Cargando…

Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous

BACKGROUND: Mechanical stress plays a crucial role in the pathogenesis of intervertebral disc degeneration (IVDD). The mechanosensitive Piezo1 ion channel can sense the changes in mechanical stress and convert the mechanical signals into chemical signals. This study aims to investigate the effect of...

Descripción completa

Detalles Bibliográficos
Autores principales: Shi, Sheng, Kang, Xing-Jian, Zhou, Zhi, He, Zhi-Min, Zheng, Shuang, He, Shi-Sheng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9125856/
https://www.ncbi.nlm.nih.gov/pubmed/35606793
http://dx.doi.org/10.1186/s13075-022-02804-y
_version_ 1784712019478315008
author Shi, Sheng
Kang, Xing-Jian
Zhou, Zhi
He, Zhi-Min
Zheng, Shuang
He, Shi-Sheng
author_facet Shi, Sheng
Kang, Xing-Jian
Zhou, Zhi
He, Zhi-Min
Zheng, Shuang
He, Shi-Sheng
author_sort Shi, Sheng
collection PubMed
description BACKGROUND: Mechanical stress plays a crucial role in the pathogenesis of intervertebral disc degeneration (IVDD). The mechanosensitive Piezo1 ion channel can sense the changes in mechanical stress and convert the mechanical signals into chemical signals. This study aims to investigate the effect of Piezo1 on the mechanical stress-induced IVDD and explore the possible mechanism. METHODS: The expression of Piezo1 and collagen II in immunohistochemical staining, cervical curvature, and the stiffness of nucleus pulpous (NP) were performed in normal and degenerated human intervertebral discs. In the experiment, high-intensity compression was applied to mimic the mechanical environment of IVDD. The cell viability, matrix macromolecules, and pro-inflammatory cytokines were examined to investigate the effect of Piezo1 on mechanical stress-treated NP cells. Additionally, autophagy condition of NP cells was detected within high-intensity compression and/or the inhibitor of Piezo1, GsMTx4. RESULTS: The up-expression of Piezo1, down-expression of Col II, elevated stiffness of NP, and poor kyphosis were observed in degenerated human intervertebral discs. High-intensity stress significantly decreased cell viability and the synthesis of extracellular matrix but increased the expression of senescence-associated proteins (p53 and p16) and pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) by mitochondrial dysfunction and suppression of autophagy. However, GsMTx4 can partly attenuate these effects. CONCLUSION: Piezo1 upregulation under excessive mechanical stress promotes the apoptosis, senescence, and pro-inflammatory cytokines of NP and leads to the loss of extracellular matrix by mitochondrial dysfunction and the suppression of autophagy; on the other hand, the inhibition of Piezo1 can partly alleviate these effects.
format Online
Article
Text
id pubmed-9125856
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-91258562022-05-24 Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous Shi, Sheng Kang, Xing-Jian Zhou, Zhi He, Zhi-Min Zheng, Shuang He, Shi-Sheng Arthritis Res Ther Research Article BACKGROUND: Mechanical stress plays a crucial role in the pathogenesis of intervertebral disc degeneration (IVDD). The mechanosensitive Piezo1 ion channel can sense the changes in mechanical stress and convert the mechanical signals into chemical signals. This study aims to investigate the effect of Piezo1 on the mechanical stress-induced IVDD and explore the possible mechanism. METHODS: The expression of Piezo1 and collagen II in immunohistochemical staining, cervical curvature, and the stiffness of nucleus pulpous (NP) were performed in normal and degenerated human intervertebral discs. In the experiment, high-intensity compression was applied to mimic the mechanical environment of IVDD. The cell viability, matrix macromolecules, and pro-inflammatory cytokines were examined to investigate the effect of Piezo1 on mechanical stress-treated NP cells. Additionally, autophagy condition of NP cells was detected within high-intensity compression and/or the inhibitor of Piezo1, GsMTx4. RESULTS: The up-expression of Piezo1, down-expression of Col II, elevated stiffness of NP, and poor kyphosis were observed in degenerated human intervertebral discs. High-intensity stress significantly decreased cell viability and the synthesis of extracellular matrix but increased the expression of senescence-associated proteins (p53 and p16) and pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β) by mitochondrial dysfunction and suppression of autophagy. However, GsMTx4 can partly attenuate these effects. CONCLUSION: Piezo1 upregulation under excessive mechanical stress promotes the apoptosis, senescence, and pro-inflammatory cytokines of NP and leads to the loss of extracellular matrix by mitochondrial dysfunction and the suppression of autophagy; on the other hand, the inhibition of Piezo1 can partly alleviate these effects. BioMed Central 2022-05-23 2022 /pmc/articles/PMC9125856/ /pubmed/35606793 http://dx.doi.org/10.1186/s13075-022-02804-y Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research Article
Shi, Sheng
Kang, Xing-Jian
Zhou, Zhi
He, Zhi-Min
Zheng, Shuang
He, Shi-Sheng
Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
title Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
title_full Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
title_fullStr Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
title_full_unstemmed Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
title_short Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
title_sort excessive mechanical stress-induced intervertebral disc degeneration is related to piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9125856/
https://www.ncbi.nlm.nih.gov/pubmed/35606793
http://dx.doi.org/10.1186/s13075-022-02804-y
work_keys_str_mv AT shisheng excessivemechanicalstressinducedintervertebraldiscdegenerationisrelatedtopiezo1overexpressiontriggeringtheimbalanceofautophagyapoptosisinhumannucleuspulpous
AT kangxingjian excessivemechanicalstressinducedintervertebraldiscdegenerationisrelatedtopiezo1overexpressiontriggeringtheimbalanceofautophagyapoptosisinhumannucleuspulpous
AT zhouzhi excessivemechanicalstressinducedintervertebraldiscdegenerationisrelatedtopiezo1overexpressiontriggeringtheimbalanceofautophagyapoptosisinhumannucleuspulpous
AT hezhimin excessivemechanicalstressinducedintervertebraldiscdegenerationisrelatedtopiezo1overexpressiontriggeringtheimbalanceofautophagyapoptosisinhumannucleuspulpous
AT zhengshuang excessivemechanicalstressinducedintervertebraldiscdegenerationisrelatedtopiezo1overexpressiontriggeringtheimbalanceofautophagyapoptosisinhumannucleuspulpous
AT heshisheng excessivemechanicalstressinducedintervertebraldiscdegenerationisrelatedtopiezo1overexpressiontriggeringtheimbalanceofautophagyapoptosisinhumannucleuspulpous