Your browser doesn't support javascript.
loading
Notch1-WISP-1 axis determines the regulatory role of mesenchymal stem cell-derived stromal fibroblasts in melanoma metastasis.
Shao, Hongwei; Cai, Long; Moller, Mecker; Issac, Biju; Zhang, Leiming; Owyong, Mark; Moscowitz, Anna Elizabeth; Vazquez-Padron, Roberto; Radtke, Freddy; Liu, Zhao-Jun.
Afiliação
  • Shao H; Department of Surgery, University of Miami School of Medicine, Miami, USA.
  • Cai L; Department of Surgery, University of Miami School of Medicine, Miami, USA.
  • Moller M; Hangzhou Red-Cross Hospital, Zhejiang, China.
  • Issac B; Department of Surgery, University of Miami School of Medicine, Miami, USA.
  • Zhang L; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
  • Owyong M; Department of Surgery, University of Miami School of Medicine, Miami, USA.
  • Moscowitz AE; Yantai University, School of Pharmacy, Shandong, China.
  • Vazquez-Padron R; Department of Surgery, University of Miami School of Medicine, Miami, USA.
  • Radtke F; Department of Surgery, University of Miami School of Medicine, Miami, USA.
  • Liu ZJ; Department of Surgery, University of Miami School of Medicine, Miami, USA.
Oncotarget ; 7(48): 79262-79273, 2016 Nov 29.
Article em En | MEDLINE | ID: mdl-27813493
ABSTRACT
Mesenchymal stem cells-derived fibroblasts (MSC-DF) constitute a significant portion of stromal fibroblasts in the tumor microenvironment (TME) and are key modulators of tumor progression. However, the molecular mechanisms that determine their tumor-regulatory function are poorly understood. Here, we uncover the Notch1 pathway as a molecular determinant that selectively controls the regulatory role of MSC-DF in melanoma metastasis. We demonstrate that the Notch1 pathway's activity is inversely correlated with the metastasis-regulating function of fibroblasts and can determine the metastasis-promoting or -suppressing phenotype of MSC-DF. When co-grafted with melanoma cells, MSC-DFNotch1-/- selectively promote, while MSC-DFN1IC+/+ preferentially suppress melanoma metastasis, but not growth, in mouse models. Consistently, conditioned media (CM) from MSC-DFNotch1-/- and MSC-DFN1IC+/+ oppositely, yet selectively regulates migration, but not growth of melanoma cells in vitro. Additionally, when co-cultured with metastatic melanoma cells in vitro, MSC-DFNotch1-/- support, while MSC-DFN1IC+/+ inhibit melanoma cells in the formation of spheroids. These findings expand the repertoire of Notch1 signaling as a molecular switch in determining the tumor metastasis-regulating function of MSC-DF. We also identified Wnt-induced secreted protein-1 (WISP-1) as a key downstream secretory mediator of Notch1 signaling to execute the influential role of MSC-DF on melanoma metastasis. These findings reveal the Notch1-WISP-1 axis as a crucial molecular determinant in governing stromal regulation of melanoma metastasis; thus, establishing this axis as a potential therapeutic target for melanoma metastasis.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Proteínas Proto-Oncogênicas / Receptor Notch1 / Proteínas de Sinalização Intercelular CCN / Células-Tronco Mesenquimais / Fibroblastos / Melanoma Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2016 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Proteínas Proto-Oncogênicas / Receptor Notch1 / Proteínas de Sinalização Intercelular CCN / Células-Tronco Mesenquimais / Fibroblastos / Melanoma Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2016 Tipo de documento: Article