Cargando…

Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells

Intervertebral disc (IVD) degeneration (IDD) is a widely recognized contributor to low back pain. Mechanical stress is a crucial etiological factor of IDD. During the process of IDD, a vicious circle is formed between abnormal mechanical stress and the damage of disc structure and function. Notably,...

Descripción completa

Detalles Bibliográficos
Autores principales: Feng, Chencheng, Yang, Minghui, Zhang, Yang, Lan, Minghong, Huang, Bo, Liu, Huan, Zhou, Yue
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5881642/
https://www.ncbi.nlm.nih.gov/pubmed/29512682
http://dx.doi.org/10.3892/ijmm.2018.3522
_version_ 1783311352706629632
author Feng, Chencheng
Yang, Minghui
Zhang, Yang
Lan, Minghong
Huang, Bo
Liu, Huan
Zhou, Yue
author_facet Feng, Chencheng
Yang, Minghui
Zhang, Yang
Lan, Minghong
Huang, Bo
Liu, Huan
Zhou, Yue
author_sort Feng, Chencheng
collection PubMed
description Intervertebral disc (IVD) degeneration (IDD) is a widely recognized contributor to low back pain. Mechanical stress is a crucial etiological factor of IDD. During the process of IDD, a vicious circle is formed between abnormal mechanical stress and the damage of disc structure and function. Notably, the pathological process of IDD is mediated by the phenotypic shift of IVD cells from an extracellular matrix anabolic phenotype to a catabolic and pro-inflammatory phenotype. Therefore, the effects of mechanical stress on the initiation and progression of IDD depend on the mechanobiology of IVD cells. Recently, disc cell senescence was identified as a new hallmark of IDD. However, the senescent response of disc cells to mechanical stress remains unknown. In this study, we found that prolonged exposure of cyclic mechanical tension (CMT) with unphysiological magnitude generated by the Flexercell tension system markedly induced premature senescence of nucleus pulposus (NP) cells. CMT augmented the DNA damage of NP cells, but did not affect the redox homeostasis of NP cells. Moreover, the p53-p21-retinoblastoma protein (Rb) pathway was activated by CMT to mediate the CMT-induced premature senescence of NP cells. The findings are beneficial to understanding the mechanism of disc cell senescence and the mechanobiology of disc cells further. It suggests that prolonged abnormal mechanical stress accelerates the establishment and progression of disc cell senescence and consequently impairs the structural and functional homeostasis of IVDs to cause IDD. Preventing the pro-senescent effect of mechanical stress on IVD cells is a promising approach to delay the process of IDD.
format Online
Article
Text
id pubmed-5881642
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-58816422018-04-12 Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells Feng, Chencheng Yang, Minghui Zhang, Yang Lan, Minghong Huang, Bo Liu, Huan Zhou, Yue Int J Mol Med Articles Intervertebral disc (IVD) degeneration (IDD) is a widely recognized contributor to low back pain. Mechanical stress is a crucial etiological factor of IDD. During the process of IDD, a vicious circle is formed between abnormal mechanical stress and the damage of disc structure and function. Notably, the pathological process of IDD is mediated by the phenotypic shift of IVD cells from an extracellular matrix anabolic phenotype to a catabolic and pro-inflammatory phenotype. Therefore, the effects of mechanical stress on the initiation and progression of IDD depend on the mechanobiology of IVD cells. Recently, disc cell senescence was identified as a new hallmark of IDD. However, the senescent response of disc cells to mechanical stress remains unknown. In this study, we found that prolonged exposure of cyclic mechanical tension (CMT) with unphysiological magnitude generated by the Flexercell tension system markedly induced premature senescence of nucleus pulposus (NP) cells. CMT augmented the DNA damage of NP cells, but did not affect the redox homeostasis of NP cells. Moreover, the p53-p21-retinoblastoma protein (Rb) pathway was activated by CMT to mediate the CMT-induced premature senescence of NP cells. The findings are beneficial to understanding the mechanism of disc cell senescence and the mechanobiology of disc cells further. It suggests that prolonged abnormal mechanical stress accelerates the establishment and progression of disc cell senescence and consequently impairs the structural and functional homeostasis of IVDs to cause IDD. Preventing the pro-senescent effect of mechanical stress on IVD cells is a promising approach to delay the process of IDD. D.A. Spandidos 2018-06 2018-02-28 /pmc/articles/PMC5881642/ /pubmed/29512682 http://dx.doi.org/10.3892/ijmm.2018.3522 Text en Copyright: © Feng 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
Feng, Chencheng
Yang, Minghui
Zhang, Yang
Lan, Minghong
Huang, Bo
Liu, Huan
Zhou, Yue
Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells
title Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells
title_full Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells
title_fullStr Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells
title_full_unstemmed Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells
title_short Cyclic mechanical tension reinforces DNA damage and activates the p53-p21-Rb pathway to induce premature senescence of nucleus pulposus cells
title_sort cyclic mechanical tension reinforces dna damage and activates the p53-p21-rb pathway to induce premature senescence of nucleus pulposus cells
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5881642/
https://www.ncbi.nlm.nih.gov/pubmed/29512682
http://dx.doi.org/10.3892/ijmm.2018.3522
work_keys_str_mv AT fengchencheng cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells
AT yangminghui cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells
AT zhangyang cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells
AT lanminghong cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells
AT huangbo cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells
AT liuhuan cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells
AT zhouyue cyclicmechanicaltensionreinforcesdnadamageandactivatesthep53p21rbpathwaytoinduceprematuresenescenceofnucleuspulposuscells