Aberrant Thyroid-Stimulating Hormone Receptor Signaling Increases VEGF-A and CXCL8 Secretion of Thyroid Cancer Cells, Contributing to Angiogenesis and Tumor Growth

Clin Cancer Res. 2019 Jan 1;25(1):414-425. doi: 10.1158/1078-0432.CCR-18-0663. Epub 2018 Oct 12.

Abstract

Purpose: Thyroid-stimulating hormone (TSH) suppression is widely used to treat well-differentiated thyroid cancer, whereas its role in poorly differentiated thyroid cancer (PDTC) is undetermined. Besides thyrocytes, TSH also binds to stromal cells, comprising tumor microenvironments. This study aimed to investigate the effects of TSH on tumor microenvironments in PDTC.

Experimental design: An ectopic tumor model using PDTC cells (BHP10-3SCp and FRO), which exhibit TSH/cAMP-independent cell growth, was treated with TSH. IHC was performed using tissue microarrays from 13 PDTCs.

Results: TSH treatment significantly enhanced tumor growth of PDTCs with increased vascularity but not that of breast cancer cells, suggesting this effect is unique to thyroid cancer cells, not stromal cells. TSH significantly upregulated VEGF-A and CXCL8 expressions in BHP10-3SCp cells via AKT and ERK signaling, resulting in higher concentrations of VEGF-A and CXCL8 in conditioned medium of TSH-treated BHP10-3SCp cells (TSH-CM) compared with controls. TSH-CM treatment enhanced tube formation potentials of endothelial cells, and blocking VEGF and/or CXCL8 reduced them. Blocking VEGF and/or CXCL8 also reduced TSH-dependent tumor growth with reduced tumor vasculature in vivo. TSH-treated tumors showed increased macrophage densities, and macrophage inhibition reduced TSH-dependent tumor growth in vivo. In human PDTCs, preoperative TSH levels were positively associated with VEGF-A and tumor size, and the expression of VEGF-A was positively correlated with CD31, CD163, and CXCL8, and their clinical poor prognosis.

Conclusions: Aberrant TSH receptor signaling modulates tumor angiogenesis by stimulating VEGF-A and CXCL8 secretion from PDTC cells and enhances tumor growth; thus, TSH suppression is beneficial for treating PDTCs.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Angiogenesis Inhibitors / pharmacology
  • Animals
  • Cell Proliferation / drug effects
  • Humans
  • Interleukin-8 / genetics*
  • Macrophages / metabolism
  • Macrophages / pathology
  • Mice
  • Neovascularization, Pathologic / drug therapy
  • Neovascularization, Pathologic / genetics*
  • Neovascularization, Pathologic / pathology
  • Receptors, Thyrotropin / antagonists & inhibitors
  • Receptors, Thyrotropin / genetics*
  • Thyroid Neoplasms / drug therapy
  • Thyroid Neoplasms / genetics*
  • Thyroid Neoplasms / pathology
  • Thyrotropin / genetics
  • Thyrotropin / metabolism
  • Tumor Microenvironment / drug effects
  • Vascular Endothelial Growth Factor A / genetics*
  • Xenograft Model Antitumor Assays

Substances

  • Angiogenesis Inhibitors
  • Interleukin-8
  • Receptors, Thyrotropin
  • Vascular Endothelial Growth Factor A
  • Thyrotropin