DOI QR코드

DOI QR Code

Pathogenesis and prognosis of primary oral squamous cell carcinoma based on microRNAs target genes: a systems biology approach

  • Taherkhani, Amir (Research Center for Molecular Medicine, Hamadan University of Medical Sciences) ;
  • Dehto, Shahab Shahmoradi (Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Hamadan University of Medical Sciences) ;
  • Jamshidi, Shokoofeh (Dental Research Center, Department of Oral and Maxillofacial Pathology, School of Dentistry, Hamadan University of Medical Sciences) ;
  • Shojaei, Setareh (Department of Oral and Maxillofacial Pathology, Faculty of Dentistry, Hamadan University of Medical Sciences)
  • Received : 2022.06.17
  • Accepted : 2022.08.30
  • Published : 2022.09.30

Abstract

Oral squamous cell carcinoma (OSCC) is the most prevalent head and neck malignancy, with frequent cervical lymph-node metastasis, leading to a poor prognosis in OSCC patients. The present study aimed to identify potential markers, including microRNAs (miRNAs) and genes, significantly involved in the etiology of early-stage OSCC. Additionally, the main OSCC's dysregulated Gene Ontology annotations and significant signaling pathways were identified. The dataset GSE45238 underwent multivariate statistical analysis in order to distinguish primary OSCC tissues from healthy oral epithelium. Differentially expressed miRNAs (DEMs) with the criteria of p-value < 0.001 and |Log2 fold change| > 1.585 were identified in the two groups, and subsequently, validated targets of DEMs were identified. A protein interaction map was constructed, hub genes were identified, significant modules within the network were illustrated, and significant pathways and biological processes associated with the clusters were demonstrated. Using the GEPI2 database, the hub genes' predictive function was assessed. Compared to the healthy controls, main OSCC had a total of 23 DEMs. In patients with head and neck squamous cell carcinoma (HNSCC), upregulation of CALM1, CYCS, THBS1, MYC, GATA6, and SPRED3 was strongly associated with a poor prognosis. In HNSCC patients, overexpression of PIK3R3, GIGYF1, and BCL2L11 was substantially correlated with a good prognosis. Besides, "proteoglycans in cancer" was the most significant pathway enriched in the primary OSCC. The present study results revealed more possible mechanisms mediating primary OSCC and may be useful in the prognosis of the patients with early-stage OSCC.

Keywords

Acknowledgement

The authors would like to appreciate the Dental Research Center, Deputy of Research and Technology, and Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan - Iran for their supports. This paper was extracted from the thesis of Shahab Shahmoradi Dehto.

References

  1. Chou CH, Chou YE, Chuang CY, Yang SF, Lin CW. Combined effect of genetic polymorphisms of AURKA and environmental factors on oral cancer development in Taiwan. PLoS One 2017;12:e0171583. https://doi.org/10.1371/journal.pone.0171583
  2. Rivera C. Essentials of oral cancer. Int J Clin Exp Pathol 2015;8:11884-11894.
  3. Reyes-Gibby CC, Anderson KO, Merriman KW, Todd KH, Shete SS, Hanna EY. Survival patterns in squamous cell carcinoma of the head and neck: pain as an independent prognostic factor for survival. J Pain 2014;15:1015-1022.
  4. Lee WH, Chen HM, Yang SF, Liang C, Peng CY, Lin FM, et al. Bacterial alterations in salivary microbiota and their association in oral cancer. Sci Rep 2017;7:16540. https://doi.org/10.1038/s41598-017-16418-x
  5. Saintigny P, Zhang L, Fan YH, El-Naggar AK, Papadimitrakopoulou VA, Feng L, et al. Gene expression profiling predicts the development of oral cancer. Cancer Prev Res (Phila) 2011;4:218-229. https://doi.org/10.1158/1940-6207.CAPR-10-0155
  6. Mehrotra R, Singh M, Kumar D, Pandey AN, Gupta RK, Sinha US. Age specific incidence rate and pathological spectrum of oral cancer in Allahabad. Indian J Med Sci 2003;57:400-404.
  7. Dave K, Ali A, Magalhaes M. Increased expression of PD-1 and PD-L1 in oral lesions progressing to oral squamous cell carcinoma: a pilot study. Sci Rep 2020;10:9705. https://doi.org/10.1038/s41598-020-66257-6
  8. Zhai J, Luo G. GATA6 induced FN1 activation promotes the proliferation, invasion and migration of oral squamous cell carcinoma cells. Mol Med Rep 2022;25:102. https://doi.org/10.3892/mmr.2022.12618
  9. Xiao M, Zhang J, Chen W, Chen W. M1-like tumor-associated macrophages activated by exosome-transferred THBS1 promote malignant migration in oral squamous cell carcinoma. J Exp Clin Cancer Res 2018;37:143. https://doi.org/10.1186/s13046-018-0815-2
  10. Wang Z, Yan J, Zou T, Gao H. MicroRNA-1294 inhibited oral squamous cell carcinoma growth by targeting c-Myc. Oncol Lett 2018;16:2243-2250.
  11. Ries LA, Eisner MP, Kosary CL, Hankey BF, Miller BA, Clegg L, et al. SEER cancer statistics review, 1975-2002. Bethesda: Natioanl Cancer Institute, 2022. Accessed 2022 Jun 10. Available from: https://seer.cancer.gov/archive/csr/1975_2002/.
  12. Torabi M, Haghani J, Hashemipour MA, Ebrahimi M. Mast cells density in hyperkeratosis, dysplastic oral mucosa and oral squamous cell carcinoma. Avicenna J Dent Res 2018;10:67-70. https://doi.org/10.34172/ajdr.2018.15
  13. Chawla JP, Iyer N, Soodan KS, Sharma A, Khurana SK, Priyadarshni P. Role of miRNA in cancer diagnosis, prognosis, therapy and regulation of its expression by Epstein-Barr virus and human papillomaviruses: with special reference to oral cancer. Oral Oncol 2015;51:731-737. https://doi.org/10.1016/j.oraloncology.2015.05.008
  14. Mattick JS, Makunin IV. Non-coding RNA. Hum Mol Genet 2006;15 Spec No 1:R17-R29. https://doi.org/10.1093/hmg/ddl046
  15. Solomon MC, Radhakrishnan RA. MicroRNA's: the vibrant performers in the oral cancer scenario. Jpn Dent Sci Rev 2020;56:85-89. https://doi.org/10.1016/j.jdsr.2020.04.001
  16. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer 2006;6:857-866. https://doi.org/10.1038/nrc1997
  17. Wienholds E, Plasterk RH. MicroRNA function in animal development. FEBS Lett 2005;579:5911-5922. https://doi.org/10.1016/j.febslet.2005.07.070
  18. Cheng G, Danquah M, Mahato RI. MicroRNAs as therapeutic targets for cancer. In: Pharmaceutical Perspectives of Cancer Therapeutics (Lu Y, Mahato RI, eds.). New York: Springer, 2009. pp. 441-444.
  19. Shiah SG, Hsiao JR, Chang WM, Chen YW, Jin YT, Wong TY, et al. Downregulated miR329 and miR410 promote the proliferation and invasion of oral squamous cell carcinoma by targeting Wnt-7b. Cancer Res 2014;74:7560-7572. https://doi.org/10.1158/0008-5472.CAN-14-0978
  20. Barrett T, Wilhite SE, Ledoux P, Evangelista C, Kim IF, Tomashevsky M, et al. NCBI GEO: archive for functional genomics data sets: update. Nucleic Acids Res 2013;41:D991-D995.
  21. R Core Team. R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing, 2013.
  22. Oskouie AA, Ahmadi MS, Taherkhani A. Identification of prognostic biomarkers in papillary thyroid cancer and developing non-invasive diagnostic models through integrated bioinformatics analysis. Microrna 2022;11:73-87. https://doi.org/10.2174/2211536611666220124115445
  23. Dweep H, Gretz N. miRWalk2.0: a comprehensive atlas of microRNA-target interactions. Nat Methods 2015;12:697. https://doi.org/10.1038/nmeth.3485
  24. Hsu SD, Lin FM, Wu WY, Liang C, Huang WC, Chan WL, et al. miRTarBase: a database curates experimentally validated microRNA-target interactions. Nucleic Acids Res 2011;39:D163-D169. https://doi.org/10.1093/nar/gkq1107
  25. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res 2021;49:D605-D612. https://doi.org/10.1093/nar/gkaa1074
  26. Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics 2011;27:431-432. https://doi.org/10.1093/bioinformatics/btq675
  27. Bayat Z, Ahmadi-Motamayel F, Parsa MS, Taherkhani A. Potential biomarkers and signaling pathways associated with the pathogenesis of primary salivary gland carcinoma: a bioinformatics study. Genomics Inform 2021;19:e42. https://doi.org/10.5808/gi.21052
  28. Reimand J, Kull M, Peterson H, Hansen J, Vilo J. g:Profiler: a webbased toolset for functional profiling of gene lists from large-scale experiments. Nucleic Acids Res 2007;35:W193-W200. https://doi.org/10.1093/nar/gkm226
  29. 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 2017;45:W98-W102. https://doi.org/10.1093/nar/gkx247
  30. Cancer Genome Atlas Research Network; Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 2013;45: 1113-1120. https://doi.org/10.1038/ng.2764
  31. Consortium GT. The Genotype-Tissue Expression (GTEx) project. Nat Genet 2013;45:580-585. https://doi.org/10.1038/ng.2653
  32. Goedhart J, Luijsterburg MS. VolcaNoseR is a web app for creating, exploring, labeling and sharing volcano plots. Sci Rep 2020;10:20560. https://doi.org/10.1038/s41598-020-76603-3
  33. Caspa Gokulan R, Devaraj H. Stem cell markers CXCR-4 and CD133 predict aggressive phenotype and their double positivity indicates poor prognosis of oral squamous cell carcinoma. Cancers (Basel) 2021;13:5895. https://doi.org/10.3390/cancers13235895
  34. Noorbakhsh F, Rezaie F, Abdolmaleki M, Ghasemi N, Abdollahi B, Nojan F, et al. Dental students' awareness about prevention, early diagnosis and referral of patients with oral cancers in Tabriz in 2018. Avicenna J Den Res 2018;10:89-94. https://doi.org/10.34172/ajdr.2018.19
  35. Bautista-Sanchez D, Arriaga-Canon C, Pedroza-Torres A, De La Rosa-Velazquez IA, Gonzalez-Barrios R, Contreras-Espinosa L, et al. The promising role of miR-21 as a cancer biomarker and its importance in RNA-based therapeutics. Mol Ther Nucleic Acids 2020;20:409-420. https://doi.org/10.1016/j.omtn.2020.03.003
  36. Nguyen P, Bar-Sela G, Sun L, Bisht KS, Cui H, Kohn E, et al. BAT3 and SET1A form a complex with CTCFL/BORIS to modulate H3K4 histone dimethylation and gene expression. Mol Cell Biol 2008;28:6720-6729. https://doi.org/10.1128/MCB.00568-08
  37. Zhang X, Gee H, Rose B, Lee CS, Clark J, Elliott M, et al. Regulation of the tumour suppressor PDCD4 by miR-499 and miR-21 in oropharyngeal cancers. BMC Cancer 2016;16:86. https://doi.org/10.1186/s12885-016-2109-4
  38. Zhu S, Si ML, Wu H, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem 2007;282: 14328-14336. https://doi.org/10.1074/jbc.M611393200
  39. Wang XS, Prensner JR, Chen G, Cao Q, Han B, Dhanasekaran SM, et al. An integrative approach to reveal driver gene fusions from paired-end sequencing data in cancer. Nat Biotechnol 2009; 27:1005-1011. https://doi.org/10.1038/nbt.1584
  40. Amirfallah A, Knutsdottir H, Arason A, Hilmarsdottir B, Johannsson OT, Agnarsson BA, et al. Hsa-miR-21-3p associates with breast cancer patient survival and targets genes in tumor suppressive pathways. PLoS One 2021;16:e0260327. https://doi.org/10.1371/journal.pone.0260327
  41. Gao Z, Liu H, Shi Y, Yin L, Zhu Y, Liu R. Identification of cancer stem cell molecular markers and effects of hsa-miR-21-3p on stemness in esophageal squamous cell carcinoma. Cancers (Basel) 2019;11:518. https://doi.org/10.3390/cancers11040518
  42. Crimi S, Falzone L, Gattuso G, Grillo CM, Candido S, Bianchi A, et al. Droplet digital PCR analysis of liquid biopsy samples unveils the diagnostic role of hsa-miR-133a-3p and hsa-miR-375-3p in oral cancer. Biology (Basel) 2020;9:379.
  43. Xu X, Chen X, Xu M, Liu X, Pan B, Qin J, et al. miR-375-3p suppresses tumorigenesis and partially reverses chemoresistance by targeting YAP1 and SP1 in colorectal cancer cells. Aging (Albany NY) 2019;11:7357-7385.
  44. Liu T, Han X, Zheng S, Liu Q, Tuerxun A, Zhang Q, et al. CALM1 promotes progression and dampens chemosensitivity to EGFR inhibitor in esophageal squamous cell carcinoma. Cancer Cell Int 2021;21:121. https://doi.org/10.1186/s12935-021-01801-6
  45. Berchtold MW, Villalobo A. The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. Biochim Biophys Acta 2014;1843:398-435. https://doi.org/10.1016/j.bbamcr.2013.10.021
  46. Vergne I, Chua J, Deretic V. Tuberculosis toxin blocking phagosome maturation inhibits a novel Ca2+/calmodulin-PI3K hVPS34 cascade. J Exp Med 2003;198:653-659. https://doi.org/10.1084/jem.20030527
  47. Chin D, Means AR. Calmodulin: a prototypical calcium sensor. Trends Cell Biol 2000;10:322-328. https://doi.org/10.1016/S0962-8924(00)01800-6
  48. Adeola HA, Smith M, Kaestner L, Blackburn JM, Zerbini LF. Novel potential serological prostate cancer biomarkers using CT100+ cancer antigen microarray platform in a multi-cultural South African cohort. Oncotarget 2016;7:13945-13964. https://doi.org/10.18632/oncotarget.7359
  49. Zhang L, Feng C, Zhou Y, Zhou Q. Dysregulated genes targeted by microRNAs and metabolic pathways in bladder cancer revealed by bioinformatics methods. Oncol Lett 2018;15:9617-9624.
  50. Zamanian Azodi M, Rezaei Tavirani M, Rezaei Tavirani M, Vafaee R, Rostami-Nejad M. Nasopharyngeal carcinoma protein interaction mapping analysis via proteomic approaches. Asian Pac J Cancer Prev 2018;19:845-851.
  51. Adrain C, Martin SJ. The mitochondrial apoptosome: a killer unleashed by the cytochrome seas. Trends Biochem Sci 2001;26: 390-397. https://doi.org/10.1016/S0968-0004(01)01844-8
  52. Barczyk K, Kreuter M, Pryjma J, Booy EP, Maddika S, Ghavami S, et al. Serum cytochrome c indicates in vivo apoptosis and can serve as a prognostic marker during cancer therapy. Int J Cancer 2005;116:167-173. https://doi.org/10.1002/ijc.21037
  53. Bayat Z, Farhadi Z, Taherkhani A. Identification of potential biomarkers associated with poor prognosis in oral squamous cell carcinoma through integrated bioinformatics analysis: a pilot study. Gene Rep 2021;24:101243. https://doi.org/10.1016/j.genrep.2021.101243
  54. Hayashido Y, Nakashima M, Urabe K, Yoshioka H, Yoshioka Y, Hamana T, et al. Role of stromal thrombospondin-1 in motility and proteolytic activity of oral squamous cell carcinoma cells. Int J Mol Med 2003;12:447-452.
  55. Alvarez AA, Axelrod JR, Whitaker RS, Isner PD, Bentley RC, Dodge RK, et al. Thrombospondin-1 expression in epithelial ovarian carcinoma: association with p53 status, tumor angiogenesis, and survival in platinum-treated patients. Gynecol Oncol 2001;82:273-278. https://doi.org/10.1006/gyno.2001.6287
  56. Tuszynski GP, Nicosia RF. Localization of thrombospondin and its cysteine-serine-valine-threonine-cysteine-glycine-specific receptor in human breast carcinoma. Lab Invest 1994;70:228-233.
  57. Patey M, Delemer B, Bellon G, Martiny L, Pluot M, Haye B. Immunohistochemical study of thrombospondin and its receptors alpha root of beta 3 and CD36 in normal thyroid and in thyroid tumours. J Clin Pathol 1999;52:895-900. https://doi.org/10.1136/jcp.52.12.895
  58. Chen CR, Kang Y, Massague J. Defective repression of c-myc in breast cancer cells: a loss at the core of the transforming growth factor beta growth arrest program. Proc Natl Acad Sci U S A 2001;98:992-999. https://doi.org/10.1073/pnas.98.3.992
  59. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316: 1039-1043. https://doi.org/10.1126/science.1141478
  60. Peng SY, Lai PL, Hsu HC. Amplification of the c-myc gene in human hepatocellular carcinoma: biologic significance. J Formos Med Assoc 1993;92:866-870.
  61. Pai R, Pai S, Lalitha R, Kumaraswamy S, Lalitha N, Johnston R, et al. Over-expression of c-Myc oncoprotein in oral squamous cell carcinoma in the South Indian population. Ecancermedicalscience 2009;3:128.
  62. Chen YJ, Lin SC, Kao T, Chang CS, Hong PS, Shieh TM, et al. Genome-wide profiling of oral squamous cell carcinoma. J Pathol 2004;204:326-332. https://doi.org/10.1002/path.1640
  63. Tremblay M, Sanchez-Ferras O, Bouchard M. GATA transcription factors in development and disease. Development 2018;145: dev164384. https://doi.org/10.1242/dev.164384
  64. Martinelli P, Carrillo-de Santa Pau E, Cox T, Sainz B Jr, Dusetti N, Greenhalf W, et al. GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut 2017;66:1665-1676. https://doi.org/10.1136/gutjnl-2015-311256
  65. Tsuji S, Kawasaki Y, Furukawa S, Taniue K, Hayashi T, Okuno M, et al. The miR-363-GATA6-Lgr5 pathway is critical for colorectal tumourigenesis. Nat Commun 2014;5:3150. https://doi.org/10.1038/ncomms4150
  66. Sulahian R, Casey F, Shen J, Qian ZR, Shin H, Ogino S, et al. An integrative analysis reveals functional targets of GATA6 transcriptional regulation in gastric cancer. Oncogene 2014;33:5637-5648. https://doi.org/10.1038/onc.2013.517
  67. Deng X, Jiang P, Chen J, Li J, Li D, He Y, et al. GATA6 promotes epithelial-mesenchymal transition and metastasis through MUC1/beta-catenin pathway in cholangiocarcinoma. Cell Death Dis 2020;11:860. https://doi.org/10.1038/s41419-020-03070-z
  68. Deng L, Liu H. MicroRNA-506 suppresses growth and metastasis of oral squamous cell carcinoma via targeting GATA6. Int J Clin Exp Med 2015;8:1862-1870.
  69. King JA, Corcoran NM, D'Abaco GM, Straffon AF, Smith CT, Poon CL, et al. Eve-3: a liver enriched suppressor of Ras/MAPK signaling. J Hepatol 2006;44:758-767. https://doi.org/10.1016/j.jhep.2005.10.031
  70. Mizoguchi M, Nutt CL, Louis DN. Mutation analysis of CBL-C and SPRED3 on 19q in human glioblastoma. Neurogenetics 2004;5:81-82. https://doi.org/10.1007/s10048-003-0164-x
  71. He Z, Gong F, Liao J, Wang Q, Su Y, Chen C, et al. Spred-3 mutation and Ras/Raf/MAPK activation confer acquired resistance to EGFR tyrosine kinase inhibitor in an EGFR mutated NSCLC cell line. Transl Cancer Res 2020;9:2542-2555. https://doi.org/10.21037/tcr.2020.03.05
  72. Dituri F, Gigante G, Scialpi R, Mancarella S, Fabregat I, Giannelli G. Proteoglycans in cancer: friends or enemies? A special focus on hepatocellular carcinoma. Cancers (Basel) 2022;14:1902. https://doi.org/10.3390/cancers14081902
  73. Espinoza-Sanchez NA, Gotte M. Role of cell surface proteoglycans in cancer immunotherapy. Semin Cancer Biol 2020;62:48-67. https://doi.org/10.1016/j.semcancer.2019.07.012
  74. Ahrens TD, Bang-Christensen SR, Jorgensen AM, Loppke C, Spliid CB, Sand NT, et al. The role of proteoglycans in cancer metastasis and circulating tumor cell analysis. Front Cell Dev Biol 2020;8:749. https://doi.org/10.3389/fcell.2020.00749
  75. Nagarajan A, Malvi P, Wajapeyee N. Heparan sulfate and heparan sulfate proteoglycans in cancer initiation and progression. Front Endocrinol (Lausanne) 2018;9:483. https://doi.org/10.3389/fendo.2018.00483
  76. Garusi E, Rossi S, Perris R. Antithetic roles of proteoglycans in cancer. Cell Mol Life Sci 2012;69:553-579. https://doi.org/10.1007/s00018-011-0816-1