Cargando…

NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells

Expression of neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin2, a recently recognized iron regulatory protein that bonds to matrix metalloproteinase 9 (MMP9), is increased in a spectrum of cancers including those of the colorectum. Using colon carcinoma cell lines stably transfected with...

Descripción completa

Detalles Bibliográficos
Autores principales: Hu, Limei, Hittelman, Walter, Lu, Tao, Ji, Ping, Arlinghaus, Ralph, Shmulevich, Ilya, Hamilton, Stanley R., Zhang, Wei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: 2009
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7770608/
https://www.ncbi.nlm.nih.gov/pubmed/19308044
http://dx.doi.org/10.1038/labinvest.2009.17
_version_ 1783629544603779072
author Hu, Limei
Hittelman, Walter
Lu, Tao
Ji, Ping
Arlinghaus, Ralph
Shmulevich, Ilya
Hamilton, Stanley R.
Zhang, Wei
author_facet Hu, Limei
Hittelman, Walter
Lu, Tao
Ji, Ping
Arlinghaus, Ralph
Shmulevich, Ilya
Hamilton, Stanley R.
Zhang, Wei
author_sort Hu, Limei
collection PubMed
description Expression of neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin2, a recently recognized iron regulatory protein that bonds to matrix metalloproteinase 9 (MMP9), is increased in a spectrum of cancers including those of the colorectum. Using colon carcinoma cell lines stably transfected with NGAL or antisense NGAL, we demonstrated that NGAL overexpression altered subcellular localization of E-cadherin and catenins, decreased E-cadherin-mediated cell-cell adhesion, enhanced cell-matrix attachment, and increased cell motility and in vitro invasion. Conversely, decrease in NGAL enhanced more aggregated growth pattern and decreased in vitro invasion. We further demonstrated that NGAL exerted these effects through alteration of the subcellular localization of Rac1 in an extracellular matrix-dependent but MMP9-independent manner. Furthermore, we observed that the NGAL overexpressing cells tolerated increased iron level in the culture environment while the NGAL underexpressing cells showed significant cell death after prolonged incubation in high iron condition. Thus, overexpressing NGAL in colon carcinomas is an important regulatory molecule that integrates extracellular environment cues, iron metabolism, and intracellular small GTPase signaling in cancer migration and invasion. NGAL may therefore be a new target for therapeutic intervention in colorectal carcinoma.
format Online
Article
Text
id pubmed-7770608
institution National Center for Biotechnology Information
language English
publishDate 2009
record_format MEDLINE/PubMed
spelling pubmed-77706082020-12-29 NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells Hu, Limei Hittelman, Walter Lu, Tao Ji, Ping Arlinghaus, Ralph Shmulevich, Ilya Hamilton, Stanley R. Zhang, Wei Lab Invest Article Expression of neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin2, a recently recognized iron regulatory protein that bonds to matrix metalloproteinase 9 (MMP9), is increased in a spectrum of cancers including those of the colorectum. Using colon carcinoma cell lines stably transfected with NGAL or antisense NGAL, we demonstrated that NGAL overexpression altered subcellular localization of E-cadherin and catenins, decreased E-cadherin-mediated cell-cell adhesion, enhanced cell-matrix attachment, and increased cell motility and in vitro invasion. Conversely, decrease in NGAL enhanced more aggregated growth pattern and decreased in vitro invasion. We further demonstrated that NGAL exerted these effects through alteration of the subcellular localization of Rac1 in an extracellular matrix-dependent but MMP9-independent manner. Furthermore, we observed that the NGAL overexpressing cells tolerated increased iron level in the culture environment while the NGAL underexpressing cells showed significant cell death after prolonged incubation in high iron condition. Thus, overexpressing NGAL in colon carcinomas is an important regulatory molecule that integrates extracellular environment cues, iron metabolism, and intracellular small GTPase signaling in cancer migration and invasion. NGAL may therefore be a new target for therapeutic intervention in colorectal carcinoma. 2009-03-23 2009-05 /pmc/articles/PMC7770608/ /pubmed/19308044 http://dx.doi.org/10.1038/labinvest.2009.17 Text en http://www.nature.com/authors/editorial_policies/license.html#terms Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms
spellingShingle Article
Hu, Limei
Hittelman, Walter
Lu, Tao
Ji, Ping
Arlinghaus, Ralph
Shmulevich, Ilya
Hamilton, Stanley R.
Zhang, Wei
NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells
title NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells
title_full NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells
title_fullStr NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells
title_full_unstemmed NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells
title_short NGAL decreases E-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through Rac1 in colon carcinoma cells
title_sort ngal decreases e-cadherin-mediated cell-cell adhesion and increases cell motility and invasion through rac1 in colon carcinoma cells
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7770608/
https://www.ncbi.nlm.nih.gov/pubmed/19308044
http://dx.doi.org/10.1038/labinvest.2009.17
work_keys_str_mv AT hulimei ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT hittelmanwalter ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT lutao ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT jiping ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT arlinghausralph ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT shmulevichilya ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT hamiltonstanleyr ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells
AT zhangwei ngaldecreasesecadherinmediatedcellcelladhesionandincreasescellmotilityandinvasionthroughrac1incoloncarcinomacells