Your browser doesn't support javascript.
loading
P-REX1-Independent, Calcium-Dependent RAC1 Hyperactivation in Prostate Cancer.
Baker, Martin J; Abba, Martín C; Garcia-Mata, Rafael; Kazanietz, Marcelo G.
Affiliation
  • Baker MJ; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, 1256 Biomedical Research Building II/III, 421 Curie Blvd., Philadelphia, PA 19104-6160, USA.
  • Abba MC; Centro de Investigaciones Inmunológicas Básicas y Aplicadas, Universidad Nacional de La Plata, La Plata CP1900, Argentina.
  • Garcia-Mata R; Department of Biological Sciences, The University of Toledo, Toledo, OH 43606, USA.
  • Kazanietz MG; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, 1256 Biomedical Research Building II/III, 421 Curie Blvd., Philadelphia, PA 19104-6160, USA.
Cancers (Basel) ; 12(2)2020 Feb 19.
Article in En | MEDLINE | ID: mdl-32092966
ABSTRACT
The GTPase Rac1 is a well-established master regulator of cell motility and invasiveness contributing to cancer metastasis. Dysregulation of the Rac1 signaling pathway, resulting in elevated motile and invasive potential, has been reported in multiple cancers. However, there are limited studies on the regulation of Rac1 in prostate cancer. Here, we demonstrate that aggressive androgen-independent prostate cancer cells display marked hyperactivation of Rac1. This hyperactivation is independent of P-Rex1 activity or its direct activators, the PI3K product PIP3 and Gßγ subunits. Furthermore, we demonstrate that the motility and invasiveness of PC3 prostate cancer cells is independent of P-Rex1, supporting the analysis of publicly available datasets indicating no correlation between high P-Rex1 expression and cancer progression in patients. Rac1 hyperactivation was not related to the presence of activating Rac1 mutations and was insensitive to overexpression of a Rac-GAP or the silencing of specific Rac-GEFs expressed in prostate cancer cells. Interestingly, active Rac1 levels in these cells were markedly reduced by elevations in intracellular calcium or by serum stimulation, suggesting the presence of an alternative means of Rac1 regulation in prostate cancer that does not involve previously established paradigms.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Cancers (Basel) Year: 2020 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Cancers (Basel) Year: 2020 Document type: Article Affiliation country: United States
...