Genome Biology (Jul 2023)

Stable maternal proteins underlie distinct transcriptome, translatome, and proteome reprogramming during mouse oocyte-to-embryo transition

  • Hongmei Zhang,
  • Shuyan Ji,
  • Ke Zhang,
  • Yuling Chen,
  • Jia Ming,
  • Feng Kong,
  • Lijuan Wang,
  • Shun Wang,
  • Zhuoning Zou,
  • Zhuqing Xiong,
  • Kai Xu,
  • Zili Lin,
  • Bo Huang,
  • Ling Liu,
  • Qiang Fan,
  • Suoqin Jin,
  • Haiteng Deng,
  • Wei Xie

DOI
https://doi.org/10.1186/s13059-023-02997-8
Journal volume & issue
Vol. 24, no. 1
pp. 1 – 25

Abstract

Read online

Abstract Background The oocyte-to-embryo transition (OET) converts terminally differentiated gametes into a totipotent embryo and is critically controlled by maternal mRNAs and proteins, while the genome is silent until zygotic genome activation. How the transcriptome, translatome, and proteome are coordinated during this critical developmental window remains poorly understood. Results Utilizing a highly sensitive and quantitative mass spectrometry approach, we obtain high-quality proteome data spanning seven mouse stages, from full-grown oocyte (FGO) to blastocyst, using 100 oocytes/embryos at each stage. Integrative analyses reveal distinct proteome reprogramming compared to that of the transcriptome or translatome. FGO to 8-cell proteomes are dominated by FGO-stockpiled proteins, while the transcriptome and translatome are more dynamic. FGO-originated proteins frequently persist to blastocyst while corresponding transcripts are already downregulated or decayed. Improved concordance between protein and translation or transcription is observed for genes starting translation upon meiotic resumption, as well as those transcribed and translated only in embryos. Concordance between protein and transcription/translation is also observed for proteins with short half-lives. We built a kinetic model that predicts protein dynamics by incorporating both initial protein abundance in FGOs and translation kinetics across developmental stages. Conclusions Through integrative analyses of datasets generated by ultrasensitive methods, our study reveals that the proteome shows distinct dynamics compared to the translatome and transcriptome during mouse OET. We propose that the remarkably stable oocyte-originated proteome may help save resources to accommodate the demanding needs of growing embryos. This study will advance our understanding of mammalian OET and the fundamental principles governing gene expression.

Keywords