Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation

Abstract Background Bone formation plays an important role in early tendon–bone healing after anterior cruciate ligament reconstruction (ACLR). Dedifferentiated osteogenic bone marrow mesenchymal stem cells (De-BMSCs) have enhanced osteogenic potential. This study aimed to investigate the effect of...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Kai Tie, Jinghang Cai, Jun Qin, Hao Xiao, Yangfan Shangguan, Hui Wang, Liaobin Chen
Formato: article
Lenguaje:EN
Publicado: BMC 2021
Materias:
Acceso en línea:https://doaj.org/article/14d4ab3d803a4b4bb4bb931faf826d0c
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:14d4ab3d803a4b4bb4bb931faf826d0c
record_format dspace
spelling oai:doaj.org-article:14d4ab3d803a4b4bb4bb931faf826d0c2021-11-21T12:04:45ZNanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation10.1186/s13287-021-02643-91757-6512https://doaj.org/article/14d4ab3d803a4b4bb4bb931faf826d0c2021-11-01T00:00:00Zhttps://doi.org/10.1186/s13287-021-02643-9https://doaj.org/toc/1757-6512Abstract Background Bone formation plays an important role in early tendon–bone healing after anterior cruciate ligament reconstruction (ACLR). Dedifferentiated osteogenic bone marrow mesenchymal stem cells (De-BMSCs) have enhanced osteogenic potential. This study aimed to investigate the effect of De-BMSCs transplantation on the promotion of bone formation at the tendon–bone interface after ACLR and to further explore the molecular mechanism of the enhanced osteogenic potential of De-BMSCs. Methods BMSCs from the femurs and tibias of New Zealand white rabbits were subjected to osteogenic induction and then cultured in medium without osteogenic factors; the obtained cell population was termed De-BMSCs. De-BMSCs were induced to undergo osteo-, chondro- and adipo-differentiation in vitro to examine the characteristics of primitive stem cells. An ACLR model with a semitendinosus tendon was established in rabbits, and the animals were divided into a control group, BMSCs group, and De-BMSCs group. At 12 weeks after surgery, the rabbits in each group were sacrificed to evaluate tendon–bone healing by histologic staining, micro-computed tomography (micro-CT) examination, and biomechanical testing. During osteogenic differentiation of De-BMSCs, an siRNA targeting nuclear factor of activated T-cells 1 (NFATc1) was used to verify the molecular mechanism of the enhanced osteogenic potential of De-BMSCs. Results De-BMSCs exhibited some properties similar to BMSCs, including multiple differentiation potential and cell surface markers. Bone formation at the tendon–bone interface in the De-BMSCs group was significantly increased, and biomechanical strength was significantly improved. During the osteogenic differentiation of De-BMSCs, the expression of Nanog and NFATc1 was synergistically increased, which promoted the interaction of NFATc1 and Osterix, resulting in increased expression of osteoblast marker genes such as COL1A, OCN, and OPN. Conclusions De-BMSCs transplantation could promote bone formation at the tendon–bone interface after ACLR and improve the biomechanical strength of the reconstruction. The Nanog/NFATc1/Osterix signaling pathway mediated the enhanced osteogenic differentiation efficiency of De-BMSCs.Kai TieJinghang CaiJun QinHao XiaoYangfan ShangguanHui WangLiaobin ChenBMCarticleDedifferentiationBMSCsOsteogenic differentiationAnterior cruciate ligamentTendon–bone healingMedicine (General)R5-920BiochemistryQD415-436ENStem Cell Research & Therapy, Vol 12, Iss 1, Pp 1-16 (2021)
institution DOAJ
collection DOAJ
language EN
topic Dedifferentiation
BMSCs
Osteogenic differentiation
Anterior cruciate ligament
Tendon–bone healing
Medicine (General)
R5-920
Biochemistry
QD415-436
spellingShingle Dedifferentiation
BMSCs
Osteogenic differentiation
Anterior cruciate ligament
Tendon–bone healing
Medicine (General)
R5-920
Biochemistry
QD415-436
Kai Tie
Jinghang Cai
Jun Qin
Hao Xiao
Yangfan Shangguan
Hui Wang
Liaobin Chen
Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation
description Abstract Background Bone formation plays an important role in early tendon–bone healing after anterior cruciate ligament reconstruction (ACLR). Dedifferentiated osteogenic bone marrow mesenchymal stem cells (De-BMSCs) have enhanced osteogenic potential. This study aimed to investigate the effect of De-BMSCs transplantation on the promotion of bone formation at the tendon–bone interface after ACLR and to further explore the molecular mechanism of the enhanced osteogenic potential of De-BMSCs. Methods BMSCs from the femurs and tibias of New Zealand white rabbits were subjected to osteogenic induction and then cultured in medium without osteogenic factors; the obtained cell population was termed De-BMSCs. De-BMSCs were induced to undergo osteo-, chondro- and adipo-differentiation in vitro to examine the characteristics of primitive stem cells. An ACLR model with a semitendinosus tendon was established in rabbits, and the animals were divided into a control group, BMSCs group, and De-BMSCs group. At 12 weeks after surgery, the rabbits in each group were sacrificed to evaluate tendon–bone healing by histologic staining, micro-computed tomography (micro-CT) examination, and biomechanical testing. During osteogenic differentiation of De-BMSCs, an siRNA targeting nuclear factor of activated T-cells 1 (NFATc1) was used to verify the molecular mechanism of the enhanced osteogenic potential of De-BMSCs. Results De-BMSCs exhibited some properties similar to BMSCs, including multiple differentiation potential and cell surface markers. Bone formation at the tendon–bone interface in the De-BMSCs group was significantly increased, and biomechanical strength was significantly improved. During the osteogenic differentiation of De-BMSCs, the expression of Nanog and NFATc1 was synergistically increased, which promoted the interaction of NFATc1 and Osterix, resulting in increased expression of osteoblast marker genes such as COL1A, OCN, and OPN. Conclusions De-BMSCs transplantation could promote bone formation at the tendon–bone interface after ACLR and improve the biomechanical strength of the reconstruction. The Nanog/NFATc1/Osterix signaling pathway mediated the enhanced osteogenic differentiation efficiency of De-BMSCs.
format article
author Kai Tie
Jinghang Cai
Jun Qin
Hao Xiao
Yangfan Shangguan
Hui Wang
Liaobin Chen
author_facet Kai Tie
Jinghang Cai
Jun Qin
Hao Xiao
Yangfan Shangguan
Hui Wang
Liaobin Chen
author_sort Kai Tie
title Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation
title_short Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation
title_full Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation
title_fullStr Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation
title_full_unstemmed Nanog/NFATc1/Osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after ACL reconstruction with De-BMSCs transplantation
title_sort nanog/nfatc1/osterix signaling pathway-mediated promotion of bone formation at the tendon–bone interface after acl reconstruction with de-bmscs transplantation
publisher BMC
publishDate 2021
url https://doaj.org/article/14d4ab3d803a4b4bb4bb931faf826d0c
work_keys_str_mv AT kaitie nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
AT jinghangcai nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
AT junqin nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
AT haoxiao nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
AT yangfanshangguan nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
AT huiwang nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
AT liaobinchen nanognfatc1osterixsignalingpathwaymediatedpromotionofboneformationatthetendonboneinterfaceafteraclreconstructionwithdebmscstransplantation
_version_ 1718419228380364800