Cargando…

Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression

Human gingival fibroblasts (HGFs) are responsible for connective tissue repair and scarring, and are exposed to mechanical forces under physiological and pathological conditions. The exact mechanisms underlying gingival tissue reconstruction under mechanical forces remain unclear. The present study...

Descripción completa

Detalles Bibliográficos
Autores principales: Nan, Lan, Zheng, Yi, Liao, Ni, Li, Songze, Wang, Yao, Chen, Zhixing, Wei, Liying, Zhao, Shuang, Mo, Shuixue
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6390077/
https://www.ncbi.nlm.nih.gov/pubmed/30664222
http://dx.doi.org/10.3892/mmr.2019.9882
_version_ 1783398067794345984
author Nan, Lan
Zheng, Yi
Liao, Ni
Li, Songze
Wang, Yao
Chen, Zhixing
Wei, Liying
Zhao, Shuang
Mo, Shuixue
author_facet Nan, Lan
Zheng, Yi
Liao, Ni
Li, Songze
Wang, Yao
Chen, Zhixing
Wei, Liying
Zhao, Shuang
Mo, Shuixue
author_sort Nan, Lan
collection PubMed
description Human gingival fibroblasts (HGFs) are responsible for connective tissue repair and scarring, and are exposed to mechanical forces under physiological and pathological conditions. The exact mechanisms underlying gingival tissue reconstruction under mechanical forces remain unclear. The present study aimfed to investigate the effects of mechanical forces on the proliferation and extracellular matrix synthesis in HGFs by establishing a 3-dimensional (3D) HGF culture model using poly(lactide-co-glycolide) (PLGA) scaffolds. HGFs were cultured in 3D PLGA scaffolds and a mechanical force of 0, 5, 15, 25 or 35 g/cm(2) was applied to HGFs for 24 h. A mechanical force of 25 g/cm(2) induced the highest proliferation rate, and thus was selected for subsequent experiments. Cell viability was determined using the MTT assay at 0, 24, 48 and 72 h. The expression levels of type I collagen (COL-1) and matrix metallopeptidase (MMP)-1 were examined by reverse transcription-quantitative polymerase chain reaction and ELISA, and transforming growth factor (TGF)-β expression was evaluated by ELISA. The application of mechanical force on HGFs cultured on the 3D PLGA scaffolds resulted in a significant increase in cell proliferation and COL-1 expression, as well as a decrease in MMP-1 expression. A TGF-β1 inhibitor was also applied, which attenuated the effects of mechanical force on HGF proliferation, and COL-1 and MMP-1 expression, thus suggesting that TGF-β signaling pathways may mediate the mechanical force-induced alterations observed in HGFs. In conclusion, these findings helped to clarify the mechanisms underlying mechanical force-induced HGF proliferation and ECM synthesis, which may promote the development of targeted therapeutics to treat various diseases, including gingival atrophy caused by orthodontic treatment.
format Online
Article
Text
id pubmed-6390077
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-63900772019-03-07 Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression Nan, Lan Zheng, Yi Liao, Ni Li, Songze Wang, Yao Chen, Zhixing Wei, Liying Zhao, Shuang Mo, Shuixue Mol Med Rep Articles Human gingival fibroblasts (HGFs) are responsible for connective tissue repair and scarring, and are exposed to mechanical forces under physiological and pathological conditions. The exact mechanisms underlying gingival tissue reconstruction under mechanical forces remain unclear. The present study aimfed to investigate the effects of mechanical forces on the proliferation and extracellular matrix synthesis in HGFs by establishing a 3-dimensional (3D) HGF culture model using poly(lactide-co-glycolide) (PLGA) scaffolds. HGFs were cultured in 3D PLGA scaffolds and a mechanical force of 0, 5, 15, 25 or 35 g/cm(2) was applied to HGFs for 24 h. A mechanical force of 25 g/cm(2) induced the highest proliferation rate, and thus was selected for subsequent experiments. Cell viability was determined using the MTT assay at 0, 24, 48 and 72 h. The expression levels of type I collagen (COL-1) and matrix metallopeptidase (MMP)-1 were examined by reverse transcription-quantitative polymerase chain reaction and ELISA, and transforming growth factor (TGF)-β expression was evaluated by ELISA. The application of mechanical force on HGFs cultured on the 3D PLGA scaffolds resulted in a significant increase in cell proliferation and COL-1 expression, as well as a decrease in MMP-1 expression. A TGF-β1 inhibitor was also applied, which attenuated the effects of mechanical force on HGF proliferation, and COL-1 and MMP-1 expression, thus suggesting that TGF-β signaling pathways may mediate the mechanical force-induced alterations observed in HGFs. In conclusion, these findings helped to clarify the mechanisms underlying mechanical force-induced HGF proliferation and ECM synthesis, which may promote the development of targeted therapeutics to treat various diseases, including gingival atrophy caused by orthodontic treatment. D.A. Spandidos 2019-03 2019-01-21 /pmc/articles/PMC6390077/ /pubmed/30664222 http://dx.doi.org/10.3892/mmr.2019.9882 Text en Copyright: © Nan et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Nan, Lan
Zheng, Yi
Liao, Ni
Li, Songze
Wang, Yao
Chen, Zhixing
Wei, Liying
Zhao, Shuang
Mo, Shuixue
Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression
title Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression
title_full Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression
title_fullStr Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression
title_full_unstemmed Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression
title_short Mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3D PLGA scaffolds via TGF-β expression
title_sort mechanical force promotes the proliferation and extracellular matrix synthesis of human gingival fibroblasts cultured on 3d plga scaffolds via tgf-β expression
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6390077/
https://www.ncbi.nlm.nih.gov/pubmed/30664222
http://dx.doi.org/10.3892/mmr.2019.9882
work_keys_str_mv AT nanlan mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT zhengyi mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT liaoni mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT lisongze mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT wangyao mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT chenzhixing mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT weiliying mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT zhaoshuang mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression
AT moshuixue mechanicalforcepromotestheproliferationandextracellularmatrixsynthesisofhumangingivalfibroblastsculturedon3dplgascaffoldsviatgfbexpression