Your browser doesn't support javascript.
loading
Prdm6 drives ductus arteriosus closure by promoting ductus arteriosus smooth muscle cell identity and contractility.
Zou, Meng; Mangum, Kevin D; Magin, Justin C; Cao, Heidi H; Yarboro, Michael T; Shelton, Elaine L; Taylor, Joan M; Reese, Jeff; Furey, Terrence S; Mack, Christopher P.
Afiliação
  • Zou M; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Mangum KD; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Magin JC; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Cao HH; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Yarboro MT; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
  • Shelton EL; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
  • Taylor JM; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Reese J; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
  • Furey TS; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
  • Mack CP; Department of Pathology and McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
JCI Insight ; 8(5)2023 03 08.
Article em En | MEDLINE | ID: mdl-36749647
ABSTRACT
Based upon our demonstration that the smooth muscle cell-selective (SMC-selective) putative methyltransferase, Prdm6, interacts with myocardin-related transcription factor-A, we examined Prdm6's role in SMCs in vivo using cell type-specific knockout mouse models. Although SMC-specific depletion of Prdm6 in adult mice was well tolerated, Prdm6 depletion in Wnt1-expressing cells during development resulted in perinatal lethality and a completely penetrant patent ductus arteriosus (DA) phenotype. Lineage tracing experiments in Wnt1Cre2 Prdm6fl/fl ROSA26LacZ mice revealed normal neural crest-derived SMC investment of the outflow tract. In contrast, myography measurements on DA segments isolated from E18.5 embryos indicated that Prdm6 depletion significantly reduced DA tone and contractility. RNA-Seq analyses on DA and ascending aorta samples at E18.5 identified a DA-enriched gene program that included many SMC-selective contractile associated proteins that was downregulated by Prdm6 depletion. Chromatin immunoprecipitation-sequencing experiments in outflow tract SMCs demonstrated that 50% of the genes Prdm6 depletion altered contained Prdm6 binding sites. Finally, using several genome-wide data sets, we identified an SMC-selective enhancer within the Prdm6 third intron that exhibited allele-specific activity, providing evidence that rs17149944 may be the causal SNP for a cardiovascular disease GWAS locus identified within the human PRDM6 gene.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Canal Arterial / Permeabilidade do Canal Arterial Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans / Pregnancy Idioma: En Revista: JCI Insight Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Canal Arterial / Permeabilidade do Canal Arterial Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans / Pregnancy Idioma: En Revista: JCI Insight Ano de publicação: 2023 Tipo de documento: Article