Sema4C modulates the migration of primary tumor-associated lymphatic endothelial cells via an ERK-mediated pathway

Exp Ther Med. 2021 Oct;22(4):1102. doi: 10.3892/etm.2021.10535. Epub 2021 Aug 2.

Abstract

Although lymphatic endothelial cells (LECs) serve a positive role in tumor lymphatic metastasis, the regulation of LECs undergoing migration similar to that of tumor cells remains poorly understood. A previous study revealed that semaphorin 4C (Sema4C) could be a marker of LECs in cervical cancer. Thus, the present study aimed to understand the mechanism via which Sema4C could promote the development of tumor-associated characteristics in LECs in cervical cancer. Primary tumor-associated LECs (TLECs) were distinguished from cervical cancer by flow cytometry. The promigratory ability was assessed using the Transwell assay. Lentivirus infection was used to alter the expression of Sema4C in TLECs. Confocal laser scanning was used to determine the infection efficiency of lentivirus infection. Sema4C/ERK/E-cadherin pathway was measured by reverse transcription-quantitative PCR and western blotting. The co-localization of Sema4C and the lymphatic marker lymphatic vessel endothelial hyaluronan receptor 1 was verified. Primary tumor-associated LECs (TLECs) were isolated from a mouse xenograft cervical tumor model. It was revealed that overexpressing Sema4C stimulated the migratory ability of TLECs, downregulated E-cadherin expression and stimulated ERK phosphorylation, whereas knocking down Sema4C had the opposite effects. The treatment of PD98059 (ERK inhibitor) blocked the pro-migratory ability of TLECs, which indicated a dependence on the ERK signaling pathway. It was identified that the Sema4C/ERK/E-cadherin pathway may be critical for the migration of TLECs, which may promote lymph node metastasis. Therefore, Sema4C could be a promising target for the treatment of cervical cancer with lymphatic metastasis.

Keywords: E-cadherin; extracellular signal-regulated kinase; semaphorin 4C; tumor-associated lymphatic endothelial cells.

Grants and funding

Funding: The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 81502238 and 81602286), the Department of Medical and Health Science Technology of Shandong Province (grant no. 2016w0345), the Department of Science Technology of Jinan City (grant no. 201705051), the China Postdoctoral Science Foundation (grant no. 2019T120594) and Natural Science Foundation General Project of Shandong Province (grant no. ZR2020MH230).