Intest Res.  2024 Apr;22(2):199-207. 10.5217/ir.2023.00162.

Unraveling molecular similarities between colorectal polyps and colorectal cancer: a systems biology approach

Affiliations
  • 1Department of Biology, School of Sciences, Razi University, Kermanshah, Islamic Republic of Iran

Abstract

Background/Aims
Colorectal cancer (CRC) and colorectal polyps are intimately linked, with polyps acting as precursors to CRC. Understanding the molecular mechanisms governing their development is crucial for advancing diagnosis and treatment. Employing a systems biology approach, we investigated the molecular similarities between polyp and CRC.
Methods
We analyzed gene expression profiles, protein-protein interactions, transcription factors, and gene ontology to identify common differentially expressed genes (DEGs) and unravel shared molecular pathways.
Results
Our analysis revealed 520 commonly dysregulated genes in polyps and CRC, serving as potential biomarkers and pivotal contributors to disease progression. Gene ontology analysis elucidated distinct biological processes associated with upregulated and downregulated DEGs in both conditions, highlighting common pathways, including signal transduction, cell adhesion, and positive regulation of cell proliferation. Moreover, protein-protein interaction networks shed light on subnetworks involved in rRNA processing, positive regulation of cell proliferation, mRNA splicing, and cell division. Transcription factor analysis identified major regulators and differentially expressed transcription factors in polyp and CRC. Notably, we identified common differentially expressed transcription factors, including ZNF217, NR3C1, KLF5, GATA6, and STAT3, with STAT3 and NR3C1 exhibiting increased expression.
Conclusions
This comprehensive analysis enriches our understanding of the molecular mechanisms underlying polyp formation and CRC development, providing potential targets for further investigation and therapeutic intervention. Our findings contribute substantively to crafting personalized strategies for refining the diagnosis and treatment of polyps and CRC.

Keyword

Colorectal neoplasms; Colorectal polyps; Systems biology; Gene expression; Protein interaction maps

Figure

  • Fig. 1. Cases the identification of differentially expressed genes (DEGs) in various colorectal disease types. (A) The bar graph provides a visual representation of the DEGs, clearly indicating the count of upregulated and downregulated genes for each specific disease type. (B) The Venn diagram illustrates the overlapping instances of DEGs among different colorectal disease types, enabling a comprehensive comparison. (C) The bar chart demonstrates the fold change in expression of 520 DEGs commonly observed across all disease types. CRC, colorectal cancer.

  • Fig. 2. The findings of the gene ontology analysis for the differentially expressed genes (DEGs). (A) The main biological processes (BP) associated with upregulated genes in CRC. (B) The main BP linked to downregulated genes in CRC. (C) The main BP connected to upregulated genes in polyps. (D) The main BP associated with downregulated genes in polyps. (E) The 10 most significant BP shared by both diseases. The number below the diagram indicates the number of genes involved in the process as a percentage of the total number of genes involved, while, the number in front of the column indicates the number of genes involved in the process. CRC, colorectal cancer.

  • Fig. 3. Protein-protein interaction networks with module annotations. The colored nodes represent the modules identified through overlapping neighborhood expansion. The tables provide annotations for the modules, with only those modules having P-values less than 0.05 being chosen. In the network, the color of the nodes indicates the presence of modules, and larger-sized nodes indicate a greater degree of connectivity. (A) Colorectal cancer (CRC) network. (B) Polyp network. (C) Common differentially expressed genes (DEGs) proteins networks.

  • Fig. 4. Three gene regulatory networks: (A) the colorectal cancer DE-TF network, (B) the polyp DE-TF network, and (C) the common DE-TF network. In these networks, blue indicates upregulated TFs, while red indicates downregulated TFs. DE-TFs, differentially expressed transcription factors.


Reference

1. Bozic D, Baralić K, Živančević K, et al. Predicting sulforaphaneinduced adverse effects in colon cancer patients via in silico investigation. Biomed Pharmacother. 2022; 146:112598.
Article
2. Alzahrani SM, Al Doghaither HA, Al-Ghafari AB. General insight into cancer: an overview of colorectal cancer (Review). Mol Clin Oncol. 2021; 15:271.
Article
3. Godkhindi AM, Gowda RM. Automated detection of polyps in CT colonography images using deep learning algorithms in colon cancer diagnosis. Proceedings of the 2017 International Conference on Energy, Communication, Data Analytics and Soft Computing (ICECDS); 2017 Aug 1-2; Chennai. IEEE; 2017. p. 1722-1728.
4. Tanwar S, Goel PM, Johri P, Divan MJ. Classification of benign and malignant colorectal polyps using pit pattern classification. Proceedings of the 4th International Conference: Innovative Advancement in Engineering & Technology (IAET); 2020 Feb 21-22; Jaipur. SSRN; 2020. https://doi.org/10.2139/ssrn.3558374.
5. Suárez J, Mata E, Guerra A, et al. Impact of the COVID-19 pandemic during Spain’s state of emergency on the diagnosis of colorectal cancer. J Surg Oncol. 2021; 123:32–36.
Article
6. González LV, de Miguel Ibáñez R, Sotos FE. Colorectal cancer prevalence and survival in Cuenca (Spain). J Gastrointest Cancer. 2023; 54:80–89.
Article
7. Niu Y, Xue J, Wu X, et al. Clinical significance of serum haptoglobin and protein disulfide-isomerase A3 in the screening, diagnosis, and staging of colorectal cancer. Front Pharmacol. 2022; 13:935500.
Article
8. Chai X, Li Y, Yin Z, et al. Association of meat subtypes with colorectal polyp prevalence: finding from the Lanxi Pre-colorectal Cancer Cohort in China. Front Nutr. 2022; 9:833571.
Article
9. Battaglin F, Naseem M, Lenz HJ, Salem ME. Microsatellite instability in colorectal cancer: overview of its clinical significance and novel perspectives. Clin Adv Hematol Oncol. 2018; 16:735–745.
10. Taieb J, Svrcek M, Cohen R, Basile D, Tougeron D, Phelip JM. Deficient mismatch repair/microsatellite unstable colorectal cancer: diagnosis, prognosis and treatment. Eur J Cancer. 2022; 175:136–157.
Article
11. Wimmer K, Etzler J. Constitutional mismatch repair-deficiency syndrome: have we so far seen only the tip of an iceberg? Hum Genet. 2008; 124:105–122.
Article
12. Goldstein J, Tran B, Ensor J, et al. Multicenter retrospective analysis of metastatic colorectal cancer (CRC) with high-level microsatellite instability (MSI-H). Ann Oncol. 2014; 25:1032–1038.
Article
13. Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017; 45–D362-D368.
Article
14. Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003; 13:2498–2504.
Article
15. Bastian M, Heymann S, Jacomy M. Gephi: an open source software for exploring and manipulating networks. Proceedings of the Third International AAAI Conference on Web and Social Media; 2009 May 17-20; San Jose, CA. AAAI Press; 2009. p. 361-362.
16. Nepusz T, Yu H, Paccanaro A. Detecting overlapping protein complexes in protein-protein interaction networks. Nat Methods. 2012; 9:471–472.
Article
17. Lachmann A, Xu H, Krishnan J, Berger SI, Mazloom AR, Ma’ayan A. ChEA: transcription factor regulation inferred from integrating genome-wide ChIP-X experiments. Bioinformatics. 2010; 26:2438–2444.
Article
18. Huang DW, Sherman BT, Zheng X, et al. Extracting biological meaning from large gene lists with DAVID. Curr Protoc Bioinformatics. 2009; 27:13.11.1–13.11.13.
Article
19. Song M, Emilsson L, Roelstraete B, Ludvigsson JF. Risk of colorectal cancer in first degree relatives of patients with colorectal polyps: nationwide case-control study in Sweden. BMJ. 2021; 373–n877.
Article
20. Wang SS, Zhai GQ, Huang ZG, et al. Nitidine chloride regulates cell function of bladder cancer in vitro through downregulating lymphocyte antigen 75. Naunyn Schmiedebergs Arch Pharmacol. 2023; 396:2071–2085.
Article
21. Yang K, Shen J, Tan FQ, Zheng XY, Xie LP. Antitumor activity of small activating RNAs induced PAWR gene activation in human bladder cancer cells. Int J Med Sci. 2021; 18:3039–3049.
Article
22. Zhang W, Zhu J, He X, et al. Exosome complex genes mediate RNA degradation and predict survival in mantle cell lymphoma. Oncol Lett. 2019; 18:5119–5128.
Article
23. Mouterde M, Daali Y, Rollason V, et al. Joint analysis of phenotypic and genomic diversity sheds light on the evolution of xenobiotic metabolism in humans. Genome Biol Evol. 2022; 14:evac167.
Article
24. Dai L, Mugaanyi J, Zhang T, et al. A pan-cancer bioinformatic analysis of the carcinogenic role of SMARCA1 in human carcinomas. PLoS One. 2022; 17:e0274823.
Article
25. Brien GL, Stegmaier K, Armstrong SA. Targeting chromatin complexes in fusion protein-driven malignancies. Nat Rev Cancer. 2019; 19:255–269.
Article
26. Chen X, Xu W, Ma Z, et al. Circ_0000215 exerts oncogenic function in nasopharyngeal carcinoma by targeting miR-512-5p. Front Cell Dev Biol. 2021; 9:688873.
Article
27. Wu F, Sun Y, Chen J, et al. The oncogenic role of APC/C activator protein Cdc20 by an integrated pan-cancer analysis in human tumors. Front Oncol. 2021; 11:721797.
Article
28. Uehara R, Yamada E, Uehara M, et al. Tgm2-p62-p53 complex may function as an autophagic regulator through Fyn and involve in diabetic kidney disease. J Endocr Soc. 2021; 5(Suppl 1):A442–A443.
Article
29. Chew NJ, Nguyen EV, Su SP, et al. FGFR3 signaling and function in triple negative breast cancer. Cell Commun Signal. 2020; 18:13.
Article
30. Li X, Wang W, Ding X. Pan-cancer investigation of psoriasisrelated BUB1B gene: genetical alteration and oncogenic immunology. Sci Rep. 2023; 13:6058.
Article
31. Hydbring P, Malumbres M, Sicinski P. Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol. 2016; 17:280–292.
Article
32. Alcalde J, Munk M, González-Muñoz M, Panina S, Berchtold MW, Villalobo A. Calmodulin downregulation in conditional knockout HeLa cells inhibits cell migration. Arch Biochem Biophys. 2021; 697:108680.
Article
33. Sun G, Qin J, Qiu Y, et al. Microarray analysis of gene expression in the ovarian cancer cell line HO-8910 with silencing of the ZNF217 gene. Mol Med Rep. 2009; 2:851–855.
Article
34. Chia NY, Deng N, Das K, et al. Regulatory crosstalk between lineage-survival oncogenes KLF5, GATA4 and GATA6 cooperatively promotes gastric cancer development. Gut. 2015; 64:707–719.
Article
35. Sun J, Zhou H, Bao X, et al. lncRNA TUG1 facilitates colorectal cancer stem cell characteristics and chemoresistance by enhancing GATA6 protein stability. Stem Cells Int. 2021; 2021:1075481.
Article
36. Li YY. The effect of a small interfering RNA targeting STAT3 gene on the biological function of human endometrial cancer cell line HEC-1B. Tumor. 2013; (12):1054–1060.
37. Chen W, Zhang J, Fu H, et al. KLF5 is activated by gene amplification in gastric cancer and is essential for gastric cell proliferation. Cells. 2021; 10:1002.
Article
38. Kim DJ, Chan KS, Sano S, Digiovanni J. Signal transducer and activator of transcription 3 (Stat3) in epithelial carcinogenesis. Mol Carcinog. 2007; 46:725–731.
Article
39. Park YY. Genomic analysis of nuclear receptors and miRNAs identifies a role for the NR3C1/miR-200 axis in colon cancer. Genes Genomics. 2021; 43:913–920.
Article
40. Xiong H, Chen Z, Lin B, et al. Naringenin regulates FKBP4/NR3C1/NRF2 axis in autophagy and proliferation of breast cancer and differentiation and maturation of dendritic cell. Front Immunol. 2022; 12:745111.
Article
Full Text Links
  • IR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr