Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity

The role of prostaglandin (PG) F2α has been scarcely studied in cancer. We have identified a new function for PGF2α in ovarian cancer, stimulating the production of Prostate Transmembrane Protein, Androgen Induced 1 (PMEPA1). We show that this induction increases cell plasticity and proliferation, e...

Full description

Bibliographic Details
Main Authors: Alba Jiménez-Segovia, Alba Mota, Alejandro Rojo-Sebastián, Beatriz Barrocal, Angela Rynne-Vidal, María-Laura García-Bermejo, Raquel Gómez-Bris, Lukas J.A.C. Hawinkels, Pilar Sandoval, Ramon Garcia-Escudero, Manuel López-Cabrera, Gema Moreno-Bueno, Manuel Fresno, Konstantinos Stamatakis
Format: Article
Language:English
Published: Elsevier 2019-11-01
Series:Neoplasia: An International Journal for Oncology Research
Online Access:http://www.sciencedirect.com/science/article/pii/S1476558619302878
id doaj-ab84a5ae9764410dbac8933e3cf2d65a
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Alba Jiménez-Segovia
Alba Mota
Alejandro Rojo-Sebastián
Beatriz Barrocal
Angela Rynne-Vidal
María-Laura García-Bermejo
Raquel Gómez-Bris
Lukas J.A.C. Hawinkels
Pilar Sandoval
Ramon Garcia-Escudero
Manuel López-Cabrera
Gema Moreno-Bueno
Manuel Fresno
Konstantinos Stamatakis
spellingShingle Alba Jiménez-Segovia
Alba Mota
Alejandro Rojo-Sebastián
Beatriz Barrocal
Angela Rynne-Vidal
María-Laura García-Bermejo
Raquel Gómez-Bris
Lukas J.A.C. Hawinkels
Pilar Sandoval
Ramon Garcia-Escudero
Manuel López-Cabrera
Gema Moreno-Bueno
Manuel Fresno
Konstantinos Stamatakis
Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity
Neoplasia: An International Journal for Oncology Research
author_facet Alba Jiménez-Segovia
Alba Mota
Alejandro Rojo-Sebastián
Beatriz Barrocal
Angela Rynne-Vidal
María-Laura García-Bermejo
Raquel Gómez-Bris
Lukas J.A.C. Hawinkels
Pilar Sandoval
Ramon Garcia-Escudero
Manuel López-Cabrera
Gema Moreno-Bueno
Manuel Fresno
Konstantinos Stamatakis
author_sort Alba Jiménez-Segovia
title Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity
title_short Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity
title_full Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity
title_fullStr Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity
title_full_unstemmed Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticity
title_sort prostaglandin f2α-induced prostate transmembrane protein, androgen induced 1 mediates ovarian cancer progression increasing epithelial plasticity
publisher Elsevier
series Neoplasia: An International Journal for Oncology Research
issn 1476-5586
publishDate 2019-11-01
description The role of prostaglandin (PG) F2α has been scarcely studied in cancer. We have identified a new function for PGF2α in ovarian cancer, stimulating the production of Prostate Transmembrane Protein, Androgen Induced 1 (PMEPA1). We show that this induction increases cell plasticity and proliferation, enhancing tumor growth through PMEPA1. Thus, PMEPA1 overexpression in ovarian carcinoma cells, significantly increased cell proliferation rates, whereas PMEPA1 silencing decreased proliferation. In addition, PMEPA1 overexpression buffered TGFβ signaling, via reduction of SMAD-dependent signaling. PMEPA1 overexpressing cells acquired an epithelial morphology, associated with higher E-cadherin expression levels while β-catenin nuclear translocation was inhibited. Notwithstanding, high PMEPA1 levels also correlated with epithelial to mesenchymal transition markers, such as vimentin and ZEB1, allowing the cells to take advantage of both epithelial and mesenchymal characteristics, gaining in cell plasticity and adaptability. Interestingly, in mouse xenografts, PMEPA1 overexpressing ovarian cells had a clear survival and proliferative advantage, resulting in higher metastatic capacity, while PMEPA1 silencing had the opposite effect. Furthermore, high PMEPA1 expression in a cohort of advanced ovarian cancer patients was observed, correlating with E-cadherin expression. Most importantly, high PMEPA1 mRNA levels were associated with lower patient survival.
url http://www.sciencedirect.com/science/article/pii/S1476558619302878
work_keys_str_mv AT albajimenezsegovia prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT albamota prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT alejandrorojosebastian prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT beatrizbarrocal prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT angelarynnevidal prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT marialauragarciabermejo prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT raquelgomezbris prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT lukasjachawinkels prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT pilarsandoval prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT ramongarciaescudero prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT manuellopezcabrera prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT gemamorenobueno prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT manuelfresno prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
AT konstantinosstamatakis prostaglandinf2ainducedprostatetransmembraneproteinandrogeninduced1mediatesovariancancerprogressionincreasingepithelialplasticity
_version_ 1725068373792915456
spelling doaj-ab84a5ae9764410dbac8933e3cf2d65a2020-11-25T01:35:07ZengElsevierNeoplasia: An International Journal for Oncology Research1476-55862019-11-01211110731084Prostaglandin F2α-induced Prostate Transmembrane Protein, Androgen Induced 1 mediates ovarian cancer progression increasing epithelial plasticityAlba Jiménez-Segovia0Alba Mota1Alejandro Rojo-Sebastián2Beatriz Barrocal3Angela Rynne-Vidal4María-Laura García-Bermejo5Raquel Gómez-Bris6Lukas J.A.C. Hawinkels7Pilar Sandoval8Ramon Garcia-Escudero9Manuel López-Cabrera10Gema Moreno-Bueno11Manuel Fresno12Konstantinos Stamatakis13Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, SpainDepartamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, Madrid, Spain; MD Anderson Cancer Center Madrid & Fundación MD Anderson Internacional, Madrid, SpainMD Anderson Cancer Center Madrid & Fundación MD Anderson Internacional, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), SpainCentro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, SpainCentro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, SpainBiomarkers and Therapeutic Targets Lab, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, SpainCentro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, SpainDepartment of Gastroenterology and Hepatology, Leiden University Medical Centre, Leiden, the NetherlandsCentro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, SpainCentro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain; Molecular Oncology Unit, CIEMAT, Madrid, Spain; Biomedical Research Institute I+12, University Hospital 12 de Octubre, Madrid 28041, SpainCentro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, SpainDepartamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas ‘Alberto Sols’ (CSIC-UAM), IdiPaz, Madrid, Spain; MD Anderson Cancer Center Madrid & Fundación MD Anderson Internacional, Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), SpainCentro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, Spain; Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IIS-P), Madrid, Spain; Corresponding authors at: Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, Spain.Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, Spain; Instituto de Investigación Sanitaria Hospital Universitario de la Princesa (IIS-P), Madrid, Spain; Corresponding authors at: Centro de Biología Molecular “Severo Ochoa” (Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid), c/ Nicolás Cabrera, 1, Campus Cantoblanco, Universidad Autónoma de Madrid, Madrid 28049, Spain.The role of prostaglandin (PG) F2α has been scarcely studied in cancer. We have identified a new function for PGF2α in ovarian cancer, stimulating the production of Prostate Transmembrane Protein, Androgen Induced 1 (PMEPA1). We show that this induction increases cell plasticity and proliferation, enhancing tumor growth through PMEPA1. Thus, PMEPA1 overexpression in ovarian carcinoma cells, significantly increased cell proliferation rates, whereas PMEPA1 silencing decreased proliferation. In addition, PMEPA1 overexpression buffered TGFβ signaling, via reduction of SMAD-dependent signaling. PMEPA1 overexpressing cells acquired an epithelial morphology, associated with higher E-cadherin expression levels while β-catenin nuclear translocation was inhibited. Notwithstanding, high PMEPA1 levels also correlated with epithelial to mesenchymal transition markers, such as vimentin and ZEB1, allowing the cells to take advantage of both epithelial and mesenchymal characteristics, gaining in cell plasticity and adaptability. Interestingly, in mouse xenografts, PMEPA1 overexpressing ovarian cells had a clear survival and proliferative advantage, resulting in higher metastatic capacity, while PMEPA1 silencing had the opposite effect. Furthermore, high PMEPA1 expression in a cohort of advanced ovarian cancer patients was observed, correlating with E-cadherin expression. Most importantly, high PMEPA1 mRNA levels were associated with lower patient survival.http://www.sciencedirect.com/science/article/pii/S1476558619302878