Oncotarget

Research Papers:

Lactate-activated macrophages induced aerobic glycolysis and epithelial-mesenchymal transition in breast cancer by regulation of CCL5-CCR5 axis: a positive metabolic feedback loop

Sensen Lin, Li Sun, Xiaodan Lyu, Xiongfei Ai, Danyu Du, Nan Su, Hongyang Li, Luyong Zhang, Jun Yu _ and Shengtao Yuan

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:110426-110443. https://doi.org/10.18632/oncotarget.22786

Metrics: PDF 2854 views  |   HTML 4010 views  |   ?  


Abstract

Sensen Lin1,*, Li Sun1,*, Xiaodan Lyu1,*, Xiongfei Ai1, Danyu Du1, Nan Su1, Hongyang Li1, Luyong Zhang1, Jun Yu2 and Shengtao Yuan1

1Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, PR China

2Department of Molecular Biology, Jiangsu Cancer Hospital, Nanjing 210009, PR China

*These authors have contributed equally to this work

Correspondence to:

Jun Yu, email: [email protected]

Shengtao Yuan, email: [email protected]

Keywords: macrophage; lactate; CCL5-CCR5 axis; glycolysis; AMPK

Received: March 21, 2017     Accepted: November 16, 2017     Published: November 30, 2017

ABSTRACT

Aberrant energy metabolism is critical for cancer progression. Tumor-associated macrophages (TAMs) can stimulate tumor angiogenesis and enhance cancer metastasis; however, the metabolic interaction between cancer cells and macrophages characterized by lactate shuttles remains unclear. Here, we showed that lactate activated human macrophages to a TAM-like phenotype and stimulated the secretion of CCL5 by activation of Notch signaling in macrophages. Reciprocally, CCL5 increased cell migration, induced cancer cell EMT, and promoted aerobic glycolysis in breast cancer cells, suggesting a positive metabolic feedback loop in the co-culture system. Inhibition of CCR5, the cognate receptor of CCL5, or neutralization of CCL5, broke the metabolic loop and decreased cancer cell migration and EMT. Inhibition of aerobic glycolysis significantly reduced breast cancer cell EMT, indicated that aerobic glycolysis was necessary for the invasive phenotype of cancer cells. We further showed that TGF-β signaling regulated the expression of CCR5 in the co-culture system, and CCL5 induced glycolysis by mediation of AMPK signaling. The expression of CCL5-CCR5 axis was highly associated with macrophage infiltration, TGF-β and p-AMPK in clinical samples. CCL5-CCR5 axis promoted breast cancer metastasis in vivo. Our findings suggested a pivotal role of CCL5-CCR5 axis in the metabolic communication between cancer cells and macrophages.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 22786