Abstract

Erythropoiesis is one of the best understood examples of cellular differentiation. Morphologically, erythroid differentiation proceeds in a nearly identical fashion between humans and mice, but recent evidence has shown that networks of gene expression governing this process are divergent between species. We undertook a systematic comparative analysis of six histone modifications and four transcriptional master regulators in primary proerythroblasts and erythroid cell lines to better understand the underlying basis of these transcriptional differences. Our analyses suggest that while chromatin structure across orthologous promoters is strongly conserved, subtle differences are associated with transcriptional divergence between species. Many transcription factor (TF) occupancy sites were poorly conserved across species (∼25% for GATA1, TAL1, and NFE2) but were more conserved between proerythroblasts and cell lines derived from the same species. We found that certain cis-regulatory modules co-occupied by GATA1, TAL1, and KLF1 are under strict evolutionary constraint and localize to genes necessary for erythroid cell identity. More generally, we show that conserved TF occupancy sites are indicative of active regulatory regions and strong gene expression that is sustained during maturation. Our results suggest that evolutionary turnover of TF binding sites associates with changes in the underlying chromatin structure, driving transcriptional divergence. We provide examples of how this framework can be applied to understand epigenomic variation in specific regulatory regions, such as the β-globin gene locus. Our findings have important implications for understanding epigenomic changes that mediate variation in cellular differentiation across species, while also providing a valuable resource for studies of hematopoiesis.

Highlights

  • Red blood cell (RBC) production is one of the best understood examples of lineage commitment and cellular differentiation [1,2,3,4]

  • While studies of this process in mouse have substantially improved our knowledge of human erythropoiesis, recent work has shown a significant divergence in global gene expression across species, suggesting that extrapolation from mouse models to human is not always straightforward

  • By globally comparing chromatin structure across primary cells and model cell lines in both species, we discovered that while chromatin structure is well conserved at orthologous promoters, subtle changes are predictive of speciesspecific gene expression

Read more

Summary

Introduction

Red blood cell (RBC) production (erythropoiesis) is one of the best understood examples of lineage commitment and cellular differentiation [1,2,3,4]. The ProEs subsequently undergo terminal erythroid differentiation into mature RBCs that enucleate, contain a significant concentration of hemoglobin, and have highly elastic cytoskeletons [3]. This differentiation process is governed by a number of transcription factors (TFs) that dynamically coordinate a complex transcriptional gene regulatory network (GRN). Extrapolation from mouse models of terminal erythroid differentiation to humans has historically been straightforward, grounded in the nearly identical morphology of mature RBCs and their precursors between species [4,5,6]. While there are many well-known examples of species-specific differences in erythroid GRNs, such as developmental variation of b-like globin gene expression, the divergent role of BCL11A during developmental hemoglobin switching, and differences in cis-regulatory modules (CRMs) regulating GATA1 transcription [7,8,9], a marked global divergence in the expression profiles of the erythroid lineage was only recently described by systematic comparative analyses of human and murine erythroid transcriptomes [10,11]

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