Please use this identifier to cite or link to this item: https://doi.org/10.3892/ijo.2012.1633
Title: The effect of Aurora kinases on cell proliferation, cell cycle regulation and metastasis in renal cell carcinoma
Authors: Li, Y
Zhou, W
Wei, L
Jin, J
Tang, K
Li, C
Teh, B.T 
Chen, X
Keywords: aurora A kinase
aurora B kinase
aurora C kinase
cyclin B
cyclin dependent kinase 1
microRNA
protein tyrosine phosphatase
tozasertib
animal model
animal tissue
antineoplastic activity
article
cell proliferation
cell viability
controlled study
down regulation
enzyme activity
enzyme inhibition
female
G2 phase cell cycle checkpoint
in vitro study
in vivo study
kidney carcinoma
metastasis
molecularly targeted therapy
nonhuman
priority journal
protein expression
tumor volume
tumor xenograft
upregulation
Animals
Carcinoma, Renal Cell
Cell Cycle
Cell Cycle Proteins
Cell Line, Tumor
Cell Proliferation
Female
Gene Expression Regulation, Neoplastic
Humans
Kidney Neoplasms
Mice
Mice, Nude
Neoplasm Metastasis
Piperazines
Protein-Serine-Threonine Kinases
RNA Interference
Transplantation, Heterologous
Tumor Burden
Issue Date: 2012
Citation: Li, Y, Zhou, W, Wei, L, Jin, J, Tang, K, Li, C, Teh, B.T, Chen, X (2012). The effect of Aurora kinases on cell proliferation, cell cycle regulation and metastasis in renal cell carcinoma. International Journal of Oncology 41 (6) : 2139-2149. ScholarBank@NUS Repository. https://doi.org/10.3892/ijo.2012.1633
Rights: Attribution 4.0 International
Abstract: Aurora kinases have been shown to be involved in the regulation of the cell cycle and are related to tumor progression. This suggests the possibility that they can serve as new anticancer targets for tumor treatment. However, the important roles that Aurora kinases and their signaling pathway play in renal cell carcinoma (RCC) are not fully understood and addressed to date. In this study, we aimed to address these questions. We observed that downregulation of Aurora kinases induced by AurA miRNA, AurB miRNA or VX680 could inhibit proliferation and metastasis, induce G2/M phase arrest in clear cell renal cell carcinoma cells and exert antitumor activity in an SN12C xenograft model. We also show that either silencing of Aurora kinases or treating the cells with VX680 could downregulate the expression of cdc25c and cyclin B/cdc2, upregulate the expression of p-cdc2 (Tyr15) via blocking the activity of ERK. All these changes may contribute to inhibition of proliferation, metastasis and G2/M arrest in ccRCC. In summary, we proved that both Aurora kinases A and B are key elements of tumor growth regulation, and inhibition of Aurora kinases may contribute to blocking ccRCC progression. We conclude that Aurora kinases could be potential therapeutic targets in the management of renal cell carcinoma.
Source Title: International Journal of Oncology
URI: https://scholarbank.nus.edu.sg/handle/10635/180815
ISSN: 1019-6439
DOI: 10.3892/ijo.2012.1633
Rights: Attribution 4.0 International
Appears in Collections:Staff Publications
Elements

Show full item record
Files in This Item:
File Description SizeFormatAccess SettingsVersion 
10_3892_ijo_2012_1633.pdf874.95 kBAdobe PDF

OPEN

NoneView/Download

Google ScholarTM

Check

Altmetric


This item is licensed under a Creative Commons License Creative Commons