Cargando…

Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro

Although controlled local inflammation is essential for adequate bone regeneration, several studies have shown that hyper-inflammatory conditions after major trauma are associated with impaired fracture healing. These hyper-inflammatory conditions include the trauma-induced systemic inflammatory res...

Descripción completa

Detalles Bibliográficos
Autores principales: Bastian, Okan W., Croes, Michiel, Alblas, Jacqueline, Koenderman, Leo, Leenen, Luke P. H., Blokhuis, Taco J.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5938347/
https://www.ncbi.nlm.nih.gov/pubmed/29765377
http://dx.doi.org/10.3389/fimmu.2018.00945
_version_ 1783320762380189696
author Bastian, Okan W.
Croes, Michiel
Alblas, Jacqueline
Koenderman, Leo
Leenen, Luke P. H.
Blokhuis, Taco J.
author_facet Bastian, Okan W.
Croes, Michiel
Alblas, Jacqueline
Koenderman, Leo
Leenen, Luke P. H.
Blokhuis, Taco J.
author_sort Bastian, Okan W.
collection PubMed
description Although controlled local inflammation is essential for adequate bone regeneration, several studies have shown that hyper-inflammatory conditions after major trauma are associated with impaired fracture healing. These hyper-inflammatory conditions include the trauma-induced systemic inflammatory response to major injury, open fractures, and significant injury to the surrounding soft tissues. The current literature suggests that increased or prolonged influx of neutrophils into the fracture hematoma may mediate impairment of bone regeneration after hyper-inflammatory conditions. The underlying mechanism remains unclear. We hypothesize that high neutrophil numbers inhibit synthesis of mineralized extracellular matrix (ECM) by bone marrow stromal cells (BMSCs). We therefore studied the effect of increasing concentrations of neutrophils on ECM synthesis by human BMSCs in vitro. Moreover, we determined how high neutrophil concentrations affect BMSC cell counts, as well as BMSC osteogenic activity determined by alkaline phosphatase (ALP) expression and ALP activity. Co-culture of BMSCs with neutrophils induced a 52% decrease in BMSC cell count (p < 0.01), a 64% decrease in the percentage of ALP+ cells (p < 0.001), a 28% decrease in total ALP activity (p < 0.01), and a significant decrease in the amount of mineralized ECM [38% decrease after 4 weeks (p < 0.05)]. Co-cultures with peripheral blood mononuclear cells and neutrophils within transwells did not induce a significant decrease in ALP activity. In conclusion, our data shows that a decreased amount of mineralized ECM became synthesized by BMSCs, when they were co-cultured with high neutrophil concentrations. Moreover, high neutrophil concentrations induced a decrease in BMSC cell counts and decreased ALP activity. Clarifying the underlying mechanism may contribute to development of therapies that augment bone regeneration or prevent impaired fracture healing after hyper-inflammatory conditions.
format Online
Article
Text
id pubmed-5938347
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-59383472018-05-14 Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro Bastian, Okan W. Croes, Michiel Alblas, Jacqueline Koenderman, Leo Leenen, Luke P. H. Blokhuis, Taco J. Front Immunol Immunology Although controlled local inflammation is essential for adequate bone regeneration, several studies have shown that hyper-inflammatory conditions after major trauma are associated with impaired fracture healing. These hyper-inflammatory conditions include the trauma-induced systemic inflammatory response to major injury, open fractures, and significant injury to the surrounding soft tissues. The current literature suggests that increased or prolonged influx of neutrophils into the fracture hematoma may mediate impairment of bone regeneration after hyper-inflammatory conditions. The underlying mechanism remains unclear. We hypothesize that high neutrophil numbers inhibit synthesis of mineralized extracellular matrix (ECM) by bone marrow stromal cells (BMSCs). We therefore studied the effect of increasing concentrations of neutrophils on ECM synthesis by human BMSCs in vitro. Moreover, we determined how high neutrophil concentrations affect BMSC cell counts, as well as BMSC osteogenic activity determined by alkaline phosphatase (ALP) expression and ALP activity. Co-culture of BMSCs with neutrophils induced a 52% decrease in BMSC cell count (p < 0.01), a 64% decrease in the percentage of ALP+ cells (p < 0.001), a 28% decrease in total ALP activity (p < 0.01), and a significant decrease in the amount of mineralized ECM [38% decrease after 4 weeks (p < 0.05)]. Co-cultures with peripheral blood mononuclear cells and neutrophils within transwells did not induce a significant decrease in ALP activity. In conclusion, our data shows that a decreased amount of mineralized ECM became synthesized by BMSCs, when they were co-cultured with high neutrophil concentrations. Moreover, high neutrophil concentrations induced a decrease in BMSC cell counts and decreased ALP activity. Clarifying the underlying mechanism may contribute to development of therapies that augment bone regeneration or prevent impaired fracture healing after hyper-inflammatory conditions. Frontiers Media S.A. 2018-05-01 /pmc/articles/PMC5938347/ /pubmed/29765377 http://dx.doi.org/10.3389/fimmu.2018.00945 Text en Copyright © 2018 Bastian, Croes, Alblas, Koenderman, Leenen and Blokhuis. https://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Bastian, Okan W.
Croes, Michiel
Alblas, Jacqueline
Koenderman, Leo
Leenen, Luke P. H.
Blokhuis, Taco J.
Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro
title Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro
title_full Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro
title_fullStr Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro
title_full_unstemmed Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro
title_short Neutrophils Inhibit Synthesis of Mineralized Extracellular Matrix by Human Bone Marrow-Derived Stromal Cells In Vitro
title_sort neutrophils inhibit synthesis of mineralized extracellular matrix by human bone marrow-derived stromal cells in vitro
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5938347/
https://www.ncbi.nlm.nih.gov/pubmed/29765377
http://dx.doi.org/10.3389/fimmu.2018.00945
work_keys_str_mv AT bastianokanw neutrophilsinhibitsynthesisofmineralizedextracellularmatrixbyhumanbonemarrowderivedstromalcellsinvitro
AT croesmichiel neutrophilsinhibitsynthesisofmineralizedextracellularmatrixbyhumanbonemarrowderivedstromalcellsinvitro
AT alblasjacqueline neutrophilsinhibitsynthesisofmineralizedextracellularmatrixbyhumanbonemarrowderivedstromalcellsinvitro
AT koendermanleo neutrophilsinhibitsynthesisofmineralizedextracellularmatrixbyhumanbonemarrowderivedstromalcellsinvitro
AT leenenlukeph neutrophilsinhibitsynthesisofmineralizedextracellularmatrixbyhumanbonemarrowderivedstromalcellsinvitro
AT blokhuistacoj neutrophilsinhibitsynthesisofmineralizedextracellularmatrixbyhumanbonemarrowderivedstromalcellsinvitro