Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop

Authors: Feifei Zhang, Kaitao Li, Mingxin Pan, Weidong Li, Juan Wu, Mingyi Li, Liang Zhao, Hui Wang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

As novel biomarkers for various cancers, microRNAs negatively regulate genes expression via promoting mRNA degradation and suppressing mRNA translation. miR-589 has been reported to be deregulated in several human cancer types. However, its biological role has not been functionally characterized in gastric cancer. Here, we aim to investigate the biological effect of miR-589 on gastric cancer and to reveal the possible mechanism.

Methods

Real-time PCR was performed to evaluate the expression of miR-589 in 34 paired normal and stomach tumor specimens, as well as gastric cell lines. Functional assays, such as wound healing, transwell assays and in vivo assays, were used to detect the biological effect of miR-589 and LIFR. We determined the role of miR-589 in gastric cancer tumorigenesis in vivo using xenograft nude models. Dual-luciferase report assays and Chromatin immunoprecipitation (ChIP) assay were performed for target evaluation, and the relationships were confirmed by western blot assay.

Result

MiR-589 expression was significantly higher in tumor tissues and gastric cancer cells than those in matched normal tissues and gastric epithelial cells, respectively. Clinically, overexpression of miR-589 is associated with tumor metastasis, invasion and poor prognosis of GC patients. Gain- and loss-of function experiments showed that miR-589 promoted cell migration, metastasis and invasion in vitro and lung metastasis in vivo. Mechanistically, we found that miR-589 directly targeted LIFR to activate PI3K/AKT/c-Jun signaling. Meanwhile, c-Jun bound to the promoter region of miR-589 and activated its transcription. Thus miR-589 regulated its expression in a feedback loop that promoted cell migration, metastasis and invasion.

Conclusion

Our study identified miR-589, as an oncogene, markedly induced cell metastasis and invasion via an atypical miR-589-LIFR-PI3K/AKT-c-Jun feedback loop, which suggested miR-589 as a potential biomarker and/or therapeutic target for the gastric cancer management.
Appendix
Available only for authorised users
Literature
2.
3.
4.
go back to reference Wagner A, et al. Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. J Clin Oncol. 2006;24(18):2903–9.CrossRefPubMed Wagner A, et al. Chemotherapy in advanced gastric cancer: a systematic review and meta-analysis based on aggregate data. J Clin Oncol. 2006;24(18):2903–9.CrossRefPubMed
5.
go back to reference Mirzaei H, et al. Circulating microRNAs as potential diagnostic biomarkers and therapeutic targets in gastric Cancer: current status and future perspectives. Curr Med Chem. 2016;23(36):4135–50.CrossRefPubMed Mirzaei H, et al. Circulating microRNAs as potential diagnostic biomarkers and therapeutic targets in gastric Cancer: current status and future perspectives. Curr Med Chem. 2016;23(36):4135–50.CrossRefPubMed
6.
go back to reference Wang M, et al. Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 2014;110(5):1199–210.CrossRefPubMedPubMedCentral Wang M, et al. Deregulated microRNAs in gastric cancer tissue-derived mesenchymal stem cells: novel biomarkers and a mechanism for gastric cancer. Br J Cancer. 2014;110(5):1199–210.CrossRefPubMedPubMedCentral
7.
go back to reference Cui L, et al. Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer. 2013;119(9):1618–26.CrossRefPubMed Cui L, et al. Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer. 2013;119(9):1618–26.CrossRefPubMed
8.
go back to reference Carlomagno N, et al. Diagnostic, predictive, prognostic, and therapeutic molecular biomarkers in third millennium: a breakthrough in gastric Cancer. Biomed Res Int. 2017;2017:7869802.CrossRefPubMedPubMedCentral Carlomagno N, et al. Diagnostic, predictive, prognostic, and therapeutic molecular biomarkers in third millennium: a breakthrough in gastric Cancer. Biomed Res Int. 2017;2017:7869802.CrossRefPubMedPubMedCentral
10.
11.
go back to reference Li J, et al. MicroRNAs as novel biological targets for detection and regulation. Chem Soc Rev. 2014;43(2):506–17.CrossRefPubMed Li J, et al. MicroRNAs as novel biological targets for detection and regulation. Chem Soc Rev. 2014;43(2):506–17.CrossRefPubMed
12.
go back to reference Song J, Meltzer S. MicroRNAs in pathogenesis, diagnosis, and treatment of gastroesophageal cancers. Gastroenterology. 2012;143(1):35–47.e2.CrossRefPubMed Song J, Meltzer S. MicroRNAs in pathogenesis, diagnosis, and treatment of gastroesophageal cancers. Gastroenterology. 2012;143(1):35–47.e2.CrossRefPubMed
13.
14.
go back to reference Ma D, et al. miR-93-5p/IFNAR1 axis promotes gastric cancer metastasis through activating the STAT3 signaling pathway. Cancer Lett. 2017;408:23–32.CrossRefPubMed Ma D, et al. miR-93-5p/IFNAR1 axis promotes gastric cancer metastasis through activating the STAT3 signaling pathway. Cancer Lett. 2017;408:23–32.CrossRefPubMed
15.
go back to reference Liu C, et al. Hypermethylation of miRNA-589 promoter leads to upregulation of HDAC5 which promotes malignancy in non-small cell lung cancer. Int J Oncol. 2017;50(6):2079–90.CrossRefPubMed Liu C, et al. Hypermethylation of miRNA-589 promoter leads to upregulation of HDAC5 which promotes malignancy in non-small cell lung cancer. Int J Oncol. 2017;50(6):2079–90.CrossRefPubMed
16.
go back to reference Zhang X, et al. miR-589-5p inhibits MAP3K8 and suppresses CD90(+) cancer stem cells in hepatocellular carcinoma. J Exp Clin Cancer Res. 2016;35(1):176.CrossRefPubMedPubMedCentral Zhang X, et al. miR-589-5p inhibits MAP3K8 and suppresses CD90(+) cancer stem cells in hepatocellular carcinoma. J Exp Clin Cancer Res. 2016;35(1):176.CrossRefPubMedPubMedCentral
17.
18.
go back to reference Chen, D.H., et al., LIFR is a breast cancer metastasis suppressor upstream of the Hippo-YAP pathway and a prognostic marker. Vol. 18. 2012. Chen, D.H., et al., LIFR is a breast cancer metastasis suppressor upstream of the Hippo-YAP pathway and a prognostic marker. Vol. 18. 2012.
19.
go back to reference Hergovich A. YAP-Hippo signalling downstream of leukemia inhibitory factor receptor: implications for breast cancer. Breast Cancer Res. 2012;14(6):326.CrossRefPubMedPubMedCentral Hergovich A. YAP-Hippo signalling downstream of leukemia inhibitory factor receptor: implications for breast cancer. Breast Cancer Res. 2012;14(6):326.CrossRefPubMedPubMedCentral
20.
go back to reference Zeng H, et al. Feedback activation of leukemia inhibitory factor receptor limits response to histone deacetylase inhibitors in breast Cancer. Cancer Cell. 2016;30(3):459–73.CrossRefPubMed Zeng H, et al. Feedback activation of leukemia inhibitory factor receptor limits response to histone deacetylase inhibitors in breast Cancer. Cancer Cell. 2016;30(3):459–73.CrossRefPubMed
21.
go back to reference Blanchard F, et al. DNA methylation controls the responsiveness of hepatoma cells to leukemia inhibitory factor. Hepatology. 2003;38(6):1516–28.CrossRefPubMed Blanchard F, et al. DNA methylation controls the responsiveness of hepatoma cells to leukemia inhibitory factor. Hepatology. 2003;38(6):1516–28.CrossRefPubMed
22.
go back to reference Okamura Y, et al. Leukemia inhibitory factor receptor (LIFR) is detected as a novel suppressor gene of hepatocellular carcinoma using double-combination array. Cancer Lett. 2010;289(2):170–7.CrossRefPubMed Okamura Y, et al. Leukemia inhibitory factor receptor (LIFR) is detected as a novel suppressor gene of hepatocellular carcinoma using double-combination array. Cancer Lett. 2010;289(2):170–7.CrossRefPubMed
23.
go back to reference Zhao J, et al. A novel long noncoding RNA-LOWEG is low expressed in gastric cancer and acts as a tumor suppressor by inhibiting cell invasion. J Cancer Res Clin Oncol. 2016;142(3):601–9.CrossRefPubMed Zhao J, et al. A novel long noncoding RNA-LOWEG is low expressed in gastric cancer and acts as a tumor suppressor by inhibiting cell invasion. J Cancer Res Clin Oncol. 2016;142(3):601–9.CrossRefPubMed
24.
go back to reference Zhao H, Wang J, S. Tony To. The phosphatidylinositol 3-kinase/Akt and c-Jun N-terminal kinase signaling in cancer: alliance or contradiction? (review). Int J Oncol. 2015;47(2):429–36.CrossRefPubMed Zhao H, Wang J, S. Tony To. The phosphatidylinositol 3-kinase/Akt and c-Jun N-terminal kinase signaling in cancer: alliance or contradiction? (review). Int J Oncol. 2015;47(2):429–36.CrossRefPubMed
26.
go back to reference Wang J, et al. miR-629-3p may serve as a novel biomarker and potential therapeutic target for lung metastases of triple-negative breast cancer. Breast Cancer Res. 2017;19(1):72.CrossRefPubMedPubMedCentral Wang J, et al. miR-629-3p may serve as a novel biomarker and potential therapeutic target for lung metastases of triple-negative breast cancer. Breast Cancer Res. 2017;19(1):72.CrossRefPubMedPubMedCentral
27.
28.
29.
go back to reference Wolf J, et al. Different soluble forms of the Interleukin-6 family signal transducer gp130 fine-tune the blockade of Interleukin-6 trans-signaling. J Biol Chem. 2016;291(31):16186–96.CrossRefPubMedPubMedCentral Wolf J, et al. Different soluble forms of the Interleukin-6 family signal transducer gp130 fine-tune the blockade of Interleukin-6 trans-signaling. J Biol Chem. 2016;291(31):16186–96.CrossRefPubMedPubMedCentral
30.
go back to reference Kosfeld A, et al. Mutations in the leukemia inhibitory factor receptor (LIFR) gene and Lifr deficiency cause urinary tract malformations. Hum Mol Genet. 2017;26(9):1716–31.CrossRefPubMed Kosfeld A, et al. Mutations in the leukemia inhibitory factor receptor (LIFR) gene and Lifr deficiency cause urinary tract malformations. Hum Mol Genet. 2017;26(9):1716–31.CrossRefPubMed
31.
go back to reference Luo Q, et al. LIFR functions as a metastasis suppressor in hepatocellular carcinoma by negatively regulating phosphoinositide 3-kinase/AKT pathway. Carcinogenesis. 2015;36(10):1201–12.CrossRefPubMed Luo Q, et al. LIFR functions as a metastasis suppressor in hepatocellular carcinoma by negatively regulating phosphoinositide 3-kinase/AKT pathway. Carcinogenesis. 2015;36(10):1201–12.CrossRefPubMed
32.
go back to reference Zhen Y, et al. miR-374a-CCND1-pPI3K/AKT-c-JUN feedback loop modulated by PDCD4 suppresses cell growth, metastasis, and sensitizes nasopharyngeal carcinoma to cisplatin. Oncogene. 2017;36(2):275–85.CrossRefPubMed Zhen Y, et al. miR-374a-CCND1-pPI3K/AKT-c-JUN feedback loop modulated by PDCD4 suppresses cell growth, metastasis, and sensitizes nasopharyngeal carcinoma to cisplatin. Oncogene. 2017;36(2):275–85.CrossRefPubMed
33.
go back to reference Peng Y, et al. Direct regulation of FOXK1 by C-Jun promotes proliferation, invasion and metastasis in gastric cancer cells. Cell Death Dis. 2016;7(11):e2480.CrossRefPubMedPubMedCentral Peng Y, et al. Direct regulation of FOXK1 by C-Jun promotes proliferation, invasion and metastasis in gastric cancer cells. Cell Death Dis. 2016;7(11):e2480.CrossRefPubMedPubMedCentral
34.
go back to reference Sui H, et al. miR200c attenuates P-gp-mediated MDR and metastasis by targeting JNK2/c-Jun signaling pathway in colorectal cancer. Mol Cancer Ther. 2014;13(12):3137–51.CrossRefPubMed Sui H, et al. miR200c attenuates P-gp-mediated MDR and metastasis by targeting JNK2/c-Jun signaling pathway in colorectal cancer. Mol Cancer Ther. 2014;13(12):3137–51.CrossRefPubMed
36.
37.
go back to reference Gokulnath M, et al. Transforming growth factor-β1 regulation of ATF-3, c-Jun and JunB proteins for activation of matrix metalloproteinase-13 gene in human breast cancer cells. Int J Biol Macromol. 2017;94(Pt A):370–7.CrossRefPubMed Gokulnath M, et al. Transforming growth factor-β1 regulation of ATF-3, c-Jun and JunB proteins for activation of matrix metalloproteinase-13 gene in human breast cancer cells. Int J Biol Macromol. 2017;94(Pt A):370–7.CrossRefPubMed
38.
go back to reference Johnson G, Nakamura K. The c-Jun kinase/stress-activated pathway: regulation, function and role in human disease. Biochim Biophys Acta. 2007;1773(8):1341–8.CrossRefPubMedPubMedCentral Johnson G, Nakamura K. The c-Jun kinase/stress-activated pathway: regulation, function and role in human disease. Biochim Biophys Acta. 2007;1773(8):1341–8.CrossRefPubMedPubMedCentral
39.
go back to reference Gustems M, et al. C-Jun/c-Fos heterodimers regulate cellular genes via a newly identified class of methylated DNA sequence motifs. Nucleic Acids Res. 2014;42(5):3059–72.CrossRefPubMed Gustems M, et al. C-Jun/c-Fos heterodimers regulate cellular genes via a newly identified class of methylated DNA sequence motifs. Nucleic Acids Res. 2014;42(5):3059–72.CrossRefPubMed
40.
go back to reference Zhao M, et al. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat Commun. 2016;7:11309.CrossRefPubMedPubMedCentral Zhao M, et al. miR-3188 regulates nasopharyngeal carcinoma proliferation and chemosensitivity through a FOXO1-modulated positive feedback loop with mTOR-p-PI3K/AKT-c-JUN. Nat Commun. 2016;7:11309.CrossRefPubMedPubMedCentral
Metadata
Title
miR-589 promotes gastric cancer aggressiveness by a LIFR-PI3K/AKT-c-Jun regulatory feedback loop
Authors
Feifei Zhang
Kaitao Li
Mingxin Pan
Weidong Li
Juan Wu
Mingyi Li
Liang Zhao
Hui Wang
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0821-4

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine