Cargando…

Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells

BACKGROUND: Mirk/Dyrk1B contributes to G0 arrest by destabilization of cyclin D1 and stabilization of p27kip1 to maintain the viability of quiescent human cancer cells, and it could be negatively regulated by mitogenic-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signa...

Descripción completa

Detalles Bibliográficos
Autores principales: Gao, Jingchun, Zhao, Yi, Lv, Yunyi, Chen, Yamin, Wei, Bing, Tian, Jianxin, Yang, Zhihai, Kong, Fandou, Pang, Jian, Liu, Jiwei, Shi, Hong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3575355/
https://www.ncbi.nlm.nih.gov/pubmed/23311607
http://dx.doi.org/10.1186/1475-2867-13-2
_version_ 1782259709261119488
author Gao, Jingchun
Zhao, Yi
Lv, Yunyi
Chen, Yamin
Wei, Bing
Tian, Jianxin
Yang, Zhihai
Kong, Fandou
Pang, Jian
Liu, Jiwei
Shi, Hong
author_facet Gao, Jingchun
Zhao, Yi
Lv, Yunyi
Chen, Yamin
Wei, Bing
Tian, Jianxin
Yang, Zhihai
Kong, Fandou
Pang, Jian
Liu, Jiwei
Shi, Hong
author_sort Gao, Jingchun
collection PubMed
description BACKGROUND: Mirk/Dyrk1B contributes to G0 arrest by destabilization of cyclin D1 and stabilization of p27kip1 to maintain the viability of quiescent human cancer cells, and it could be negatively regulated by mitogenic-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling. This study was performed to investigate the effect of Mirk/Dyrk1B on cell cycle and survival of human cancer cells involving MAPK/ERK signaling. METHODS: The correlations between Mirk/Dyrk1B expression and active ERK1/2 detected by western blot in both ovarian cancer and non-small cell lung cancer (NSCLC) cells were analyzed by simple regression. Mirk/Dyrk1B unique phosphopeptides with sites associated with Mirk/Dyrk1B protein were isolated and quantitated by liquid chromatography coupled to tandem mass/mass spectrometry (LC-MS/MS) proteomics analysis. The human cancer cells were treated with small interfering RNAs (siRNAs) and/or U0126, an inhibitor of MEK for indicated duration, followed by investigating the alterations of cell cycle and apoptosis as well as related proteins examined by flow cytometry and Western blot, respectively. RESULTS: Our study demonstrated the widely expressed Mirk/Dyrk1B proteins in the human cancer cells were positively correlated with the levels of activated ERK1/2. Moreover, Mirk/Dyrk1B protein expressions consistent with the tyrosine autophosphorylated levels in the human cancer cells were increased by U0126 or growth factor-depleted culture. Conversely, knockdown of Mirk/Dyrk1B by siRNA led to up-regulated activation of c-Raf-MEK-ERK1/2 pathway and subsequent changes in cell cycle proteins (cyclin D1, p27kip1), accompanied by increased growth rate and cells from G0/G1 into S of cell cycle which could be blocked by U0126 in a dose-dependent manner, indicating Mirk/Dyrk1B may sequester MAPK/ERK pathway, and vice versa. Whereas, combined Mirk siRNA and U0126 induced cell apoptosis in the human cancer cells. CONCLUSIONS: These data together show that Mirk/Dyrk1B mediates cell cycle and survival via interacting with MAPK/ERK signals and simultaneous inhibition of both pathways may be a novel therapeutic target for human cancer.
format Online
Article
Text
id pubmed-3575355
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-35753552013-02-19 Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells Gao, Jingchun Zhao, Yi Lv, Yunyi Chen, Yamin Wei, Bing Tian, Jianxin Yang, Zhihai Kong, Fandou Pang, Jian Liu, Jiwei Shi, Hong Cancer Cell Int Primary Research BACKGROUND: Mirk/Dyrk1B contributes to G0 arrest by destabilization of cyclin D1 and stabilization of p27kip1 to maintain the viability of quiescent human cancer cells, and it could be negatively regulated by mitogenic-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling. This study was performed to investigate the effect of Mirk/Dyrk1B on cell cycle and survival of human cancer cells involving MAPK/ERK signaling. METHODS: The correlations between Mirk/Dyrk1B expression and active ERK1/2 detected by western blot in both ovarian cancer and non-small cell lung cancer (NSCLC) cells were analyzed by simple regression. Mirk/Dyrk1B unique phosphopeptides with sites associated with Mirk/Dyrk1B protein were isolated and quantitated by liquid chromatography coupled to tandem mass/mass spectrometry (LC-MS/MS) proteomics analysis. The human cancer cells were treated with small interfering RNAs (siRNAs) and/or U0126, an inhibitor of MEK for indicated duration, followed by investigating the alterations of cell cycle and apoptosis as well as related proteins examined by flow cytometry and Western blot, respectively. RESULTS: Our study demonstrated the widely expressed Mirk/Dyrk1B proteins in the human cancer cells were positively correlated with the levels of activated ERK1/2. Moreover, Mirk/Dyrk1B protein expressions consistent with the tyrosine autophosphorylated levels in the human cancer cells were increased by U0126 or growth factor-depleted culture. Conversely, knockdown of Mirk/Dyrk1B by siRNA led to up-regulated activation of c-Raf-MEK-ERK1/2 pathway and subsequent changes in cell cycle proteins (cyclin D1, p27kip1), accompanied by increased growth rate and cells from G0/G1 into S of cell cycle which could be blocked by U0126 in a dose-dependent manner, indicating Mirk/Dyrk1B may sequester MAPK/ERK pathway, and vice versa. Whereas, combined Mirk siRNA and U0126 induced cell apoptosis in the human cancer cells. CONCLUSIONS: These data together show that Mirk/Dyrk1B mediates cell cycle and survival via interacting with MAPK/ERK signals and simultaneous inhibition of both pathways may be a novel therapeutic target for human cancer. BioMed Central 2013-01-11 /pmc/articles/PMC3575355/ /pubmed/23311607 http://dx.doi.org/10.1186/1475-2867-13-2 Text en Copyright ©2013 Gao et al.; licensee BioMed Central Ltd. http://creativecommons.org/licenses/by/2.0 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Primary Research
Gao, Jingchun
Zhao, Yi
Lv, Yunyi
Chen, Yamin
Wei, Bing
Tian, Jianxin
Yang, Zhihai
Kong, Fandou
Pang, Jian
Liu, Jiwei
Shi, Hong
Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells
title Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells
title_full Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells
title_fullStr Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells
title_full_unstemmed Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells
title_short Mirk/Dyrk1B mediates G0/G1 to S phase cell cycle progression and cell survival involving MAPK/ERK signaling in human cancer cells
title_sort mirk/dyrk1b mediates g0/g1 to s phase cell cycle progression and cell survival involving mapk/erk signaling in human cancer cells
topic Primary Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3575355/
https://www.ncbi.nlm.nih.gov/pubmed/23311607
http://dx.doi.org/10.1186/1475-2867-13-2
work_keys_str_mv AT gaojingchun mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT zhaoyi mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT lvyunyi mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT chenyamin mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT weibing mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT tianjianxin mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT yangzhihai mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT kongfandou mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT pangjian mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT liujiwei mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells
AT shihong mirkdyrk1bmediatesg0g1tosphasecellcycleprogressionandcellsurvivalinvolvingmapkerksignalinginhumancancercells