The bacterial protein azurin impairs invasion and FAK/Src signaling in P-cadherin-overexpressing breast cancer cell models

PLoS One. 2013 Jul 19;8(7):e69023. doi: 10.1371/journal.pone.0069023. Print 2013.

Abstract

P-cadherin overexpression occurs in about 30% of all breast carcinomas, being a poor prognostic factor for breast cancer patients. In a cellular background of wild-type E-cadherin, we have previously shown that its expression promotes invasion, motility and migration of breast cancer cells due to the induced secretion of metalloproteases (MMPs) to the extracellular medium and to the concomitant shedding of a pro-invasive soluble form of this protein (sP-cad). Azurin is secreted by Pseudomonas aeruginosa and induces in vitro and in vivo cytotoxicity after its preferential penetration in human cancer cells relative to normal cells. Three different breast cancer cell lines, MCF-7/AZ.Mock, MCF-7/AZ.Pcad and SUM149 were treated with sub-killing doses of azurin. Invasion of these cells was measured using Matrigel Invasion Assays and MTT assays were performed to determine cell viability upon treatment and the effects on cadherins expression was determined by Western blot and Immunofluorescence. Gelatin Zymography was used to determine activity of MMP2 in the conditioned media of azurin treated and untreated cells and the phosphorylation levels of intracellular signaling proteins were determined by Western blot. The invasive phenotype of these breast cancer cells was significantly reduced by azurin. Azurin (50-100 µM) also caused a specific decrease on P-cadherin protein levels from 30-50% in MCF-7/AZ.Pcad and SUM149 breast cancer cell lines, but the levels of E-cadherin remain unaltered. More, the levels of sP-cad and the activity of MMP2 were reduced in the extracellular media of azurin treated cells and we also observed a decrease in the phosphorylation levels of both FAK and Src proteins. Our data show that azurin specifically targets P-cadherin, not E-cadherin, abrogating P-cadherin-mediated invasive effects and signaling. Therefore, azurin could possibly be considered a therapeutic tool to treat poor-prognosis breast carcinomas overexpressing P-cadherin in a wild type E-cadherin context.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Azurin / pharmacology*
  • Bacterial Proteins / pharmacology
  • Breast Neoplasms / genetics*
  • Breast Neoplasms / metabolism*
  • Breast Neoplasms / pathology
  • Cadherins / genetics*
  • Cadherins / metabolism
  • Cell Line, Tumor
  • Cell Movement / drug effects
  • Cell Survival / drug effects
  • Enzyme Activation / drug effects
  • Female
  • Focal Adhesion Kinase 1 / metabolism*
  • Gene Expression*
  • Humans
  • MCF-7 Cells
  • Matrix Metalloproteinase 2 / metabolism
  • Signal Transduction / drug effects*
  • src-Family Kinases / metabolism*

Substances

  • Bacterial Proteins
  • Cadherins
  • Azurin
  • Focal Adhesion Kinase 1
  • src-Family Kinases
  • Matrix Metalloproteinase 2

Grants and funding

The work presented was supported by a scientific project (PTDC/EBBBIO/100326/2008) financed by the Portuguese Science and Technology Foundation (FCT). FCT also provides research grants as follows: PhD research grant for Nuno Bernardes (SFRH/BD/48763/2008) and post-doc research grant to Ana Sofia Ribeiro (SFRH/BPD/75705/2011) and Programa Ciência 2007 for Joana Paredes [POPH - QREN - Tipology 4.2]. IPATIMUP is an Associate Laboratory of the Portuguese Ministry of Science, Technology and Higher Education and is partially supported by FCT. Work at ITQB was supported by grant PEst-OE/EQB/LA0004/2011, also from FCT. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.