Horm Metab Res
DOI: 10.1055/a-2209-0771
Original Article: Endocrine Research

Identification of Key Genes and Related Drugs of Adrenocortical Carcinoma by Integrated Bioinformatics Analysis

1   The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
,
Xiao-chun Zeng
1   The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
,
Kui-rong Ji
2   Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
,
Ling-yi Zhang
2   Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
,
Xiao-min Chen
2   Department of Endocrinology, Zhongshan Hospital Xiamen University, Xiamen, China
› Institutsangaben

Abstract

Adrenocortical carcinoma (ACC) is a malignant carcinoma with an extremely poor prognosis, and its pathogenesis remains to be understood to date, necessitating further investigation. This study aims to discover biomarkers and potential therapeutic agents for ACC through bioinformatics, enhancing clinical diagnosis and treatment strategies. Differentially expressed genes (DEGs) between ACC and normal adrenal cortex were screened out from the GSE19750 and GSE90713 datasets available in the GEO database. An online Venn diagram tool was utilized to identify the common DEGs between the two datasets. The identified DEGs were subjected to functional assessment, pathway enrichment, and identification of hub genes by performing the protein-protein interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The differences in the expressions of hub genes between ACC and normal adrenal cortex were validated at the GEPIA2 website, and the association of these genes with the overall patient survival was also assessed. Finally, on the QuartataWeb website, drugs related to the identified hub genes were determined. A total of 114 DEGs, 10 hub genes, and 69 known drugs that could interact with these genes were identified. The GO and KEGG analyses revealed a close association of the identified DEGs with cellular signal transduction. The 10 hub genes identified were overexpressed in ACC, in addition to being significantly associated with adverse prognosis in ACC. Three genes and the associated known drugs were identified as potential targets for ACC treatment.

Supplementary Material



Publikationsverlauf

Eingereicht: 25. September 2023

Angenommen nach Revision: 01. November 2023

Artikel online veröffentlicht:
18. Dezember 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Zheng S, Cherniack AD, Dewal N. et al. Comprehensive pan-genomic characterization of adrenocortical carcinoma. Cancer Cell 2016; 29: 723-736
  • 2 Jasim S, Habra MA. Management of adrenocortical carcinoma. Curr Oncol Rep 2019; 21: 1-11
  • 3 Gambella A, Volante M, Papotti M. Histopathologic features of adrenal cortical carcinoma. Adv Anat Pathol 2023; 30: 34-46
  • 4 Bussey KJ, Demeure MJ. Toward a pathway-centered approach for the treatment of adrenocortical carcinoma. Curr Opin Oncol 2011; 23: 34-44
  • 5 Stigliano A, Cerquetti L, Lardo P. et al. New insights and future perspectives in the therapeutic strategy of adrenocortical carcinoma. Oncol Rep 2017; 37: 1301-1311
  • 6 Long SE, Miller BS. Adrenocortical cancer treatment. Surg Clin North Am 2019; 99: 759-771
  • 7 Stojadinovic A, Ghossein RA, Hoos A. et al. Adrenocortical carcinoma: clinical, morphologic, and molecular characterization. J Clin Oncol 2002; 20: 941-950
  • 8 Wilmouth JJ, Olabe J, Garcia-Garcia D. et al. Sexually dimorphic activation of innate antitumor immunity prevents adrenocortical carcinoma development. Sci Adv 2022; 8: eadd0422
  • 9 Shimada H, Yamazaki Y, Sugawara A. et al. Molecular mechanisms of functional adrenocortical adenoma and carcinoma: genetic characterization and intracellular signaling pathway. Biomedicines 2021; 9: 892
  • 10 Pitsava G, Maria AG, Faucz FR. Disorders of the adrenal cortex: Genetic and molecular aspects. Front Endocrinol (Lausanne) 2022; 13: 931389
  • 11 Georgantzoglou N, Kokkali S, Tsourouflis G. et al. Tumor microenvironment in adrenocortical carcinoma: barrier to immunotherapy success?. Cancers (Basel) 2021; 13: 1798
  • 12 Perge P, Decmann Á, Pezzani R. et al. Analysis of circulating extracellular vesicle-associated microRNAs in cortisol-producing adrenocortical tumors. Endocrine 2018; 59: 280-287
  • 13 Murtha TD, Brown TC, Rubinstein JC. et al. Overexpression of cytochrome P450 2A6 in adrenocortical carcinoma. Surgery 2017; 161: 1667-1674
  • 14 Bussey KJ, Bapat A, Linnehan C. et al. Targeting polo-like kinase 1, a regulator of p53, in the treatment of adrenocortical carcinoma. Clin Transl Med 2016; 5: 1-14
  • 15 Huang YG, Wang Y, Zhu R-J. et al. EMS1/DLL4-Notch signaling axis augments cell cycle-mediated tumorigenesis and progress in human adrenocortical carcinoma. Front Oncol 2021; 11: 771579
  • 16 Landwehr LS, Altieri B, Schreiner J. et al. Interplay between glucocorticoids and tumor-infiltrating lymphocytes on the prognosis of adrenocortical carcinoma. J Immunother Cancer 2020; 8: e000469
  • 17 Lin Z, Tan C, Qiu Q. et al. Ubiquitin-specific protease 22 is a deubiquitinase of CCNB1. Cell Discov 2015; 1: 1-16
  • 18 Liu WT, Chen C, Lu IC. et al. MJ-66 induces malignant glioma cells G2/M phase arrest and mitotic catastrophe through regulation of cyclin B1/Cdk1 complex. Neuropharmacology 2014; 86: 219-227
  • 19 Fang Y, Yu H, Liang X. et al. Chk1-induced CCNB1 overexpression promotes cell proliferation and tumor growth in human colorectal cancer. Cancer Biol Ther 2014; 15: 1268-1279
  • 20 Chen EB, Qin X, Peng K. et al. HnRNPR-CCNB1/CENPF axis contributes to gastric cancer proliferation and metastasis. Aging (Albany NY) 2019; 11: 7473
  • 21 Gu J, Liu X, Li J. et al. MicroRNA-144 inhibits cell proliferation, migration and invasion in human hepatocellular carcinoma by targeting CCNB1. Cancer Cell Int 2019; 19: 1-10
  • 22 Zhao M, Kim Y, Yoon B. et al. Expression profiling of cyclin B1 and D1 in cervical carcinoma. Exp Oncol 2006; 28: 44-48
  • 23 Moon SJ, Kim JH, Kong SH. et al. Protein expression of cyclin B1, transferrin receptor, and fibronectin is correlated with the prognosis of adrenal cortical carcinoma. Endocrinol Metab 2020; 35: 132-141
  • 24 Lei Cy, Wang W, Zhu Yt. et al. The decrease of cyclin B2 expression inhibits invasion and metastasis of bladder cancer. Urol Oncol 2016; 34: 237.e1-237.e10
  • 25 Sun C, Lowe S, Ma S. et al. CCNB2 expression correlates with worse outcomes in breast cancer patients: a pooled analysis. Women Health 2022; 62: 655-663
  • 26 Wu S, Su R, Jia H. Cyclin B2 (CCNB2) stimulates the proliferation of triple-negative breast cancer (TNBC) cells in vitro and in vivo. Dis Markers 2021; 5511041
  • 27 Ikeya A, Nakashima M, Yamashita M. et al. CCNB2 and AURKA overexpression may cause atypical mitosis in Japanese cortisol-producing adrenocortical carcinoma with TP53 somatic variant. PLoS One 2020; 15: e0231665
  • 28 Wang A, Dai H, Gong Y. et al. ANLN-induced EZH2 upregulation promotes pancreatic cancer progression by mediating miR-218-5p/LASP1 signaling axis. J Exp Clin Cancer Res 2019; 38: 1-20
  • 29 Wang G, Shen W, Cui L. et al. Overexpression of Anillin (ANLN) is correlated with colorectal cancer progression and poor prognosis. Cancer Biomark 2016; 16: 459-465
  • 30 Zhang L, Wei Y, He Y. et al. Clinical implication and immunological landscape analyses of ANLN in pan-cancer: a new target for cancer research. Cancer Med 2023; 12: 4907-4920
  • 31 Zhang X, Li L, Huang S. et al. Comprehensive analysis of ANLN in human tumors: a prognostic biomarker associated with cancer immunity. Oxid Med Cell Longev 2022; 5322929
  • 32 Liu K, Cui L, Li C. et al. Pan-cancer analysis of the prognostic and immunological role of ANLN: an onco-immunological biomarker. Front Genet 2022; 13: 922472
  • 33 Zhou H, Zou M, Ding X. et al. Role of Bclaf1 in promoting adrenocortical carcinoma proliferation: a study combining the use of bioinformatics and molecular events. Cancer Manag Res 2021; 13: 6785-6795
  • 34 Ren L, Yang Y, Li W. et al. CDK1 serves as a therapeutic target of adrenocortical carcinoma via regulating epithelial–mesenchymal transition, G2/M phase transition, and PANoptosis. J Transl Med 2022; 20: 444
  • 35 Huang Yg, Li D, Wang L. et al. CENPF/CDK1 signaling pathway enhances the progression of adrenocortical carcinoma by regulating the G2/M-phase cell cycle. J Transl Med 2022; 20: 78
  • 36 Sunada S, Saito H, Zhang D. et al. CDK1 inhibitor controls G2/M phase transition and reverses DNA damage sensitivity. Biochem Biophys Res Commun 2021; 550: 56-61
  • 37 Wang J, Che W, Wang W. et al. CDKN3 promotes tumor progression and confers cisplatin resistance via RAD51 in esophageal cancer. Cancer Manag Res 2019; 11: 3253-3264
  • 38 Li Y, Ji S, Fu LY. et al. Knockdown of cyclin-dependent kinase inhibitor 3 inhibits proliferation and invasion in human gastric cancer cells. Oncol Res 2017; 25: 721-731
  • 39 Liu D, Zhang J, Wu Y. et al. YY1 suppresses proliferation and migration of pancreatic ductal adenocarcinoma by regulating the CDKN3/MdM2/P53/P21 signaling pathway. Int J Cancer 2018; 142: 1392-1404
  • 40 Fan C, Chen L, Huang Q. et al. Overexpression of major CDKN3 transcripts is associated with poor survival in lung adenocarcinoma. Br J Cancer 2015; 113: 1735-1743
  • 41 Li WH, Zhang L, Wu YH. CDKN3 regulates cisplatin resistance to colorectal cancer through TIPE1. Eur Rev Med Pharmacol Sci 2020; 24: 3614-3623
  • 42 Xiao H, Xu D, Chen P. et al. Identification of five genes as a potential biomarker for predicting progress and prognosis in adrenocortical carcinoma. J Cancer 2018; 9: 4484
  • 43 Zhang T, Guo J, Gu J. et al. KIAA0101 is a novel transcriptional target of FoxM1 and is involved in the regulation of hepatocellular carcinoma microvascular invasion by regulating epithelial-mesenchymal transition. J Cancer 2019; 10: 3501
  • 44 Wang X, Jung YS, Jun S. et al. PAF-Wnt signaling-induced cell plasticity is required for maintenance of breast cancer cell stemness. Nat Commun 2016; 7: 10633
  • 45 Jain M, Zhang L, Patterson EE. et al. KIAA0101 is overexpressed, and promotes growth and invasion in adrenal cancer. PLoS One 2011; 6: e26866
  • 46 Li J, Dallmayer M, Kirchner T. et al. PRC1: linking cytokinesis, chromosomal instability, and cancer evolution. Trends Cancer 2018; 4: 59-73
  • 47 Xu T, Wang X, Jia X. et al. Overexpression of protein regulator of cytokinesis 1 facilitates tumor growth and indicates unfavorable prognosis of patients with colon cancer. Cancer Cell Int 2020; 20: 1-12
  • 48 Hanselmann S, Gertzmann D, Shin WJ. et al. Expression of the cytokinesis regulator PRC1 results in p53-pathway activation in A549 cells but does not directly regulate gene expression in the nucleus. Cell Cycle 2023; 22: 419-432
  • 49 Chen J, Rajasekaran M, Xia H. et al. The microtubule-associated protein PRC1 promotes early recurrence of hepatocellular carcinoma in association with the Wnt/β-catenin signalling pathway. Gut 2016; 65: 1522-1534
  • 50 Zhan P, Zhang B. Xi Gm et al. PRC1 contributes to tumorigenesis of lung adenocarcinoma in association with the Wnt/β-catenin signaling pathway. Mol Cancer 2017; 16: 1-15
  • 51 Wang X, Wang J, Lyu L. et al. Oncogenic role and potential regulatory mechanism of topoisomerase IIα in a pan-cancer analysis. Sci Rep 2022; 12: 11161
  • 52 Roca E, Berruti A, Sbiera S. et al. Topoisomerase 2α and thymidylate synthase expression in adrenocortical cancer. Endocr Relat Cancer 2017; 24: 319-327
  • 53 Liu T, Zhang H, Yi S. et al. Mutual regulation of MDM 4 and TOP 2A in cancer cell proliferation. Mol Oncol 2019; 13: 1047-1058
  • 54 Jain M, Zhang L, He M. et al. TOP2A is overexpressed in and a therapeutic target for adrenocortical carcinoma. Endocr Relat Cancer 2013; 20: 361
  • 55 Zhang F, Ye J, Guo W. et al. TYMS-TM4SF4 axis promotes the progression of colorectal cancer by EMT and upregulating stem cell marker. Am J Cancer Res 2022; 12: 1009
  • 56 Ciszewski WM, Chmielewska-Kassassir M, Wozniak LA. et al. Thymidylate synthase overexpression drives the invasive phenotype in colon Cancer cells. Biomedicines 2022; 10: 1267
  • 57 Siddiqui MA, Gollavilli PN, Ramesh V. et al. Thymidylate synthase drives the phenotypes of epithelial-to-mesenchymal transition in non-small cell lung cancer. Br J Cancer 2021; 124: 281-289
  • 58 Xing Z, Luo Z, Yang H. et al. Screening and identification of key biomarkers in adrenocortical carcinoma based on bioinformatics analysis. Oncol Lett 2019; 18: 4667-4676
  • 59 Dastsooz H, Cereda M, Donna D. et al. A comprehensive bioinformatics analysis of UBE2C in cancers. Int J Mol Sci 2019; 20: 2228
  • 60 Liu Y, Zhao R, Chi S. et al. UBE2C is upregulated by estrogen and promotes epithelial–mesenchymal transition via p53 in endometrial cancer. Mol Cancer Res 2020; 18: 204-215
  • 61 Wang Y, Huang F, Liu M. et al. UBE2C mRNA expression controlled by miR-300 and HuR determines its oncogenic role in gastric cancer. Biochem Biophys Res Commun 2021; 534: 597-603
  • 62 Jiang X, Yuan Y, Tang L. et al. Comprehensive pan-cancer analysis of the prognostic and immunological roles of the METTL3/lncRNA-SNHG1/miRNA-140-3p/UBE2C axis. Front Cell. Develop Biol 2021; 9: 765772
  • 63 D’Angelo NA, Noronha MA, Câmara MC. et al. Doxorubicin nanoformulations on therapy against cancer: An overview from the last 10 years. Biomat Adv 2022; 133: 112623
  • 64 Kushiro K, Watanabe S, Goto Y. et al. Efficacy and safety of amrubicin therapy after chemoimmunotherapy in small cell lung cancer patients. Transl Lung Cancer Res 2022; 11: 1858
  • 65 Wu J, Duan L, Zhang L. et al. Fotemustine, teniposide and dexamethasone versus high-dose methotrexate plus cytarabine in newly diagnosed primary CNS lymphoma: a randomised phase 2 trial. J Neurooncol 2018; 140: 427-434
  • 66 Kreft D, Wang Y, Rattay M. et al. Binding mechanism of anti-cancer chemotherapeutic drug mitoxantrone to DNA characterized by magnetic tweezers. J Nanobiotechnol 2018; 16: 1-7
  • 67 Mohammad Hadi L, Yaghini E, MacRobert AJ. et al. Synergy between photodynamic therapy and dactinomycin chemotherapy in 2D and 3D ovarian cancer cell cultures. Int J Mol Sci 2020; 21: 3203
  • 68 Liang B, Li N, Zhang S. et al. Idarubicin-loaded methoxy poly (ethylene glycol)-b-poly (l-lactide-co-glycolide) nanoparticles for enhancing cellular uptake and promoting antileukemia activity. Int J Nanomed 2019; 14: 543-556
  • 69 Hu W, Lv K, Teng R. et al. Pegylated liposomal doxorubicin versus epirubicin as adjuvant therapy for stage I–III breast cancer. Front Genet 2021; 12: 746114
  • 70 Zálešák F, Bon DJD, Pospíšil J. Lignans and neolignans: plant secondary metabolites as a reservoir of biologically active substances. Pharmacol Res 2019; 146: 104284
  • 71 Sader S, Wu C. Computational analysis of Amsacrine resistance in human topoisomerase II alpha mutants (R487K and E571K) using homology modeling, docking and all-atom molecular dynamics simulation in explicit solvent. J Mol Graph Model 2017; 72: 209-219
  • 72 Muti M, Muti M. Electrochemical monitoring of the interaction between anticancer drug and DNA in the presence of antioxidant. Talanta 2018; 178: 1033-1039
  • 73 Sharma P, Zargar-Shoshtari K, Sexton WJ. Valrubicin in refractory non-muscle invasive bladder cancer. Expert Rev Anticancer Ther 2015; 15: 1379-1387
  • 74 Jennifer CZ, Sara Mohamed J, Salma A. et al. Pralatrexate injection for the treatment of patients with relapsed or refractory peripheral T-cell lymphoma. Expert Rev Hematol 2020; 13: 577-583
  • 75 de Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol 2014; 741: 8-16
  • 76 Siddiqui NS, Godara A, Byrne MM. et al. Capecitabine for the treatment of pancreatic cancer. Expert Opin Pharmacother 2019; 20: 399-409
  • 77 Jiang F, Yang K, Wan X-R. et al. Reproductive outcomes after floxuridine-based regimens for gestational trophoblastic neoplasia: A retrospective cohort study in a national referral center in China. Gynecol Oncol 2020; 159: 464-469
  • 78 Chang WW, Wang BY, Chen SH. et al. miR-145-5p Targets Sp1 in non-small cell lung cancer cells and links to BMI1 induced pemetrexed resistance and epithelial–mesenchymal transition. Int J Mol Sci 2022; 23: 15352
  • 79 Hsieh MY, Chen G, Chang DC. et al. The impact of metronomic adjuvant chemotherapy in patients with advanced oral cancer. Ann Surg Oncol 2018; 25: 2091-2097
  • 80 Chun KS, Joo SH. Modulation of reactive oxygen species to overcome 5-fluorouracil resistance. Biomol Ther (Seoul) 2022; 30: 479
  • 81 Casneuf V, Borbath I, Van den Eynde M. et al. Joint Belgian recommendation on screening for DPD-deficiency in patients treated with 5-FU, capecitabine (and tegafur). Acta Clin Belg 2022; 77: 346-352
  • 82 Jia HJ, Zhou M, Vashisth MK. et al. Trifluridine induces HUVECs senescence by inhibiting mTOR-dependent autophagy. Biochem Biophys Res Commun 2022; 610: 119-126
  • 83 Hu C, Chen X, Lin X. et al. Raltitrexed regulates proliferation and apoptosis of HGC-27 cells by upregulating RSK4. BMC Pharmacol Toxicol 2022; 23: 1-9
  • 84 Kuter DJ, Rogers KA, Boxer MA. et al. Fostamatinib for the treatment of warm antibody autoimmune hemolytic anemia: Phase 2, multicenter, open-label study. Am J Hematol 2022; 97: 691-699
  • 85 Herman S, Barr P, McAuley E. et al. Fostamatinib inhibits B-cell receptor signaling, cellular activation and tumor proliferation in patients with relapsed and refractory chronic lymphocytic leukemia. Leukemia 2013; 27: 1769-1773