1. Academic Validation
  2. Semaphorin 7A promotes endothelial to mesenchymal transition through ATF3 mediated TGF-β2/Smad signaling

Semaphorin 7A promotes endothelial to mesenchymal transition through ATF3 mediated TGF-β2/Smad signaling

  • Cell Death Dis. 2020 Aug 10;11(8):695. doi: 10.1038/s41419-020-02818-x.
Lei Hong  # 1 2 3 Fengchan Li  # 2 Chaojun Tang  # 2 Ling Li 2 Lili Sun 1 Xiaoqiang Li 4 Li Zhu 5
Affiliations

Affiliations

  • 1 Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China.
  • 2 Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China.
  • 3 Department of Vascular Surgery, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, Anhui, China.
  • 4 Department of Vascular Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, China. [email protected].
  • 5 Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Suzhou Key Laboratory of Thrombosis and Vascular Biology, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China. [email protected].
  • # Contributed equally.
Abstract

Endothelial to mesenchymal transition (EndMT) is an important pathological change in many diseases. Semaphorin7A (Sema7A) has been reported to regulate nerve and vessel homeostasis, but its role in EndMT remains unclear. Here we investigate the effect of Sema7A on EndMT and the underlying mechanism. Sema7A-overexpressed human umbilical vein endothelial cells (Sema7A-HUVECs) were generated and showed lower levels of endothelial cell markers and higher levels of mesenchymal cell markers indicating the occurrence of EndMT. RNA-sequencing analysis showed a total of 1168 upregulated genes and 886 downregulated genes. Among them, most of the molecules associated with EndMT were upregulated in Sema7A-HUVECs. Mechanistically, Sema7A-HUVECs showed a higher TGF-β2 expression and activated TGF-β/Smad Signaling. Importantly, Sema7A overexpression upregulated activating transcription factor 3 (ATF3) that was found to selectively bind the promotor region of TGF-β2, but not TGF-β1, promoting TGF-β2 transcription, which was further confirmed by ATF3-siRNA knockdown approach. Blocking β1 Integrin, a known Sema7A receptor, alleviated the expression of ATF3, TGF-β2, and EndMT in Sema7A-overexpressed HUVECs, implying a role of β1 Integrin/ATF3/TGF-β2 axis in mediating Sema7A-induced EndMT. Using Sema7A-deficient mice and the partial carotid artery ligation (PCL) model, we showed that Sema7A deletion attenuated EndMT induced by blood flow disturbance in vivo. In conclusion, Sema7A promotes TGF-β2 secretion by upregulating transcription factor ATF3 in a β1 integrin-dependent manner, and thus facilitates EndMT through TGF/Smad signaling, implying Sema7A as a potential therapeutic target for EndMT-related vascular diseases.

Figures
Products