Sirtuin 1 functionally and physically interacts with disruptor of telomeric silencing-1 to regulate alpha-ENaC transcription in collecting duct

J Biol Chem. 2009 Jul 31;284(31):20917-26. doi: 10.1074/jbc.M109.020073. Epub 2009 Jun 2.

Abstract

Aldosterone increases renal tubular Na+ absorption in large part by increasing transcription of the epithelial Na(+) channel alpha-subunit (alpha-ENaC) expressed in the apical membrane of collecting duct principal cells. We recently reported that a complex containing the histone H3K79 methyltransferase disruptor of telomeric silencing-1 (Dot1) associates with and represses the alpha-ENaC promoter in mouse inner medullary collecting duct mIMCD3 cells, and that aldosterone acts to disrupt this complex and its inhibitory effects (Zhang, W., Xia, X., Reisenauer, M. R., Rieg, T., Lang, F., Kuhl, D., Vallon, V., and Kone, B. C. (2007) J. Clin. Invest. 117, 773-783). Here we demonstrate that the NAD(+)-dependent deacetylase sirtuin 1 (Sirt1) functionally and physically interacts with Dot1 to enhance the distributive activity of Dot1 on H3K79 methylation and thereby represses alpha-ENaC transcription in mIMCD3 cells. Sirt1 overexpression inhibited basal alpha-ENaC mRNA expression and alpha-ENaC promoter activity, surprisingly in a deacetylase-independent manner. The ability of Sirt1 to inhibit alpha-ENaC transcription was retained in a truncated Sirt1 construct expressing only its N-terminal domain. Conversely, Sirt1 knockdown enhanced alpha-ENaC mRNA levels and alpha-ENaC promoter activity, and inhibited global H3K79 methylation, particularly H3K79 trimethylation, in chromatin associated with the alpha-ENaC promoter. Sirt1 and Dot1 co-immunoprecipitated from mIMCD3 cells and colocalized in the nucleus. Sirt1 immunoprecipitated from chromatin associated with regions of the alpha-ENaC promoter known to associate with Dot1. Aldosterone inhibited Sirt1 association at two of these regions, as well as Sirt1 mRNA expression, in a coordinate manner with induction of alpha-ENaC transcription. Overexpressed Sirt1 inhibited aldosterone induction of alpha-ENaC transcription independent of effects on mineralocorticoid receptor trans-activation. These data identify Sirt1 as a novel modulator of alpha-ENaC, Dot1, and the aldosterone signaling pathway.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Aldosterone / pharmacology
  • Animals
  • Chromatin / metabolism
  • Epithelial Sodium Channels / genetics*
  • Epithelial Sodium Channels / metabolism
  • Gene Expression Regulation / drug effects
  • Gene Knockdown Techniques
  • Histone Deacetylases / metabolism
  • Histone-Lysine N-Methyltransferase
  • Histones / metabolism
  • Immunoprecipitation
  • Kidney Tubules, Collecting / cytology
  • Kidney Tubules, Collecting / drug effects
  • Kidney Tubules, Collecting / metabolism*
  • Lysine / metabolism
  • Methylation / drug effects
  • Methyltransferases / metabolism*
  • Mice
  • Promoter Regions, Genetic / genetics
  • Protein Binding / drug effects
  • RNA, Messenger / genetics
  • RNA, Messenger / metabolism
  • Receptors, Mineralocorticoid / metabolism
  • Sirtuin 1
  • Sirtuins / genetics
  • Sirtuins / metabolism*
  • Transcription, Genetic* / drug effects

Substances

  • Chromatin
  • Epithelial Sodium Channels
  • Histones
  • RNA, Messenger
  • Receptors, Mineralocorticoid
  • Aldosterone
  • Dot1l protein, mouse
  • Methyltransferases
  • Histone-Lysine N-Methyltransferase
  • Sirt1 protein, mouse
  • Sirtuin 1
  • Sirtuins
  • Histone Deacetylases
  • Lysine