4.7 Article

The H3.3 chaperone Hira complex orchestrates oocyte developmental competence

Journal

DEVELOPMENT
Volume 149, Issue 5, Pages -

Publisher

COMPANY BIOLOGISTS LTD
DOI: 10.1242/dev.200044

Keywords

Hira complex; Histone H3.3; Oocyte-to-embryo transition; Zygotic genome activation; Competent oocyte

Funding

  1. Medical Research Council [MR/N022556/1]
  2. Wellcome Trust-University of Edinburgh Institutional Strategic Support Fund
  3. Deanery of Clinical Sciences, College of Medicine and Veterinary Medicine of the University of Edinburgh
  4. Scottish Government
  5. National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development [R01HD102533]
  6. U.S. Department of Agriculture-National Institute of Food and Agriculture [2019-67016-29863, W4171]
  7. Audubon Center for Research of Endangered Species
  8. University of Edinburgh

Ask authors/readers for more resources

Oocyte developmental competence and embryogenesis rely on the H3.3 chaperone Hira complex, which regulates chromatin condensation and transcriptional quiescence.
Successful reproduction requires an oocyte competent to sustain early embryo development. By the end of oogenesis, the oocyte has entered a transcriptionally silenced state, the mechanisms and significance of which remain poorly understood. Histone H3.3, a histone H3 variant, has unique cell cycle-independent functions in chromatin structure and gene expression. Here, we have characterised the H3.3 chaperone Hira/Cabin1/Ubn1 complex, showing that loss of function of any of these subunits causes early embryogenesis failure in mouse. Transcriptome and nascent RNA analyses revealed that transcription is aberrantly silenced in mutant oocytes. Histone marks, including H3K4me3 and H3K9me3, are reduced and chromatin accessibility is impaired in Hira/Cabin1 mutants. Misregulated genes in mutant oocytes include Zscan4d, a two-cell specific gene involved in zygote genome activation. Overexpression of Zscan4 in the oocyte partially recapitulates the phenotypes of Hira mutants and Zscan4 knockdown in Cabin1 mutant oocytes partially restored their developmental potential, illustrating that temporal and spatial expression of Zscan4 is fine-tuned at the oocyte-to-embryo transition. Thus, the H3.3 chaperone Hira complex has a maternal effect function in oocyte developmental competence and embryogenesis, through modulating chromatin condensation and transcriptional quiescence.

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