METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia

Abstract N6-methyladenosine (m6A) is the most prevalent reversible modification in eukaryotic mRNA, and it plays a critical role in tumor progression. The purpose of this study was to investigate the function and regulatory mechanisms of the methyltransferase METTL3 in renal cell carcinoma (RCC). ME...

Full description

Bibliographic Details
Main Authors: Yimeng Chen, Yichen He, Zhengsheng Li, Nan Zhang, Cuixing Zhou, Xiaozhou He, Dong Xue
Format: Article
Language:English
Published: Nature Publishing Group 2024-01-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-023-06411-w
_version_ 1797349625847349248
author Yimeng Chen
Yichen He
Zhengsheng Li
Nan Zhang
Cuixing Zhou
Xiaozhou He
Dong Xue
author_facet Yimeng Chen
Yichen He
Zhengsheng Li
Nan Zhang
Cuixing Zhou
Xiaozhou He
Dong Xue
author_sort Yimeng Chen
collection DOAJ
description Abstract N6-methyladenosine (m6A) is the most prevalent reversible modification in eukaryotic mRNA, and it plays a critical role in tumor progression. The purpose of this study was to investigate the function and regulatory mechanisms of the methyltransferase METTL3 in renal cell carcinoma (RCC). METTL3 expression was upregulated and predicted a poor prognosis in patients with advanced RCC. METTL3 facilitated the proliferation, migration, and invasion of RCC cells, depending on its methylase activity. METTL3 positively regulated the expression of PLOD2, and both genes were triggered under prolonged hypoxia. Mechanistically, hypoxia-induced the binding of HIF-1α to the METTL3 promoter, which enhanced its transcriptional activity. METTL3-mediated m6A modifications of PLOD2 mRNA at 3’UTR region, promoting the translation of PLOD2 protein. Furthermore, silencing METTL3 impaired RCC progression in vitro. In vivo, administration of highly potent and selective METTL3 inhibitor STM2457 showed anti-tumor effects, whereas AAV9-mediated re-transduction of PLOD2 largely abolished the above phenomenon in a subcutaneous mouse model. These findings reveal that hypoxia and HIF-driven METTL3 transcription promote RCC progression by increasing PLOD2 expression in an m6A-dependent manner, suggesting that METTL3 may serve as a novel pharmaceutical intervention for RCC.
first_indexed 2024-03-08T12:33:01Z
format Article
id doaj.art-149ecd98a0494e4d9d30c4b5fd0b7ac3
institution Directory Open Access Journal
issn 2041-4889
language English
last_indexed 2024-03-08T12:33:01Z
publishDate 2024-01-01
publisher Nature Publishing Group
record_format Article
series Cell Death and Disease
spelling doaj.art-149ecd98a0494e4d9d30c4b5fd0b7ac32024-01-21T12:37:17ZengNature Publishing GroupCell Death and Disease2041-48892024-01-0115111310.1038/s41419-023-06411-wMETTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxiaYimeng Chen0Yichen He1Zhengsheng Li2Nan Zhang3Cuixing Zhou4Xiaozhou He5Dong Xue6Department of Urology, The Third Affiliated Hospital of Soochow UniversityDepartment of Urology, The Third Affiliated Hospital of Soochow UniversityDepartment of Urology, The Third Affiliated Hospital of Soochow UniversityDepartment of Urology, The Third Affiliated Hospital of Soochow UniversityDepartment of Urology, The Third Affiliated Hospital of Soochow UniversityDepartment of Urology, The Third Affiliated Hospital of Soochow UniversityDepartment of Urology, The Third Affiliated Hospital of Soochow UniversityAbstract N6-methyladenosine (m6A) is the most prevalent reversible modification in eukaryotic mRNA, and it plays a critical role in tumor progression. The purpose of this study was to investigate the function and regulatory mechanisms of the methyltransferase METTL3 in renal cell carcinoma (RCC). METTL3 expression was upregulated and predicted a poor prognosis in patients with advanced RCC. METTL3 facilitated the proliferation, migration, and invasion of RCC cells, depending on its methylase activity. METTL3 positively regulated the expression of PLOD2, and both genes were triggered under prolonged hypoxia. Mechanistically, hypoxia-induced the binding of HIF-1α to the METTL3 promoter, which enhanced its transcriptional activity. METTL3-mediated m6A modifications of PLOD2 mRNA at 3’UTR region, promoting the translation of PLOD2 protein. Furthermore, silencing METTL3 impaired RCC progression in vitro. In vivo, administration of highly potent and selective METTL3 inhibitor STM2457 showed anti-tumor effects, whereas AAV9-mediated re-transduction of PLOD2 largely abolished the above phenomenon in a subcutaneous mouse model. These findings reveal that hypoxia and HIF-driven METTL3 transcription promote RCC progression by increasing PLOD2 expression in an m6A-dependent manner, suggesting that METTL3 may serve as a novel pharmaceutical intervention for RCC.https://doi.org/10.1038/s41419-023-06411-w
spellingShingle Yimeng Chen
Yichen He
Zhengsheng Li
Nan Zhang
Cuixing Zhou
Xiaozhou He
Dong Xue
METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia
Cell Death and Disease
title METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia
title_full METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia
title_fullStr METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia
title_full_unstemmed METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia
title_short METTL3 facilitates renal cell carcinoma progression by PLOD2 m6A-methylation under prolonged hypoxia
title_sort mettl3 facilitates renal cell carcinoma progression by plod2 m6a methylation under prolonged hypoxia
url https://doi.org/10.1038/s41419-023-06411-w
work_keys_str_mv AT yimengchen mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia
AT yichenhe mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia
AT zhengshengli mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia
AT nanzhang mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia
AT cuixingzhou mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia
AT xiaozhouhe mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia
AT dongxue mettl3facilitatesrenalcellcarcinomaprogressionbyplod2m6amethylationunderprolongedhypoxia