4.8 Article

Transcriptional landscape of the human cell cycle

Publisher

NATL ACAD SCIENCES
DOI: 10.1073/pnas.1617636114

Keywords

GRO-seq; nascent RNA; transcriptional regulation; epigenetics; cell cycle

Funding

  1. National Natural Science Foundation of China [31329003]
  2. Princess Margaret Cancer Foundation
  3. Canada Foundation for Innovation
  4. Ontario Research Fund [CFI32372]
  5. Natural Sciences and Engineering Research Council of Canada [498706]
  6. NIH [1R01GM099409]
  7. US Public Health Service [R01CA63113, R01CA173023]
  8. German National Academy of Sciences Leopoldina Fellowship
  9. Office of Management, Information, and Research (OMIR) Early Researcher Award
  10. Terry Fox New Investigator Award
  11. Canadian Institutes of Health Research (CIHR) New Investigator Salary Award

Ask authors/readers for more resources

Steady-state gene expression across the cell cycle has been studied extensively. However, transcriptional gene regulation and the dynamics of histone modification at different cell-cycle stages are largely unknown. By applying a combination of global nuclear run-on sequencing (GRO-seq), RNA sequencing (RNA-seq), and histone-modification Chip sequencing (ChIP-seq), we depicted a comprehensive transcriptional landscape at the G0/G1, G1/S, and M phases of breast cancer MCF-7 cells. Importantly, GRO-seq and RNA-seq analysis identified different cell-cycle-regulated genes, suggesting a lag between transcription and steady-state expression during the cell cycle. Interestingly, we identified genes actively transcribed at early M phase that are longer in length and have low expression and are accompanied by a global increase in active histone 3 lysine 4 methylation (H3K4me2) and histone 3 lysine 27 acetylation (H3K27ac) modifications. In addition, we identified 2,440 cell-cycle-regulated enhancer RNAs (eRNAs) that are strongly associated with differential active transcription but not with stable expression levels across the cell cycle. Motif analysis of dynamic eRNAs predicted Kruppel-like factor 4 (KLF4) as a key regulator of G1/S transition, and this identification was validated experimentally. Taken together, our combined analysis characterized the transcriptional and histone-modification profile of the human cell cycle and identified dynamic transcriptional signatures across the cell cycle.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available