Human Papillomavirus 16 (HPV-16), HPV-18, and HPV-31 E6 Override the Normal Phosphoregulation of E6AP Enzymatic Activity

J Virol. 2017 Oct 27;91(22):e01390-17. doi: 10.1128/JVI.01390-17. Print 2017 Nov 15.

Abstract

The human papillomavirus (HPV) E6 oncoproteins recruit the cellular ubiquitin ligase E6AP/UBE3A to target cellular substrates for proteasome-mediated degradation, and one consequence of this activity is the E6 stimulation of E6AP autoubiquitination and degradation. Recent studies identified an autism-linked mutation within E6AP at T485, which was identified as a protein kinase A phosphoacceptor site and which could directly regulate E6AP ubiquitin ligase activity. In this study, we have analyzed how T485-mediated regulation of E6AP might affect E6 targeting of some of its known substrates. We show that modulation of T485 has no effect on the ability of E6 to direct either p53 or Dlg for degradation. Furthermore, T485 regulation has no effect on HPV-16 or HPV-31 E6-induced autodegradation of E6AP but does affect HPV-18 E6-induced autodegradation of E6AP. In cells derived from cervical cancers, we find low levels of both phosphorylated and nonphosphorylated E6AP in the nucleus. However, ablation of E6 results in a dramatic accumulation of phospho-E6AP in the cytoplasm, whereas nonphosphorylated E6AP accumulates primarily in the nucleus. Interestingly, E6AP phosphorylation at T485 confers association with 14-3-3 proteins, and this interaction seems to be important, in part, for the ability of E6 to recruit phospho-E6AP into the nucleus. These results demonstrate that HPV E6 overrides the normal phosphoregulation of E6AP, both in terms of its enzymatic activity and its subcellular distribution.IMPORTANCE Recent reports demonstrate the importance of phosphoregulation of E6AP for its normal enzymatic activity. Here, we show that HPV E6 is capable of overriding this regulation and can promote degradation of p53 and Dlg regardless of the phosphorylation status of E6AP. Furthermore, E6 interaction with E6AP also significantly alters how E6AP is subject to autodegradation and suggests that this is not a simple stimulation of an already-existing activity but rather a redirection of E6AP activity toward itself. Furthermore, E6-mediated regulation of the subcellular distribution of phospho-E6AP appears to be dependent, in part, upon the 14-3-3 family of proteins.

Keywords: E6AP; HPV E6; phosphorylation.

MeSH terms

  • Adaptor Proteins, Signal Transducing / genetics
  • Adaptor Proteins, Signal Transducing / metabolism
  • Cell Nucleus / genetics
  • Cell Nucleus / metabolism*
  • Cell Nucleus / virology
  • Cytoplasm / genetics
  • Cytoplasm / metabolism*
  • Cytoplasm / virology
  • DNA-Binding Proteins / genetics
  • DNA-Binding Proteins / metabolism*
  • Discs Large Homolog 1 Protein
  • HEK293 Cells
  • Human papillomavirus 16 / genetics
  • Human papillomavirus 16 / metabolism*
  • Human papillomavirus 18 / genetics
  • Human papillomavirus 18 / metabolism*
  • Human papillomavirus 31 / genetics
  • Human papillomavirus 31 / metabolism*
  • Humans
  • Membrane Proteins / genetics
  • Membrane Proteins / metabolism
  • Oncogene Proteins, Viral / genetics
  • Oncogene Proteins, Viral / metabolism*
  • Phosphorylation
  • Protein Transport
  • Proteolysis
  • Repressor Proteins / genetics
  • Repressor Proteins / metabolism*
  • Tumor Suppressor Protein p53 / genetics
  • Tumor Suppressor Protein p53 / metabolism

Substances

  • Adaptor Proteins, Signal Transducing
  • DLG1 protein, human
  • DNA-Binding Proteins
  • Discs Large Homolog 1 Protein
  • E6 protein, Human papillomavirus type 16
  • E6 protein, Human papillomavirus type 18
  • E6 protein, Human papillomavirus type 31
  • Membrane Proteins
  • Oncogene Proteins, Viral
  • Repressor Proteins
  • TP53 protein, human
  • Tumor Suppressor Protein p53