Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation

Detalhes bibliográficos
Autor(a) principal: Carrascal, M
Data de Publicação: 2018
Outros Autores: Silva, M, Ramalho, J, Pen, C, Martins, M, Pascoal, C, Amaral, C, Serrano, I, Oliveira, MJ, Sackstein, R, Videira, P
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.17/3949
Resumo: Breast cancer tissue overexpresses fucosylated glycans, such as sialyl-Lewis X/A (sLeX/A ), and α-1,3/4-fucosyltransferases (FUTs) in relation to increased disease progression and metastasis. These glycans in tumor circulating cells mediate binding to vascular E-selectin, initiating tumor extravasation. However, their role in breast carcinogenesis is still unknown. Here, we aimed to define the contribution of the fucosylated structures, including sLeX/A , to cell adhesion, cell signaling, and cell proliferation in invasive ductal carcinomas (IDC), the most frequent type of breast cancer. We first analyzed expression of E-selectin ligands in IDC tissue and established primary cell cultures from the tissue. We observed strong reactivity with E-selectin and anti-sLeX/A antibodies in both IDC tissue and cell lines, and expression of α-1,3/4 FUTs FUT4, FUT5, FUT6, FUT10, and FUT11. To further assess the role of fucosylation in IDC biology, we immortalized a primary IDC cell line with human telomerase reverse transcriptase to create the 'CF1_T cell line'. Treatment with 2-fluorofucose (2-FF), a fucosylation inhibitor, completely abrogated its sLeX/A expression and dramatically reduced adherence of CF1_T cells to E-selectin under hemodynamic flow conditions. In addition, 2-FF-treated CF1_T cells showed a reduced migratory ability, as well as decreased cell proliferation rate. Notably, 2-FF treatment lowered the growth factor expression of CF1_T cells, prominently for FGF2, vascular endothelial growth factor, and transforming growth factor beta, and negatively affected activation of signal-regulating protein kinases 1 and 2 and p38 mitogen-activated protein kinase signaling pathways. These data indicate that fucosylation licenses several malignant features of IDC, such as cell adhesion, migration, proliferation, and growth factor expression, contributing to tumor progression.
id RCAP_740ead46929b3b4b1ca7c8a5e46b5256
oai_identifier_str oai:repositorio.chlc.min-saude.pt:10400.17/3949
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str
spelling Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK ActivationHSJ ANPATAdultAgedFemaleHumansBreast Neoplasms / enzymology*Middle AgedBreast Neoplasms / pathologyCarcinoma, Ductal, Breast / enzymology*Carcinoma, Ductal, Breast / pathologyCell Adhesion / drug effectsCell Line, TumorCell Proliferation / drug effectsE-Selectin / geneticsE-Selectin / metabolism*Fucose / analogs & derivativesFucose / pharmacologyFucosyltransferases / antagonists & inhibitors*LigandsMAP Kinase Signaling System / drug effectsNeoplasm InvasivenessOligosaccharides / metabolism*Primary Cell CultureSialyl Lewis X Antigenp38 Mitogen-Activated Protein Kinases / geneticsp38 Mitogen-Activated Protein Kinases / metabolism*Breast cancer tissue overexpresses fucosylated glycans, such as sialyl-Lewis X/A (sLeX/A ), and α-1,3/4-fucosyltransferases (FUTs) in relation to increased disease progression and metastasis. These glycans in tumor circulating cells mediate binding to vascular E-selectin, initiating tumor extravasation. However, their role in breast carcinogenesis is still unknown. Here, we aimed to define the contribution of the fucosylated structures, including sLeX/A , to cell adhesion, cell signaling, and cell proliferation in invasive ductal carcinomas (IDC), the most frequent type of breast cancer. We first analyzed expression of E-selectin ligands in IDC tissue and established primary cell cultures from the tissue. We observed strong reactivity with E-selectin and anti-sLeX/A antibodies in both IDC tissue and cell lines, and expression of α-1,3/4 FUTs FUT4, FUT5, FUT6, FUT10, and FUT11. To further assess the role of fucosylation in IDC biology, we immortalized a primary IDC cell line with human telomerase reverse transcriptase to create the 'CF1_T cell line'. Treatment with 2-fluorofucose (2-FF), a fucosylation inhibitor, completely abrogated its sLeX/A expression and dramatically reduced adherence of CF1_T cells to E-selectin under hemodynamic flow conditions. In addition, 2-FF-treated CF1_T cells showed a reduced migratory ability, as well as decreased cell proliferation rate. Notably, 2-FF treatment lowered the growth factor expression of CF1_T cells, prominently for FGF2, vascular endothelial growth factor, and transforming growth factor beta, and negatively affected activation of signal-regulating protein kinases 1 and 2 and p38 mitogen-activated protein kinase signaling pathways. These data indicate that fucosylation licenses several malignant features of IDC, such as cell adhesion, migration, proliferation, and growth factor expression, contributing to tumor progression.WileyRepositório do Centro Hospitalar Universitário de Lisboa Central, EPECarrascal, MSilva, MRamalho, JPen, CMartins, MPascoal, CAmaral, CSerrano, IOliveira, MJSackstein, RVideira, P2022-01-04T11:28:45Z20182018-01-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.17/3949engMol Oncol. 2018 May;12(5):579-593.10.1002/1878-0261.12163.info:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-03-10T09:44:42ZPortal AgregadorONG
dc.title.none.fl_str_mv Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
title Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
spellingShingle Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
Carrascal, M
HSJ ANPAT
Adult
Aged
Female
Humans
Breast Neoplasms / enzymology*
Middle Aged
Breast Neoplasms / pathology
Carcinoma, Ductal, Breast / enzymology*
Carcinoma, Ductal, Breast / pathology
Cell Adhesion / drug effects
Cell Line, Tumor
Cell Proliferation / drug effects
E-Selectin / genetics
E-Selectin / metabolism*
Fucose / analogs & derivatives
Fucose / pharmacology
Fucosyltransferases / antagonists & inhibitors*
Ligands
MAP Kinase Signaling System / drug effects
Neoplasm Invasiveness
Oligosaccharides / metabolism*
Primary Cell Culture
Sialyl Lewis X Antigen
p38 Mitogen-Activated Protein Kinases / genetics
p38 Mitogen-Activated Protein Kinases / metabolism*
title_short Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
title_full Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
title_fullStr Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
title_full_unstemmed Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
title_sort Inhibition of Fucosylation in Human Invasive Ductal Carcinoma Reduces E-Selectin Ligand Expression, Cell Proliferation, and ERK1/2 and p38 MAPK Activation
author Carrascal, M
author_facet Carrascal, M
Silva, M
Ramalho, J
Pen, C
Martins, M
Pascoal, C
Amaral, C
Serrano, I
Oliveira, MJ
Sackstein, R
Videira, P
author_role author
author2 Silva, M
Ramalho, J
Pen, C
Martins, M
Pascoal, C
Amaral, C
Serrano, I
Oliveira, MJ
Sackstein, R
Videira, P
author2_role author
author
author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv Repositório do Centro Hospitalar Universitário de Lisboa Central, EPE
dc.contributor.author.fl_str_mv Carrascal, M
Silva, M
Ramalho, J
Pen, C
Martins, M
Pascoal, C
Amaral, C
Serrano, I
Oliveira, MJ
Sackstein, R
Videira, P
dc.subject.por.fl_str_mv HSJ ANPAT
Adult
Aged
Female
Humans
Breast Neoplasms / enzymology*
Middle Aged
Breast Neoplasms / pathology
Carcinoma, Ductal, Breast / enzymology*
Carcinoma, Ductal, Breast / pathology
Cell Adhesion / drug effects
Cell Line, Tumor
Cell Proliferation / drug effects
E-Selectin / genetics
E-Selectin / metabolism*
Fucose / analogs & derivatives
Fucose / pharmacology
Fucosyltransferases / antagonists & inhibitors*
Ligands
MAP Kinase Signaling System / drug effects
Neoplasm Invasiveness
Oligosaccharides / metabolism*
Primary Cell Culture
Sialyl Lewis X Antigen
p38 Mitogen-Activated Protein Kinases / genetics
p38 Mitogen-Activated Protein Kinases / metabolism*
topic HSJ ANPAT
Adult
Aged
Female
Humans
Breast Neoplasms / enzymology*
Middle Aged
Breast Neoplasms / pathology
Carcinoma, Ductal, Breast / enzymology*
Carcinoma, Ductal, Breast / pathology
Cell Adhesion / drug effects
Cell Line, Tumor
Cell Proliferation / drug effects
E-Selectin / genetics
E-Selectin / metabolism*
Fucose / analogs & derivatives
Fucose / pharmacology
Fucosyltransferases / antagonists & inhibitors*
Ligands
MAP Kinase Signaling System / drug effects
Neoplasm Invasiveness
Oligosaccharides / metabolism*
Primary Cell Culture
Sialyl Lewis X Antigen
p38 Mitogen-Activated Protein Kinases / genetics
p38 Mitogen-Activated Protein Kinases / metabolism*
description Breast cancer tissue overexpresses fucosylated glycans, such as sialyl-Lewis X/A (sLeX/A ), and α-1,3/4-fucosyltransferases (FUTs) in relation to increased disease progression and metastasis. These glycans in tumor circulating cells mediate binding to vascular E-selectin, initiating tumor extravasation. However, their role in breast carcinogenesis is still unknown. Here, we aimed to define the contribution of the fucosylated structures, including sLeX/A , to cell adhesion, cell signaling, and cell proliferation in invasive ductal carcinomas (IDC), the most frequent type of breast cancer. We first analyzed expression of E-selectin ligands in IDC tissue and established primary cell cultures from the tissue. We observed strong reactivity with E-selectin and anti-sLeX/A antibodies in both IDC tissue and cell lines, and expression of α-1,3/4 FUTs FUT4, FUT5, FUT6, FUT10, and FUT11. To further assess the role of fucosylation in IDC biology, we immortalized a primary IDC cell line with human telomerase reverse transcriptase to create the 'CF1_T cell line'. Treatment with 2-fluorofucose (2-FF), a fucosylation inhibitor, completely abrogated its sLeX/A expression and dramatically reduced adherence of CF1_T cells to E-selectin under hemodynamic flow conditions. In addition, 2-FF-treated CF1_T cells showed a reduced migratory ability, as well as decreased cell proliferation rate. Notably, 2-FF treatment lowered the growth factor expression of CF1_T cells, prominently for FGF2, vascular endothelial growth factor, and transforming growth factor beta, and negatively affected activation of signal-regulating protein kinases 1 and 2 and p38 mitogen-activated protein kinase signaling pathways. These data indicate that fucosylation licenses several malignant features of IDC, such as cell adhesion, migration, proliferation, and growth factor expression, contributing to tumor progression.
publishDate 2018
dc.date.none.fl_str_mv 2018
2018-01-01T00:00:00Z
2022-01-04T11:28:45Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10400.17/3949
url http://hdl.handle.net/10400.17/3949
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv Mol Oncol. 2018 May;12(5):579-593.
10.1002/1878-0261.12163.
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Wiley
publisher.none.fl_str_mv Wiley
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv
repository.mail.fl_str_mv
_version_ 1777302277906759680