Cargando…

A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network

microRNAs (miRNAs) are non-coding RNAs that regulate gene expression post-transcriptionally, and mounting evidence supports the prevalence and functional significance of their interplay with transcription factors (TFs). Here we describe the identification of a regulatory circuit between muscle miRNA...

Descripción completa

Detalles Bibliográficos
Autores principales: Lu, Leina, Zhou, Liang, Chen, Eric Z., Sun, Kun, Jiang, Peiyong, Wang, Lijun, Su, Xiaoxi, Sun, Hao, Wang, Huating
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2012
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3271076/
https://www.ncbi.nlm.nih.gov/pubmed/22319554
http://dx.doi.org/10.1371/journal.pone.0027596
_version_ 1782222650652753920
author Lu, Leina
Zhou, Liang
Chen, Eric Z.
Sun, Kun
Jiang, Peiyong
Wang, Lijun
Su, Xiaoxi
Sun, Hao
Wang, Huating
author_facet Lu, Leina
Zhou, Liang
Chen, Eric Z.
Sun, Kun
Jiang, Peiyong
Wang, Lijun
Su, Xiaoxi
Sun, Hao
Wang, Huating
author_sort Lu, Leina
collection PubMed
description microRNAs (miRNAs) are non-coding RNAs that regulate gene expression post-transcriptionally, and mounting evidence supports the prevalence and functional significance of their interplay with transcription factors (TFs). Here we describe the identification of a regulatory circuit between muscle miRNAs (miR-1, miR-133 and miR-206) and Yin Yang 1 (YY1), an epigenetic repressor of skeletal myogenesis in mouse. Genome-wide identification of potential down-stream targets of YY1 by combining computational prediction with expression profiling data reveals a large number of putative miRNA targets of YY1 during skeletal myoblasts differentiation into myotubes with muscle miRs ranking on top of the list. The subsequent experimental results demonstrate that YY1 indeed represses muscle miRs expression in myoblasts and the repression is mediated through multiple enhancers and recruitment of Polycomb complex to several YY1 binding sites. YY1 regulating miR-1 is functionally important for both C2C12 myogenic differentiation and injury-induced muscle regeneration. Furthermore, we demonstrate that miR-1 in turn targets YY1, thus forming a negative feedback loop. Together, these results identify a novel regulatory circuit required for skeletal myogenesis and reinforce the idea that regulatory circuitries involving miRNAs and TFs are prevalent mechanisms.
format Online
Article
Text
id pubmed-3271076
institution National Center for Biotechnology Information
language English
publishDate 2012
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-32710762012-02-08 A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network Lu, Leina Zhou, Liang Chen, Eric Z. Sun, Kun Jiang, Peiyong Wang, Lijun Su, Xiaoxi Sun, Hao Wang, Huating PLoS One Research Article microRNAs (miRNAs) are non-coding RNAs that regulate gene expression post-transcriptionally, and mounting evidence supports the prevalence and functional significance of their interplay with transcription factors (TFs). Here we describe the identification of a regulatory circuit between muscle miRNAs (miR-1, miR-133 and miR-206) and Yin Yang 1 (YY1), an epigenetic repressor of skeletal myogenesis in mouse. Genome-wide identification of potential down-stream targets of YY1 by combining computational prediction with expression profiling data reveals a large number of putative miRNA targets of YY1 during skeletal myoblasts differentiation into myotubes with muscle miRs ranking on top of the list. The subsequent experimental results demonstrate that YY1 indeed represses muscle miRs expression in myoblasts and the repression is mediated through multiple enhancers and recruitment of Polycomb complex to several YY1 binding sites. YY1 regulating miR-1 is functionally important for both C2C12 myogenic differentiation and injury-induced muscle regeneration. Furthermore, we demonstrate that miR-1 in turn targets YY1, thus forming a negative feedback loop. Together, these results identify a novel regulatory circuit required for skeletal myogenesis and reinforce the idea that regulatory circuitries involving miRNAs and TFs are prevalent mechanisms. Public Library of Science 2012-02-01 /pmc/articles/PMC3271076/ /pubmed/22319554 http://dx.doi.org/10.1371/journal.pone.0027596 Text en Lu et al. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Lu, Leina
Zhou, Liang
Chen, Eric Z.
Sun, Kun
Jiang, Peiyong
Wang, Lijun
Su, Xiaoxi
Sun, Hao
Wang, Huating
A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network
title A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network
title_full A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network
title_fullStr A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network
title_full_unstemmed A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network
title_short A Novel YY1-miR-1 Regulatory Circuit in Skeletal Myogenesis Revealed by Genome-Wide Prediction of YY1-miRNA Network
title_sort novel yy1-mir-1 regulatory circuit in skeletal myogenesis revealed by genome-wide prediction of yy1-mirna network
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3271076/
https://www.ncbi.nlm.nih.gov/pubmed/22319554
http://dx.doi.org/10.1371/journal.pone.0027596
work_keys_str_mv AT luleina anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT zhouliang anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT chenericz anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT sunkun anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT jiangpeiyong anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT wanglijun anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT suxiaoxi anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT sunhao anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT wanghuating anovelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT luleina novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT zhouliang novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT chenericz novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT sunkun novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT jiangpeiyong novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT wanglijun novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT suxiaoxi novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT sunhao novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork
AT wanghuating novelyy1mir1regulatorycircuitinskeletalmyogenesisrevealedbygenomewidepredictionofyy1mirnanetwork