GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.

<h4>Background</h4>TGFß overproduction in cancer cells is one of the main characteristics of late tumor progression being implicated in metastasis, tumor growth, angiogenesis and immune response. We investigated the therapeutic efficacy of anti-TGFß peptides in the control of angiogenesi...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Francesca Margheri, Nicola Schiavone, Laura Papucci, Lucia Magnelli, Simona Serratì, Anastasia Chillà, Anna Laurenzana, Francesca Bianchini, Lido Calorini, Eugenio Torre, Javier Dotor, Esperanza Feijoo, Gabriella Fibbi, Mario Del Rosso
Formato: article
Lenguaje:EN
Publicado: Public Library of Science (PLoS) 2012
Materias:
R
Q
Acceso en línea:https://doaj.org/article/9be95842e3a64f8a9bd8cf6d6c21ae83
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:9be95842e3a64f8a9bd8cf6d6c21ae83
record_format dspace
spelling oai:doaj.org-article:9be95842e3a64f8a9bd8cf6d6c21ae832021-11-18T08:06:53ZGDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.1932-620310.1371/journal.pone.0050342https://doaj.org/article/9be95842e3a64f8a9bd8cf6d6c21ae832012-01-01T00:00:00Zhttps://www.ncbi.nlm.nih.gov/pmc/articles/pmid/23226264/?tool=EBIhttps://doaj.org/toc/1932-6203<h4>Background</h4>TGFß overproduction in cancer cells is one of the main characteristics of late tumor progression being implicated in metastasis, tumor growth, angiogenesis and immune response. We investigated the therapeutic efficacy of anti-TGFß peptides in the control of angiogenesis elicited by conditional over-expression of TGFß.<h4>Methods</h4>We have inserted in human MCF7 mammary-cancer cells a mutated TGFß gene in a tetracycline-repressible vector to obtain conditional expression of mature TGFß upon transient transfection, evaluated the signaling pathways involved in TGFß-dependent endothelial cells activation and the efficacy of anti-TGFß peptides in the control of MCF7-TGFß-dependent angiogenesis.<h4>Results</h4>TGFß over-expression induced in MCF7 several markers of the epithelial-to-mesenchymal transition. Conditioned-medium of TGFß-transfected MCF7 stimulated angiogenesis in vivo and in vitro by subsequent activation of SMAD2/3 and SMAD1/5 signaling in endothelial cells, as well as SMAD4 nuclear translocation, resulting in over-expression of the pro-angiogenic growth and differentiation factor-5 (GDF5). Inhibition or silencing of GDF5 in TGFß-stimulated EC resulted in impairment of GDF5 expression and of TGFß-dependent urokinase-plasminogen activator receptor (uPAR) overproduction, leading to angiogenesis impairment. Two different TGFß antagonist peptides inhibited all the angiogenesis-related properties elicited in EC by exogenous and conditionally-expressed TGFß in vivo and in vitro, including SMAD1/5 phosphorylation, SMAD4 nuclear translocation, GDF5 and uPAR overexpression. Antagonist peptides and anti-GDF5 antibodies efficiently inhibited in vitro and in vivo angiogenesis.<h4>Conclusions</h4>TGFß produced by breast cancer cells induces in endothelial cells expression of GDF5, which in turn stimulates angiogenesis both in vitro and in vivo. Angiogenesis activation is rapid and the involved mechanism is totally opposed to the old and controversial dogma about the AKL5/ALK1 balance. The GDF-dependent pro-angiogenic effects of TGFß are controlled by anti-TGFß peptides and anti-GDF5 antibodies, providing a basis to develop targeted clinical studies.Francesca MargheriNicola SchiavoneLaura PapucciLucia MagnelliSimona SerratìAnastasia ChillàAnna LaurenzanaFrancesca BianchiniLido CaloriniEugenio TorreJavier DotorEsperanza FeijooGabriella FibbiMario Del RossoPublic Library of Science (PLoS)articleMedicineRScienceQENPLoS ONE, Vol 7, Iss 11, p e50342 (2012)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
Francesca Margheri
Nicola Schiavone
Laura Papucci
Lucia Magnelli
Simona Serratì
Anastasia Chillà
Anna Laurenzana
Francesca Bianchini
Lido Calorini
Eugenio Torre
Javier Dotor
Esperanza Feijoo
Gabriella Fibbi
Mario Del Rosso
GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.
description <h4>Background</h4>TGFß overproduction in cancer cells is one of the main characteristics of late tumor progression being implicated in metastasis, tumor growth, angiogenesis and immune response. We investigated the therapeutic efficacy of anti-TGFß peptides in the control of angiogenesis elicited by conditional over-expression of TGFß.<h4>Methods</h4>We have inserted in human MCF7 mammary-cancer cells a mutated TGFß gene in a tetracycline-repressible vector to obtain conditional expression of mature TGFß upon transient transfection, evaluated the signaling pathways involved in TGFß-dependent endothelial cells activation and the efficacy of anti-TGFß peptides in the control of MCF7-TGFß-dependent angiogenesis.<h4>Results</h4>TGFß over-expression induced in MCF7 several markers of the epithelial-to-mesenchymal transition. Conditioned-medium of TGFß-transfected MCF7 stimulated angiogenesis in vivo and in vitro by subsequent activation of SMAD2/3 and SMAD1/5 signaling in endothelial cells, as well as SMAD4 nuclear translocation, resulting in over-expression of the pro-angiogenic growth and differentiation factor-5 (GDF5). Inhibition or silencing of GDF5 in TGFß-stimulated EC resulted in impairment of GDF5 expression and of TGFß-dependent urokinase-plasminogen activator receptor (uPAR) overproduction, leading to angiogenesis impairment. Two different TGFß antagonist peptides inhibited all the angiogenesis-related properties elicited in EC by exogenous and conditionally-expressed TGFß in vivo and in vitro, including SMAD1/5 phosphorylation, SMAD4 nuclear translocation, GDF5 and uPAR overexpression. Antagonist peptides and anti-GDF5 antibodies efficiently inhibited in vitro and in vivo angiogenesis.<h4>Conclusions</h4>TGFß produced by breast cancer cells induces in endothelial cells expression of GDF5, which in turn stimulates angiogenesis both in vitro and in vivo. Angiogenesis activation is rapid and the involved mechanism is totally opposed to the old and controversial dogma about the AKL5/ALK1 balance. The GDF-dependent pro-angiogenic effects of TGFß are controlled by anti-TGFß peptides and anti-GDF5 antibodies, providing a basis to develop targeted clinical studies.
format article
author Francesca Margheri
Nicola Schiavone
Laura Papucci
Lucia Magnelli
Simona Serratì
Anastasia Chillà
Anna Laurenzana
Francesca Bianchini
Lido Calorini
Eugenio Torre
Javier Dotor
Esperanza Feijoo
Gabriella Fibbi
Mario Del Rosso
author_facet Francesca Margheri
Nicola Schiavone
Laura Papucci
Lucia Magnelli
Simona Serratì
Anastasia Chillà
Anna Laurenzana
Francesca Bianchini
Lido Calorini
Eugenio Torre
Javier Dotor
Esperanza Feijoo
Gabriella Fibbi
Mario Del Rosso
author_sort Francesca Margheri
title GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.
title_short GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.
title_full GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.
title_fullStr GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.
title_full_unstemmed GDF5 regulates TGFß-dependent angiogenesis in breast carcinoma MCF-7 cells: in vitro and in vivo control by anti-TGFß peptides.
title_sort gdf5 regulates tgfß-dependent angiogenesis in breast carcinoma mcf-7 cells: in vitro and in vivo control by anti-tgfß peptides.
publisher Public Library of Science (PLoS)
publishDate 2012
url https://doaj.org/article/9be95842e3a64f8a9bd8cf6d6c21ae83
work_keys_str_mv AT francescamargheri gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT nicolaschiavone gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT laurapapucci gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT luciamagnelli gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT simonaserrati gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT anastasiachilla gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT annalaurenzana gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT francescabianchini gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT lidocalorini gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT eugeniotorre gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT javierdotor gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT esperanzafeijoo gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT gabriellafibbi gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
AT mariodelrosso gdf5regulatestgfßdependentangiogenesisinbreastcarcinomamcf7cellsinvitroandinvivocontrolbyantitgfßpeptides
_version_ 1718422183757217792