Role of NELF‑B in supporting epithelial‑mesenchymal transition and cell proliferation during hepatocellular carcinoma progression

Funding Sponsor

American University in Cairo

Author's Department

Biotechnology Program

Find in your Library

https://doi.org/10.3892/ol.2021.13022

Document Type

Research Article

Publication Title

Oncology Letters

Publication Date

11-1-2021

doi

10.3892/ol.2021.13022

Abstract

Negative elongation factor‑B (NELF‑B), also known as cofactor of BRCA1 (COBRA1), is one of the four subunits of the NELF complex. It interacts with BRCA1, in addition to other transcription complexes in various tissues. The NELF complex represses the transcription of several genes by stalling RNA polymerase II during the early phase of transcription elongation. The role of NELF‑B in liver cancer and hepatocellular carcinoma (HCC), the most prevalent type of liver cancer, remains to be elucidated. It has been previously demonstrated that silencing of NELF‑B inhibits the proliferation and migration of HepG2 cells. The present study aimed to investigate the consequences of ectopic expression and silencing of NELF‑B in liver cancer HepG2 and SNU449 cell lines. Functional assays were performed to examine the effects on gene and protein expression, viability, migration and invasion of cells. Overexpression of NELF‑B did not alter the proliferation and migration of HepG2 cells, or the expression of tested genes, indicating that overexpression alone may not be sufficient for altering these features in HepG2 cells. By contrast, knockdown of NELF‑B in SNU449 cells resulted in decreased cell proliferation, together with induction of apoptosis and decreased expression levels of Ki‑67 and survivin, which are markers of proliferation and inhibition of apoptosis, respectively. Additionally, silencing of NELF‑B resulted in a significant decrease in the hallmarks of epithelial‑mesenchymal transition (EMT), including cell migration and invasion, and decreased the expression levels of EMT markers, such as N‑cadherin, vimentin and β‑catenin. Decreased expression levels of forkhead box F2 transcription factor and increased mRNA levels of trefoil factor 1, a putative tumor suppressor, were also detected following the silencing of NELF‑B. The current results demonstrated that NELF‑B enhanced the manifestation of most hallmarks of cancer, including cell proliferation, migration, invasion and inhibition of apoptosis, indicating its critical role in the progression of HCC.

This document is currently not available here.

Share

COinS