Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments

Background: The biological mechanisms that contribute to atrophic long bone non-union are poorly understood. Multipotential mesenchymal stromal cells (MSCs) are key contributors to bone formation and are recognised as important mediators of blood vessel formation. This study examines the role of MSC...

Full description

Bibliographic Details
Main Authors: R. J. Cuthbert, E. Jones, C. Sanjurjo-Rodríguez, A. Lotfy, P. Ganguly, S. M. Churchman, P. Kastana, H. B. Tan, D. McGonagle, E. Papadimitriou, P. V. Giannoudis
Format: Article
Language:English
Published: MDPI AG 2020-05-01
Series:Journal of Clinical Medicine
Subjects:
Online Access:https://www.mdpi.com/2077-0383/9/6/1628
_version_ 1797566807148593152
author R. J. Cuthbert
E. Jones
C. Sanjurjo-Rodríguez
A. Lotfy
P. Ganguly
S. M. Churchman
P. Kastana
H. B. Tan
D. McGonagle
E. Papadimitriou
P. V. Giannoudis
author_facet R. J. Cuthbert
E. Jones
C. Sanjurjo-Rodríguez
A. Lotfy
P. Ganguly
S. M. Churchman
P. Kastana
H. B. Tan
D. McGonagle
E. Papadimitriou
P. V. Giannoudis
author_sort R. J. Cuthbert
collection DOAJ
description Background: The biological mechanisms that contribute to atrophic long bone non-union are poorly understood. Multipotential mesenchymal stromal cells (MSCs) are key contributors to bone formation and are recognised as important mediators of blood vessel formation. This study examines the role of MSCs in tissue formation at the site of atrophic non-union. Materials and Methods: Tissue and MSCs from non-union sites (<i>n</i> = 20) and induced periosteal (IP) membrane formed following the Masquelet bone reconstruction technique (<i>n</i> = 15) or bone marrow (<i>n</i> = 8) were compared. MSC content, differentiation, and influence on angiogenesis were measured in vitro. Cell content and vasculature measurements were performed by flow cytometry and histology, and gene expression was measured by quantitative polymerase chain reaction (qPCR). Results: MSCs from non-union sites had comparable differentiation potential to bone marrow MSCs. Compared with induced periosteum, non-union tissue contained similar proportion of colony-forming cells, but a greater proportion of pericytes (<i>p</i> = 0.036), and endothelial cells (<i>p</i> = 0.016) and blood vessels were more numerous (<i>p</i> = 0.001) with smaller luminal diameter (<i>p</i> = 0.046). MSCs showed marked differences in angiogenic transcripts depending on the source, and those from induced periosteum, but not non-union tissue, inhibited early stages of in vitro angiogenesis. Conclusions: In vitro, non-union site derived MSCs have no impairment of differentiation capacity, but they differ from IP-derived MSCs in mediating angiogenesis. Local MSCs may thus be strongly implicated in the formation of the immature vascular network at the non-union site. Attention should be given to their angiogenic support profile when selecting MSCs for regenerative therapy.
first_indexed 2024-03-10T19:32:35Z
format Article
id doaj.art-510df18d1e564e628b465326924728d0
institution Directory Open Access Journal
issn 2077-0383
language English
last_indexed 2024-03-10T19:32:35Z
publishDate 2020-05-01
publisher MDPI AG
record_format Article
series Journal of Clinical Medicine
spelling doaj.art-510df18d1e564e628b465326924728d02023-11-20T02:00:37ZengMDPI AGJournal of Clinical Medicine2077-03832020-05-0196162810.3390/jcm9061628Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic EnvironmentsR. J. Cuthbert0E. Jones1C. Sanjurjo-Rodríguez2A. Lotfy3P. Ganguly4S. M. Churchman5P. Kastana6H. B. Tan7D. McGonagle8E. Papadimitriou9P. V. Giannoudis10Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKBiotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, EgyptLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKDepartment of Pharmacy, School of Health Sciences, University of Patras, Patras 265 04, GreeceLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKDepartment of Pharmacy, School of Health Sciences, University of Patras, Patras 265 04, GreeceLeeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UKBackground: The biological mechanisms that contribute to atrophic long bone non-union are poorly understood. Multipotential mesenchymal stromal cells (MSCs) are key contributors to bone formation and are recognised as important mediators of blood vessel formation. This study examines the role of MSCs in tissue formation at the site of atrophic non-union. Materials and Methods: Tissue and MSCs from non-union sites (<i>n</i> = 20) and induced periosteal (IP) membrane formed following the Masquelet bone reconstruction technique (<i>n</i> = 15) or bone marrow (<i>n</i> = 8) were compared. MSC content, differentiation, and influence on angiogenesis were measured in vitro. Cell content and vasculature measurements were performed by flow cytometry and histology, and gene expression was measured by quantitative polymerase chain reaction (qPCR). Results: MSCs from non-union sites had comparable differentiation potential to bone marrow MSCs. Compared with induced periosteum, non-union tissue contained similar proportion of colony-forming cells, but a greater proportion of pericytes (<i>p</i> = 0.036), and endothelial cells (<i>p</i> = 0.016) and blood vessels were more numerous (<i>p</i> = 0.001) with smaller luminal diameter (<i>p</i> = 0.046). MSCs showed marked differences in angiogenic transcripts depending on the source, and those from induced periosteum, but not non-union tissue, inhibited early stages of in vitro angiogenesis. Conclusions: In vitro, non-union site derived MSCs have no impairment of differentiation capacity, but they differ from IP-derived MSCs in mediating angiogenesis. Local MSCs may thus be strongly implicated in the formation of the immature vascular network at the non-union site. Attention should be given to their angiogenic support profile when selecting MSCs for regenerative therapy.https://www.mdpi.com/2077-0383/9/6/1628MSCsfracture healingnon-unioninduced periosteumosteogenesisregenerative medicine
spellingShingle R. J. Cuthbert
E. Jones
C. Sanjurjo-Rodríguez
A. Lotfy
P. Ganguly
S. M. Churchman
P. Kastana
H. B. Tan
D. McGonagle
E. Papadimitriou
P. V. Giannoudis
Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments
Journal of Clinical Medicine
MSCs
fracture healing
non-union
induced periosteum
osteogenesis
regenerative medicine
title Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments
title_full Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments
title_fullStr Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments
title_full_unstemmed Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments
title_short Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments
title_sort regulation of angiogenesis discriminates tissue resident mscs from effective and defective osteogenic environments
topic MSCs
fracture healing
non-union
induced periosteum
osteogenesis
regenerative medicine
url https://www.mdpi.com/2077-0383/9/6/1628
work_keys_str_mv AT rjcuthbert regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT ejones regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT csanjurjorodriguez regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT alotfy regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT pganguly regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT smchurchman regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT pkastana regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT hbtan regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT dmcgonagle regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT epapadimitriou regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments
AT pvgiannoudis regulationofangiogenesisdiscriminatestissueresidentmscsfromeffectiveanddefectiveosteogenicenvironments