Your browser doesn't support javascript.
loading
A Role for TGFß Signaling in Preclinical Osteolytic Estrogen Receptor-Positive Breast Cancer Bone Metastases Progression.
Cheng, Julia N; Frye, Jennifer B; Whitman, Susan A; Kunihiro, Andrew G; Pandey, Ritu; Funk, Janet L.
Afiliação
  • Cheng JN; Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85724, USA.
  • Frye JB; Department of Medicine, University of Arizona, Tucson, AZ 85724, USA.
  • Whitman SA; Department of Medicine, University of Arizona, Tucson, AZ 85724, USA.
  • Kunihiro AG; Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85724, USA.
  • Pandey R; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA.
  • Funk JL; Department of Medicine, University of Arizona, Tucson, AZ 85724, USA.
Int J Mol Sci ; 22(9)2021 Apr 24.
Article em En | MEDLINE | ID: mdl-33923316
ABSTRACT
While tumoral Smad-mediated transforming growth factor ß (TGFß) signaling drives osteolytic estrogen receptor α-negative (ER-) breast cancer bone metastases (BMETs) in preclinical models, its role in ER+ BMETs, representing the majority of clinical BMETs, has not been documented. Experiments were undertaken to examine Smad-mediated TGFß signaling in human ER+ cells and bone-tropic behavior following intracardiac inoculation of estrogen (E2)-supplemented female nude mice. While all ER+ tumor cells tested (ZR-75-1, T47D, and MCF-7-derived) expressed TGFß receptors II and I, only cells with TGFß-inducible Smad signaling (MCF-7) formed osteolytic BMETs in vivo. Regulated secretion of PTHrP, an osteolytic factor expressed in >90% of clinical BMETs, also tracked with osteolytic potential; TGFß and E2 each induced PTHrP in bone-tropic or BMET-derived MCF-7 cells, with the combination yielding additive effects, while in cells not forming BMETs, PTHrP was not induced. In vivo treatment with 1D11, a pan-TGFß neutralizing antibody, significantly decreased osteolytic ER+ BMETs in association with a decrease in bone-resorbing osteoclasts at the tumor-bone interface. Thus, TGFß may also be a driver of ER+ BMET osteolysis. Moreover, additive pro-osteolytic effects of tumoral E2 and TGFß signaling could at least partially explain the greater propensity for ER+ tumors to form BMETs, which are primarily osteolytic.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Temas: Geral / Tipos_de_cancer / Outros_tipos Base de dados: MEDLINE Assunto principal: Osteoclastos / Osteólise / Neoplasias Ósseas / Neoplasias da Mama / Receptores de Estrogênio / Regulação Neoplásica da Expressão Gênica / Fator de Crescimento Transformador beta Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Revista: Int J Mol Sci Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Temas: Geral / Tipos_de_cancer / Outros_tipos Base de dados: MEDLINE Assunto principal: Osteoclastos / Osteólise / Neoplasias Ósseas / Neoplasias da Mama / Receptores de Estrogênio / Regulação Neoplásica da Expressão Gênica / Fator de Crescimento Transformador beta Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Revista: Int J Mol Sci Ano de publicação: 2021 Tipo de documento: Article País de afiliação: Estados Unidos