Abstract

Gastric carcinoma is the third leading cause of lethal cancer worldwide. Previous studies showed that Notch1 receptor intracellular domain (N1IC), the activated form of Notch1 receptor, promotes gastric cancer progression. It has been demonstrated that a significant cross-talk interplays between Notch pathways and microRNAs (miRNAs) in controlling tumorigenesis. This study identified an intronic microRNA-151 (miR-151), which consists of two mature miRNAs, miR-151-3p and miR-151-5p, as a Notch1 receptor-induced miRNA in gastric cancer cells. Activation of Notch1 pathway enhanced expressions of miR-151 and its host gene, focal adhesion kinase (FAK), in gastric cancer cells. The levels of miR-151 in gastric cancer samples were higher than those of adjacent non-tumor samples. Activated Notch1 pathway induced CBF1-dependent FAK promoter activity. The ectopic expression of miR-151 promoted growth and progression of SC-M1 gastric cancer cells including cell viability and colony formation, migration, and invasion abilities. Activated Notch1 pathway could augment progression of gastric cancer cells through miR-151-5p and FAK. The mRNA levels of pluripotency genes, Nanog and SOX-2, tumorsphere formation ability, tumor growth, and lung metastasis of SC-M1 cells were elevated by activated Notch1 pathway through miR-151-5p. Furthermore, miR-151-5p could target 3′-untranslated region (3′-UTR) of p53 mRNA and down-regulate p53 level in SC-M1 cells. Mechanistically, Notch1/miR-151-5p axis contributed to progression of SC-M1 cells through down-regulation of p53 which in turn repressed FAK promoter activity. Taken together, these results suggest that Notch1 pathway and miR-151-5p interplay with p53 in a reciprocal regulation loop in controlling gastric carcinogenesis.

Highlights

  • Gastric carcinoma is one of the most common malignant diseases and the third leading cause of cancerrelated deaths in the world [1]

  • To identify the Notch1 receptor-induced miRNAs in gastric cancer cells, miRNA quantitative real-time PCR analyses were performed in Notch1 receptor intracellular domain (N1IC)-expressing SC-M1 (SC-M1/HA-N1IC) cells and control cells

  • Western blot analyses demonstrated that focal adhesion kinase (FAK) and pFAK Y397 levels were increased by N1IC in SC-M1/HA-N1IC, K562/HA-N1IC, and HEK293/myc-N1IC cells (Figure 1C)

Read more

Summary

Introduction

Gastric carcinoma is one of the most common malignant diseases and the third leading cause of cancerrelated deaths in the world [1]. Notch pathways play pivotal roles in tumorigenesis [2, 3]. There are four Notch receptor paralogues (Notch1-4) and five Notch ligands in mammals [2, 3]. Notch receptors are cleaved to release Notch receptor intracellular domains, the activated forms of Notch www.impactjournals.com/oncotarget receptors. Notch receptor intracellular domains are translocated into nucleus to activate expression levels of target genes via both C promoter binding factor-1 (CBF1)/ recombination signal binding protein-Jk (RBP-Jk)dependent and-independent pathways [2, 3]. Notch and Notch pathways have been shown to promote tumorigenesis [4, 5]. Notch receptor expression was associated with gastric cancer development [6] and Notch receptor promoted gastric cancer growth [7]

Methods
Results
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