1
|
Xu H, Wei K, Ni J, Deng X, Wang Y, Xiang T, Song F, Wang Q, Niu Y, Jiang F, Wang J, Sheng L, Dai J. Matrix stiffness regulates nucleus pulposus cell glycolysis by MRTF-A-dependent mechanotransduction. Bone Res 2025; 13:23. [PMID: 39952914 PMCID: PMC11828926 DOI: 10.1038/s41413-025-00402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 02/17/2025] Open
Abstract
Increased matrix stiffness of nucleus pulposus (NP) tissue is a main feature of intervertebral disc degeneration (IVDD) and affects various functions of nucleus pulposus cells (NPCs). Glycolysis is the main energy source for NPC survival, but the effects and underlying mechanisms of increased extracellular matrix (ECM) stiffness on NPC glycolysis remain unknown. In this study, hydrogels with different stiffness were established to mimic the mechanical environment of NPCs. Notably, increased matrix stiffness in degenerated NP tissues from IVDD patients was accompanied with impaired glycolysis, and NPCs cultured on rigid substrates exhibited a reduction in glycolysis. Meanwhile, RNA sequencing analysis showed altered cytoskeleton-related gene expression in NPCs on rigid substrates. Myocardin-related transcription factor A (MRTF-A) is a transcriptional coactivator in mechanotransduction mainly responding to cytoskeleton remodeling, which was activated and translocated to the nucleus under rigid substrate and was upregulated during IVDD progression. Furthermore, gas chromatography-mass spectrometry (GC-MS) analysis revealed that MRTF-A overexpression reduced NPC glycolytic metabolite abundance and identified a correlation with AMPK pathway. Mechanistically, rigid substrates and MRTF-A overexpression inhibited Kidins220 expression and AMPK phosphorylation in NPCs, whereas MRTF-A inhibition, treated with the MRTF-A inhibitor CCG, partially rescued NP tissue degeneration and glycolytic enzyme expression. Our data demonstrate that MRTF-A is a critical regulator that responds to increased matrix stiffness in IVDD, and MRTF-A activation reduces NPC glycolysis by down-regulating Kidins220 and inhibiting AMPK phosphorylation.
Collapse
Affiliation(s)
- Haoran Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Wei
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinhao Ni
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuexing Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Taiyang Xiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fanglong Song
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qianliang Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanping Niu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fengxian Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Lei Sheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Jun Dai
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Su KK, Yu DC, Cao XF, Li P, Chang L, Yu XL, Li ZQ, Li M. Bone Marrow Mesenchymal Stem Cell-Derived Exosomes Alleviate Nuclear Pulposus Cells Degeneration Through the miR-145a-5p/USP31/HIF-1α Signaling Pathway. Stem Cell Rev Rep 2024; 20:2268-2282. [PMID: 39212824 DOI: 10.1007/s12015-024-10781-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Bone marrow mesenchymal stem cell (BMSC)-derived exosomes possess therapeutic potential against degenerative diseases. This study aimed to investigate the effects of BMSC-derived exosomes on intervertebral disc degeneration (IVDD) and explore the underlying molecular mechanisms. Through transcriptome sequencing and histological analysis, we observed a significant increase in HIF-1α expression in degenerative nucleus pulposus (NP) tissues. The addition of HIF-1α resulted in elevated expression of inflammatory factors IL-1β and IL-6, higher levels of matrix-degrading enzyme MMP13, and lower expression of aggrecan in NP cells. Co-culturing with BMSCs diminished the expression of HIF-1α, MMP13, IL-1β, and IL-6 in degenerative NP cells induced by overload pressure. miRNA chip analysis and PCR validation revealed that miR-145a-5p was the primary miRNA carried by BMSC-derived exosomes. Overexpression of miR-145a-5p was effective in minimizing the expression of HIF-1α, MMP13, IL-1β, and IL-6 in degenerative NP cells. Luciferase reporter assays confirmed USP31 as the target gene of miR-145a-5p, and the regulation of NP cells by BMSC-derived exosomes via miR-145a-5p was dependent on USP31. In conclusion, BMSC-derived exosomes alleviated IVDD through the miR-145a-5p/USP31/HIF-1α signaling pathway, providing valuable insights into the treatment of IVDD.
Collapse
Affiliation(s)
- Kang-Kang Su
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China
| | - De-Chen Yu
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China
| | - Xiong-Fei Cao
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China
| | - Pan Li
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China
| | - Le Chang
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China
| | - Xiao-Lei Yu
- Department of Cardiology, Air Force Medical University Tangdu Hospital, Xi'an710000, China
| | - Zhi-Quan Li
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China.
| | - Mo Li
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an710000, China.
| |
Collapse
|
3
|
Park J, Soh H, Jo S, Weon S, Lee SH, Park JA, Lee MK, Kim TH, Sung IH, Lee JK. Scaffold-induced compression enhances ligamentization potential of decellularized tendon graft reseeded with ACL-derived cells. iScience 2023; 26:108521. [PMID: 38162024 PMCID: PMC10755058 DOI: 10.1016/j.isci.2023.108521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Anterior cruciate ligament (ACL) reconstruction is often performed using a tendon graft. However, the predominant synthesis of fibrotic scar tissue (type III collagen) occurs during the healing process of the tendon graft, resulting in a significantly lower mechanical strength than that of normal ACL tissue. In this study, ACL-derived cells were reseeded to the tendon graft, and scaffold-induced compression was applied to test whether the compressive force results in superior cell survival and integration. Given nanofiber polycaprolactone (PCL) scaffold-induced compression, ACL-derived cells reseeded to a tendon graft demonstrated superior cell survival and integration and resulted in higher gene expression levels of type I collagen compared to non-compressed cell-allograft composites in vitro. Translocation of Yes-associated protein (YAP) into the nucleus was correlated with higher expression of type I collagen in the compression group. These data support the hypothesis of a potential role of mechanotransduction in the ligamentization process.
Collapse
Affiliation(s)
- Jinsung Park
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Hyunsoo Soh
- Department of Orthopaedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Subin Weon
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Seung Hoon Lee
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Jeong-Ah Park
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
| | - Myung-Kyu Lee
- Department of Research and Development, Korea Public Tissue Bank, Seongnam-si, Gyeonggi-do, Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Disease, Seoul, Republic of Korea
| | - Il-Hoon Sung
- Department of Orthopaedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| | - Jin Kyu Lee
- Hanyang University Institute for Rheumatology Research, Seoul, Republic of Korea
- Department of Orthopaedic Surgery, Hanyang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
4
|
Ke W, Wang B, Liao Z, Song Y, Li G, Ma L, Wang K, Li S, Hua W, Yang C. Matrix stiffness induces Drp1-mediated mitochondrial fission through Piezo1 mechanotransduction in human intervertebral disc degeneration. J Transl Med 2023; 21:711. [PMID: 37817199 PMCID: PMC10563269 DOI: 10.1186/s12967-023-04590-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/04/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Extracellular matrix stiffness is emerging as a crucial mechanical cue that drives the progression of various diseases, such as cancer, fibrosis, and inflammation. The matrix stiffness of the nucleus pulposus (NP) tissues increase gradually during intervertebral disc degeneration (IDD), while the mechanism through which NP cells sense and react to matrix stiffness remains unclear. In addition, mitochondrial dynamics play a key role in various cellular functions. An in-depth investigation of the pathogenesis of IDD can provide new insights for the development of effective therapies. In this study, we aim to investigate the effects of matrix stiffness on mitochondrial dynamics in IDD. METHODS To build the gradient stiffness model, NP cells were cultured on polystyrene plates with different stiffness. Western blot analysis, and immunofluorescence staining were used to detect the expression of mitochondrial dynamics-related proteins. Flow cytometry was used to detect the mitochondrial membrane potential and intracellular Ca2+ levels. Apoptosis related proteins, ROS level, and TUNEL staining were performed to assess the effect of substrate stiffness on NP cells. RESULTS Stiff substrate increased phosphorylation of dynamin-related protein 1 (Drp1) at Ser616 by activating extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, which promoted mitochondrial fission and apoptosis in NP cells. Furthermore, Piezo1 activation was involved in the regulation of the post-translational modifications of Drp1 and mitochondrial fission caused by matrix stiffness. Inhibition of Piezo1 and ERK1/2 can effectively reduce stiffness-induced ROS elevation and apoptosis in NP cells. CONCLUSIONS Our results revealed that stiff substrate causes Piezo1 activation and Ca2+ influx, results in ERK1/2 activation and phosphorylation of Drp1 at S616, and finally leads to mitochondrial fission and apoptosis in NP cells. These findings reveal a new mechanism of mechanotransduction in NP cells, providing novel insights into the development of therapies for treating IDD.
Collapse
Affiliation(s)
- Wencan Ke
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bingjin Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
5
|
Chen Z, Lv Z, Zhuang Y, Saiding Q, Yang W, Xiong W, Zhang Z, Chen H, Cui W, Zhang Y. Mechanical Signal-Tailored Hydrogel Microspheres Recruit and Train Stem Cells for Precise Differentiation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2300180. [PMID: 37230467 DOI: 10.1002/adma.202300180] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/31/2023] [Indexed: 05/27/2023]
Abstract
The aberrant mechanical microenvironment in degenerated tissues induces misdirection of cell fate, making it challenging to achieve efficient endogenous regeneration. Herein, a hydrogel microsphere-based synthetic niche with integrated cell recruitment and targeted cell differentiation properties via mechanotransduction is constructed . Through the incorporation of microfluidics and photo-polymerization strategies, fibronectin (Fn) modified methacrylated gelatin (GelMA) microspheres are prepared with the independently tunable elastic modulus (1-10Kpa) and ligand density (2 and 10 µg mL-1 ), allowing a wide range of cytoskeleton modulation to trigger the corresponding mechanobiological signaling. The combination of the soft matrix (2Kpa) and low ligand density (2 µg mL-1 ) can support the nucleus pulposus (NP)-like differentiation of intervertebral disc (IVD) progenitor/stem cells by translocating Yes-associated protein (YAP), without the addition of inducible biochemical factors. Meanwhile, platelet-derived growth factor-BB (PDGF-BB) is loaded onto Fn-GelMA microspheres (PDGF@Fn-GelMA) via the heparin-binding domain of Fn to initiate endogenous cell recruitment. In in vivo experiments, hydrogel microsphere-niche maintained the IVD structure and stimulated matrix synthesis. Overall, this synthetic niche with cell recruiting and mechanical training capabilities offered a promising strategy for endogenous tissue regeneration.
Collapse
Affiliation(s)
- Zehao Chen
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
- School of Mechatronic Engineering and Automation, Shanghai University, Nanchen Road 333, Shanghai, 200444, P. R. China
| | - Zhendong Lv
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wu Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wei Xiong
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Zhen Zhang
- School of Mechatronic Engineering and Automation, Shanghai University, Nanchen Road 333, Shanghai, 200444, P. R. China
| | - Hao Chen
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yuhui Zhang
- Department of Spine Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, P. R. China
| |
Collapse
|
6
|
Zhang GZ, Li L, Luo ZB, Zhang CY, Wang YG, Kang XW. Identification and experimental validation of key extracellular proteins as potential targets in intervertebral disc degeneration. Bone Joint Res 2023; 12:522-535. [PMID: 37661086 PMCID: PMC10475329 DOI: 10.1302/2046-3758.129.bjr-2022-0369.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Aims This study aimed, through bioinformatics analysis and in vitro experiment validation, to identify the key extracellular proteins of intervertebral disc degeneration (IDD). Methods The gene expression profile of GSE23130 was downloaded from the Gene Expression Omnibus (GEO) database. Extracellular protein-differentially expressed genes (EP-DEGs) were screened by protein annotation databases, and we used Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to analyze the functions and pathways of EP-DEGs. STRING and Cytoscape were used to construct protein-protein interaction (PPI) networks and identify hub EP-DEGs. NetworkAnalyst was used to analyze transcription factors (TFs) and microRNAs (miRNAs) that regulate hub EP-DEGs. A search of the Drug Signatures Database (DSigDB) for hub EP-DEGs revealed multiple drug molecules and drug-target interactions. Results A total of 56 EP-DEGs were identified in the differential expression analysis. EP-DEGs were enriched in the extracellular structure organization, ageing, collagen-activated signalling pathway, PI3K-Akt signalling pathway, and AGE-RAGE signalling pathway. PPI network analysis showed that the top ten hub EP-DEGs are closely related to IDD. Correlation analysis also demonstrated a significant correlation between the ten hub EP-DEGs (p<0.05), which were selected to construct TF-gene interaction and TF-miRNA coregulatory networks. In addition, ten candidate drugs were screened for the treatment of IDD. Conclusion The findings clarify the roles of extracellular proteins in IDD and highlight their potential as promising novel therapeutic targets.
Collapse
Affiliation(s)
- Guang-Zhi Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Lei Li
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Zhang-Bin Luo
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Cang-Yu Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Yong-Gang Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| | - Xue-Wen Kang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou, China
- The International Cooperation Base of Gansu Province for The Pain Research in Spinal Disorders, Lanzhou, China
| |
Collapse
|
7
|
Ke W, Liao Z, Liang H, Tong B, Song Y, Li G, Ma L, Wang K, Feng X, Li S, Hua W, Wang B, Yang C. Stiff Substrate Induces Nucleus Pulposus Cell Ferroptosis via YAP and N-Cadherin Mediated Mechanotransduction. Adv Healthc Mater 2023; 12:e2300458. [PMID: 37022980 DOI: 10.1002/adhm.202300458] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/26/2023] [Indexed: 04/07/2023]
Abstract
Increased tissue stiffness is associated with various pathological processes, such as fibrosis, inflammation, and aging. The matrix stiffness of the nucleus pulposus (NP) tissues increases gradually during intervertebral disc degeneration (IDD), while the mechanism through which NP cells sense and react to matrix stiffness remains unclear. In this study, the results indicate that ferroptosis is involved in stiff substrate-induced NP cell death. The expression of acyl-CoA synthetase long-chain family member 4 (ACSL4) increases in NP cells of the stiff group, which mediates lipid peroxidation and ferroptosis in NP cells. In addition, stiff substrate activates the hippo signaling cascade and induces the nuclear translocation of yes-associated protein (YAP). Interestingly, inhibition of YAP is efficient to reverse the increase of ACSL4 expression caused by matrix stiffness. Furthermore, stiff substrate suppresses the expression of N-cadherin in NP cells. N-cadherin overexpression can inhibit YAP nuclear translocation via the formation of the N-cadherin/β-catenin/YAP complex, and reverse matrix stiffness-induced ferroptosis in NP cells. Finally, the effects of YAP inhibition and N-cadherin overexpression on IDD progression are further illustrated in animal models. These findings reveal a new mechanism of mechanotransduction in NP cells, providing novel insights into the development of therapies for the treatment of IDD.
Collapse
Affiliation(s)
- Wencan Ke
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bide Tong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liang Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobo Feng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bingjin Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
8
|
Lazaro-Pacheco D, Mohseni M, Rudd S, Cooper-White J, Holsgrove TP. The role of biomechanical factors in models of intervertebral disc degeneration across multiple length scales. APL Bioeng 2023; 7:021501. [PMID: 37180733 PMCID: PMC10168717 DOI: 10.1063/5.0137698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/24/2023] [Indexed: 05/16/2023] Open
Abstract
Low back pain is the leading cause of disability, producing a substantial socio-economic burden on healthcare systems worldwide. Intervertebral disc (IVD) degeneration is a primary cause of lower back pain, and while regenerative therapies aimed at full functional recovery of the disc have been developed in recent years, no commercially available, approved devices or therapies for the regeneration of the IVD currently exist. In the development of these new approaches, numerous models for mechanical stimulation and preclinical assessment, including in vitro cell studies using microfluidics, ex vivo organ studies coupled with bioreactors and mechanical testing rigs, and in vivo testing in a variety of large and small animals, have emerged. These approaches have provided different capabilities, certainly improving the preclinical evaluation of these regenerative therapies, but challenges within the research environment, and compromises relating to non-representative mechanical stimulation and unrealistic test conditions, remain to be resolved. In this review, insights into the ideal characteristics of a disc model for the testing of IVD regenerative approaches are first assessed. Key learnings from in vivo, ex vivo, and in vitro IVD models under mechanical loading stimulation to date are presented alongside the merits and limitations of each model based on the physiological resemblance to the human IVD environment (biological and mechanical) as well as the possible feedback and output measurements for each approach. When moving from simplified in vitro models to ex vivo and in vivo approaches, the complexity increases resulting in less controllable models but providing a better representation of the physiological environment. Although cost, time, and ethical constraints are dependent on each approach, they escalate with the model complexity. These constraints are discussed and weighted as part of the characteristics of each model.
Collapse
Affiliation(s)
- Daniela Lazaro-Pacheco
- Department of Engineering, University of Exeter, Harrison Building, Streatham Campus, North Park Road, Exeter EX4 4QF, United Kingdom
| | - Mina Mohseni
- School of Chemical Engineering, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Samuel Rudd
- School of Chemical Engineering, The University of Queensland, St. Lucia QLD 4072, Australia
| | | | - Timothy Patrick Holsgrove
- Department of Engineering, University of Exeter, Harrison Building, Streatham Campus, North Park Road, Exeter EX4 4QF, United Kingdom
| |
Collapse
|
9
|
Silwal P, Nguyen-Thai AM, Mohammad HA, Wang Y, Robbins PD, Lee JY, Vo NV. Cellular Senescence in Intervertebral Disc Aging and Degeneration: Molecular Mechanisms and Potential Therapeutic Opportunities. Biomolecules 2023; 13:686. [PMID: 37189433 PMCID: PMC10135543 DOI: 10.3390/biom13040686] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Closely associated with aging and age-related disorders, cellular senescence (CS) is the inability of cells to proliferate due to accumulated unrepaired cellular damage and irreversible cell cycle arrest. Senescent cells are characterized by their senescence-associated secretory phenotype that overproduces inflammatory and catabolic factors that hamper normal tissue homeostasis. Chronic accumulation of senescent cells is thought to be associated with intervertebral disc degeneration (IDD) in an aging population. This IDD is one of the largest age-dependent chronic disorders, often associated with neurological dysfunctions such as, low back pain, radiculopathy, and myelopathy. Senescent cells (SnCs) increase in number in the aged, degenerated discs, and have a causative role in driving age-related IDD. This review summarizes current evidence supporting the role of CS on onset and progression of age-related IDD. The discussion includes molecular pathways involved in CS such as p53-p21CIP1, p16INK4a, NF-κB, and MAPK, and the potential therapeutic value of targeting these pathways. We propose several mechanisms of CS in IDD including mechanical stress, oxidative stress, genotoxic stress, nutritional deprivation, and inflammatory stress. There are still large knowledge gaps in disc CS research, an understanding of which will provide opportunities to develop therapeutic interventions to treat age-related IDD.
Collapse
Affiliation(s)
- Prashanta Silwal
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Allison M. Nguyen-Thai
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Haneef Ahamed Mohammad
- Department of Health Information Management, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Yanshan Wang
- Department of Health Information Management, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Paul D. Robbins
- Institute of the Biology of Aging and Metabolism and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joon Y. Lee
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nam V. Vo
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
10
|
Myo1b Promotes Premature Endothelial Senescence and Dysfunction via Suppressing Autophagy: Implications for Vascular Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:4654083. [PMID: 36654782 PMCID: PMC9842418 DOI: 10.1155/2023/4654083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Endothelial cell (EC) senescence characterized by an irreversible growth arrest leading to endothelial dysfunction has been implicated in vascular aging and aging-associated cardiovascular diseases. Autophagy plays a crucial role in the modulation of cellular senescence. Our previous showed that myosin 1b (Myo1b), one family of nonfilamentous class-1 myosin, was reported to be involved in the modulation of human smooth muscle cell senescence. However, the role of Myo1b in the modulation of EC senescence with links to autophagy has yet to be elucidated. In this study, we sought to explore the role of Myo1b in endothelial senescence and further elucidate the underlying mechanisms. Here, we show prominent upregulation of Myo1b in senescent ECs in comparison with nonsenescence ECs in both mRNA and protein expression levels. Silencing Myo1b in senescent cells ameliorates endothelial dysfunctions and reverses endothelial senescence phenotypic changes such as senescence-associated-β-galactosidase activity, cyclin-dependent kinase inhibitor p21WAF1, expression of vascular adhesion molecule-1 (VCAM1) and intercellular adhesion molecule-1 (ICAM1), and the senescence-associated cytokines. In contrast, in nonsenescent cells, overexpressing Myo1b promotes endothelial senescence and suppresses autophagy through the impairment of autophagosome and lysosome fusion. The interaction between Myo1b and LRRK2 through Myo1b tail domain promotes intracellular calcium elevation, which results in the inhibition of autophagic flux. In vitro and in vivo aging models, Myo1b knockdown in senescent ECs and wild type-aged mice is able to enhance autophagy and ameliorate aging-associated endothelial dysfunction. Taken together, our studies reveal a new function for Myo1b, that is, to couple LRRK2 assembly to promote an increase in intracellular calcium level, which impairs the autophagosome-lysosome fusion, and ultimately the promotion of EC senescence and vascular aging.
Collapse
|
11
|
Cheng F, Wang C, Ji Y, Yang B, Shu J, Shi K, Wang L, Wang S, Zhang Y, Huang X, Zhou X, Xia K, Liang C, Chen Q, Li F. Partial reprogramming strategy for intervertebral disc rejuvenation by activating energy switch. Aging Cell 2022; 21:e13577. [PMID: 35266272 PMCID: PMC9009234 DOI: 10.1111/acel.13577] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 01/08/2023] Open
Abstract
Rejuvenation of nucleus pulposus cells (NPCs) in degenerative discs can reverse intervertebral disc degeneration (IDD). Partial reprogramming is used to rejuvenate aging cells and ameliorate progression of aging tissue to avoiding formation of tumors by classical reprogramming. Understanding the effects and potential mechanisms of partial reprogramming in degenerative discs provides insights for development of new therapies for IDD treatment. The findings of the present study show that partial reprogramming through short‐term cyclic expression of Oct‐3/4, Sox2, Klf4, and c‐Myc (OSKM) inhibits progression of IDD, and significantly reduces senescence related phenotypes in aging NPCs. Mechanistically, short‐term induction of OSKM in aging NPCs activates energy metabolism as a “energy switch” by upregulating expression of Hexokinase 2 (HK2) ultimately promoting redistribution of cytoskeleton and restoring the aging state in aging NPCs. These findings indicate that partial reprogramming through short‐term induction of OSKM has high therapeutic potential in the treatment of IDD.
Collapse
Affiliation(s)
- Feng Cheng
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Chenggui Wang
- Department of Orthopedics The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University Wenzhou China
| | - Yufei Ji
- Department of Gastrointestinal Surgery Xiamen Cancer Center The First Affiliated Hospital of Xiamen University Xiamen China
| | - Biao Yang
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Jiawei Shu
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Kesi Shi
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Lulu Wang
- Laboratory of Metabolism and Cell Fate Guangzhou Institutes of Biomedicine and Health Chinese Academy of Sciences Guangzhou China
| | - Shaoke Wang
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Yuang Zhang
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Xianpeng Huang
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Xiaopeng Zhou
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Kaishun Xia
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Chengzhen Liang
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Qixin Chen
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| | - Fangcai Li
- Department of Orthopedics Surgery The Second Affiliated Hospital School of Medicine Zhejiang University Hangzhou China
- Orthopedics Research Institute of Zhejiang University Hangzhou China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province Hangzhou China
| |
Collapse
|
12
|
Tsukamoto S, Chiam KH, Asakawa T, Sawasaki K, Takesue N, Sakamoto N. Compressive forces driven by lateral actin fibers are a key to the nuclear deformation under uniaxial cell-substrate stretching. Biochem Biophys Res Commun 2022; 597:37-43. [PMID: 35123264 DOI: 10.1016/j.bbrc.2022.01.107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/10/2023]
Abstract
Cells sense the direction of mechanical stimuli including substrate stretching and show morphological and functional responses. The nuclear deformation with respect to the direction of mechanical stimuli is thought of as a vital factor in mechanosensitive intracellular signaling and gene transcription, but the detailed relationship between the direction of stimuli and nuclear deformation behavior is not fully solved yet. Here, we assessed the role of actin cytoskeletons in nuclear deformation caused by cell substrate stretching with different directions. Cells on a PDMS stretching chamber were subjected to a step-strain and changes of long- and short-axes of nucleus before and after stretching were evaluated in terms of nuclear orientation against the direction of stretching. Nuclei oriented parallel to the stretching direction showed elongation and shrinkage in the long and short axes, respectively, and vice versa. However, calculation of the aspect ratio (ratio of long- and short-axes) changes revealed orientation-depend nuclear deformation: The nucleus oriented parallel to the stretching direction showed a greater aspect ratio change than it aligned in the perpendicular direction of the stretching. A decrease in actin cytoskeletal tension significantly changed the nuclear deformation only in the short axis direction, thereby abolishing the orientation-depend deformation of the nucleus. These results suggest that lateral compressive forces exerted by the actin cytoskeleton is a key factor of orientation-depend deformation in short axis of the nucleus under the cell-substrate stretching condition, and may be crucial for mechano-sensing and responses to the cell-substrate stretching direction.
Collapse
Affiliation(s)
- Shingo Tsukamoto
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan; Bioinformatics Institute, A∗STAR, Singapore.
| | | | - Takumi Asakawa
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Kaoru Sawasaki
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Naoyuki Takesue
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Tokyo Metropolitan University, Hachioji, Tokyo, Japan.
| |
Collapse
|
13
|
Wang D, Peng P, Dudek M, Hu X, Xu X, Shang Q, Wang D, Jia H, Wang H, Gao B, Zheng C, Mao J, Gao C, He X, Cheng P, Wang H, Zheng J, Hoyland JA, Meng QJ, Luo Z, Yang L. Restoring the dampened expression of the core clock molecule BMAL1 protects against compression-induced intervertebral disc degeneration. Bone Res 2022; 10:20. [PMID: 35217644 PMCID: PMC8881495 DOI: 10.1038/s41413-022-00187-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/09/2022] Open
Abstract
The circadian clock participates in maintaining homeostasis in peripheral tissues, including intervertebral discs (IVDs). Abnormal mechanical loading is a known risk factor for intervertebral disc degeneration (IDD). Based on the rhythmic daily loading pattern of rest and activity, we hypothesized that abnormal mechanical loading could dampen the IVD clock, contributing to IDD. Here, we investigated the effects of abnormal loading on the IVD clock and aimed to inhibit compression-induced IDD by targeting the core clock molecule brain and muscle Arnt-like protein-1 (BMAL1). In this study, we showed that BMAL1 KO mice exhibit radiographic features similar to those of human IDD and that BMAL1 expression was negatively correlated with IDD severity by systematic analysis based on 149 human IVD samples. The intrinsic circadian clock in the IVD was dampened by excessive loading, and BMAL1 overexpression by lentivirus attenuated compression-induced IDD. Inhibition of the RhoA/ROCK pathway by Y-27632 or melatonin attenuated the compression-induced decrease in BMAL1 expression. Finally, the two drugs partially restored BMAL1 expression and alleviated IDD in a diurnal compression model. Our results first show that excessive loading dampens the circadian clock of nucleus pulposus tissues via the RhoA/ROCK pathway, the inhibition of which potentially protects against compression-induced IDD by preserving BMAL1 expression. These findings underline the importance of the circadian clock for IVD homeostasis and provide a potentially effective therapeutic strategy for IDD.
Collapse
Affiliation(s)
- Dong Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Pandi Peng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.,Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China
| | - Michal Dudek
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK.,Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, M13 9PL, UK
| | - Xueyu Hu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xiaolong Xu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Qiliang Shang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Di Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Haoruo Jia
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Han Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Bo Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chao Zheng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jianxin Mao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chu Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xin He
- Department of Medicine Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Pengzhen Cheng
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Huanbo Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jianmin Zheng
- Radiology Department, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Judith A Hoyland
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Qing-Jun Meng
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK.,Wellcome Centre for Cell Matrix Research, University of Manchester, Manchester, M13 9PL, UK
| | - Zhuojing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China. .,Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China.
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China. .,Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China.
| |
Collapse
|
14
|
Ke W, Wang B, Hua W, Song Y, Lu S, Luo R, Li G, Wang K, Liao Z, Xiang Q, Li S, Wu X, Zhang Y, Yang C. The distinct roles of myosin IIA and IIB under compression stress in nucleus pulposus cells. Cell Prolif 2021; 54:e12987. [PMID: 33415745 PMCID: PMC7848961 DOI: 10.1111/cpr.12987] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/08/2020] [Accepted: 12/23/2020] [Indexed: 12/24/2022] Open
Abstract
Objectives Inappropriate or excessive compression applied to intervertebral disc (IVD) contributes substantially to IVD degeneration. The actomyosin system plays a leading role in responding to mechanical stimuli. In the present study, we investigated the roles of myosin II isoforms in the compression stress‐induced senescence of nucleus pulposus (NP) cells. Material and methods Nucleus pulposus cells were exposed to 1.0 MPa compression for 0, 12, 24 or 36 hours. Immunofluorescence and co‐immunoprecipitation analysis were used to measure the interaction of myosin IIA and IIB with actin. Western blot analysis and immunofluorescence staining were used to detect nuclear expression and nuclear localization of MRTF‐A. In addition, the expression levels of p‐RhoA/RhoA, ROCK1/2 and p‐MLC/MLC were measured in human NP cells under compression stress and in degenerative IVD tissues. Results Compression stress increased the interaction of myosin IIA and actin, while the interaction of myosin IIB and actin was reduced. The actomyosin cytoskeleton remodelling was involved in the compression stress‐induced fibrotic phenotype mediated by MRTF‐A nuclear translocation and inhibition of proliferation in NP cells. Furthermore, RhoA/ROCK1 pathway activation mediated compression stress‐induced human NP cells senescence by regulating the interaction of myosin IIA and IIB with actin. Conclusions We for the first time investigated the regulation of actomyosin cytoskeleton in human NP cells under compression stress. It provided new insights into the development of therapy for effectively inhibiting IVD degeneration.
Collapse
Affiliation(s)
- Wencan Ke
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingjin Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbin Hua
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Saideng Lu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rongjin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiwei Liao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xiang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinghuo Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yukun Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|