Uncoupling of the LKB1-AMPKalpha energy sensor pathway by growth factors and oncogenic BRAF

PLoS One. 2009;4(3):e4771. doi: 10.1371/journal.pone.0004771. Epub 2009 Mar 10.

Abstract

Background: Understanding the biochemical mechanisms contributing to melanoma development and progression is critical for therapeutical intervention. LKB1 is a multi-task Ser/Thr kinase that phosphorylates AMPK controlling cell growth and apoptosis under metabolic stress conditions. Additionally, LKB1(Ser428) becomes phosphorylated in a RAS-Erk1/2-p90(RSK) pathway dependent manner. However, the connection between the RAS pathway and LKB1 is mostly unknown.

Methodology/principal findings: Using the UV induced HGF transgenic mouse melanoma model to investigate the interplay among HGF signaling, RAS pathway and PI3K pathway in melanoma, we identified LKB1 as a protein directly modified by HGF induced signaling. A variety of molecular techniques and tissue culture revealed that LKB1(Ser428) (Ser431 in the mouse) is constitutively phosphorylated in BRAF(V600E) mutant melanoma cell lines and spontaneous mouse tumors with high RAS pathway activity. Interestingly, BRAF(V600E) mutant melanoma cells showed a very limited response to metabolic stress mediated by the LKB1-AMPK-mTOR pathway. Here we show for the first time that RAS pathway activation including BRAF(V600E) mutation promotes the uncoupling of AMPK from LKB1 by a mechanism that appears to be independent of LKB1(Ser428) phosphorylation. Notably, the inhibition of the RAS pathway in BRAF(V600E) mutant melanoma cells recovered the complex formation and rescued the LKB1-AMPKalpha metabolic stress-induced response, increasing apoptosis in cooperation with the pro-apoptotic proteins Bad and Bim, and the down-regulation of Mcl-1.

Conclusions/significance: These data demonstrate that growth factor treatment and in particular oncogenic BRAF(V600E) induces the uncoupling of LKB1-AMPKalpha complexes providing at the same time a possible mechanism in cell proliferation that engages cell growth and cell division in response to mitogenic stimuli and resistance to low energy conditions in tumor cells. Importantly, this mechanism reveals a new level for therapeutical intervention particularly relevant in tumors harboring a deregulated RAS-Erk1/2 pathway.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • AMP-Activated Protein Kinases / drug effects
  • AMP-Activated Protein Kinases / metabolism*
  • Animals
  • Apoptosis
  • Apoptosis Regulatory Proteins / genetics
  • Hepatocyte Growth Factor / pharmacology*
  • Intercellular Signaling Peptides and Proteins / pharmacology
  • Melanoma, Experimental / pathology
  • Mice
  • Mice, Transgenic
  • Mutation, Missense
  • Phosphatidylinositol 3-Kinases / metabolism
  • Phosphorylation
  • Protein Serine-Threonine Kinases / metabolism*
  • Proto-Oncogene Proteins B-raf / genetics
  • Proto-Oncogene Proteins B-raf / physiology*
  • Signal Transduction / drug effects*
  • ras Proteins / metabolism

Substances

  • Apoptosis Regulatory Proteins
  • Intercellular Signaling Peptides and Proteins
  • Hepatocyte Growth Factor
  • Braf protein, mouse
  • Protein Serine-Threonine Kinases
  • Proto-Oncogene Proteins B-raf
  • Stk11 protein, mouse
  • AMP-Activated Protein Kinases
  • ras Proteins