Characterization and assessment of lung and bone marrow derived endothelial cells and their bone regenerative potential

dc.contributor.authorde Lima Perini, Mariana Moraes
dc.contributor.authorValuch, Conner R.
dc.contributor.authorDadwal, Ushashi C.
dc.contributor.authorAwosanya, Olatundun D.
dc.contributor.authorMostardo, Sarah L.
dc.contributor.authorBlosser, Rachel J.
dc.contributor.authorKnox, Adam M.
dc.contributor.authorMcGuire, Anthony C.
dc.contributor.authorBattina, Hanisha L.
dc.contributor.authorNazzal, Murad
dc.contributor.authorKacena, Melissa A.
dc.contributor.authorLi, Jiliang
dc.contributor.departmentBiology, School of Scienceen_US
dc.date.accessioned2023-01-11T17:53:54Z
dc.date.available2023-01-11T17:53:54Z
dc.date.issued2022
dc.description.abstractAngiogenesis is important for successful fracture repair. Aging negatively affects the number and activity of endothelial cells (ECs) and subsequently leads to impaired bone healing. We previously showed that implantation of lung-derived endothelial cells (LECs) improved fracture healing in rats. In this study, we characterized and compared neonatal lung and bone marrow-derived endothelial cells (neonatal LECs and neonatal BMECs) and further asses3sed if implantation of neonatal BMECs could enhance bone healing in both young and aged mice. We assessed neonatal EC tube formation, proliferation, and wound migration ability in vitro in ECs isolated from the bone marrow and lungs of neonatal mice. The in vitro studies demonstrated that both neonatal LECs and neonatal BMECs exhibited EC traits. To test the function of neonatal ECs in vivo, we created a femoral fracture in young and aged mice and implanted a collagen sponge to deliver neonatal BMECs at the fracture site. In the mouse fracture model, endochondral ossification was delayed in aged control mice compared to young controls. Neonatal BMECs significantly improved endochondral bone formation only in aged mice. These data suggest BMECs have potential to enhance aged bone healing. Compared to LECs, BMECs are more feasible for translational cell therapy and clinical applications in bone repair. Future studies are needed to examine the fate and function of BMECs implanted into the fracture sites.en_US
dc.eprint.versionFinal published versionen_US
dc.identifier.citationde Lima Perini, M. M., Valuch, C. R., Dadwal, U. C., Awosanya, O. D., Mostardo, S. L., Blosser, R. J., Knox, A. M., McGuire, A. C., Battina, H. L., Nazzal, M., Kacena, M. A., Li, J. (2022). Characterization and assessment of lung and bone marrow derived endothelial cells and their bone regenerative potential. Frontiers in Endocrinology. https://doi.org/10.3389/fendo.2022.935391en_US
dc.identifier.urihttps://hdl.handle.net/1805/30920
dc.language.isoenen_US
dc.publisherFrontiersen_US
dc.relation.isversionof10.3389/fendo.2022.935391en_US
dc.relation.journalFrontiers in Endocrinologyen_US
dc.rightsAttribution 4.0 International*
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/*
dc.sourcePublisheren_US
dc.subjectagingen_US
dc.subjectfracture healingen_US
dc.subjectendothelial progenitor cellsen_US
dc.titleCharacterization and assessment of lung and bone marrow derived endothelial cells and their bone regenerative potentialen_US
dc.typeArticleen_US
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
fendo-13-935391.pdf
Size:
6.69 MB
Format:
Adobe Portable Document Format
Description:
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.99 KB
Format:
Item-specific license agreed upon to submission
Description: