Cancer Res Treat.  2018 Apr;50(2):382-397. 10.4143/crt.2016.551.

Effect of Necrosis on the miRNA-mRNA Regulatory Network in CRT-MG Human Astroglioma Cells

Affiliations
  • 1Department of Physiology, Ewha Womans University School of Medicine, Seoul, Korea. yc@ewha.ac.kr
  • 2Tissue Injury Defense Research Center, Ewha Womans University School of Medicine, Seoul, Korea.
  • 3Department of Biochemistry, Ewha Womans University School of Medicine, Seoul, Korea.
  • 4Department of Internal Medicine, Ewha Womans University School of Medicine, Seoul, Korea.
  • 5Department of Pathology, Ewha Womans University School of Medicine, Seoul, Korea.

Abstract

PURPOSE
Glioblastoma multiforme (GBM) is the most common adult primary intracranial tumor. The remarkable features of GBM include central necrosis. MicroRNAs (miRNAs) have been considered as diagnostic/prognostic biomarkers for many cancers, including glioblastoma. However, the effect of necrosis on the miRNA expression profile and predicted miRNA-mRNA regulatory information remain unclear. The purpose of this study is to examine the effect of necrotic cells on the modulation of miRNA and mRNA expression profiles and miRNA-mRNA network in CRT-MG cells.
MATERIALS AND METHODS
We used human astroglioma cells, CRT-MG, treated with necrotic CRT-MG cells to examine the effect of necrosis on the modulation of miRNA and mRNA by next-generation sequencing. For preparation of necrotic cells, CRT-MG cells were frozen and thawed through cycle of liquid nitrogen-water bath. The putative miRNA-mRNA regulatory relationship was inferred through target information, using miRDB.
RESULTS
The necrotic cells induced dysregulation of 106 miRNAs and 887 mRNAs. Among them, 11 miRNAs that had a negative correlation value of p < 0.05 by the hypergeometric test were screened, and their target mRNAs were analyzed by Gene Ontology enrichment analysis. Using the Kyoto Encyclopedia of Genes and Genomes database, we also found several necrotic cell treatment-activated pathways that were modulated by relevant gene targets of differentially expressed miRNAs.
CONCLUSION
Our result demonstrated that dysregulation of miRNA and mRNA expression profiles occurs when GBM cells are exposed to necrotic cells, suggesting that several miRNAs may have the potential to be used as biomarkers for predicting GBM progression and pathogenesis.

Keyword

Glioblastoma; Necrosis; MicroRNA

MeSH Terms

Adult
Astrocytoma*
Baths
Biomarkers
Gene Ontology
Genome
Glioblastoma
Humans*
MicroRNAs
Necrosis*
RNA, Messenger
Biomarkers
MicroRNAs
RNA, Messenger

Figure

  • Fig. 1. Graphical representation of the 106 miRNAs differentially expressed between necrotic cell-treated and untreated CRT-MG cells. (A) Clustering of the 106 most differentially upregulated and downregulated miRNAs for classification between the necrotic cell–treated (NC) and untreated CRT-MG cells (control). Complete linkage hierarchical clustering was performed with the Euclidian distance measure. The NC and control cells clustered separately. The colors in the heat map represent the normalized expression values, with lower expression values being colored in shades of green and higher expression values in shades of red. (B) Counts of mature miRNAs upregulated or downregulated in NC cells. (C) Scatter plot showing the normalized miRNA volume of all conditions analyzed (y-axis) and changes in miRNAs expression between the NC and control cells. The top five most significantly dysregulated miRNAs are marked. (D) Quantitative real-time polymerase chain reaction analysis was performed glioblastoma multiforme tissues from the near-necrosis tumor area (Nec) and from non-necrosis tumor area (Tm). Data are presented as fold induction compared with non-necrosis tumor area. *p < 0.05, **p < 0.01 vs. Tm.

  • Fig. 2. Graphical representation of the 887 mRNAs differentially expressed between necrotic cell–treated (NC) and untreated CRT-MG cells. (A) Clustering of the 887 most differentially upregulated and downregulated mRNAs for classification between the NC and untreated CRT-MG cells (control). Complete linkage hierarchical clustering was performed with the Euclidian distance measure. The NC and control cells clustered separately. The colors in the heatmap represent the normalized expression values, with lower expression values being colored in shades of green and higher expression values in shades of red. (B) Counts of mRNAs upregulated or downregulated in NC cells. (C) Scatter plot showing the normalized mRNA volume of all conditions analyzed (y-axis) and changes in mRNA expression between the NC and control cells. The top five most significantly dysregulated genes are marked. (D, E) Validation of mRNA analyzed reverse transcription–polymerase chain reaction (RT-PCR) and quantitative reverse transcription–polymerase chain reaction (qRT-PCR). (D) The expression level of six upregulated mRNA (CXCL3, CCL2, IL1A, TNIP1 [transcript variant 6 and 10], and ALOX5AP) and three downregulated mRNA (CYBRD1, COL2A1, and CNR1) were validated by RT-PCR. GAPDH gene was evaluated as external control. (E) Two upregulated mRNA (ICAM1 and LCN2) and three downregulated mRNA (POSTN, PCP4, and LYPD1) were determined by qRT-PCR. Data are presented as fold induction compared with untreated necrotic cells. **p < 0.01, ***p < 0.001 vs. control.

  • Fig. 3. Pie chart representing the distribution of negatively correlated miRNA-mRNA pairs. Pie chart depicting the distribution of negatively correlated miRNA-mRNA pairs within the three color-coded groups. Eighty-eight miRNAs were found to have at least one negatively regulated miRNA-mRNA pair for significant mRNAs, and 11 miRNAs a negative correlation with p < 0.05 by the hypergeometric test. a)Upregulated miRNAs–downregulated mRNAs in necrotic cell–treated CRT-MG with p < 0.05 by the hypergeometric test, b)Downregulated miRNAs–upregulated mRNAs in necrotic cell–treated CRT-MG with p < 0.05 by the hypergeometric test, c)Negatively regulated miRNA-mRNA pairs with p ≥ 0.05 by the hypergeometric test.

  • Fig. 4. Gene Ontology (GO) enrichment analysis for negatively correlated miRNA-mRNA. The 188 genes that were upregulated and downregulated in necrotic cell–treated CRT-MG cells, with p < 0.05 by the hypergeometric test, were subjected to GO functional enrichment analysis. To study the regulatory effects of the significant negatively correlated miRNA-mRNA pairs, and to further explore the function of the predicted target genes. These mRNAs were included in the biological process, cell component, and molecular function classifications.

  • Fig. 5. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis for negatively correlated miRNA-mRNA pairs. The 188 genes that were upregulated and downregulated in necrotic cell–treated CRT-MG cells, with p < 0.05 by the hypergeometric test, were subjected to KEGG database analysis. These mRNAs were related to various biological pathways of metabolism (red), genetic information (green), environmental information (orange), cellular processes (gray), organismal system (purple), and human diseases (pink). The colors in the enrichment map represent the significant p-values of the geneset enrichment, with high significant values (p ≤ 0.001) being colored in blue shade. PI3K, phosphoinositide 3-kinase; ECM, extracellular matrix; AMPK, AMP-activated protein kinase.


Reference

References

1. Scheithauer BW. Development of the WHO classification of tumors of the central nervous system: a historical perspective. Brain Pathol. 2009; 19:551–64.
Article
2. Nakada M, Kita D, Watanabe T, Hayashi Y, Teng L, Pyko IV, et al. Aberrant signaling pathways in glioma. Cancers (Basel). 2011; 3:3242–78.
Article
3. Hammoud MA, Sawaya R, Shi W, Thall PF, Leeds NE. Prognostic significance of preoperative MRI scans in glioblastoma multiforme. J Neurooncol. 1996; 27:65–73.
Article
4. Raza SM, Lang FF, Aggarwal BB, Fuller GN, Wildrick DM, Sawaya R. Necrosis and glioblastoma: a friend or a foe? A review and a hypothesis. Neurosurgery. 2002; 51:2–12.
Article
5. Ahn SH, Park H, Ahn YH, Kim S, Cho MS, Kang JL, et al. Necrotic cells influence migration and invasion of glioblastoma via NF-kappaB/AP-1-mediated IL-8 regulation. Sci Rep. 2016; 6:24552.
Article
6. Barboriak DP, Provenzale JM. Evaluation of software for registration of contrast-enhanced brain MR images in patients with glioblastoma multiforme. AJR Am J Roentgenol. 2002; 179:245–50.
Article
7. Inui M, Martello G, Piccolo S. MicroRNA control of signal transduction. Nat Rev Mol Cell Biol. 2010; 11:252–63.
Article
8. Que T, Song Y, Liu Z, Zheng S, Long H, Li Z, et al. Decreased miRNA-637 is an unfavorable prognosis marker and promotes glioma cell growth, migration and invasion via direct targeting Akt1. Oncogene. 2015; 34:4952–63.
Article
9. Hu N, Zhang J, Cui W, Kong G, Zhang S, Yue L, et al. miR-520b regulates migration of breast cancer cells by targeting hepatitis B X-interacting protein and interleukin-8. J Biol Chem. 2011; 286:13714–22.
Article
10. LeBlanc VC, Morin P. Exploring miRNA-associated signatures with diagnostic relevance in glioblastoma multiforme and breast cancer patients. J Clin Med. 2015; 4:1612–30.
Article
11. Chiosea S, Jelezcova E, Chandran U, Acquafondata M, McHale T, Sobol RW, et al. Up-regulation of dicer, a component of the microRNA machinery, in prostate adenocarcinoma. Am J Pathol. 2006; 169:1812–20.
Article
12. Qu JQ, Yi HM, Ye X, Zhu JF, Yi H, Li LN, et al. MiRNA-203 reduces nasopharyngeal carcinoma radioresistance by targeting IL8/AKT signaling. Mol Cancer Ther. 2015; 14:2653–64.
Article
13. Jansson MD, Lund AH. MicroRNA and cancer. Mol Oncol. 2012; 6:590–610.
Article
14. Chen B, Li H, Zeng X, Yang P, Liu X, Zhao X, et al. Roles of microRNA on cancer cell metabolism. J Transl Med. 2012; 10:228.
Article
15. Di Leva G, Garofalo M, Croce CM. MicroRNAs in cancer. Annu Rev Pathol. 2014; 9:287–314.
Article
16. Dong L, Li Y, Han C, Wang X, She L, Zhang H. miRNA microarray reveals specific expression in the peripheral blood of glioblastoma patients. Int J Oncol. 2014; 45:746–56.
Article
17. Xia Z, Liu F, Zhang J, Liu L. Decreased expression of miRNA-204-5p contributes to glioma progression and promotes glioma cell growth, migration and invasion. PLoS One. 2015; 10:e0132399.
Article
18. Fabbri E, Brognara E, Montagner G, Ghimenton C, Eccher A, Cantu C, et al. Regulation of IL-8 gene expression in gliomas by microRNA miR-93. BMC Cancer. 2015; 15:661.
Article
19. Mucaj V, Lee SS, Skuli N, Giannoukos DN, Qiu B, Eisinger-Mathason TS, et al. MicroRNA-124 expression counteracts pro-survival stress responses in glioblastoma. Oncogene. 2015; 34:2204–14.
Article
20. Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-κB pathway. Int Immunol. 2000; 12:1539–46.
Article
21. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 2001; 25:402–8.
22. Harris MA, Clark J, Ireland A, Lomax J, Ashburner M, Foulger R, et al. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 2004; 32:D258–61.
23. Kanehisa M, Goto S, Kawashima S, Nakaya A. The KEGG databases at GenomeNet. Nucleic Acids Res. 2002; 30:42–6.
Article
24. Xu G, Li JY. Differential expression of PDGFRB and EGFR in microvascular proliferation in glioblastoma. Tumour Biol. 2016; 37:10577–86.
Article
25. Li P, Teng F, Gao F, Zhang M, Wu J, Zhang C. Identification of circulating microRNAs as potential biomarkers for detecting acute ischemic stroke. Cell Mol Neurobiol. 2015; 35:433–47.
Article
26. Plaisier CL, O'Brien S, Bernard B, Reynolds S, Simon Z, Toledo CM, et al. Causal mechanistic regulatory network for glioblastoma deciphered using systems genetics network analysis. Cell Syst. 2016; 3:172–86.
Article
27. Liang ML, Hsieh TH, Ng KH, Tsai YN, Tsai CF, Chao ME, et al. Downregulation of miR-137 and miR-6500-3p promotes cell proliferation in pediatric high-grade gliomas. Oncotarget. 2016; 7:19723–37.
Article
28. Munoz L, Yeung YT, Grewal T. Oncogenic Ras modulates p38 MAPK-mediated inflammatory cytokine production in glioblastoma cells. Cancer Biol Ther. 2016; 17:355–63.
Article
29. Ouchi R, Okabe S, Migita T, Nakano I, Seimiya H. Senescence from glioma stem cell differentiation promotes tumor growth. Biochem Biophys Res Commun. 2016; 470:275–81.
Article
30. Zhang B, Shi L, Lu S, Sun X, Liu Y, Li H, et al. Autocrine IL-8 promotes F-actin polymerization and mediate mesenchymal transition via ELMO1-NF-kappaB-Snail signaling in glioma. Cancer Biol Ther. 2015; 16:898–911.
31. Zhou J, Yi L, Ouyang Q, Xu L, Cui H, Xu M. Neurotensin signaling regulates stem-like traits of glioblastoma stem cells through activation of IL-8/CXCR1/STAT3 pathway. Cell Signal. 2014; 26:2896–902.
Article
32. Bertaut A, Truntzer C, Madkouri R, Kaderbhai CG, Derangere V, Vincent J, et al. Blood baseline neutrophil count predicts bevacizumab efficacy in glioblastoma. Oncotarget. 2016; 7:70948–58.
Article
33. Wang L, Qin H, Li L, Zhang Y, Tu Y, Feng F, et al. Overexpression of CCL20 and its receptor CCR6 predicts poor clinical prognosis in human gliomas. Med Oncol. 2012; 29:3491–7.
Article
34. Brandenburg S, Muller A, Turkowski K, Radev YT, Rot S, Schmidt C, et al. Resident microglia rather than peripheral macrophages promote vascularization in brain tumors and are source of alternative pro-angiogenic factors. Acta Neuropathol. 2016; 131:365–78.
Article
35. Bruyere C, Mijatovic T, Lonez C, Spiegl-Kreinecker S, Berger W, Kast RE, et al. Temozolomide-induced modification of the CXC chemokine network in experimental gliomas. Int J Oncol. 2011; 38:1453–64.
Article
36. Sielska M, Przanowski P, Wylot B, Gabrusiewicz K, Maleszewska M, Kijewska M, et al. Distinct roles of CSF family cytokines in macrophage infiltration and activation in glioma progression and injury response. J Pathol. 2013; 230:310–21.
Article
37. Knebel FH, Albuquerque RC, Massaro RR, Maria-Engler SS, Campa A. Dual effect of serum amyloid A on the invasiveness of glioma cells. Mediators Inflamm. 2013; 2013:509089.
Article
38. Bouwens TA, Trouw LA, Veerhuis R, Dirven CM, Lamfers ML, Al-Khawaja H. Complement activation in glioblastoma multiforme pathophysiology: evidence from serum levels and presence of complement activation products in tumor tissue. J Neuroimmunol. 2015; 278:271–6.
Article
39. Li M, Bolduc AR, Hoda MN, Gamble DN, Dolisca SB, Bolduc AK, et al. The indoleamine 2,3-dioxygenase pathway controls complement-dependent enhancement of chemo-radiation therapy against murine glioblastoma. J Immunother Cancer. 2014; 2:21.
Article
40. Kammerer R, Buchner A, Palluch P, Pongratz T, Oboukhovskij K, Beyer W, et al. Induction of immune mediators in glioma and prostate cancer cells by non-lethal photodynamic therapy. PLoS One. 2011; 6:e21834.
Article
41. Li Q, Chen B, Cai J, Sun Y, Wang G, Li Y, et al. Comparative analysis of matrix metalloproteinase family members reveals that MMP9 predicts survival and response to temozolomide in patients with primary glioblastoma. PLoS One. 2016; 11:e0151815.
Article
42. Joseph JV, van Roosmalen IA, Busschers E, Tomar T, Conroy S, Eggens-Meijer E, et al. Serum-induced differentiation of glioblastoma neurospheres leads to enhanced migration/invasion capacity that is associated with increased MMP9. PLoS One. 2015; 10:e0145393.
Article
43. Musumeci G, Magro G, Cardile V, Coco M, Marzagalli R, Castrogiovanni P, et al. Characterization of matrix metalloproteinase-2 and -9, ADAM-10 and N-cadherin expression in human glioblastoma multiforme. Cell Tissue Res. 2015; 362:45–60.
Article
44. Wang F, Xiao W, Sun J, Han D, Zhu Y. MiRNA-181c inhibits EGFR-signaling-dependent MMP9 activation via suppressing Akt phosphorylation in glioblastoma. Tumour Biol. 2014; 35:8653–8.
Article
45. Hwang JS, Jung EH, Kwon MY, Han IO. Glioma-secreted soluble factors stimulate microglial activation: The role of interleukin-1beta and tumor necrosis factor-alpha. J Neuroimmunol. 2016; 298:165–71.
46. Kryvdiuk IV, Minchenko DO, Hlushchak NA, Ratushna OO, Karbovskyi LL, Minchenko OH. Inhibition of IRE1 modifies effect of glucose deprivation on the expression of TNFα-related genes in U87 glioma cells. Ukr Biochem J. 2015; 87:36–51.
Article
47. Kore RA, Abraham EC. Inflammatory cytokines, interleukin-1 beta and tumor necrosis factor-alpha, upregulated in glioblastoma multiforme, raise the levels of CRYAB in exosomes secreted by U373 glioma cells. Biochem Biophys Res Commun. 2014; 453:326–31.
Article
48. Rahme GJ, Zhang Z, Young AL, Cheng C, Bivona EJ, Fiering SN, et al. PDGF engages an E2F-USP1 signaling pathway to support ID2-mediated survival of proneural glioma cells. Cancer Res. 2016; 76:2964–76.
Article
49. Bao G, Wang N, Li R, Xu G, Liu P, He B. MiR-508-5p inhibits the progression of glioma by targeting glycoprotein nonmetastatic melanoma B. Neurochem Res. 2016; 41:1684–90.
Article
50. Qu J, Rizak JD, Fan Y, Guo X, Li J, Huma T, et al. Establishment and partial characterization of a human tumor cell line, GBM-HSF, from a glioblastoma multiforme. Hum Cell. 2014; 27:129–36.
Article
51. McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer: a new therapeutic opportunity. Nat Rev Cancer. 2005; 5:505–15.
52. Totoki Y, Yoshida A, Hosoda F, Nakamura H, Hama N, Ogura K, et al. Unique mutation portraits and frequent COL2A1 gene alteration in chondrosarcoma. Genome Res. 2014; 24:1411–20.
Article
53. Bedoya F, Meneu JC, Macias MI, Moreno A, Enriquez-De-Salamanca R, Gonzalez EM, et al. Mutation in CNR1 gene and VEGF expression in esophageal cancer. Tumori. 2009; 95:68–75.
Article
54. Plaisier CL, Pan M, Baliga NS. A miRNA-regulatory network explains how dysregulated miRNAs perturb oncogenic processes across diverse cancers. Genome Res. 2012; 22:2302–14.
Article
55. Zhou X, Xu X, Wang J, Lin J, Chen W. Identifying miRNA/mRNA negative regulation pairs in colorectal cancer. Sci Rep. 2015; 5:12995.
Article
56. Thurnherr T, Mah WC, Lei Z, Jin Y, Rozen SG, Lee CG. Differentially Expressed miRNAs in hepatocellular carcinoma target genes in the genetic information processing and metabolism pathways. Sci Rep. 2016; 6:20065.
Article
57. Zhong M, Bian Z, Wu Z. miR-30a suppresses cell migration and invasion through downregulation of PIK3CD in colorectal carcinoma. Cell Physiol Biochem. 2013; 31:209–18.
Article
58. Shi Y, Chen C, Zhang X, Liu Q, Xu JL, Zhang HR, et al. Primate-specific miR-663 functions as a tumor suppressor by targeting PIK3CD and predicts the prognosis of human glioblastoma. Clin Cancer Res. 2014; 20:1803–13.
Article
59. Jones BA, Beamer M, Ahmed S. Fractalkine/CX3CL1: a potential new target for inflammatory diseases. Mol Interv. 2010; 10:263–70.
Article
60. Tardaguila M, Mira E, Garcia-Cabezas MA, Feijoo AM, Quintela-Fandino M, Azcoitia I, et al. CX3CL1 promotes breast cancer via transactivation of the EGF pathway. Cancer Res. 2013; 73:4461–73.
Article
61. Ferretti E, Pistoia V, Corcione A. Role of fractalkine/CX3CL1 and its receptor in the pathogenesis of inflammatory and malignant diseases with emphasis on B cell malignancies. Mediators Inflamm. 2014; 2014:480941.
Article
62. Marchesi F, Locatelli M, Solinas G, Erreni M, Allavena P, Mantovani A. Role of CX3CR1/CX3CL1 axis in primary and secondary involvement of the nervous system by cancer. J Neuroimmunol. 2010; 224:39–44.
Article
Full Text Links
  • CRT
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