13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Human papillomavirus type 16 E7 maintains elevated levels of the cdc25A tyrosine phosphatase during deregulation of cell cycle arrest.

      Journal of Biology
      3T3 Cells, Animals, Binding Sites, CDC2-CDC28 Kinases, Cell Cycle, Cell Cycle Proteins, Cell Differentiation, Cell Division, Cells, Cultured, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinases, metabolism, DNA-Binding Proteins, E2F Transcription Factors, Enzyme Induction, Histone Deacetylase 1, Histone Deacetylases, Humans, Keratinocytes, cytology, enzymology, virology, Mice, Oncogene Proteins, Viral, chemistry, Papillomaviridae, Papillomavirus E7 Proteins, Promoter Regions, Genetic, genetics, Protein Binding, Protein Structure, Tertiary, Protein-Serine-Threonine Kinases, RNA, Messenger, Response Elements, Retinoblastoma Protein, Transcription Factors, Transcriptional Activation, Transfection, cdc25 Phosphatases, biosynthesis

      Read this article at

      ScienceOpenPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Essential to the oncogenic properties of human papillomavirus type 16 (HPV-16) are the activities encoded by the early gene product E7. HPV-16 E7 (E7.16) binds to cellular factors involved in cell cycle regulation and differentiation. These include the retinoblastoma tumor suppressor protein (Rb) and histone deacetylase (HDAC) complexes. While the biological significance of these interactions remains unclear, E7 is believed to help maintain cells in a proliferative state, thus establishing an environment that is conducive to viral replication. Most pathways that govern cell growth converge on downstream effectors. Among these is the cdc25A tyrosine phosphatase. cdc25A is required for G(1)/S transition, and its deregulation is associated with carcinogenesis. Considering the importance of cdc25A in cell cycle progression, it represents a relevant target for viral oncoproteins. Accordingly, the present study focuses on the putative deregulation of cdc25A by E7.16. Our results indicate that E7.16 can impede growth arrest induced during serum starvation and keratinocyte differentiation. Importantly, these E7-specific phenotypes correlate with elevated cdc25A steady-state levels. Reporter assays performed with NIH 3T3 cell lines and human keratinocytes indicate that E7 can transactivate the cdc25A promoter. In addition, transcriptional activation by E7.16 requires the distal E2F site within the cdc25A promoter. We further demonstrate that the ability of E7 to abrogate cell cycle arrest, activate cdc25A transcription, and increase cdc25A protein levels requires intact Rb and HDAC-1 binding domains. Finally, by using the cdk inhibitor roscovitine, we reveal that E7 activates the cdc25A promoter independently of cell cycle progression and cdk activity. Consequently, we propose that E7.16 can directly target cdc25A transcription and maintains cdc25A gene expression by disrupting Rb/E2F/HDAC-1 repressor complexes.

          Related collections

          Author and article information

          Comments

          Comment on this article