Cargando…

Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing

Three dimension (3D) printed scaffolds have been shown to be superior in promoting tissue repair, but the cell‐level specific regulatory network activated by 3D printing scaffolds with different material components to form a symbiosis niche have not been systematically revealed. Here, three typical...

Descripción completa

Detalles Bibliográficos
Autores principales: Ji, Ce, Qiu, Minglong, Ruan, Huitong, Li, Cuidi, Cheng, Liang, Wang, Juan, Li, Changwei, Qi, Jin, Cui, Wenguo, Deng, Lianfu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8922091/
https://www.ncbi.nlm.nih.gov/pubmed/35040587
http://dx.doi.org/10.1002/advs.202105194
_version_ 1784669456435249152
author Ji, Ce
Qiu, Minglong
Ruan, Huitong
Li, Cuidi
Cheng, Liang
Wang, Juan
Li, Changwei
Qi, Jin
Cui, Wenguo
Deng, Lianfu
author_facet Ji, Ce
Qiu, Minglong
Ruan, Huitong
Li, Cuidi
Cheng, Liang
Wang, Juan
Li, Changwei
Qi, Jin
Cui, Wenguo
Deng, Lianfu
author_sort Ji, Ce
collection PubMed
description Three dimension (3D) printed scaffolds have been shown to be superior in promoting tissue repair, but the cell‐level specific regulatory network activated by 3D printing scaffolds with different material components to form a symbiosis niche have not been systematically revealed. Here, three typical 3D printed scaffolds, including natural polymer hydrogel (gelatin‐methacryloyl, GelMA), synthetic polymer material (polycaprolactone, PCL), and bioceramic (β‐tricalcium phosphate, β‐TCP), are fabricated to explore the regulating effect of the symbiotic microenvironment during bone healing. Enrichment analysis show that hydrogel promotes tissue regeneration and reconstruction by improving blood vessel generation by enhancing oxygen transport and red blood cell development. The PCL scaffold regulates cell proliferation and differentiation by promoting cellular senescence, cell cycle and deoxyribonucleic acid (DNA) replication pathways, accelerating the process of endochondral ossification, and the formation of callus. The β‐TCP scaffold can specifically enhance the expression of osteoclast differentiation and extracellular space pathway genes to promote the differentiation of osteoclasts and promote the process of bone remodeling. In these processes, specific biomaterial properties can be used to guide cell behavior and regulate molecular network in the symbiotic microenvironment to reduce the barriers of regeneration and repair.
format Online
Article
Text
id pubmed-8922091
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-89220912022-03-21 Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing Ji, Ce Qiu, Minglong Ruan, Huitong Li, Cuidi Cheng, Liang Wang, Juan Li, Changwei Qi, Jin Cui, Wenguo Deng, Lianfu Adv Sci (Weinh) Research Articles Three dimension (3D) printed scaffolds have been shown to be superior in promoting tissue repair, but the cell‐level specific regulatory network activated by 3D printing scaffolds with different material components to form a symbiosis niche have not been systematically revealed. Here, three typical 3D printed scaffolds, including natural polymer hydrogel (gelatin‐methacryloyl, GelMA), synthetic polymer material (polycaprolactone, PCL), and bioceramic (β‐tricalcium phosphate, β‐TCP), are fabricated to explore the regulating effect of the symbiotic microenvironment during bone healing. Enrichment analysis show that hydrogel promotes tissue regeneration and reconstruction by improving blood vessel generation by enhancing oxygen transport and red blood cell development. The PCL scaffold regulates cell proliferation and differentiation by promoting cellular senescence, cell cycle and deoxyribonucleic acid (DNA) replication pathways, accelerating the process of endochondral ossification, and the formation of callus. The β‐TCP scaffold can specifically enhance the expression of osteoclast differentiation and extracellular space pathway genes to promote the differentiation of osteoclasts and promote the process of bone remodeling. In these processes, specific biomaterial properties can be used to guide cell behavior and regulate molecular network in the symbiotic microenvironment to reduce the barriers of regeneration and repair. John Wiley and Sons Inc. 2022-01-18 /pmc/articles/PMC8922091/ /pubmed/35040587 http://dx.doi.org/10.1002/advs.202105194 Text en © 2022 The Authors. Advanced Science published by Wiley‐VCH GmbH https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Articles
Ji, Ce
Qiu, Minglong
Ruan, Huitong
Li, Cuidi
Cheng, Liang
Wang, Juan
Li, Changwei
Qi, Jin
Cui, Wenguo
Deng, Lianfu
Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing
title Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing
title_full Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing
title_fullStr Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing
title_full_unstemmed Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing
title_short Transcriptome Analysis Revealed the Symbiosis Niche of 3D Scaffolds to Accelerate Bone Defect Healing
title_sort transcriptome analysis revealed the symbiosis niche of 3d scaffolds to accelerate bone defect healing
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8922091/
https://www.ncbi.nlm.nih.gov/pubmed/35040587
http://dx.doi.org/10.1002/advs.202105194
work_keys_str_mv AT jice transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT qiuminglong transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT ruanhuitong transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT licuidi transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT chengliang transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT wangjuan transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT lichangwei transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT qijin transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT cuiwenguo transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing
AT denglianfu transcriptomeanalysisrevealedthesymbiosisnicheof3dscaffoldstoacceleratebonedefecthealing