Integrative analysis of PTEN-related hub genes and validating drug targets for colorectal cancer
Gang Wang, Zhi Min Zhu, Kun Wang
Integrative analysis of PTEN-related hub genes and validating drug targets for colorectal cancer
Colorectal cancer (CRC) is a heterogeneous disease and one of the most prevalent malignancies worldwide. Previous research has indicated that phosphatase and tensin homolog (PTEN)-related genes found in CRC may serve as potential biomarkers for individualized treatment options. The present study aimed to examine the association between PTEN-related genes and the prognosis of CRC patients by evaluating the significance of PTEN-related hub genes and determining potential mechanisms and genes associated with them. Gene expression profiles and clinical information were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. At present, PTEN mutations have been identified in 7% of CRC patients, according to the most recent TCGA data. Differential expression analysis revealed 54 genes as differentially expressed genes (DEGs) between PTEN-related genes and GEO databases (GSE39582 and GSE6263). Further gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted on PTEN-related DEGs. The prognostic efficacy of the PTEN-related DEG signature was assessed using Kaplan–Meier survival and receiver operating characteristic curve analyses. Bioinformatics methods were utilized to analyze the correlation between PTEN-related DEGs and CRC prognosis, survival, and drug efficacy. Through these analyses, eight prognostic-related PTEN-related hub genes (PPARGC1A, NTRK2, ANK2, PLCB4, STC2, PLAU, CDKN1A, and HPGDS) were identified and a risk prognosis model was constructed. Notably, NTRK2 and HPGDS were found to affect drug treatment response in CRC. Targeting these prognostic-related PTEN-related hub genes can regulate cell death signaling, which may benefit the prognosis of CRC patients and improve drug sensitivity.
colorectal cancer / hub genes / individualized treatment / PTEN / TCGA
[1] |
Brody H. Colorectal cancer. Nature. 2015;521:S1.
|
[2] |
Danielsen SA, Eide PW, Nesbakken A, Guren T, Leithe E, Lothe RA. Portrait of the PI3K/AKT pathway in colorectal cancer. Biochim Biophys Acta. 2015;1855:104-121.
|
[3] |
Courtney KD, Corcoran RB, Engelman JA. The PI3K pathway as drug target in human cancer. J Clin Oncol. 2010;28:1075-1083.
|
[4] |
Cai JC, Li R, Xu XN, Zhang L, Lian R, Fang LS. CK1 alpha suppresses lung tumour growth by stabilizing PTEN and inducing autophagy. Nat Cell Biol. 2018;20:465-478.
|
[5] |
Lee YR, Chen M, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat Rev Mol Cell Biol. 2018;19:547-562.
|
[6] |
Li J, Yen C, Liaw D, et al. PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 1997;275:1943-1947.
|
[7] |
Steck PA, Pershouse MA, Jasser SA, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat Genet. 1997;15:356-362.
|
[8] |
Salmena L, Carracedo A, Pandolfi PP. Tenets of PTEN tumor suppression. Cell. 2008;133:403-414.
|
[9] |
Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nat Genet. 1998;19:348-355.
|
[10] |
Podsypanina K, Ellenson LH, Nemes A, et al. Mutation of Pten/Mmac1 in mice causes neoplasia in multiple organ systems. Proc Natl Acad Sci U S A. 1999;96:1563-1568.
|
[11] |
Alimonti A, Carracedo A, Clohessy JG, et al. Subtle variations in Pten dose determine cancer susceptibility. Nat Genet. 2010;42:454-458.
|
[12] |
Yang W, Soares J, Greninger P, et al. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41(Database issue):D955-D961.
|
[13] |
Reimand J, Isser R, Voisin V, et al. Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap. Nat Protoc. 2019;14(2):482-517.
|
[14] |
Li Q, Yunpeng Bai L, Lyle T, et al. Mechanism of PRL2 phosphatase-mediated PTEN degradation and tumorigenesis. Proc Natl Acad Sci U S A. 2020;117(34):20538-20548.
|
[15] |
Chang H, Cai Z, Roberts TM. The mechanisms underlying PTEN loss in human tumors suggest potential therapeutic opportunities. Biomolecules. 2019;9(11):713.
|
[16] |
Zhang Y, Wang J, Wu D. Il-21-secreting hucmscs combined with mir-200c inhibit tumor growth and metastasis via repression of wnt/β-catenin signaling and epithelial–mesenchymal transition in epithelial ovarian cancer. Oncotargets Ther. 2018;11:2037-2050.
|
[17] |
Zhao D, Lu X, Wang G. Synthetic essentiality of chromatin remodelling factor chd1 in pten-deficient cancer. Nature. 2017;542(7642):484-488.
|
[18] |
Qi C, Zhou T, Bai Y, et al. China special issue on gastrointestinal tumor-NTRK fusion in a large real-world population and clinical utility of circulating tumor DNA genotyping to guide TRK inhibitor treatment. Int J Cancer. 2023;153:1916-1927.
CrossRef
Google scholar
|
[19] |
Khotskaya YB, Holla VR, Farago AF, Mills Shaw KR, Meric-Bernstam F, Hong DS. Targeting TRK family proteins in cancer. Pharmacol Ther. 2017;173:58-66.
|
[20] |
Xu Y, Jiang WG, Wang HC, et al. BDNF activates TrkB/PLCγ1 signaling pathway to promote proliferation and invasion of ovarian cancer cells through inhibition of apoptosis. Eur Rev Med Pharmacol Sci. 2019;23:5093-5100.
|
[21] |
Li W, Lu Y, Yu X, Yong M, Ma D, Gao Q. Detection of exosomal tyrosine receptor kinase B as a potential biomarker in ovarian cancer. J Cell Biochem. 2019;120(4):6361-6369.
|
[22] |
Wang X, Xu Z, Chen X, Ren X, Wei J, Zhou S. A tropomyosin receptor kinase family protein, NTRK2 is a potential predictive biomarker for lung adenocarcinoma. Peer J. 2019;7:e7125.
|
[23] |
Suh K, Carlson JJ, Xia F, Williamson T, Sullivan SD. The potential long-term comparative effectiveness of larotrectinib vs standard of care for treatment of metastatic TRK fusion thyroid cancer, colorectal cancer, and soft tissue sarcoma. J Manag Care Spec Pharm. 2022;28(6):622-630.
|
[24] |
Suh K, Carlson JJ, Xia F, Williamson T, Sullivan SD. Comparative effectiveness of larotrectinib versus entrectinib for the treatment of metastatic NTRK gene fusion cancers. J Comp Eff Res. 2022;11(14):1011-1019.
|
[25] |
Woodcock J, LaVange LM. Master protocols to study multiple therapies, multiple diseases, or both. N Engl J Med. 2017;377(1):62-70.
|
[26] |
Rosati G, Aprile G, Colombo A, et al. Colorectal cancer heterogeneity and the impact on precision medicine and therapy efficacy. Biomedicine. 2022;10(5):1035.
|
[27] |
Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol. 2020;21(4):531-540.
|
[28] |
Harada G, Drilon A. TRK inhibitor activity and resistance in TRK fusion-positive cancers in adults. Cancer Genet. 2022;264-265:33-39.
|
[29] |
Meng L, Liu B, Ji R, Jiang X, Yan X, Xin Y. Targeting the BDNF/TrkB pathway for the treatment of tumors. Oncol Lett. 2019;17(2):2031-2039.
|
[30] |
Thomaz A, Pinheiro KV, Souza BK, et al. Antitumor activities and cellular changes induced by TrkB inhibition in medulloblastoma. Front Pharmacol. 2019;10:698.
|
[31] |
Olson KL, Holt MC, Ciske FL, et al. Novel amide and imidazole compounds as potent hematopoietic prostaglandin D2 synthase inhibitors. Bioorg Med Chem Lett. 2021;34:127759.
|
[32] |
Kanaoka Y, Ago H, Inagaki E, et al. Cloning and crystal structure of hematopoietic prostaglandin D synthase. Cell. 1997;90(6):1085-1095.
|
[33] |
Seo MJ, Oh DK. Prostaglandin synthases: molecular characterization and involvement in prostaglandin biosynthesis. Prog Lipid Res. 2017;66:50-68.
|
[34] |
Zhang J, Zhang Y, Gong H, et al. Genetic mapping using 1.4M SNP array refined loci for fatty acid composition traits in Chinese Erhualian and Bamaxiang pigs. J Anim Breed Genet. 2017;134(6):472-483.
|
[35] |
Wang J, Mak O. Induction of apoptosis in non-small cell lung carcinoma A549 cells by PGD2 metabolite, 15d-PGJ2. Cell Biol Int. 2011;35(11):1089-1096.
|
[36] |
Lyon AM, Tesmer JJ. Structural insights into phospholipase C-beta function. Mol Pharmacol. 2013;84(4):488-500.
|
[37] |
Castano E, Yildirim S, Fáberová V, et al. Nuclear phosphoinositides-versatile regulators of genome functions. Cell. 2019;8(7):649.
|
[38] |
Lee C-C, Lee A-W, Wei P-L, Liu Y-S, Chang Y-J, Huang C-Y. In silico analysis to identify miR-1271-5p/PLCB4 (phospholipase C Beta 4) axis mediated oxaliplatin resistance in metastatic colorectal cancer. Sci Rep. 2023;13:4366.
|
[39] |
Wang G, Yang Y, Wang Y-Z, Zhu Z-M, Yin P-H, Ke X. The effects and mechanisms of fatty acid metabolism mediated glycolysis regulated by betulinic acid loaded nanoliposomes in colorectal cancer. Oncol Rep. 2020;44(6):2595-2609.
|
[40] |
Wang G, Wang Y-Z, Yu Y, Yin P-H, Xu K, Zhang H. The anti-tumor effect and mechanism of triterpenoids in Rhus chinensis Mill. on reversing effector CD8+ T-cells dysfunction by targeting glycolysis pathways in colorectal cancer. Integr Cancer Ther. 2021;20:15347354211017219.
|
[41] |
Wang G, Yang Y, Li Z-M, Zhu Z-M, Wang Z-J, Tao M-F. Triterpenoids of Rhus chinensis supressed colorectal cancer progress by enhancing antitumor immunity and CD8+T cells tumor infiltration. Nutr Cancer. 2022;74(7):2550-2564.
|
[42] |
Wang G, Yang Y, Yi D, et al. Eudragit S100 prepared pH-responsive liposomes-loaded betulinic acid against colorectal cancer in vitro and in vivo. J Liposome Res. 2022;32(3):250-264.
|
[43] |
Wang G, Wang JJ, Xu XN, Shi F, Fu XL. Targeting cellular energy metabolism- mediated ferroptosis by small molecule compounds for colorectal cancer therapy. J Drug Target. 2022;30(8):819-832.
|
[44] |
Hashemzadeh S, Arabzadeh AA, Estiar MA, et al. Clinical utility of measuring expression levels of Stanniocalcin 2 in patients with colorectal cancer. Med Oncol. 2014;31(10):237.
|
[45] |
Zhang C, Chen S, Ma X, et al. Upregulation of STC2 in colorectal cancer and its clinicopathological significance. Onco Targets Ther. 2019;12:1249-1258.
|
[46] |
Wang J, Sahengbieke S, Xu X, et al. Gene expression analyses identify a relationship between stanniocalcin 2 and the malignant behavior of colorectal cancer. Onco Targets Ther. 2018;11:7155-7168.
|
[47] |
Sunita BS, Sen A, Suhag V. To evaluate immunoreactivity of cyclooxygenase-2 in cases of endometrial carcinoma and correlate it with expression of p53 and vascular endothelial growth factor. J Cancer Res Ther. 2018;14(6):1366-1372.
|
[48] |
Miyazaki S, Kikuchi H, Iino I, et al. Anti-VEGF antibody therapy induces tumor hypoxia and stanniocalcin 2 expression and potentiates growth of human colon cancer xenografts. Int J Cancer. 2014;135(2):295-307.
|
[49] |
Yuan Q, Zhan L, Zhang L-L, et al. Stanniocalcin 2 induces oxaliplatin resistance in colorectal cancer cells by upregulating P-glycoprotein. Can J Physiol Pharmacol. 2016;94(9):929-935.
|
[50] |
Wu J, Lai M, Shao C, Wang J, Wei JJ. STC2 overexpression mediated by HMGA2 is a biomarker for aggressiveness of high-grade serous ovarian cancer. Oncol Rep. 2015;34(3):1494-1502.
|
/
〈 | 〉 |