Multiple delivery of siRNA against endoglin into murine mammary adenocarcinoma prevents angiogenesis and delays tumor growth

PLoS One. 2013 Mar 5;8(3):e58723. doi: 10.1371/journal.pone.0058723. Print 2013.

Abstract

Endoglin is a transforming growth factor-β (TGF- β) co-receptor that participates in the activation of a signaling pathway that mediates endothelial cell proliferation and migration in angiogenic tumor vasculature. Therefore, silencing of endoglin expression is an attractive approach for antiangiogenic therapy of tumors. The aim of our study was to evaluate the therapeutic potential of small interfering RNA (siRNA) molecules against endoglin in vitro and in vivo. Therapeutic potential in vitro was assessed in human and murine endothelial cells (HMEC-1, 2H11) by determining endoglin expression level, cell proliferation and tube formation. In vivo, the therapeutic potential of siRNA molecules was evaluated in TS/A mammary adenocarcinoma growing in BALB/c mice. Results of our study showed that siRNA molecules against endoglin have a good antiangiogenic therapeutic potential in vitro, as expression of endoglin mRNA and protein levels in mouse and human microvascular endothelial cells after lipofection were efficiently reduced, which resulted in the inhibition of endothelial cell proliferation and tube formation. In vivo, silencing of endoglin with triple electrotransfer of siRNA molecules into TS/A mammary adenocarcinoma also significantly reduced the mRNA levels, number of tumor blood vessels and the growth of tumors. The obtained results demonstrate that silencing of endoglin is a promising antiangiogenic therapy of tumors that could not be used as single treatment, but as an adjunct to the established cytotoxic treatment approaches.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenocarcinoma / blood supply
  • Adenocarcinoma / pathology
  • Adenocarcinoma / therapy*
  • Animals
  • Cell Line, Tumor
  • Cell Proliferation
  • Electroporation
  • Endoglin
  • Endothelial Cells / metabolism
  • Female
  • Gene Expression
  • Humans
  • Intracellular Signaling Peptides and Proteins / genetics*
  • Intracellular Signaling Peptides and Proteins / metabolism
  • Mammary Neoplasms, Experimental / blood supply
  • Mammary Neoplasms, Experimental / pathology
  • Mammary Neoplasms, Experimental / therapy*
  • Mice
  • Mice, Inbred BALB C
  • Neoplasm Transplantation
  • Neovascularization, Pathologic / genetics
  • Neovascularization, Pathologic / prevention & control
  • RNA Interference
  • RNA, Small Interfering / genetics*
  • Transfection
  • Tumor Burden

Substances

  • Endoglin
  • Eng protein, mouse
  • Intracellular Signaling Peptides and Proteins
  • RNA, Small Interfering

Grants and funding

The authors acknowledge the financial support from the state budget by the Slovenian Research Agency (program no. P3-0003, project no. J3-4211). Research was conducted in the scope of EBAM European Associated Laboratory (LEA). This manuscript is a result of the networking efforts of the COST Action TD1104. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.