Your browser doesn't support javascript.
loading
Arsenic Trioxide Decreases Lymphangiogenesis by Inducing Apoptotic Pathways and Inhibition of Important Endothelial Cell Receptors.
Hrgovic, Igor; Zöller, Eva; Doll, Monika; Hailemariam-Jahn, Tsige; Jakob, Thilo; Kaufmann, Roland; Meissner, Markus; Kleemann, Johannes.
  • Hrgovic I; Department of Dermatology and Allergy, Experimental Dermatology and Allergy Research Group, University Medical Center Giessen, Justus Liebig University, 35392 Giessen, Germany.
  • Zöller E; Department of Dermatology, Venereology and Allergy, Goethe University, 60596 Frankfurt am Main, Germany.
  • Doll M; Department of Dermatology, Venereology and Allergy, Goethe University, 60596 Frankfurt am Main, Germany.
  • Hailemariam-Jahn T; Department of Dermatology, Venereology and Allergy, Goethe University, 60596 Frankfurt am Main, Germany.
  • Jakob T; Department of Dermatology and Allergy, Experimental Dermatology and Allergy Research Group, University Medical Center Giessen, Justus Liebig University, 35392 Giessen, Germany.
  • Kaufmann R; Department of Dermatology, Venereology and Allergy, Goethe University, 60596 Frankfurt am Main, Germany.
  • Meissner M; Department of Dermatology, Venereology and Allergy, Goethe University, 60596 Frankfurt am Main, Germany.
  • Kleemann J; Department of Dermatology, Venereology and Allergy, Goethe University, 60596 Frankfurt am Main, Germany.
Curr Issues Mol Biol ; 46(1): 67-80, 2023 Dec 21.
Article en En | MEDLINE | ID: mdl-38275666
ABSTRACT
Tumor-induced lymphangiogenesis is strongly associated with the formation of tumor metastasis. Therefore, the regulation of lymphangiogenesis offers a promising target in cancer therapy. Arsenic trioxide (ATO) is highly effective in the treatment of patients with acute promyelocytic leukemia (APL). As ATO mediates anti-angiogenic effects on endothelial and tumor cells, we aimed to explore the impact of ATO on lymphangiogenesis in human lymphatic endothelial cells (LEC). The BrdU assay and flow cytometry analysis were used to evaluate the influence of ATO on the proliferation and cell cycle distribution of LECs. The lymphatic suppression effects of ATO were investigated in vitro using the lymphatic tube formation assay. The effects of ATO on apoptosis, mitochondrial membrane potential and endothelial cell receptors were investigated by Western blotting, ELISA, flow cytometry and qRT-PCR. The treatment of LECs with ATO attenuated cell proliferation, blocked tube formation and induced subG0/G1 arrest in LECs, thus suggesting enhanced apoptosis. Although subG0/G1 arrest was accompanied by the upregulation of p21 and p53, ATO treatment did not lead to visible cell cycle arrest in LECs. In addition, ATO caused apoptosis via the release of cytochrome c from mitochondria, activating caspases 3, 8 and 9; downregulating the anti-apoptotic proteins survivin, XIAP and cIAP-2; and upregulating the pro-apoptotic protein Fas. Furthermore, we observed that ATO inhibited the VEGF-induced proliferation of LECs, indicating that pro-survival VEGF/VEGFR signaling was affected by ATO treatment. Finally, we found that ATO inhibited the expression of the important endothelial cell receptors VEGFR-2, VEGFR-3, Tie-2 and Lyve-1. In conclusion, we demonstrate that ATO inhibits lymphangiogenesis by activating apoptotic pathways and inhibiting important endothelial cell receptors, which suggests that this drug should be further evaluated in the treatment of tumor-associated lymphangiogenesis.
Palabras clave