Cargando…

Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading

Knee osteoarthritis (KOA) is a degenerative disease resulting from mechanical overload, where direct physical impacts on chondrocytes play a crucial role in disease development by inducing inflammation and extracellular matrix degradation. However, the signaling cascades that sense these physical im...

Descripción completa

Detalles Bibliográficos
Autores principales: Cai, Xiaomin, Warburton, Christopher, Perez, Olivia F., Wang, Ying, Ho, Lucy, Finelli, Christina, Ehlen, Quinn T., Wu, Chenzhou, Rodriguez, Carlos D., Kaplan, Lee, Best, Thomas M., Huang, Chun-Yuh, Meng, Zhipeng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cold Spring Harbor Laboratory 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10473729/
https://www.ncbi.nlm.nih.gov/pubmed/37662374
http://dx.doi.org/10.1101/2023.06.09.544419
_version_ 1785100326464913408
author Cai, Xiaomin
Warburton, Christopher
Perez, Olivia F.
Wang, Ying
Ho, Lucy
Finelli, Christina
Ehlen, Quinn T.
Wu, Chenzhou
Rodriguez, Carlos D.
Kaplan, Lee
Best, Thomas M.
Huang, Chun-Yuh
Meng, Zhipeng
author_facet Cai, Xiaomin
Warburton, Christopher
Perez, Olivia F.
Wang, Ying
Ho, Lucy
Finelli, Christina
Ehlen, Quinn T.
Wu, Chenzhou
Rodriguez, Carlos D.
Kaplan, Lee
Best, Thomas M.
Huang, Chun-Yuh
Meng, Zhipeng
author_sort Cai, Xiaomin
collection PubMed
description Knee osteoarthritis (KOA) is a degenerative disease resulting from mechanical overload, where direct physical impacts on chondrocytes play a crucial role in disease development by inducing inflammation and extracellular matrix degradation. However, the signaling cascades that sense these physical impacts and induce the pathogenic transcriptional programs of KOA remain to be defined, which hinders the identification of novel therapeutic approaches. Recent studies have implicated a crucial role of Hippo signaling in osteoarthritis. Since Hippo signaling senses mechanical cues, we aimed to determine its role in chondrocyte responses to mechanical overload. Here we show that mechanical loading induces the expression of inflammatory and matrix-degrading genes by activating the nuclear factor-kappaB (NFκB) pathway in a Hippo-dependent manner. Applying mechanical compressional force to 3-dimensional cultured chondrocytes activated NFκB and induced the expression of NFκB target genes for inflammation and matrix degradation (i.e., IL1β and ADAMTS4). Interestingly, deleting the Hippo pathway effector YAP or activating YAP by deleting core Hippo kinases LATS1/2 blocked the NFκB pathway activation induced by mechanical loading. Consistently, treatment with a LATS1/2 kinase inhibitor abolished the upregulation of IL1β and ADAMTS4 caused by mechanical loading. Mechanistically, mechanical loading activates Protein Kinase C (PKC), which activates NFκB p65 by phosphorylating its Serine 536. Furthermore, the mechano-activation of both PKC and NFκB p65 is blocked in LATS1/2 or YAP knockout cells, indicating that the Hippo pathway is required by this mechanoregulation. Additionally, the mechanical loading-induced phosphorylation of NFκB p65 at Ser536 is blocked by the LATS1/2 inhibitor Lats-In-1 or the PKC inhibitor AEB-071. Our study suggests that the interplay of the Hippo signaling and PKC controls NFκB-mediated inflammation and matrix degradation in response to mechanical loading. Chemical inhibitors targeting Hippo signaling or PKC can prevent the mechanoresponses of chondrocytes associated with inflammation and matrix degradation, providing a novel therapeutic strategy for KOA.
format Online
Article
Text
id pubmed-10473729
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Cold Spring Harbor Laboratory
record_format MEDLINE/PubMed
spelling pubmed-104737292023-09-02 Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading Cai, Xiaomin Warburton, Christopher Perez, Olivia F. Wang, Ying Ho, Lucy Finelli, Christina Ehlen, Quinn T. Wu, Chenzhou Rodriguez, Carlos D. Kaplan, Lee Best, Thomas M. Huang, Chun-Yuh Meng, Zhipeng bioRxiv Article Knee osteoarthritis (KOA) is a degenerative disease resulting from mechanical overload, where direct physical impacts on chondrocytes play a crucial role in disease development by inducing inflammation and extracellular matrix degradation. However, the signaling cascades that sense these physical impacts and induce the pathogenic transcriptional programs of KOA remain to be defined, which hinders the identification of novel therapeutic approaches. Recent studies have implicated a crucial role of Hippo signaling in osteoarthritis. Since Hippo signaling senses mechanical cues, we aimed to determine its role in chondrocyte responses to mechanical overload. Here we show that mechanical loading induces the expression of inflammatory and matrix-degrading genes by activating the nuclear factor-kappaB (NFκB) pathway in a Hippo-dependent manner. Applying mechanical compressional force to 3-dimensional cultured chondrocytes activated NFκB and induced the expression of NFκB target genes for inflammation and matrix degradation (i.e., IL1β and ADAMTS4). Interestingly, deleting the Hippo pathway effector YAP or activating YAP by deleting core Hippo kinases LATS1/2 blocked the NFκB pathway activation induced by mechanical loading. Consistently, treatment with a LATS1/2 kinase inhibitor abolished the upregulation of IL1β and ADAMTS4 caused by mechanical loading. Mechanistically, mechanical loading activates Protein Kinase C (PKC), which activates NFκB p65 by phosphorylating its Serine 536. Furthermore, the mechano-activation of both PKC and NFκB p65 is blocked in LATS1/2 or YAP knockout cells, indicating that the Hippo pathway is required by this mechanoregulation. Additionally, the mechanical loading-induced phosphorylation of NFκB p65 at Ser536 is blocked by the LATS1/2 inhibitor Lats-In-1 or the PKC inhibitor AEB-071. Our study suggests that the interplay of the Hippo signaling and PKC controls NFκB-mediated inflammation and matrix degradation in response to mechanical loading. Chemical inhibitors targeting Hippo signaling or PKC can prevent the mechanoresponses of chondrocytes associated with inflammation and matrix degradation, providing a novel therapeutic strategy for KOA. Cold Spring Harbor Laboratory 2023-06-11 /pmc/articles/PMC10473729/ /pubmed/37662374 http://dx.doi.org/10.1101/2023.06.09.544419 Text en https://creativecommons.org/licenses/by-nc-nd/4.0/This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which allows reusers to copy and distribute the material in any medium or format in unadapted form only, for noncommercial purposes only, and only so long as attribution is given to the creator.
spellingShingle Article
Cai, Xiaomin
Warburton, Christopher
Perez, Olivia F.
Wang, Ying
Ho, Lucy
Finelli, Christina
Ehlen, Quinn T.
Wu, Chenzhou
Rodriguez, Carlos D.
Kaplan, Lee
Best, Thomas M.
Huang, Chun-Yuh
Meng, Zhipeng
Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading
title Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading
title_full Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading
title_fullStr Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading
title_full_unstemmed Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading
title_short Hippo Signaling Modulates the Inflammatory Response of Chondrocytes to Mechanical Compressive Loading
title_sort hippo signaling modulates the inflammatory response of chondrocytes to mechanical compressive loading
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10473729/
https://www.ncbi.nlm.nih.gov/pubmed/37662374
http://dx.doi.org/10.1101/2023.06.09.544419
work_keys_str_mv AT caixiaomin hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT warburtonchristopher hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT perezoliviaf hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT wangying hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT holucy hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT finellichristina hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT ehlenquinnt hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT wuchenzhou hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT rodriguezcarlosd hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT kaplanlee hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT bestthomasm hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT huangchunyuh hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading
AT mengzhipeng hipposignalingmodulatestheinflammatoryresponseofchondrocytestomechanicalcompressiveloading