Cargando…

The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation

BACKGROUND: Hepatocellular carcinoma (HCC) is a very common malignant tumor. Long noncoding RNAs (lncRNAs) enable discoveries of new therapeutic tumor targets. We aimed to study the role and potential regulatory mechanisms of the lncRNA KIF9-AS1 in HCC. METHODS: CCK-8, scratch assay, and flow cytome...

Descripción completa

Detalles Bibliográficos
Autores principales: Yu, Yong, Lu, Xianghong, Yan, Yang, Wang, Yonggang, Meng, Jiangyun, Tian, Shufeng, Mu, Jinsong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9584731/
https://www.ncbi.nlm.nih.gov/pubmed/36276278
http://dx.doi.org/10.1155/2022/3888798
_version_ 1784813336488050688
author Yu, Yong
Lu, Xianghong
Yan, Yang
Wang, Yonggang
Meng, Jiangyun
Tian, Shufeng
Mu, Jinsong
author_facet Yu, Yong
Lu, Xianghong
Yan, Yang
Wang, Yonggang
Meng, Jiangyun
Tian, Shufeng
Mu, Jinsong
author_sort Yu, Yong
collection PubMed
description BACKGROUND: Hepatocellular carcinoma (HCC) is a very common malignant tumor. Long noncoding RNAs (lncRNAs) enable discoveries of new therapeutic tumor targets. We aimed to study the role and potential regulatory mechanisms of the lncRNA KIF9-AS1 in HCC. METHODS: CCK-8, scratch assay, and flow cytometry were used to detect cell proliferation, migration, and apoptosis, respectively. Bax, Bcl-2, ERK, and pERK expression were measured by western blotting. StarBase predicted KIF9-AS1 expression in HCC and paracancerous tissues. RPISeq predicted the interaction score of KIF9-AS1 and DNMT1, and MethyPrimer revealed the CpG island distribution in the RAI2 promoter. MSP was performed to measure RAI2 methylation. RIP and ChIP were performed to examine lncRNA KIF9-AS1, DNMT1, and RAI2 interactions. Finally, the effect of KIF9-AS1 knockdown on HCC was verified with nude mice. RESULTS: We found that KIF9-AS1 expression was increased in HCC tissues. KIF9-AS1 knockdown inhibited the proliferation and migration, and facilitated the apoptosis of HCC cells. lncRNA KIF9-AS1-mediated RAI2 expression led to DNMT1 recruitment and regulated RAI2 DNA methylation. RAI2 overexpression inhibited the proliferation and migration and promoted the apoptosis of HCC cells. KIF9-AS1 knockdown inhibited subcutaneous tumor formation in vivo. CONCLUSION: This study shows that KIF9-AS1 accelerates HCC growth by inducing DNMT1 promotion of RAI2 DNA methylation.
format Online
Article
Text
id pubmed-9584731
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-95847312022-10-21 The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation Yu, Yong Lu, Xianghong Yan, Yang Wang, Yonggang Meng, Jiangyun Tian, Shufeng Mu, Jinsong J Oncol Research Article BACKGROUND: Hepatocellular carcinoma (HCC) is a very common malignant tumor. Long noncoding RNAs (lncRNAs) enable discoveries of new therapeutic tumor targets. We aimed to study the role and potential regulatory mechanisms of the lncRNA KIF9-AS1 in HCC. METHODS: CCK-8, scratch assay, and flow cytometry were used to detect cell proliferation, migration, and apoptosis, respectively. Bax, Bcl-2, ERK, and pERK expression were measured by western blotting. StarBase predicted KIF9-AS1 expression in HCC and paracancerous tissues. RPISeq predicted the interaction score of KIF9-AS1 and DNMT1, and MethyPrimer revealed the CpG island distribution in the RAI2 promoter. MSP was performed to measure RAI2 methylation. RIP and ChIP were performed to examine lncRNA KIF9-AS1, DNMT1, and RAI2 interactions. Finally, the effect of KIF9-AS1 knockdown on HCC was verified with nude mice. RESULTS: We found that KIF9-AS1 expression was increased in HCC tissues. KIF9-AS1 knockdown inhibited the proliferation and migration, and facilitated the apoptosis of HCC cells. lncRNA KIF9-AS1-mediated RAI2 expression led to DNMT1 recruitment and regulated RAI2 DNA methylation. RAI2 overexpression inhibited the proliferation and migration and promoted the apoptosis of HCC cells. KIF9-AS1 knockdown inhibited subcutaneous tumor formation in vivo. CONCLUSION: This study shows that KIF9-AS1 accelerates HCC growth by inducing DNMT1 promotion of RAI2 DNA methylation. Hindawi 2022-10-13 /pmc/articles/PMC9584731/ /pubmed/36276278 http://dx.doi.org/10.1155/2022/3888798 Text en Copyright © 2022 Yong Yu et al. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Yu, Yong
Lu, Xianghong
Yan, Yang
Wang, Yonggang
Meng, Jiangyun
Tian, Shufeng
Mu, Jinsong
The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation
title The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation
title_full The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation
title_fullStr The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation
title_full_unstemmed The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation
title_short The lncRNA KIF9-AS1 Accelerates Hepatocellular Carcinoma Growth by Recruiting DNMT1 to Promote RAI2 DNA Methylation
title_sort lncrna kif9-as1 accelerates hepatocellular carcinoma growth by recruiting dnmt1 to promote rai2 dna methylation
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9584731/
https://www.ncbi.nlm.nih.gov/pubmed/36276278
http://dx.doi.org/10.1155/2022/3888798
work_keys_str_mv AT yuyong thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT luxianghong thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT yanyang thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT wangyonggang thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT mengjiangyun thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT tianshufeng thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT mujinsong thelncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT yuyong lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT luxianghong lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT yanyang lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT wangyonggang lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT mengjiangyun lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT tianshufeng lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation
AT mujinsong lncrnakif9as1accelerateshepatocellularcarcinomagrowthbyrecruitingdnmt1topromoterai2dnamethylation