Cargando…

SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA

OBJECTIVE: The dysregulation of ribosome biogenesis is associated with the progression of numerous tumors, including hepatocellular carcinoma (HCC). Small nucleolar RNAs (snoRNAs) regulate ribosome biogenesis by guiding the modification of ribosomal RNAs (rRNAs). However, the underlying mechanism of...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Zhiyong, Pang, Yanan, Jia, Yin, Qin, Qin, Wang, Rui, Li, Wei, Jing, Jie, Liu, Haidong, Liu, Shanrong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Compuscript 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8763008/
https://www.ncbi.nlm.nih.gov/pubmed/33710804
http://dx.doi.org/10.20892/j.issn.2095-3941.2020.0343
_version_ 1784633866750787584
author Liu, Zhiyong
Pang, Yanan
Jia, Yin
Qin, Qin
Wang, Rui
Li, Wei
Jing, Jie
Liu, Haidong
Liu, Shanrong
author_facet Liu, Zhiyong
Pang, Yanan
Jia, Yin
Qin, Qin
Wang, Rui
Li, Wei
Jing, Jie
Liu, Haidong
Liu, Shanrong
author_sort Liu, Zhiyong
collection PubMed
description OBJECTIVE: The dysregulation of ribosome biogenesis is associated with the progression of numerous tumors, including hepatocellular carcinoma (HCC). Small nucleolar RNAs (snoRNAs) regulate ribosome biogenesis by guiding the modification of ribosomal RNAs (rRNAs). However, the underlying mechanism of this process in HCC remains elusive. METHODS: RNA immunoprecipitation and sequencing were used to analyze RNAs targeted by ribosome proteins. The biological functions of SNORA23 were examined in HCC cells and a xenograft mouse model. To elucidate the underlying mechanisms, the 2′-O-ribose methylation level of rRNAs was evaluated by qPCR, and the key proteins in the PI3K/Akt/mTOR pathway were detected using Western blot. RESULTS: Twelve snoRNAs were found to co-exist in 4 cancer cell lines using RPS6 pull-down assays. SNORA23 was downregulated in HCC and correlated with the poor prognoses of HCC patients. SNORA23 inhibited the proliferation, migration, and invasion of HCC cells both in vitro and in vivo. We also found that SNORA23 regulated ribosome biogenesis by impairing 2′-O-ribose methylation of cytidine(4506) of 28S rRNA. Furthermore, SNORA23, which is regulated by the PI3K/Akt/mTOR signaling pathway, significantly inhibited the phosphorylation of 4E binding protein 1. SNORA23 and rapamycin blocked the PI3K/AKT/mTOR signaling pathway and impaired HCC growth in vivo. CONCLUSIONS: SNORA23 exhibited antitumor effects in HCC and together with rapamycin, provided a promising therapeutic strategy for HCC treatment.
format Online
Article
Text
id pubmed-8763008
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Compuscript
record_format MEDLINE/PubMed
spelling pubmed-87630082022-02-07 SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA Liu, Zhiyong Pang, Yanan Jia, Yin Qin, Qin Wang, Rui Li, Wei Jing, Jie Liu, Haidong Liu, Shanrong Cancer Biol Med Original Article OBJECTIVE: The dysregulation of ribosome biogenesis is associated with the progression of numerous tumors, including hepatocellular carcinoma (HCC). Small nucleolar RNAs (snoRNAs) regulate ribosome biogenesis by guiding the modification of ribosomal RNAs (rRNAs). However, the underlying mechanism of this process in HCC remains elusive. METHODS: RNA immunoprecipitation and sequencing were used to analyze RNAs targeted by ribosome proteins. The biological functions of SNORA23 were examined in HCC cells and a xenograft mouse model. To elucidate the underlying mechanisms, the 2′-O-ribose methylation level of rRNAs was evaluated by qPCR, and the key proteins in the PI3K/Akt/mTOR pathway were detected using Western blot. RESULTS: Twelve snoRNAs were found to co-exist in 4 cancer cell lines using RPS6 pull-down assays. SNORA23 was downregulated in HCC and correlated with the poor prognoses of HCC patients. SNORA23 inhibited the proliferation, migration, and invasion of HCC cells both in vitro and in vivo. We also found that SNORA23 regulated ribosome biogenesis by impairing 2′-O-ribose methylation of cytidine(4506) of 28S rRNA. Furthermore, SNORA23, which is regulated by the PI3K/Akt/mTOR signaling pathway, significantly inhibited the phosphorylation of 4E binding protein 1. SNORA23 and rapamycin blocked the PI3K/AKT/mTOR signaling pathway and impaired HCC growth in vivo. CONCLUSIONS: SNORA23 exhibited antitumor effects in HCC and together with rapamycin, provided a promising therapeutic strategy for HCC treatment. Compuscript 2022-01-15 2022-01-15 /pmc/articles/PMC8763008/ /pubmed/33710804 http://dx.doi.org/10.20892/j.issn.2095-3941.2020.0343 Text en Copyright: © 2022, Cancer Biology & Medicine https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY) 4.0 (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution and reproduction in any medium, provided the original author and source are credited.
spellingShingle Original Article
Liu, Zhiyong
Pang, Yanan
Jia, Yin
Qin, Qin
Wang, Rui
Li, Wei
Jing, Jie
Liu, Haidong
Liu, Shanrong
SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA
title SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA
title_full SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA
title_fullStr SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA
title_full_unstemmed SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA
title_short SNORA23 inhibits HCC tumorigenesis by impairing the 2′-O-ribose methylation level of 28S rRNA
title_sort snora23 inhibits hcc tumorigenesis by impairing the 2′-o-ribose methylation level of 28s rrna
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8763008/
https://www.ncbi.nlm.nih.gov/pubmed/33710804
http://dx.doi.org/10.20892/j.issn.2095-3941.2020.0343
work_keys_str_mv AT liuzhiyong snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT pangyanan snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT jiayin snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT qinqin snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT wangrui snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT liwei snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT jingjie snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT liuhaidong snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna
AT liushanrong snora23inhibitshcctumorigenesisbyimpairingthe2oribosemethylationlevelof28srrna