CXCR4/TGF-β1 mediated hepatic stellate cells differentiation into carcinoma-associated fibroblasts and promoted liver metastasis of colon cancer

HX Tan, WZ Gong, K Zhou, ZG Xiao, FT Hou… - Cancer biology & …, 2020 - Taylor & Francis
HX Tan, WZ Gong, K Zhou, ZG Xiao, FT Hou, T Huang, L Zhang, HY Dong, WL Zhang, Y Liu…
Cancer biology & therapy, 2020Taylor & Francis
Background: Liver metastasis of colon cancer is strongly affected by the tumor
microenvironment (TME), with interactions between tumor cells and cancer-associated
fibroblasts (CAFs) in particular. TGF-β is well known for its ability to mediate the CAF
phenotype, and CXCR4 expression is closely correlated to poor prognosis in CRC. The
relationship between these two signaling pathways remains to be delineated in liver
metastasis of colon cancer. Methods: Immunohistochemistry was employed to investigate …
Abstract
Background: Liver metastasis of colon cancer is strongly affected by the tumor microenvironment (TME), with interactions between tumor cells and cancer-associated fibroblasts (CAFs) in particular. TGF-β is well known for its ability to mediate the CAF phenotype, and CXCR4 expression is closely correlated to poor prognosis in CRC. The relationship between these two signaling pathways remains to be delineated in liver metastasis of colon cancer.
Methods: Immunohistochemistry was employed to investigate CXCR4 expression in 45 human specimens of primary colorectal cancer (CRC) and liver metastasis. The functions of SDF-1 released by hepatic stellate cells (HSCs) on CXCR4 and TGF-β1 in CRC cells were investigated in vitro. The effects of CRC on HSCs differentiation into CAFs were confirmed using co-culture technology and expression analysis of CAFs markers by qPCR, western blot and immunofluorescence. The involvement of CXCR4 and TGF-β1 was verified with addition of CXCR4 inhibitor AMD3100 and TGF-β1 inhibitor cyclophosphamide (Cy) both in vitro and in vivo.
Results: There were more CXCR4-positive cells at the liver metastatic tissues compared to the primary sites. CRC cells activated and transformed HSCs to CAFs after co-cultivating with HSCs. Activated HSCs stimulated TGF-β1 secretion from CRC cells after co-culture with CRC cells in vitro. Moreover, the expression of CAFs markers was increasing in the activated HSCs. In a mouse hepatic metastasis model, treated with AMD3100 or Cy blocked the metastatic potential of HCT116 cells and the hepatic CAFs differentiation.
Conclusions: These results indicated that CXCR4/TGF-β1 axis plays an important role in CRC liver metastasis through mediating HSCs differentiation into CAFs, providing preclinical evidences that blockade of the axis might be beneficial for anti-metastasis therapy in CRC.
Taylor & Francis Online