4.7 Article

Neural stem/precursor cells dynamically change their epigenetic landscape to differentially respond to BMP signaling for fate switching during brain development

Journal

GENES & DEVELOPMENT
Volume 35, Issue 21-22, Pages 1431-1444

Publisher

COLD SPRING HARBOR LAB PRESS, PUBLICATIONS DEPT
DOI: 10.1101/gad.348797.121

Keywords

neural stem; precursor cell; differentiation; bone morphogenetic protein; Smad; neuron; astrocyte; epigenetic modifications; chromatin accessibility

Funding

  1. Ministry of Education, Culture, Sports, Science, and Technology/Japan Society for the Promotion of Science KAKENHI [JP16H06527, JP16K21734, JP26710003, JP20K06875]
  2. Japan Science and Technology Agency CREST [JPMJCR16G1]
  3. Naito Foundation
  4. Platform for Advanced Genome Science (PAGS) [JP17H05647]

Ask authors/readers for more resources

This study reveals a sophisticated regulation of NPC fate change during cortical development, where the interplay between cell-extrinsic cues and -intrinsic epigenetic programs plays a critical role. Smads, downstream transcription factors from BMP signaling, target different genomic regions in neurogenic and gliogenic NPCs, highlighting the importance of their role in regulating NPC properties.
In this study, Katada et al. investigated NPC fate regulation and, using multiple genome-wide analyses, they demonstrate that Smads, transcription factors that act downstream from BMP signaling, target dramatically different genomic regions in neurogenic and gliogenic NPCs. Their results show the regulation of NPC property change mediated by the interplay between cell-extrinsic cues and -intrinsic epigenetic programs during cortical development. During neocortical development, tight regulation of neurogenesis-to-astrogenesis switching of neural precursor cells (NPCs) is critical to generate a balanced number of each neural cell type for proper brain functions. Accumulating evidence indicates that a complex array of epigenetic modifications and the availability of extracellular factors control the timing of neuronal and astrocytic differentiation. However, our understanding of NPC fate regulation is still far from complete. Bone morphogenetic proteins (BMPs) are renowned as cytokines that induce astrogenesis of gliogenic late-gestational NPCs. They also promote neurogenesis of mid-gestational NPCs, although the underlying mechanisms remain elusive. By performing multiple genome-wide analyses, we demonstrate that Smads, transcription factors that act downstream from BMP signaling, target dramatically different genomic regions in neurogenic and gliogenic NPCs. We found that histone H3K27 trimethylation and DNA methylation around Smad-binding sites change rapidly as gestation proceeds, strongly associated with the alteration of accessibility of Smads to their target binding sites. Furthermore, we identified two lineage-specific Smad-interacting partners-Sox11 for neurogenic and Sox8 for astrocytic differentiation-that further ensure Smad-regulated fate-specific gene induction. Our findings illuminate an exquisite regulation of NPC property change mediated by the interplay between cell-extrinsic cues and -intrinsic epigenetic programs during cortical development.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available