Pancreatic cell plasticity and cancer initiation induced by oncogenic Kras is completely dependent on wild-type PI 3-kinase p110α

  1. Julie Guillermet-Guibert1,2,3
  1. 1UMR1037, Le Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm, F-31000 Toulouse, France;
  2. 2UMR1037, CRCT, Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France;
  3. 3Equipe Labellisée Ligue Contre le Cancer, F-31000 Toulouse, France;
  4. 4Department of Internal Medicine 2, Technische Universität München, 81675 Munich, Germany;
  5. 5UMR1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Inserm, F-31000 Toulouse, France; Université Toulouse III-Paul Sabatier, F-31000 Toulouse, France;
  6. 6Cell Signaling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom;
  7. 7German Cancer Consortium (DKTK), 69120 Heidelberg, Germany;
  8. 8German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
  1. Corresponding author: julie.guillermet{at}inserm.fr

Abstract

Increased PI 3-kinase (PI3K) signaling in pancreatic ductal adenocarcinoma (PDAC) correlates with poor prognosis, but the role of class I PI3K isoforms during its induction remains unclear. Using genetically engineered mice and pharmacological isoform-selective inhibitors, we found that the p110α PI3K isoform is a major signaling enzyme for PDAC development induced by a combination of genetic and nongenetic factors. Inactivation of this single isoform blocked the irreversible transition of exocrine acinar cells into pancreatic preneoplastic ductal lesions by oncogenic Kras and/or pancreatic injury. Hitting the other ubiquitous isoform, p110β, did not prevent preneoplastic lesion initiation. p110α signaling through small GTPase Rho and actin cytoskeleton controls the reprogramming of acinar cells and regulates cell morphology in vivo and in vitro. Finally, p110α was necessary for pancreatic ductal cancers to arise from Kras-induced preneoplastic lesions by increasing epithelial cell proliferation in the context of mutated p53. Here we identify an in vivo context in which p110α cellular output differs depending on the epithelial transformation stage and demonstrate that the PI3K p110α is required for PDAC induced by oncogenic Kras, the key driver mutation of PDAC. These data are critical for a better understanding of the development of this lethal disease that is currently without efficient treatment.

Keywords

Footnotes

  • Received July 25, 2014.
  • Accepted October 27, 2014.

This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents

Life Science Alliance