Cancer Res Treat.  2017 Oct;49(4):947-959. 10.4143/crt.2016.280.

Upregulation of MicroRNA-1246 Is Associated with BRAF Inhibitor Resistance in Melanoma Cells with Mutant BRAF

Affiliations
  • 1Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, Korea. mikelee@inu.ac.kr
  • 2Genome Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.

Abstract

PURPOSE
Intrinsic and acquired resistance limit the therapeutic benefits of inhibitors of oncogenic BRAF in melanoma. To identify microRNAs (miRNAs) associated with resistance to a BRAF inhibitor, we compared miRNA expression levels in three cell lines with different BRAF inhibitor sensitivity.
MATERIALS AND METHODS
miRNA microarray analysis was conducted to compare miRNA expression levels. Real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR) was performed to confirm the expression of differentially expressed miRNAs. The cellular effects of miR-1246 were further examined by MTT assay, immunoblotting analysis, cell cycle analysis, flow cytometric assay of apoptosis, and autophagy assay.
RESULTS
The miRNA microarray analysis and qRT-PCR identified five miRNAs (miR-3617, miR-92a-1, miR-1246, miR-193b-3p, and miR-17-3p) with expression that was consistently altered in two BRAF inhibitor-resistant cell lines. Among the five miRNAs, a miR-1246 mimic significantly reduced the antiproliferative effects of the BRAF inhibitor PLX4720 in BRAF inhibitor-resistant A375P (A375P/Mdr) cells, suggesting that miR-1246 upregulation confers acquired resistance to BRAF inhibition. In particular, apoptosis was identified as a major type of cell death in miR-1246-transfected cells; however, necrosis predominated in mimic-control-transfected cells, indicating that the resistance to PLX4720 in miR-1246 mimic-transfected cells is predominantly due to a reduction in necrosis. Furthermore, we found that miR-1246 promoted G2/M arrest through autophagy as a way to escape cell death by necrosis and apoptosis in response to PLX4720. The promotion of BRAF inhibitor resistance by miR-1246 was associated with lowered levels of p-ERK.
CONCLUSION
These results suggest that miR-1246 may be a potential therapeutic target in melanoma with acquired resistance to BRAF inhibitors.

Keyword

MicroRNAs; BRAF; PLX 4720; Drug resistance; Melanoma; Microarray analysis

MeSH Terms

Apoptosis
Autophagy
Cell Cycle
Cell Death
Cell Line
Drug Resistance
Immunoblotting
Melanoma*
Microarray Analysis
MicroRNAs
Necrosis
Polymerase Chain Reaction
United Nations
Up-Regulation*
MicroRNAs

Figure

  • Fig. 1. Validation of miRNA microarray data using miRNA-specific real-time quantitative reverse-transcription polymerase chain reaction. Real-time reverse-transcription polymerase chain reaction was conducted to measure the mRNA expression levels of miR-17-3-p, miR-92a-1, miR-3617, miR-1246, and miR-193b-3p. The mean threshold cycle (Ct) was determined based on triplicate reactions. The 2−ΔΔCt method was used to calculate the fold differences in miRNA expression among the tested samples. The expression of the target genes was normalized to GAPDH expression. **p < 0.01 according to Dunnett’s t test in relative to BRAF inhibitor–sensitive A375P cells.

  • Fig. 2. MicroRNA-1246 controls the resistance to the BRAF inhibitor in melanoma cells. A375P (A), A375P/Mdr (B), and SK-MEL-2 (C) cells were transiently transfected with a miR-1246 mimic or inhibitor for 24 hours and then treated with PLX4720 in 96-well plates for 3 days. Cell growth was then evaluated using a MTT assay. The data represent the means (standard deviation) of quadruplicates from one of three representative experiments (A, C) or of three independent experiments (B). **p < 0.01 and *p < 0.05 compared with the negative control, according to Dunnett’s t test.

  • Fig. 3. Transfection with miR-1246 leads to decreased cell growth and G2/M arrest. A375P/Mdr cells were transiently transfected with miR-1246 mimic or control RNA for 24 hours, then treated with PLX4720 for 48 hours (A, B) or 72 hours (C). (A) Cell cycle progression was assessed by staining fixed cells with propidium iodide and by flow cytometric analysis (with proper preparation of the cells). The vertical and horizontal axes represent the cell number and DNA fluorescence intensity, respectively. The percentages of cells in individual cell cycle phases were quantified using the Cell Quest Pro software. The presented results are representative of at least three independent experiments. (B) Whole cell extracts were prepared at 48 hours after PLX4720 treatment. The expression levels of p21Cip1 and p27Kip1 were assessed by immunoblotting. β-Actin was assessed as a control for protein loading. The results presented are representative of two independent experiments. (C) Apoptosis was evaluated by staining with annexin V 72 hours after transfection. The flow cytometric profile shows the annexin V–FITC staining on the X-axis and the propidium iodide (PI) staining on the Y-axis. The numbers represent the percentage of cells in each quadrant. The presented results are representative of at least three independent experiments.

  • Fig. 4. Impaired autophagic flux during PLX4720-induced growth inhibition of A375P/Mdr cells. After transient transfection with a miR-1246 mimic or control RNA for 24 hours, autophagic flux was monitored in cells exposed to 20 μM PLX4720 for 48 hours. Autophagy was then detected by flow cytometric analysis of cells incubated with Cyto-ID green fluorescent probes, which detect autophagic vacuoles. (A) A representative histogram of the flow cytometric analysis is shown. Data are representative of at least two independent experiments. (B) The values are expressed as a fold increase, where the value observed in control RNA–transfected cells was set to 1.0. The upper panel shows a representative histogram of flow cytometric analysis. The lower panel shows the summarized data describing the percentage of cells positive for Cyto-ID fluorescence (n=3). CTL, control. *p < 0.05 compared with the vehicle control, as determined by Dunnett’s t test

  • Fig. 5. Effect of miR-1246 on MEK-ERK signaling. A375P/Mdr cells were transiently transfected with a miR-1246 mimic or control RNA for 24 hours. Cell lysates were then prepared after treatment with the indicated concentrations of PLX4720 for 24 hours. The phosphorylated forms of MEK and ERK were detected by immunoblotting using anti–p-MEK and anti–p-ERK antibodies. The same blots were stripped and reprobed with anti-MEK and anti-ERK antibodies to confirm similar expression levels of MEK and ERK proteins in all lanes. The numbers listed below each band indicate the phosphorylated protein/total protein ratios determined using the Image Lab software. Data are representative of at least three independent experiments.


Reference

References

1. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002; 417:949–54.
Article
2. Holderfield M, Deuker MM, McCormick F, McMahon M. Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat Rev Cancer. 2014; 14:455–67.
Article
3. Kim YK, Ahn SK, Lee M. Differential sensitivity of melanoma cell lines with differing B-Raf mutational status to the new oncogenic B-Raf kinase inhibitor UI-152. Cancer Lett. 2012; 320:215–24.
Article
4. Rizos H, Menzies AM, Pupo GM, Carlino MS, Fung C, Hyman J, et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin Cancer Res. 2014; 20:1965–77.
Article
5. Johannessen CM, Boehm JS, Kim SY, Thomas SR, Wardwell L, Johnson LA, et al. COT drives resistance to RAF inhibition through MAP kinase pathway reactivation. Nature. 2010; 468:968–72.
6. Ahn JH, Han BI, Lee M. Induction of resistance to BRAF inhibitor is associated with the inability of Spry2 to inhibit BRAF-V600E activity in BRAF mutant cells. Biomol Ther (Seoul). 2015; 23:320–6.
Article
7. Chandarlapaty S. Negative feedback and adaptive resistance to the targeted therapy of cancer. Cancer Discov. 2012; 2:311–9.
Article
8. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004; 116:281–97.
9. Ma J, Dong C, Ji C. MicroRNA and drug resistance. Cancer Gene Ther. 2010; 17:523–31.
Article
10. Pinto R, Strippoli S, De Summa S, Albano A, Azzariti A, Guida G, et al. MicroRNA expression in BRAF-mutated and wildtype metastatic melanoma and its correlation with response duration to BRAF inhibitors. Expert Opin Ther Targets. 2015; 19:1027–35.
Article
11. Stark MS, Bonazzi VF, Boyle GM, Palmer JM, Symmons J, Lanagan CM, et al. miR-514a regulates the tumour suppressor NF1 and modulates BRAFi sensitivity in melanoma. Oncotarget. 2015; 6:17753–63.
Article
12. Ahn JH, Lee M. Autophagy-dependent survival of mutant B-Raf melanoma cells selected for resistance to apoptosis induced by inhibitors against oncogenic B-Raf. Biomol Ther (Seoul). 2013; 21:114–20.
Article
13. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001; 25:402–8.
14. Oeste CL, Seco E, Patton WF, Boya P, Perez-Sala D. Interactions between autophagic and endo-lysosomal markers in endothelial cells. Histochem Cell Biol. 2013; 139:659–70.
Article
15. Jang GH, Kim NY, Lee M. Low inducible expression of p21Cip1 confers resistance to paclitaxel in BRAF mutant melanoma cells with acquired resistance to BRAF inhibitor. Mol Cell Biochem. 2015; 406:53–62.
Article
16. Ahn JH, Lee YW, Ahn SK, Lee M. Oncogenic BRAF inhibitor UAI-201 induces cell cycle arrest and autophagy in BRAF mutant glioma cells. Life Sci. 2014; 104:38–46.
Article
17. Lee JS, Lee GM. Monitoring of autophagy in Chinese hamster ovary cells using flow cytometry. Methods. 2012; 56:375–82.
Article
18. Segura MF, Greenwald HS, Hanniford D, Osman I, Hernando E. MicroRNA and cutaneous melanoma: from discovery to prognosis and therapy. Carcinogenesis. 2012; 33:1823–32.
Article
19. Mishra RR, Kneitz S, Schartl M. Comparative analysis of melanoma deregulated miRNAs in the medaka and Xiphophorus pigment cell cancer models. Comp Biochem Physiol C Toxicol Pharmacol. 2014; 163:64–76.
Article
20. Zhu J, Dong H, Zhang Q, Zhang S. Combined assays for serum carcinoembryonic antigen and microRNA-17-3p offer improved diagnostic potential for stage I/II colon cancer. Mol Clin Oncol. 2015; 3:1315–8.
Article
21. Bar M, Wyman SK, Fritz BR, Qi J, Garg KS, Parkin RK, et al. MicroRNA discovery and profiling in human embryonic stem cells by deep sequencing of small RNA libraries. Stem Cells. 2008; 26:2496–505.
Article
22. Zhang Q, Cao LY, Cheng SJ, Zhang AM, Jin XS, Li Y. p53-induced microRNA-1246 inhibits the cell growth of human hepatocellular carcinoma cells by targeting NFIB. Oncol Rep. 2015; 33:1335–41.
Article
23. Sun Z, Meng C, Wang S, Zhou N, Guan M, Bai C, et al. MicroRNA-1246 enhances migration and invasion through CADM1 in hepatocellular carcinoma. BMC Cancer. 2014; 14:616.
Article
24. Huang W, Li H, Luo R. The microRNA-1246 promotes metastasis in non-small cell lung cancer by targeting cytoplasmic polyadenylation element-binding protein 4. Diagn Pathol. 2015; 10:127.
Article
25. Chen J, Yao D, Zhao S, He C, Ding N, Li L, et al. MiR-1246 promotes SiHa cervical cancer cell proliferation, invasion, and migration through suppression of its target gene thrombospondin 2. Arch Gynecol Obstet. 2014; 290:725–32.
Article
26. Hasegawa S, Eguchi H, Nagano H, Konno M, Tomimaru Y, Wada H, et al. MicroRNA-1246 expression associated with CCNG2-mediated chemoresistance and stemness in pancreatic cancer. Br J Cancer. 2014; 111:1572–80.
Article
27. Zhang Y, Liao JM, Zeng SX, Lu H. p53 downregulates Down syndrome-associated DYRK1A through miR-1246. EMBO Rep. 2011; 12:811–7.
Article
28. Eisenberg-Lerner A, Bialik S, Simon HU, Kimchi A. Life and death partners: apoptosis, autophagy and the cross-talk between them. Cell Death Differ. 2009; 16:966–75.
Article
29. Kawabe T. G2 checkpoint abrogators as anticancer drugs. Mol Cancer Ther. 2004; 3:513–9.
30. Roskoski R Jr. MEK1/2 dual-specificity protein kinases: structure and regulation. Biochem Biophys Res Commun. 2012; 417:5–10.
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