1
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
2
|
Li H, Kelley J, Ye Y, Ye ZW, Townsend DM, Zhang J, Wu Y. REDOX Imbalance and Oxidative Stress in the Intervertebral Disc: The Effect of Mechanical Stress and Cigarette Smoking on ER Stress and Mitochondrial Dysfunction. Cells 2025; 14:613. [PMID: 40277939 PMCID: PMC12025608 DOI: 10.3390/cells14080613] [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: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Low back pain is a widespread condition that significantly impacts quality of life, with intervertebral disc degeneration (IDD) being a major contributing factor. However, the underlying mechanisms of IDD remain poorly understood, necessitating further investigation. Environmental risk factors, such as mechanical stress and cigarette smoke, elevate reactive oxygen species levels from both endogenous and exogenous sources, leading to redox imbalance and oxidative stress. The endoplasmic reticulum (ER) and mitochondria, two key organelles responsible for protein folding and energy production, respectively, are particularly vulnerable to oxidative stress. Under oxidative stress conditions, ER stress and mitochondrial dysfunction occur, resulting in unfolded protein response activation, impaired biosynthetic processes, and disruptions in the tricarboxylic acid cycle and electron transport chain, ultimately compromising energy metabolism. Prolonged and excessive ER stress can further trigger apoptosis through ER-mitochondrial crosstalk. Given the unique microenvironment of the intervertebral disc (IVD)-characterized by hypoxia, glucose starvation, and region-specific cellular heterogeneity-the differential effects of environmental stressors on distinct IVD cell populations require further investigation. This review explores the potential mechanisms through which environmental risk factors alter IVD cell activities, contributing to IDD progression, and discusses future therapeutic strategies aimed at mitigating disc degeneration.
Collapse
Affiliation(s)
- Hui Li
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
| | - Joshua Kelley
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
| | - Yiqing Ye
- Department of Orthopaedics and Physical Medicine & Rehabilitation, Medical University of South Carolina, Charleston, SC 29425, USA
- Academic Magnet High School, North Charleston, SC 29405, USA
| | - Zhi-Wei Ye
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danyelle M. Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jie Zhang
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yongren Wu
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA; (H.L.); (J.K.)
- Department of Orthopaedics and Physical Medicine & Rehabilitation, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
3
|
Fukuoka S, Adachi N, Ouchi E, Ikemoto H, Okumo T, Ishikawa F, Onda H, Sunagawa M. Mechanoreceptor Piezo1 channel-mediated interleukin expression in conjunctival epithelial cells: Linking mechanical stress to ocular inflammation. Ocul Surf 2025; 36:56-68. [PMID: 39778715 DOI: 10.1016/j.jtos.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/27/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
PURPOSE Mechanical stress on the ocular surface, such as from eye-rubbing, has been reported to lead to inflammation and various ocular conditions. We hypothesized that the mechanosensitive Piezo1 channel in the conjunctival epithelium contributes to the inflammatory response at the ocular surface after receiving mechanical stimuli. METHODS Human conjunctival epithelial cells (HConjECs) were treated with Yoda1, a Piezo1-specific agonist, and various allergens to measure cytokine expression levels using qRT-PCR. Piezo1 activation-induced intracellular signaling pathways were also investigated by Western blot. Mechanical stretching experiments were conducted to simulate Piezo1 activation in HConjECs. Specificity of Piezo1 was confirmed by PIEZO1 knockdown and GsMTx4. In in vivo studies, using immunohistochemistry, rats were administered Yoda1 eye drops to examine the inflammatory response in the conjunctiva and Piezo1-induced signaling activation. RESULTS HConjECs expressed functional Piezo1 channel which was the dominant mechanoreceptor among putative channels and whose activation significantly increased IL-6 and IL-8 expression through the p38 MAPK-CREB pathway. Piezo1-induced [Ca2+]i elevation was crucial for the production of IL-6. The Yoda1-induced inflammatory responses were blocked by PIEZO1 knockdown. Mechanical stretching mimicked these effects, which were suppressed by GsMTx4. In vivo, Yoda1 administration led to increased phospho-p38 MAPK, phospho-CREB, and IL-6 in the rat conjunctival epithelium, with significant neutrophil infiltration. CONCLUSION Mechanical stress-induced Piezo1 channel activation in conjunctival epithelial cells can cause ocular inflammation by upregulating pro-inflammatory cytokines via the p38 MAPK-CREB pathway and promoting neutrophil infiltration. These findings suggest that mechanical stimuli on ocular surface tissues are significant risk factors for ocular inflammation.
Collapse
Affiliation(s)
- Seiya Fukuoka
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan; Department of Ophthalmology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Naoki Adachi
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan.
| | - Erika Ouchi
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Hideshi Ikemoto
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Takayuki Okumo
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Fumihiro Ishikawa
- Center for Biotechnology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Hidetoshi Onda
- Department of Ophthalmology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| | - Masataka Sunagawa
- Department of Physiology, Showa University Graduate School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo, 142-8555, Japan
| |
Collapse
|
4
|
Ao X, Li K, Chen Y, Lai W, Lian Z, Wei Z, Wang L, Zhang Z, Huang M. Inhibition of TrkB-BDNF positive feedback loop attenuates intervertebral disc degeneration and low back pain in a composite mouse model. Brain Behav Immun 2025; 128:37-53. [PMID: 40147506 DOI: 10.1016/j.bbi.2025.03.029] [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] [Received: 12/19/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) is a significant contributor to low back pain (LBP); however, the specific mechanisms involved remain unclear. Herein, a novel LBP mouse model was developed by integrating a bipedal standing model with a lumbar spine instability model (BS + LSI). This model effectively reproduced the behavioral characteristics of LBP and the pathological features of IVDD. Notably, a higher degree of degeneration and innervation in the endplates were observed in the BS + LSI mice. Transcriptome analysis revealed a significant upregulation of Ntrk2, the gene encoding TrkB, in the intervertebral discs of BS + LSI mice. Immunohistochemical staining further confirmed elevated expression of TrkB and its ligand BDNF in the endplates of these mice. Moreover, cyclic tensile strain (CTS) (20 %, 0.1 Hz, 24 h) upregulated TrkB expression and activated NF-κB signaling pathway to promote inflammatory responses in endplate chondrocytes. siBDNF transfection or treatment with the TrkB inhibitor ANA-12 effectively inhibited these pathological changes. Mechanistically, TrkB promoted BDNF expression by enhancing CREB phosphorylation, thereby establishing a TrkB-CREB-BDNF positive feedback loop. In vivo injection of ANA-12 significantly alleviated endplate inflammation and LBP-related behaviors in BS + LSI mice. Thus, an effective and replicable mouse model of LBP was established to identify TrkB as both the receptor for and an upstream regulator of BDNF, making it a crucial target for interventions to alleviate CEP inflammation and LBP.
Collapse
Affiliation(s)
- Xiang Ao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Kun Li
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| | - Yujie Chen
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| | - Weiyi Lai
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| | - Zhengnan Lian
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| | - Zhengnong Wei
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| | - Liang Wang
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Minjun Huang
- Department of Spine Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, PR China; Academy of Orthopaedics·Guangdong Province, Guangzhou, Guangdong, PR China.
| |
Collapse
|
5
|
Wang Y, Zhang W, Yang Y, Qin J, Wang R, Wang S, Fu W, Niu Q, Wang Y, Li C, Li H, Zhou Y, Liu M. Osteopontin deficiency promotes cartilaginous endplate degeneration by enhancing the NF-κB signaling to recruit macrophages and activate the NLRP3 inflammasome. Bone Res 2024; 12:53. [PMID: 39242551 PMCID: PMC11379908 DOI: 10.1038/s41413-024-00355-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 09/09/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is a major cause of discogenic pain, and is attributed to the dysfunction of nucleus pulposus, annulus fibrosus, and cartilaginous endplate (CEP). Osteopontin (OPN), a glycoprotein, is highly expressed in the CEP. However, little is known on how OPN regulates CEP homeostasis and degeneration, contributing to the pathogenesis of IDD. Here, we investigate the roles of OPN in CEP degeneration in a mouse IDD model induced by lumbar spine instability and its impact on the degeneration of endplate chondrocytes (EPCs) under pathological conditions. OPN is mainly expressed in the CEP and decreases with degeneration in mice and human patients with severe IDD. Conditional Spp1 knockout in EPCs of adult mice enhances age-related CEP degeneration and accelerates CEP remodeling during IDD. Mechanistically, OPN deficiency increases CCL2 and CCL5 production in EPCs to recruit macrophages and enhances the activation of NLRP3 inflammasome and NF-κB signaling by facilitating assembly of IRAK1-TRAF6 complex, deteriorating CEP degeneration in a spatiotemporal pattern. More importantly, pharmacological inhibition of the NF-κB/NLRP3 axis attenuates CEP degeneration in OPN-deficient IDD mice. Overall, this study highlights the importance of OPN in maintaining CEP and disc homeostasis, and proposes a promising therapeutic strategy for IDD by targeting the NF-κB/NLRP3 axis.
Collapse
Affiliation(s)
- Yanqiu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wanqian Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yi Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Jinghao Qin
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ruoyu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Wenjuan Fu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Qin Niu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yanxia Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hongli Li
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China.
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
6
|
Chen H, Zhou Q, Pu X, Wang N, Wang S, Feng Z, Wang B, Zhu Z, Qiu Y, Sun X. Association between vertebral endplate defects and patient-reported symptoms: an immunohistochemical study investigating the COX-2/PGE-2/EP-4 axis. Spine J 2024; 24:1407-1415. [PMID: 38631491 DOI: 10.1016/j.spinee.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/07/2024] [Accepted: 04/07/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND CONTEXT Vertebral endplate defects are often implicated in degenerative disc disorders, yet their connection to patient-reported symptoms remains unclear. COX-2 and PGE-2 are known for their roles in inflammation and pain, with EP-4 receptor involvement in pain signaling. Examining their expression in vertebral endplate tissues may provide insights into pathomechanism of low back pain. PURPOSE To investigate the association between endplate defects and patient-reported symptoms and to further clarify the role of the COX-2/PGE-2/EP-4 axis in the pathogenesis of chronic low back pain. STUDY DESIGN/SETTING Retrospective study. PATIENT SAMPLE A total of 71 patients who had undergone single-level L4/5 or L5/S1 modified laminectomy decompression preserving proximal upper laminae and transforaminal lumbar interbody fusion surgery were included in this study, including 18 patients diagnosed with lumbar disc herniation, 19 with lumbar disc herniation accompanied by degenerative lumbar spinal stenosis, and 34 with degenerative spondylolisthesis. OUTCOME MEASURES Demographic data, Pfirrmann grade, Modic changes, endplate defect score, visual analog scale (VAS) for back and leg pain, and Oswestry Disability Index (ODI) before surgery, 3-month and 6-month follow-up, and the percentage of immune-positive cells (COX-2, PGE-2, and EP-4) in endplate tissue sections. METHODS Patients were divided into defect and nondefect groups according to endplate morphology on lumbar MR. All intraoperative endplate specimens were immediately fixed in 10% formaldehyde, and then embedded in paraffin 3 days later for tissue sections. The outcome measures were compared between the defect group and nondefect group. Data were analyzed using independent t-tests and χ² tests. Pearson's rank correlation test was used to assess correlations between patient-reported symptoms and the percentage of immune-positive cells in the groups. Multivariable logistic regression models using the forward stepwise likelihood ratio method were used to identify the factors that were independently associated with endplate defects. RESULTS The age of Defect group was significantly higher than that of nondefect group (52.5±7.7 vs 57.2±9.1. p=.024). There were no significant differences in gender, diagnosis, BMI, comorbidities, or surgical level between the two groups. Modic changes (Type Ⅱ/Type Ⅲ) were more common in patients of Defect group than nondefect group (38.5% vs 11.1%, p<.001), and so was disc degeneration (Pfirrmann grade Ⅳ/Ⅴ) (69.2% vs 33.3%, p<.001). Defect group had significantly higher VAS-Back (6.5±2.0 vs 4.9±1.6, p<.001) and ODI scores (62.9±10.7 vs 45.2±14.8, p<.001) than nondefect group, while there was no significant differences between the two groups during the 3 and 6-month follow-up after surgery. Histologically, Defect group was characterized by upregulation of COX-2, PGE-2, and EP-4 in endplate tissue sections. Both in defect and nondefect groups, VAS-Back showed moderate positive correlations with the expressions of COX-2 (r=0.643; r=0.558, p both<.001), PGE-2 (r=0.611; r=0.640, p both<.001), and EP-4 (r=0.643; r=0.563, p both<.001). Multivariate regression analyses reveled that percentage of COX-2-positive cells was associated with endplate defects (OR=1.509, 95%CI [1.048-2.171], p=.027), as well as percentage of PGE-2-positive (OR=1.291, 95%CI [1.106-1.508], p=.001) and EP-4-positive cells (OR=1.284, 95%CI [1.048∼2.171], p=.003). CONCLUSIONS Patients with endplate defects had worse quality of life, more severe disc degeneration and Modic changes, and up-regulated COX-2/PGE-2/EP-4 axis expression in cartilage endplates in patients with defected endplates. Inflammatory factors may significantly contribute to the onset and progression of chronic low back pain in patients with endplate defects, consequently impacting patient-reported symptoms.
Collapse
Affiliation(s)
- Haojie Chen
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qingshuang Zhou
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Xiaojiang Pu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Nannan Wang
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, China
| | - Sinian Wang
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhenhua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Bin Wang
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xu Sun
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
7
|
Salman A, Khawaja AS, Baig K, Zehra U. Comparing contribution of bony and cartilaginous endplate changes to intervertebral disc degeneration. Pak J Med Sci 2024; 40:1516-1522. [PMID: 39092047 PMCID: PMC11255830 DOI: 10.12669/pjms.40.7.8762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/11/2024] [Accepted: 03/29/2024] [Indexed: 08/04/2024] Open
Abstract
Objective To compare the degenerative features of cartilaginous endplate with bony endplate in association with intervertebral degeneration in local population at radiographic, macroscopic and microscopic level in human motion segments. Methods This cross-sectional descriptive study examined 59 lumbar spine motion segments from adult male cadavers at the Department of Anatomy, University of Health Sciences, Lahore, between May and September, 2022. Radiographic assessment observed bony endplate (BEP) for the presence of sclerosis & osteophytes and degeneration scores from 1-8 were assigned. Macroscopic assessment was done to evaluate BEP, cartilaginous endplate (CEP) and IVD, and scores ranged from 1to 28 for BEP, 1-4 for CEP and 1-64 for IVD were assigned. Microscopic assessment revealed degeneration scores of CEP ranged from 1-42 and 1-30 for IVD. Segments with BEP defects were also identified on radiographs & macroscopy. Results Significant correlations were observed between the total degeneration scores of BEP with IVD and CEP scores (r=0.88 and r=0.909, respectively, p<0.001). Similarly, the total degeneration scores of the CEP is also significantly correlated with total IVD (r=0.86, p<0.001). Additionally, the samples with BEP defects were having higher IVD degeneration scores (p<0.001). Conclusion This study, for the first time identifies that there exists a critical association of bony and cartilaginous endplate with intervertebral disc degeneration individually in the same tissue sections using multi-dimension assessment methods. Degeneration in any of the components of VEP is consonantly associated with IVD degeneration. The BEP & CEP, though, they are unique structures but are interlinked with each other structurally and functionally.
Collapse
Affiliation(s)
- Amber Salman
- Amber Salman, MPhil. Department of Anatomy, University of Health Sciences, Lahore, Pakistan
| | - Asma Sajjad Khawaja
- Asma Khwaja, MBBS. Department of Anatomy, University of Health Sciences, Lahore, Pakistan
| | - Kashif Baig
- Kashif Biag, FCPS. Department of Pathology, Aziz Fatima Medical and Dental College, Faisalabad, Pakistan
| | - Uruj Zehra
- Uruj Zehra, PhD. Department of Anatomy, University of Health Sciences, Lahore, Pakistan
| |
Collapse
|
8
|
Lei L, Wen Z, Cao M, Zhang H, Ling SKK, Fu BSC, Qin L, Xu J, Yung PSH. The emerging role of Piezo1 in the musculoskeletal system and disease. Theranostics 2024; 14:3963-3983. [PMID: 38994033 PMCID: PMC11234281 DOI: 10.7150/thno.96959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/15/2024] [Indexed: 07/13/2024] Open
Abstract
Piezo1, a mechanosensitive ion channel, has emerged as a key player in translating mechanical stimuli into biological signaling. Its involvement extends beyond physiological and pathological processes such as lymphatic vessel development, axon growth, vascular development, immunoregulation, and blood pressure regulation. The musculoskeletal system, responsible for structural support, movement, and homeostasis, has recently attracted attention regarding the significance of Piezo1. This review aims to provide a comprehensive summary of the current research on Piezo1 in the musculoskeletal system, highlighting its impact on bone formation, myogenesis, chondrogenesis, intervertebral disc homeostasis, tendon matrix cross-linking, and physical activity. Additionally, we explore the potential of targeting Piezo1 as a therapeutic approach for musculoskeletal disorders, including osteoporosis, muscle atrophy, intervertebral disc degeneration, and osteoarthritis.
Collapse
Affiliation(s)
- Lei Lei
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zhenkang Wen
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mingde Cao
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Haozhi Zhang
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Samuel Ka-Kin Ling
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bruma Sai-Chuen Fu
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ling Qin
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Patrick Shu-Hang Yung
- Musculoskeletal Research Laboratory and Centre of Musculoskeletal Aging and Regeneration, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Yuan X, Zhao X, Wang W, Li C. Mechanosensing by Piezo1 and its implications in the kidney. Acta Physiol (Oxf) 2024; 240:e14152. [PMID: 38682304 DOI: 10.1111/apha.14152] [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: 09/21/2023] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
Piezo1 is an essential mechanosensitive transduction ion channel in mammals. Its unique structure makes it capable of converting mechanical cues into electrical and biological signals, modulating biological and (patho)physiological processes in a wide variety of cells. There is increasing evidence demonstrating that the piezo1 channel plays a vital role in renal physiology and disease conditions. This review summarizes the current evidence on the structure and properties of Piezo1, gating modulation, and pharmacological characteristics, with special focus on the distribution and (patho)physiological significance of Piezo1 in the kidney, which may provide insights into potential treatment targets for renal diseases involving this ion channel.
Collapse
Affiliation(s)
- Xi Yuan
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoduo Zhao
- Department of Pathology, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Weidong Wang
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chunling Li
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Institute of Hypertension, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
10
|
Xiang P, Luo ZP, Che YJ. Insights into the mechanical microenvironment within the cartilaginous endplate: An emerging role in maintaining disc homeostasis and normal function. Heliyon 2024; 10:e31162. [PMID: 38803964 PMCID: PMC11128916 DOI: 10.1016/j.heliyon.2024.e31162] [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: 12/26/2023] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Biomechanical factors are strongly linked with the emergence and development of intervertebral disc degeneration (IVDD). The intervertebral disc (IVD), as a unique enclosed biomechanical structure, exhibits distinct mechanical properties within its substructures. Damage to the mechanical performance of any substructure can disrupt the overall mechanical function of the IVD. Endplate degeneration serves as a significant precursor to IVDD. The endplate (EP) structure, especially the cartilaginous endplate (CEP), serves as a conduit for nutrient and metabolite transport in the IVD. It is inevitably influenced by its nutritional environment, mechanical loading, cytokines and extracellular components. Currently, reports on strategies targeting the CEP for the prevention and treatment of IVDD are scarce. This is due to two primary reasons: first, limited knowledge of the biomechanical microenvironment surrounding the degenerated CEP cells; and second, innovative biological treatment strategies, such as implanting active cells (disc or mesenchymal stem cells) or modulating natural cell activity through the addition of therapeutic factors or genes to treat IVDD often overlook a critical aspect-the restoration of the nutrient supply function and mechanical microenvironment of the endplate. Therefore, restoring the healthy structure of the CEP and maintaining a stable mechanical microenvironment within the EP are crucial for the prevention of IVDD and the repair of degenerated IVDs. We present a comprehensive literature review on the mechanical microenvironment characteristics of cartilage endplates and their associated mechanical signaling pathways. Our aim is to provide valuable insights into the development and implementation of strategies to prevent IVDD by delaying or reversing CEP degeneration.
Collapse
Affiliation(s)
- Pan Xiang
- Department of Orthopaedics, The First Affiliated Hospital of SooChow University, Suzhou, Jiangsu, 215000, PR China
| | - Zong-Ping Luo
- Department of Orthopaedics, The First Affiliated Hospital of SooChow University, Suzhou, Jiangsu, 215000, PR China
| | - Yan-Jun Che
- Orthopedics and Sports Medicine Center, The Affiliated Suzhou Hospital of Nanjing Medical University, 242 Guangji Road, Suzhou, Jiangsu, 215008, PR China
| |
Collapse
|
11
|
Liu C, Gao X, Lou J, Li H, Chen Y, Chen M, Zhang Y, Hu Z, Chang X, Luo M, Zhai Y, Li C. Aberrant mechanical loading induces annulus fibrosus cells apoptosis in intervertebral disc degeneration via mechanosensitive ion channel Piezo1. Arthritis Res Ther 2023; 25:117. [PMID: 37420255 PMCID: PMC10327399 DOI: 10.1186/s13075-023-03093-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 06/16/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) is closely associated with the structural damage in the annulus fibrosus (AF). Aberrant mechanical loading is an important inducement of annulus fibrosus cells (AFCs) apoptosis, which contributes to the AF structural damage and aggravates IVDD, but the underlying mechanism is still unclear. This study aims to investigate the mechanism of a mechanosensitive ion channel protein Piezo1 in aberrant mechanical loading-induced AFCs apoptosis and IVDD. METHODS Rats were subjected to lumbar instability surgery to induce the unbalanced dynamic and static forces to establish the lumbar instability model. MRI and histological staining were used to evaluate the IVDD degree. A cyclic mechanical stretch (CMS)-stimulated AFCs apoptosis model was established by a Flexcell system in vitro. Tunel staining, mitochondrial membrane potential (MMP) detection, and flow cytometry were used to evaluate the apoptosis level. The activation of Piezo1 was detected using western blot and calcium fluorescent probes. Chemical activator Yoda1, chemical inhibitor GSMTx4, and a lentiviral shRNA-Piezo1 system (Lv-Piezo1) were utilized to regulate the function of Piezo1. High-throughput RNA sequencing (RNA-seq) was used to explore the mechanism of Piezo1-induced AFCs apoptosis. The Calpain activity and the activation of Calpain2/Bax/Caspase3 axis were evaluated by the Calpain activity kit and western blot with the siRNA-mediated Calapin1 or Calpain2 knockdown. Intradiscal administration of Lv-Piezo1 was utilized to evaluate the therapeutic effect of Piezo1 silencing in IVDD rats. RESULTS Lumbar instability surgery promoted the expression of Piezo1 in AFCs and stimulated IVDD in rats 4 weeks after surgery. CMS elicited distinct apoptosis of AFCs, with enhanced Piezo1 activation. Yoda1 further promoted CMS-induced apoptosis of AFCs, while GSMTx4 and Lv-Piezo1 exhibited opposite effects. RNA-seq showed that knocking down Piezo1 inhibited the calcium signaling pathway. CMS enhanced Calpain activity and elevated the expression of BAX and cleaved-Caspase3. Calpain2, but not Calpain1 knockdown, inhibited the expression of BAX and cleaved-Caspase3 and alleviated AFCs apoptosis. Lv-Piezo1 significantly alleviated the progress of IVDD in rats after lumbar instability surgery. CONCLUSIONS Aberrant mechanical loading induces AFCs apoptosis to promote IVDD by activating Piezo1 and downstream Calpain2/BAX/Caspase3 pathway. Piezo1 is expected to be a potential therapeutic target in treating IVDD.
Collapse
Affiliation(s)
- Chenhao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
- Department of Orthopedics, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai, China
| | - Xiaoxin Gao
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Jinhui Lou
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Haiyin Li
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Yuxuan Chen
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
- Center of Traumatic Orthopedics, People's Liberation Army 990 Hospital, Xinyang, 464000, Henan, China
| | - Molong Chen
- Department of Orthopedics/Sports Medicine Center, The First Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
| | - Yuyao Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Zhilei Hu
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Xian Chang
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China
| | - Menglin Luo
- Clinical Laboratory, Qinghai Provincial People's Hospital, Xining, 810007, Qinghai, China
| | - Yu Zhai
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China.
| | - Changqing Li
- Department of Orthopedics, The Second Affiliated Hospital of Army Medical University (The Third Military Medical University), Chongqing, 400038, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400038, China.
| |
Collapse
|
12
|
Dienes B, Bazsó T, Szabó L, Csernoch L. The Role of the Piezo1 Mechanosensitive Channel in the Musculoskeletal System. Int J Mol Sci 2023; 24:ijms24076513. [PMID: 37047487 PMCID: PMC10095409 DOI: 10.3390/ijms24076513] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
Since the recent discovery of the mechanosensitive Piezo1 channels, many studies have addressed the role of the channel in various physiological or even pathological processes of different organs. Although the number of studies on their effects on the musculoskeletal system is constantly increasing, we are still far from a precise understanding. In this review, the knowledge available so far regarding the musculoskeletal system is summarized, reviewing the results achieved in the field of skeletal muscles, bones, joints and cartilage, tendons and ligaments, as well as intervertebral discs.
Collapse
|
13
|
Keaveny TM, Adams AL, Fischer H, Brara HS, Burch S, Guppy KH, Kopperdahl DL. Increased risks of vertebral fracture and reoperation in primary spinal fusion patients who test positive for osteoporosis by Biomechanical Computed Tomography analysis. Spine J 2023; 23:412-424. [PMID: 36372353 DOI: 10.1016/j.spinee.2022.10.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND CONTEXT While osteoporosis is a risk factor for adverse outcomes in spinal fusion patients, diagnosing osteoporosis reliably in this population has been challenging due to degenerative changes and spinal deformities. Addressing that challenge, biomechanical computed tomography analysis (BCT) is a CT-based diagnostic test for osteoporosis that measures both bone mineral density and bone strength (using finite element analysis) at the spine; CT scans taken for spinal evaluation or previous care can be repurposed for the analysis. PURPOSE Assess the effectiveness of BCT for preoperatively identifying spinal fusion patients with osteoporosis who are at high risk of reoperation or vertebral fracture. STUDY DESIGN Observational cohort study in a multi-center integrated managed care system using existing data from patient medical records and imaging archives. PATIENT SAMPLE We studied a randomly sampled subset of all adult patients who had any type of primary thoracic (T4 or below) or lumbar fusion between 2005 and 2018. For inclusion, patients with accessible study data needed a preop CT scan without intravenous contrast that contained images (before any instrumentation) of the upper instrumented vertebral level. OUTCOME MEASURES Reoperation for any reason (primary outcome) or a newly documented vertebral fracture (secondary outcome) occurring up to 5 years after the primary surgery. METHODS All study data were extracted using available coded information and CT scans from the medical records. BCT was performed at a centralized lab blinded to the clinical outcomes; patients could test positive for osteoporosis based on either low values of bone strength (vertebral strength ≤ 4,500 N women or 6,500 N men) and/or bone mineral density (vertebral trabecular bone mineral density ≤ 80 mg/cm3 both sexes). Cox proportional hazard ratios were adjusted by age, presence of obesity, and whether the fusion was long (four or more levels fused) or short (3 or fewer levels fused); Kaplan-Meier survival was compared by the log rank test. This project was funded by NIH (R44AR064613) and all physician co-authors and author 1 received salary support from their respective departments. Author 6 is employed by, and author 1 has equity in and consults for, the company that provides the BCT test; the other authors declare no conflicts of interest. RESULTS For the 469 patients analyzed (298 women, 171 men), median follow-up time was 44.4 months, 11.1% had a reoperation (median time 14.5 months), and 7.7% had a vertebral fracture (median time 2.0 months). Overall, 25.8% of patients tested positive for osteoporosis and no patients under age 50 tested positive. Compared to patients without osteoporosis, those testing positive were at almost five-fold higher risk for vertebral fracture (adjusted hazard ratio 4.7, 95% confidence interval = 2.2-9.7; p<.0001 Kaplan-Meier survival). Of those positive-testing patients, those who tested positive concurrently for low values of both bone strength and bone mineral density (12.6% of patients overall) were at almost four-fold higher risk for reoperation (3.7, 1.9-7.2; Kaplan-Meier survival p<.0001); the remaining positive-testing patients (those who tested positive for low values of either bone strength or bone mineral density but not both) were not at significantly higher risk for reoperation (1.6, 0.7-3.7) but were for vertebral fracture (4.3, 1.9-10.2). For both clinical outcomes, risk remained high for patients who underwent short or long fusion. CONCLUSION In a real-world clinical setting, BCT was effective in identifying primary spinal fusion patients aged 50 or older with osteoporosis who were at elevated risks of reoperation and vertebral fracture.
Collapse
Affiliation(s)
- Tony M Keaveny
- Departments of Mechanical Engineering and Bioengineering, University of California, Berkeley, CA, USA.
| | - Annette L Adams
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Heidi Fischer
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Harsimran S Brara
- Department of Neurosurgery, Southern California Permanente Medical Group, Los Angeles, CA, USA
| | - Shane Burch
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Kern H Guppy
- Department of Neurosurgery, Kaiser Permanente Medical Group, Sacramento, CA, USA
| | | |
Collapse
|
14
|
Savadipour A, Palmer D, Ely EV, Collins KH, Garcia-Castorena JM, Harissa Z, Kim YS, Oestrich A, Qu F, Rashidi N, Guilak F. The role of PIEZO ion channels in the musculoskeletal system. Am J Physiol Cell Physiol 2023; 324:C728-C740. [PMID: 36717101 PMCID: PMC10027092 DOI: 10.1152/ajpcell.00544.2022] [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: 12/06/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
PIEZO1 and PIEZO2 are mechanosensitive cation channels that are highly expressed in numerous tissues throughout the body and exhibit diverse, cell-specific functions in multiple organ systems. Within the musculoskeletal system, PIEZO1 functions to maintain muscle and bone mass, sense tendon stretch, and regulate senescence and apoptosis in response to mechanical stimuli within cartilage and the intervertebral disc. PIEZO2 is essential for transducing pain and touch sensations as well as proprioception in the nervous system, which can affect musculoskeletal health. PIEZO1 and PIEZO2 have been shown to act both independently as well as synergistically in different cell types. Conditions that alter PIEZO channel mechanosensitivity, such as inflammation or genetic mutations, can have drastic effects on these functions. For this reason, therapeutic approaches for PIEZO-related disease focus on altering PIEZO1 and/or PIEZO2 activity in a controlled manner, either through inhibition with small molecules, or through dietary control and supplementation to maintain a healthy cell membrane composition. Although many opportunities to better understand PIEZO1 and PIEZO2 remain, the studies summarized in this review highlight how crucial PIEZO channels are to musculoskeletal health and point to promising possible avenues for their modulation as a therapeutic target.
Collapse
Affiliation(s)
- Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Daniel Palmer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Erica V Ely
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Jaquelin M Garcia-Castorena
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| | - Yu Seon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Arin Oestrich
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Feini Qu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children - St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
15
|
Guo S, Wang C, Xiao C, Gu Q, Long L, Wang X, Xu H, Li S. Role of the mechanosensitive piezo1 channel in intervertebral disc degeneration. Clin Physiol Funct Imaging 2023; 43:59-70. [PMID: 36400723 DOI: 10.1111/cpf.12798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/23/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022]
Abstract
Intervertebral disc degeneration (IDD) is a multifactorial skeletal disease involving mechanical, genetic, systemic, and biological factors, and it is characterized by apoptosis of the nucleus pulposus cells and breakdown of the extracellular matrix (ECM), which will impair the structure and function of the intervertebral disc (IVD), and cause low back pain. Recently, the piezo1 is recognized as a critical mechanically activated ion channel of IDD. Numerous studies have reported that the piezo1 ion channel was aberrantly activated in the degenerated disc tissues and deeply participated in the pathogenesis of IDD. Inactivating or interfering with the piezo1 channel could effectively prevent the progression of IDD under the experimental conditions. It may be a promising target for the prevention and treatment of the disabling disease. Therefore, we have to make a comprehensive investigation and understanding of the mechanisms and functions of the piezo1 in the biomechanics of the spine. This study mainly elucidates the role of the piezo1 channel in IDD, which may facilitate the development of therapeutic targets for this disease.
Collapse
Affiliation(s)
- Sheng Guo
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chenglong Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Changming Xiao
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qinwen Gu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Longhai Long
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoqiang Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Houping Xu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Sen Li
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
16
|
Wu J, Chen Y, Liao Z, Liu H, Zhang S, Zhong D, Qiu X, Chen T, Su D, Ke X, Wan Y, Zhou T, Su P. Self-amplifying loop of NF-κB and periostin initiated by PIEZO1 accelerates mechano-induced senescence of nucleus pulposus cells and intervertebral disc degeneration. Mol Ther 2022; 30:3241-3256. [PMID: 35619555 PMCID: PMC9552911 DOI: 10.1016/j.ymthe.2022.05.021] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/05/2022] [Accepted: 05/21/2022] [Indexed: 11/21/2022] Open
Abstract
Abnormal mechanical load is a main risk factor of intervertebral disc degeneration (IDD), and cellular senescence is a pathological change in IDD. In addition, extracellular matrix (ECM) stiffness promotes human nucleus pulposus cells (hNPCs) senescence. However, the molecular mechanism underlying mechano-induced cellular senescence and IDD progression is not yet fully elucidated. First, we demonstrated that mechano-stress promoted hNPCs senescence via NF-κB signaling. Subsequently, we identified periostin as the main mechano-responsive molecule in hNPCs through unbiased sequencing, which was transcriptionally upregulated by NF-κB p65; moreover, secreted periostin by senescent hNPCs further promoted senescence and upregulated the catabolic process in hNPCs through activating NF-κB, forming a positive loop. Both Postn (encoding periostin) knockdown via siRNA and periostin inactivation via neutralizing antibodies alleviated IDD and NPCs senescence. Furthermore, we found that mechano-stress initiated the positive feedback of NF-κB and periostin via PIEZO1. PIEZO1 activation by Yoda1 induced severe IDD in rat tails without compression, and Postn knockdown alleviated the Yoda1-induced IDD in vivo. Here, we reported for the first time that self-amplifying loop of NF-κB and periostin initiated via PIEZO1 under mechano-stress accelerated NPCs senescence, leading to IDD. Furthermore, periostin neutralizing antibodies, which may serve as potential therapeutic agents for IDD, interrupted this loop.
Collapse
Affiliation(s)
- Jinna Wu
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Yuyu Chen
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Zhiheng Liao
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Hengyu Liu
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Shun Zhang
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Dongmei Zhong
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xianjian Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Taiqiu Chen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510120, China
| | - Deying Su
- Guangdong Provincial Key Laboratory of Proteomics and State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaona Ke
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Yong Wan
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China
| | - Taifeng Zhou
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China.
| | - Peiqiang Su
- Department of Spine Surgery, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, No.58 Zhongshan 2(nd) Road, Yuexiu District, Guangzhou 510080, China.
| |
Collapse
|
17
|
Li Y, Wu H, Li Z, Li B, Zhu M, Chen D, Ye F, Yu B, Huang Y. Species variation in the cartilaginous endplate of the lumbar intervertebral disc. JOR Spine 2022; 5:e1218. [PMID: 36203863 PMCID: PMC9520767 DOI: 10.1002/jsp2.1218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUNDS Cartilaginous endplate (CEP) plays an essential role in intervertebral disc (IVD) health and disease. The aim was to compare the CEP structure of lumbar IVD and to reveal the detailed pattern of integration between the CEP and bony endplate (BEP) from different species. METHODS A total of 34 IVDs (5 human, 5 goat, 8 pig, 8 rabbit, and 8 rat IVDs) were collected, fixed and midsagittally cut; in each IVD, one-half was used for histological staining to observe the CEP morphology, and the other half was used for scanning electron microscopy (SEM) analysis to measure the diameters and distributions of collagen fibers in the central and peripheral CEP areas and to observe the pattern of CEP-BEP integration from different species. RESULTS The human, pig, goat, and rabbit IVDs had the typical BEP-CEP structure, but the rat CEP was directly connected with the growth plate. Human CEP was the thickest (896.95 ± 87.71 μm) among these species, followed by pig, goat, rat, and rabbit CEPs. Additionally, the mean cellular density of the rabbit CEP was the highest, which was 930 ± 202 per mm2, followed by the rat, goat, pig, and human CEPs. In all the species, the collagen fiber diameter in the peripheral area was much bigger than that in the central area. The collagen fiber diameters of CEP from the human, pig, goat, and rat were distributed between 35 nm and 65 nm. The BEP and CEP were connected by the collagen from the CEP, aggregating into bundles or cross links with each other to form a network, and anchored to BEP. CONCLUSIONS Significant differences in the thickness, cellular density, and collagen characterization of CEPs from different species were demonstrated; the integration of BEP-CEP in humans, pigs, goats, and rabbits was mainly achieved by the collagen bundles anchoring system, while the typical BEP-CEP interface did not exist in rats.
Collapse
Affiliation(s)
- Yun‐He Li
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
| | - Hai‐Long Wu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
| | - Zhen Li
- AO Research Institute DavosDavosSwitzerland
| | - Bin‐Bin Li
- Department of Human Anatomy & HistoembryologyHangzhou Normal UniversityHangzhouChina
| | - Man Zhu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
| | - Di Chen
- Research Center for Computer‐aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Fei‐Hong Ye
- Hangzhou Zhigu Research Center for Tissue Engineering and Regenerative MedicineHangzhouChina
| | - Bin‐Sheng Yu
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
- Institute of Orthopaedics, Peking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| | - Yong‐Can Huang
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine SurgeryPeking University Shenzhen HospitalShenzhenChina
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, National & Local Joint Engineering Research Center of Orthopaedic BiomaterialsPeking University Shenzhen HospitalShenzhenChina
- Institute of Orthopaedics, Peking University Shenzhen HospitalShenzhen Peking University‐The Hong Kong University of Science and Technology Medical CenterShenzhenChina
| |
Collapse
|
18
|
Shi S, Kang XJ, Zhou Z, He ZM, Zheng S, He SS. Excessive mechanical stress-induced intervertebral disc degeneration is related to Piezo1 overexpression triggering the imbalance of autophagy/apoptosis in human nucleus pulpous. Arthritis Res Ther 2022; 24:119. [PMID: 35606793 PMCID: PMC9125856 DOI: 10.1186/s13075-022-02804-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Sheng Shi
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| | - Xing-Jian Kang
- School of Stomatology, Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Zhi Zhou
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| | - Zhi-Min He
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China
| | - Shuang Zheng
- School of Medicine, Shanghai University, Shanghai, 200444, People's Republic of China.
| | - Shi-Sheng He
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, People's Republic of China.
- Spinal Pain Research Institute, Tongji University School of Medicine, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
19
|
Ling Z, Liu Y, Wang Z, Zhang Z, Chen B, Yang J, Zeng B, Gao Y, Jiang C, Huang Y, Zou X, Wang X, Wei F. Single-Cell RNA-Seq Analysis Reveals Macrophage Involved in the Progression of Human Intervertebral Disc Degeneration. Front Cell Dev Biol 2022; 9:833420. [PMID: 35295968 PMCID: PMC8918513 DOI: 10.3389/fcell.2021.833420] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Intervertebral disc degeneration (IDD) has been considered as the primary pathological mechanism that underlies low back pain. Understanding the molecular mechanisms underlying human IDD is imperative for making strategies to treat IDD-related diseases. Herein, we report the molecular programs, lineage progression patterns, and paths of cellular communications during the progression of IDD using single-cell RNA sequencing (scRNA-seq) on nucleus pulposus (NP) cells from patients with different grades of IDD undergoing discectomy. New subtypes of cells and cell-type-specific gene signatures of the metabolic homeostatic NP cells (Met NPC), adhesive NP cells (Adh NPC), inflammatory response NP cells (IR NPC), endoplasmic reticulum stress NP cells (ERS NPC), fibrocartilaginous NP cells (Fc NPC), and CD70 and CD82+ progenitor NP cells (Pro NPC) were identified. In the late stage of IDD, the IR NPC and Fc NPC account for a large proportion of NPC. Importantly, immune cells including macrophages, T cells, myeloid progenitors, and neutrophils were also identified, and further analysis showed that significant intercellular interaction between macrophages and Pro NPC occurred via MIF (macrophage migration inhibitory factor) and NF-kB signaling pathways during the progression of IDD. In addition, dynamic polarization of macrophage M1 and M2 cell subtypes was found in the progression of IDD, and gene set functional enrichment analysis suggested a significant role of the macrophage polarization in regulating cell metabolism, especially the Pro NPC. Finally, we found that the NP cells in the late degenerative stage were mainly composed of the cell types related to inflammatory and endoplasmic reticulum (ER) response, and fibrocartilaginous activity. Our results provided new insights into the identification of NP cell populations at single-cell resolution and at the relatively whole-transcriptome scale, accompanied by cellular communications between immune cells and NP cells, and discriminative markers in relation to specific cell subsets. These new findings present clues for effective and functional manipulation of human IDD-related bioremediation and healthcare.
Collapse
Affiliation(s)
- Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Liu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bolin Chen
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiaming Yang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Baozhu Zeng
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yu Gao
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chang Jiang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yulin Huang
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiuhui Wang
- Department of Orthopaedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- *Correspondence: Fuxin Wei, ; Xiuhui Wang,
| | - Fuxin Wei
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
- *Correspondence: Fuxin Wei, ; Xiuhui Wang,
| |
Collapse
|
20
|
Liu H, Hu J, Zheng Q, Feng X, Zhan F, Wang X, Xu G, Hua F. Piezo1 Channels as Force Sensors in Mechanical Force-Related Chronic Inflammation. Front Immunol 2022; 13:816149. [PMID: 35154133 PMCID: PMC8826255 DOI: 10.3389/fimmu.2022.816149] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Mechanical damage is one of the predisposing factors of inflammation, and it runs through the entire inflammatory pathological process. Repeated or persistent damaging mechanical irritation leads to chronic inflammatory diseases. The mechanism of how mechanical forces induce inflammation is not fully understood. Piezo1 is a newly discovered mechanically sensitive ion channel. The Piezo1 channel opens in response to mechanical stimuli, transducing mechanical signals into an inflammatory cascade in the cell leading to tissue inflammation. A large amount of evidence shows that Piezo1 plays a vital role in the occurrence and progression of chronic inflammatory diseases. This mini-review briefly presents new evidence that Piezo1 responds to different mechanical stresses to trigger inflammation in various tissues. The discovery of Piezo1 provides new insights for the treatment of chronic inflammatory diseases related to mechanical stress. Inhibiting the transduction of damaging mechanical signals into inflammatory signals can inhibit inflammation and improve the outcome of inflammation at an early stage. The pharmacology of Piezo1 has shown bright prospects. The development of tissue-specific Piezo1 drugs for clinical use may be a new target for treating chronic inflammation.
Collapse
Affiliation(s)
- Hailin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaojin Feng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fenfang Zhan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xifeng Wang
- Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guohai Xu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Anesthesiology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|