We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Expression patterns and a prognostic model of m6A-associated regulators in prostate adenocarcinoma

    Song Ou-Yang

    Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430030, China

    Department of Urology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832008, China

    ,
    Ji-Hong Liu

    Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430030, China

    &
    Qin-Zhang Wang

    *Author for correspondence:

    E-mail Address: wqz1969@sina.com.cn

    Department of Urology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832008, China

    Published Online:https://doi.org/10.2217/bmm-2020-0095

    Aim: To study the expression patterns and prognostic value of the m6A-associated regulators in prostate adenocarcinoma (PRAD). Materials & methods: The mRNA expression and clinical data were downloaded from ‘The Cancer Genome Atlas database’. The m6A-associated variants were downloaded from m6AVar database, and combined with 14 common m6A regulators for subsequent analysis. One-way analysis of variance, univariate Cox regression analysis and least absolute shrinkage and selection operator algorithm were successively applied to obtain the ultimate regulators and prognostic model. Finally, consensus clustering, protein–protein interaction (PPI) and enrichment analysis were performed. Result: Nine regulators were obtained. PRAD patients could be classified into two risk groups and subclasses with significant survival differences by the prognostic model and consensus clustering, respectively. Conclusion: All these nine regulators were related to prognosis in PRAD, and could be used as clinical biomarkers.

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global Cancer Statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68(6), 394–424 (2018).
    • 2. Nebbioso A, Tambaro FP, Dell'aversana C, Altucci L. Cancer epigenetics: moving forward. PLoS Genet. 14(6), e1007362 (2018).
    • 3. Pan Y, Ma P, Liu Y, Li W, Shu Y. Multiple functions of m(6)A RNA methylation in cancer. J. Hematol. Oncol. 11(1), 48 (2018).
    • 4. Wu Y, Sarkissyan M, Vadgama JV. Epigenetics in breast and prostate cancer. Methods Mol. Biol. 1238, 425–466 (2015).
    • 5. Zhao L, Duan YT, Lu P et al. Epigenetic targets and their inhibitors in cancer therapy. Curr. Top. Med. Chem. 18(28), 2395–2419 (2018).
    • 6. Morena F, Argentati C, Bazzucchi M, Emiliani C, Martino S. Above the epitranscriptome: RNA modifications and stem cell identity. Genes (Basel) 9(7), 329 (2018).
    • 7. Dai D, Wang H, Zhu L, Jin H, Wang X. N6-methyladenosine links RNA metabolism to cancer progression. Cell Death Dis. 9(2), 124 (2018).
    • 8. Batista PJ. The RNA modification N(6)-methyladenosine and its implications in human disease. Genomics Proteomics Bioinformatics 15(3), 154–163 (2017).
    • 9. Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 149(7), 1635–1646 (2012).
    • 10. Chai RC, Wu F, Wang QX et al. m(6)A RNA methylation regulators contribute to malignant progression and have clinical prognostic impact in gliomas. Aging (Albany NY) 11(4), 1204–1225 (2019). • Evaluates the relationship between m6A RNA methylation regulators and malignant progression clinical prognostic impact in gliomas.
    • 11. Lin S, Choe J, Du P, Triboulet R, Gregory RI. The m(6)A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell 62(3), 335–345 (2016).
    • 12. Zhang C, Samanta D, Lu H et al. Hypoxia induces the breast cancer stem cell phenotype by HIF-dependent and ALKBH5-mediated m(6)A-demethylation of NANOG mRNA. Proc. Natl Acad. Sci USA 113(14), E2047–2056 (2016).
    • 13. Mounir M, Lucchetta M, Silva TC et al. New functionalities in the TCGAbiolinks package for the study and integration of cancer data from GDC and GTEx. PLoS Comput. Biol. 15(3), e1006701 (2019). • This package can able to access The National Cancer Institute (NCI) Genomic Data Commons (GDC) thorough its GDC Application Programming Interface (API) to search, download and prepare relevant data for analysis in R, including the ‘The Cancer Genome Atlas’ and ‘Genotype-Tissue Expression’ database.
    • 14. Zheng Y, Nie P, Peng D et al. m6AVar: a database of functional variants involved in m6A modification. Nucleic Acids Res. 46(D1), D139–D145 (2018). • The m6AVar database is specifically designed to collect functional variants that can significantly alter the status of m6A, including the m6A-associated cancer somatic mutations.
    • 15. Friedman J, Hastie T, Tibshirani R. Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33(1), 1–22 (2010). • This package fits least absolute shrinkage and selection operator and elastic-net model paths for regression, logistic and multinomial regression using coordinate descent.
    • 16. Uhlen M, Fagerberg L, Hallstrom BM et al. Proteomics. tissue-based map of the human proteome. Science 347(6220), 1260419 (2015).
    • 17. Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics 26(12), 1572–1573 (2010). • This package provides an algorithm for determining cluster count and membership by stability evidence in unsupervised analysis.
    • 18. Szklarczyk D, Gable AL, Lyon D et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47(D1), D607–D613 (2019). • This package provides an algorithm for determining cluster count and membership by stability evidence in unsupervised analysis.
    • 19. Robinson MD, Mccarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26(1), 139–140 (2010).
    • 20. Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16(5), 284–287 (2012). • This R package implements methods to analyze and visualize functional profiles of gene and gene clusters.
    • 21. Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in gene expression regulation. Cell 169(7), 1187–1200 (2017).
    • 22. Deng X, Su R, Feng X, Wei M, Chen J. Role of N(6)-methyladenosine modification in cancer. Curr. Opin. Genet. Dev. 48, 1–7 (2018).
    • 23. Meyer KD, Jaffrey SR. The dynamic epitranscriptome: n6-methyladenosine and gene expression control. Nat. Rev. Mol. Cell Biol. 15(5), 313–326 (2014).
    • 24. Alarcón CR, Goodarzi H, Lee H, Liu X, Tavazoie S, Tavazoie SF. HNRNPA2B1 is a mediator of m(6)A-dependent nuclear RNA processing events. Cell 162(6), 1299–1308 (2015).
    • 25. Chen T, Hao Y-J, Zhang Y et al. m(6)A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. Cell Stem Cell 16(3), 289–301 (2015).
    • 26. Huarte M. The emerging role of lncRNAs in cancer. Nat. Med. 21(11), 1253–1261 (2015).
    • 27. Hayes J, Peruzzi PP, Lawler S. MicroRNAs in cancer: biomarkers, functions and therapy. Trends Mol. Med. 20(8), 460–469 (2014).
    • 28. Meng Y, Li S, Gu D et al. Genetic variants in m6A modification genes are associated with colorectal cancer risk. Carcinogenesis 41(1), 8–17 (2020). • Evaluates the relationship between genetic variants in m6A modification and predictors risk in colorectal cancer.
    • 29. Li E, Wei B, Wang X, Kang R. METTL3 enhances cell adhesion through stabilizing integrin β1 mRNA via an m6A-HuR-dependent mechanism in prostatic carcinoma. Am. J. Cancer Res. 10(3), 1012–1025 (2020).
    • 30. Yuan Y, Du Y, Wang L, Liu X. The M6A methyltransferase METTL3 promotes the development and progression of prostate carcinoma via mediating MYC methylation. J. Cancer 11(12), 3588–3595 (2020).
    • 31. Ma X-X, Cao Z-G, Zhao S-L. m6A methyltransferase METTL3 promotes the progression of prostate cancer via m6A-modified LEF1. Eur. Rev. Med. Pharmacol. Sci. 24(7), 3565–3571 (2020).
    • 32. Barros-Silva D, Lobo J, Guimarães-Teixeira C et al. VIRMA-dependent N6-methyladenosine modifications regulate the expression of long non-coding RNAs CCAT1 and CCAT2 in prostate cancer. Cancers (Basel) 12(4), 771 (2020).
    • 33. Xiong G, Li L, Chen X et al. Up-regulation of CKAP2L expression promotes lung adenocarcinoma invasion and is associated with poor prognosis. Onco Targets Ther. 12, 1171–1180 (2019).
    • 34. Wang M, Xie T, Wu Y et al. Identification of RFC5 as a novel potential prognostic biomarker in lung cancer through bioinformatics analysis. Oncol. Lett. 16(4), 4201–4210 (2018).
    • 35. Levrier C, Rockstroh A, Gabrielli B et al. Discovery of thalicthuberine as a novel antimitotic agent from nature that disrupts microtubule dynamics and induces apoptosis in prostate cancer cells. Cell Cycle 17(5), 652–668 (2018).
    • 36. Mukhtar E, Adhami VM, Sechi M, Mukhtar H. Dietary flavonoid fisetin binds to beta-tubulin and disrupts microtubule dynamics in prostate cancer cells. Cancer Lett. 367(2), 173–183 (2015).
    • 37. Ho SM, Rao R, To S, Schoch E, Tarapore P. Bisphenol A and its analogues disrupt centrosome cycle and microtubule dynamics in prostate cancer. Endocr. Relat. Cancer 24(2), 83–96 (2017).
    • 38. Bochtler T, Kartal-Kaess M, Granzow M et al. Micronucleus formation in human cancer cells is biased by chromosome size. Genes Chromosomes Cancer 58(6), 392–395 (2019).
    • 39. Zhang BN, Bueno Venegas A, Hickson ID, Chu WK. DNA replication stress and its impact on chromosome segregation and tumorigenesis. Semin. Cancer Biol. 55, 61–69 (2019).
    • 40. Pan HW, Su HH, Hsu CW, Huang GJ, Wu TT. Targeted TPX2 increases chromosome missegregation and suppresses tumor cell growth in human prostate cancer. Onco Targets Ther. 10, 3531–3543 (2017).
    • 41. Wenzel ES, Singh ATK. Cell-cycle checkpoints and aneuploidy on the path to cancer. In Vivo 32(1), 1–5 (2018).
    • 42. Jacobberger JW, Sramkoski RM, Stefan T, Woost PG. Multiparameter cell cycle analysis. Methods Mol. Biol. 1678, 203–247 (2018).
    • 43. Nam RK, Benatar T, Wallis CJD et al. MicroRNA-139 is a predictor of prostate cancer recurrence and inhibits growth and migration of prostate cancer cells through cell cycle arrest and targeting IGF1R and AXL. Prostate 79(12), 1422–1438 (2019).
    • 44. Georgescu C, Corbin JM, Thibivilliers S et al. A TMEFF2-regulated cell cycle derived gene signature is prognostic of recurrence risk in prostate cancer. BMC Cancer 19(1), 423 (2019).
    • 45. Rubicz R, Zhao S, April C et al. Expression of cell cycle-regulated genes and prostate cancer prognosis in a population-based cohort. Prostate 75(13), 1354–1362 (2015).
    • 46. Wan J, Zhang J, Zhang J. Expression of p53 and its mechanism in prostate cancer. Oncol. Lett. 16(1), 378–382 (2018).