Trefoil Factor-3 (TFF3) Stimulates De Novo Angiogenesis in Mammary Carcinoma both Directly and Indirectly via IL-8/CXCR2

PLoS One. 2015 Nov 11;10(11):e0141947. doi: 10.1371/journal.pone.0141947. eCollection 2015.

Abstract

Mammary carcinoma cells produce pro-angiogenic factors to stimulate angiogenesis and tumor growth. Trefoil factor-3 (TFF3) is an oncogene secreted from mammary carcinoma cells and associated with poor prognosis. Herein, we demonstrate that TFF3 produced in mammary carcinoma cells functions as a promoter of tumor angiogenesis. Forced expression of TFF3 in mammary carcinoma cells promoted proliferation, survival, invasion and in vitro tubule formation of human umbilical vein endothelial cells (HUVEC). MCF7-TFF3 cells with forced expression of TFF3 generated tumors with enhanced microvessel density as compared to tumors formed by vector control cells. Depletion of TFF3 in mammary carcinoma cells by siRNA concordantly decreased the angiogenic behavior of HUVEC. Forced expression of TFF3 in mammary carcinoma cells stimulated IL-8 transcription and subsequently enhanced IL-8 expression in both mammary carcinoma cells and HUVEC. Depletion of IL-8 in mammary carcinoma cells with forced expression of TFF3, or antibody inhibition of IL-8, partially abrogated mammary carcinoma cell TFF3-stimulated HUVEC angiogenic behavior in vitro, as did inhibition of the IL-8 receptor, CXCR2. Depletion of STAT3 by siRNA in MCF-7 cells with forced expression of TFF3 partially diminished the angiogenic capability of TFF3 on stimulation of cellular processes of HUVEC. Exogenous recombinant hTFF3 also directly promoted the angiogenic behavior of HUVEC. Hence, TFF3 is a potent angiogenic factor and functions as a promoter of de novo angiogenesis in mammary carcinoma, which may co-coordinate with the growth promoting and metastatic actions of TFF3 in mammary carcinoma to enhance tumor progression.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / genetics
  • Blotting, Western
  • Breast Neoplasms / blood supply
  • Breast Neoplasms / genetics
  • Breast Neoplasms / metabolism*
  • Cell Line, Tumor
  • Cell Movement / genetics
  • Cell Proliferation / genetics
  • Cells, Cultured
  • Coculture Techniques
  • Female
  • Human Umbilical Vein Endothelial Cells / metabolism
  • Humans
  • Interleukin-8 / genetics
  • Interleukin-8 / metabolism*
  • MCF-7 Cells
  • Mice, Inbred BALB C
  • Mice, Nude
  • Microscopy, Fluorescence
  • Neovascularization, Pathologic / genetics
  • Neovascularization, Pathologic / metabolism*
  • Peptides / genetics
  • Peptides / metabolism*
  • RNA Interference
  • Receptors, Interleukin-8B / genetics
  • Receptors, Interleukin-8B / metabolism*
  • Reverse Transcriptase Polymerase Chain Reaction
  • STAT3 Transcription Factor / genetics
  • STAT3 Transcription Factor / metabolism
  • Transplantation, Heterologous
  • Trefoil Factor-3

Substances

  • Interleukin-8
  • Peptides
  • Receptors, Interleukin-8B
  • STAT3 Transcription Factor
  • TFF3 protein, human
  • Trefoil Factor-3

Grants and funding

This work was supported by the Cancer Science Institute of Singapore through grants from the Ministry of Education and the National Research Foundation of Singapore, by a grant from the National Medical Research Council of Singapore [R-713-000-163-511], the National Key Scientific Program of China (2012CB934002, 2010CB912804), National Natural Science Foundation of China (81272925, 31201022, 81302322). PEL was also supported by The Chinese Academy of Sciences President’s International Fellowship Initiative (PIFI) (Grant No. 2015VBA031).