Unknown

Dataset Information

0

IL-3R-alpha blockade inhibits tumor endothelial cell-derived extracellular vesicle (EV)-mediated vessel formation by targeting the ?-catenin pathway.


ABSTRACT: The proangiogenic cytokine Interleukin-3 (IL-3) is released by inflammatory cells in breast and ovarian cancer tissue microenvironments and also acts as an autocrine factor for human breast and kidney tumor-derived endothelial cells (TECs). We have previously shown that IL-3-treated endothelial cells (ECs) release extracellular vesicles (EVs), which serve as a paracrine mechanism for neighboring ECs, by transferring active molecules. The impact of an anti-IL-3R-alpha blocking antibody on the proangiogenic effect of EVs released from TECs (anti-IL-3R-EVs) has therefore been investigated in this study. We have found that anti-IL-3R-EV treatment prevented neovessel formation and, more importantly, also induced the regression of in vivo TEC-derived neovessels. Two miRs that target the canonical wingless (Wnt)/?-catenin pathway, at different levels, were found to be differentially regulated when comparing the miR-cargo of naive TEC-derived EVs (EVs) and anti-IL-3R-EVs. miR-214-3p, which directly targets ?-catenin, was found to be upregulated, whereas miR-24-3p, which targets adenomatous polyposis coli (APC) and glycogen synthase kinase-3? (GSK3?), was found to be downregulated. In fact, upon their transfer into the cell, low ?-catenin content and high levels of the two members of the "?-catenin destruction complex" were detected. Moreover, c-myc downregulation was found in TECs treated with anti-IL-3R-EVs, pre-miR-214-3p-EVs and antago-miR-24-3p-EVs, which is consistent with network analyses of miR-214-3p and miR-24-3p gene targeting. Finally, in vivo studies have demonstrated the impaired growth of vessels in pre-miR-214-3p-EV- and antago-miR-24-3p-EV-treated animals. These effects became much more evident when combo treatment was applied. The results of the present study identify the canonical Wnt/?-catenin pathway as a relevant mechanism of TEC-derived EV proangiogenic action. Furthermore, we herein provide evidence that IL-3R blockade may yield some significant advantages, than miR targeting, in inhibiting the proangiogenic effects of naive TEC-derived EVs by changing TEC-EV-miR cargo.

SUBMITTER: Lombardo G 

PROVIDER: S-EPMC5861089 | biostudies-other | 2018 Mar

REPOSITORIES: biostudies-other

altmetric image

Publications

IL-3R-alpha blockade inhibits tumor endothelial cell-derived extracellular vesicle (EV)-mediated vessel formation by targeting the β-catenin pathway.

Lombardo Giusy G   Gili Maddalena M   Grange Cristina C   Cavallari Claudia C   Dentelli Patrizia P   Togliatto Gabriele G   Taverna Daniela D   Camussi Giovanni G   Brizzi Maria Felice MF  

Oncogene 20171214 9


The proangiogenic cytokine Interleukin-3 (IL-3) is released by inflammatory cells in breast and ovarian cancer tissue microenvironments and also acts as an autocrine factor for human breast and kidney tumor-derived endothelial cells (TECs). We have previously shown that IL-3-treated endothelial cells (ECs) release extracellular vesicles (EVs), which serve as a paracrine mechanism for neighboring ECs, by transferring active molecules. The impact of an anti-IL-3R-alpha blocking antibody on the pro  ...[more]

Similar Datasets

| S-EPMC6246790 | biostudies-literature
| S-EPMC8254805 | biostudies-literature
| S-EPMC4393420 | biostudies-literature
| S-EPMC9266738 | biostudies-literature
| S-EPMC5341127 | biostudies-literature
| S-EPMC7034442 | biostudies-literature
| S-EPMC6479947 | biostudies-literature
| S-EPMC7726798 | biostudies-literature
| S-EPMC6733813 | biostudies-other
| S-EPMC5326622 | biostudies-literature