Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation

Although skeletal myogenesis is largely controlled by myogenic regulatory factors, epigenetic modifications have recently emerged as an essential regulatory mechanism of gene expression. Molecular regulation of stem cell differentiation is exerted through both genetic and epigenetic factors over dis...

Full description

Bibliographic Details
Main Author: Hamed, Munerah
Other Authors: Li, Qiao
Format: Others
Language:en
Published: Université d'Ottawa / University of Ottawa 2019
Subjects:
RXR
Online Access:http://hdl.handle.net/10393/39559
http://dx.doi.org/10.20381/ruor-23802
id ndltd-uottawa.ca-oai-ruor.uottawa.ca-10393-39559
record_format oai_dc
spelling ndltd-uottawa.ca-oai-ruor.uottawa.ca-10393-395592019-08-31T04:38:32Z Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation Hamed, Munerah Li, Qiao Histone acetyltransferase p300 Nuclear receptor RXR Gene regulation Chromatin Stem cell differentiation Epigenetics Although skeletal myogenesis is largely controlled by myogenic regulatory factors, epigenetic modifications have recently emerged as an essential regulatory mechanism of gene expression. Molecular regulation of stem cell differentiation is exerted through both genetic and epigenetic factors over distal enhancer regions. Understanding the mechanistic action of active or poised enhancers is therefore, imperative for the control of stem cell differentiation. Based on the genome-wide co-occurrence of different epigenetic marks in proliferating myoblasts, we have generated a chromatin state model to profile differentiation- and rexinoid-responsive histone acetylation in early myoblast differentiation. Here, we delineate the functional mode of transcription regulators during early myogenic differentiation using genome-wide chromatin state association. We define a role of transcriptional coactivator p300, when recruited by muscle master regulator MyoD, in the establishment and regulation of myogenic loci at the onset of myoblast differentiation. In addition, we reveal an enrichment of loci-specific histone acetylation at p300 associated active or poised enhancers, mainly when enlisted by MyoD. We have previously established that bexarotene, a clinically approved agonist of retinoid X receptor (RXR), promotes the specification and differentiation of skeletal muscle lineage. Hence, we investigated the genome-wide impact of rexinoids on myogenic differentiation and uncovered a new mechanism of rexinoid action, which is mediated by the nuclear receptor and largely reconciled through direct regulation of MyoD gene expression. In addition, we determined rexinoid-responsive residue-specific histone acetylation at a distinct chromatin state associated with MyoD and myogenin. Finally, through ChIP-seq and RNA-seq analyses, we have identified dystroglycan (Dag1) as a differentiation-dependent and a rexinoid-responsive model target, and we revealed a possible co-regulation of Dag1 by p300 and MyoD accompanied by enrichment of loci-specific histone acetylation. Taken together, we provide novel molecular insights into the regulation of myogenic enhancers by p300 in concert with MyoD. Furthermore, we provide novel mechanistic perceptions into the interplay between RXR signaling and chromatin states pertinent to myogenic programs in early myoblast differentiation. Our studies present a valuable insight for driving condition-specific chromatin state or enhancers pharmacologically to treat muscle-related diseases and for the identification of additional myogenic targets and molecular interactions for therapeutic development. 2019-08-30T15:39:04Z 2019-08-30 Thesis http://hdl.handle.net/10393/39559 http://dx.doi.org/10.20381/ruor-23802 en application/pdf Université d'Ottawa / University of Ottawa
collection NDLTD
language en
format Others
sources NDLTD
topic Histone acetyltransferase
p300
Nuclear receptor
RXR
Gene regulation
Chromatin
Stem cell differentiation
Epigenetics
spellingShingle Histone acetyltransferase
p300
Nuclear receptor
RXR
Gene regulation
Chromatin
Stem cell differentiation
Epigenetics
Hamed, Munerah
Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation
description Although skeletal myogenesis is largely controlled by myogenic regulatory factors, epigenetic modifications have recently emerged as an essential regulatory mechanism of gene expression. Molecular regulation of stem cell differentiation is exerted through both genetic and epigenetic factors over distal enhancer regions. Understanding the mechanistic action of active or poised enhancers is therefore, imperative for the control of stem cell differentiation. Based on the genome-wide co-occurrence of different epigenetic marks in proliferating myoblasts, we have generated a chromatin state model to profile differentiation- and rexinoid-responsive histone acetylation in early myoblast differentiation. Here, we delineate the functional mode of transcription regulators during early myogenic differentiation using genome-wide chromatin state association. We define a role of transcriptional coactivator p300, when recruited by muscle master regulator MyoD, in the establishment and regulation of myogenic loci at the onset of myoblast differentiation. In addition, we reveal an enrichment of loci-specific histone acetylation at p300 associated active or poised enhancers, mainly when enlisted by MyoD. We have previously established that bexarotene, a clinically approved agonist of retinoid X receptor (RXR), promotes the specification and differentiation of skeletal muscle lineage. Hence, we investigated the genome-wide impact of rexinoids on myogenic differentiation and uncovered a new mechanism of rexinoid action, which is mediated by the nuclear receptor and largely reconciled through direct regulation of MyoD gene expression. In addition, we determined rexinoid-responsive residue-specific histone acetylation at a distinct chromatin state associated with MyoD and myogenin. Finally, through ChIP-seq and RNA-seq analyses, we have identified dystroglycan (Dag1) as a differentiation-dependent and a rexinoid-responsive model target, and we revealed a possible co-regulation of Dag1 by p300 and MyoD accompanied by enrichment of loci-specific histone acetylation. Taken together, we provide novel molecular insights into the regulation of myogenic enhancers by p300 in concert with MyoD. Furthermore, we provide novel mechanistic perceptions into the interplay between RXR signaling and chromatin states pertinent to myogenic programs in early myoblast differentiation. Our studies present a valuable insight for driving condition-specific chromatin state or enhancers pharmacologically to treat muscle-related diseases and for the identification of additional myogenic targets and molecular interactions for therapeutic development.
author2 Li, Qiao
author_facet Li, Qiao
Hamed, Munerah
author Hamed, Munerah
author_sort Hamed, Munerah
title Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation
title_short Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation
title_full Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation
title_fullStr Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation
title_full_unstemmed Characterization of the Epigenetic Signature Underlying Early Myogenic Differentiation
title_sort characterization of the epigenetic signature underlying early myogenic differentiation
publisher Université d'Ottawa / University of Ottawa
publishDate 2019
url http://hdl.handle.net/10393/39559
http://dx.doi.org/10.20381/ruor-23802
work_keys_str_mv AT hamedmunerah characterizationoftheepigeneticsignatureunderlyingearlymyogenicdifferentiation
_version_ 1719241874740346880