Oncotarget

Priority Research Papers:

AP4 is required for mitogen- and c-MYC-induced cell cycle progression

Rene Jackstadt _ and Heiko Hermeking

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2014; 5:7316-7327. https://doi.org/10.18632/oncotarget.2348

Metrics: PDF 2640 views  |   HTML 2946 views  |   ?  


Abstract

Rene Jackstadt1 and Heiko Hermeking1,2,3

1 Experimental and Molecular Pathology, Institute of Pathology, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, D-80337 Munich, Germany

2 German Cancer Consortium (DKTK), D-69120 Heidelberg, Germany

3 German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany

Correspondence to:

Prof. Heiko Hermeking, e-mail: [email protected]

Keywords: AP4, AP-4, TFAP4, c-MYC, cell cycle, cytokinesis

Received: August 8, 2014     Revised and Accepted: August 11, 2014     Published: August 19, 2014

ABSTRACT

AP4 represents a c-MYC-inducible bHLH-LZ transcription factor, which displays elevated expression in many types of tumors. We found that serum-starved AP4-deficient mouse embryo fibroblasts (MEFs) were unable to resume proliferation and showed a delayed S-phase entry after restimulation. Furthermore, they accumulated as tetraploid cells due to a cytokinesis defect. In addition, AP4 was required for c-MYC-induced cell cycle re-entry. AP4-deficient MEFs displayed decreased expression of CDK2 (cyclin-dependent kinase 2), which we characterized as a conserved and direct AP4 target. Activation of an AP4 estrogen receptor fusion protein (AP4-ER) enhanced proliferation of human diploid fibroblasts in a CDK2-dependent manner. However, in contrast to c-MYC-ER, AP4-ER activation was not sufficient to induce cell cycle re-entry or apoptosis in serum-starved MEFs. AP4-deficiency was accompanied by increased spontaneous and c-MYC-induced DNA damage in MEFs. Furthermore, c-MYC-induced apoptosis was decreased in AP4-deficient MEFs, suggesting that induction of apoptosis by c-MYC is linked to its ability to activate AP4 and thereby cell cycle progression. Taken together, these results indicate that AP4 is a central mediator and coordinator of cell cycle progression in response to mitogenic signals and c-MYC activation. Therefore, inhibition of AP4 function may represent a therapeutic approach to block tumor cell proliferation.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 2348