Oncotarget

Research Papers:

The roles of RRP15 in nucleolar formation, ribosome biogenesis and checkpoint control in human cells

Zhixiong Dong, Changjun Zhu, Qimin Zhan and Wei Jiang _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:13240-13252. https://doi.org/10.18632/oncotarget.14658

Metrics: PDF 2300 views  |   HTML 2556 views  |   ?  


Abstract

Zhixiong Dong1,2, Changjun Zhu1,2, Qimin Zhan3, Wei Jiang1,3

1Key Laboratory of Molecular and Cellular Systems Biology, Tianjin Normal University, Tianjin 300387, China

2Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin 300387, China

3State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China

Correspondence to:

Wei Jiang, email: [email protected]

Changjun Zhu, email: [email protected]

Keywords: RRP15, nucleolus, ribosome biogenesis, nucleolar stress, checkpoint control

Received: June 07, 2016     Accepted: December 27, 2016     Published: January 14, 2017

ABSTRACT

The nucleolus controls ribosome biogenesis and its perturbation induces nucleolar stress that inhibits cell cycle progression and activates checkpoint responses. Here, we investigate the roles of ribosomal RNA processing protein, RRP15, in nucleolar formation, ribosome biogenesis, cell cycle progression and checkpoint control in human cells. RRP15 is localized in the nucleolus and required for nucleolar formation. In contrast to the budding yeast Rrp15p that was reported as a component of pre-60S subunits, RRP15 is found in both pre-40S and pre-60S subunits and involved in regulating rRNA transcription and ribosome biogenesis. Perturbation of RRP15 induces nucleolar stress that activates RPL5/RPL11/5S rRNA (RP)-Mdm2-p53 axis checkpoint response and arrests cells at G1-G1/S in p53-proficient non-transformed RPE1 cells but not in p53-deficient HeLa and MCF7 tumor cells. Instead, p53-deficient HeLa and MCF7 cells with RRP15-dependent nucleolar stress enter S-phase with S-phase perturbation that activates ATR-Chk1- γH2AX axis DNA replication/damage checkpoint response, delaying S-G2/M progression and, ultimately, causing cell death. The selective checkpoint response, cell cycle inhibition and/or cytotoxicity induced by RRP15-dependent nucleolar stress in p53-proficient non-transformed cells and p53-deficient tumor cells suggest that RRP15 might be a potential target for cancer therapy.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 14658