Cargando…

Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation

Although purinergic signaling is important in regulation of immune responses, the therapeutic potential of it in the tumor microenvironment is little defined. In this study, we demonstrate that UDP/P2Y(6) signaling facilitates breast cancer metastasis both in vitro and in vivo. We found that P2Y(6)...

Descripción completa

Detalles Bibliográficos
Autores principales: Ma, Xiaobin, Pan, Xinhua, Wei, Yinglei, Tan, Binhe, Yang, Linli, Ren, Hua, Qian, Min, Du, Bing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5045376/
https://www.ncbi.nlm.nih.gov/pubmed/27074554
http://dx.doi.org/10.18632/oncotarget.8664
_version_ 1782457104684023808
author Ma, Xiaobin
Pan, Xinhua
Wei, Yinglei
Tan, Binhe
Yang, Linli
Ren, Hua
Qian, Min
Du, Bing
author_facet Ma, Xiaobin
Pan, Xinhua
Wei, Yinglei
Tan, Binhe
Yang, Linli
Ren, Hua
Qian, Min
Du, Bing
author_sort Ma, Xiaobin
collection PubMed
description Although purinergic signaling is important in regulation of immune responses, the therapeutic potential of it in the tumor microenvironment is little defined. In this study, we demonstrate that UDP/P2Y(6) signaling facilitates breast cancer metastasis both in vitro and in vivo. We found that P2Y(6) is not only aberrantly expressed and mutated in most tumor types, but also highly correlated with poor prognosis in breast cancer patients. Furthermore, the migration and invasion of breast cancer cells was obviously increased by UDP and blocked by P2Y(6) specific inhibitor MRS2578 and P2Y(6) shRNA. Similar results was also found in breast cancer cell metastasis mouse model. Interestingly, the endogenous agonist UDP was released significantly by doxorubicin treated cells. In addition, the expression and enzyme activity of MMP-9 were both promoted by UDP and inhibited by MRS2578 or P2Y(6) shRNA. Furthermore, UDP-induced cell invasion was blocked by an MMP-9 inhibitor. Mechanistically, the MAPKs and NF-κB signaling pathways, known to be involved in regulation of MMP-9 expression, were both activated by UDP. Taken together, our study reveals a relationship between extracellular danger signals and breast cancer metastasis, which suggests the potential therapeutic significance of UDP/P2Y(6) signaling in cancer therapy.
format Online
Article
Text
id pubmed-5045376
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-50453762016-10-13 Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation Ma, Xiaobin Pan, Xinhua Wei, Yinglei Tan, Binhe Yang, Linli Ren, Hua Qian, Min Du, Bing Oncotarget Research Paper Although purinergic signaling is important in regulation of immune responses, the therapeutic potential of it in the tumor microenvironment is little defined. In this study, we demonstrate that UDP/P2Y(6) signaling facilitates breast cancer metastasis both in vitro and in vivo. We found that P2Y(6) is not only aberrantly expressed and mutated in most tumor types, but also highly correlated with poor prognosis in breast cancer patients. Furthermore, the migration and invasion of breast cancer cells was obviously increased by UDP and blocked by P2Y(6) specific inhibitor MRS2578 and P2Y(6) shRNA. Similar results was also found in breast cancer cell metastasis mouse model. Interestingly, the endogenous agonist UDP was released significantly by doxorubicin treated cells. In addition, the expression and enzyme activity of MMP-9 were both promoted by UDP and inhibited by MRS2578 or P2Y(6) shRNA. Furthermore, UDP-induced cell invasion was blocked by an MMP-9 inhibitor. Mechanistically, the MAPKs and NF-κB signaling pathways, known to be involved in regulation of MMP-9 expression, were both activated by UDP. Taken together, our study reveals a relationship between extracellular danger signals and breast cancer metastasis, which suggests the potential therapeutic significance of UDP/P2Y(6) signaling in cancer therapy. Impact Journals LLC 2016-04-09 /pmc/articles/PMC5045376/ /pubmed/27074554 http://dx.doi.org/10.18632/oncotarget.8664 Text en Copyright: © 2016 Ma et al. http://creativecommons.org/licenses/by/2.5/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Ma, Xiaobin
Pan, Xinhua
Wei, Yinglei
Tan, Binhe
Yang, Linli
Ren, Hua
Qian, Min
Du, Bing
Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation
title Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation
title_full Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation
title_fullStr Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation
title_full_unstemmed Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation
title_short Chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through P2Y(6) activation
title_sort chemotherapy-induced uridine diphosphate release promotes breast cancer metastasis through p2y(6) activation
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5045376/
https://www.ncbi.nlm.nih.gov/pubmed/27074554
http://dx.doi.org/10.18632/oncotarget.8664
work_keys_str_mv AT maxiaobin chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT panxinhua chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT weiyinglei chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT tanbinhe chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT yanglinli chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT renhua chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT qianmin chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation
AT dubing chemotherapyinduceduridinediphosphatereleasepromotesbreastcancermetastasisthroughp2y6activation