Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1

An important objective of vascularized tissue regeneration is to develop agents for osteonecrosis. We aimed to identify the pro-angiogenic and osteogenic efficacy of adipose tissue-derived (AD) pericytes combined with Nel-like protein-1 (NELL-1) to investigate the therapeutic effects on osteonecrosi...

Full description

Bibliographic Details
Main Authors: Hyun-Ju An, Kyung Rae Ko, Minjung Baek, Yoonhui Jeong, Hyeon Hae Lee, Hyungkyung Kim, Do Kyung Kim, So-Young Lee, Soonchul Lee
Format: Article
Language:English
Published: MDPI AG 2021-08-01
Series:Cells
Subjects:
Online Access:https://www.mdpi.com/2073-4409/10/9/2244
id doaj-7494499e3ca649bc851a74511a08da62
record_format Article
spelling doaj-7494499e3ca649bc851a74511a08da622021-09-25T23:52:08ZengMDPI AGCells2073-44092021-08-01102244224410.3390/cells10092244Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1Hyun-Ju An0Kyung Rae Ko1Minjung Baek2Yoonhui Jeong3Hyeon Hae Lee4Hyungkyung Kim5Do Kyung Kim6So-Young Lee7Soonchul Lee8Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, KoreaDepartment of Orthopaedic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, KoreaDepartment of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, KoreaDepartment of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, KoreaDepartment of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, KoreaDepartment of Pathology, Kyung Hee University Hospital at Gangdong, Kyung Hee University, College of Medicine, 892 Dongnam-Ro, Gangdong-gu, Seoul 05278, KoreaCHA Graduate School of Medicine, 120 Hyeryong-Ro, Pocheon-si 11160, Gyeonggi-do, KoreaDepartment of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, KoreaDepartment of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-Ro, Seongnam-si 13496, Gyeonggi-do, KoreaAn important objective of vascularized tissue regeneration is to develop agents for osteonecrosis. We aimed to identify the pro-angiogenic and osteogenic efficacy of adipose tissue-derived (AD) pericytes combined with Nel-like protein-1 (NELL-1) to investigate the therapeutic effects on osteonecrosis. Tube formation and cell migration were assessed to determine the pro-angiogenic efficacy. Vessel formation was evaluated in vivo using the chorioallantoic membrane assay. A mouse model with a 2.5 mm necrotic bone fragment in the femoral shaft was used as a substitute for osteonecrosis in humans. Bone formation was assessed radiographically (plain radiographs, three-dimensional images, and quantitative analyses), and histomorphometric analyses were performed. To identify factors related to the effects of NELL-1, analysis using microarrays, qRT-PCR, and Western blotting was performed. The results for pro-angiogenic efficacy evaluation identified synergistic effects of pericytes and NELL-1 on tube formation, cell migration, and vessel formation. For osteogenic efficacy analysis, the mouse model for osteonecrosis was treated in combination with pericytes and NELL-1, and the results showed maximum bone formation using radiographic images and quantitative analyses, compared with other treatment groups and showed robust bone and vessel formation using histomorphometric analysis. We identified an association between FGF2 and the effects of NELL-1 using array-based analysis. Thus, combinatorial therapy using AD pericytes and NELL-1 may have potential as a novel treatment for osteonecrosis.https://www.mdpi.com/2073-4409/10/9/2244pericytes<i>Nel</i>-like protein-1angiogenesisbone regenerationbone formationosteonecrosis
collection DOAJ
language English
format Article
sources DOAJ
author Hyun-Ju An
Kyung Rae Ko
Minjung Baek
Yoonhui Jeong
Hyeon Hae Lee
Hyungkyung Kim
Do Kyung Kim
So-Young Lee
Soonchul Lee
spellingShingle Hyun-Ju An
Kyung Rae Ko
Minjung Baek
Yoonhui Jeong
Hyeon Hae Lee
Hyungkyung Kim
Do Kyung Kim
So-Young Lee
Soonchul Lee
Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1
Cells
pericytes
<i>Nel</i>-like protein-1
angiogenesis
bone regeneration
bone formation
osteonecrosis
author_facet Hyun-Ju An
Kyung Rae Ko
Minjung Baek
Yoonhui Jeong
Hyeon Hae Lee
Hyungkyung Kim
Do Kyung Kim
So-Young Lee
Soonchul Lee
author_sort Hyun-Ju An
title Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1
title_short Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1
title_full Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1
title_fullStr Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1
title_full_unstemmed Pro-Angiogenic and Osteogenic Effects of Adipose Tissue-Derived Pericytes Synergistically Enhanced by Nel-like Protein-1
title_sort pro-angiogenic and osteogenic effects of adipose tissue-derived pericytes synergistically enhanced by nel-like protein-1
publisher MDPI AG
series Cells
issn 2073-4409
publishDate 2021-08-01
description An important objective of vascularized tissue regeneration is to develop agents for osteonecrosis. We aimed to identify the pro-angiogenic and osteogenic efficacy of adipose tissue-derived (AD) pericytes combined with Nel-like protein-1 (NELL-1) to investigate the therapeutic effects on osteonecrosis. Tube formation and cell migration were assessed to determine the pro-angiogenic efficacy. Vessel formation was evaluated in vivo using the chorioallantoic membrane assay. A mouse model with a 2.5 mm necrotic bone fragment in the femoral shaft was used as a substitute for osteonecrosis in humans. Bone formation was assessed radiographically (plain radiographs, three-dimensional images, and quantitative analyses), and histomorphometric analyses were performed. To identify factors related to the effects of NELL-1, analysis using microarrays, qRT-PCR, and Western blotting was performed. The results for pro-angiogenic efficacy evaluation identified synergistic effects of pericytes and NELL-1 on tube formation, cell migration, and vessel formation. For osteogenic efficacy analysis, the mouse model for osteonecrosis was treated in combination with pericytes and NELL-1, and the results showed maximum bone formation using radiographic images and quantitative analyses, compared with other treatment groups and showed robust bone and vessel formation using histomorphometric analysis. We identified an association between FGF2 and the effects of NELL-1 using array-based analysis. Thus, combinatorial therapy using AD pericytes and NELL-1 may have potential as a novel treatment for osteonecrosis.
topic pericytes
<i>Nel</i>-like protein-1
angiogenesis
bone regeneration
bone formation
osteonecrosis
url https://www.mdpi.com/2073-4409/10/9/2244
work_keys_str_mv AT hyunjuan proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT kyungraeko proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT minjungbaek proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT yoonhuijeong proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT hyeonhaelee proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT hyungkyungkim proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT dokyungkim proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT soyounglee proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
AT soonchullee proangiogenicandosteogeniceffectsofadiposetissuederivedpericytessynergisticallyenhancedbynellikeprotein1
_version_ 1717367762628640769