell lung cancer and colorectal cancer: Similarities and main differences for prognosis and therapies. Crit Rev Oncol Hematol. 2020;146:102859.

    PubMed  Google Scholar 

  12. Smoot ME, et al. Cytoscape 28 new features for data integration and network visualization. Bioinformatics. 2011;27(3):431–2.

    PubMed  CAS  Google Scholar 

  13. Chin C, et al. Cytohubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 2014;8(Suppl 4):S11.

  14. Tang Z, et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45:98–102.

    Google Scholar 

  15. Lamb J. The Connectivity Map: a new tool for biomedical research. Nat Rev Cancer. 2007;7(1):54–60.

    PubMed  CAS  Google Scholar 

  16. Wu D, Wang X. Application of clinical bioinformatics in lung cancer-specific biomarkers. Cancer Metastasis Rev. 2015;34(2):209–16.

    PubMed  CAS  Google Scholar 

  17. Liang Y, et al. Gene expression profiling reveals molecularly and clinically distinct subtypes of glioblastoma multiforme. Proc Natl Acad Sci U S A. 2005;102(16):5814–9.

    PubMed  PubMed Central  CAS  Google Scholar 

  18. Bahrami A, et al. Therapeutic Potential of Targeting PI3K/AKT Pathway in Treatment of Colorectal Cancer: Rational and Progress. J Cell Biochem. 2018;119(3):2460–9.

    PubMed  CAS  Google Scholar 

  19. Ishaque N, et al. Whole genome sequencing puts forward hypotheses on metastasis evolution and therapy in colorectal cancer. Nat Commun. 2018;9(1):4782.

    PubMed  PubMed Central  Google Scholar 

  20. Stelzl U, et al. A human protein-protein interaction network: a resource for annotating the proteome. Cell. 2005;122(6):957–68.

    PubMed  CAS  Google Scholar 

  21. Zou X, et al. Up-regulation of type I collagen during tumorigenesis of colorectal cancer revealed by quantitative proteomic analysis. J Proteomics. 2013;94:473–85.

    PubMed  CAS  Google Scholar 

  22. Rodia MT, et al. LGALS4, CEACAM6, TSPAN8, and COL1A2: Blood Markers for Colorectal Cancer-Validation in a Cohort of Subjects With Positive Fecal Immunochemical Test Result. Clin Colorectal Cancer. 2018;17(2):217–28.

    Google Scholar 

  23. Yu Y, et al. The inhibitory effects of COL1A2 on colorectal cancer cell proliferation, migration, and invasion. J Cancer. 2018;9(16):2953–62.

    PubMed  PubMed Central  Google Scholar 

  24. Ao R, et al. 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. 2018;119(6):4420–34.

    PubMed  CAS  Google Scholar 

  25. Slattery ML, et al. The PI3K/AKT signaling pathway: Associations of miRNAs with dysregulated gene expression in colorectal cancer. Mol Carcinog. 2018;57(2):243–61.

    PubMed  CAS  Google Scholar 

  26. Tian Q, et al. THBS2 is a biomarker for AJCC stages and a strong prognostic indicator in colorectal cancer. J BUON. 2018;23(5):1331–6.

    PubMed  Google Scholar 

  27. Sun XY, et al. MiR-93-5p promotes cervical cancer progression by targeting THBS2/MMPS signal pathway. Eur Rev Med Pharmacol Sci. 2019;23(12):5113–211.

    PubMed  Google Scholar 

  28. Lambert E, et al. TIMPs as multifacial proteins. Crit Rev Oncol Hematol. 2004;49(3):187–98.

    PubMed  Google Scholar 

  29. Song G, et al. TIMP1 is a prognostic marker for the progression and metastasis of colon cancer through FAK-PI3K/AKT and MAPK pathway. J Exp Clin Cancer Res. 2016;35(1):148.

    PubMed  PubMed Central  Google Scholar 

  30. Pellegrini P, et al. Simultaneous measurement of soluble carcinoembryonic antigen and the tissue inhibitor of metalloproteinase TIMP1 serum levels for use as markers of pre-invasive to invasive colorectal cancer. Cancer Immunol Immunother. 2000;49(7):388–94.

    PubMed  CAS  Google Scholar 

  31. Eckfeld C, et al. Functional disparities within the TIMP family in cancer: hints from molecular divergence. Cancer Metastasis Rev. 2019;38(3):469–81.

    PubMed  CAS  Google Scholar 

  32. Prokopchuk O, et al. Elevated systemic levels of the matrix metalloproteinase inhibitor TIMP-1 correlate with clinical markers of cachexia in patients with chronic pancreatitis and pancreatic cancer. BMC Cancer. 2018;18(1):128.

    PubMed  PubMed Central  Google Scholar 

  33. Dabkeviciene D, et al. The role of interleukin-8 (CXCL8) and CXCR2 in acquired chemoresistance of human colorectal carcinoma cells HCT116. Med Oncol. 2015;32(12):258.

    PubMed  Google Scholar 

  34. Pączek S, et al. CXCL-8 in Preoperative Colorectal Cancer Patients: Significance for Diagnosis and Cancer Progression. Int J Mol Sci. 2020;21(6):2040.

    PubMed Central  Google Scholar 

  35. Ha H, Debnath B, Neamati N. Role of the CXCL8-CXCR1/2 Axis in Cancer and Inflammatory Diseases. Theranostics. 2017;7(6):1543–88.

    PubMed  PubMed Central  CAS  Google Scholar 

  36. Lee YS, et al. Interleukin-8 and its receptor CXCR2 in the tumour microenvironment promote colon cancer growth, progression and metastasis. Br J Cancer. 2012;106(11):1833–41.

    PubMed  PubMed Central  CAS  Google Scholar 

  37. Ning Y, et al. Interleukin-8 is associated with proliferation, migration, angiogenesis and chemosensitivity in vitro and in vivo in colon cancer cell line models. Int J Cancer. 2011;128(9):2038–49.

    PubMed  PubMed Central  CAS  Google Scholar 

  38. Moody TW, et al. VIP-ellipticine derivatives inhibit the growth of breast cancer cells. Life Sci. 2002;71(9):1005–144.

    PubMed  CAS  Google Scholar 

  39. Duan C, et al. Piperlongumine induces gastric cancer cell apoptosis and G2/M cell cycle arrest both in vitro and in vivo. Tumour Biol. 2016;37(8):10793–804.

    PubMed  CAS  Google Scholar 

  40. Hall JA, et al. Novobiocin Analogues That Inhibit the MAPK Pathway. J Med Chem. 2016;59(3):925–33.

    PubMed  PubMed Central  CAS  Google Scholar 

  41. Al-Saeedi FJ. Study of the cytotoxicity of asiaticoside on rats and tumour cells. BMC Cancer. 2014;14:220.

    PubMed  PubMed Central  Google Scholar 

  42. Eckschlager T, et al. Histone Deacetylase Inhibitors as Anticancer Drugs. Int J Mol Sci. 2017;18(7):1414.

    PubMed Central  Google Scholar 

  43. Lu C, Wang W, El-Deiry WS. Non-genotoxic anti-neoplastic effects of ellipticine derivative NSC176327 in p53-deficient human colon carcinoma cells involve stimulation of p73. Cancer Biol Ther. 2008;7(12):2039–46.

    PubMed  PubMed Central  CAS  Google Scholar 

  44. Kuo YC, et al. Ellipticine induces apoptosis through p53-dependent pathway in human hepatocellular carcinoma HepG2 cells. Life Sci. 2006;78(22):2550–7.

    PubMed  CAS  Google Scholar 

  45. Martinkova E, et al. alpha5beta1 integrin antagonists reduce chemotherapy-induced premature senescence and facilitate apoptosis in human glioblastoma cells. Int J Cancer. 2010;127(5):1240–8.

    PubMed  CAS  Google Scholar 

Download references

Funding

This work was supported by the National Natural Science Foundation of China (No. 31371290), the Frontier and Key Technology Innovation Project of Guangdong Province (No.2014B010118003), and the Science and Technology Planning Project of Guangdong Province (No. 2015B010129008).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. DEG identification and gene functional enrichment analysis were performed by JX and LX. The PPI network construction and hub genes verification were performed by MG, LQ, CD, and ZL. The active small molecules selection was finished by YW, JX, and QW. The first draft of the manuscript was written by ZZ, XL, and WW. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Wenhui Wang or Xiaoyan Li.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

12032_2020_1425_MOESM1_ESM.tif

Supplementary file1 Supplementary Fig. 1 Venn diagram of the common DEGs among GSE9348, GSE35279 and GSE106582. a 78 common upregulated genes. b 88 common downregulated genes (TIF 11085 kb)

Supplementary file2 (DOCX 15 kb)

Supplementary file3 (DOCX 16 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zheng, Z., Xie, J., Xiong, L. et al. Identification of candidate biomarkers and therapeutic drugs of colorectal cancer by integrated bioinformatics analysis. Med Oncol 37, 104 (2020). https://doi.org/10.1007/s12032-020-01425-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12032-020-01425-2

Keyword

Navigation