Cargando…

Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model

BACKGROUND: Bone fracture healing is dependent upon the rapid migration and engraftment of bone marrow (BM) progenitor and stem cells to the site of injury. Stromal cell-derived factor-1 plays a crucial role in recruiting BM cells expressing its receptor CXCR4. Recently, a CXCR4 antagonist, plerixaf...

Descripción completa

Detalles Bibliográficos
Autores principales: Asiedu, Kingsley O., Ferdousi, Munira, Ton, Phuongnga T., Adler, Stephen S., Choyke, Peter L., Sato, Noriko
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Berlin Heidelberg 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6292830/
https://www.ncbi.nlm.nih.gov/pubmed/30547233
http://dx.doi.org/10.1186/s13550-018-0463-8
_version_ 1783380441040945152
author Asiedu, Kingsley O.
Ferdousi, Munira
Ton, Phuongnga T.
Adler, Stephen S.
Choyke, Peter L.
Sato, Noriko
author_facet Asiedu, Kingsley O.
Ferdousi, Munira
Ton, Phuongnga T.
Adler, Stephen S.
Choyke, Peter L.
Sato, Noriko
author_sort Asiedu, Kingsley O.
collection PubMed
description BACKGROUND: Bone fracture healing is dependent upon the rapid migration and engraftment of bone marrow (BM) progenitor and stem cells to the site of injury. Stromal cell-derived factor-1 plays a crucial role in recruiting BM cells expressing its receptor CXCR4. Recently, a CXCR4 antagonist, plerixafor, has been used to mobilize BM cells into the blood in efforts to enhance cell migration to sites of injury presumably improving healing. In this study, we employed zirconium-89 ((89)Zr)-oxine-labeled BM cells imaged with positron emission tomography (PET)/computed tomography (CT) to visualize and quantitate BM cell trafficking following acute bone injury and to investigate the effect of plerixafor on BM cell homing. Unilateral 1-mm incisions were created in the distal tibia of mice either on the same day (d0) or 24 h (d1) after (89)Zr-oxine-labeled BM cell transfer (n = 4–6, 2–2.3 × 10(7) cells at 9.65–15.7 kBq/10(6) cells). Serial microPET/CT imaging was performed and migration of (89)Zr-labeled cells to the bone injury was quantified. The effects of three daily doses of plerixafor on cell trafficking were evaluated beginning on the day of fracture generation (n = 4–6). The labeled cells localizing to the fracture were analyzed by flow cytometry and immunohistochemistry. RESULTS: In d0- and d1-fracture groups, 0.7% and 1.7% of administered BM cells accumulated within the fracture, respectively. Plerixafor treatment reduced BM cell migration to the fracture by approximately one-third (p < 0.05 for both fracture groups). Flow cytometry analysis of donor cells collected from the injured site revealed a predominance of CD45(+) stem/progenitor cell populations and subsequent histological analysis demonstrated the presence of donor cells engrafted within sites of fracture repair. CONCLUSION: (89)Zr-oxine labeling enabled visualization and quantitation of BM cell recruitment to acute fractures and further demonstrated that plerixafor plays an inhibitory role in this recruitment. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13550-018-0463-8) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6292830
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Springer Berlin Heidelberg
record_format MEDLINE/PubMed
spelling pubmed-62928302018-12-28 Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model Asiedu, Kingsley O. Ferdousi, Munira Ton, Phuongnga T. Adler, Stephen S. Choyke, Peter L. Sato, Noriko EJNMMI Res Original Research BACKGROUND: Bone fracture healing is dependent upon the rapid migration and engraftment of bone marrow (BM) progenitor and stem cells to the site of injury. Stromal cell-derived factor-1 plays a crucial role in recruiting BM cells expressing its receptor CXCR4. Recently, a CXCR4 antagonist, plerixafor, has been used to mobilize BM cells into the blood in efforts to enhance cell migration to sites of injury presumably improving healing. In this study, we employed zirconium-89 ((89)Zr)-oxine-labeled BM cells imaged with positron emission tomography (PET)/computed tomography (CT) to visualize and quantitate BM cell trafficking following acute bone injury and to investigate the effect of plerixafor on BM cell homing. Unilateral 1-mm incisions were created in the distal tibia of mice either on the same day (d0) or 24 h (d1) after (89)Zr-oxine-labeled BM cell transfer (n = 4–6, 2–2.3 × 10(7) cells at 9.65–15.7 kBq/10(6) cells). Serial microPET/CT imaging was performed and migration of (89)Zr-labeled cells to the bone injury was quantified. The effects of three daily doses of plerixafor on cell trafficking were evaluated beginning on the day of fracture generation (n = 4–6). The labeled cells localizing to the fracture were analyzed by flow cytometry and immunohistochemistry. RESULTS: In d0- and d1-fracture groups, 0.7% and 1.7% of administered BM cells accumulated within the fracture, respectively. Plerixafor treatment reduced BM cell migration to the fracture by approximately one-third (p < 0.05 for both fracture groups). Flow cytometry analysis of donor cells collected from the injured site revealed a predominance of CD45(+) stem/progenitor cell populations and subsequent histological analysis demonstrated the presence of donor cells engrafted within sites of fracture repair. CONCLUSION: (89)Zr-oxine labeling enabled visualization and quantitation of BM cell recruitment to acute fractures and further demonstrated that plerixafor plays an inhibitory role in this recruitment. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13550-018-0463-8) contains supplementary material, which is available to authorized users. Springer Berlin Heidelberg 2018-12-13 /pmc/articles/PMC6292830/ /pubmed/30547233 http://dx.doi.org/10.1186/s13550-018-0463-8 Text en © The Author(s). 2018 Open AccessThis is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.
spellingShingle Original Research
Asiedu, Kingsley O.
Ferdousi, Munira
Ton, Phuongnga T.
Adler, Stephen S.
Choyke, Peter L.
Sato, Noriko
Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
title Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
title_full Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
title_fullStr Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
title_full_unstemmed Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
title_short Bone marrow cell homing to sites of acute tibial fracture: (89)Zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
title_sort bone marrow cell homing to sites of acute tibial fracture: (89)zr-oxine cell labeling with positron emission tomographic imaging in a mouse model
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6292830/
https://www.ncbi.nlm.nih.gov/pubmed/30547233
http://dx.doi.org/10.1186/s13550-018-0463-8
work_keys_str_mv AT asiedukingsleyo bonemarrowcellhomingtositesofacutetibialfracture89zroxinecelllabelingwithpositronemissiontomographicimaginginamousemodel
AT ferdousimunira bonemarrowcellhomingtositesofacutetibialfracture89zroxinecelllabelingwithpositronemissiontomographicimaginginamousemodel
AT tonphuongngat bonemarrowcellhomingtositesofacutetibialfracture89zroxinecelllabelingwithpositronemissiontomographicimaginginamousemodel
AT adlerstephens bonemarrowcellhomingtositesofacutetibialfracture89zroxinecelllabelingwithpositronemissiontomographicimaginginamousemodel
AT choykepeterl bonemarrowcellhomingtositesofacutetibialfracture89zroxinecelllabelingwithpositronemissiontomographicimaginginamousemodel
AT satonoriko bonemarrowcellhomingtositesofacutetibialfracture89zroxinecelllabelingwithpositronemissiontomographicimaginginamousemodel