Correlation Engine 2.0
Clear Search sequence regions


  • chromatin (2)
  • expressed genes (1)
  • factors (2)
  • female (1)
  • humans (1)
  • mice (1)
  • prosencephalon (1)
  • protein human (1)
  • RAI1 (7)
  • rats (2)
  • regulates (1)
  • rna (1)
  • Sizes of these terms reflect their relevance to your search.

    Long-lasting forms of synaptic plasticity such as synaptic scaling are critically dependent on transcription. Activity-dependent transcriptional dynamics in neurons, however, remain incompletely characterized because most previous efforts relied on measurement of steady-state mRNAs. Here, we use nascent RNA sequencing to profile transcriptional dynamics of primary neuron cultures undergoing network activity shifts. We find pervasive transcriptional changes, in which ∼45% of expressed genes respond to network activity shifts. We further link retinoic acid-induced 1 (RAI1), the Smith-Magenis syndrome gene, to the transcriptional program driven by reduced network activity. Remarkable agreement among nascent transcriptomes, dynamic chromatin occupancy of RAI1, and electrophysiological properties of Rai1-deficient neurons demonstrates the essential roles of RAI1 in suppressing synaptic upscaling in the naive network, while promoting upscaling triggered by activity silencing. These results highlight the utility of bona fide transcription profiling to discover mechanisms of activity-dependent chromatin remodeling that underlie normal and pathological synaptic plasticity. Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.

    Citation

    Patricia M Garay, Alex Chen, Takao Tsukahara, Jean Carlos Rodríguez Díaz, Rafi Kohen, J Christian Althaus, Margarete A Wallner, Roman J Giger, Kevin S Jones, Michael A Sutton, Shigeki Iwase. RAI1 Regulates Activity-Dependent Nascent Transcription and Synaptic Scaling. Cell reports. 2020 Aug 11;32(6):108002

    Expand section icon Mesh Tags

    Expand section icon Substances


    PMID: 32783930

    View Full Text