Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.

Neutrophil gelatinase-associated lipocalin (NGAL) is emerging as a mediator of various biological and pathological states. However, the specific biological role of this molecule remains unclear, as it serves as a biomarker for many conditions. The high sensitivity of NGAL as a biomarker coupled with...

Full description

Bibliographic Details
Main Authors: Gaetano La Manna, Giulia Ghinatti, Pier Luigi Tazzari, Francesco Alviano, Francesca Ricci, Irene Capelli, Vania Cuna, Paola Todeschini, Eugenio Brunocilla, Pasqualepaolo Pagliaro, Laura Bonsi, Sergio Stefoni
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2014-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3937322?pdf=render
id doaj-ce91df7a66984a0abf13e54164ee9e21
record_format Article
spelling doaj-ce91df7a66984a0abf13e54164ee9e212020-11-24T21:51:02ZengPublic Library of Science (PLoS)PLoS ONE1932-62032014-01-0192e8949710.1371/journal.pone.0089497Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.Gaetano La MannaGiulia GhinattiPier Luigi TazzariFrancesco AlvianoFrancesca RicciIrene CapelliVania CunaPaola TodeschiniEugenio BrunocillaPasqualepaolo PagliaroLaura BonsiSergio StefoniNeutrophil gelatinase-associated lipocalin (NGAL) is emerging as a mediator of various biological and pathological states. However, the specific biological role of this molecule remains unclear, as it serves as a biomarker for many conditions. The high sensitivity of NGAL as a biomarker coupled with relatively low specificity may hide important biological roles. Data point toward an acute compensatory, protective role for NGAL in response to adverse cellular stresses, including inflammatory and oxidative stress. The aim of this study was to understand whether NGAL modulates the T-cell response through regulation of the human leukocyte antigen G (HLA-G) complex, which is a mediator of tolerance.Peripheral blood mononuclear cells (PBMCs) were obtained from eight healthy donors and isolated by centrifugation on a Ficoll gradient. All donors gave informed consent. PBMCs were treated with four different concentrations of NGAL (40-320 ng/ml) in an iron-loaded or iron-free form. Changes in cell phenotype were analyzed by flow cytometry. NGAL stimulated expression of HLA-G on CD4+ T cells in a dose- and iron-dependent manner. Iron deficiency prevented NGAL-mediated effects, such that HLA-G expression was unaltered. Furthermore, NGAL treatment affected stimulation of regulatory T cells and in vitro expansion of CD4(+) CD25(+) FoxP3(+) cells. An NGAL neutralizing antibody limited HLA-G expression and significantly decreased the percentage of CD4(+) CD25(+) FoxP3(+) cells.We provide in vitro evidence that NGAL is involved in cellular immunity. The potential role of NGAL as an immunomodulatory molecule is based on its ability to induce immune tolerance by upregulating HLA-G expression and expansion of T-regulatory cells in healthy donors. Future studies should further evaluate the role of NGAL in immunology and immunomodulation and its possible relationship to immunosuppressive therapy efficacy, tolerance induction in transplant patients, and other immunological disorders.http://europepmc.org/articles/PMC3937322?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Gaetano La Manna
Giulia Ghinatti
Pier Luigi Tazzari
Francesco Alviano
Francesca Ricci
Irene Capelli
Vania Cuna
Paola Todeschini
Eugenio Brunocilla
Pasqualepaolo Pagliaro
Laura Bonsi
Sergio Stefoni
spellingShingle Gaetano La Manna
Giulia Ghinatti
Pier Luigi Tazzari
Francesco Alviano
Francesca Ricci
Irene Capelli
Vania Cuna
Paola Todeschini
Eugenio Brunocilla
Pasqualepaolo Pagliaro
Laura Bonsi
Sergio Stefoni
Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.
PLoS ONE
author_facet Gaetano La Manna
Giulia Ghinatti
Pier Luigi Tazzari
Francesco Alviano
Francesca Ricci
Irene Capelli
Vania Cuna
Paola Todeschini
Eugenio Brunocilla
Pasqualepaolo Pagliaro
Laura Bonsi
Sergio Stefoni
author_sort Gaetano La Manna
title Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.
title_short Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.
title_full Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.
title_fullStr Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.
title_full_unstemmed Neutrophil gelatinase-associated lipocalin increases HLA-G(+)/FoxP3(+) T-regulatory cell population in an in vitro model of PBMC.
title_sort neutrophil gelatinase-associated lipocalin increases hla-g(+)/foxp3(+) t-regulatory cell population in an in vitro model of pbmc.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2014-01-01
description Neutrophil gelatinase-associated lipocalin (NGAL) is emerging as a mediator of various biological and pathological states. However, the specific biological role of this molecule remains unclear, as it serves as a biomarker for many conditions. The high sensitivity of NGAL as a biomarker coupled with relatively low specificity may hide important biological roles. Data point toward an acute compensatory, protective role for NGAL in response to adverse cellular stresses, including inflammatory and oxidative stress. The aim of this study was to understand whether NGAL modulates the T-cell response through regulation of the human leukocyte antigen G (HLA-G) complex, which is a mediator of tolerance.Peripheral blood mononuclear cells (PBMCs) were obtained from eight healthy donors and isolated by centrifugation on a Ficoll gradient. All donors gave informed consent. PBMCs were treated with four different concentrations of NGAL (40-320 ng/ml) in an iron-loaded or iron-free form. Changes in cell phenotype were analyzed by flow cytometry. NGAL stimulated expression of HLA-G on CD4+ T cells in a dose- and iron-dependent manner. Iron deficiency prevented NGAL-mediated effects, such that HLA-G expression was unaltered. Furthermore, NGAL treatment affected stimulation of regulatory T cells and in vitro expansion of CD4(+) CD25(+) FoxP3(+) cells. An NGAL neutralizing antibody limited HLA-G expression and significantly decreased the percentage of CD4(+) CD25(+) FoxP3(+) cells.We provide in vitro evidence that NGAL is involved in cellular immunity. The potential role of NGAL as an immunomodulatory molecule is based on its ability to induce immune tolerance by upregulating HLA-G expression and expansion of T-regulatory cells in healthy donors. Future studies should further evaluate the role of NGAL in immunology and immunomodulation and its possible relationship to immunosuppressive therapy efficacy, tolerance induction in transplant patients, and other immunological disorders.
url http://europepmc.org/articles/PMC3937322?pdf=render
work_keys_str_mv AT gaetanolamanna neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT giuliaghinatti neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT pierluigitazzari neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT francescoalviano neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT francescaricci neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT irenecapelli neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT vaniacuna neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT paolatodeschini neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT eugeniobrunocilla neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT pasqualepaolopagliaro neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT laurabonsi neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
AT sergiostefoni neutrophilgelatinaseassociatedlipocalinincreaseshlagfoxp3tregulatorycellpopulationinaninvitromodelofpbmc
_version_ 1725880960156696576