Oncotarget

Research Papers:

Transcriptional regulation of the potential tumor suppressor ABI3 gene in thyroid carcinomas: interplay between methylation and NKX2-1 availability

Lais Moraes, Ana Luiza R. Galrão, Ileana Rubió and Janete M. Cerutti _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:25960-25970. https://doi.org/10.18632/oncotarget.8416

Metrics: PDF 1532 views  |   HTML 2056 views  |   ?  


Abstract

Lais Moraes1, Ana Luiza R. Galrão1, Ileana Rubió2, Janete M. Cerutti1

1Genetic Bases of Thyroid Tumors Laboratory, Division of Genetics, Department of Morphology and Genetics, Escola Paulista de Medicina, Universidade Federal de São Paulo, SP, Brazil

2Department of Biological Sciences, Universidade Federal de São Paulo, SP, Brazil

Correspondence to:

Janete M. Cerutti, e-mail: [email protected]

Keywords: ABI3, NKX2-1, DNA methylation, follicular thyroid carcinoma, cancer-specific differentially methylated region (cDMR)

Received: August 21, 2015     Accepted: March 12, 2016     Published: March 27, 2016

ABSTRACT

We previously reported that ABI3 expression was decreased in thyroid cancer tissues and that ectopic expression of ABI3 in a follicular thyroid carcinoma cell line delayed cell cycle progression and inhibited cell proliferation, invasion, migration and tumor formation in athymic mice. These data indicated that ABI3 is a tumor suppressor gene; however the mechanism through which ABI3 is silenced in thyroid carcinomas is unknown. We here show that treatment of four follicular thyroid carcinoma cell lines with 5-aza-dC induced demethylation of a specific region of the ABI3 promoter and restored ABI3 expression. In contrast, 5-aza-dC treatment did not restore ABI3 expression in a non-thyroid cell line, suggesting a tissue-specific regulation. We additionally show that 8 CpG sites located within the ABI3 promoter are hypermethylated in most thyroid carcinoma samples and the degree of methylation correlated with ABI3 expression. Narrowing the region to specific CpG sites, the CpG4-6 sites showed the largest difference between benign and malignant lesions. In silico analysis revealed that these CpG sites flank a canonical binding site for NKX2-1, a thyroid specific transcriptional factor. Analysis of thyroid samples shows a correlation between NKX2-1 and ABI3 expression. In vitro assays demonstrate that NKX2-1 was required for ABI3 expression. Luciferase assay further confirmed the promoter activity of this region, which was increased when the cells were co-transfected with NKX2-1. Our study shows that the transcriptional silencing of ABI3 in cancer cells occurs via methylation and uncovered a previously unrecognized role for NKX2-1 in the regulation of ABI3.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 8416