CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo

Standard

CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo. / Wegwitz, Florian; Lenfert, Eva; Gerstel, Daniela; von Ehrenstein, Lena; Einhoff, Julia; Schmidt, Geske; Logsdon, Matthew; Brandner, Johanna; Tiegs, Gisa; Beauchemin, Nicole; Wagener, Christoph; Deppert, Wolfgang; Horst, Andrea Kristina.

in: ONCOTARGET, Jahrgang 7, Nr. 39, 27.09.2016, S. 63730-63746.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Wegwitz, F, Lenfert, E, Gerstel, D, von Ehrenstein, L, Einhoff, J, Schmidt, G, Logsdon, M, Brandner, J, Tiegs, G, Beauchemin, N, Wagener, C, Deppert, W & Horst, AK 2016, 'CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo', ONCOTARGET, Jg. 7, Nr. 39, S. 63730-63746. https://doi.org/10.18632/oncotarget.11650

APA

Wegwitz, F., Lenfert, E., Gerstel, D., von Ehrenstein, L., Einhoff, J., Schmidt, G., Logsdon, M., Brandner, J., Tiegs, G., Beauchemin, N., Wagener, C., Deppert, W., & Horst, A. K. (2016). CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo. ONCOTARGET, 7(39), 63730-63746. https://doi.org/10.18632/oncotarget.11650

Vancouver

Wegwitz F, Lenfert E, Gerstel D, von Ehrenstein L, Einhoff J, Schmidt G et al. CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo. ONCOTARGET. 2016 Sep 27;7(39):63730-63746. https://doi.org/10.18632/oncotarget.11650

Bibtex

@article{24b91d71e098480dab404d31e2bd91ad,
title = "CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo",
abstract = "We analyzed the molecular basis for carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-controlled inhibition of epithelial-mesenchymal transition (EMT) in a mouse model for mammary adenocarcinoma (WAP-T mice). We demonstrate that silencing of CEACAM1 in WAP-T tumor-derived G-2 cells induces epithelial-mesenchymal plasticity (EMP), as evidenced by typical changes of gene expression, morphology and increased invasion. In contrast, reintroduction of CEACAM1 into G-2 cells reversed up-regulation of genes imposing mesenchymal transition, as well as cellular invasion. We identified the Wnt-pathway as target for CEACAM1-mediated repression of EMT. Importantly, β-catenin phosphorylation status and transcriptional activity strongly depend on CEACAM1 expression: CEACAM1high G-2 cells displayed enhanced phosphorylation of β-catenin at S33/S37/T41 and decreased phosphorylation at Y86, thereby inhibiting canonical Wnt/β-catenin signaling. We identified Src-homology 2 domain-containing phosphatase 2 (SHP-2) as a critical binding partner of CEACAM1 that could modulate β-catenin Y86 phosphorylation. Hence, CEACAM1 serves as a scaffold that controls membrane proximal β-catenin signaling. In vivo, mammary tumors of WAP-T/CEACAM1null mice displayed increased nuclear translocation of β-catenin and a dramatically enhanced metastasis rate compared to WAP-T mice. Hence, CEACAM1 controls EMT in vitro and in vivo by site-specific regulation of β-catenin phosphorylation. Survival analyses of human mammary carcinoma patients corroborated these data, indicating that CEACAM1 is a prognostic marker for breast cancer survival.",
author = "Florian Wegwitz and Eva Lenfert and Daniela Gerstel and {von Ehrenstein}, Lena and Julia Einhoff and Geske Schmidt and Matthew Logsdon and Johanna Brandner and Gisa Tiegs and Nicole Beauchemin and Christoph Wagener and Wolfgang Deppert and Horst, {Andrea Kristina}",
year = "2016",
month = sep,
day = "27",
doi = "10.18632/oncotarget.11650",
language = "English",
volume = "7",
pages = "63730--63746",
journal = "ONCOTARGET",
issn = "1949-2553",
publisher = "IMPACT JOURNALS LLC",
number = "39",

}

RIS

TY - JOUR

T1 - CEACAM1 controls the EMT switch in murine mammary carcinoma in vitro and in vivo

AU - Wegwitz, Florian

AU - Lenfert, Eva

AU - Gerstel, Daniela

AU - von Ehrenstein, Lena

AU - Einhoff, Julia

AU - Schmidt, Geske

AU - Logsdon, Matthew

AU - Brandner, Johanna

AU - Tiegs, Gisa

AU - Beauchemin, Nicole

AU - Wagener, Christoph

AU - Deppert, Wolfgang

AU - Horst, Andrea Kristina

PY - 2016/9/27

Y1 - 2016/9/27

N2 - We analyzed the molecular basis for carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-controlled inhibition of epithelial-mesenchymal transition (EMT) in a mouse model for mammary adenocarcinoma (WAP-T mice). We demonstrate that silencing of CEACAM1 in WAP-T tumor-derived G-2 cells induces epithelial-mesenchymal plasticity (EMP), as evidenced by typical changes of gene expression, morphology and increased invasion. In contrast, reintroduction of CEACAM1 into G-2 cells reversed up-regulation of genes imposing mesenchymal transition, as well as cellular invasion. We identified the Wnt-pathway as target for CEACAM1-mediated repression of EMT. Importantly, β-catenin phosphorylation status and transcriptional activity strongly depend on CEACAM1 expression: CEACAM1high G-2 cells displayed enhanced phosphorylation of β-catenin at S33/S37/T41 and decreased phosphorylation at Y86, thereby inhibiting canonical Wnt/β-catenin signaling. We identified Src-homology 2 domain-containing phosphatase 2 (SHP-2) as a critical binding partner of CEACAM1 that could modulate β-catenin Y86 phosphorylation. Hence, CEACAM1 serves as a scaffold that controls membrane proximal β-catenin signaling. In vivo, mammary tumors of WAP-T/CEACAM1null mice displayed increased nuclear translocation of β-catenin and a dramatically enhanced metastasis rate compared to WAP-T mice. Hence, CEACAM1 controls EMT in vitro and in vivo by site-specific regulation of β-catenin phosphorylation. Survival analyses of human mammary carcinoma patients corroborated these data, indicating that CEACAM1 is a prognostic marker for breast cancer survival.

AB - We analyzed the molecular basis for carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-controlled inhibition of epithelial-mesenchymal transition (EMT) in a mouse model for mammary adenocarcinoma (WAP-T mice). We demonstrate that silencing of CEACAM1 in WAP-T tumor-derived G-2 cells induces epithelial-mesenchymal plasticity (EMP), as evidenced by typical changes of gene expression, morphology and increased invasion. In contrast, reintroduction of CEACAM1 into G-2 cells reversed up-regulation of genes imposing mesenchymal transition, as well as cellular invasion. We identified the Wnt-pathway as target for CEACAM1-mediated repression of EMT. Importantly, β-catenin phosphorylation status and transcriptional activity strongly depend on CEACAM1 expression: CEACAM1high G-2 cells displayed enhanced phosphorylation of β-catenin at S33/S37/T41 and decreased phosphorylation at Y86, thereby inhibiting canonical Wnt/β-catenin signaling. We identified Src-homology 2 domain-containing phosphatase 2 (SHP-2) as a critical binding partner of CEACAM1 that could modulate β-catenin Y86 phosphorylation. Hence, CEACAM1 serves as a scaffold that controls membrane proximal β-catenin signaling. In vivo, mammary tumors of WAP-T/CEACAM1null mice displayed increased nuclear translocation of β-catenin and a dramatically enhanced metastasis rate compared to WAP-T mice. Hence, CEACAM1 controls EMT in vitro and in vivo by site-specific regulation of β-catenin phosphorylation. Survival analyses of human mammary carcinoma patients corroborated these data, indicating that CEACAM1 is a prognostic marker for breast cancer survival.

U2 - 10.18632/oncotarget.11650

DO - 10.18632/oncotarget.11650

M3 - SCORING: Journal article

C2 - 27572314

VL - 7

SP - 63730

EP - 63746

JO - ONCOTARGET

JF - ONCOTARGET

SN - 1949-2553

IS - 39

ER -