A calcium-dependent switch in a CREST-BRG1 complex regulates activity-dependent gene expression.

CREST plays a critical role in activity-dependent development, but its mechanism of action is not well understood. Here, we show that a CREST-BRG1 complex regulates promoter activation by orchestrating a calcium-dependent release of a repressor complex and a recruitment of an activator complex. In resting neurons, transcription of the c-fos ...
promoter is inhibited by BRG1-dependent recruitment of a phospho-Rb-HDAC repressor complex. Upon calcium influx, Rb becomes dephosphorylated at serine 795 by calcineurin, which leads to release of the repressor complex. At the same time, there is increased recruitment of CBP to the promoter by a CREST-dependent mechanism, which leads to transcriptional activation. The CREST-BRG1 also binds to the NR2B promoter, and activity-dependent induction of NR2B expression involves a release of HDAC1 and recruitment of CBP, suggesting that this mechanism may be generally involved in regulating calcium-dependent transcription of neuronal genes.
Mesh Terms:
Animals, Calcium, Cells, Cultured, Cerebral Cortex, Chromatin Immunoprecipitation, DNA Helicases, Embryo, Mammalian, Gene Expression Regulation, Green Fluorescent Proteins, Histone Deacetylases, Humans, Neurons, Nuclear Proteins, Promoter Regions, Genetic, Proto-Oncogene Proteins c-fos, Rats, Trans-Activators, Transcription Factors, Transcriptional Activation, Transfection, Two-Hybrid System Techniques
Neuron
Date: Dec. 10, 2008
Download Curated Data For This Publication
104169
Switch View:
  • Interactions 8