Cargando…

Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy

Established studies proved that mechanical compression loading had multiple effects on the biological behavior of the intervertebral disc (IVD). However, the regulating mechanism involved in this process remains unclear. The current study is aimed at exploring the potential bioregulators and signali...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yiyang, Hu, Yanzhu, Wang, Haoming, Liu, Ningyuan, Luo, Lei, Zhao, Chen, Zhou, Dandan, Tong, Hang, Li, Pei, Zhou, Qiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8270705/
https://www.ncbi.nlm.nih.gov/pubmed/34306312
http://dx.doi.org/10.1155/2021/6192498
_version_ 1783720849342201856
author Wang, Yiyang
Hu, Yanzhu
Wang, Haoming
Liu, Ningyuan
Luo, Lei
Zhao, Chen
Zhou, Dandan
Tong, Hang
Li, Pei
Zhou, Qiang
author_facet Wang, Yiyang
Hu, Yanzhu
Wang, Haoming
Liu, Ningyuan
Luo, Lei
Zhao, Chen
Zhou, Dandan
Tong, Hang
Li, Pei
Zhou, Qiang
author_sort Wang, Yiyang
collection PubMed
description Established studies proved that mechanical compression loading had multiple effects on the biological behavior of the intervertebral disc (IVD). However, the regulating mechanism involved in this process remains unclear. The current study is aimed at exploring the potential bioregulators and signaling pathways involved in the compression-associated biological changes of nucleus pulposus (NP) cells. Tandem mass tag- (TMT-) based quantitative proteomics was exerted to analyze the differentially expressed proteins (DEPs) and signal pathways among the different groups of NP cells cultured under noncompression, low-compression (LC), and high-compression (HC) loading. Eight potential protective bioregulators for the NP cell survival under different compression loading were predicted by the proteomics, among which macrophage migration inhibitory factor (MIF) and oxidative stress-related pathways were selected for further evaluation, due to its similar function in regulating the fate of the cartilage endplate- (CEP-) derived cells. We found that deficiency of MIF accentuates the accumulation of ROS, mitochondrial dysfunction, and senescence of NP cells under overloaded mechanical compression. The potential molecular mechanism involved in this process is related to the mitophagy regulating role of MIF. Our findings provide a better understanding of the regulatory role of mechanical compression on the cellular fate commitment and matrix metabolism of NP, and the potential strategies for treating disc degenerative diseases via using MIF-regulating agents.
format Online
Article
Text
id pubmed-8270705
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-82707052021-07-22 Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy Wang, Yiyang Hu, Yanzhu Wang, Haoming Liu, Ningyuan Luo, Lei Zhao, Chen Zhou, Dandan Tong, Hang Li, Pei Zhou, Qiang Oxid Med Cell Longev Research Article Established studies proved that mechanical compression loading had multiple effects on the biological behavior of the intervertebral disc (IVD). However, the regulating mechanism involved in this process remains unclear. The current study is aimed at exploring the potential bioregulators and signaling pathways involved in the compression-associated biological changes of nucleus pulposus (NP) cells. Tandem mass tag- (TMT-) based quantitative proteomics was exerted to analyze the differentially expressed proteins (DEPs) and signal pathways among the different groups of NP cells cultured under noncompression, low-compression (LC), and high-compression (HC) loading. Eight potential protective bioregulators for the NP cell survival under different compression loading were predicted by the proteomics, among which macrophage migration inhibitory factor (MIF) and oxidative stress-related pathways were selected for further evaluation, due to its similar function in regulating the fate of the cartilage endplate- (CEP-) derived cells. We found that deficiency of MIF accentuates the accumulation of ROS, mitochondrial dysfunction, and senescence of NP cells under overloaded mechanical compression. The potential molecular mechanism involved in this process is related to the mitophagy regulating role of MIF. Our findings provide a better understanding of the regulatory role of mechanical compression on the cellular fate commitment and matrix metabolism of NP, and the potential strategies for treating disc degenerative diseases via using MIF-regulating agents. Hindawi 2021-07-01 /pmc/articles/PMC8270705/ /pubmed/34306312 http://dx.doi.org/10.1155/2021/6192498 Text en Copyright © 2021 Yiyang 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, Yiyang
Hu, Yanzhu
Wang, Haoming
Liu, Ningyuan
Luo, Lei
Zhao, Chen
Zhou, Dandan
Tong, Hang
Li, Pei
Zhou, Qiang
Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy
title Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy
title_full Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy
title_fullStr Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy
title_full_unstemmed Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy
title_short Deficiency of MIF Accentuates Overloaded Compression-Induced Nucleus Pulposus Cell Oxidative Damage via Depressing Mitophagy
title_sort deficiency of mif accentuates overloaded compression-induced nucleus pulposus cell oxidative damage via depressing mitophagy
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8270705/
https://www.ncbi.nlm.nih.gov/pubmed/34306312
http://dx.doi.org/10.1155/2021/6192498
work_keys_str_mv AT wangyiyang deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT huyanzhu deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT wanghaoming deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT liuningyuan deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT luolei deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT zhaochen deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT zhoudandan deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT tonghang deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT lipei deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy
AT zhouqiang deficiencyofmifaccentuatesoverloadedcompressioninducednucleuspulposuscelloxidativedamageviadepressingmitophagy