Cargando…

The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells

The molecular basis of epithelial ovarian cancer (EOC) dissemination is still poorly understood. Previously, we identified the mannose receptor LY75 gene as hypomethylated in high-grade (HG) serous EOC tumors, compared to normal ovarian tissues. LY75 represents endocytic receptor expressed on dendri...

Descripción completa

Detalles Bibliográficos
Autores principales: Faddaoui, Adnen, Bachvarova, Magdalena, Plante, Marie, Gregoire, Jean, Renaud, Marie-Claude, Sebastianelli, Alexandra, Gobeil, Stephane, Morin, Chantale, Macdonald, Elizabeth, Vanderhyden, Barbara, Bachvarov, Dimcho
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4924702/
https://www.ncbi.nlm.nih.gov/pubmed/26871602
http://dx.doi.org/10.18632/oncotarget.7288
_version_ 1782439905679376384
author Faddaoui, Adnen
Bachvarova, Magdalena
Plante, Marie
Gregoire, Jean
Renaud, Marie-Claude
Sebastianelli, Alexandra
Gobeil, Stephane
Morin, Chantale
Macdonald, Elizabeth
Vanderhyden, Barbara
Bachvarov, Dimcho
author_facet Faddaoui, Adnen
Bachvarova, Magdalena
Plante, Marie
Gregoire, Jean
Renaud, Marie-Claude
Sebastianelli, Alexandra
Gobeil, Stephane
Morin, Chantale
Macdonald, Elizabeth
Vanderhyden, Barbara
Bachvarov, Dimcho
author_sort Faddaoui, Adnen
collection PubMed
description The molecular basis of epithelial ovarian cancer (EOC) dissemination is still poorly understood. Previously, we identified the mannose receptor LY75 gene as hypomethylated in high-grade (HG) serous EOC tumors, compared to normal ovarian tissues. LY75 represents endocytic receptor expressed on dendritic cells and so far, has been primarily studied for its role in antigen processing and presentation. Here we demonstrate that LY75 is overexpressed in advanced EOC and that LY75 suppression induces mesenchymal-to-epithelial transition (MET) in EOC cell lines with mesenchymal morphology (SKOV3 and TOV112), accompanied by reduction of their migratory and invasive capacity in vitro and enhanced tumor cell colonization and metastatic growth in vivo. LY75 knockdown in SKOV3 cells also resulted in predominant upregulation of functional pathways implicated in cell proliferation and metabolism, while pathways associated with cell signaling and adhesion, complement activation and immune response were mostly suppressed. Moreover, LY75 suppression had an opposite effect on EOC cell lines with epithelial phenotype (A2780s and OV2008), by directing epithelial-to-mesenchymal transition (EMT) associated with reduced capacity for in vivo EOC cell colonization, as similar/identical signaling pathways were reversely regulated, when compared to mesenchymal LY75 knockdown EOC cells. To our knowledge, this is the first report of a gene displaying such pleiotropic effects in sustaining the cellular phenotype of EOC cells and points to novel functions of this receptor in modulating EOC dissemination. Our data also support previous findings regarding the superior capacity of epithelial cancer cells in metastatic colonization of distant sites, compared to cancer cells with mesenchymal-like morphology.
format Online
Article
Text
id pubmed-4924702
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-49247022016-07-13 The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells Faddaoui, Adnen Bachvarova, Magdalena Plante, Marie Gregoire, Jean Renaud, Marie-Claude Sebastianelli, Alexandra Gobeil, Stephane Morin, Chantale Macdonald, Elizabeth Vanderhyden, Barbara Bachvarov, Dimcho Oncotarget Research Paper The molecular basis of epithelial ovarian cancer (EOC) dissemination is still poorly understood. Previously, we identified the mannose receptor LY75 gene as hypomethylated in high-grade (HG) serous EOC tumors, compared to normal ovarian tissues. LY75 represents endocytic receptor expressed on dendritic cells and so far, has been primarily studied for its role in antigen processing and presentation. Here we demonstrate that LY75 is overexpressed in advanced EOC and that LY75 suppression induces mesenchymal-to-epithelial transition (MET) in EOC cell lines with mesenchymal morphology (SKOV3 and TOV112), accompanied by reduction of their migratory and invasive capacity in vitro and enhanced tumor cell colonization and metastatic growth in vivo. LY75 knockdown in SKOV3 cells also resulted in predominant upregulation of functional pathways implicated in cell proliferation and metabolism, while pathways associated with cell signaling and adhesion, complement activation and immune response were mostly suppressed. Moreover, LY75 suppression had an opposite effect on EOC cell lines with epithelial phenotype (A2780s and OV2008), by directing epithelial-to-mesenchymal transition (EMT) associated with reduced capacity for in vivo EOC cell colonization, as similar/identical signaling pathways were reversely regulated, when compared to mesenchymal LY75 knockdown EOC cells. To our knowledge, this is the first report of a gene displaying such pleiotropic effects in sustaining the cellular phenotype of EOC cells and points to novel functions of this receptor in modulating EOC dissemination. Our data also support previous findings regarding the superior capacity of epithelial cancer cells in metastatic colonization of distant sites, compared to cancer cells with mesenchymal-like morphology. Impact Journals LLC 2016-02-09 /pmc/articles/PMC4924702/ /pubmed/26871602 http://dx.doi.org/10.18632/oncotarget.7288 Text en Copyright: © 2016 Faddaoui 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
Faddaoui, Adnen
Bachvarova, Magdalena
Plante, Marie
Gregoire, Jean
Renaud, Marie-Claude
Sebastianelli, Alexandra
Gobeil, Stephane
Morin, Chantale
Macdonald, Elizabeth
Vanderhyden, Barbara
Bachvarov, Dimcho
The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
title The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
title_full The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
title_fullStr The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
title_full_unstemmed The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
title_short The mannose receptor LY75 (DEC205/CD205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
title_sort mannose receptor ly75 (dec205/cd205) modulates cellular phenotype and metastatic potential of ovarian cancer cells
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4924702/
https://www.ncbi.nlm.nih.gov/pubmed/26871602
http://dx.doi.org/10.18632/oncotarget.7288
work_keys_str_mv AT faddaouiadnen themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT bachvarovamagdalena themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT plantemarie themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT gregoirejean themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT renaudmarieclaude themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT sebastianellialexandra themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT gobeilstephane themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT morinchantale themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT macdonaldelizabeth themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT vanderhydenbarbara themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT bachvarovdimcho themannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT faddaouiadnen mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT bachvarovamagdalena mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT plantemarie mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT gregoirejean mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT renaudmarieclaude mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT sebastianellialexandra mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT gobeilstephane mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT morinchantale mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT macdonaldelizabeth mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT vanderhydenbarbara mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells
AT bachvarovdimcho mannosereceptorly75dec205cd205modulatescellularphenotypeandmetastaticpotentialofovariancancercells