Profiling the role of m6A effectors in the regulation of pluripotent reprogramming

Abstract The N6-methyladenosine (m6A) RNA modification plays essential roles in multiple biological processes, including stem cell fate determination. To explore the role of the m6A modification in pluripotent reprogramming, we used RNA-seq to map m6A effectors in human iPSCs, fibroblasts, and H9 ES...

Full description

Bibliographic Details
Main Authors: Wenjun Wang, Lei Zhou, Hui Li, Tingge Sun, Xue Wen, Wei Li, Miguel A. Esteban, Andrew R. Hoffman, Ji-Fan Hu, Jiuwei Cui
Format: Article
Language:English
Published: BMC 2024-04-01
Series:Human Genomics
Subjects:
Online Access:https://doi.org/10.1186/s40246-024-00597-6
_version_ 1797219640391237632
author Wenjun Wang
Lei Zhou
Hui Li
Tingge Sun
Xue Wen
Wei Li
Miguel A. Esteban
Andrew R. Hoffman
Ji-Fan Hu
Jiuwei Cui
author_facet Wenjun Wang
Lei Zhou
Hui Li
Tingge Sun
Xue Wen
Wei Li
Miguel A. Esteban
Andrew R. Hoffman
Ji-Fan Hu
Jiuwei Cui
author_sort Wenjun Wang
collection DOAJ
description Abstract The N6-methyladenosine (m6A) RNA modification plays essential roles in multiple biological processes, including stem cell fate determination. To explore the role of the m6A modification in pluripotent reprogramming, we used RNA-seq to map m6A effectors in human iPSCs, fibroblasts, and H9 ESCs, as well as in mouse ESCs and fibroblasts. By integrating the human and mouse RNA-seq data, we found that 19 m6A effectors were significantly upregulated in reprogramming. Notably, IGF2BPs, particularly IGF2BP1, were among the most upregulated genes in pluripotent cells, while YTHDF3 had high levels of expression in fibroblasts. Using quantitative PCR and Western blot, we validated the pluripotency-associated elevation of IGF2BPs. Knockdown of IGF2BP1 induced the downregulation of stemness genes and exit from pluripotency. Proteome analysis of cells collected at both the beginning and terminal states of the reprogramming process revealed that the IGF2BP1 protein was positively correlated with stemness markers SOX2 and OCT4. The eCLIP-seq target analysis showed that IGF2BP1 interacted with the coding sequence (CDS) and 3’UTR regions of the SOX2 transcripts, in agreement with the location of m6A modifications. This study identifies IGF2BP1 as a vital pluripotency-associated m6A effector, providing new insight into the interplay between m6A epigenetic modifications and pluripotent reprogramming.
first_indexed 2024-04-24T12:36:51Z
format Article
id doaj.art-0538708e10884724abb35f2ecfeb727e
institution Directory Open Access Journal
issn 1479-7364
language English
last_indexed 2024-04-24T12:36:51Z
publishDate 2024-04-01
publisher BMC
record_format Article
series Human Genomics
spelling doaj.art-0538708e10884724abb35f2ecfeb727e2024-04-07T11:24:47ZengBMCHuman Genomics1479-73642024-04-0118111410.1186/s40246-024-00597-6Profiling the role of m6A effectors in the regulation of pluripotent reprogrammingWenjun Wang0Lei Zhou1Hui Li2Tingge Sun3Xue Wen4Wei Li5Miguel A. Esteban6Andrew R. Hoffman7Ji-Fan Hu8Jiuwei Cui9Cancer Center, The First Hospital of Jilin UniversityCancer Center, The First Hospital of Jilin UniversityCancer Center, The First Hospital of Jilin UniversityCancer Center, The First Hospital of Jilin UniversityCancer Center, The First Hospital of Jilin UniversityCancer Center, The First Hospital of Jilin UniversityGuangzhou Institutes of Biomedicine and Health, Chinese Academy of SciencesVA Palo Alto Health Care System, Stanford University School of MedicineCancer Center, The First Hospital of Jilin UniversityCancer Center, The First Hospital of Jilin UniversityAbstract The N6-methyladenosine (m6A) RNA modification plays essential roles in multiple biological processes, including stem cell fate determination. To explore the role of the m6A modification in pluripotent reprogramming, we used RNA-seq to map m6A effectors in human iPSCs, fibroblasts, and H9 ESCs, as well as in mouse ESCs and fibroblasts. By integrating the human and mouse RNA-seq data, we found that 19 m6A effectors were significantly upregulated in reprogramming. Notably, IGF2BPs, particularly IGF2BP1, were among the most upregulated genes in pluripotent cells, while YTHDF3 had high levels of expression in fibroblasts. Using quantitative PCR and Western blot, we validated the pluripotency-associated elevation of IGF2BPs. Knockdown of IGF2BP1 induced the downregulation of stemness genes and exit from pluripotency. Proteome analysis of cells collected at both the beginning and terminal states of the reprogramming process revealed that the IGF2BP1 protein was positively correlated with stemness markers SOX2 and OCT4. The eCLIP-seq target analysis showed that IGF2BP1 interacted with the coding sequence (CDS) and 3’UTR regions of the SOX2 transcripts, in agreement with the location of m6A modifications. This study identifies IGF2BP1 as a vital pluripotency-associated m6A effector, providing new insight into the interplay between m6A epigenetic modifications and pluripotent reprogramming.https://doi.org/10.1186/s40246-024-00597-6m6AEpigeneticsIGF2BPsiPSCStem cellsReprogramming
spellingShingle Wenjun Wang
Lei Zhou
Hui Li
Tingge Sun
Xue Wen
Wei Li
Miguel A. Esteban
Andrew R. Hoffman
Ji-Fan Hu
Jiuwei Cui
Profiling the role of m6A effectors in the regulation of pluripotent reprogramming
Human Genomics
m6A
Epigenetics
IGF2BPs
iPSC
Stem cells
Reprogramming
title Profiling the role of m6A effectors in the regulation of pluripotent reprogramming
title_full Profiling the role of m6A effectors in the regulation of pluripotent reprogramming
title_fullStr Profiling the role of m6A effectors in the regulation of pluripotent reprogramming
title_full_unstemmed Profiling the role of m6A effectors in the regulation of pluripotent reprogramming
title_short Profiling the role of m6A effectors in the regulation of pluripotent reprogramming
title_sort profiling the role of m6a effectors in the regulation of pluripotent reprogramming
topic m6A
Epigenetics
IGF2BPs
iPSC
Stem cells
Reprogramming
url https://doi.org/10.1186/s40246-024-00597-6
work_keys_str_mv AT wenjunwang profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT leizhou profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT huili profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT tinggesun profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT xuewen profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT weili profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT miguelaesteban profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT andrewrhoffman profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT jifanhu profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming
AT jiuweicui profilingtheroleofm6aeffectorsintheregulationofpluripotentreprogramming