Your browser doesn't support javascript.
loading
Multiple mechanisms regulate H3 acetylation of enhancers in response to thyroid hormone.
Præstholm, Stine M; Siersbæk, Majken S; Nielsen, Ronni; Zhu, Xuguang; Hollenberg, Anthony N; Cheng, Sheue-Yann; Grøntved, Lars.
Afiliação
  • Præstholm SM; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
  • Siersbæk MS; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
  • Nielsen R; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
  • Zhu X; Laboratory of Molecular Biology, CCR, NCI, NIH, Bethesda, Maryland, United States of America.
  • Hollenberg AN; Division of Endocrinology, Diabetes and Metabolism Weill Cornell Medicine, New York, New York, United States of America.
  • Cheng SY; Laboratory of Molecular Biology, CCR, NCI, NIH, Bethesda, Maryland, United States of America.
  • Grøntved L; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
PLoS Genet ; 16(5): e1008770, 2020 05.
Article em En | MEDLINE | ID: mdl-32453730
ABSTRACT
Hormone-dependent activation of enhancers includes histone hyperacetylation and mediator recruitment. Histone hyperacetylation is mostly explained by a bimodal switch model, where histone deacetylases (HDACs) disassociate from chromatin, and histone acetyl transferases (HATs) are recruited. This model builds on decades of research on steroid receptor regulation of transcription. Yet, the general concept of the bimodal switch model has not been rigorously tested genome wide. We have used a genomics approach to study enhancer hyperacetylation by the thyroid hormone receptor (TR), described to operate as a bimodal switch. H3 acetylation, HAT and HDAC ChIP-seq analyses of livers from hypo- and hyperthyroid wildtype, TR deficient and NCOR1 disrupted mice reveal three types of thyroid hormone (T3)-regulated enhancers. One subset of enhancers is bound by HDAC3-NCOR1 in the absence of hormone and constitutively occupy TR and HATs irrespective of T3 levels, suggesting a poised enhancer state in absence of hormone. In presence of T3, HDAC3-NCOR1 dissociates from these enhancers leading to histone hyperacetylation, suggesting a histone acetylation rheostat function of HDACs at poised enhancers. Another subset of enhancers, not occupied by HDACs, is hyperacetylated in a T3-dependent manner, where TR is recruited to chromatin together with HATs. Lastly, a subset of enhancers, is not occupied directly by TR yet requires TR for histone hyperacetylation. This indirect enhancer activation involves co-association with TR bound enhancers within super-enhancers or topological associated domains. Collectively, this demonstrates various mechanisms controlling hormone-dependent transcription and adds significant details to the otherwise simple bimodal switch model.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Hormônios Tireóideos / Receptores dos Hormônios Tireóideos / Histonas / Elementos Facilitadores Genéticos / Histona Acetiltransferases Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Hormônios Tireóideos / Receptores dos Hormônios Tireóideos / Histonas / Elementos Facilitadores Genéticos / Histona Acetiltransferases Tipo de estudo: Prognostic_studies Limite: Animals Idioma: En Ano de publicação: 2020 Tipo de documento: Article