scholarly journals Quantitative Proteomics Analysis Reveals Novel Insights into Mechanisms of Action of Long Noncoding RNA Hox Transcript Antisense Intergenic RNA (HOTAIR) in HeLa Cells

2015 ◽  
Vol 14 (6) ◽  
pp. 1447-1463 ◽  
Author(s):  
Peng Zheng ◽  
Qian Xiong ◽  
Ying Wu ◽  
Ying Chen ◽  
Zhuo Chen ◽  
...  
2021 ◽  
pp. 2100031
Author(s):  
Peng Zheng ◽  
Chenglinzi Liu ◽  
Yaoqin Wu ◽  
Ruifeng Xu ◽  
Ying Chen ◽  
...  

2018 ◽  
Vol 7 (1) ◽  
pp. 84-92 ◽  
Author(s):  
Murtaza Hasan ◽  
Ghazala Mustafa ◽  
Javed Iqbal ◽  
Muhammad Ashfaq ◽  
Nasir Mahmood

Here, we have investigated the comparative quantitative proteomics analysis of the molecular response of HeLa cells to biocompatible Fe2C@C nanoparticles (NPs) using 16O/18O isotopic labelling of the cell culture.


2019 ◽  
Vol 10 ◽  
Author(s):  
Yingzi Wang ◽  
Emmanuel Wirekoh Arthur ◽  
Na Liu ◽  
Xiaofang Li ◽  
Wenjing Xiang ◽  
...  

2020 ◽  
Vol 295 (17) ◽  
pp. 5626-5639 ◽  
Author(s):  
Ryoma Yoneda ◽  
Naomi Ueda ◽  
Kousuke Uranishi ◽  
Masataka Hirasaki ◽  
Riki Kurokawa

pncRNA-D is an irradiation-induced 602-nt long noncoding RNA transcribed from the promoter region of the cyclin D1 (CCND1) gene. CCND1 expression is predicted to be inhibited through an interplay between pncRNA-D and RNA-binding protein TLS/FUS. Because the pncRNA-D–TLS interaction is essential for pncRNA-D–stimulated CCND1 inhibition, here we studied the possible role of RNA modification in this interaction in HeLa cells. We found that osmotic stress induces pncRNA-D by recruiting RNA polymerase II to its promoter. pncRNA-D was highly m6A-methylated in control cells, but osmotic stress reduced the methylation and also arginine methylation of TLS in the nucleus. Knockdown of the m6A modification enzyme methyltransferase-like 3 (METTL3) prolonged the half-life of pncRNA-D, and among the known m6A recognition proteins, YTH domain-containing 1 (YTHDC1) was responsible for binding m6A of pncRNA-D. Knockdown of METTL3 or YTHDC1 also enhanced the interaction of pncRNA-D with TLS, and results from RNA pulldown assays implicated YTHDC1 in the inhibitory effect on the TLS–pncRNA-D interaction. CRISPR/Cas9-mediated deletion of candidate m6A site decreased the m6A level in pncRNA-D and altered its interaction with the RNA-binding proteins. Of note, a reduction in the m6A modification arrested the cell cycle at the G0/G1 phase, and pncRNA-D knockdown partially reversed this arrest. Moreover, pncRNA-D induction in HeLa cells significantly suppressed cell growth. Collectively, these findings suggest that m6A modification of the long noncoding RNA pncRNA-D plays a role in the regulation of CCND1 gene expression and cell cycle progression.


Sign in / Sign up

Export Citation Format

Share Document