Cargando…

Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway

BACKGROUND: Intervertebral disc degeneration (IVDD) is one of the major causes of low back pain and motor deficiency. Nucleus pulposus (NP) degeneration plays a key role in the process of IVDD. The mechanical and biological interactions involved in NP degeneration have not been elucidated. The prese...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Dandan, Chen, Yuanzhen, Cao, Shengnan, Ren, Pengcheng, Shi, Haojun, Li, Huazhong, Xie, Liangyu, Huang, Weimin, Shi, Bin, Han, Jinxiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7990548/
https://www.ncbi.nlm.nih.gov/pubmed/33815660
http://dx.doi.org/10.1155/2021/6699326
_version_ 1783669095190757376
author Wang, Dandan
Chen, Yuanzhen
Cao, Shengnan
Ren, Pengcheng
Shi, Haojun
Li, Huazhong
Xie, Liangyu
Huang, Weimin
Shi, Bin
Han, Jinxiang
author_facet Wang, Dandan
Chen, Yuanzhen
Cao, Shengnan
Ren, Pengcheng
Shi, Haojun
Li, Huazhong
Xie, Liangyu
Huang, Weimin
Shi, Bin
Han, Jinxiang
author_sort Wang, Dandan
collection PubMed
description BACKGROUND: Intervertebral disc degeneration (IVDD) is one of the major causes of low back pain and motor deficiency. Nucleus pulposus (NP) degeneration plays a key role in the process of IVDD. The mechanical and biological interactions involved in NP degeneration have not been elucidated. The present study is aimed at investigating the effect and mechanism of cyclic mechanical stretch in regulating the function and degeneration of NP cells. METHODS: NP cells were subjected to cyclic tensile stress (10% deformation) of 0.1 Hz for 8640 cycles. Cell proliferation was conducted through the MTT assay. The cell cycle and apoptosis were detected by flow cytometry. A gene expression profile chip was used to analyze the differentially expressed genes between the tensile stress group and the control group. Enrichment analysis of Gene Ontology (GO) annotation and signaling pathways were analyzed. Western blot and RNA interference were carried out to investigate the role of the ITGA2/PI3K/AKT pathway in the effect of cyclic mechanical stretch on NP cells. RESULTS: NP cells exhibited a greater (P < 0.05) growth rate in the tensile stress group compared to the control group. Cyclic mechanical stress significantly promoted the cell cycle transition of NP cells from the S phase to the G2/M phase. A fewer proportion of apoptotic cells were found in the tensile stress group (P < 0.05), indicating that cyclic mechanical stretch inhibits NP cell apoptosis. Microarray analysis revealed 689 significant differentially expressed genes between the two groups (P < 0.05), of which 333 genes were upregulated and another 356 genes were downregulated. Cyclic mechanical stretch altered the expression of 31 genes involved in the ITGA2/PI3K/AKT pathway and remarkably promoted this pathway in NP cells. Downregulation of ITGA2 and AKT further demonstrated that the PI3K/AKT pathway was responsible for the proliferation and COL2A1 expression of NP cells upon cyclic mechanical stretch. CONCLUSIONS: Cyclic mechanical stretch promoted the proliferation and cell cycle and reversely inhibited the apoptosis of NP cells. Cyclic mechanical stretch promoted COL2A1 expression and ameliorated the degeneration of NP cells via regulation of the ITGA2/PI3K/AKT signaling pathway. Our results may provide a potential target and a possibility of IVDD disease treatment by ameliorating the degenerative changes.
format Online
Article
Text
id pubmed-7990548
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-79905482021-04-01 Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway Wang, Dandan Chen, Yuanzhen Cao, Shengnan Ren, Pengcheng Shi, Haojun Li, Huazhong Xie, Liangyu Huang, Weimin Shi, Bin Han, Jinxiang Oxid Med Cell Longev Research Article BACKGROUND: Intervertebral disc degeneration (IVDD) is one of the major causes of low back pain and motor deficiency. Nucleus pulposus (NP) degeneration plays a key role in the process of IVDD. The mechanical and biological interactions involved in NP degeneration have not been elucidated. The present study is aimed at investigating the effect and mechanism of cyclic mechanical stretch in regulating the function and degeneration of NP cells. METHODS: NP cells were subjected to cyclic tensile stress (10% deformation) of 0.1 Hz for 8640 cycles. Cell proliferation was conducted through the MTT assay. The cell cycle and apoptosis were detected by flow cytometry. A gene expression profile chip was used to analyze the differentially expressed genes between the tensile stress group and the control group. Enrichment analysis of Gene Ontology (GO) annotation and signaling pathways were analyzed. Western blot and RNA interference were carried out to investigate the role of the ITGA2/PI3K/AKT pathway in the effect of cyclic mechanical stretch on NP cells. RESULTS: NP cells exhibited a greater (P < 0.05) growth rate in the tensile stress group compared to the control group. Cyclic mechanical stress significantly promoted the cell cycle transition of NP cells from the S phase to the G2/M phase. A fewer proportion of apoptotic cells were found in the tensile stress group (P < 0.05), indicating that cyclic mechanical stretch inhibits NP cell apoptosis. Microarray analysis revealed 689 significant differentially expressed genes between the two groups (P < 0.05), of which 333 genes were upregulated and another 356 genes were downregulated. Cyclic mechanical stretch altered the expression of 31 genes involved in the ITGA2/PI3K/AKT pathway and remarkably promoted this pathway in NP cells. Downregulation of ITGA2 and AKT further demonstrated that the PI3K/AKT pathway was responsible for the proliferation and COL2A1 expression of NP cells upon cyclic mechanical stretch. CONCLUSIONS: Cyclic mechanical stretch promoted the proliferation and cell cycle and reversely inhibited the apoptosis of NP cells. Cyclic mechanical stretch promoted COL2A1 expression and ameliorated the degeneration of NP cells via regulation of the ITGA2/PI3K/AKT signaling pathway. Our results may provide a potential target and a possibility of IVDD disease treatment by ameliorating the degenerative changes. Hindawi 2021-03-16 /pmc/articles/PMC7990548/ /pubmed/33815660 http://dx.doi.org/10.1155/2021/6699326 Text en Copyright © 2021 Dandan Wang et al. https://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Wang, Dandan
Chen, Yuanzhen
Cao, Shengnan
Ren, Pengcheng
Shi, Haojun
Li, Huazhong
Xie, Liangyu
Huang, Weimin
Shi, Bin
Han, Jinxiang
Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway
title Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway
title_full Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway
title_fullStr Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway
title_full_unstemmed Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway
title_short Cyclic Mechanical Stretch Ameliorates the Degeneration of Nucleus Pulposus Cells through Promoting the ITGA2/PI3K/AKT Signaling Pathway
title_sort cyclic mechanical stretch ameliorates the degeneration of nucleus pulposus cells through promoting the itga2/pi3k/akt signaling pathway
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7990548/
https://www.ncbi.nlm.nih.gov/pubmed/33815660
http://dx.doi.org/10.1155/2021/6699326
work_keys_str_mv AT wangdandan cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT chenyuanzhen cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT caoshengnan cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT renpengcheng cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT shihaojun cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT lihuazhong cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT xieliangyu cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT huangweimin cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT shibin cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway
AT hanjinxiang cyclicmechanicalstretchamelioratesthedegenerationofnucleuspulposuscellsthroughpromotingtheitga2pi3kaktsignalingpathway