Cargando…

Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21

Radiotherapy with or without concurrent chemotherapy is the standard treatment for nasopharyngeal carcinoma (NPC) patients, whose efficacy is limited partly by intrinsic and acquired radioresistance. DNA methyltransferase 3B (DNMT3B) has been reported to participate in tumorigenesis via DNA methylat...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Cheng, Guo, Ergang, Ming, Jun, Sun, Wei, Nie, Xin, Sun, Lu, Peng, Shan, Luo, Min, Liu, Dongbo, Zhang, Linli, Mei, Qi, Long, Guoxian, Hu, Guangyuan, Hu, Guoqing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society of Gene & Cell Therapy 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7200625/
https://www.ncbi.nlm.nih.gov/pubmed/32382655
http://dx.doi.org/10.1016/j.omto.2020.04.007
_version_ 1783529373824974848
author Wu, Cheng
Guo, Ergang
Ming, Jun
Sun, Wei
Nie, Xin
Sun, Lu
Peng, Shan
Luo, Min
Liu, Dongbo
Zhang, Linli
Mei, Qi
Long, Guoxian
Hu, Guangyuan
Hu, Guoqing
author_facet Wu, Cheng
Guo, Ergang
Ming, Jun
Sun, Wei
Nie, Xin
Sun, Lu
Peng, Shan
Luo, Min
Liu, Dongbo
Zhang, Linli
Mei, Qi
Long, Guoxian
Hu, Guangyuan
Hu, Guoqing
author_sort Wu, Cheng
collection PubMed
description Radiotherapy with or without concurrent chemotherapy is the standard treatment for nasopharyngeal carcinoma (NPC) patients, whose efficacy is limited partly by intrinsic and acquired radioresistance. DNA methyltransferase 3B (DNMT3B) has been reported to participate in tumorigenesis via DNA methylation, but its role in mediating progression and radioresistance of NPC remains unclear. Therefore, we conducted the following studies to explore the relationship between DNMT3B and NPC. Here, we found that DNMT3B was elevated in NPC tissues and predicted the poor prognosis of NPC patients. We demonstrated for the first time that ionizing radiation could induce DNMT3B, which might be one of the reasons for radioresistance. Silencing of DNMT3B inhibited migration and invasion via suppressing epithelial-mesenchymal transition (EMT) in NPC cells. Furthermore, silencing DNMT3B restored and activated p53 and p21 via DNA demethylation, which led to cell cycle arrest and apoptosis, resulting in increased radiosensitivity of NPC both in vitro and in vivo. DNMT3B functions as a novel oncogene in the radioresistance of NPC through regulating EMT, cell cycle, and apoptosis. Therefore, DNMT3B could be a potential target for NPC treatment.
format Online
Article
Text
id pubmed-7200625
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher American Society of Gene & Cell Therapy
record_format MEDLINE/PubMed
spelling pubmed-72006252020-05-07 Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21 Wu, Cheng Guo, Ergang Ming, Jun Sun, Wei Nie, Xin Sun, Lu Peng, Shan Luo, Min Liu, Dongbo Zhang, Linli Mei, Qi Long, Guoxian Hu, Guangyuan Hu, Guoqing Mol Ther Oncolytics Article Radiotherapy with or without concurrent chemotherapy is the standard treatment for nasopharyngeal carcinoma (NPC) patients, whose efficacy is limited partly by intrinsic and acquired radioresistance. DNA methyltransferase 3B (DNMT3B) has been reported to participate in tumorigenesis via DNA methylation, but its role in mediating progression and radioresistance of NPC remains unclear. Therefore, we conducted the following studies to explore the relationship between DNMT3B and NPC. Here, we found that DNMT3B was elevated in NPC tissues and predicted the poor prognosis of NPC patients. We demonstrated for the first time that ionizing radiation could induce DNMT3B, which might be one of the reasons for radioresistance. Silencing of DNMT3B inhibited migration and invasion via suppressing epithelial-mesenchymal transition (EMT) in NPC cells. Furthermore, silencing DNMT3B restored and activated p53 and p21 via DNA demethylation, which led to cell cycle arrest and apoptosis, resulting in increased radiosensitivity of NPC both in vitro and in vivo. DNMT3B functions as a novel oncogene in the radioresistance of NPC through regulating EMT, cell cycle, and apoptosis. Therefore, DNMT3B could be a potential target for NPC treatment. American Society of Gene & Cell Therapy 2020-04-21 /pmc/articles/PMC7200625/ /pubmed/32382655 http://dx.doi.org/10.1016/j.omto.2020.04.007 Text en © 2020 The Author(s) http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Article
Wu, Cheng
Guo, Ergang
Ming, Jun
Sun, Wei
Nie, Xin
Sun, Lu
Peng, Shan
Luo, Min
Liu, Dongbo
Zhang, Linli
Mei, Qi
Long, Guoxian
Hu, Guangyuan
Hu, Guoqing
Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21
title Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21
title_full Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21
title_fullStr Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21
title_full_unstemmed Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21
title_short Radiation-Induced DNMT3B Promotes Radioresistance in Nasopharyngeal Carcinoma through Methylation of p53 and p21
title_sort radiation-induced dnmt3b promotes radioresistance in nasopharyngeal carcinoma through methylation of p53 and p21
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7200625/
https://www.ncbi.nlm.nih.gov/pubmed/32382655
http://dx.doi.org/10.1016/j.omto.2020.04.007
work_keys_str_mv AT wucheng radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT guoergang radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT mingjun radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT sunwei radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT niexin radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT sunlu radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT pengshan radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT luomin radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT liudongbo radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT zhanglinli radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT meiqi radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT longguoxian radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT huguangyuan radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21
AT huguoqing radiationinduceddnmt3bpromotesradioresistanceinnasopharyngealcarcinomathroughmethylationofp53andp21