Oncotarget

Research Papers:

Centrosome aberrations in human mammary epithelial cells driven by cooperative interactions between p16INK4a deficiency and telomere-dependent genotoxic stress

Daniel Domínguez, Purificación Feijoo, Aina Bernal, Amaia Ercilla, Neus Agell, Anna Genescà and Laura Tusell _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:28238-28256. https://doi.org/10.18632/oncotarget.4958

Metrics: PDF 1272 views  |   HTML 2341 views  |   ?  


Abstract

Daniel Domínguez1, Purificación Feijoo1, Aina Bernal1, Amaia Ercilla2, Neus Agell2, Anna Genescà1 and Laura Tusell1

1 Cell Biology Unit, Department of Cell Biology, Physiology and Immunology, Bioscience School, Universitat Autònoma de Barcelona, Bellaterra, Spain

2 Departament de Biologia Cellular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, IDIBAPS, Barcelona, Spain

Correspondence to:

Laura Tusell, email:

Keywords: centrosome aberrations, CDKN2A, human mammary epithelial cells, telomere dysfunction

Received: November 07, 2014 Accepted: July 07, 2015 Published: July 22, 2015

Abstract

Virtually all human cancers display chromosome instability (CIN), a condition in which chromosomes are gained or lost at a high rate. CIN occurs early in cancer development where it may undermine the advance of the neoplastic disease. With the aim of establishing the mechanisms underlying CIN in cancer, we investigated possible links between telomere-dysfunction and centrosome defects, which were seen to coincide in early in breast carcinogenesis using human mammary epithelial cells (HMECs). In this study, we show that TP53 proficient vHMECs cells develop centrosome aberrations when telomere-dysfunction genotoxic stress is produced in the presence of a defective p16INK4a setting and in parallel with an activation of the DNA damage checkpoint response. These aberrations consist of the accumulation of centrosomes in polyploid vHMECs, plus centriole overduplication in both diploid and polyploid cells, thus reflecting that distinct mechanisms underlie the generation of centrosome aberrations in vHMECs. Transduction of vHMEC with hTERT, which rescued the telomere dysfunction phenotype and consequently reduced DNA damage checkpoint activation, led to a progressive reduction of centrosome aberrations with cell culture, both in diploid and in polyploid vHMECs. Radiation-induced DNA damage also raised centrosome aberrations in vHMEC-hTERT. Collectively, our results, using vHMECs define a model where p16INK4a deficiency along with short dysfunctional telomeres cooperatively engenders centrosome abnormalities before p53 function is compromised.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 4958