Abstract

BackgroundOur previous work demonstrated that lncRNA-MALAT1 was overexpressed in recurrent colorectal cancer (CRC) and metastatic sites in post-surgical patients. However, the upstream regulatory mechanism of MALAT1 is not well-defined. Histone demethylase JMJD2C holds great potential of epigenetic regulating mechanism in tumor diseases, especially the moderating effect on the promoter activity of targeted genes associated closely with tumor development. Therefore, we herein investigated whether JMJD2C could epigeneticly regulate the promoter activity of MALAT1 and the downstream β-catenin signaling pathway, thereby affecting the metastatic abilities of CRC cells.MethodsJMJD2C expressions in human CRC samples were detected by real-time PCR and immunohistochemistry staining. Gene silencing and overexpressing efficiencies of JMJD2C were confirmed by real-time PCR and western blot. The migration of CRC cells in vitro were tested by transwell and wound healing assays. The protein expression and cellular localization of JMJD2C and β-catenin were characterized by immunofluorescence staining and western blot. The histone methylation level of MALAT1 promoter region (H3K9me3 and H3K36me3) was tested by ChIP-PCR assays. The promoter activity of MALAT1 was detected by luciferase reporter assay. The expressions of MALAT1 and the downstream β-catenin signaling pathway related genes in CRC cells were detected by real-time PCR and western blot, respectively. The nude mice tail vein metastasis model was established to observe the effect of JMJD2C on the lung metastasis of CRC cells in vivo.ResultsOur present results indicated that histone demethylase JMJD2C was overexpressed in matched CRC tumor tissues of primary and metastatic foci, and CRC patients with lower JMJD2C expression in primary tumors had better prognosis with longer OS (Overall Survival). The following biological function observation suggested that JMJD2C promoted CRC metastasis in vitro and in vivo. Further molecular mechanism investigation demonstrated that JMJD2C protein translocated into the nuclear, lowered the histone methylation level of MALAT1 promoter in the sites of H3K9me3 and H3K36me3, up-regulated the expression of MALAT1, and enhanced the β-catenin signaling pathway in CRC cells.ConclusionOur data demonstrated that JMJD2C could enhance the metastatic abilities of CRC cells in vitro and in vivo by regulating the histone methylation level of MALAT1 promoter, thereby up-regulating the expression of MALAT1 and enhancing the activity of β-catenin signaling pathway, providing that JMJD2C might be a novel therapeutic target for CRC metastasis.

Highlights

  • Our previous work demonstrated that Long non-coding RNAs (lncRNAs)-Metastasis associated in lung adenocarcinoma transcript 1 (MALAT1) was overexpressed in recurrent colorectal cancer (CRC) and metastatic sites in post-surgical patients

  • Our data demonstrated that Jumonji domain-containing protein 2C (JMJD2C) could enhance the metastatic abilities of CRC cells in vitro and in vivo by regulating the histone methylation level of MALAT1 promoter, thereby up-regulating the expression of MALAT1 and enhancing the activity of β-catenin signaling pathway, providing that JMJD2C might be a novel therapeutic target for CRC metastasis

  • JMJD2C enhanced nuclear translocation of β-catenin and activated β-catenin signaling in CRC Our previous studies have demonstrated that MALAT1 could enhance the β-catenin signaling pathway [6, 7]; we further investigated the role of JMJD2C on regulating the expressions of β-catenin signaling pathway related proteins including β-catenin, c-Myc, and Integrin β-like 1 (ITGBL1)

Read more

Summary

Introduction

Our previous work demonstrated that lncRNA-MALAT1 was overexpressed in recurrent colorectal cancer (CRC) and metastatic sites in post-surgical patients. We investigated whether JMJD2C could epigeneticly regulate the promoter activity of MALAT1 and the downstream β-catenin signaling pathway, thereby affecting the metastatic abilities of CRC cells. Metastasis associated in lung adenocarcinoma transcript 1 (MALAT1) is one of these functional lncRNAs, which is highly expressed in CRC patients. We have successfully demonstrated that MALAT1 could promote tumor growth and metastasis in CRC by binding to SFPQ (PTB-associated splicing factor) and releasing oncogene PTBP-2 (polypyrimidine tract binding protein) from the SFPQ/PTBP-2 complex [5], and increase the nuclear translocation of β-catenin from cytoplasm, thereby activating downstream genes of β-catenin signaling pathway [6, 7]. The upstream regulatory mechanism of MALAT1 is not well-elucidated

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call