S-nitrosylation of HDAC2 regulates the expression of the chromatin-remodeling factor Brm during radial neuron migration

Proc Natl Acad Sci U S A. 2013 Feb 19;110(8):3113-8. doi: 10.1073/pnas.1218126110. Epub 2013 Jan 28.

Abstract

Dynamic epigenetic modifications play a key role in mediating the expression of genes required for neuronal development. We previously identified nitric oxide (NO) as a signaling molecule that mediates S-nitrosylation of histone deacetylase 2 (HDAC2) and epigenetic changes in neurons. Here, we show that HDAC2 nitrosylation regulates neuronal radial migration during cortical development. Bead-array analysis performed in the developing cortex revealed that brahma (Brm), a subunit of the ATP-dependent chromatin-remodeling complex BRG/brahma-associated factor, is one of the genes regulated by S-nitrosylation of HDAC2. In the cortex, expression of a mutant form of HDAC2 that cannot be nitrosylated dramatically inhibits Brm expression. Our study identifies NO and HDAC2 nitrosylation as part of a signaling pathway that regulates cortical development and the expression of Brm in neurons.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Movement*
  • Cell Separation
  • Cerebral Cortex / cytology
  • Cerebral Cortex / metabolism
  • Chromatin Assembly and Disassembly*
  • Electroporation
  • Female
  • Flow Cytometry
  • Histone Deacetylase 2 / metabolism*
  • Mice
  • Neurons / cytology*
  • Nitric Oxide / metabolism*
  • Pregnancy
  • Signal Transduction
  • Transcription Factors / metabolism*

Substances

  • Smarca2 protein, mouse
  • Transcription Factors
  • Nitric Oxide
  • Hdac2 protein, mouse
  • Histone Deacetylase 2