Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration

Abstract Replacement of lost cranial bone (partly mesodermal and partly neural crest‐derived) is challenging and includes the use of nonviable allografts. To revitalize allografts, bone marrow‐derived mesenchymal stromal cells (mesoderm‐derived BM‐MSCs) have been used with limited success. We hypoth...

Full description

Bibliographic Details
Main Authors: Juliane D. Glaeser, Phillip Behrens, Tina Stefanovic, Khosrowdad Salehi, Angela Papalamprou, Wafa Tawackoli, Melodie F. Metzger, Samuel Eberlein, Trevor Nelson, Yasaman Arabi, Kevin Kim, Robert H. Baloh, Shiran Ben‐David, Doron Cohn‐Schwartz, Robert Ryu, Hyun W. Bae, Zulma Gazit, Dmitriy Sheyn
Format: Article
Language:English
Published: Wiley 2021-05-01
Series:Stem Cells Translational Medicine
Subjects:
MSC
Online Access:https://doi.org/10.1002/sctm.20-0364
id doaj-623fd8f367c6432984c32584341c5193
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Juliane D. Glaeser
Phillip Behrens
Tina Stefanovic
Khosrowdad Salehi
Angela Papalamprou
Wafa Tawackoli
Melodie F. Metzger
Samuel Eberlein
Trevor Nelson
Yasaman Arabi
Kevin Kim
Robert H. Baloh
Shiran Ben‐David
Doron Cohn‐Schwartz
Robert Ryu
Hyun W. Bae
Zulma Gazit
Dmitriy Sheyn
spellingShingle Juliane D. Glaeser
Phillip Behrens
Tina Stefanovic
Khosrowdad Salehi
Angela Papalamprou
Wafa Tawackoli
Melodie F. Metzger
Samuel Eberlein
Trevor Nelson
Yasaman Arabi
Kevin Kim
Robert H. Baloh
Shiran Ben‐David
Doron Cohn‐Schwartz
Robert Ryu
Hyun W. Bae
Zulma Gazit
Dmitriy Sheyn
Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
Stem Cells Translational Medicine
allograft
bone healing
cranial repair
MSC
neural crest cells
author_facet Juliane D. Glaeser
Phillip Behrens
Tina Stefanovic
Khosrowdad Salehi
Angela Papalamprou
Wafa Tawackoli
Melodie F. Metzger
Samuel Eberlein
Trevor Nelson
Yasaman Arabi
Kevin Kim
Robert H. Baloh
Shiran Ben‐David
Doron Cohn‐Schwartz
Robert Ryu
Hyun W. Bae
Zulma Gazit
Dmitriy Sheyn
author_sort Juliane D. Glaeser
title Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
title_short Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
title_full Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
title_fullStr Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
title_full_unstemmed Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
title_sort neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integration
publisher Wiley
series Stem Cells Translational Medicine
issn 2157-6564
2157-6580
publishDate 2021-05-01
description Abstract Replacement of lost cranial bone (partly mesodermal and partly neural crest‐derived) is challenging and includes the use of nonviable allografts. To revitalize allografts, bone marrow‐derived mesenchymal stromal cells (mesoderm‐derived BM‐MSCs) have been used with limited success. We hypothesize that coating of allografts with induced neural crest cell‐mesenchymal progenitor cells (iNCC‐MPCs) improves implant‐to‐bone integration in mouse cranial defects. Human induced pluripotent stem cells were reprogramed from dermal fibroblasts, differentiated to iNCCs and then to iNCC‐MPCs. BM‐MSCs were used as reference. Cells were labeled with luciferase (Luc2) and characterized for MSC consensus markers expression, differentiation, and risk of cellular transformation. A calvarial defect was created in non‐obese diabetic/severe combined immunodeficiency (NOD/SCID) mice and allografts were implanted, with or without cell coating. Bioluminescence imaging (BLI), microcomputed tomography (μCT), histology, immunofluorescence, and biomechanical tests were performed. Characterization of iNCC‐MPC‐Luc2 vs BM‐MSC‐Luc2 showed no difference in MSC markers expression and differentiation in vitro. In vivo, BLI indicated survival of both cell types for at least 8 weeks. At week 8, μCT analysis showed enhanced structural parameters in the iNCC‐MPC‐Luc2 group and increased bone volume in the BM‐MSC‐Luc2 group compared to controls. Histology demonstrated improved integration of iNCC‐MPC‐Luc2 allografts compared to BM‐MSC‐Luc2 group and controls. Human osteocalcin and collagen type 1 were detected at the allograft‐host interphase in cell‐seeded groups. The iNCC‐MPC‐Luc2 group also demonstrated improved biomechanical properties compared to BM‐MSC‐Luc2 implants and cell‐free controls. Our results show an improved integration of iNCC‐MPC‐Luc2‐coated allografts compared to BM‐MSC‐Luc2 and controls, suggesting the use of iNCC‐MPCs as potential cell source for cranial bone repair.
topic allograft
bone healing
cranial repair
MSC
neural crest cells
url https://doi.org/10.1002/sctm.20-0364
work_keys_str_mv AT julianedglaeser neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT phillipbehrens neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT tinastefanovic neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT khosrowdadsalehi neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT angelapapalamprou neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT wafatawackoli neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT melodiefmetzger neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT samueleberlein neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT trevornelson neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT yasamanarabi neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT kevinkim neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT roberthbaloh neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT shiranbendavid neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT doroncohnschwartz neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT robertryu neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT hyunwbae neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT zulmagazit neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
AT dmitriysheyn neuralcrestderivedmesenchymalprogenitorcellsenhancecranialallograftintegration
_version_ 1721527152794402816
spelling doaj-623fd8f367c6432984c32584341c51932021-04-14T16:20:02ZengWileyStem Cells Translational Medicine2157-65642157-65802021-05-0110579780910.1002/sctm.20-0364Neural crest‐derived mesenchymal progenitor cells enhance cranial allograft integrationJuliane D. Glaeser0Phillip Behrens1Tina Stefanovic2Khosrowdad Salehi3Angela Papalamprou4Wafa Tawackoli5Melodie F. Metzger6Samuel Eberlein7Trevor Nelson8Yasaman Arabi9Kevin Kim10Robert H. Baloh11Shiran Ben‐David12Doron Cohn‐Schwartz13Robert Ryu14Hyun W. Bae15Zulma Gazit16Dmitriy Sheyn17Orthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USADepartment of Orthopedics Cedars‐Sinai Medical Center Los Angeles California USADepartment of Orthopedics Cedars‐Sinai Medical Center Los Angeles California USADepartment of Orthopedics Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USABoard of Governors Regenerative Medicine Institute Cedars‐Sinai Medical Center Los Angeles California USABoard of Governors Regenerative Medicine Institute Cedars‐Sinai Medical Center Los Angeles California USABoard of Governors Regenerative Medicine Institute Cedars‐Sinai Medical Center Los Angeles California USADepartment of Surgery Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USABoard of Governors Regenerative Medicine Institute Cedars‐Sinai Medical Center Los Angeles California USAOrthopaedic Stem Cell Research Laboratory Cedars‐Sinai Medical Center Los Angeles California USAAbstract Replacement of lost cranial bone (partly mesodermal and partly neural crest‐derived) is challenging and includes the use of nonviable allografts. To revitalize allografts, bone marrow‐derived mesenchymal stromal cells (mesoderm‐derived BM‐MSCs) have been used with limited success. We hypothesize that coating of allografts with induced neural crest cell‐mesenchymal progenitor cells (iNCC‐MPCs) improves implant‐to‐bone integration in mouse cranial defects. Human induced pluripotent stem cells were reprogramed from dermal fibroblasts, differentiated to iNCCs and then to iNCC‐MPCs. BM‐MSCs were used as reference. Cells were labeled with luciferase (Luc2) and characterized for MSC consensus markers expression, differentiation, and risk of cellular transformation. A calvarial defect was created in non‐obese diabetic/severe combined immunodeficiency (NOD/SCID) mice and allografts were implanted, with or without cell coating. Bioluminescence imaging (BLI), microcomputed tomography (μCT), histology, immunofluorescence, and biomechanical tests were performed. Characterization of iNCC‐MPC‐Luc2 vs BM‐MSC‐Luc2 showed no difference in MSC markers expression and differentiation in vitro. In vivo, BLI indicated survival of both cell types for at least 8 weeks. At week 8, μCT analysis showed enhanced structural parameters in the iNCC‐MPC‐Luc2 group and increased bone volume in the BM‐MSC‐Luc2 group compared to controls. Histology demonstrated improved integration of iNCC‐MPC‐Luc2 allografts compared to BM‐MSC‐Luc2 group and controls. Human osteocalcin and collagen type 1 were detected at the allograft‐host interphase in cell‐seeded groups. The iNCC‐MPC‐Luc2 group also demonstrated improved biomechanical properties compared to BM‐MSC‐Luc2 implants and cell‐free controls. Our results show an improved integration of iNCC‐MPC‐Luc2‐coated allografts compared to BM‐MSC‐Luc2 and controls, suggesting the use of iNCC‐MPCs as potential cell source for cranial bone repair.https://doi.org/10.1002/sctm.20-0364allograftbone healingcranial repairMSCneural crest cells