Abstract

BackgroundMetastasis is one of the main reasons for treatment failure in endometrial cancer. Notably, high mobility group AT-hook 2 (HMGA2) has been recognized as a driving factor of tumour metastasis. microRNAs (miRNAs) are powerful posttranscriptional regulators of HMGA2.MethodsThe binding sites of miR-302a-5p and miR-367-3p on HMGA2 mRNA were identified using bioinformatics prediction software and were validated via luciferase assay. The expression levels of miR-302a-5p and miR-367-3p were detected using quantitative real-time PCR and in situ hybridization. Western blotting and immunohistochemistry were used to detect the levels of HMGA2 and epithelial-mesenchymal transition pathway-related proteins. Co-immunoprecipitation was used to detect protein interactions. The roles of miR-302a-5p and miR-367-3p in the regulation of HMGA2 during the progression of endometrial cancer were investigated using both in vitro and in vivo assays.ResultsIn the present study, high HMGA2 expression was correlated with poor clinical outcomes in endometrial cancer. The binding sites of miRNAs on HMGA2 mRNA were identified using bioinformatics prediction software and were validated via luciferase assay. In the endometrial cancer cell lines Ishikawa and HEC-1A, the overexpression of miR-302a-5p/367-3p significantly inhibited the expression of HMGA2 mRNA. In endometrial cancer tissues, we showed that miR-302a-5p and miR-367-3p were significantly downregulated and thus inversely correlated with HMGA2. The miR-302a-5p and miR-367-3p expression levels were closely correlated with FIGO stage and lymph node metastasis. High expression of miR-302a-5p/367-3p was correlated with high survival rates in endometrial cancer. In addition, miR-302a-5p/367-3p suppressed the malignant behaviour of endometrial carcinoma cells via the inhibition of HMGA2 expression.ConclusionOur findings indicate that miR-302a-5p/367-3p-mediated expression of HMGA2 regulates the malignant behaviour of endometrial carcinoma cells, which suggests that the miR-302a-5p/367-3p-HMGA2 axis may be a predictive biomarker of endometrial cancer metastasis and patient survival and a potential therapeutic target in metastatic endometrial cancer.

Highlights

  • Metastasis is one of the main reasons for treatment failure in endometrial cancer

  • The results showed that High mobility group AT-hook 2 (HMGA2) expression was significantly associated with the clinicopathological features

  • HMGA2 is a functional target of miR-302a-5p/367-3p in the regulation of epithelial-mesenchymal transition (EMT)-associated proteins in Ishikawa and HEC-1A cell lines To further study the mechanism by which miR-302a-5p/ 367-3p and HMGA2 regulate cell migration and invasion, we examined the levels of EMT-associated proteins in Ishikawa and HEC-1A cells

Read more

Summary

Introduction

Metastasis is one of the main reasons for treatment failure in endometrial cancer. High mobility group AT-hook 2 (HMGA2) has been recognized as a driving factor of tumour metastasis. For patients with advanced metastatic or recurrent endometrial cancer, treatment failure is still high due to the loss of opportunity for surgery. The potential mechanisms in the development of endometrial cancer, especially those involved in tumour metastasis, have become the basis for the development of molecular targeted therapy for endometrial cancer, which is essential for the treatment of this type of cancer. High mobility group AT-hook 2 (HMGA2) is a chromatin-binding protein that organizes protein complexes on enhancers of various genes to regulate gene expression and cell differentiation [5]. The mechanisms by which HMGA2 regulates the biological characteristics of endometrial cancer as well as the factors involved remain undefined

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