Your browser doesn't support javascript.
loading
FGF1 supports glycolytic metabolism through the estrogen receptor in endocrine-resistant and obesity-associated breast cancer.
Castillo-Castrejon, Marisol; Sankofi, Barbara Mensah; Murguia, Stevi Johnson; Udeme, Abasi-Ama; Cen, Hoaning Howard; Xia, Yi Han; Thomas, Nisha S; Berry, William L; Jones, Kenneth L; Richard, Vincent R; Zahedi, Rene P; Borchers, Christoph H; Johnson, James D; Wellberg, Elizabeth A.
Afiliación
  • Castillo-Castrejon M; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Sankofi BM; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Murguia SJ; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Udeme AA; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Cen HH; Life Sciences Institute, University of British Columbia, Vancouver, Canada.
  • Xia YH; Life Sciences Institute, University of British Columbia, Vancouver, Canada.
  • Thomas NS; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Berry WL; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Jones KL; Department of Pathology, University of Oklahoma Health Sciences Center, 975 NE 10th Street BRC 309, Oklahoma City, OK, 73104, USA.
  • Richard VR; Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital and McGill University, Montreal, QC, Canada.
  • Zahedi RP; Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB, R3E 3P4, Canada.
  • Borchers CH; Department of Internal Medicine, University of Manitoba, Winnipeg, MB, R3E 3P4, Canada.
  • Johnson JD; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, R3E 0J9, Canada.
  • Wellberg EA; CancerCare Manitoba Research Institute, Winnipeg, MB, R3E 0V9, Canada.
Breast Cancer Res ; 25(1): 99, 2023 08 22.
Article en En | MEDLINE | ID: mdl-37608351
ABSTRACT

BACKGROUND:

Obesity increases breast cancer risk and breast cancer-specific mortality, particularly for people with estrogen receptor (ER)-positive tumors. Body mass index (BMI) is used to define obesity, but it may not be the best predictor of breast cancer risk or prognosis on an individual level. Adult weight gain is an independent indicator of breast cancer risk. Our previous work described a murine model of obesity, ER-positive breast cancer, and weight gain and identified fibroblast growth factor receptor (FGFR) as a potential driver of tumor progression. During adipose tissue expansion, the FGF1 ligand is produced by hypertrophic adipocytes as a stimulus to stromal preadipocytes that proliferate and differentiate to provide additional lipid storage capacity. In breast adipose tissue, FGF1 production may stimulate cancer cell proliferation and tumor progression.

METHODS:

We explored the effects of FGF1 on ER-positive endocrine-sensitive and resistant breast cancer and compared that to the effects of the canonical ER ligand, estradiol. We used untargeted proteomics, specific immunoblot assays, gene expression profiling, and functional metabolic assessments of breast cancer cells. The results were validated in tumors from obese mice and breast cancer datasets from women with obesity.

RESULTS:

FGF1 stimulated ER phosphorylation independently of estradiol in cells that grow in obese female mice after estrogen deprivation treatment. Phospho- and total proteomic, genomic, and functional analyses of endocrine-sensitive and resistant breast cancer cells show that FGF1 promoted a cellular phenotype characterized by glycolytic metabolism. In endocrine-sensitive but not endocrine-resistant breast cancer cells, mitochondrial metabolism was also regulated by FGF1. Comparison of gene expression profiles indicated that tumors from women with obesity shared hallmarks with endocrine-resistant breast cancer cells.

CONCLUSIONS:

Collectively, our data suggest that one mechanism by which obesity and weight gain promote breast cancer progression is through estrogen-independent ER activation and cancer cell metabolic reprogramming, partly driven by FGF/FGFR. The first-line treatment for many patients with ER-positive breast cancer is inhibition of estrogen synthesis using aromatase inhibitors. In women with obesity who are experiencing weight gain, locally produced FGF1 may activate ER to promote cancer cell metabolic reprogramming and tumor progression independently of estrogen.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Asunto principal: Neoplasias de la Mama / Receptores de Estrógenos / Factor 1 de Crecimiento de Fibroblastos Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Animals Idioma: En Revista: Breast Cancer Res Asunto de la revista: NEOPLASIAS Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Asunto principal: Neoplasias de la Mama / Receptores de Estrógenos / Factor 1 de Crecimiento de Fibroblastos Tipo de estudio: Prognostic_studies / Risk_factors_studies Límite: Animals Idioma: En Revista: Breast Cancer Res Asunto de la revista: NEOPLASIAS Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos