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
|
Paik S, Kim JK, Shin HJ, Park EJ, Kim IS, Jo EK. Updated insights into the molecular networks for NLRP3 inflammasome activation. Cell Mol Immunol 2025:10.1038/s41423-025-01284-9. [PMID: 40307577 DOI: 10.1038/s41423-025-01284-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Over the past decade, significant advances have been made in our understanding of how NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasomes are activated. These findings provide detailed insights into the transcriptional and posttranslational regulatory processes, the structural-functional relationship of the activation processes, and the spatiotemporal dynamics of NLRP3 activation. Notably, the multifaceted mechanisms underlying the licensing of NLRP3 inflammasome activation constitute a focal point of intense research. Extensive research has revealed the interactions of NLRP3 and its inflammasome components with partner molecules in terms of positive and negative regulation. In this Review, we provide the current understanding of the complex molecular networks that play pivotal roles in regulating NLRP3 inflammasome priming, licensing and assembly. In addition, we highlight the intricate and interconnected mechanisms involved in the activation of the NLRP3 inflammasome and the associated regulatory pathways. Furthermore, we discuss recent advances in the development of therapeutic strategies targeting the NLRP3 inflammasome to identify potential therapeutics for NLRP3-associated inflammatory diseases. As research continues to uncover the intricacies of the molecular networks governing NLRP3 activation, novel approaches for therapeutic interventions against NLRP3-related pathologies are emerging.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Biochemistry and Cell Biology, Eulji University School of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea.
| |
Collapse
|
3
|
Ye B, Xu D, Zhong L, Wang Y, Wang W, Xu H, Han X, Min J, Wu G, Huang W, Liang G. Ubiquitin-specific protease 25 improves myocardial ischemia-reperfusion injury by deubiquitinating NLRP3 and negatively regulating NLRP3 inflammasome activity in cardiomyocytes. Clin Transl Med 2025; 15:e70243. [PMID: 39985261 PMCID: PMC11845855 DOI: 10.1002/ctm2.70243] [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: 08/15/2024] [Revised: 12/21/2024] [Accepted: 02/12/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Myocardial ischemia/reperfusion injury (MI/RI) restricts the effect of myocardial reperfusion therapy and lacks effective prevention and treatment methods. Deubiquitinating enzymes (DUBs), especially members of the ubiquitin-specific protease (USP) family of DUBs, are key proteins in the ubiquitination modification process and play a vital role in MI/RI. Therefore, we aimed to investigate the role of USP25, as a member of the USP family, in MI/RI and its molecular mechanism. METHODS Transcriptome sequencing was applied to evaluate the differential expression of USP families during hypoxia/reoxygenation (H/R) and validated in human and mouse heart samples and cardiomyocytes by performing quantitative polymerase chain reaction. Wild-type or USP25-/- mice were used to develop the MI/RI model. Co-immunoprecipitation (Co-IP) combined with liquid chromatography-tandem mass spectrometry analysis was used to screen the potential substrate protein of USP25 in H/R-induced cardiomyocyte injury. TUNEL and Hoechst/propidium iodide staining and western blot were used to detect the level of pyroptosis. In addition, cardiomyocyte-specific USP25 overexpression in NLRP3-/- mice with AAV9 vectors was used to validate the biological function of USP25 and NLRP3 interaction. RESULTS We found that the expression level of USP25 was significantly decreased in I/R-induced mouse heart tissues and primary cardiomyocytes in a time-dependent manner. USP25 deficiency exacerbated MI/RI and aggravated I/R-induced cardiac remodelling in mice. Mechanistically, USP25 directly binds to NLRP3 protein and K63-linkedly deubiquitinates NLRP3 at residue K243 via its active site C178, thus hindering NLRP3-ASC interaction and ASC oligomerization to inhibit NLRP3 activation and pyroptosis in cardiomyocytes. We further showed that the overexpression of USP25 in cardiomyocytes ameliorated MI/RI in mice, whereas this protective effect disappeared when NLRP3 is knocked out. CONCLUSIONS Our study demonstrated that USP25 ameliorates MI/RI by regulating NLRP3 activation and its mediated pyroptosis. This finding extends the protective role of USP25 in cardiovascular disease and provides an experimental basis for future USP25-based drug development for the treatment of MI/RI. KEY POINTS The deubiquitinating enzyme USP25 was down-regulated both in myocardial ischemia/reperfusion injury (MI/RI) myocardium tissues. The deficiency of USP25 worsened exacerbated MI/RI in mice, whereas the overexpression of USP25 in cardiomyocytes mitigated this pathological phenotype. USP25 directly interacts with the NLRP3 protein and deubiquitinates it via K63 linkage at residue K243 through its active site C178, thus affecting NLRP3-ASC interaction and ASC oligomerization to inhibit NLRP3 activation and pyroptosis in cardiomyocytes.
Collapse
Affiliation(s)
- Bozhi Ye
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiangChina
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Diyun Xu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
- Chemical Biology Research Center, School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| | - Lingfeng Zhong
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Yi Wang
- School of Pharmaceutical SciencesHangzhou Normal UniversityHangzhouZhejiangChina
| | - Wei Wang
- Affiliated Yongkang First People's HospitalHangzhou Medical CollegeYongkangZhejiangChina
| | - Haowen Xu
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiangChina
| | - Xue Han
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiangChina
| | - Julian Min
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiangChina
| | - Gaojun Wu
- Department of Cardiology and the Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated HospitalWenzhou Medical UniversityWenzhouZhejiangChina
| | - Wenhai Huang
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiangChina
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- School of Pharmaceutical SciencesHangzhou Medical CollegeHangzhouZhejiangChina
- Chemical Biology Research Center, School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
4
|
Liao Y, Kong Y, Chen H, Xia J, Zhao J, Zhou Y. Unraveling the priming phase of NLRP3 inflammasome activation: Molecular insights and clinical relevance. Int Immunopharmacol 2025; 146:113821. [PMID: 39674000 DOI: 10.1016/j.intimp.2024.113821] [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: 09/13/2024] [Revised: 11/10/2024] [Accepted: 12/05/2024] [Indexed: 12/16/2024]
Abstract
The NLRP3 inflammasome plays a pivotal role in the innate immune response. Its activation involves a two-step mechanism that consists of priming and activation. The priming of the NLRP3 inflammasome is a vital initial phase necessary for its activation and subsequent involvement in the immune response, though its understanding varies across studies. Recent research has identified key proteins that influence the priming process, revealing a sophisticated regulatory network. This review provides a comprehensive review of the priming phase of NLRP3 inflammasome activation, with a particular focus on the underlying molecular mechanisms, including transcriptional regulation, orchestration of the phosphorylation status, deubiquitination and the relationships with the inflammation-associated diseases. Understanding the intricacies of NLRP3 inflammasome priming not only elucidates fundamental aspects of immune regulation, but also provides potential avenues for therapeutic intervention in inflammatory diseases.
Collapse
Affiliation(s)
- Yonghong Liao
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China; National Center of Technology Innovation for Pigs, 402460, Rongchang, Chongqing, China
| | - Yueyao Kong
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China
| | - Hongyu Chen
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China
| | - Jing Xia
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China
| | - Jianjun Zhao
- College of Animal Science and Technology, Southwest University, 402460 Chongqing, China
| | - Yang Zhou
- College of Veterinary Medicine, Southwest University, 402460 Chongqing, China; National Center of Technology Innovation for Pigs, 402460, Rongchang, Chongqing, China.
| |
Collapse
|
5
|
Sang P, Li X, Wang Z. Bone Mesenchymal Stem Cells Inhibit Oxidative Stress-Induced Pyroptosis in Annulus Fibrosus Cells to Alleviate Intervertebral Disc Degeneration Based on Matric Hydrogels. Appl Biochem Biotechnol 2024; 196:8043-8057. [PMID: 38676833 DOI: 10.1007/s12010-024-04953-z] [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: 04/16/2024] [Indexed: 04/29/2024]
Abstract
Intervertebral disc degeneration (IVDD) is the primary cause of low back pain. Stem cell transplantation may be a possible approach to promote IVDD. This study was aimed to investigate the role of bone mesenchymal stem cells (BMSCs) in IVDD and the molecular mechanism. Annulus fibrosus cells (AFCs) were treated with tert-butyl hydroperoxide (TBHP) to induce oxidative stress injury. AFC biological functions were analyzed using a lactate dehydrogenase kit, enzyme-linked immunosorbent assay, flow cytometry, and western blot. The molecular mechanisms of BMSC functions were assessed using quantitative real-time PCR, western blot, immunoprecipitation (IP), co-IP, GST pull-down, and cycloheximide treatment. Furthermore, the impacts of BMSCs in IVDD progression in vivo were evaluated by magnetic resonance imaging (MRI) and H&E analysis. BMSCs inhibited TBHP-induced inflammation and pyroptosis in AFCs. Knockdown of SIRT1 reversed the effects on inflammation and pyroptosis of BMSCs. Moreover, SIRT1 promoted the deacetylation of ASC rather than NLRP3. SIRT1 interacted with ASC to reduce its protein stability, thereby negatively regulating ASC protein levels. In addition, BMSCs alleviated LPS-induced IVDD based on matrix hydrogels. BMSCs inhibited oxidative stress-induced pyroptosis and inflammation in AFCs, thereby alleviating IVDD, suggesting that BMSCs may contribute to treating intervertebral disc generation.
Collapse
Affiliation(s)
- Ping Sang
- Department of Spine Surgery, Jilin Provincial People's Hospital, No. 1183, Gongnong Road, Changchun, 130021, Jilin, China.
| | - Xuepeng Li
- Department of Spine Surgery, Jilin Provincial People's Hospital, No. 1183, Gongnong Road, Changchun, 130021, Jilin, China
| | - Ziyu Wang
- Department of Spine Surgery, Jilin Provincial People's Hospital, No. 1183, Gongnong Road, Changchun, 130021, Jilin, China
| |
Collapse
|
6
|
Zhou YR, Dang JJ, Yang QC, Sun ZJ. The regulation of pyroptosis by post-translational modifications: molecular mechanisms and therapeutic targets. EBioMedicine 2024; 109:105420. [PMID: 39476537 PMCID: PMC11564932 DOI: 10.1016/j.ebiom.2024.105420] [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: 05/21/2024] [Revised: 08/23/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Pyroptosis, a type of programmed cell death mediated by gasdermin family proteins, releases a large amount of immune stimulatory substances, which further contribute to inflammation and elicit an adaptive immune response against tumours and pathogens. And it occurs through multiple pathways that involve the activation of specific caspases and the cleavage of gasdermins. Post-translational modifications (PTMs) could influence the chemical properties of the modified residues and neighbouring regions, ultimately affecting the activity, stability, and functions of proteins to regulate pyroptosis. Many studies have been conducted to explore the influence of PTMs on the regulation of pyroptosis. In this review, we provide a comprehensive summary of different types of PTMs that influence pyroptosis, along with their corresponding modifying enzymes. Moreover, it elaborates on the specific contributions of different PTMs to pyroptosis and delves into how the regulation of these modifications can be leveraged for therapeutic interventions in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Yi-Rao Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Jun-Jie Dang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Qi-Chao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
7
|
Liang Z, Damianou A, Vendrell I, Jenkins E, Lassen FH, Washer SJ, Grigoriou A, Liu G, Yi G, Lou H, Cao F, Zheng X, Fernandes RA, Dong T, Tate EW, Di Daniel E, Kessler BM. Proximity proteomics reveals UCH-L1 as an essential regulator of NLRP3-mediated IL-1β production in human macrophages and microglia. Cell Rep 2024; 43:114152. [PMID: 38669140 DOI: 10.1016/j.celrep.2024.114152] [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: 11/10/2023] [Revised: 02/28/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Activation of the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome complex is an essential innate immune signaling mechanism. To reveal how human NLRP3 inflammasome assembly and activation are controlled, in particular by components of the ubiquitin system, proximity labeling, affinity purification, and RNAi screening approaches were performed. Our study provides an intricate time-resolved molecular map of different phases of NLRP3 inflammasome activation. Also, we show that ubiquitin C-terminal hydrolase 1 (UCH-L1) interacts with the NACHT domain of NLRP3. Downregulation of UCH-L1 decreases pro-interleukin-1β (IL-1β) levels. UCH-L1 chemical inhibition with small molecules interfered with NLRP3 puncta formation and ASC oligomerization, leading to altered IL-1β cleavage and secretion, particularly in microglia cells, which exhibited elevated UCH-L1 expression as compared to monocytes/macrophages. Altogether, we profiled NLRP3 inflammasome activation dynamics and highlight UCH-L1 as an important modulator of NLRP3-mediated IL-1β production, suggesting that a pharmacological inhibitor of UCH-L1 may decrease inflammation-associated pathologies.
Collapse
Affiliation(s)
- Zhu Liang
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK; Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK.
| | - Andreas Damianou
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK; Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Iolanda Vendrell
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK; Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Edward Jenkins
- Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Frederik H Lassen
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford OX3 7LF, UK
| | - Sam J Washer
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Athina Grigoriou
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK; Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Guihai Liu
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Gangshun Yi
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Hantao Lou
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| | - Fangyuan Cao
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Xiaonan Zheng
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Ricardo A Fernandes
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Tao Dong
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, UK
| | - Elena Di Daniel
- Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Benedikt M Kessler
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK; Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK.
| |
Collapse
|
8
|
Que Y, Wong C, Qiu J, Gao W, Lin Y, Zhou H, Gao B, Li P, Deng Z, Shi H, Hu W, Liu S, Peng Y, Su P, Xu C, Liang A, Qiu X, Huang D. Maslinic acid alleviates intervertebral disc degeneration by inhibiting the PI3K/AKT and NF-κB signaling pathways. Acta Biochim Biophys Sin (Shanghai) 2024; 56:776-788. [PMID: 38495003 PMCID: PMC11187486 DOI: 10.3724/abbs.2024027] [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: 11/09/2023] [Accepted: 01/04/2024] [Indexed: 03/19/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is the cause of low back pain (LBP), and recent research has suggested that inflammatory cytokines play a significant role in this process. Maslinic acid (MA), a natural compound found in olive plants ( Olea europaea), has anti-inflammatory properties, but its potential for treating IDD is unclear. The current study aims to investigate the effects of MA on TNFα-induced IDD in vitro and in other in vivo models. Our findings suggest that MA ameliorates the imbalance of the extracellular matrix (ECM) and mitigates senescence by upregulating aggrecan and collagen II levels as well as downregulating MMP and ADAMTS levels in nucleus pulposus cells (NPCs). It can also impede the progression of IDD in rats. We further find that MA significantly affects the PI3K/AKT and NF-κB pathways in TNFα-induced NPCs determined by RNA-seq and experimental verification, while the AKT agonist Sc-79 eliminates these signaling cascades. Furthermore, molecular docking simulation shows that MA directly binds to PI3K. Dysfunction of the PI3K/AKT pathway and ECM metabolism has also been confirmed in clinical specimens of degenerated nucleus pulposus. This study demonstrates that MA may hold promise as a therapeutic agent for alleviating ECM metabolism disorders and senescence to treat IDD.
Collapse
Affiliation(s)
- Yichen Que
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Chipiu Wong
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Jincheng Qiu
- Panyu Hospital of Chinese MedicineDepartment of Minimally Invasive Spine SurgeryGuangzhou511408China
| | - Wenjie Gao
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Youxi Lin
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Hang Zhou
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Bo Gao
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Pengfei Li
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Zhihuai Deng
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Huihong Shi
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Wenjun Hu
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Song Liu
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Yan Peng
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Peiqiang Su
- Department of Orthopedic Surgerythe First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou510080China
| | - Caixia Xu
- Research Centre for Translational Medicinethe First Affiliated Hospital of Sun Yat-sen UniversityGuangzhou510006China
| | - Anjing Liang
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Xianjian Qiu
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| | - Dongsheng Huang
- Department of Orthopedic SurgerySun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou510120China
| |
Collapse
|
9
|
Wei D, Tian X, Ren Z, Liu Z, Sun C. Mechanistic insights into the role of USP14 in adipose tissue macrophage recruitment and insulin resistance in obesity. Int J Biol Macromol 2024; 267:131645. [PMID: 38631582 DOI: 10.1016/j.ijbiomac.2024.131645] [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: 01/21/2024] [Revised: 03/28/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
Diet-induced obesity can cause metabolic syndromes. The critical link in disease progression is adipose tissue macrophage (ATM) recruitment, which drives low-level inflammation, triggering adipocyte dysfunction. It is unclear whether ubiquitin-specific proteinase 14 (USP14) affects metabolic disorders by mediating adipose tissue inflammation. In the present study, we showed that USP14 is highly expressed in ATMs of obese human patients and diet-induced obese mice. Mouse USP14 overexpression aggravated obesity-related insulin resistance by increasing the levels of pro-inflammatory ATMs, leading to adipose tissue inflammation, excessive lipid accumulation, and hepatic steatosis. In contrast, USP14 knockdown in adipose tissues alleviated the phenotypes induced by a high-fat diet. Co-culture experiments showed that USP14 deficiency in macrophages led to decreased adipocyte lipid deposition and enhanced insulin sensitivity, suggesting that USP14 plays an important role in ATMs. Mechanistically, USP14 interacted with TNF receptor-associated 6, preventing K48-linked ubiquitination as well as proteasome degradation, leading to increased pro-inflammatory polarization of macrophages. In contrast, the pharmacological inhibition of USP14 significantly ameliorated diet-induced hyperlipidemia and insulin resistance in mice. Our results demonstrated that macrophage USP14 restriction constitutes a key constraint on the pro-inflammatory M1 phenotype, thereby inhibiting obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Dongqin Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Xin Tian
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Zeyu Ren
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Zunhai Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shanxi, China.
| |
Collapse
|
10
|
Zhu D, Liang H, Du Z, Liu Q, Li G, Zhang W, Wu D, Zhou X, Song Y, Yang C. Altered Metabolism and Inflammation Driven by Post-translational Modifications in Intervertebral Disc Degeneration. RESEARCH (WASHINGTON, D.C.) 2024; 7:0350. [PMID: 38585329 PMCID: PMC10997488 DOI: 10.34133/research.0350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a prevalent cause of low back pain and a leading contributor to disability. IVDD progression involves pathological shifts marked by low-grade inflammation, extracellular matrix remodeling, and metabolic disruptions characterized by heightened glycolytic pathways, mitochondrial dysfunction, and cellular senescence. Extensive posttranslational modifications of proteins within nucleus pulposus cells and chondrocytes play crucial roles in reshaping the intervertebral disc phenotype and orchestrating metabolism and inflammation in diverse contexts. This review focuses on the pivotal roles of phosphorylation, ubiquitination, acetylation, glycosylation, methylation, and lactylation in IVDD pathogenesis. It integrates the latest insights into various posttranslational modification-mediated metabolic and inflammatory signaling networks, laying the groundwork for targeted proteomics and metabolomics for IVDD treatment. The discussion also highlights unexplored territories, emphasizing the need for future research, particularly in understanding the role of lactylation in intervertebral disc health, an area currently shrouded in mystery.
Collapse
Affiliation(s)
- Dingchao Zhu
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Zhi Du
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Qian Liu
- College of Life Sciences,
Wuhan University, Wuhan 430072, Hubei Province, China
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Di Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xingyu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
11
|
Tang S, Geng Y, Wang Y, Lin Q, Yu Y, Li H. The roles of ubiquitination and deubiquitination of NLRP3 inflammasome in inflammation-related diseases: A review. BIOMOLECULES & BIOMEDICINE 2024; 24:708-721. [PMID: 38193803 PMCID: PMC11293225 DOI: 10.17305/bb.2023.9997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024]
Abstract
The inflammatory response is a natural immune response that prevents microbial invasion and repairs damaged tissues. However, excessive inflammatory responses can lead to various inflammation-related diseases, posing a significant threat to human health. The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome is a vital mediator in the activation of the inflammatory cascade. Targeting the hyperactivation of the NLRP3 inflammasome may offer potential strategies for the prevention or treatment of inflammation-related diseases. It has been established that the ubiquitination and deubiquitination modifications of the NLRP3 inflammasome can provide protective effects in inflammation-related diseases. These modifications modulate several pathological processes, including excessive inflammatory responses, pyroptosis, abnormal autophagy, proliferation disorders, and oxidative stress damage. Therefore, this review discusses the regulation of NLRP3 inflammasome activation by ubiquitination and deubiquitination modifications, explores the role of these modifications in inflammation-related diseases, and examines the potential underlying mechanisms.
Collapse
Affiliation(s)
- Shaokai Tang
- School of Physical Education, Yanshan University, Qinhuangdao, China
| | - Yuanwen Geng
- School of Physical Education, Yanshan University, Qinhuangdao, China
| | - Yawei Wang
- School of Public Administration, Yanshan University, Qinhuangdao, China
| | - Qinqin Lin
- School of Physical Education, Yanshan University, Qinhuangdao, China
- School of Public Administration, Yanshan University, Qinhuangdao, China
| | - Yirong Yu
- School of Physical Education, Yanshan University, Qinhuangdao, China
| | - Hao Li
- School of Physical Education, Yanshan University, Qinhuangdao, China
| |
Collapse
|
12
|
Yang S, Zhang Y, Peng Q, Meng B, Wang J, Sun H, Chen L, Dai R, Zhang L. Regulating pyroptosis by mesenchymal stem cells and extracellular vesicles: A promising strategy to alleviate intervertebral disc degeneration. Biomed Pharmacother 2024; 170:116001. [PMID: 38128182 DOI: 10.1016/j.biopha.2023.116001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is a main cause of low back pain (LBP), which can lead to disability and thus generate a heavy burden on society. IVDD is characterized by a decrease in nucleus pulposus cells (NPCs) and endogenous mesenchymal stem cells (MSCs), degradation of the extracellular matrix, macrophage infiltration, and blood vessel and nerve ingrowth. To date, the therapeutic approaches regarding IVDD mainly include conservative treatment and surgical intervention. However, both can only relieve symptoms rather than stop or revert the progression of IVDD, since the pathogenesis of IVDD is not yet clear. Pyroptosis, which is characterized by Caspase family dependence and conducted by the Gasdermin family, is a newly discovered mode of programmed cell death. Pyroptosis has been observed in NPCs, annulus fibrosus cells (AFCs), chondrocytes, MSCs, macrophages, vascular endothelial cells and neurons and may contribute to IVDD. MSCs are a kind of pluripotent stem cell that can be found in almost all tissues. MSCs have a strong ability to secrete extracellular vesicles (EVs), which contain exosomes, microvesicles and apoptotic bodies. EVs derived from MSCs play an important role in pyroptosis regulation and could be beneficial for alleviating IVDD. This review focuses on clarifying the regulation of pyroptosis to improve IVDD by MSCs and EVs derived from MSCs.
Collapse
Affiliation(s)
- Sheng Yang
- Department of Orthopedics, Graduate School of Dalian Medical University, Dalian 116044, China
| | - Yongbo Zhang
- Department of Orthopedics, Graduate School of Dalian Medical University, Dalian 116044, China
| | - Qing Peng
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Bo Meng
- Department of Orthopedics, Graduate School of Dalian Medical University, Dalian 116044, China
| | - Jiabo Wang
- Department of Orthopedics, Huai'an 82 Hospital, Huai'an 223003, China
| | - Hua Sun
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Liuyang Chen
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Rui Dai
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou 225001, China
| | - Liang Zhang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
13
|
Gao J, Gao Z. The regulatory role and mechanism of USP14 in endothelial cell pyroptosis induced by coronary heart disease. Clin Hemorheol Microcirc 2024; 86:495-508. [PMID: 38073382 DOI: 10.3233/ch-232003] [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] [Indexed: 05/12/2024]
Abstract
OBJECTIVE The present study probes into the role and mechanism of ubiquitin specific peptidase 14 (USP14) in coronary heart disease (CHD)-triggered endothelial cell pyroptosis. METHODS An in vitro CHD model was established by inducing human coronary artery endothelial cells (HCAECs) with oxidized low-density lipoprotein (ox-LDL). HCAECs were transfected with si-USP14, followed by evaluation of cell viability by CCK-8 assay, detection of lactate dehydrogenase (LDH) activity by assay kit, detection of USP14, miR-15b-5p, NLRP3, GSDMD-N, and Cleaved-Caspase-1 expressions by qRT-PCR or Western blot, as well as IL-1β and IL-18 concentrations by ELISA. Co-IP confirmed the binding between USP14 and NLRP3. The ubiquitination level of NLRP3 in cells was measured after protease inhibitor MG132 treatment. Dual-luciferase reporter assay verified the targeting relationship between miR-15b-5p and USP14. RESULTS USP14 and NLRP3 were highly expressed but miR-15b-5p was poorly expressed in ox-LDL-exposed HCAECs. USP14 silencing strengthened the viability of ox-LDL-exposed HCAECs, reduced the intracellular LDH activity, and diminished the NLRP3, GSDMD-N, Cleaved-Caspase-1, IL-1β, and IL-18 expressions. USP14 bound to NLRP3 protein and curbed its ubiquitination. Repression of NLRP3 ubiquitination counteracted the inhibitory effect of USP14 silencing on HCAEC pyroptosis. miR-15b-5p restrained USP14 transcription and protein expression. miR-15b-5p overexpression alleviated HCAEC pyroptosis by suppressing USP14/NLRP3. CONCLUSION USP14 stabilizes NLRP3 protein expression through deubiquitination, thereby facilitating endothelial cell pyroptosis in CHD. miR-15b-5p restrains endothelial cell pyroptosis by targeting USP14 expression.
Collapse
Affiliation(s)
- Jie Gao
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, China
| | - Zhao Gao
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi'an, China
| |
Collapse
|
14
|
Kordi N, Sanaei M, Akraminia P, Yavari S, Saydi A, Abadi FK, Heydari N, Jung F, Karami S. PANoptosis and cardiovascular disease: The preventive role of exercise training. Clin Hemorheol Microcirc 2024; 88:499-512. [PMID: 39269827 DOI: 10.3233/ch-242396] [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] [Indexed: 09/15/2024]
Abstract
Regulated cell death, including pyroptosis, apoptosis, and necroptosis, is vital for the body's defense system. Recent research suggests that these three types of cell death are interconnected, giving rise to a new concept called PANoptosis. PANoptosis has been linked to various diseases, making it crucial to comprehend its mechanism for effective treatments. PANoptosis is controlled by upstream receptors and molecular signals, which form polymeric complexes known as PANoptosomes. Cell death combines necroptosis, apoptosis, and pyroptosis and cannot be fully explained by any of these processes alone. Understanding pyroptosis, apoptosis, and necroptosis is essential for understanding PANoptosis. Physical exercise has been shown to suppress pyroptotic, apoptotic, and necroptotic signaling pathways by reducing inflammatory factors, proapoptotic factors, and necroptotic factors such as caspases and TNF-alpha. This ultimately leads to a decrease in cardiac structural remodeling. The beneficial effects of exercise on cardiovascular health may be attributed to its ability to inhibit these cell death pathways.
Collapse
Affiliation(s)
- Negin Kordi
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | | | - Peyman Akraminia
- Department of Sports Physiology, Faculty of Physical Education and Sports Sciences, Islamic Azad University, South Tehran Branch, Iran
| | - Sajad Yavari
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Ali Saydi
- Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Fatemeh Khamis Abadi
- Department of Sport Physiology, Faculty of Human Sciences, Islamic Azad University, Borujerd, Iran
| | - Naser Heydari
- Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| | - Friedrich Jung
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Sajad Karami
- Faculty of Physical Education and Sport Sciences, Shahid Rajaee Teacher Training University, Tehran, Iran
| |
Collapse
|
15
|
Duan Y, Yu C, Kuang W, Li J, Qiu S, Ni S, Chen Z. Mesenchymal stem cell exosomes inhibit nucleus pulposus cell apoptosis via the miR-125b-5p/TRAF6/NF-κB pathway axis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1938-1949. [PMID: 37964606 PMCID: PMC10753375 DOI: 10.3724/abbs.2023241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/07/2023] [Indexed: 11/16/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is the pathological basis of a range of degenerative spinal diseases and is the primary cause of lower back pain. Mesenchymal stem cell (MSC) transplantation inhibits IVDD progression. However, the specific mechanisms that underlie these effects remain unclear. In this study, candidate microRNAs (miRNAs) are screened using bioinformatics and high-throughput sequencing. TNF-α is used to induce nucleus pulposus cell (NPC) degeneration. MSC-derived exosomes (MSC-exosomes) are obtained using high-speed centrifugation and identified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blot analysis. Cell viability is determined by CCK-8 assay. Flow cytometry and TUNEL assays are used to detect cell apoptosis. The expression levels of miR-125b-5p are detected by RT-qPCR, and a dual-luciferase gene reporter assay confirms the downstream target genes of miR-125b-5p. Protein expression is determined by western blot analysis. Rat models are used to validate the function of miR-125b-5p in MSC-exosomes. The results show that miR-125b-5p is expressed at low levels in degenerated disc tissues compared with that in normal disc tissues; however, it is highly expressed in MSC-exosomes. Furthermore, MSC-exosomes are efficiently taken up by NPCs while miR-125b-5p is delivered into NPCs; thus, MSC-exosomes act as inhibitors of apoptosis in NPCs. Overexpression of miR-125b-5p downregulates TRAF6 expression and inhibits NF-κB activation. However, TRAF6 overexpression reverses these effects of miR-125b-5p. We demonstrate that MSC-exosomes attenuate IVDD in vivo by delivering miR-125b-5p. MSC-exosomes can deliver miR-125b-5p to target TRAF6, inhibit NF-κB activation, and attenuate the progression of IVDD.
Collapse
Affiliation(s)
- Yang Duan
- Department of Spinal SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| | - Cheng Yu
- Department of Spinal SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| | - Wenhao Kuang
- Department of Spinal SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| | - Jianjun Li
- Department of Spinal SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| | - Sujun Qiu
- Department of Spinal SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| | - Songjia Ni
- Department of Orthopedics and TraumatologyZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| | - Zhong Chen
- Department of Spinal SurgeryZhujiang HospitalSouthern Medical UniversityGuangzhou510260China
| |
Collapse
|
16
|
Qin Y, Zhao W. Posttranslational modifications of NLRP3 and their regulatory roles in inflammasome activation. Eur J Immunol 2023; 53:e2350382. [PMID: 37382218 DOI: 10.1002/eji.202350382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/09/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
The NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is a multimolecular complex that plays a fundamental role in inflammation. Optimal activation of NLRP3 inflammasome is crucial for host defense against pathogens and the maintenance of immune homeostasis. Aberrant NLRP3 inflammasome activity has been implicated in various inflammatory diseases. Posttranslational modifications (PTMs) of NLRP3, a key inflammasome sensor, play critical roles in directing inflammasome activation and controlling the severity of inflammation and inflammatory diseases, such as arthritis, peritonitis, inflammatory bowel disease, atherosclerosis, and Parkinson's disease. Various NLRP3 PTMs, including phosphorylation, ubiquitination, and SUMOylation, could direct inflammasome activation and control inflammation severity by affecting the protein stability, ATPase activity, subcellular localization, and oligomerization of NLRP3 as well as the association between NLRP3 and other inflammasome components. Here, we provide an overview of the PTMs of NLRP3 and their roles in controlling inflammation and summarize potential anti-inflammatory drugs targeting NLRP3 PTMs.
Collapse
Affiliation(s)
- Ying Qin
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wei Zhao
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|