Return to search

Release of soluble E-cadherin and its angiogenic role in ovarian cancer

Ovarian cancer is the most lethal gynecological cancer. This is mainly due to widespread peritoneal dissemination and malignant ascites, in which angiogenesis, the formation of new blood vessels, is critical to both ascites development and its metastasis. Loss of E-cadherin is a well-established marker that characterizes the progression of metastatic tumors, including ovarian cancer. The release of a soluble form of E-cadherin (sE-cad) has been frequently associated with a rapid reduction of functional E-cadherin at the cell surface. Importantly, sE-cad is significantly present in ascites from women with stage III/IV ovarian cancer when compared to women with benign ovarian cysts. However, despite the clinical significance, most studies have focused on its role in weakening cell-cell adhesion, whether sE-cad itself has any biological function is not fully understood. Here it is shown for the first time a potent angiogenic role for sE-cad released from ovarian carcinoma. Soluble form of E-cadherin promoted the migration, permeability, and tubulogenesis of endothelial cells. These activities were also observed with a sE-cad/Fc chimera, and targeted inhibition using E-cadherin blocking antibodies completely prevented the sE-cad mediated effects. In addition, it was further revealed that sE-cad could be released from ovarian cancer cells in form of exosomes, a form of extracellular vesicles that play an important role in distant intercellular communication. sE-cad-positive exosomes were able to stimulate the angiogenic phenotype in vitro and functional neovascularization in a Matrigel implant model in vivo. The use of E-cadherin blocking antibodies resulted in diminished angiogenesis, confirming that the effect was sE-cad-positive exosomes specific. In search of the underlying mechanism by which sE-cad-positive exosomes promoted angiogenesis in endothelial cells which lacked E-cadherin, sE-cad was found to heterodimerize with VE-cadherin. This effect was associated with constitutive activation of phosphatidylinositol 3-kinase (PI3K)/Akt and its effector β-catenin, but not p120 catenin. Similarly, the angiogenic phenotype could be reversed by inhibition of VE-cadherin, PI3K/Akt and β-catenin. A mass spectrometric proteomic analysis of the isolated exosomes revealed distinct membrane-bound proteases, especially disintegrin and metalloproteinase 10 (ADAM10) and matrix metalloproteinase 25 (MMP25) commonly associated with ovarian cancer progression, are implicated in sE-cad production. Small interfering RNA-mediated down-regulation of ADAM10 and MMP25 significantly inhibited sE-cad production. Moreover, hepatocyte growth factor, a multifaceted cytokine which is frequently elevated in ovarian cancer ascites, was shown to increase the expression of ADAM10 and MMP25 concomitant with an elevated level of sE-cad. Together, these results uncover a novel angiogenic role of sE-cad and a new mechanism of the action of sE-cad in tumor progression. / published_or_final_version / Biological Sciences / Doctoral / Doctor of Philosophy

Identiferoai:union.ndltd.org:HKU/oai:hub.hku.hk:10722/202370
Date January 2014
CreatorsTang, Kei-shuen, 鄧紀旋
ContributorsWong, AST
PublisherThe University of Hong Kong (Pokfulam, Hong Kong)
Source SetsHong Kong University Theses
LanguageEnglish
Detected LanguageEnglish
TypePG_Thesis
RightsCreative Commons: Attribution 3.0 Hong Kong License, The author retains all proprietary rights, (such as patent rights) and the right to use in future works.
RelationHKU Theses Online (HKUTO)

Page generated in 0.0023 seconds