Abstract

Abstract Treating metastatic disease remains a major obstacle in oncology. Neuropeptide Y (NPY) and its Y5 receptor (Y5R) are expressed in a number of malignancies, including pediatric tumors Ewing sarcoma (ES) and neuroblastoma (NB). Our previous clinical and experimental data implicated NPY/Y5R axis in ES and NB metastasis. We have shown that Y5R is highly expressed in angioinvasive NB cells, while elevated release of NPY from NB tumors associates with metastatic disease and poor clinical outcome. Additionally, hypoxia-induced over-activation of NPY/Y5R pathway in ES tumors, which express particularly high levels of Y5R, led to formation of polyploid cells that give rise to chromosomally unstable, metastatic progeny. This phenomenon was reproduced in CHO-K1 cells over-expressing Y5R, which became polyploid due to a cytokinesis defect. Altogether, our data suggested a role of the NPY/Y5R pathway in the regulation of cell migration and cytokinesis, processes essential in tumor growth and dissemination. Thus, the goal of our study was to determine mechanisms of NPY/Y5R actions. We have found that in both CHO-K1/Y5R transfectants and ES cells, Y5R stimulation activated an essential cytoskeleton regulator, RhoA, activity of which is tightly controlled in a spatial and temporal manner during both cell migration and cytokinesis. This data was confirmed by strong co-localization of Y5R and active RhoA in all cell types. In non-dividing cells, expression of Y5R and active RhoA was limited, while their levels were higher in mitotic cells. Notably, in CHO-K1 cells over-expressing Y5R, both Y5R and RhoA accumulated in the narrow zone of the cleavage furrow at the abscission phase of cytokinesis. Such RhoA activation at this stage was previously shown to trigger cytokinesis defects. The concurrent changes in subcellular localization of Y5R and active RhoA were also observed in migratory cells. In single cell migration, both Y5R and active RhoA accumulated on the trailing and leading edges of the cells, while in the cells migrating collectively their expression was elevated in those present at the front of the moving colony. Strong Y5R/active Rho-A co-localization was also observed in membrane blebs of cells in amoebal migration mode. The role of Y5R in stimulation of cell migration was confirmed by transwell assay, in which Y5R antagonist significantly inhibited NPY-induced migration of NB and ES cells. Altogether, our data support the role of NPY/Y5R/RhoA pathway in regulation of cytoskeleton functions. The stimulation of this pathway promotes cell migration, while its over-activation may lead to cytokinesis defects and chromosomal instability, both of which are essential processes in tumor progression. Further studies are required to determine if these functions of the NPY/Y5R/RhoA pathway contribute to metastasis and validate Y5R as a potential therapeutic target. Citation Format: Nouran Abualsaud, Congyi Lu, Akanksha Mahajan, Abrar Bakr, Shiya Zhu, Lindsay Caprio, Sung Hyeok Hong, Joanna Kitlinska. The role of neuropeptide Y and its Y5 receptor in RhoA-mediated regulation of cytokinesis and cell motility [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4143.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call