NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma

Abstract The dynamic changes of RNA N6-methyladenosine (m6A) during cancer progression participate in various cellular processes. However, less is known about a possible direct connection between upstream regulator and m6A modification, and therefore affects oncogenic progression. Here, we have iden...

Full description

Bibliographic Details
Main Authors: Zhongting Mei, Zhihua Shen, Jiaying Pu, Qian Liu, Guoxin Liu, Xuting He, Yang Wang, Jinrui Yue, Shiyu Ge, Tao Li, Ye Yuan, Lei Yang
Format: Article
Language:English
Published: BMC 2024-01-01
Series:Cell Communication and Signaling
Subjects:
Online Access:https://doi.org/10.1186/s12964-023-01321-y
_version_ 1797349786580418560
author Zhongting Mei
Zhihua Shen
Jiaying Pu
Qian Liu
Guoxin Liu
Xuting He
Yang Wang
Jinrui Yue
Shiyu Ge
Tao Li
Ye Yuan
Lei Yang
author_facet Zhongting Mei
Zhihua Shen
Jiaying Pu
Qian Liu
Guoxin Liu
Xuting He
Yang Wang
Jinrui Yue
Shiyu Ge
Tao Li
Ye Yuan
Lei Yang
author_sort Zhongting Mei
collection DOAJ
description Abstract The dynamic changes of RNA N6-methyladenosine (m6A) during cancer progression participate in various cellular processes. However, less is known about a possible direct connection between upstream regulator and m6A modification, and therefore affects oncogenic progression. Here, we have identified that a key enzyme in N4-acetylcytidine (ac4C) acetylation NAT10 is highly expressed in human osteosarcoma tissues, and its knockdown enhanced m6A contents and significantly suppressed osteosarcoma cell growth, migration and invasion. Further results revealed that NAT10 silence inhibits mRNA stability and translation of m6A reader protein YTHDC1, and displayed an increase in glucose uptake, a decrease in lactate production and pyruvate content. YTHDC1 recognizes differential m6A sites on key enzymes of glycolysis phosphofructokinase (PFKM) and lactate dehydrogenase A (LDHA) mRNAs, which suppress glycolysis pathway by increasing mRNA stability of them in an m6A methylation-dependent manner. YTHDC1 partially abrogated the inhibitory effect caused by NAT10 knockdown in tumor models in vivo, lentiviral overexpression of YTHDC1 partially restored the reduced stability of YTHDC1 caused by lentiviral depleting NAT10 at the cellular level. Altogether, we found ac4C driven RNA m6A modification can positively regulate the glycolysis of cancer cells and reveals a previously unrecognized signaling axis of NAT10/ac4C-YTHDC1/m6A-LDHA/PFKM in osteosarcoma. Video Abstract
first_indexed 2024-03-08T12:35:28Z
format Article
id doaj.art-78bec7fc68504b9184354cedba69468b
institution Directory Open Access Journal
issn 1478-811X
language English
last_indexed 2024-03-08T12:35:28Z
publishDate 2024-01-01
publisher BMC
record_format Article
series Cell Communication and Signaling
spelling doaj.art-78bec7fc68504b9184354cedba69468b2024-01-21T12:28:20ZengBMCCell Communication and Signaling1478-811X2024-01-0122111410.1186/s12964-023-01321-yNAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcomaZhongting Mei0Zhihua Shen1Jiaying Pu2Qian Liu3Guoxin Liu4Xuting He5Yang Wang6Jinrui Yue7Shiyu Ge8Tao Li9Ye Yuan10Lei Yang11Department of Pharmacology, (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical UniversityDepartment of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityDepartment of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityDepartment of Pharmacology, (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical UniversityDepartment of Pharmacology, (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical UniversityDepartment of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityDepartment of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityDepartment of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityDepartment of Pharmacology, (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical UniversityDepartment of Pharmacology, (The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical UniversityDepartment of Clinical Pharmacology, College of Pharmacy, Harbin Medical UniversityDepartment of Orthopedics, The First Affiliated Hospital of Harbin Medical UniversityAbstract The dynamic changes of RNA N6-methyladenosine (m6A) during cancer progression participate in various cellular processes. However, less is known about a possible direct connection between upstream regulator and m6A modification, and therefore affects oncogenic progression. Here, we have identified that a key enzyme in N4-acetylcytidine (ac4C) acetylation NAT10 is highly expressed in human osteosarcoma tissues, and its knockdown enhanced m6A contents and significantly suppressed osteosarcoma cell growth, migration and invasion. Further results revealed that NAT10 silence inhibits mRNA stability and translation of m6A reader protein YTHDC1, and displayed an increase in glucose uptake, a decrease in lactate production and pyruvate content. YTHDC1 recognizes differential m6A sites on key enzymes of glycolysis phosphofructokinase (PFKM) and lactate dehydrogenase A (LDHA) mRNAs, which suppress glycolysis pathway by increasing mRNA stability of them in an m6A methylation-dependent manner. YTHDC1 partially abrogated the inhibitory effect caused by NAT10 knockdown in tumor models in vivo, lentiviral overexpression of YTHDC1 partially restored the reduced stability of YTHDC1 caused by lentiviral depleting NAT10 at the cellular level. Altogether, we found ac4C driven RNA m6A modification can positively regulate the glycolysis of cancer cells and reveals a previously unrecognized signaling axis of NAT10/ac4C-YTHDC1/m6A-LDHA/PFKM in osteosarcoma. Video Abstracthttps://doi.org/10.1186/s12964-023-01321-yOsteosarcomaac4C acetylationm6A methylationNAT10GlycolysisYTHDC1
spellingShingle Zhongting Mei
Zhihua Shen
Jiaying Pu
Qian Liu
Guoxin Liu
Xuting He
Yang Wang
Jinrui Yue
Shiyu Ge
Tao Li
Ye Yuan
Lei Yang
NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma
Cell Communication and Signaling
Osteosarcoma
ac4C acetylation
m6A methylation
NAT10
Glycolysis
YTHDC1
title NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma
title_full NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma
title_fullStr NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma
title_full_unstemmed NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma
title_short NAT10 mediated ac4C acetylation driven m6A modification via involvement of YTHDC1-LDHA/PFKM regulates glycolysis and promotes osteosarcoma
title_sort nat10 mediated ac4c acetylation driven m6a modification via involvement of ythdc1 ldha pfkm regulates glycolysis and promotes osteosarcoma
topic Osteosarcoma
ac4C acetylation
m6A methylation
NAT10
Glycolysis
YTHDC1
url https://doi.org/10.1186/s12964-023-01321-y
work_keys_str_mv AT zhongtingmei nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT zhihuashen nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT jiayingpu nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT qianliu nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT guoxinliu nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT xutinghe nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT yangwang nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT jinruiyue nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT shiyuge nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT taoli nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT yeyuan nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma
AT leiyang nat10mediatedac4cacetylationdrivenm6amodificationviainvolvementofythdc1ldhapfkmregulatesglycolysisandpromotesosteosarcoma