Cargando…

Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4

Evidence indicates that microRNAs (miRNAs) play vital roles in regulating osteogenic differentiation and bone formation. Methods: Here, we show that a polyethyleneimine (PEI)-functionalized graphene oxide (GO) complex efficiently loaded with the miR-214 inhibitor is assembled into silk fibroin/hydro...

Descripción completa

Detalles Bibliográficos
Autores principales: Ou, Lingling, Lan, Yong, Feng, Zhiqiang, Feng, Longbao, Yang, Junjie, Liu, Yu, Bian, Liming, Tan, Jiali, Lai, Renfa, Guo, Rui
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6599658/
https://www.ncbi.nlm.nih.gov/pubmed/31285777
http://dx.doi.org/10.7150/thno.34676
_version_ 1783430977464303616
author Ou, Lingling
Lan, Yong
Feng, Zhiqiang
Feng, Longbao
Yang, Junjie
Liu, Yu
Bian, Liming
Tan, Jiali
Lai, Renfa
Guo, Rui
author_facet Ou, Lingling
Lan, Yong
Feng, Zhiqiang
Feng, Longbao
Yang, Junjie
Liu, Yu
Bian, Liming
Tan, Jiali
Lai, Renfa
Guo, Rui
author_sort Ou, Lingling
collection PubMed
description Evidence indicates that microRNAs (miRNAs) play vital roles in regulating osteogenic differentiation and bone formation. Methods: Here, we show that a polyethyleneimine (PEI)-functionalized graphene oxide (GO) complex efficiently loaded with the miR-214 inhibitor is assembled into silk fibroin/hydroxyapatite (SF/HAP) scaffolds that spatially control the release of the miR-214 inhibitor. Results: SF/HAP/GO scaffolds with nanosized GO show high mechanical strength, and their hierarchical microporous structures promote cell adhesion and growth. The SF/HAP/GO-PEI scaffolds loaded with mir-214 inhibitor (SF/HAP/GPM) were tested for their ability to enhance osteogenic differentiation by inhibiting the expression of miR-214 while inversely increasing the expression of activating transcription factor 4 (ATF4) and activating the Akt and ERK1/2 signaling pathways in mouse osteoblastic cells (MC3T3-E1) in vitro. Similarly, the scaffolds activated the osteoblastic activity of endogenous osteoblast cells to repair critical-sized bone defects in rats without the need for loading osteoblast cells. Conclusion: This technology is used to increase osteogenic differentiation and mineralized bone formation in bone defects, which helps to achieve cell-free scaffold-based miRNA-inhibitor therapy for bone tissue engineering.
format Online
Article
Text
id pubmed-6599658
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-65996582019-07-08 Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4 Ou, Lingling Lan, Yong Feng, Zhiqiang Feng, Longbao Yang, Junjie Liu, Yu Bian, Liming Tan, Jiali Lai, Renfa Guo, Rui Theranostics Research Paper Evidence indicates that microRNAs (miRNAs) play vital roles in regulating osteogenic differentiation and bone formation. Methods: Here, we show that a polyethyleneimine (PEI)-functionalized graphene oxide (GO) complex efficiently loaded with the miR-214 inhibitor is assembled into silk fibroin/hydroxyapatite (SF/HAP) scaffolds that spatially control the release of the miR-214 inhibitor. Results: SF/HAP/GO scaffolds with nanosized GO show high mechanical strength, and their hierarchical microporous structures promote cell adhesion and growth. The SF/HAP/GO-PEI scaffolds loaded with mir-214 inhibitor (SF/HAP/GPM) were tested for their ability to enhance osteogenic differentiation by inhibiting the expression of miR-214 while inversely increasing the expression of activating transcription factor 4 (ATF4) and activating the Akt and ERK1/2 signaling pathways in mouse osteoblastic cells (MC3T3-E1) in vitro. Similarly, the scaffolds activated the osteoblastic activity of endogenous osteoblast cells to repair critical-sized bone defects in rats without the need for loading osteoblast cells. Conclusion: This technology is used to increase osteogenic differentiation and mineralized bone formation in bone defects, which helps to achieve cell-free scaffold-based miRNA-inhibitor therapy for bone tissue engineering. Ivyspring International Publisher 2019-06-09 /pmc/articles/PMC6599658/ /pubmed/31285777 http://dx.doi.org/10.7150/thno.34676 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Ou, Lingling
Lan, Yong
Feng, Zhiqiang
Feng, Longbao
Yang, Junjie
Liu, Yu
Bian, Liming
Tan, Jiali
Lai, Renfa
Guo, Rui
Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4
title Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4
title_full Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4
title_fullStr Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4
title_full_unstemmed Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4
title_short Functionalization of SF/HAP Scaffold with GO-PEI-miRNA inhibitor Complexes to Enhance Bone Regeneration through Activating Transcription Factor 4
title_sort functionalization of sf/hap scaffold with go-pei-mirna inhibitor complexes to enhance bone regeneration through activating transcription factor 4
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6599658/
https://www.ncbi.nlm.nih.gov/pubmed/31285777
http://dx.doi.org/10.7150/thno.34676
work_keys_str_mv AT oulingling functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT lanyong functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT fengzhiqiang functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT fenglongbao functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT yangjunjie functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT liuyu functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT bianliming functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT tanjiali functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT lairenfa functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4
AT guorui functionalizationofsfhapscaffoldwithgopeimirnainhibitorcomplexestoenhanceboneregenerationthroughactivatingtranscriptionfactor4