Abstract

BackgroundEsophageal cancer (EC) is the eighth most common cause of cancer-associated mortality in humans. Recent studies have revealed the important roles of microRNAs (miRs) in mediating tumor initiation and progression. miR-216a has been found to be involved in the progression of EC, but the underlying mechanisms remain largely unknown. The aim of this study is to explore the mechanism of miR-216a and the downstream molecules in esophageal cancer.Materials and MethodsThe degree of methylation of miR-216a promoter in EC tissues and cell lines was determined with methylation specific polymerase chain reaction (MSP). The levels of miR-216a and HMGB3 in EC cells were quantified by quantitative PCR (qPCR) and Western blot (WB). EC cell lines were treated with DNA methylation inhibitor 5-aza-2’-deoxycytidine (5-AZ), miR-216a mimics, and HMGB3 siRNA to explore the effects of miR-216a and HMGB3 on the proliferation, migration, invasion, and apoptosis of cells. Dual-luciferase reporter assay was employed to verify the binding of miR-216a to the 3’UTR of HMGB2 mRNA.ResultsThe promoter of MiR-216a was hypermethylated and the expression of miR-216a was down-regulated in EC, while HMGB3 was up-regulated. Dual luciferase reporter assay confirmed the binding of miR-216a to the 3’UTR of HMGB3 mRNA. Demethylated miR-216a and miR-216a mimics elevated miR-216a expression and down-regulated HMGB3, as well as inhibited cell proliferation, migration, and invasion. Inhibiting the expression of HMGB3 played an important role in inducing apoptosis, suppressing cell expansion, and down-regulating the activity of Wnt/β-catenin pathway.ConclusionsHypermethylation in the promoter of miR-216a upregulated HMGB3 and the Wnt/β-catenin pathway, resulting in enhanced EC progression.

Highlights

  • Esophageal cancer (EC), a common cancer with high metastasis [1], is mainly classified into esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC)

  • Inhibiting the expression of High mobility group box 3 (HMGB3) played an important role in inducing apoptosis, suppressing cell expansion, and down-regulating the activity of Wnt/b-catenin pathway

  • HMGB3 plays a role in regulating cell proliferation and apoptosis, and its abnormal expression may result in malignant phenotype of breast cancer [17,18,19,20,21,22]

Read more

Summary

Introduction

Esophageal cancer (EC), a common cancer with high metastasis [1], is mainly classified into esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC). Esophageal carcinogenesis is associated with microRNA (miRNA) regulation [6,7,8,9], so understanding the expression and function of miRNAs in EC is helpful to promote the early diagnosis and treatment of EC. It has been reported to be downregulated in EC, which improves cancer cell viability and promotes the progression of EC [14, 15]. HMGB3 plays a role in regulating cell proliferation and apoptosis, and its abnormal expression may result in malignant phenotype of breast cancer [17,18,19,20,21,22]. The aim of this study is to explore the mechanism of miR-216a and the downstream molecules in esophageal cancer

Objectives
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