Cargando…

Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death

Runt-related transcription factor 2 (RUNX2) has been considered to be one of master regulators for osteoblast differentiation and bone formation. Recently, we have described that RUNX2 attenuates p53/TAp73-dependent cell death of human osteosarcoma U2OS cells bearing wild-type p53 in response to adr...

Descripción completa

Detalles Bibliográficos
Autores principales: Sugimoto, H, Nakamura, M, Yoda, H, Hiraoka, K, Shinohara, K, Sang, M, Fujiwara, K, Shimozato, O, Nagase, H, Ozaki, T
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4981025/
https://www.ncbi.nlm.nih.gov/pubmed/27551445
http://dx.doi.org/10.1038/cddiscovery.2015.10
_version_ 1782447557640716288
author Sugimoto, H
Nakamura, M
Yoda, H
Hiraoka, K
Shinohara, K
Sang, M
Fujiwara, K
Shimozato, O
Nagase, H
Ozaki, T
author_facet Sugimoto, H
Nakamura, M
Yoda, H
Hiraoka, K
Shinohara, K
Sang, M
Fujiwara, K
Shimozato, O
Nagase, H
Ozaki, T
author_sort Sugimoto, H
collection PubMed
description Runt-related transcription factor 2 (RUNX2) has been considered to be one of master regulators for osteoblast differentiation and bone formation. Recently, we have described that RUNX2 attenuates p53/TAp73-dependent cell death of human osteosarcoma U2OS cells bearing wild-type p53 in response to adriamycin. In this study, we have asked whether RUNX2 silencing could enhance gemcitabine (GEM) sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells. Under our experimental conditions, GEM treatment increased the expression level of p53 family TAp63, whereas RUNX2 was reduced following GEM exposure, indicating that there exists an inverse relationship between the expression level of TAp63 and RUNX2 following GEM exposure. To assess whether TAp63 could be involved in the regulation of GEM sensitivity of AsPC-1 cells, small interfering RNA-mediated knockdown of TAp63 was performed. As expected, silencing of TAp63 significantly prohibited GEM-dependent cell death as compared with GEM-treated non-silencing cells. As TAp63 was negatively regulated by RUNX2, we sought to examine whether RUNX2 knockdown could enhance the sensitivity to GEM. Expression analysis demonstrated that depletion of RUNX2 apparently stimulates the expression of TAp63, as well as proteolytic cleavage of poly ADP ribose polymerase (PARP) after GEM exposure, and further augmented GEM-mediated induction of p53/TAp63-target genes, such as p21(WAF1), PUMA and NOXA, relative to GEM-treated control-transfected cells, implying that RUNX2 has a critical role in the regulation of GEM resistance through the downregulation of TAp63. Notably, ablation of TAp63 gave a decrease in number of γH2AX-positive cells in response to GEM relative to control-transfected cells following GEM exposure. Consistently, GEM-dependent phosphorylation of ataxia telangiectasia-mutated protein was remarkably impaired in TAp63 knockdown cells. Collectively, our present findings strongly suggest that RUNX2-mediated repression of TAp63 contributes at least in part to GEM resistance of AsPC-1 cells, and thus silencing of RUNX2 may be a novel strategy to enhance the efficacy of GEM in p53-deficient pancreatic cancer cells.
format Online
Article
Text
id pubmed-4981025
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-49810252016-08-22 Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death Sugimoto, H Nakamura, M Yoda, H Hiraoka, K Shinohara, K Sang, M Fujiwara, K Shimozato, O Nagase, H Ozaki, T Cell Death Discov Article Runt-related transcription factor 2 (RUNX2) has been considered to be one of master regulators for osteoblast differentiation and bone formation. Recently, we have described that RUNX2 attenuates p53/TAp73-dependent cell death of human osteosarcoma U2OS cells bearing wild-type p53 in response to adriamycin. In this study, we have asked whether RUNX2 silencing could enhance gemcitabine (GEM) sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells. Under our experimental conditions, GEM treatment increased the expression level of p53 family TAp63, whereas RUNX2 was reduced following GEM exposure, indicating that there exists an inverse relationship between the expression level of TAp63 and RUNX2 following GEM exposure. To assess whether TAp63 could be involved in the regulation of GEM sensitivity of AsPC-1 cells, small interfering RNA-mediated knockdown of TAp63 was performed. As expected, silencing of TAp63 significantly prohibited GEM-dependent cell death as compared with GEM-treated non-silencing cells. As TAp63 was negatively regulated by RUNX2, we sought to examine whether RUNX2 knockdown could enhance the sensitivity to GEM. Expression analysis demonstrated that depletion of RUNX2 apparently stimulates the expression of TAp63, as well as proteolytic cleavage of poly ADP ribose polymerase (PARP) after GEM exposure, and further augmented GEM-mediated induction of p53/TAp63-target genes, such as p21(WAF1), PUMA and NOXA, relative to GEM-treated control-transfected cells, implying that RUNX2 has a critical role in the regulation of GEM resistance through the downregulation of TAp63. Notably, ablation of TAp63 gave a decrease in number of γH2AX-positive cells in response to GEM relative to control-transfected cells following GEM exposure. Consistently, GEM-dependent phosphorylation of ataxia telangiectasia-mutated protein was remarkably impaired in TAp63 knockdown cells. Collectively, our present findings strongly suggest that RUNX2-mediated repression of TAp63 contributes at least in part to GEM resistance of AsPC-1 cells, and thus silencing of RUNX2 may be a novel strategy to enhance the efficacy of GEM in p53-deficient pancreatic cancer cells. Nature Publishing Group 2015-08-10 /pmc/articles/PMC4981025/ /pubmed/27551445 http://dx.doi.org/10.1038/cddiscovery.2015.10 Text en Copyright © 2015 Cell Death Differentiation Association http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Sugimoto, H
Nakamura, M
Yoda, H
Hiraoka, K
Shinohara, K
Sang, M
Fujiwara, K
Shimozato, O
Nagase, H
Ozaki, T
Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death
title Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death
title_full Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death
title_fullStr Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death
title_full_unstemmed Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death
title_short Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death
title_sort silencing of runx2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer aspc-1 cells through the stimulation of tap63-mediated cell death
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4981025/
https://www.ncbi.nlm.nih.gov/pubmed/27551445
http://dx.doi.org/10.1038/cddiscovery.2015.10
work_keys_str_mv AT sugimotoh silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT nakamuram silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT yodah silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT hiraokak silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT shinoharak silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT sangm silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT fujiwarak silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT shimozatoo silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT nagaseh silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath
AT ozakit silencingofrunx2enhancesgemcitabinesensitivityofp53deficienthumanpancreaticcanceraspc1cellsthroughthestimulationoftap63mediatedcelldeath