Cargando…

NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling

Neuromedin U (NMU) was originally named based on its strong uterine contractile activity, but little is known regarding its signaling/functions in utero. We identified that NMU and one of its receptors, NMUR2, are not only present in normal uterine endometrium but also co-expressed in endometrial ca...

Descripción completa

Detalles Bibliográficos
Autores principales: Lin, Ting-Yu, Wu, Fang-Ju, Chang, Chia-Lin, Li, Zhongyou, Luo, Ching-Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4891116/
https://www.ncbi.nlm.nih.gov/pubmed/26849234
http://dx.doi.org/10.18632/oncotarget.7169
_version_ 1782435225149636608
author Lin, Ting-Yu
Wu, Fang-Ju
Chang, Chia-Lin
Li, Zhongyou
Luo, Ching-Wei
author_facet Lin, Ting-Yu
Wu, Fang-Ju
Chang, Chia-Lin
Li, Zhongyou
Luo, Ching-Wei
author_sort Lin, Ting-Yu
collection PubMed
description Neuromedin U (NMU) was originally named based on its strong uterine contractile activity, but little is known regarding its signaling/functions in utero. We identified that NMU and one of its receptors, NMUR2, are not only present in normal uterine endometrium but also co-expressed in endometrial cancer tissues, where the NMU level is correlated with the malignant grades and survival of patients. Cell-based assays further confirmed that NMU signaling can promote cell motility and proliferation of endometrial cancer cells derived from grade II tumors. Activation of NMU pathway in these endometrial cancer cells is required in order to sustain expression of various adhesion molecules, such as CD44 and integrin alpha1, as well as production of their corresponding extracellular matrix ligands, hyaluronan and collagen IV; it also increased the activity of SRC and its downstream proteins RHOA and RAC1. Thus, it is concluded that NMU pathway positively controls the adhesion signaling-SRC-Rho GTPase axis in the tested endometrial cancer cells and that changes in cell motility and proliferation can occur when there is manipulation of NMU signaling in these cells either in vitro or in vivo. Intriguingly, this novel mechanism also explains how NMU signaling promotes the EGFR-driven and TGFβ receptor-driven mesenchymal transitions. Through the above axis, NMU signaling not only can promote malignancy of the tested endometrial cancer cells directly, but also helps these cells to become more sensitive to niche growth factors in their microenvironment.
format Online
Article
Text
id pubmed-4891116
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-48911162016-06-23 NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling Lin, Ting-Yu Wu, Fang-Ju Chang, Chia-Lin Li, Zhongyou Luo, Ching-Wei Oncotarget Research Paper Neuromedin U (NMU) was originally named based on its strong uterine contractile activity, but little is known regarding its signaling/functions in utero. We identified that NMU and one of its receptors, NMUR2, are not only present in normal uterine endometrium but also co-expressed in endometrial cancer tissues, where the NMU level is correlated with the malignant grades and survival of patients. Cell-based assays further confirmed that NMU signaling can promote cell motility and proliferation of endometrial cancer cells derived from grade II tumors. Activation of NMU pathway in these endometrial cancer cells is required in order to sustain expression of various adhesion molecules, such as CD44 and integrin alpha1, as well as production of their corresponding extracellular matrix ligands, hyaluronan and collagen IV; it also increased the activity of SRC and its downstream proteins RHOA and RAC1. Thus, it is concluded that NMU pathway positively controls the adhesion signaling-SRC-Rho GTPase axis in the tested endometrial cancer cells and that changes in cell motility and proliferation can occur when there is manipulation of NMU signaling in these cells either in vitro or in vivo. Intriguingly, this novel mechanism also explains how NMU signaling promotes the EGFR-driven and TGFβ receptor-driven mesenchymal transitions. Through the above axis, NMU signaling not only can promote malignancy of the tested endometrial cancer cells directly, but also helps these cells to become more sensitive to niche growth factors in their microenvironment. Impact Journals LLC 2016-02-03 /pmc/articles/PMC4891116/ /pubmed/26849234 http://dx.doi.org/10.18632/oncotarget.7169 Text en Copyright: © 2016 Lin 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
Lin, Ting-Yu
Wu, Fang-Ju
Chang, Chia-Lin
Li, Zhongyou
Luo, Ching-Wei
NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling
title NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling
title_full NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling
title_fullStr NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling
title_full_unstemmed NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling
title_short NMU signaling promotes endometrial cancer cell progression by modulating adhesion signaling
title_sort nmu signaling promotes endometrial cancer cell progression by modulating adhesion signaling
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4891116/
https://www.ncbi.nlm.nih.gov/pubmed/26849234
http://dx.doi.org/10.18632/oncotarget.7169
work_keys_str_mv AT lintingyu nmusignalingpromotesendometrialcancercellprogressionbymodulatingadhesionsignaling
AT wufangju nmusignalingpromotesendometrialcancercellprogressionbymodulatingadhesionsignaling
AT changchialin nmusignalingpromotesendometrialcancercellprogressionbymodulatingadhesionsignaling
AT lizhongyou nmusignalingpromotesendometrialcancercellprogressionbymodulatingadhesionsignaling
AT luochingwei nmusignalingpromotesendometrialcancercellprogressionbymodulatingadhesionsignaling