Abstract

Neural cell fate acquisition is mediated by transcription factors expressed in nascent neuroectoderm, including Geminin and members of the Zic transcription factor family. However, regulatory networks through which this occurs are not well defined. Here, we identified Geminin-associated chromatin locations in embryonic stem cells and Geminin- and Zic1-associated locations during neural fate acquisition at a genome-wide level. We determined how Geminin deficiency affected histone acetylation at gene promoters during this process. We integrated these data to demonstrate that Geminin associates with and promotes histone acetylation at neurodevelopmental genes, while Geminin and Zic1 bind a shared gene subset. Geminin- and Zic1-associated genes exhibit embryonic nervous system-enriched expression and encode other regulators of neural development. Both Geminin and Zic1-associated peaks are enriched for Zic1 consensus binding motifs, while Zic1-bound peaks are also enriched for Sox3 motifs, suggesting co-regulatory potential. Accordingly, we found that Geminin and Zic1 could cooperatively activate the expression of several shared targets encoding transcription factors that control neurogenesis, neural plate patterning, and neuronal differentiation. We used these data to construct gene regulatory networks underlying neural fate acquisition. Establishment of this molecular program in nascent neuroectoderm directly links early neural cell fate acquisition with regulatory control of later neurodevelopment.

Highlights

  • Transcription factors[8,9,10,14,15,16,17] and is one of several cellular inhibitors of genome re-replication within each cell cycle[18]

  • We found that chromatin association of Gmnn changed during the ES-NE transition, but that most Gmnn-associated genes in both contexts exhibited embryonic central nervous system (CNS)-enriched expression

  • These Gmnn-associated genes were preferentially enriched for gene ontology (GO) terms associated with neural development, neurogenesis, and neuronal differentiation

Read more

Summary

Introduction

Transcription factors[8,9,10,14,15,16,17] and is one of several cellular inhibitors of genome re-replication within each cell cycle[18]. We used chromatin immunoprecipitation (ChIP) and generation sequencing (ChIP-seq) to define Gmnn-associated chromatin locations in ES cells and ES-derived neuroectoderm (NE) at a genome-wide level We compared these Gmnn-associated gene profiles with effects of Gmnn deficiency on histone acetylation of promoters during neural fate acquisition. This work defines unique and shared targets of Gmnn and Zic[1], which act with other nTFs to link transcriptional control of early neural fate acquisition with activation of gene expression programs driving several aspects of later neural development

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call