Your browser doesn't support javascript.
loading
RON (MST1R) and HGFL (MST1) Co-Overexpression Supports Breast Tumorigenesis through Autocrine and Paracrine Cellular Crosstalk.
Hunt, Brian G; Jones, Angelle; Lester, Carissa; Davis, James C; Benight, Nancy M; Waltz, Susan E.
Afiliação
  • Hunt BG; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Jones A; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Lester C; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Davis JC; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Benight NM; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
  • Waltz SE; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
Cancers (Basel) ; 14(10)2022 May 19.
Article em En | MEDLINE | ID: mdl-35626096
ABSTRACT

BACKGROUND:

Aberrant RON signaling is present in numerous cancers including breast cancer. Evidence suggests that the ligand, hepatocyte growth factor-like (HGFL), is also overexpressed in breast cancer. RON (MST1R) and HGFL (MST1) genes are located on human chromosome 3 and mouse chromosome 9 respectively and are found near each other in both species. Based on co-expression patterns, we posited that RON and HGFL are co-regulated and that coordinate upregulation drives aggressive tumorigenesis.

METHODS:

Mouse models were used to establish the functional significance of RON and HGFL co-overexpression on the activation of tumor cells and tumor-associated macrophages in breast cancer. TCGA and METABRIC gene expression and alteration data were used to query the relationships between MST1R and MST1 in breast cancer.

RESULTS:

In tumor models, physiologic sources of HGFL modestly improve Arginase-1+ (M2) macrophage recruitment to the tumor proper. Tumor-cell produced HGFL functions in autocrine to sustain tumor cell RON activation and MAPK-dependent secretion of chemotactic factors and in paracrine to activate RON on macrophages and to promote breast cancer stem cell self-renewal. In silico analyses support that RON and HGFL are co-expressed across virtually all cancer types including breast cancer and that common genomic alterations do not appear to be drivers of RON/HGFL co-overexpression.

CONCLUSIONS:

Co-overexpression of RON and HGFL in breast cancer cells (augmented by physiologic sources of HGFL) promotes tumorigenesis through autocrine-mediated RON activation/RON-dependent secretome changes and paracrine activation of macrophage RON to promote breast cancer stem cell self-renewal.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article