Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway

Recent evidence suggested that N6-methyladenosine (m<sup>6</sup>A) methylation can determine m<sup>6</sup>A-modified mRNA fate and play an important role in skeletal muscle development. It was well known that transforming growth factor beta 1 (TGFβ1) is involved in a variety...

Full description

Bibliographic Details
Main Authors: Kaiping Deng, Zhipeng Liu, Xiaodan Li, Zhen Zhang, Yixuan Fan, Qunhao Huang, Yanli Zhang, Feng Wang
Format: Article
Language:English
Published: MDPI AG 2023-03-01
Series:Cells
Subjects:
Online Access:https://www.mdpi.com/2073-4409/12/7/1005
_version_ 1797608185826115584
author Kaiping Deng
Zhipeng Liu
Xiaodan Li
Zhen Zhang
Yixuan Fan
Qunhao Huang
Yanli Zhang
Feng Wang
author_facet Kaiping Deng
Zhipeng Liu
Xiaodan Li
Zhen Zhang
Yixuan Fan
Qunhao Huang
Yanli Zhang
Feng Wang
author_sort Kaiping Deng
collection DOAJ
description Recent evidence suggested that N6-methyladenosine (m<sup>6</sup>A) methylation can determine m<sup>6</sup>A-modified mRNA fate and play an important role in skeletal muscle development. It was well known that transforming growth factor beta 1 (TGFβ1) is involved in a variety of cellular processes, such as proliferation, differentiation, and apoptosis. However, little is known about the m<sup>6</sup>A-mediated TGFβ1 regulation in myogenesis. Here, we observed an increase in endogenous TGFβ1 expression and activity during myotube differentiation. However, the knockdown of TGFβ1 inhibits the proliferation and induces cell apoptosis of myoblast. Moreover, we found that m<sup>6</sup>A in 5′-untranslated regions (5′UTR) of TGFβ1 promote its decay and inhibit its expression, leading to the blockage of the TGFβ1/SMAD2 signaling pathway. Furthermore, the targeted specific demethylation of TGFβ1 m<sup>6</sup>A using dCas13b-FTO significantly increased the TGFβ1-mediated activity of the SMAD2 signaling pathway, promoting myoblast proliferation. These findings suggest that TGFβ1 is an essential regulator of myoblast growth that is negatively regulated by m<sup>6</sup>A. Overall, these results highlight the critical role of m<sup>6</sup>A-mediated post-transcriptional regulation in myogenesis.
first_indexed 2024-03-11T05:40:48Z
format Article
id doaj.art-703ddba947564d0da2e646fd4167a647
institution Directory Open Access Journal
issn 2073-4409
language English
last_indexed 2024-03-11T05:40:48Z
publishDate 2023-03-01
publisher MDPI AG
record_format Article
series Cells
spelling doaj.art-703ddba947564d0da2e646fd4167a6472023-11-17T16:27:56ZengMDPI AGCells2073-44092023-03-01127100510.3390/cells12071005Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling PathwayKaiping Deng0Zhipeng Liu1Xiaodan Li2Zhen Zhang3Yixuan Fan4Qunhao Huang5Yanli Zhang6Feng Wang7Institute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaInstitute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaInstitute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaInstitute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaInstitute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaAnimal Husbandry and Veterinary Station of Haimen District, Nantong 226100, ChinaInstitute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaInstitute of Sheep and Goat Science, Nanjing Agricultural University, Nanjing 210095, ChinaRecent evidence suggested that N6-methyladenosine (m<sup>6</sup>A) methylation can determine m<sup>6</sup>A-modified mRNA fate and play an important role in skeletal muscle development. It was well known that transforming growth factor beta 1 (TGFβ1) is involved in a variety of cellular processes, such as proliferation, differentiation, and apoptosis. However, little is known about the m<sup>6</sup>A-mediated TGFβ1 regulation in myogenesis. Here, we observed an increase in endogenous TGFβ1 expression and activity during myotube differentiation. However, the knockdown of TGFβ1 inhibits the proliferation and induces cell apoptosis of myoblast. Moreover, we found that m<sup>6</sup>A in 5′-untranslated regions (5′UTR) of TGFβ1 promote its decay and inhibit its expression, leading to the blockage of the TGFβ1/SMAD2 signaling pathway. Furthermore, the targeted specific demethylation of TGFβ1 m<sup>6</sup>A using dCas13b-FTO significantly increased the TGFβ1-mediated activity of the SMAD2 signaling pathway, promoting myoblast proliferation. These findings suggest that TGFβ1 is an essential regulator of myoblast growth that is negatively regulated by m<sup>6</sup>A. Overall, these results highlight the critical role of m<sup>6</sup>A-mediated post-transcriptional regulation in myogenesis.https://www.mdpi.com/2073-4409/12/7/1005m<sup>6</sup>AdCas13bTGFβ1cell proliferationmyoblast
spellingShingle Kaiping Deng
Zhipeng Liu
Xiaodan Li
Zhen Zhang
Yixuan Fan
Qunhao Huang
Yanli Zhang
Feng Wang
Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway
Cells
m<sup>6</sup>A
dCas13b
TGFβ1
cell proliferation
myoblast
title Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway
title_full Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway
title_fullStr Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway
title_full_unstemmed Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway
title_short Targeted Demethylation of the TGFβ1 mRNA Promotes Myoblast Proliferation via Activating the SMAD2 Signaling Pathway
title_sort targeted demethylation of the tgfβ1 mrna promotes myoblast proliferation via activating the smad2 signaling pathway
topic m<sup>6</sup>A
dCas13b
TGFβ1
cell proliferation
myoblast
url https://www.mdpi.com/2073-4409/12/7/1005
work_keys_str_mv AT kaipingdeng targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT zhipengliu targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT xiaodanli targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT zhenzhang targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT yixuanfan targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT qunhaohuang targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT yanlizhang targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway
AT fengwang targeteddemethylationofthetgfb1mrnapromotesmyoblastproliferationviaactivatingthesmad2signalingpathway