Cargando…

Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study

Bone is an organ with high natural regenerative capacity and most fractures heal spontaneously when appropriate fracture fixation is provided. However, additional treatment is required for patients with large segmental defects exceeding the endogenous healing potential and for patients suffering fro...

Descripción completa

Detalles Bibliográficos
Autores principales: Herrmann, Marietta, Zeiter, Stephan, Eberli, Ursula, Hildebrand, Maria, Camenisch, Karin, Menzel, Ursula, Alini, Mauro, Verrier, Sophie, Stadelmann, Vincent A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5816045/
https://www.ncbi.nlm.nih.gov/pubmed/29484293
http://dx.doi.org/10.3389/fbioe.2018.00005
_version_ 1783300604596060160
author Herrmann, Marietta
Zeiter, Stephan
Eberli, Ursula
Hildebrand, Maria
Camenisch, Karin
Menzel, Ursula
Alini, Mauro
Verrier, Sophie
Stadelmann, Vincent A.
author_facet Herrmann, Marietta
Zeiter, Stephan
Eberli, Ursula
Hildebrand, Maria
Camenisch, Karin
Menzel, Ursula
Alini, Mauro
Verrier, Sophie
Stadelmann, Vincent A.
author_sort Herrmann, Marietta
collection PubMed
description Bone is an organ with high natural regenerative capacity and most fractures heal spontaneously when appropriate fracture fixation is provided. However, additional treatment is required for patients with large segmental defects exceeding the endogenous healing potential and for patients suffering from fracture non-unions. These cases are often associated with insufficient vascularization. Transplantation of CD34+ endothelial progenitor cells (EPCs) has been successfully applied to promote neovascularization of bone defects, however including extensive ex vivo manipulation of cells. Here, we hypothesized, that treatment with granulocyte colony-stimulating factor (G-CSF) may improve bone healing by mobilization of CD34+ progenitor cells into the circulation, which in turn may facilitate vascularization at the defect site. In this pilot study, we aimed to characterize the different cell populations mobilized by G-CSF and investigate the influence of cell mobilization on the healing of a critical size femoral defect in rats. Cell mobilization was investigated by flow cytometry at different time points after five consecutive daily G-CSF injections. In a pilot study, bone healing of a 4.5-mm critical femoral defect in F344 rats was compared between a saline-treated control group and a G-CSF treatment group. In vivo microcomputed tomography and histology were applied to compare bone formation in both treatment groups. Our data revealed that leukocyte counts show a peak increase at the first day after the last G-CSF injection. In addition, we found that CD34+ progenitor cells, including EPCs, were significantly enriched at day 1, and further increased at day 5 and day 11. Upregulation of monocytes, granulocytes and macrophages peaked at day 1. G-CSF treatment significantly increased bone volume and bone density in the defect, which was confirmed by histology. Our data show that different cell populations are mobilized by G-CSF treatment in cell specific patterns. Although in this pilot study no bridging of the critical defect was observed, significantly improved bone formation by G-CSF treatment was clearly shown.
format Online
Article
Text
id pubmed-5816045
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-58160452018-02-26 Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study Herrmann, Marietta Zeiter, Stephan Eberli, Ursula Hildebrand, Maria Camenisch, Karin Menzel, Ursula Alini, Mauro Verrier, Sophie Stadelmann, Vincent A. Front Bioeng Biotechnol Bioengineering and Biotechnology Bone is an organ with high natural regenerative capacity and most fractures heal spontaneously when appropriate fracture fixation is provided. However, additional treatment is required for patients with large segmental defects exceeding the endogenous healing potential and for patients suffering from fracture non-unions. These cases are often associated with insufficient vascularization. Transplantation of CD34+ endothelial progenitor cells (EPCs) has been successfully applied to promote neovascularization of bone defects, however including extensive ex vivo manipulation of cells. Here, we hypothesized, that treatment with granulocyte colony-stimulating factor (G-CSF) may improve bone healing by mobilization of CD34+ progenitor cells into the circulation, which in turn may facilitate vascularization at the defect site. In this pilot study, we aimed to characterize the different cell populations mobilized by G-CSF and investigate the influence of cell mobilization on the healing of a critical size femoral defect in rats. Cell mobilization was investigated by flow cytometry at different time points after five consecutive daily G-CSF injections. In a pilot study, bone healing of a 4.5-mm critical femoral defect in F344 rats was compared between a saline-treated control group and a G-CSF treatment group. In vivo microcomputed tomography and histology were applied to compare bone formation in both treatment groups. Our data revealed that leukocyte counts show a peak increase at the first day after the last G-CSF injection. In addition, we found that CD34+ progenitor cells, including EPCs, were significantly enriched at day 1, and further increased at day 5 and day 11. Upregulation of monocytes, granulocytes and macrophages peaked at day 1. G-CSF treatment significantly increased bone volume and bone density in the defect, which was confirmed by histology. Our data show that different cell populations are mobilized by G-CSF treatment in cell specific patterns. Although in this pilot study no bridging of the critical defect was observed, significantly improved bone formation by G-CSF treatment was clearly shown. Frontiers Media S.A. 2018-02-12 /pmc/articles/PMC5816045/ /pubmed/29484293 http://dx.doi.org/10.3389/fbioe.2018.00005 Text en Copyright © 2018 Herrmann, Zeiter, Eberli, Hildebrand, Camenisch, Menzel, Alini, Verrier and Stadelmann. http://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 Bioengineering and Biotechnology
Herrmann, Marietta
Zeiter, Stephan
Eberli, Ursula
Hildebrand, Maria
Camenisch, Karin
Menzel, Ursula
Alini, Mauro
Verrier, Sophie
Stadelmann, Vincent A.
Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study
title Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study
title_full Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study
title_fullStr Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study
title_full_unstemmed Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study
title_short Five Days Granulocyte Colony-Stimulating Factor Treatment Increases Bone Formation and Reduces Gap Size of a Rat Segmental Bone Defect: A Pilot Study
title_sort five days granulocyte colony-stimulating factor treatment increases bone formation and reduces gap size of a rat segmental bone defect: a pilot study
topic Bioengineering and Biotechnology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5816045/
https://www.ncbi.nlm.nih.gov/pubmed/29484293
http://dx.doi.org/10.3389/fbioe.2018.00005
work_keys_str_mv AT herrmannmarietta fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT zeiterstephan fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT eberliursula fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT hildebrandmaria fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT camenischkarin fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT menzelursula fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT alinimauro fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT verriersophie fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy
AT stadelmannvincenta fivedaysgranulocytecolonystimulatingfactortreatmentincreasesboneformationandreducesgapsizeofaratsegmentalbonedefectapilotstudy