Abstract

The chromatin remodeling complexes chromatin accessibility complex and ATP-utilizing chromatin assembly and remodeling factor (ACF) combine the ATPase ISWI with the signature subunit ACF1. These enzymes catalyze well-studied nucleosome sliding reactions in vitro, but how their actions affect physiological gene expression remains unclear. Here, we explored the influence of Drosophila melanogaster chromatin accessibility complex/ACF on transcription by using complementary gain- and loss-of-function approaches. Targeting ACF1 to multiple reporter genes inserted at many different genomic locations revealed a context-dependent inactivation of poorly transcribed reporters in repressive chromatin. Accordingly, single-embryo transcriptome analysis of an Acf knock-out allele showed that only lowly expressed genes are derepressed in the absence of ACF1. Finally, the nucleosome arrays in Acf-deficient chromatin show loss of physiological regularity, particularly in transcriptionally inactive domains. Taken together, our results highlight that ACF1-containing remodeling factors contribute to the establishment of an inactive ground state of the genome through chromatin organization.

Highlights

  • The chromatin accessibility complex (CHRAC) and the related ATPutilizing chromatin assembly and remodeling factor (ACF) are prototypic nucleosome sliding factors purified originally from extracts of Drosophila melanogaster embryos [1, 2]

  • Because ISWI is present in several other nucleosome remodelers [5], ACF1 serves as the signature regulatory subunit for the two complexes

  • Successful tethering of ACF1-GAL4DBD alone (GAL4) DNA-binding domain (GAL4DBD) in early embryos was confirmed by chromatin immunoprecipitation (ChIP)–quantitative PCR (Fig 1C)

Read more

Summary

Introduction

The chromatin accessibility complex (CHRAC) and the related ATPutilizing chromatin assembly and remodeling factor (ACF) are prototypic nucleosome sliding factors purified originally from extracts of Drosophila melanogaster embryos [1, 2]. ACF associates with two histone-fold subunits, CHRAC-14 and CHRAC-16, to form CHRAC [3]. Both complexes have very similar nucleosome sliding activity in vitro [4]. Because ISWI is present in several other nucleosome remodelers [5], ACF1 serves as the signature regulatory subunit for the two complexes. ISWI and ACF1 bind target nucleosomes and flanking linker DNA. Substrate binding and ATP hydrolysis cycles trigger conformation changes in the remodeler that disrupt histone–DNA interactions and eventually displace the intact histone octamer along the DNA, effectively sliding a nucleosome [6, 7, 8, 9, 10, 11, 12] (for review, see reference 13)

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