全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

miR-149 Inhibits Non-Small-Cell Lung Cancer Cells EMT by Targeting FOXM1

DOI: 10.1155/2013/506731

Full-Text   Cite this paper   Add to My Lib

Abstract:

MicroRNAs (miRNAs) have been implied to play crucial roles for epithelial-to-mesenchymal transition (EMT) of non-small-cell lung cancer cells (NSCLC cells). Here we found that the expression of miR-149, downregulated in lung cancer, was inversely correlated with invasive capability and the EMT phenotype of NSCLC cells. miR-149 inhibited EMT in NSCLC cells. Furthermore, we demonstrated that miR-149 directly targeted Forkhead box M1 (FOXM1), and FOXM1 was involved in the EMT induced by TGF-β1 in A549 cells. Overexpression of FOXM1 restored EMT process inhibited by miR-149. Our work suggested that miR-149 might be an EMT suppressor in NSCLC cells. 1. Introduction Lung cancer was the most frequently occurring malignant cancer, and the leading death cause of lung cancer was metastasis. Tumor metastasis involved several steps including separation, migration, invasion, and formation of a new tumor nodule. Epithelial-to-mesenchymal transition (EMT) played key role in the process of cancer metastasis [1]. EMT not only changed cell morphology but also induced cells to acquire essential new functions like migration and invasion. EMT was characterized by different regulations of epithelial and mesenchymal genes. The increase of mesenchymal markers vimentin and N-cadherin and the loss of epithelial marker E-cadherin were associated with EMT [2, 3]. Transforming growth factor-β1 (TGF-β1) had been suggested to be an important inducer of EMT in various cancers [4]. MicroRNAs (miRNAs) were a class of small noncoding RNAs, ~18–25 nucleotides, which regulated gene expression posttranscriptionally via inhibiting translation or inducing target mRNA degradation. Dysregulation of miRNAs occurred in a variety of cancers, and several miRNAs acted as essential modulators for EMT in non-small-cell lung cancer (NSCLC). Previous studies reported a downregulation of miR-149 in NSCLC, and miR-149 functioned as a tumor suppressor in human glioma and gastric cancer [5–7]. However, the role of miR-149 in the EMT of NSCLC cells remained poorly understood. In the present study, we found that miR-149 was significantly downregulated and the expression of miR-149 was inversely correlated with the invasive capability and EMT phenotype of NSCLC cells. Ectopic expression of miR-149 inhibited EMT in NSCLC cells. Furthermore, we found that miR-149 directly targeted FOXM1, a potential metastasis promoter. 2. Materials and Methods 2.1. Cell Lines Four NSCLC cell lines including A549, Calu3, Calu1, and H1299 were obtained from ATCC. These cells were grown in RPMI 1640 media (Whitaker Biomedical

References

[1]  D. Gao, L. T. Vahdat, S. Wong, J. C. Chang, and V. Mittal, “Microenvironmental regulation of epithelial-mesenchymal transitions in cancer,” Cancer Research, vol. 72, no. 19, pp. 4883–4889, 2012.
[2]  M. A. Kim, H. S. Lee, H. E. Lee, J. H. Kim, H. K. Yang, and W. H. Kim, “Prognostic importance of epithelial-mesenchymal transition-related protein expression in gastric carcinoma,” Histopathology, vol. 54, no. 4, pp. 442–451, 2009.
[3]  N. S. Nagathihalli and N. B. Merchant, “Src-mediated regulation of E-cadherin and EMT in pancreatic cancer,” Frontiers in Bioscience, vol. 17, pp. 2059–2069, 2012.
[4]  J. Fuxe and M. C. Karlsson, “TGF-beta-induced epithelial-mesenchymal transition: a link between cancer and inflammation,” Seminars in Cancer Biology, vol. 22, pp. 455–461, 2012.
[5]  R. Mallick, S. Patnaik, and S. Yendamuri, “MicroRNAs and lung cancer: biology and applications in diagnosis and prognosis,” Journal of Carcinogenesis, vol. 9, article no. 8, 2010.
[6]  V. Zaninovic, T. Zamora, and K. Tajima, “Origins of T-cell leukaemia virus,” Nature, vol. 344, no. 6264, p. 299, 1990.
[7]  S. J. Pan, S. K. Zhan, B. G. Pei, Q. F. Sun, L. G. Bian, and B. M. Sun, “MicroRNA-149 inhibits proliferation and invasion of glioma cells via blockade of AKT1 signaling,” International Journal of Immunopathology and Pharmacology, vol. 25, pp. 871–881, 2012.
[8]  J. Koshiol, M. Rotunno, D. Consonni et al., “Chronic obstructive pulmonary disease and altered risk of lung cancer in a population-based case-control study,” PLoS ONE, vol. 4, no. 10, Article ID e7380, 2009.
[9]  M. Angulo, E. Lecuona, and J. I. Sznajder, “Role of MicroRNAs in lung disease,” Archivos de Bronconeumología, vol. 48, pp. 325–330, 2012.
[10]  G. P. Gupta and J. Massagué, “Cancer Metastasis: building a Framework,” Cell, vol. 127, no. 4, pp. 679–695, 2006.
[11]  P. Y. Lin, S. L. Yu, and P. C. Yang, “MicroRNA in lung cancer,” British Journal of Cancer, vol. 103, no. 8, pp. 1144–1148, 2010.
[12]  C. C. Solomides, B. J. Evans, J. M. Navenot, R. Vadigepalli, S. C. Peiper, and Z. X. Wang, “MicroRNA profiling in lung cancer reveals new molecular markers for diagnosis,” Acta Cytologica, vol. 56, pp. 645–654, 2012.
[13]  Y. K. Zhang, W. Y. Zhu, J. Y. He, et al., “miRNAs expression profiling to distinguish lung squamous-cell carcinoma from adenocarcinoma subtypes,” Journal of Cancer Research and Clinical Oncology, vol. 138, no. 10, pp. 1641–1650, 2012.
[14]  L. He, J. M. Thomson, M. T. Hemann et al., “A microRNA polycistron as a potential human oncogene,” Nature, vol. 435, no. 7043, pp. 828–833, 2005.
[15]  M. Cao, M. Seike, C. Soeno, et al., “MiR-23a regulates TGF-β-induced epithelial-mesenchymal transition by targeting E-cadherin in lung cancer cells,” International Journal of Oncology, vol. 41, no. 13, pp. 869–875, 2012.
[16]  X. Liu, L. F. Sempere, H. Ouyang et al., “MicroRNA-31 functions as an oncogenic microRNA in mouse and human lung cancer cells by repressing specific tumor suppressors,” Journal of Clinical Investigation, vol. 120, no. 4, pp. 1298–1309, 2010.
[17]  S. Brabletz and T. Brabletz, “The ZEB/miR-200 feedback loop-a motor of cellular plasticity in development and cancer?” EMBO Reports, vol. 11, no. 9, pp. 670–677, 2010.
[18]  J. Li, Y. Wang, J. Luo, et al., “miR-134 inhibits epithelial to mesenchymal transition by targeting FOXM1 in non-small cell lung cancer cells,” FEBS Letters, vol. 586, no. 20, pp. 3761–3765, 2012.
[19]  J. Zhang, Y. F. Liu, and Y. Gan, “Lack of association between miR-149 C T polymorphism and cancer susceptibility: a meta-analysis based on 4,677 cases and 4,830 controls,” Molecular Biology Reports, vol. 39, no. 9, pp. 8749–8753, 2012.
[20]  Y. Wang, X. Zheng, Z. Zhang, et al., “MicroRNA-149 inhibits proliferation and cell cycle progression through the targeting of ZBTB2 in human gastric cancer,” PLoS ONE, vol. 7, Article ID e41693, 2012.
[21]  D. Li, P. Chen, X. Y. Li, et al., “Grade-specific expression profiles of miRNAs/mRNAs and docking study in human grade I-III astrocytomas,” OMICS, vol. 15, no. 10, pp. 673–682, 2011.
[22]  L. Jin, W. L. Hu, C. C. Jiang, et al., “MicroRNA-149*, a p53-responsive microRNA, functions as an oncogenic regulator in human melanoma,” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 38, pp. 15840–15845, 2011.
[23]  Z. Luo, L. Zhang, Z. Li, et al., “An in silico analysis of dynamic changes in microRNA expression profiles in stepwise development of nasopharyngeal carcinoma,” BMC Medical Genomics, vol. 5, p. 3, 2012.
[24]  Y. Wang, L. Wen, S. H. Zhao, Z. H. Ai, J. Z. Guo, and W. C. Liu, “FoxM1 expression is significantly associated with cisplatin-based chemotherapy resistance and poor prognosis in advanced non-small cell lung cancer patients,” Lung Cancer, vol. 79, pp. 173–179, 2013.
[25]  M. K. Hui, K. W. Chan, J. M. Luk, et al., “Cytoplasmic Forkhead box M1 (FoxM1) in esophageal squamous cell carcinoma significantly correlates with pathological disease stage,” World Journal of Surgery, vol. 36, no. 1, pp. 90–97, 2012.
[26]  J. Millour, D. Constantinidou, A. V. Stavropoulou et al., “FOXM1 is a transcriptional target of ERα and has a critical role in breast cancer endocrine sensitivity and resistance,” Oncogene, vol. 29, no. 20, pp. 2983–2995, 2010.
[27]  C. Y. Koo, K. W. Muir, and E. W. Lam, “FOXM1: from cancer initiation to progression and treatment,” Biochimica et Biophysica Acta, vol. 1819, pp. 28–37, 2012.
[28]  M. Priller, J. P?schl, L. Abr?o, et al., “Expression of FoxM1 is required for the proliferation of medulloblastoma cells and indicates worse survival of patients,” Clinical Cancer Research, vol. 17, no. 21, pp. 6791–6801, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413