Cargando…

Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism

Osteoblasts have the capacity to perceive and transduce mechanical signals, and thus, regulate the mRNA and protein expression of a variety of genes associated with osteogenesis. Cytoskeletal reconstruction, as one of the earliest perception events for external mechanical stimulation, has previously...

Descripción completa

Detalles Bibliográficos
Autores principales: GAO, JIE, FU, SHANMIN, ZENG, ZHAOBIN, LI, FEIFEI, NIU, QIANNAN, JING, DA, FENG, XUE
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4918615/
https://www.ncbi.nlm.nih.gov/pubmed/27177232
http://dx.doi.org/10.3892/mmr.2016.5239
_version_ 1782439139726065664
author GAO, JIE
FU, SHANMIN
ZENG, ZHAOBIN
LI, FEIFEI
NIU, QIANNAN
JING, DA
FENG, XUE
author_facet GAO, JIE
FU, SHANMIN
ZENG, ZHAOBIN
LI, FEIFEI
NIU, QIANNAN
JING, DA
FENG, XUE
author_sort GAO, JIE
collection PubMed
description Osteoblasts have the capacity to perceive and transduce mechanical signals, and thus, regulate the mRNA and protein expression of a variety of genes associated with osteogenesis. Cytoskeletal reconstruction, as one of the earliest perception events for external mechanical stimulation, has previously been demonstrated to be essential for mechanotransduction in bone cells. However, the mechanism by which mechanical signals induce cytoskeletal deformation remains poorly understood. The actin-binding protein, cofilin, promotes the depolymerization of actin and is understood to be important in the regulation of activities in various cell types, including endothelial, neuronal and muscle cells. However, to the best of our knowledge, the importance of cofilin in osteoblastic mechanotransduction has not been previously investigated. In the present study, osteoblast-like MG-63 cells were subjected to physiological cyclic stretch stimulation (12% elongation) for 1, 4, 8, 12 and 24 h, and the expression levels of cofilin and osteogenesis-associated genes were quantified with reverse transcription-quantitative polymerase chain reaction, immunofluorescence staining and western blotting analyses. Additionally, knockdown of cofilin using RNA interference was conducted, and the mRNA levels of osteogenesis-associated genes were compared between osteoblast-like cells in the presence and absence of cofilin gene knockdown. The results of the present study demonstrated that cyclic stretch stimulates the expression of genes associated with osteoblastic activities in MG-63 cells, including alkaline phosphatase (ALP), osteocalcin (OCN), runt-related transcription factor 2 (Runx2) and collagen-1 (COL-1). Cyclic stretch also regulates the mRNA and protein expression of cofilin in MG-63 cells. Furthermore, stretch-induced increases in the levels of osteogenesis-associated genes, including ALP, OCN, Runx2 and COL-1, were reduced following cofilin gene knockdown. Together, these results demonstrate that cofilin is involved in the regulation of mechanical load-induced osteogenesis and, to the best of our knowledge, provides the first evidence demonstrating the importance of cofilin in osteoblastic mechanotransduction.
format Online
Article
Text
id pubmed-4918615
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-49186152016-07-11 Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism GAO, JIE FU, SHANMIN ZENG, ZHAOBIN LI, FEIFEI NIU, QIANNAN JING, DA FENG, XUE Mol Med Rep Articles Osteoblasts have the capacity to perceive and transduce mechanical signals, and thus, regulate the mRNA and protein expression of a variety of genes associated with osteogenesis. Cytoskeletal reconstruction, as one of the earliest perception events for external mechanical stimulation, has previously been demonstrated to be essential for mechanotransduction in bone cells. However, the mechanism by which mechanical signals induce cytoskeletal deformation remains poorly understood. The actin-binding protein, cofilin, promotes the depolymerization of actin and is understood to be important in the regulation of activities in various cell types, including endothelial, neuronal and muscle cells. However, to the best of our knowledge, the importance of cofilin in osteoblastic mechanotransduction has not been previously investigated. In the present study, osteoblast-like MG-63 cells were subjected to physiological cyclic stretch stimulation (12% elongation) for 1, 4, 8, 12 and 24 h, and the expression levels of cofilin and osteogenesis-associated genes were quantified with reverse transcription-quantitative polymerase chain reaction, immunofluorescence staining and western blotting analyses. Additionally, knockdown of cofilin using RNA interference was conducted, and the mRNA levels of osteogenesis-associated genes were compared between osteoblast-like cells in the presence and absence of cofilin gene knockdown. The results of the present study demonstrated that cyclic stretch stimulates the expression of genes associated with osteoblastic activities in MG-63 cells, including alkaline phosphatase (ALP), osteocalcin (OCN), runt-related transcription factor 2 (Runx2) and collagen-1 (COL-1). Cyclic stretch also regulates the mRNA and protein expression of cofilin in MG-63 cells. Furthermore, stretch-induced increases in the levels of osteogenesis-associated genes, including ALP, OCN, Runx2 and COL-1, were reduced following cofilin gene knockdown. Together, these results demonstrate that cofilin is involved in the regulation of mechanical load-induced osteogenesis and, to the best of our knowledge, provides the first evidence demonstrating the importance of cofilin in osteoblastic mechanotransduction. D.A. Spandidos 2016-07 2016-05-10 /pmc/articles/PMC4918615/ /pubmed/27177232 http://dx.doi.org/10.3892/mmr.2016.5239 Text en Copyright: © Gao 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
GAO, JIE
FU, SHANMIN
ZENG, ZHAOBIN
LI, FEIFEI
NIU, QIANNAN
JING, DA
FENG, XUE
Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
title Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
title_full Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
title_fullStr Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
title_full_unstemmed Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
title_short Cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
title_sort cyclic stretch promotes osteogenesis-related gene expression in osteoblast-like cells through a cofilin-associated mechanism
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4918615/
https://www.ncbi.nlm.nih.gov/pubmed/27177232
http://dx.doi.org/10.3892/mmr.2016.5239
work_keys_str_mv AT gaojie cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism
AT fushanmin cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism
AT zengzhaobin cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism
AT lifeifei cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism
AT niuqiannan cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism
AT jingda cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism
AT fengxue cyclicstretchpromotesosteogenesisrelatedgeneexpressioninosteoblastlikecellsthroughacofilinassociatedmechanism