Oncotarget

Research Papers:

A Phosphatidic Acid (PA) conveyor system of continuous intracellular transport from cell membrane to nucleus maintains EGF receptor homeostasis

Karen M. Henkels, Taylor E. Miller, Ramya Ganesan, Brandon A. Wilkins, Kristen Fite and Julian Gomez-Cambronero _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:47002-47017. https://doi.org/10.18632/oncotarget.9685

Metrics: PDF 2213 views  |   HTML 2475 views  |   ?  


Abstract

Karen M. Henkels2, Taylor E. Miller2, Ramya Ganesan2, Brandon A. Wilkins2, Kristen Fite2 and Julian Gomez-Cambronero1,2

1 Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women Hospital and Harvard Medical School, Boston, MA, USA

2 Department of Biochemistry and Molecular Biology, Wright State University Boonshoft School of Medicine, Dayton, Ohio, USA

Correspondence to:

Julian Gomez-Cambronero, email:

Keywords: mammalian cells, breast cancer, gene expression, cell signaling, phospholipids

Received: April 14, 2016 Accepted: May 14, 2016 Published: May 31, 2016

Abstract

The intracellular concentration of the mitogen phosphatidic acid (PA) must be maintained at low levels until the need arises for cell proliferation. How temporal and spatial trafficking of PA affects its target proteins in the different cellular compartments is not fully understood. We report that in cancer cells, PA cycles back and forth from the cellular membrane to the nucleus, affecting the function of epidermal growth factor (EGF), in a process that involves PPARα/LXRα signaling. Upon binding to its ligand, EGF receptor (EGFR)-initiated activation of phospholipase D (PLD) causes a spike in intracellular PA production that forms vesicles transporting EGFR from early endosomes (EEA1 marker) and prolonged internalization in late endosomes and Golgi (RCAS marker). Cells incubated with fluorescent-labeled PA (NBD-PA) show PA in “diffuse” locations throughout the cytoplasm, punctae (small, <0.1 μm) vesicles) and large (>0.5 μm) vesicles that co-localize with EGFR. We also report that PPARα/LXRα form heterodimers that bind to new Responsive Elements (RE) in the EGFR promoter. Nuclear PA enhances EGFR expression, a role compatible with the mitogenic ability of the phospholipid. Newly made EGFR is packaged into PA recycling vesicles (Rab11 marker) and transported back to the cytoplasm and plasma membrane. However, a PLD+PA combination impedes binding of PPARα/LXRα to the EGFR promoter. Thus, if PA levels inside the nucleus reach a certain threshold (>100 nM) PA outcompetes the nuclear receptors and transcription is inhibited. This new signaling function of PLD-PA targeting EGFR trafficking and biphasically modulating its transcription, could explain cell proliferation initiation and its maintenance in cancer cells.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 9685