Abstract

Within the AGC kinase superfamily, gene fusions resulting from chromosomal rearrangements have been most frequently described for protein kinase C (PKC), with gene fragments encoding either the C-terminal catalytic domain or the N-terminal regulatory moiety fused to other genes. Kinase fusions that eliminate regulatory domains are typically gain of function and often oncogenic. However, several quality control pathways prevent accumulation of aberrant PKC, suggesting that PKC fusions may paradoxically be loss of function. To explore this topic, we used biochemical, cellular, and genome editing approaches to investigate the function of fusions that retain the portion of the gene encoding either the catalytic domain or regulatory domain of PKC. Overexpression studies revealed that PKC catalytic domain fusions were constitutively active but vulnerable to degradation. Genome editing of endogenous genes to generate a cancer-associated PKC fusion resulted in cells with detectable levels of fusion transcript but no detectable protein. Hence, PKC catalytic domain fusions are paradoxically loss of function as a result of their instability, preventing appreciable accumulation of protein in cells. Overexpression of a PKC regulatory domain fusion suppressed both basal and agonist-induced endogenous PKC activity, acting in a dominant-negative manner by competing for diacylglycerol. For both catalytic and regulatory domain fusions, the PKC component of the fusion proteins mediated the effects of the full-length fusions on the parameters examined, suggesting that the partner protein is dispensable in these contexts. Taken together, our findings reveal that PKC gene fusions are distinct from oncogenic fusions and present a mechanism by which loss of PKC function occurs in cancer.

Highlights

  • In a multitude of diseases, including metabolic disorders, neurodegeneration, and most notably, cancer [4,5,6]

  • In order to assess the landscape of protein kinase C (PKC) fusions in cancer, we queried the literature and several online databases to curate a comprehensive list of all PKC gene fusions identified from patient tumor samples (Table S1) [18, 23,24,25,26,27,28,29,30,31,32,33,34,35,36,37]

  • We addressed whether the fusion proteins exhibited catalytic activity, as assessed with the genetically encoded, fluorescence resonance energy transfer (FRET)-based reporter for PKC activity, C kinase activity reporter (CKAR) [46]

Read more

Summary

Introduction

In a multitude of diseases, including metabolic disorders, neurodegeneration, and most notably, cancer [4,5,6]. Whereas wild-type PKC was constitutively phosphorylated at all three processing sites, the full-length fusion proteins TANC2-PKCα, CTL1-PKCα, and GGA2-PKCβII were not (Fig. 2B). To further probe the basal activity of the fusion proteins, COS7 cells expressing either CKAR alone (white) or coexpressing mCherry-tagged PKC were treated with a PKC inhibitor alone (BisIV or Gӧ6983) (Fig. 2D).

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call