1
|
Hao T, Pei Z, Hu S, Zhao Z, He W, Wang J, Jiang L, Ariben J, Wu L, Yang X, Wang L, Wu Y, Chen X, Li Q, Yang H, Li S, Wang X, Sun M, Zhang B. Identification of osteoarthritis-associated chondrocyte subpopulations and key gene-regulating drugs based on multi-omics analysis. Sci Rep 2025; 15:12448. [PMID: 40216809 PMCID: PMC11992032 DOI: 10.1038/s41598-025-90694-w] [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: 10/08/2024] [Accepted: 02/14/2025] [Indexed: 04/14/2025] Open
Abstract
The mechanism by which chondrocytes respond to mechanical stress in joints significantly affects the balance and function of cartilage. This study aims to characterize osteoarthritis-associated chondrocyte subpopulations and key gene targets for regulatory drugs. To begin, single-cell and transcriptome datasets were obtained from the Gene Expression Omnibus (GEO) database. Cell communication and pseudo-temporal analysis, as well as High-dimensional Weighted Gene Co-expression Network Analysis (hdWGCNA), were conducted on the single-cell data to identify key chondrocyte subtypes and module genes. Subsequently, Consensus Cluster Plus analysis was utilized to identify distinct disease subgroups within the osteoarthritis (OA) training dataset based on the key module genes. Furthermore, differential gene expression analysis and GO/KEGG pathway enrichment analysis were performed on the identified subgroups. To screen for hub genes associated with OA, a combination of 10 machine learning algorithms and 113 algorithm compositions was integrated. Additionally, the immune and pathway scores of the training dataset samples were evaluated using the ESTIMATE, MCP-counter, and ssGSEA algorithms to establish the relationship between the hub genes and immune and pathways. Following this, a network depicting the interaction between the hub genes and transcription factors was constructed based on the Network Analyst database. Moreover, the hub genes were subjected to drug prediction and molecular docking using the RNAactDrug database and AutoDockTools. Finally, real-time fluorescence quantitative PCR (RT-qPCR) was employed to detect the expression of hub genes in the plasma samples collected from osteoarthritis patients and healthy adults. In the OA sample, there is a significant increase in the proportion of prehypertrophic chondrocytes (preHTC), particularly in subgroups 6, 7, and 9. We defined these subgroups as OA_PreHTC subgroups. The OA_PreHTC subgroup exhibits a higher communication intensity with proliferative-related pathways such as ANGPTL and TGF-β. Furthermore, two OA disease subgroups were identified in the training set samples. This led to the identification of 411 differentially expressed genes (DEGs) related to osteoarthritis, 2485 DEGs among subgroups, as well as 238 intersecting genes and 5 hub genes (MMP13, FAM26F, CHI3L1, TAC1, and CKS2). RT-qPCR results indicate significant differences in the expression levels of five hub genes and their related TFs in the clinical blood samples of OA patients compared to the healthy control group (NC). Moreover, these five hub genes are positively associated with inflammatory pathways such as TNF-α, JAK-STAT3, and inflammatory response, while being negatively associated with proliferation pathways like WNT and KRAS. Additionally, the five hub genes are positively associated with neutrophils, activated CD4 T cell, gamma delta T cell, and regulatory T cell, while being negatively associated with CD56dim natural killer cell and Type 17T helper cell. Molecular docking results reveal that CAY10603, Tenulin, T0901317, and Nonactin exhibit high binding activity to CHI3L1, suggesting their potential as therapeutic drugs for OA. The OA_PreHTC subgroups plays a crucial role in the occurrence and development of osteoarthritis (OA). Five hub genes may exert their effects on OA through interactions with PreHTC cells, other chondrocytes, and immune cells, playing a role in inhibiting cell proliferation and stimulating inflammation, thus having high diagnostic value for OA. Additionally, CAY10603, Tenulin, T0901317, and Nonactin have potential therapeutic effects for OA patients.
Collapse
Affiliation(s)
- Ting Hao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Zhiwei Pei
- Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, People's Republic of China
| | - Sile Hu
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Zhenqun Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Wanxiong He
- Sanya People's Hospital, No. 558 Jiefang Road, Sanya City, Hainan Province, People's Republic of China
| | - Jing Wang
- Baotou Medical College Bayannur Clinical Medical College, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Liuchang Jiang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Jirigala Ariben
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Lina Wu
- Aier Eye Hospital, Tianjin University, No. 102 Fukang Road, Tianjin, 300000, People's Republic of China
| | - Xiaolong Yang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Leipeng Wang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Yonggang Wu
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Xiaofeng Chen
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Qiang Li
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Haobo Yang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Siqin Li
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Xing Wang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China.
| | - Mingqi Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| | - Baoxin Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
- Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, People's Republic of China.
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| |
Collapse
|
2
|
Chalmers JD, Metersky M, Aliberti S, Morgan L, Fucile S, Lauterio M, McDonald PP. Neutrophilic inflammation in bronchiectasis. Eur Respir Rev 2025; 34:240179. [PMID: 40174958 PMCID: PMC11962982 DOI: 10.1183/16000617.0179-2024] [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: 08/06/2024] [Accepted: 01/11/2025] [Indexed: 04/04/2025] Open
Abstract
Noncystic fibrosis bronchiectasis, hereafter referred to as bronchiectasis, is a chronic, progressive lung disease that can affect people of all ages. Patients with clinically significant bronchiectasis have chronic cough and sputum production, as well as recurrent respiratory infections, fatigue and impaired health-related quality of life. The pathophysiology of bronchiectasis has been described as a vicious vortex of chronic inflammation, recurring airway infection, impaired mucociliary clearance and progressive lung damage that promotes the development and progression of the disease. This review describes the pivotal role of neutrophil-driven inflammation in the pathogenesis and progression of bronchiectasis. Delayed neutrophil apoptosis and increased necrosis enhance dysregulated inflammation in bronchiectasis and failure to resolve this contributes to chronic, sustained inflammation. The excessive release of neutrophil serine proteases, such as neutrophil elastase, cathepsin G and proteinase 3, promotes a protease-antiprotease imbalance that correlates with increased inflammation in bronchiectasis and contributes to disease progression. While there are currently no licensed therapies to treat bronchiectasis, this review will explore the evolving evidence for neutrophilic inflammation as a novel treatment target with meaningful clinical benefits.
Collapse
Affiliation(s)
- James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Dundee, UK
| | - Mark Metersky
- University of Connecticut School of Medicine, Farmington, CT, USA
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
| | - Lucy Morgan
- Department of Respiratory Medicine, Concord Clinical School, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
3
|
Moulin D, Sellam J, Berenbaum F, Guicheux J, Boutet MA. The role of the immune system in osteoarthritis: mechanisms, challenges and future directions. Nat Rev Rheumatol 2025; 21:221-236. [PMID: 40082724 DOI: 10.1038/s41584-025-01223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 03/16/2025]
Abstract
Osteoarthritis (OA) is a chronic joint disease that has long been considered a simple wear-and-tear condition. Over the past decade, research has revealed that various inflammatory features of OA, such as low-grade peripheral inflammation and synovitis, contribute substantially to the pathophysiology of the disease. Technological advances in the past 5 years have revealed a large diversity of innate and adaptive immune cells in the joints, particularly in the synovium and infrapatellar fat pad. Notably, the presence of synovial lymphoid structures, circulating autoantibodies and alterations in memory T cell and B cell populations have been documented in OA. These data indicate a potential contribution of self-reactivity to the disease pathogenesis, blurring the often narrow and inaccurate line between chronic inflammatory and autoimmune diseases. The diverse immune changes associated with OA pathogenesis can vary across disease phenotypes, and a better characterization of their underlying molecular endotypes will be key to stratifying patients, designing novel therapeutic approaches and ultimately ameliorating treatment allocation. Furthermore, examining both articular and systemic alterations, including changes in the gut-joint axis and microbial dysbiosis, could open up novel avenues for OA management.
Collapse
Affiliation(s)
- David Moulin
- Université de Lorraine, CNRS, IMoPA, Nancy, France.
- CHRU-Nancy, IHU INFINY, Nancy, France.
| | - Jérémie Sellam
- Department of Rheumatology, Saint-Antoine Hospital, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université UMRS 938, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Francis Berenbaum
- Department of Rheumatology, Saint-Antoine Hospital, Centre de Recherche Saint-Antoine, Inserm, Sorbonne Université UMRS 938, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, INSERM, CHU Nantes, UMR1229 Regenerative Medicine and Skeleton, RMeS, Nantes, France
| | - Marie-Astrid Boutet
- Nantes Université, Oniris, INSERM, CHU Nantes, UMR1229 Regenerative Medicine and Skeleton, RMeS, Nantes, France.
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London, UK.
| |
Collapse
|
4
|
Hu K, Song M, Song T, Jia X, Song Y. Osteoimmunology in Osteoarthritis: Unraveling the Interplay of Immunity, Inflammation, and Joint Degeneration. J Inflamm Res 2025; 18:4121-4142. [PMID: 40125089 PMCID: PMC11930281 DOI: 10.2147/jir.s514002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease influenced by multiple factors, with its etiology arising from intricate interactions among mechanical stress, inflammatory processes, and disruptions in bone metabolism. Recent research in bone immunology indicates that immune-mediated mechanisms significantly contribute to the progression of OA, highlighting the interactions among immune cells, cytokine networks, and bone components. Immune cells interact with osteoclasts, osteoblasts, and chondrocytes in a variety of ways. These interactions foster a pro-inflammatory microenvironment, contributing to cartilage breakdown, synovial inflammation, and the sclerosis of subchondral bone. In this article, we present a comprehensive review of bone immunology in OA, focusing on the critical role of immune cells and their cytokine-mediated feedback loops in the pathophysiology of OA. In addition, we are exploring novel therapeutic strategies targeting bone immune pathways, including macrophage polarization, T-cell differentiation, and stem cell therapy to restore the metabolic balance between immunity and bone. By integrating cutting-edge research in bone immunology, this review integrates the latest advancements in bone immunology to construct a comprehensive framework for unraveling the pathogenesis of OA, laying a theoretical foundation for the development of innovative precision therapies.
Collapse
Affiliation(s)
- Kangyi Hu
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Min Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Ting Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Xiao Jia
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| | - Yongjia Song
- Clinical College of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, People’s Republic of China
| |
Collapse
|
5
|
Bhutada S, Hoyle A, Piuzzi NS, Apte SS. Degradomics defines proteolysis information flow from human knee osteoarthritis cartilage to matched synovial fluid and the contributions of secreted proteases ADAMTS5, MMP13 and CMA1 to articular cartilage breakdown. Osteoarthritis Cartilage 2025; 33:116-127. [PMID: 39293776 DOI: 10.1016/j.joca.2024.09.002] [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: 05/29/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVES Proteolytic cartilage extracellular matrix breakdown is a major mechanism of articular cartilage loss in osteoarthritis (OA) pathogenesis. We sought to determine the overlap of proteolytic peptides in matched knee OA cartilage and synovial fluid on a proteome-wide scale to increase the prospective biomarker repertoire and to attribute proteolytic cleavages to specific secreted proteases. DESIGN Matched human knee OA cartilage and synovial fluid (n = 5) were analyzed by N-terminomics using Terminal Amine Isotopic Labeling of Substrates (TAILS), comprising labeling and enrichment of protein N-termini, high-resolution mass spectrometry and positional peptide mapping. Donor non-OA articular cartilage was digested with CMA1, MMP13 or ADAMTS5, and TAILS was used to identify cleavage sites, which were matched against cartilage and synovial fluid degradomes. RESULTS Of over 20,000 cleaved peptides in the combined OA cartilage and synovial fluid degradomes, 677 peptides, originating from 153 proteins, were present in all cartilage and synovial fluid samples. CMA1, MMP13 and ADAMTS5 digestion of cartilage identified numerous cleavage sites for each protease and distinct cleavage site preferences. Peptides resulting from the activities of these proteases were detected in OA cartilage and synovial fluid. CONCLUSIONS Proteolytic fragments from both cartilage and circulating proteins are detectable by synovial fluid degradomics. CMA1, MMP13 and ADAMTS5 activity profiles in cartilage are distinct from each other and the previously determined HtrA1 profile. This work expands the proteolytic biomarker space for OA investigation, suggests that multiple, diverse proteases contribute to cartilage destruction, and demonstrates that their specific contributions can each be defined by multiple biomarkers.
Collapse
Affiliation(s)
- Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, USA; Musculoskeletal Research Center, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Hoyle
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Nicolas S Piuzzi
- Musculoskeletal Research Center, Cleveland Clinic, Cleveland, OH, USA; Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, USA; Musculoskeletal Research Center, Cleveland Clinic, Cleveland, OH, USA; Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
6
|
Xu C, Wang S, Chen X, Zhang T, Ni Z, Ji G, Wang F. Causal associations between circulating immune cells and osteoarthritis: A bidirectional mendelian randomization study. Int Immunopharmacol 2024; 142:113156. [PMID: 39278062 DOI: 10.1016/j.intimp.2024.113156] [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: 06/06/2024] [Revised: 08/05/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
PURPOSE Osteoarthritis (OA) is a common degenerative joint disease, with its etiology remaining poorly understood. Our study aims to explore the causal associations between immune cells and OA, with the goal of generating a new perspective for targeted intervention strategies. METHODS A bidirectional two-sample Mendelian randomization (MR) analysis was performed to estimate the causality between multiple circulating immune cells and different sites of OA. The immune cell traits analyzed included the counts of circulating white blood cells (WBC), lymphocytes, monocytes, neutrophils, eosinophils, and basophils, as well as certain subsets of T and B lymphocytes. The OA types included were OA at any site, knee OA, hip OA, spine OA, thumb OA, and hand OA. Inverse-variance weighted (IVW), MR-Egger, weight median and weight mode were used to evaluate causal effects, with IVW being the main analysis method. Sensitivity analyses were conducted to assess heterogeneity and pleiotropy. RESULTS Our findings indicated that resting regulatory T cell (Treg) absolute counts (AC) were causally associated with an increased risk for spine OA [odds ratio (OR), 1.051; 95 % confidence interval (CI), 1.018-1.086; P=0.0005, PFDR=0.0350], and spine OA showed a positive causal relationship with the neutrophils count (OR, 1.104; 95 %CI, 1.032-1.181; P=0.0039, PFDR=0.0233). Besides, OA at any site was correlated with a rise in circulating eosinophils count (OR, 1.05; 95 %CI, 1.021-1.079; P=0.0007, PFDR=0.0041), while knee OA was associated with decreased total WBC (OR, 0.945; 95 %CI, 0.912-0.979; P=0.0016, PFDR=0.0048) and monocytes counts (OR, 0.958; 95 %CI, 0.934-0.982; P=0.0007, PFDR = 0.0041). No evidence of heterogeneity or horizontal pleiotropy was detected. CONCLUSIONS Our study has demonstrated the causal associations between multiple immune cells and diverse joint OA. These results highlight the intricate interplay between immune cells and OA, suggesting potential targets for therapeutic interventions to manage disease progression and alleviate symptoms.
Collapse
Affiliation(s)
- Chenyue Xu
- Department of Joint Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, Hebei, China.
| | - Shengjie Wang
- Department of Joint Surgery, Harrison International Peace Hospital, Hengshui 053000, Hebei, China.
| | - Xiaobo Chen
- Department of Joint Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, Hebei, China.
| | - Tianhang Zhang
- Hebei Medical University, Shijiazhuang 050051, Hebei, China.
| | - Zhengyi Ni
- Department of Joint Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, Hebei, China.
| | - Gang Ji
- Department of Joint Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, Hebei, China.
| | - Fei Wang
- Department of Joint Surgery, Hebei Medical University Third Hospital, Shijiazhuang 050051, Hebei, China.
| |
Collapse
|
7
|
Scherr F, Darisipudi MN, Börner FR, Austermeier S, Hoffmann F, Eberhardt M, Abdurrahman G, Saade C, von Eggeling F, Kasper L, Holtfreter S, Bröker BM, Kiehntopf M. Alpha-1-antitrypsin as novel substrate for S. aureus' Spl proteases - implications for virulence. Front Immunol 2024; 15:1481181. [PMID: 39628483 PMCID: PMC11611844 DOI: 10.3389/fimmu.2024.1481181] [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: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 12/06/2024] Open
Abstract
Background The serine protease like (Spl) proteases of Staphylococcus aureus are a family of six proteases whose function and impact on virulence are poorly understood. Here we propose alpha-1-antitrypsin (AAT), an important immunomodulatory serine protease inhibitor as target of SplD, E and F. AAT is an acute phase protein, interacting with many proteases and crucial for prevention of excess tissue damage by neutrophil elastase during the innate immune response to infections. Methods We used MALDI-TOF-MS to identify the cleavage site of Spl proteases within AAT's reactive center loop (RCL) and LC-MS/MS to quantify the resulting peptide cleavage product in in vitro digestions of AAT and heterologous expressed proteases or culture supernatants from different S. aureus strains. We further confirmed proteolytic cleavage and formation of a covalent complex with Western Blots, investigated AAT's inhibitory potential against Spls and examined the NETosis inhibitory activity of AAT-Spl-digestions. Results SplD, E and F, but not A or B, cleave AAT in its RCL, resulting in the release of a peptide consisting of AAT's C-terminal 36 amino acids (C36). Synthetic C36, as well as AAT-SplD/E/F-digestions exhibit NETosis inhibition. Only SplE, but not D or F, was partly inhibited by AAT, forming a covalent complex. Conclusion We unraveled a new virulence trait of S. aureus, where SplD/E/F cleave and inactivate AAT while the cleavage product C36 inhibits NETosis.
Collapse
Affiliation(s)
- Franziska Scherr
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | | | - Friedemann R. Börner
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute Jena (HKI), Jena, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, Matrix-assisted Laser Desorption/Ionization (MALDI) Imaging and Clinical Biophotonics, Jena University Hospital, Jena, Germany
| | - Martin Eberhardt
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Christopher Saade
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, Matrix-assisted Laser Desorption/Ionization (MALDI) Imaging and Clinical Biophotonics, Jena University Hospital, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute Jena (HKI), Jena, Germany
- Institute of Novel and Emerging Infectious Diseases (INNT), Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| |
Collapse
|
8
|
Panichi V, Costantini S, Grasso M, Arciola CR, Dolzani P. Innate Immunity and Synovitis: Key Players in Osteoarthritis Progression. Int J Mol Sci 2024; 25:12082. [PMID: 39596150 PMCID: PMC11594236 DOI: 10.3390/ijms252212082] [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: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoarthritis (OA) is a chronic progressive disease of the joint. Although representing the most frequent cause of disability in the elderly, OA remains partly obscure in its pathogenic mechanisms and is still the orphan of resolutive therapies. The concept of what was once considered a "wear and tear" of articular cartilage is now that of an inflammation-related disease that affects over time the whole joint. The attention is increasingly focused on the synovium. Even from the earliest clinical stages, synovial inflammation (or synovitis) is a crucial factor involved in OA progression and a major player in pain onset. The release of inflammatory molecules in the synovium mediates disease progression and worsening of clinical features. The activation of synovial tissue-resident cells recalls innate immunity cells from the bloodstream, creating a proinflammatory milieu that fuels and maintains a damaging condition of low-grade inflammation in the joint. In such a context, cellular and molecular inflammatory behaviors in the synovium could be the primum movens of the structural and functional alterations of the whole joint. This paper focuses on and discusses the involvement of innate immunity cells in synovitis and their role in the progression of OA.
Collapse
Affiliation(s)
- Veronica Panichi
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Silvia Costantini
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40136 Bologna, Italy; (S.C.); (M.G.)
| | - Merimma Grasso
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40136 Bologna, Italy; (S.C.); (M.G.)
| | - Carla Renata Arciola
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, 40136 Bologna, Italy; (S.C.); (M.G.)
- Laboratory of Immunorheumatology and Tissue Regeneration, Laboratory of Pathology of Implant Infections, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Paolo Dolzani
- Laboratory of Immunorheumatology and Tissue Regeneration, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| |
Collapse
|
9
|
Barter MJ, Turner DA, Rice SJ, Hines M, Lin H, Falconer AMD, McDonnell E, Soul J, Arques MDC, Europe-Finner GN, Rowan AD, Young DA, Wilkinson DJ. SERPINA3 is a marker of cartilage differentiation and is essential for the expression of extracellular matrix genes during early chondrogenesis. Matrix Biol 2024; 133:33-42. [PMID: 39097037 DOI: 10.1016/j.matbio.2024.07.004] [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: 11/15/2023] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024]
Abstract
Serine proteinase inhibitors (serpins) are a family of structurally similar proteins which regulate many diverse biological processes from blood coagulation to extracellular matrix (ECM) remodelling. Chondrogenesis involves the condensation and differentiation of mesenchymal stem cells (MSCs) into chondrocytes which occurs during early development. Here, and for the first time, we demonstrate that one serpin, SERPINA3 (gene name SERPINA3, protein also known as alpha-1 antichymotrypsin), plays a critical role in chondrogenic differentiation. We observed that SERPINA3 expression was markedly induced at early time points during in vitro chondrogenesis. We examined the expression of SERPINA3 in human cartilage development, identifying significant enrichment of SERPINA3 in developing cartilage compared to total limb, which correlated with well-described markers of cartilage differentiation. When SERPINA3 was silenced using siRNA, cartilage pellets were smaller and contained lower proteoglycan as determined by dimethyl methylene blue assay (DMMB) and safranin-O staining. Consistent with this, RNA sequencing revealed significant downregulation of genes associated with cartilage ECM formation perturbing chondrogenesis. Conversely, SERPINA3 silencing had a negligible effect on the gene expression profile during osteogenesis suggesting the role of SERPINA3 is specific to chondrocyte differentiation. The global effect on cartilage formation led us to investigate the effect of SERPINA3 silencing on the master transcriptional regulator of chondrogenesis, SOX9. Indeed, we observed that SOX9 protein levels were markedly reduced at early time points suggesting a role for SERPINA3 in regulating SOX9 expression and activity. In summary, our data support a non-redundant role for SERPINA3 in enabling chondrogenesis via regulation of SOX9 levels.
Collapse
Affiliation(s)
- Matthew J Barter
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - David A Turner
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby St, Liverpool L7 8TX, UK
| | - Sarah J Rice
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Mary Hines
- Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby St, Liverpool L7 8TX, UK
| | - Hua Lin
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Adrian M D Falconer
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Euan McDonnell
- Computational Biology Facility, University of Liverpool, MerseyBio, Crown Street, Liverpool L69 7ZB, UK
| | - Jamie Soul
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK; Computational Biology Facility, University of Liverpool, MerseyBio, Crown Street, Liverpool L69 7ZB, UK
| | - Maria Del Carmen Arques
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - G Nicholas Europe-Finner
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Andrew D Rowan
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - David A Young
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - David J Wilkinson
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK; Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby St, Liverpool L7 8TX, UK.
| |
Collapse
|
10
|
Zhang S, Zhong Y, Wang X, Jiang W, Chen X, Kang Y, Li Z, Liao W, Zheng L, Sheng P, Zhang Z. Association of peripheral inflammatory indicators with osteoarthritis risk. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100496. [PMID: 39021876 PMCID: PMC11254169 DOI: 10.1016/j.ocarto.2024.100496] [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: 01/17/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Objectives Numerous studies have established the role of inflammation in osteoarthritis (OA) progression, yet limited research explores the association between systemic inflammatory indicators and pre-diagnosis OA risk. This study aimed to investigate the association between peripheral inflammatory indicators and the risk of OA using data from the UK Biobank. Methods The study analyzed data from 417,507 participants in the UK Biobank, including neutrophil count, lymphocyte count, monocyte count, platelet count, and C-reactive protein meter. Additionally, derived ratios such as NLR(neutrophils-lymphocytes ratio), PLR(Platelets-lymphocytes ratio), SII(systemic immune-inflammation index), and LMR (lymphocytes-monocytes ratio) were examined. Cox proportional hazards models and restricted cubic spline models were used to assess both linear and nonlinear associations. Results Over a mean follow-up period of 12.7 years, a total of 49,509 OA events were identified. The findings revealed that CRP (HR:1.06, 95%CI:1.05-1.07), NLR (HR:1.02, 95%CI:1.01-1.03), PLR (HR:1.02, 95%CI:1.01-1.03), and SII (HR:1.03, 95%CI:1.01-1.04) were associated with an increased risk of OA, while LMR (HR:0.97, 95%CI:0.96-0.99) showed a significant negative correlation with OA risk. Subgroup analyses further emphasized that these associations were significant across most of the population. Although neutrophils, lymphocytes, monocytes, and platelets showed a nominal association with the risk of OA, the results were unreliable, especially for specific joint OA. Conclusion The study provides evidence of a significant association between elevated peripheral inflammatory indicators and OA risk. These findings underscore the importance of low-grade chronic inflammation in OA development. The potential clinical utility of these indicators as early predictors of OA is suggested, warranting further exploration.
Collapse
Affiliation(s)
- Shiyong Zhang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yanlin Zhong
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xudong Wang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wei Jiang
- Department of Bone and Joint, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Xicong Chen
- The Tenth Department of Orthopedics, Foshan Hospital of Traditional Chinese Medicine, China
| | - Yunze Kang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhiwen Li
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiming Liao
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Linli Zheng
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
11
|
Zou Y, Liu C, Wang Z, Li G, Xiao J. Neural and immune roles in osteoarthritis pain: Mechanisms and intervention strategies. J Orthop Translat 2024; 48:123-132. [PMID: 39220678 PMCID: PMC11363721 DOI: 10.1016/j.jot.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Pain is the leading symptom for most individuals with osteoarthritis (OA), a complex condition marked by joint discomfort. Recently, the dynamic interplay between the nervous and immune systems has become a focal point for understanding pain regulation. Despite this, there is still a substantial gap in our comprehensive understanding of the neuroimmune interactions and their effects on pain in OA. This review examines the bidirectional influences between immune cells and nerves in OA progression. It explores current approaches that target neuroimmune pathways, including promoting M2 macrophage polarization and specific neuronal receptor targeting, for effective pain reduction. Translational potential statement This review provides a comprehensive overview of the mechanisms underlying the interplay between the immune system and nervous system during the progression of OA, as well as their contributions to pain. Additionally, it compiles existing intervention strategies targeting neuroimmunity for the treatment of OA pain. This information offers valuable insights for researchers seeking to address the challenge of OA pain.
Collapse
Affiliation(s)
- Yi Zou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Changyu Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Zhenggang Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Guanghui Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| | - Jun Xiao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, Hubei, 430030, China
| |
Collapse
|
12
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Kraus VB, Hsueh MF. Molecular biomarker approaches to prevention of post-traumatic osteoarthritis. Nat Rev Rheumatol 2024; 20:272-289. [PMID: 38605249 DOI: 10.1038/s41584-024-01102-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/13/2024]
Abstract
Up to 50% of individuals develop post-traumatic osteoarthritis (PTOA) within 10 years following knee-joint injuries such as anterior cruciate ligament rupture or acute meniscal tear. Lower-extremity PTOA prevalence is estimated to account for ≥12% of all symptomatic osteoarthritis (OA), or approximately 5.6 million cases in the USA. With knowledge of the inciting event, it might be possible to 'catch PTOA in the act' with sensitive imaging and soluble biomarkers and thereby prevent OA sequelae by early intervention. Existing biomarker data in the joint-injury literature can provide insights into the pathogenesis and early risk trajectory related to PTOA and can help to elucidate a research agenda for preventing or slowing the onset of PTOA. Non-traumatic OA and PTOA have many clinical, radiological and genetic similarities, and efforts to understand early risk trajectories in PTOA might therefore contribute to the identification and classification of early non-traumatic OA, which is the most prevalent form of OA.
Collapse
Affiliation(s)
- Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA.
| | - Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
14
|
Qi B, Wang Z, Cao Y, Zhao H. Study on the treatment of osteoarthritis by acupuncture combined with traditional Chinese medicine based on pathophysiological mechanism: A review. Medicine (Baltimore) 2024; 103:e37483. [PMID: 38579081 PMCID: PMC10994424 DOI: 10.1097/md.0000000000037483] [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: 01/01/2024] [Accepted: 02/13/2024] [Indexed: 04/07/2024] Open
Abstract
Osteoarthritis (OA) is a major contributor to disability and social costs in the elderly. As the population ages and becomes increasingly obese, the incidence of the disease is higher than in previous decades. In recent years, important progress has been made in the causes and pathogenesis of OA pain. Modern medical treatment modalities mainly include the specific situation of the patient and focus on the core treatment, including self-management and education, exercise, and related weight loss. As an important part of complementary and alternative medicine, TCM has remarkable curative effect, clinical safety, and diversity of treatment methods in the treatment of OA. Traditional Chinese Medicine treatment of OA has attracted worldwide attention. Therefore, this article will study the pathophysiological mechanism of OA based on modern medicine, and explore the treatment of OA by acupuncture combined with Chinese Medicine.
Collapse
Affiliation(s)
- Biao Qi
- Shenzhen Baoan District Shiyan People’s Hospital, Shenzhen, China
| | - Zeyu Wang
- Shenzhen Pingshan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Ying Cao
- Shenzhen Pingshan District Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Haishen Zhao
- Community Health Service Center of Nanhui New Town, Shanghai, China
| |
Collapse
|
15
|
Li X, Chen W, Liu D, Chen P, Wang S, Li F, Chen Q, Lv S, Li F, Chen C, Guo S, Yuan W, Li P, Hu Z. Pathological progression of osteoarthritis: a perspective on subchondral bone. Front Med 2024; 18:237-257. [PMID: 38619691 DOI: 10.1007/s11684-024-1061-y] [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: 11/21/2023] [Accepted: 01/17/2024] [Indexed: 04/16/2024]
Abstract
Osteoarthritis (OA) is a degenerative bone disease associated with aging. The rising global aging population has led to a surge in OA cases, thereby imposing a significant socioeconomic burden. Researchers have been keenly investigating the mechanisms underlying OA. Previous studies have suggested that the disease starts with synovial inflammation and hyperplasia, advancing toward cartilage degradation. Ultimately, subchondral-bone collapse, sclerosis, and osteophyte formation occur. This progression is deemed as "top to bottom." However, recent research is challenging this perspective by indicating that initial changes occur in subchondral bone, precipitating cartilage breakdown. In this review, we elucidate the epidemiology of OA and present an in-depth overview of the subchondral bone's physiological state, functions, and the varied pathological shifts during OA progression. We also introduce the role of multifunctional signal pathways (including osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK), and chemokine (CXC motif) ligand 12 (CXCL12)/CXC motif chemokine receptor 4 (CXCR4)) in the pathology of subchondral bone and their role in the "bottom-up" progression of OA. Using vivid pattern maps and clinical images, this review highlights the crucial role of subchondral bone in driving OA progression, illuminating its interplay with the condition.
Collapse
Affiliation(s)
- Xuefei Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Wenhua Chen
- Research and Development Center of Chinese Medicine Resources and Biotechnology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dan Liu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pinghua Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shiyun Wang
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangfang Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qian Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Shunyi Lv
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Fangyu Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chen Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Suxia Guo
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Weina Yuan
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Pan Li
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhijun Hu
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
16
|
Chen J, Wang X, Liu Y, Zhang X. Recent advances on neutrophil dysregulation in the pathogenesis of rheumatic diseases. Curr Opin Rheumatol 2024; 36:142-147. [PMID: 37916474 DOI: 10.1097/bor.0000000000000986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
PURPOSE OF REVIEW The exact pathogenic mechanisms of rheumatic diseases (RMD) remain largely unknown. Increasing evidence highlights a pathogenic role of neutrophil dysregulation in the development of RMD. RECENT FINDINGS The purpose of this review is to present a current overview of recent advancements in understanding the role of neutrophil dysfunction in the development of RMD. Additionally, this review will discuss strategies for targeting pathways associated with neutrophil dysregulation as potential treatments for RMD. One specific aspect of neutrophil dysregulation, known as neutrophil extracellular traps (NETs), will be explored. NETs have been found to contribute to chronic pulmonary inflammation and fibrosis, as well as serve as DNA scaffolds for binding autoantigens, including both citrullinated and carbamylated autoantigens. Putative therapies, such as 6-gingerol or defibrotide, have demonstrated beneficial effects in the treatment of RMD by suppressing NETs formation. SUMMARY Recent advances have significantly reinforced the crucial role of neutrophil dysregulation in the pathogenesis of RMD. A deeper understanding of the potential mechanisms underlying this pathogenic process would aid in the development of more precise and effective targeting strategies, thus ultimately improving the outcomes of RMD.
Collapse
Affiliation(s)
- Jianing Chen
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine
| | - Xinyu Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine
| | - Yudong Liu
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine
| |
Collapse
|
17
|
Han X, Bai F, Li P, Bai X, Zhang Y, Wang W. Identification of novel potential drugs for the treatment and prevention of osteoarthritis. Biochem Biophys Rep 2024; 37:101647. [PMID: 38304574 PMCID: PMC10830515 DOI: 10.1016/j.bbrep.2024.101647] [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: 10/23/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
Objectives Osteoarthritis (OA) is characterized by a high prevalence, poor prognosis, and a propensity to lead to disability. Despite the availability of standard therapies, they are associated with potential side effects and don't provide a complete cure for patients. Consequently, there is an urgent demand for the development of novel drugs. Method The gene expression profiles (GSE64394, GSE178557 and GSE215039) of normal and OA chondrocytes samples were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified by the "LIMMA" R package. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment were conducted using the R package clusterProfiler. A protein-protein (PPI) interaction network was performed to identify hub genes by using the Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape. Small molecule compounds linked to OA were predicted through the NetworkAnalyst platform. Finally, molecular docking was conducted using AutoDock and Pymol software. Results We identified 98 DEGs primarily implicated in endochondral ossification, extracellular matrix degradation, and Wnt signaling pathways. 23 DEGs were closely associated with OA, and 10 hub genes were found to be potential drug targets for OA. Two new targeted compounds, tetrachlorodibenzodioxin (TCDD) and valproic acid (VPA), were screened. And they both exhibited strong binding affinity to their respective targets. Conclusions Reducing exposure to TCDD could be a crucial strategy in preventing OA, and VPA has gained recognition as a novel drug candidate for OA treatment.
Collapse
Affiliation(s)
- Xiaosong Han
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Fan Bai
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Peng Li
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Xiaojin Bai
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Yanli Zhang
- Biological Medicine Research and Development Center, Yangtze Delta of Zhejiang, Hangzhou, Zhejiang, 314006, China
| | - Wenmin Wang
- Biological Medicine Research and Development Center, Yangtze Delta of Zhejiang, Hangzhou, Zhejiang, 314006, China
| |
Collapse
|
18
|
Wang M, Wu Y, Li G, Lin Q, Zhang W, Liu H, Su J. Articular cartilage repair biomaterials: strategies and applications. Mater Today Bio 2024; 24:100948. [PMID: 38269053 PMCID: PMC10806349 DOI: 10.1016/j.mtbio.2024.100948] [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: 10/16/2023] [Revised: 12/09/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Articular cartilage injury is a frequent worldwide disease, while effective treatment is urgently needed. Due to lack of blood vessels and nerves, the ability of cartilage to self-repair is limited. Despite the availability of various clinical treatments, unfavorable prognoses and complications remain prevalent. However, the advent of tissue engineering and regenerative medicine has generated considerable interests in using biomaterials for articular cartilage repair. Nevertheless, there remains a notable scarcity of comprehensive reviews that provide an in-depth exploration of the various strategies and applications. Herein, we present an overview of the primary biomaterials and bioactive substances from the tissue engineering perspective to repair articular cartilage. The strategies include regeneration, substitution, and immunization. We comprehensively delineate the influence of mechanically supportive scaffolds on cellular behavior, shedding light on emerging scaffold technologies, including stimuli-responsive smart scaffolds, 3D-printed scaffolds, and cartilage bionic scaffolds. Biologically active substances, including bioactive factors, stem cells, extracellular vesicles (EVs), and cartilage organoids, are elucidated for their roles in regulating the activity of chondrocytes. Furthermore, the composite bioactive scaffolds produced industrially to put into clinical use, are also explicitly presented. This review offers innovative solutions for treating articular cartilage ailments and emphasizes the potential of biomaterials for articular cartilage repair in clinical translation.
Collapse
Affiliation(s)
- Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Qiushui Lin
- Department of Spine Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Wencai Zhang
- Department of Orthopedics, The First Affiliated Hospital Jinan University, Guangzhou, 510632, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
19
|
Engelmaier A, Prodinger G, Weber A. Selective and sensitive measurement of human neutrophil elastase in clinical samples based on a novel assay principle for protease activity measurement. J Pharm Biomed Anal 2023; 229:115376. [PMID: 37011552 DOI: 10.1016/j.jpba.2023.115376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Imbalances between proteases and protease inhibitors have been associated with several pathological conditions including emphysema as seen in α1-antitrypsin deficiency. For this pathological condition, unimpeded neutrophil elastase activity has been ascribed a pivotal role in the destruction of lung tissue and thus in disease progression. Therefore, low, or non-quantifiable neutrophil elastase (NE) activity levels determined in bronchoalveolar lavage solutions indicate the success of α1-antitrypsin (AAT) augmentation therapy as NE activity will be erased. To overcome the known limitations of available elastase activity assays regarding sensitivity and selectivity, we developed a new elastase activity assay, which fundamentally relies on the highly specific complex formation between AAT and active elastase. Plate-bound AAT captured active elastase from the sample undergoing complex formation, followed by the immunological detection of human NE. This assay principle facilitated the measurement of low pM amounts of active human NE. The data of the assay performance check demonstrated adequate accuracy and precision profiles meeting currently accepted best practices for this activity assay, which can be classified as a ligand-binding assay. Furthermore, spike-recovery studies at low human NE levels, carried out for three human bronchoalveolar samples, resulted in recoveries within the 100 ± 20% range, while good linearity and parallelism of the samples' dilution-response curves was observed. Altogether, complemented by the data of selectivity and robustness studies and the accuracy and precision profile obtained in buffer, this newly developed human NE activity assay was demonstrated to perform accurately and precisely in clinically relevant samples.
Collapse
|
20
|
Talian I, Laputková G, Schwartzová V. Identification of crucial salivary proteins/genes and pathways involved in pathogenesis of temporomandibular disorders. OPEN CHEM 2022. [DOI: 10.1515/chem-2022-0249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Abstract
Temporomandibular disorder (TMD) is a collective term for a group of conditions that lead to impairment of the function of the temporomandibular joint. The proteins/genes and signaling pathways associated with TMD are still poorly understood. The aim of this study was to identify key differentially expressed salivary proteins/genes (DEGs) associated with TMD progression using LC-MS/MS coupled with a bioinformatics approach. The protein–protein interaction network was obtained from the STRING database and the hub genes were identified using Cytoscape including cytoHubba and MCODE plug-ins. In addition, enrichment of gene ontology functions and the Reactome signaling pathway was performed. A total of 140 proteins/genes were differentially expressed. From cluster analysis, a set of 20 hub genes were significantly modulated: ALB, APOA1, B2M, C3, CAT, CLU, CTSD, ENO1, GSN, HBB, HP, HSPA8, LTF, LYZ, MMP9, S100A9, SERPINA1, TF, TPI1, and TXN. Two enriched signaling pathways, glycolysis and gluconeogenesis, and tryptophan signaling pathway involving the hub genes CAT, ENO1, and TPI1 have been identified. The rest of the hub genes were mainly enriched in the innate immune system and antimicrobial peptides signaling pathways. In summary, hub DEGs and the signaling pathways identified here have elucidated the molecular mechanisms of TMD pathogenesis.
Collapse
Affiliation(s)
- Ivan Talian
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P. J. Šafárik , Košice , 040 11 , Slovak Republic
| | - Galina Laputková
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of P. J. Šafárik , Košice , 040 11 , Slovak Republic
| | - Vladimíra Schwartzová
- Clinic of Stomatology and Maxillofacial Surgery, Faculty of Medicine, University of P. J. Šafárik and Louis Pasteur University Hospital , Košice , 041 90 , Slovak Republic
| |
Collapse
|
21
|
Wilkinson DJ. The serine proteinase HtrA1 is ubiquitous and abundant in osteoarthritic joints, but what is it doing? Osteoarthritis Cartilage 2022; 30:1015-1018. [PMID: 35381345 DOI: 10.1016/j.joca.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/24/2022] [Indexed: 02/02/2023]
Affiliation(s)
- David J Wilkinson
- Department of Musculoskeletal Biology and Ageing Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 W Derby St, Liverpool L7 8TX, UK.
| |
Collapse
|
22
|
The Involvement of Neutrophils in the Pathophysiology and Treatment of Osteoarthritis. Biomedicines 2022; 10:biomedicines10071604. [PMID: 35884909 PMCID: PMC9313259 DOI: 10.3390/biomedicines10071604] [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: 06/14/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is a chronic disability that significantly impairs quality of life. OA is one of the most prevalent joint pathologies in the world, characterized by joint pain and stiffness due to the degeneration of articular cartilage and the remodeling of subchondral bone. OA pathogenesis is unique in that it involves simultaneous reparative and degradative mechanisms. Low-grade inflammation as opposed to high-grade allows for this coexistence. Previously, macrophages and T cells have been identified as playing major roles in the inflammation and destruction of OA joints, but recent studies have demonstrated that neutrophils also contribute to the pathogenesis. Neutrophils are the first immune cells to enter the synovium after joint injury, and neutrophilic activity is indispensably a requisite for the progression of OA. Neutrophils act through multiple mechanisms including tissue degeneration via neutrophil elastase (NE), osteophyte development, and the release of inflammatory cytokines and chemokines. As the actions of neutrophils in OA are discovered, the potential for novel therapeutic targets as well as diagnostic methods are revealed. The use of chondrogenic progenitor cells (CPCs), microRNAs, and exosomes are among the newest therapeutic advances in OA treatment, and this review reveals how they can be used to mitigate destructive neutrophil activity.
Collapse
|
23
|
Cross-Tissue Analysis Using Machine Learning to Identify Novel Biomarkers for Knee Osteoarthritis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:9043300. [PMID: 35785145 PMCID: PMC9246600 DOI: 10.1155/2022/9043300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 04/28/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
Abstract
Background Knee osteoarthritis (KOA) is a common degenerative joint disease. In this study, we aimed to identify new biomarkers of KOA to improve the accuracy of diagnosis and treatment. Methods GSE98918 and GSE51588 were downloaded from the Gene Expression Omnibus database as training sets, with a total of 74 samples. Gene differences were analyzed by Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, and Disease Ontology enrichment analyses for the differentially expressed genes (DEGs), and GSEA enrichment analysis was carried out for the training gene set. Through least absolute shrinkage and selection operator regression analysis, the support vector machine recursive feature elimination algorithm, and gene expression screening, the range of DEGs was further reduced. Immune infiltration analysis was carried out, and the prediction results of the combined biomarker logistic regression model were verified with GSE55457. Results In total, 84 DEGs were identified through differential gene expression analysis. The five biomarkers that were screened further showed significant differences in cartilage, subchondral bone, and synovial tissue. The diagnostic accuracy of the model synthesized using five biomarkers through logistic regression was better than that of a single biomarker and significantly better than that of a single clinical trait. Conclusions CX3CR1, SLC7A5, ARL4C, TLR7, and MTHFD2 might be used as novel biomarkers to improve the accuracy of KOA disease diagnosis, monitor disease progression, and improve the efficacy of clinical treatment.
Collapse
|
24
|
Wang Z, Le H, Wang Y, Liu H, Li Z, Yang X, Wang C, Ding J, Chen X. Instructive cartilage regeneration modalities with advanced therapeutic implantations under abnormal conditions. Bioact Mater 2022; 11:317-338. [PMID: 34977434 PMCID: PMC8671106 DOI: 10.1016/j.bioactmat.2021.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/19/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
The development of interdisciplinary biomedical engineering brings significant breakthroughs to the field of cartilage regeneration. However, cartilage defects are considerably more complicated in clinical conditions, especially when injuries occur at specific sites (e.g., osteochondral tissue, growth plate, and weight-bearing area) or under inflammatory microenvironments (e.g., osteoarthritis and rheumatoid arthritis). Therapeutic implantations, including advanced scaffolds, developed growth factors, and various cells alone or in combination currently used to treat cartilage lesions, address cartilage regeneration under abnormal conditions. This review summarizes the strategies for cartilage regeneration at particular sites and pathological microenvironment regulation and discusses the challenges and opportunities for clinical transformation.
Collapse
Affiliation(s)
- Zhonghan Wang
- Department of Plastic and Reconstruct Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, PR China
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Hanxiang Le
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Yanbing Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Zuhao Li
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Xiaoyu Yang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, PR China
| | - Chenyu Wang
- Department of Plastic and Reconstruct Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| |
Collapse
|
25
|
Bigalke A, Sponholz C, Schnabel C, Bauer M, Kiehntopf M. Multiplex quantification of C-terminal alpha-1-antitrypsin peptides provides a novel approach for characterizing systemic inflammation. Sci Rep 2022; 12:3844. [PMID: 35264629 PMCID: PMC8907207 DOI: 10.1038/s41598-022-07752-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/09/2023] Open
Abstract
C-terminal peptides (CAAPs) of the highly abundant serine protease alpha-1-antitrypsin (A1AT) have been identified at various lengths in several human materials and have been proposed to serve as putative biomarkers for a variety of diseases. CAAPs are enzymatically formed and these enzymatic activities are often associated with excessive immune responses (e.g. sepsis, allergies). However, most of those CAAPs have been either detected using in vitro incubation experiments or in human materials which are not easily accessible. To gain a comprehensive understanding about the occurrence and function of CAAPs in health and disease, a LC-MS/MS method for the simultaneous detection of nine CAAPs was developed and validated for human plasma (EDTA and lithium-heparin) and serum. Using this newly developed method, we were able to detect and quantify five CAAPs in healthy individuals thereby providing an initial proof for the presence of C36, C37, C40 and C44 in human blood. Concentrations of four CAAPs in a clinical test cohort of patients suffering from sepsis were significantly higher compared to healthy controls. These results reveal that in addition to C42 other fragments of A1AT seem to play a crucial role during systemic infections. The proposed workflow is simple, rapid and robust; thus this method could be used as diagnostic tool in routine clinical chemistry as well as for research applications for elucidating the diagnostic potential of CAAPs in numerous diseases. To this end, we also provide an overview about the current state of knowledge for CAAPs identified in vitro and in vivo.
Collapse
Affiliation(s)
- Arite Bigalke
- grid.275559.90000 0000 8517 6224Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Christoph Sponholz
- grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Claudia Schnabel
- grid.275559.90000 0000 8517 6224Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Therapy, Jena University Hospital, Jena, Germany
| | - Michael Kiehntopf
- grid.275559.90000 0000 8517 6224Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Jena, Germany
| |
Collapse
|
26
|
Young DA, Barter MJ, Soul J. Osteoarthritis year in review: genetics, genomics, epigenetics. Osteoarthritis Cartilage 2022; 30:216-225. [PMID: 34774787 PMCID: PMC8811265 DOI: 10.1016/j.joca.2021.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In this review, we have highlighted the advances over the past year in genetics, genomics and epigenetics in the field of osteoarthritis (OA). METHODS A literature search of PubMed was performed using the criteria: "osteoarthritis" and one of the following terms "genetic(s), genomic(s), epigenetic(s), polymorphism, noncoding ribonucleic acid (RNA), microRNA, long noncoding RNA, lncRNA, circular RNA, RNA sequencing (RNA-seq), single cell sequencing, transcriptomics, or deoxyribonucleic acid (DNA) methylation between April 01, 2020 and April 30, 2021. RESULTS In total we identified 765 unique publications, which eventually reduced to 380 of relevance to the field as judged by two assessors. Many of these studies included multiple search terms. We summarised advances relating to genetics, functional genetics, genomics and epigenetics, focusing on our personal key papers during the year. CONCLUSIONS This year few studies have identified new genetic variants contributing to OA susceptibility, but a focus has been on refining risk loci or their functional validation. The use of new technologies together with investigating the cross-talk between multiple tissue types, greater sample sizes and/or better patient classification (OA subtypes) will continue to increase our knowledge of disease mechanisms and progress towards understanding and treating OA.
Collapse
Affiliation(s)
- D A Young
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK.
| | - M J Barter
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - J Soul
- Skeletal Research Group, Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
27
|
Kaneva MK. Neutrophil elastase and its inhibitors-overlooked players in osteoarthritis. FEBS J 2021; 289:113-116. [PMID: 34580987 DOI: 10.1111/febs.16194] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022]
Abstract
Cartilage homeostasis is maintained by a delicate balance between anabolism and catabolism. In osteoarthritis, pathological biomechanics or injury triggers cartilage breakdown, nonresolving synovial inflammation, and bone changes, causing reduced joint mobility and incapacitating pain. Undoubtedly, the most important cartilage degrading collagenase during osteoarthritis, matrix metalloproteinase (MMP)-13, is activated by an unlikely player: neutrophil elastase. Although primarily associated with inflammatory arthritis, neutrophil elastase is present in the osteoarthritic joint, and through activating MMP-13, spurs a cascade of events leading not just to the aberrant destruction of the cartilage itself, but to the proteolysis of its own inhibitor, alpha-1-antitrypsin, as described in the new study by Wilkinson et al. Endowed with potent chondrogenic and cartilage-protective properties, the loss of alpha-1-antitrypsin from cartilage will have major consequences for osteoarthritis progression, and strategies to prevent its loss, or replace it, might provide an innovative treatment opportunity that should not be ignored. Comment on: https://doi.org/10.1111/febs.16127.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, UK.,Centre for Inflammation and Therapeutic Innovation (CiTI), Queen Mary University of London, UK
| |
Collapse
|