Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality

Szmyd, Radoslaw ; Niska-Blakie, Joanna ; Diril, M Kasim ; Renck Nunes, Patrícia ; Tzelepis, Konstantinos ; Lacroix, Aurélie ; van Hul, Noémi ; Deng, Lih-Wen ; Matos, Joao and Dreesen, Oliver , et al. (2019) In Oncogene 38(7). p.998-1018
Abstract

Cell cycle regulation, especially faithful DNA replication and mitosis, are crucial to maintain genome stability. Cyclin-dependent kinase (CDK)/cyclin complexes drive most processes in cellular proliferation. In response to DNA damage, cell cycle surveillance mechanisms enable normal cells to arrest and undergo repair processes. Perturbations in genomic stability can lead to tumor development and suggest that cell cycle regulators could be effective targets in anticancer therapy. However, many clinical trials ended in failure due to off-target effects of the inhibitors used. Here, we investigate in vivo the importance of WEE1- and MYT1-dependent inhibitory phosphorylation of mammalian CDK1. We generated Cdk1AF knockin mice, in which two... (More)

Cell cycle regulation, especially faithful DNA replication and mitosis, are crucial to maintain genome stability. Cyclin-dependent kinase (CDK)/cyclin complexes drive most processes in cellular proliferation. In response to DNA damage, cell cycle surveillance mechanisms enable normal cells to arrest and undergo repair processes. Perturbations in genomic stability can lead to tumor development and suggest that cell cycle regulators could be effective targets in anticancer therapy. However, many clinical trials ended in failure due to off-target effects of the inhibitors used. Here, we investigate in vivo the importance of WEE1- and MYT1-dependent inhibitory phosphorylation of mammalian CDK1. We generated Cdk1AF knockin mice, in which two inhibitory phosphorylation sites are replaced by the non-phosphorylatable amino acids T14A/Y15F. We uncovered that monoallelic expression of CDK1AF is early embryonic lethal in mice and induces S phase arrest accompanied by γH2AX and DNA damage checkpoint activation in mouse embryonic fibroblasts (MEFs). The chromosomal fragmentation in Cdk1AF MEFs does not rely on CDK2 and is partly caused by premature activation of MUS81-SLX4 structure-specific endonuclease complexes, as well as untimely onset of chromosome condensation followed by nuclear lamina disassembly. We provide evidence that tumor development in liver expressing CDK1AF is inhibited. Interestingly, the regulatory mechanisms that impede cell proliferation in CDK1AF expressing cells differ partially from the actions of the WEE1 inhibitor, MK-1775, with p53 expression determining the sensitivity of cells to the drug response. Thus, our work highlights the importance of improved therapeutic strategies for patients with various cancer types and may explain why some patients respond better to WEE1 inhibitors.

(Less)
Please use this url to cite or link to this publication:
@article{aaee7ae0-1a1d-4320-bbdf-b247bc7dcf50,
  abstract     = {{<p>Cell cycle regulation, especially faithful DNA replication and mitosis, are crucial to maintain genome stability. Cyclin-dependent kinase (CDK)/cyclin complexes drive most processes in cellular proliferation. In response to DNA damage, cell cycle surveillance mechanisms enable normal cells to arrest and undergo repair processes. Perturbations in genomic stability can lead to tumor development and suggest that cell cycle regulators could be effective targets in anticancer therapy. However, many clinical trials ended in failure due to off-target effects of the inhibitors used. Here, we investigate in vivo the importance of WEE1- and MYT1-dependent inhibitory phosphorylation of mammalian CDK1. We generated Cdk1AF knockin mice, in which two inhibitory phosphorylation sites are replaced by the non-phosphorylatable amino acids T14A/Y15F. We uncovered that monoallelic expression of CDK1AF is early embryonic lethal in mice and induces S phase arrest accompanied by γH2AX and DNA damage checkpoint activation in mouse embryonic fibroblasts (MEFs). The chromosomal fragmentation in Cdk1AF MEFs does not rely on CDK2 and is partly caused by premature activation of MUS81-SLX4 structure-specific endonuclease complexes, as well as untimely onset of chromosome condensation followed by nuclear lamina disassembly. We provide evidence that tumor development in liver expressing CDK1AF is inhibited. Interestingly, the regulatory mechanisms that impede cell proliferation in CDK1AF expressing cells differ partially from the actions of the WEE1 inhibitor, MK-1775, with p53 expression determining the sensitivity of cells to the drug response. Thus, our work highlights the importance of improved therapeutic strategies for patients with various cancer types and may explain why some patients respond better to WEE1 inhibitors.</p>}},
  author       = {{Szmyd, Radoslaw and Niska-Blakie, Joanna and Diril, M Kasim and Renck Nunes, Patrícia and Tzelepis, Konstantinos and Lacroix, Aurélie and van Hul, Noémi and Deng, Lih-Wen and Matos, Joao and Dreesen, Oliver and Bisteau, Xavier and Kaldis, Philipp}},
  issn         = {{1476-5594}},
  keywords     = {{Amino Acid Substitution; Animals; CDC2 Protein Kinase/genetics; Cell Cycle Proteins/genetics; DNA-Binding Proteins/genetics; Embryo Loss/enzymology; Embryo, Mammalian/enzymology; Enzyme Activation; Mice; Mice, Transgenic; Mitosis; Mutation, Missense; Nuclear Proteins/genetics; Protein-Tyrosine Kinases/genetics; S Phase; Transcription Factors/genetics}},
  language     = {{eng}},
  number       = {{7}},
  pages        = {{998--1018}},
  publisher    = {{Nature Publishing Group}},
  series       = {{Oncogene}},
  title        = {{Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality}},
  url          = {{http://dx.doi.org/10.1038/s41388-018-0464-0}},
  doi          = {{10.1038/s41388-018-0464-0}},
  volume       = {{38}},
  year         = {{2019}},
}