Cargando…

Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells

Endometrial cancer is the most commonly diagnosed gynecologic malignancy worldwide; yet the tumor microenvironment, especially the fibroblast cells surrounding the cancer cells, is poorly understood. We established four primary cultures of fibroblasts from human endometrial cancer tissues (cancer-as...

Descripción completa

Detalles Bibliográficos
Autores principales: Subramaniam, Kavita S., Tham, Seng Tian, Mohamed, Zahurin, Woo, Yin Ling, Mat Adenan, Noor Azmi, Chung, Ivy
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3724864/
https://www.ncbi.nlm.nih.gov/pubmed/23922669
http://dx.doi.org/10.1371/journal.pone.0068923
_version_ 1782476738460123136
author Subramaniam, Kavita S.
Tham, Seng Tian
Mohamed, Zahurin
Woo, Yin Ling
Mat Adenan, Noor Azmi
Chung, Ivy
author_facet Subramaniam, Kavita S.
Tham, Seng Tian
Mohamed, Zahurin
Woo, Yin Ling
Mat Adenan, Noor Azmi
Chung, Ivy
author_sort Subramaniam, Kavita S.
collection PubMed
description Endometrial cancer is the most commonly diagnosed gynecologic malignancy worldwide; yet the tumor microenvironment, especially the fibroblast cells surrounding the cancer cells, is poorly understood. We established four primary cultures of fibroblasts from human endometrial cancer tissues (cancer-associated fibroblasts, CAFs) using antibody-conjugated magnetic bead isolation. These relatively homogenous fibroblast cultures expressed fibroblast markers (CD90, vimentin and alpha-smooth muscle actin) and hormonal (estrogen and progesterone) receptors. Conditioned media collected from CAFs induced a dose-dependent proliferation of both primary cultures and cell lines of endometrial cancer in vitro (175%) when compared to non-treated cells, in contrast to those from normal endometrial fibroblast cell line (51%) (P<0.0001). These effects were not observed in fibroblast culture derived from benign endometrial hyperplasia tissues, indicating the specificity of CAFs in affecting endometrial cancer cell proliferation. To determine the mechanism underlying the differential fibroblast effects, we compared the activation of PI3K/Akt and MAPK/Erk pathways in endometrial cancer cells following treatment with normal fibroblasts- and CAFs-conditioned media. Western blot analysis showed that the expression of both phosphorylated forms of Akt and Erk were significantly down-regulated in normal fibroblasts-treated cells, but were up-regulated/maintained in CAFs-treated cells. Treatment with specific inhibitors LY294002 and U0126 reversed the CAFs-mediated cell proliferation (P<0.0001), suggesting for a role of these pathways in modulating endometrial cancer cell proliferation. Rapamycin, which targets a downstream molecule in PI3K pathway (mTOR), also suppressed CAFs-induced cell proliferation by inducing apoptosis. Cytokine profiling analysis revealed that CAFs secrete higher levels of macrophage chemoattractant protein (MCP)-1, interleukin (IL)-6, IL-8, RANTES and vascular endothelial growth factor (VEGF) than normal fibroblasts. Our data suggests that in contrast to normal fibroblasts, CAFs may exhibit a pro-tumorigenic effect in the progression of endometrial cancer, and PI3K/Akt and MAPK/Erk signaling may represent critical regulators in how endometrial cancer cells respond to their microenvironment.
format Online
Article
Text
id pubmed-3724864
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-37248642013-08-06 Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells Subramaniam, Kavita S. Tham, Seng Tian Mohamed, Zahurin Woo, Yin Ling Mat Adenan, Noor Azmi Chung, Ivy PLoS One Research Article Endometrial cancer is the most commonly diagnosed gynecologic malignancy worldwide; yet the tumor microenvironment, especially the fibroblast cells surrounding the cancer cells, is poorly understood. We established four primary cultures of fibroblasts from human endometrial cancer tissues (cancer-associated fibroblasts, CAFs) using antibody-conjugated magnetic bead isolation. These relatively homogenous fibroblast cultures expressed fibroblast markers (CD90, vimentin and alpha-smooth muscle actin) and hormonal (estrogen and progesterone) receptors. Conditioned media collected from CAFs induced a dose-dependent proliferation of both primary cultures and cell lines of endometrial cancer in vitro (175%) when compared to non-treated cells, in contrast to those from normal endometrial fibroblast cell line (51%) (P<0.0001). These effects were not observed in fibroblast culture derived from benign endometrial hyperplasia tissues, indicating the specificity of CAFs in affecting endometrial cancer cell proliferation. To determine the mechanism underlying the differential fibroblast effects, we compared the activation of PI3K/Akt and MAPK/Erk pathways in endometrial cancer cells following treatment with normal fibroblasts- and CAFs-conditioned media. Western blot analysis showed that the expression of both phosphorylated forms of Akt and Erk were significantly down-regulated in normal fibroblasts-treated cells, but were up-regulated/maintained in CAFs-treated cells. Treatment with specific inhibitors LY294002 and U0126 reversed the CAFs-mediated cell proliferation (P<0.0001), suggesting for a role of these pathways in modulating endometrial cancer cell proliferation. Rapamycin, which targets a downstream molecule in PI3K pathway (mTOR), also suppressed CAFs-induced cell proliferation by inducing apoptosis. Cytokine profiling analysis revealed that CAFs secrete higher levels of macrophage chemoattractant protein (MCP)-1, interleukin (IL)-6, IL-8, RANTES and vascular endothelial growth factor (VEGF) than normal fibroblasts. Our data suggests that in contrast to normal fibroblasts, CAFs may exhibit a pro-tumorigenic effect in the progression of endometrial cancer, and PI3K/Akt and MAPK/Erk signaling may represent critical regulators in how endometrial cancer cells respond to their microenvironment. Public Library of Science 2013-07-26 /pmc/articles/PMC3724864/ /pubmed/23922669 http://dx.doi.org/10.1371/journal.pone.0068923 Text en © 2013 Subramaniam et al http://creativecommons.org/licenses/by/4.0/ 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 properly credited.
spellingShingle Research Article
Subramaniam, Kavita S.
Tham, Seng Tian
Mohamed, Zahurin
Woo, Yin Ling
Mat Adenan, Noor Azmi
Chung, Ivy
Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells
title Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells
title_full Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells
title_fullStr Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells
title_full_unstemmed Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells
title_short Cancer-Associated Fibroblasts Promote Proliferation of Endometrial Cancer Cells
title_sort cancer-associated fibroblasts promote proliferation of endometrial cancer cells
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3724864/
https://www.ncbi.nlm.nih.gov/pubmed/23922669
http://dx.doi.org/10.1371/journal.pone.0068923
work_keys_str_mv AT subramaniamkavitas cancerassociatedfibroblastspromoteproliferationofendometrialcancercells
AT thamsengtian cancerassociatedfibroblastspromoteproliferationofendometrialcancercells
AT mohamedzahurin cancerassociatedfibroblastspromoteproliferationofendometrialcancercells
AT wooyinling cancerassociatedfibroblastspromoteproliferationofendometrialcancercells
AT matadenannoorazmi cancerassociatedfibroblastspromoteproliferationofendometrialcancercells
AT chungivy cancerassociatedfibroblastspromoteproliferationofendometrialcancercells