Cargando…

Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis

INTRODUCTION: Mammary tumorigenesis is associated with the increased expression of several proteins in the focal adhesion complex, including focal adhesion kinase (FAK) and various integrins. Aberrant expression of these molecules occurs concomitant with the conversion of TGF-β function from a tumor...

Descripción completa

Detalles Bibliográficos
Autores principales: Wendt, Michael K, Schiemann, William P
Formato: Texto
Lenguaje:English
Publicado: BioMed Central 2009
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2790843/
https://www.ncbi.nlm.nih.gov/pubmed/19740433
http://dx.doi.org/10.1186/bcr2360
_version_ 1782175137034928128
author Wendt, Michael K
Schiemann, William P
author_facet Wendt, Michael K
Schiemann, William P
author_sort Wendt, Michael K
collection PubMed
description INTRODUCTION: Mammary tumorigenesis is associated with the increased expression of several proteins in the focal adhesion complex, including focal adhesion kinase (FAK) and various integrins. Aberrant expression of these molecules occurs concomitant with the conversion of TGF-β function from a tumor suppressor to a tumor promoter. We previously showed that interaction between β(3 )integrin and TβR-II facilitates TGF-β-mediated oncogenic signaling, epithelial-mesenchymal transition (EMT), and metastasis. However, the molecular mechanisms by which the focal adhesion complex contributes to β(3 )integrin:TβR-II signaling and the oncogenic conversion of TGF-β remain poorly understood. METHODS: FAK expression and activity were inhibited in normal and malignant mammary epithelial cells (MECs) either genetically by using lentiviral-mediated delivery of shRNAs against FAK, or pharmacologically through in vitro and in vivo use of the FAK inhibitors, PF-562271 and PF-573228. Altered Smad2/3 and p38 MAPK activation, migration, EMT, and invasion in response to TGF-β(1 )were monitored in FAK-manipulated cells. TβR-II expression was increased in metastatic breast cancer cells by retroviral transduction, and the metastasis of FAK- and TβR-II-manipulated tumors was monitored by using bioluminescent imaging. RESULTS: TGF-β stimulation of MECs stabilized and activated FAK in a β(3 )integrin- and Src-dependent manner. Furthermore, by using the human MCF10A breast cancer progression model, we showed that increased FAK expression in metastatic breast cancer cells mirrored the acquisition of enhanced activation of p38 MAPK by TGF-β. Administering FAK inhibitors or rendering metastatic breast cancer cells FAK deficient abrogated the interaction between β(3 )integrin and TβR-II, thereby preventing TGF-β from (a) activating p38 MAPK; (b) stimulating MEC invasion, migration, and EMT; and (c) inducing early primary tumor dissemination to the lungs. Finally, in contrast to FAK depletion, adjuvant FAK chemotherapy of mammary tumors decreased their growth in part by diminished macrophage tumor infiltration. CONCLUSIONS: Our studies identify an essential function for FAK in mediating the interaction between β(3 )integrin and TβR-II, and thus in facilitating the oncogenic conversion of TGF-β required for mammary tumor metastasis. Furthermore, this study establishes chemotherapeutic targeting of FAK as an effective, two-pronged approach in preventing tumor progression both by decreasing innate immune cell infiltration, and by inhibiting early TGF-β-dependent metastasis.
format Text
id pubmed-2790843
institution National Center for Biotechnology Information
language English
publishDate 2009
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-27908432009-12-10 Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis Wendt, Michael K Schiemann, William P Breast Cancer Res Research article INTRODUCTION: Mammary tumorigenesis is associated with the increased expression of several proteins in the focal adhesion complex, including focal adhesion kinase (FAK) and various integrins. Aberrant expression of these molecules occurs concomitant with the conversion of TGF-β function from a tumor suppressor to a tumor promoter. We previously showed that interaction between β(3 )integrin and TβR-II facilitates TGF-β-mediated oncogenic signaling, epithelial-mesenchymal transition (EMT), and metastasis. However, the molecular mechanisms by which the focal adhesion complex contributes to β(3 )integrin:TβR-II signaling and the oncogenic conversion of TGF-β remain poorly understood. METHODS: FAK expression and activity were inhibited in normal and malignant mammary epithelial cells (MECs) either genetically by using lentiviral-mediated delivery of shRNAs against FAK, or pharmacologically through in vitro and in vivo use of the FAK inhibitors, PF-562271 and PF-573228. Altered Smad2/3 and p38 MAPK activation, migration, EMT, and invasion in response to TGF-β(1 )were monitored in FAK-manipulated cells. TβR-II expression was increased in metastatic breast cancer cells by retroviral transduction, and the metastasis of FAK- and TβR-II-manipulated tumors was monitored by using bioluminescent imaging. RESULTS: TGF-β stimulation of MECs stabilized and activated FAK in a β(3 )integrin- and Src-dependent manner. Furthermore, by using the human MCF10A breast cancer progression model, we showed that increased FAK expression in metastatic breast cancer cells mirrored the acquisition of enhanced activation of p38 MAPK by TGF-β. Administering FAK inhibitors or rendering metastatic breast cancer cells FAK deficient abrogated the interaction between β(3 )integrin and TβR-II, thereby preventing TGF-β from (a) activating p38 MAPK; (b) stimulating MEC invasion, migration, and EMT; and (c) inducing early primary tumor dissemination to the lungs. Finally, in contrast to FAK depletion, adjuvant FAK chemotherapy of mammary tumors decreased their growth in part by diminished macrophage tumor infiltration. CONCLUSIONS: Our studies identify an essential function for FAK in mediating the interaction between β(3 )integrin and TβR-II, and thus in facilitating the oncogenic conversion of TGF-β required for mammary tumor metastasis. Furthermore, this study establishes chemotherapeutic targeting of FAK as an effective, two-pronged approach in preventing tumor progression both by decreasing innate immune cell infiltration, and by inhibiting early TGF-β-dependent metastasis. BioMed Central 2009 2009-09-09 /pmc/articles/PMC2790843/ /pubmed/19740433 http://dx.doi.org/10.1186/bcr2360 Text en Copyright ©2009 Wendt and Schiemann; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium provided the original work is properly cited.
spellingShingle Research article
Wendt, Michael K
Schiemann, William P
Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis
title Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis
title_full Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis
title_fullStr Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis
title_full_unstemmed Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis
title_short Therapeutic targeting of the focal adhesion complex prevents oncogenic TGF-β signaling and metastasis
title_sort therapeutic targeting of the focal adhesion complex prevents oncogenic tgf-β signaling and metastasis
topic Research article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2790843/
https://www.ncbi.nlm.nih.gov/pubmed/19740433
http://dx.doi.org/10.1186/bcr2360
work_keys_str_mv AT wendtmichaelk therapeutictargetingofthefocaladhesioncomplexpreventsoncogenictgfbsignalingandmetastasis
AT schiemannwilliamp therapeutictargetingofthefocaladhesioncomplexpreventsoncogenictgfbsignalingandmetastasis