Abstract

BackgroundPromoters and enhancers are cis-regulatory DNA sequences that control specificity and quantity of transcription. Both are rich on clusters of cis-acting sites that interact with sequence-specific DNA-binding transcription factors (TFs). At the level of chromatin, these regions display increased nuclease sensitivity, reduced nucleosome density, including nucleosome-free regions, and specific combinations of posttranslational modifications of core histone proteins. Together, “open” and “closed” chromatins represent transcriptionally active and repressed states of individual genes, respectively. Cellular differentiation is marked by changes in local chromatin structure. Lens morphogenesis, regulated by TF Pax6, includes differentiation of epithelial precursor cells into lens fibers in parallel with differentiation of epithelial precursors into the mature lens epithelium.ResultsUsing ATAC-seq, we investigated dynamics of chromatin changes during mouse lens fibers and epithelium differentiation. Tissue-specific features of these processes are demonstrated via comparative studies of embryonic stem cells, forebrain, and liver chromatins. Unbiased analysis reveals cis-regulatory logic of lens differentiation through known (e.g., AP-1, Ets, Hsf4, Maf, and Pax6 sites) and novel (e.g., CTCF, Tead, and NF1) motifs. Twenty-six DNA-binding TFs, recognizing these cis-motifs, are markedly up-regulated in differentiating lens fibers. As specific examples, our ATAC-seq data uncovered both the regulatory regions and TF binding motifs in Foxe3, Prox1, and Mip loci that are consistent with previous, though incomplete, experimental data. A cross-examination of Pax6 binding with ATAC-seq data demonstrated that Pax6 bound to both open (H3K27ac and P300-enriched) and closed chromatin domains in lens and forebrain.ConclusionsOur study has generated the first lens chromatin accessibility maps that support a general model of stage-specific chromatin changes associated with transcriptional activities of batteries of genes required for lens fiber cell formation. Analysis of active (or open) promoters and enhancers reveals important cis-DNA motifs that establish the molecular foundation for temporally and spatially regulated gene expression in lens. Together, our data and models open new avenues for the field to conduct mechanistic studies of transcriptional control regions, reconstruction of gene regulatory networks that govern lens morphogenesis, and identification of cataract-causing mutations in noncoding sequences.

Highlights

  • Cellular differentiation is driven by the coordinated expression of batteries of genes that encode proteins required for cellular specialization and function

  • Principal component analysis of the top 500 peaks with biggest variances separated the eight samples into four groups and organized them into two differentiation pathways that matched the known biology of lens differentiation (Fig. 1e): lens fiber cell differentiation (E14.5 epithelium → E14.5 fibers → P0.5 fibers) and epithelium maturation (E14.5 epithelium → P0.5 epithelium)

  • We found that genes with Pax6 binding at open chromatin regions exhibited significantly higher expression than genes with Pax6 binding at closed chromatin regions in P0.5 lens epithelium, P0.5 lens fibers, and E12.5 forebrain [16] (Mann–Whitney U test, p < 0.05; Fig. 8d)

Read more

Summary

Introduction

Cellular differentiation is driven by the coordinated expression of batteries of genes that encode proteins required for cellular specialization and function. Tissue specificity of transcription is primarily regulated by a combinatorial action of sequence-specific DNA-binding transcription factors and their interactions with promoters and distal enhancers [1, 2]. Both promoters and enhancers of transcriptionally active genes display increased sensitivity to nuclease digestion [3] and are located to “open” chromatin domains. Transcriptionally repressed genes are often organized within “closed” chromatin domains, marked by different histone modifications (e.g., H3K27me and H3K9me3) [2, 7]. At the level of chromatin, these regions display increased nuclease sensitivity, reduced nucleosome density, including nucleosome-free regions, and specific combinations of posttranslational modifications of core histone proteins. Lens morphogenesis, regulated by TF Pax, includes differentiation of epithelial precursor cells into lens fibers in parallel with differentiation of epithelial precursors into the mature lens epithelium

Objectives
Methods
Results
Discussion
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