Abstract

BackgroundEndocrine FGF21 and FGF19 target adipocytes and hepatocytes through betaKlotho (KLB) and FGFR tyrosine kinases effecting glucose, lipid and energy metabolism. Both factors alleviate obesity and metabolic abnormalities which are contributing factors to breast tumor progression. Genomic manipulation of hepatic FGFR4 has uncovered roles of endocrine FGF signaling in both metabolic and cellular homeostasis. Here we determined whether systemic and microenvironmental metabolic alterations caused by the FGFR4 deficiency affect tumorigenesis in breast where FGFR4 is negligible. Breast tumors were induced in the bigenic mice with ablation of FGFR4 and overexpression of TGFα that activates Her2 in the ductal and lobular epithelium surrounded by adipocytes. Mammary tumorigenesis and alterations in systemic and breast microenvironmental metabolic parameters and regulatory pathways were analyzed.ResultsAblation of FGFR4 had no effect on cellular homeostasis and Her2 activity of normal breast tissue. However, the absence of FGFR4 reduced TGFα–driven breast tumor incidence and progression and improved host survival. Notable increases in hepatic and serum FGF21, ileal FGF15/19, adiponectin and adipsin, and decreases in systemic Fetuin A, IGF-1, IGFBP-1, RBP4 and TIMP1 were observed. The ablation affected adipogenesis and secretory function of adipocytes as well as lipogenesis, glycolysis and energy homeostasis associated with the functions of mitochondria, ER and peroxisomes in the breast and tumor foci. Treatment with a chemical inhibitor of NAMPT involved in the pathways inhibited the growth and survival of breast tumor cells and tumor-initiating cell-containing spheres. The FGFR4 ablation also caused elevation of inflammatory factors in the breast.ConclusionsAlthough the primary role of FGFR4 in metabolism occurs in hepatocytes, its ablation results in a net inhibitory effect on mammary tumor progression. We suggest that the tumor-delaying effect of FGFR4 deficiency may be in large part due to elevated anti-obesogenic FGF21 that triggers tumor-suppressing signals from both peripheral and breast adipocytes. The predominant changes in metabolic pathways suggested roles of metabolic effects from both peripheral and breast adipocytes on metabolic reprogramming in breast epithelial cells that contribute to the suppression of tumor progression. These results provide new insights into the contribution of systemic and microenvironmental metabolic effects controlled by endocrine FGF signaling to breast carcinogenesis.

Highlights

  • Endocrine FGF21 and FGF19 target adipocytes and hepatocytes through betaKlotho (KLB) and Fibroblast growth factor receptors (FGFRs) tyrosine kinases effecting glucose, lipid and energy metabolism

  • The inter-organ cross-talking endocrine axis from hepatic FGF21 to adipocyte FGFR1-KLB is a stress-responsive pathway leading to the correction of deranged glucose, lipid and energy metabolism that benefit the organism during conditions of stress-induced pathologies such as obesity, diabetes, fatty liver diseases, malignant transformation and insults from toxins and infection [13,17,18,19,20]

  • The FGFR4 deficiency causes increases in systemic factors, including the adipocyte-targeting hepatic stress hormone FGF21 and ileal FGF15, accompanied by increases in the tumor-suppressive adipokine adiponectin and decreases in tumor-promoting adipokines insulin-like growth factor (IGF)-1 and TIMP1 that are controlled by anti-obesogenic FGF21

Read more

Summary

Introduction

Endocrine FGF21 and FGF19 target adipocytes and hepatocytes through betaKlotho (KLB) and FGFR tyrosine kinases effecting glucose, lipid and energy metabolism. The inter-organ cross-talking endocrine axis from hepatic FGF21 to adipocyte FGFR1-KLB is a stress-responsive pathway leading to the correction of deranged glucose, lipid and energy metabolism that benefit the organism during conditions of stress-induced pathologies such as obesity, diabetes, fatty liver diseases, malignant transformation and insults from toxins and infection [13,17,18,19,20] Some of these pathologies are risk factors for tumorigenesis [21,22,23,24]. They provide novel opportunities for the treatment of obesity, type 2 diabetes, hypophosphatemia and diverse types of cancer where aberrant metabolism is a complicating contributor

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call