Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (230)

Search Parameters:
Keywords = synovium

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
7 pages, 1460 KiB  
Interesting Images
A Lipoma Arborescens Probably Causing Significant Osteoarthritis of the Elbow in a Young Man
by Won-Jong Bahk, Seungyup Shin, Junho Jang, Kyung Jin Seo, Yongju Kim and Hyunjung Kim
Diagnostics 2025, 15(15), 1888; https://doi.org/10.3390/diagnostics15151888 - 28 Jul 2025
Viewed by 227
Abstract
Lipoma arborescens (LA) is a rare, non-neoplastic, intra-articular, mass-like lesion with villous lipomatous proliferation that replaces and distends the synovium, particularly in the knee joint. A few cases have been sporadically reported to affect the shoulder, elbow, wrist, hip, and ankle. The authors [...] Read more.
Lipoma arborescens (LA) is a rare, non-neoplastic, intra-articular, mass-like lesion with villous lipomatous proliferation that replaces and distends the synovium, particularly in the knee joint. A few cases have been sporadically reported to affect the shoulder, elbow, wrist, hip, and ankle. The authors would like to present a rare and unique case of LA in the elbow joint with significant osteoarthritis in a 24-year-old young man, which suggests that a longstanding pre-existing LA can give rise to severe degenerative arthritis even in young patients unless diagnosed early and adequately treated. Full article
(This article belongs to the Section Clinical Diagnosis and Prognosis)
Show Figures

Figure 1

60 pages, 3898 KiB  
Review
The Therapeutic Potential of Phytochemicals Unlocks New Avenues in the Management of Rheumatoid Arthritis
by Kalina A. Nikolova-Ganeva, Nikolina M. Mihaylova, Lidiya A. Kechidzhieva, Kristina I. Ivanova, Alexander S. Zarkov, Daniel L. Parzhanov, Momchil M. Ivanov and Andrey S. Marchev
Int. J. Mol. Sci. 2025, 26(14), 6813; https://doi.org/10.3390/ijms26146813 - 16 Jul 2025
Viewed by 527
Abstract
Rheumatoid arthritis (RA) is a progressive and systemic autoimmune disease, characterized by a chronic inflammatory process, affecting the lining of the synovial joints, many body organs/systems, and blood vessels. Its pathological hallmarks are hyperplasic synovium, bone erosion, and progressive joint destruction. Rheumatoid arthritis [...] Read more.
Rheumatoid arthritis (RA) is a progressive and systemic autoimmune disease, characterized by a chronic inflammatory process, affecting the lining of the synovial joints, many body organs/systems, and blood vessels. Its pathological hallmarks are hyperplasic synovium, bone erosion, and progressive joint destruction. Rheumatoid arthritis affects over 20 million people, with a worldwide prevalence of 0.5–1.0%, exhibiting gender, ethnic, and geographical differences. The progressive disability severely impairs physical motion and quality of life and is finally leading to a shortened life span. The pathogenesis of RA is a complex and still poorly understood process in which genetic and environmental factors are principally associated. Current treatment mostly relies on conventional/non-biological disease-modifying anti-rheumatic drugs (cDMARDs), analgesics, non-steroidal anti-inflammatory drugs, glucocorticoids, steroids, immunosuppresants, and biologic DMARDs, which only control inflammation and pain. Along with side effects (drug toxicity and intolerance), these anti-rheumatic drugs possess limited efficacy. Therefore, the discovery of novel multi-target therapeutics with an improved safety profile that function as inhibitors of RA-linked signaling systems are in high demand, and this is in the interest of both patients and clinicians. Plant-derived extracts, nutritional supplements, dietary medicine, and molecules with anti-inflammatory activity represent promising adjuvant agents or alternatives for RA therapeutics. This review not only aims to discuss the basic features of RA pathogenesis, risk factors, and signaling pathways but also highlights the research progress in pre-clinical RA in in vitro and in vivo models, revealing new avenues in the management of the disease in terms of comprehensive multidisciplinary strategies originating from medicinal plants and plant-derived molecules. Full article
(This article belongs to the Special Issue Natural Products as Multitarget Agents in Human Diseases)
Show Figures

Graphical abstract

22 pages, 2521 KiB  
Article
Assessment of Feasibility of the M2 Macrophage-Based Adoptive Gene Transfer Strategy for Osteoarthritis with a Mouse Model
by Matilda H.-C. Sheng, David J. Baylink, Charles H. Rundle and Kin-Hing William Lau
Cells 2025, 14(14), 1067; https://doi.org/10.3390/cells14141067 - 11 Jul 2025
Viewed by 396
Abstract
Current osteoarthritis (OA) therapies fail to yield long-term clinical benefits, due in part to the lack of a mechanism for the targeted and confined delivery of therapeutics to OA joints. This study evaluates if M2 macrophages are effective cell vehicles for the targeted [...] Read more.
Current osteoarthritis (OA) therapies fail to yield long-term clinical benefits, due in part to the lack of a mechanism for the targeted and confined delivery of therapeutics to OA joints. This study evaluates if M2 macrophages are effective cell vehicles for the targeted and confined delivery of therapeutic genes to OA joints. CT bioluminescence in vivo cell tracing and fluorescent microscopy reveal that intraarticularly injected M2 macrophages were recruited to and retained at inflamed synovia. The feasibility of an M2 macrophage-based adoptive gene transfer strategy for OA was assessed using IL-1Ra as the therapeutic gene in a mouse tibial plateau injury model. Mouse M2 macrophages were transduced with lentiviral vectors expressing IL-1Ra or GFP. The transduced macrophages were intraarticularly injected into injured joints at 7 days post-injury and OA progression was monitored with plasma COMP and histology at 4 weeks. The IL-1Ra-expressing M2 macrophage treatment reduced plasma COMP, increased the area and width of the articular cartilage layer, decreased synovium thickness, and reduced the OARSI OA score without affecting the osteophyte maturity and meniscus scores when compared to the GFP-expressing M2 macrophage-treated or PBS-treated controls. When the treatment was given at 5 weeks post-injury, at which time OA should have developed, the IL-1Ra-M2 macrophage treatment also reduced plasma COMP, had a greater articular cartilage area and width, decreased synovial thickness, and reduced the OARSI OA score without an effect on the meniscus and osteophyte maturity scores at 8 weeks post-injury. In conclusion, the IL-1Ra-M2 macrophage treatment, given before or after OA was developed, delayed OA progression, indicating that the M2 macrophage-based adoptive gene transfer strategy for OA is tenable. Full article
(This article belongs to the Section Cell and Gene Therapy)
Show Figures

Figure 1

9 pages, 1777 KiB  
Article
Patient-Derived Explants of Osteoarthritic Synovium as Ex Vivo Model for Preclinical Research
by Claudia D’Oria, Gilberto Cincinelli, Ramona Bason, Federica Pisati, Francesca Simoncello, Isabella Scotti, Laura Giudice, Ilaria Suardi, Paolo Ferrua, Chiara Fossati, Pietro Simone Randelli, Roberto Caporali, Massimiliano Pagani and Francesca Ingegnoli
Int. J. Mol. Sci. 2025, 26(14), 6665; https://doi.org/10.3390/ijms26146665 - 11 Jul 2025
Viewed by 276
Abstract
Osteoarthritis (OA) is the most common chronic arthropathy worldwide. OA synovitis is a common feature that predicts the development and progression of symptoms and joint damage. Although the OA synovium is a target for novel therapies, the development of ex vivo models remains [...] Read more.
Osteoarthritis (OA) is the most common chronic arthropathy worldwide. OA synovitis is a common feature that predicts the development and progression of symptoms and joint damage. Although the OA synovium is a target for novel therapies, the development of ex vivo models remains an area requiring further research. We aim to develop a 3D tissue explant culture model of human OA synovium that preserves the architecture and cellular heterogeneity of the original tissue in vitro. We derived tissue explant models from seven patients with OA and followed the culture for up to 10 days, assessing their morphology and cellular composition by immunohistochemistry (IHC) and flow cytometry, respectively. IHC analysis of explant cultures showed that tissue integrity and viability were maintained in our in vitro system. Furthermore, cellular heterogeneity was essentially unchanged when considering CD4+ T cells, CD8+ T cells, and myeloid fractions in our model. No significant variation was observed in the CD90+ and CD90-CD55+ fractions, which also maintained an activated state as indicated by high levels of FAP expression. An ex vivo OA synovial tissue explant model can maintain pathological tissue integrity for 10 days in culture. This simple and reliable culture system may be useful for analyzing the pathogenesis of OA disease and for the development and testing of therapeutic drugs. Full article
(This article belongs to the Special Issue Recent Advances in Osteoarthritis Pathways and Biomarker Research)
Show Figures

Figure 1

22 pages, 2943 KiB  
Article
Identification of Genes Linked to Meniscal Degeneration in Osteoarthritis: An In Silico Analysis
by Aliki-Alexandra Papageorgiou, Charalampos Balis and Ioanna Papathanasiou
Int. J. Mol. Sci. 2025, 26(14), 6651; https://doi.org/10.3390/ijms26146651 - 11 Jul 2025
Viewed by 299
Abstract
Meniscal degradation is considered a driver of osteoarthritis (OA) progression, but the underlying mechanisms leading to age-related meniscus degeneration remain unknown. This study aimed to identify key genes and pathways involved in meniscal degradation through a computational analysis. Gene expression profiles were obtained [...] Read more.
Meniscal degradation is considered a driver of osteoarthritis (OA) progression, but the underlying mechanisms leading to age-related meniscus degeneration remain unknown. This study aimed to identify key genes and pathways involved in meniscal degradation through a computational analysis. Gene expression profiles were obtained from the Gene Expression Omnibus (GEO) database. Differential expression gene (DEG) analysis was performed using DESeq2 accompanied by functional enrichment analysis, protein–protein interaction (PPI) and clustering analysis. Additionally, gene set enrichment analysis (GSEA) was performed. A total of 85 mRNAs (DEMs) and 8 long non-coding RNAs (DE LncRNAs) were found to be differentially expressed in OA meniscus tissues. Among 85 DEMs, 12 genes were found to be known OA-related genes, whereas 15 genes acted as transcription regulators, including RUNX2 and TBX4, which were identified as effector genes for OA. Enrichment analysis revealed the implication of DEMs in cartilage-degradation-related processes, including inflammatory pathways, lipid metabolism, extracellular matrix organization and superoxide/nitric oxide metabolic processes. Target genes of DE lncRNAs were found to be involved in chondrocyte differentiation and pathways related to cartilage degradation. A comparative analysis of meniscus, synovium and cartilage datasets identified three genes (GJB2, PAQR5 and CLEC12A) as being differentially expressed across all three OA-affected tissues, which were implicated in inflammatory and cholesterol metabolism processes. Our results support that shared mechanisms lead to meniscal and cartilage degradation during OA progression, providing further insights into the processes underlying OA pathogenesis and potential therapeutic targets for knee OA. Full article
(This article belongs to the Special Issue Computer Analysis for Molecular Pathological Research)
Show Figures

Figure 1

28 pages, 1727 KiB  
Review
Computational and Imaging Approaches for Precision Characterization of Bone, Cartilage, and Synovial Biomolecules
by Rahul Kumar, Kyle Sporn, Vibhav Prabhakar, Ahab Alnemri, Akshay Khanna, Phani Paladugu, Chirag Gowda, Louis Clarkson, Nasif Zaman and Alireza Tavakkoli
J. Pers. Med. 2025, 15(7), 298; https://doi.org/10.3390/jpm15070298 - 9 Jul 2025
Viewed by 656
Abstract
Background/Objectives: Degenerative joint diseases (DJDs) involve intricate molecular disruptions within bone, cartilage, and synovial tissues, often preceding overt radiographic changes. These tissues exhibit complex biomolecular architectures and their degeneration leads to microstructural disorganization and inflammation that are challenging to detect with conventional imaging [...] Read more.
Background/Objectives: Degenerative joint diseases (DJDs) involve intricate molecular disruptions within bone, cartilage, and synovial tissues, often preceding overt radiographic changes. These tissues exhibit complex biomolecular architectures and their degeneration leads to microstructural disorganization and inflammation that are challenging to detect with conventional imaging techniques. This review aims to synthesize recent advances in imaging, computational modeling, and sequencing technologies that enable high-resolution, non-invasive characterization of joint tissue health. Methods: We examined advanced modalities including high-resolution MRI (e.g., T1ρ, sodium MRI), quantitative and dual-energy CT (qCT, DECT), and ultrasound elastography, integrating them with radiomics, deep learning, and multi-scale modeling approaches. We also evaluated RNA-seq, spatial transcriptomics, and mass spectrometry-based proteomics for omics-guided imaging biomarker discovery. Results: Emerging technologies now permit detailed visualization of proteoglycan content, collagen integrity, mineralization patterns, and inflammatory microenvironments. Computational frameworks ranging from convolutional neural networks to finite element and agent-based models enhance diagnostic granularity. Multi-omics integration links imaging phenotypes to gene and protein expression, enabling predictive modeling of tissue remodeling, risk stratification, and personalized therapy planning. Conclusions: The convergence of imaging, AI, and molecular profiling is transforming musculoskeletal diagnostics. These synergistic platforms enable early detection, multi-parametric tissue assessment, and targeted intervention. Widespread clinical integration requires robust data infrastructure, regulatory compliance, and physician education, but offers a pathway toward precision musculoskeletal care. Full article
(This article belongs to the Special Issue Cutting-Edge Diagnostics: The Impact of Imaging on Precision Medicine)
Show Figures

Figure 1

16 pages, 1041 KiB  
Review
The Role of SLPI Gene-Mediated Inflammation in Osteoarthritis
by Mahmuda Siddika Shefa and Wanil Kim
Biomolecules 2025, 15(7), 909; https://doi.org/10.3390/biom15070909 - 20 Jun 2025
Viewed by 937
Abstract
Osteoarthritis (OA) is a degenerative disease of joint tissue characterized by the breaking down of cartilage and resulting changes in synovium and bone. Mechanics and biology interact in a feed-forward manner in that imbalanced joint loading leads to tissue degeneration and vice versa. [...] Read more.
Osteoarthritis (OA) is a degenerative disease of joint tissue characterized by the breaking down of cartilage and resulting changes in synovium and bone. Mechanics and biology interact in a feed-forward manner in that imbalanced joint loading leads to tissue degeneration and vice versa. Amid numerous genetic factors, the Secretory Leukocyte Protease Inhibitor (SLPI) gene encodes a protein that plays a crucial role in inhibiting proteases, modulating inflammation, promoting tissue repair, and regulating immune responses. In the context of OA, SLPI has been identified as a key regulator in joint homeostasis. The release of SLPI in human tissues is augmented by pro-inflammatory factors. Such factors include cytokines released during infection or inflammatory processes, such as interleukins-1 and 6 (IL-1 and IL-6), and tumor necrosis factor alpha (TNF-α) released in many inflammatory rheumatic diseases. In this work, a comprehensive review of SLPI-mediated inflammation in OA, its biological functions, and its association with OA is described, providing a foundation for future investigations into its potential therapeutic use. As there is no effective strategy to treat or prevent OA in clinic, further investigation is encouraged to explore the translational possibility of SLPI for drug development. Full article
(This article belongs to the Section Molecular Biomarkers)
Show Figures

Figure 1

26 pages, 2956 KiB  
Review
Unraveling Osteoarthritis: Mechanistic Insights and Emerging Therapies Targeting Pain and Inflammation
by Muskan Alad, Fajer Yousef, Laura M. Epure, Angelina Lui, Michael P. Grant, Geraldine Merle, Nicoletta Eliopoulos, Jake Barralet, John Antoniou and Fackson Mwale
Biomolecules 2025, 15(6), 874; https://doi.org/10.3390/biom15060874 - 16 Jun 2025
Viewed by 1588
Abstract
Osteoarthritis (OA) is now widely recognized not merely as a cartilage-centric disease but as a multifactorial disorder affecting the entire joint as an organ, including the articular cartilage, subchondral bone, synovium, ligaments, menisci, and the innervating neural elements. This review explores the complex [...] Read more.
Osteoarthritis (OA) is now widely recognized not merely as a cartilage-centric disease but as a multifactorial disorder affecting the entire joint as an organ, including the articular cartilage, subchondral bone, synovium, ligaments, menisci, and the innervating neural elements. This review explores the complex pathophysiology of OA with a focus on the emerging mechanisms of pain and inflammation that extend beyond articular cartilage degradation. Joint inflammation driven by immune activation in response to cellular stress signals promotes the release of pro-inflammatory mediators and catabolic enzymes. Key signaling pathways such as NF-κB, MAPKs, and JAK/STAT amplify these responses, and pain is sustained through peripheral and central sensitization, contributing to exacerbating symptoms even in the absence of visible joint damage. This review also integrates molecular and cellular mechanisms to highlight innovative therapies aimed at modifying both the structural damage and neurosensory drivers of pain. These approaches offer the potential to not only alleviate symptoms but also alter disease progression, signaling a move toward personalized, mechanism-based treatments. Given the intricate interactions among joint tissues, immune activation, and sensory processing, a comprehensive strategy that targets both structural degeneration and neuroinflammation is essential for the future of OA management. Emphasizing the joint as an integrated organ, we advocate for translational research linking molecular pathology with clinically meaningful outcomes. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

24 pages, 12086 KiB  
Article
Integrative Spatial Proteomics and Single-Cell RNA Sequencing Unveil Molecular Complexity in Rheumatoid Arthritis for Novel Therapeutic Targeting
by Xue Wang, Fei Wang, Archana S. Iyer, Heather Knight, Lori J. Duggan, Yingli Yang, Liang Jin, Baoliang Cui, Yupeng He, Jan Schejbal, Lucy A. Phillips, Bohdan P. Harvey, Sílvia Sisó and Yu Tian
Proteomes 2025, 13(2), 17; https://doi.org/10.3390/proteomes13020017 - 22 May 2025
Viewed by 3338
Abstract
Understanding the heterogeneity of Rheumatoid Arthritis (RA) and identifying therapeutic targets remain challenging using traditional bulk transcriptomics alone, as it lacks the spatial and protein-level resolution needed to fully capture disease and tissue complexities. In this study, we applied Laser Capture Microdissection (LCM) [...] Read more.
Understanding the heterogeneity of Rheumatoid Arthritis (RA) and identifying therapeutic targets remain challenging using traditional bulk transcriptomics alone, as it lacks the spatial and protein-level resolution needed to fully capture disease and tissue complexities. In this study, we applied Laser Capture Microdissection (LCM) coupled with mass spectrometry-based proteomics to analyze histopathological niches of the RA synovium, enabling the identification of protein expression profiles of the diseased synovial lining and sublining microenvironments compared to their healthy counterparts. In this respect, key pathogenetic RA proteins like membrane proteins (TYROBP, AOC3, SLC16A3, TCIRG1, and NCEH1), and extracellular matrix (ECM) proteins (PLOD2, OGN, and LUM) showed different expression patterns in diseased synovium compartments. To enhance our understanding of cellular dynamics within the dissected regions, we further integrated the proteomic dataset with single-cell RNA sequencing (scRNA-seq), and deduced cell type enrichment, including T cells, fibroblasts, NK cells, myeloid cells, B cells, and synovial endothelial cells. By combining high-resolution spatial proteomics and transcriptomic analyses, we provide novel insights into the molecular mechanisms driving RA, and highlight potential protein targets for therapeutic intervention. This integrative approach offers a more comprehensive view of RA synovial pathology, and mitigates the limitations of traditional bulk transcriptomics in target discovery. Full article
Show Figures

Figure 1

15 pages, 2493 KiB  
Article
Inertial and Linear Re-Absorption Effects on a Synovial Fluid Flow Through a Lubricated Knee Joint
by Abdul Majeed Siddiqui, Khadija Maqbool, Afifa Ahmed and Amer Bilal Mann
Lubricants 2025, 13(5), 196; https://doi.org/10.3390/lubricants13050196 - 27 Apr 2025
Viewed by 473
Abstract
This study examines the flow dynamics of synovial fluid within a lubricated knee joint during movement, incorporating the effect of inertia and linear re-absorption at the synovial membrane. The fluid behavior is modeled using a couple-stress fluid framework, which accounts for mechanical phenomena [...] Read more.
This study examines the flow dynamics of synovial fluid within a lubricated knee joint during movement, incorporating the effect of inertia and linear re-absorption at the synovial membrane. The fluid behavior is modeled using a couple-stress fluid framework, which accounts for mechanical phenomena and employs a lubricated membrane. synovial membrane plays a crucial role in reducing drag and enhancing joint lubrication for the formation of a uniform lubrication layer over the cartilage surfaces. The mathematical model of synovial fluid flow through the knee joint presents a set of non-linear partial differential equations solved by a recursive approach and inverse method through the software Mathematica 11. The results indicate that synovial fluid flow generates high pressure and shear stress away from the entry point due to the combined effects of inertial forces, linear re-absorption, and micro-rotation within the couple-stress fluid. Axial flow intensifies at the center of the knee joint during activity in the presence of linear re-absorption and molecular rotation, while transverse flow increases away from the center and near to synovium due to its permeability. These findings provide critical insights for biomedical engineers to quantify pressure and stress distributions in synovial fluid to design artificial joints. Full article
Show Figures

Figure 1

17 pages, 2608 KiB  
Article
Inactivated Viral Vaccine BBV87 Protects Against Chikungunya Virus Challenge in a Non-Human Primate Model
by Sarah L. Kempster, Deborah Ferguson, Claire Ham, Joanna Hall, Adrian Jenkins, Elaine Giles, Simon L. Priestnall, Alejandro Suarez-Bonnet, Pierre Roques, Roger Le Grand, Sumathy Kandaswamy, Sushant Sahastrabuddhe, Libia Milena Hernandez, Sunee Chuasuwan, Hyeon Seon Ahn, Deok Ryun Kim, Anh Wartel, Raphaël M. Zellweger, Neil Berry and Neil Almond
Viruses 2025, 17(4), 550; https://doi.org/10.3390/v17040550 - 10 Apr 2025
Viewed by 848
Abstract
Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitos that poses a threat to global public health and for which there is an urgent need for widespread access to globally licensed vaccines. Here, we demonstrate that an inactivated CHIKV vaccine (BBV87) protects against [...] Read more.
Chikungunya virus (CHIKV) is an alphavirus transmitted by mosquitos that poses a threat to global public health and for which there is an urgent need for widespread access to globally licensed vaccines. Here, we demonstrate that an inactivated CHIKV vaccine (BBV87) protects against systemic infection with CHIKV in a non-human primate (NHP) challenge model. Groups of five cynomolgus macaques received two doses of 20 µg BBV87 vaccine or saline alone (28 days apart). Twenty-eight days after the second immunisation, all animals were challenged with CHIKV. All controls were productively infected with detectable viremia and pathological responses following challenge, including altered thermoregulation, haematological and cytokine changes. Critically, the histopathological analysis of finger joints identified areas of inflammation in the synovium. By contrast vaccinated macaques had no detectable viremia and none of the pathological changes were reported in control animals. This study demonstrates that a 20 µg dose of BBV87 vaccine confers robust protection in vivo, both on the acquisition of infection and pathology. Full article
(This article belongs to the Special Issue Antiviral Development for Emerging and Re-Emerging Viruses)
Show Figures

Figure 1

22 pages, 6353 KiB  
Article
SwinDAF3D: Pyramid Swin Transformers with Deep Attentive Features for Automated Finger Joint Segmentation in 3D Ultrasound Images for Rheumatoid Arthritis Assessment
by Jianwei Qiu, Grigorios M. Karageorgos, Xiaorui Peng, Soumya Ghose, Zhaoyuan Yang, Aaron Dentinger, Zhanpeng Xu, Janggun Jo, Siddarth Ragupathi, Guan Xu, Nada Abdulaziz, Girish Gandikota, Xueding Wang and David Mills
Bioengineering 2025, 12(4), 390; https://doi.org/10.3390/bioengineering12040390 - 5 Apr 2025
Viewed by 912
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that can cause severe joint damage and functional impairment. Ultrasound imaging has shown promise in providing real-time assessment of synovium inflammation associated with the early stages of RA. Accurate segmentation of the synovium region and [...] Read more.
Rheumatoid arthritis (RA) is a chronic autoimmune disease that can cause severe joint damage and functional impairment. Ultrasound imaging has shown promise in providing real-time assessment of synovium inflammation associated with the early stages of RA. Accurate segmentation of the synovium region and quantification of inflammation-specific imaging biomarkers are crucial for assessing and grading RA. However, automatic segmentation of the synovium in 3D ultrasound is challenging due to ambiguous boundaries, variability in synovium shape, and inhomogeneous intensity distribution. In this work, we introduce a novel network architecture, Swin Transformers with Deep Attentive Features for 3D segmentation (SwinDAF3D), which integrates Swin Transformers into a Deep Attentive Features framework. The developed architecture leverages the hierarchical structure and shifted windows of Swin Transformers to capture rich, multi-scale and attentive contextual information, improving the modeling of long-range dependencies and spatial hierarchies in 3D ultrasound images. In a six-fold cross-validation study with 3D ultrasound images of RA patients’ finger joints (n = 72), our SwinDAF3D model achieved the highest performance with a Dice Score (DSC) of 0.838 ± 0.013, an Intersection over Union (IoU) of 0.719 ± 0.019, and Surface Dice Score (SDSC) of 0.852 ± 0.020, compared to 3D UNet (DSC: 0.742 ± 0.025; IoU: 0.589 ± 0.031; SDSC: 0.661 ± 0.029), DAF3D (DSC: 0.813 ± 0.017; IoU: 0.689 ± 0.022; SDSC: 0.817 ± 0.013), Swin UNETR (DSC: 0.808 ± 0.025; IoU: 0.678 ± 0.032; SDSC: 0.822 ± 0.039), UNETR++ (DSC: 0.810 ± 0.014; IoU: 0.684 ± 0.018; SDSC: 0.829 ± 0.027) and TransUNet (DSC: 0.818 ± 0.013; IoU: 0.692 ± 0.017; SDSC: 0.815 ± 0.016) models. This ablation study demonstrates the effectiveness of combining a Swin Transformers feature pyramid with a deep attention mechanism, improving the segmentation accuracy of the synovium in 3D ultrasound. This advancement shows great promise in enabling more efficient and standardized RA screening using ultrasound imaging. Full article
Show Figures

Figure 1

18 pages, 2586 KiB  
Article
Synovial Fibroblast Extracellular Vesicles Induce Inflammation via Delivering miR-21-5p in Osteoarthritis
by Vasileios Konteles, Ioanna Papathanasiou, Maria Tzetis, Anastasios Kriebardis and Aspasia Tsezou
Cells 2025, 14(7), 519; https://doi.org/10.3390/cells14070519 - 31 Mar 2025
Cited by 2 | Viewed by 820
Abstract
Small extracellular vesicles (sEVs) derived from different osteoarthritic (OA) tissues regulate OA-related biological processes through transporting their content (proteins, miRNAs, etc.) to recipient cells. This study aimed to characterize the miRNA profile of synovial fibroblasts-derived small EVs (FS_OA_sEVs) and investigate their role in [...] Read more.
Small extracellular vesicles (sEVs) derived from different osteoarthritic (OA) tissues regulate OA-related biological processes through transporting their content (proteins, miRNAs, etc.) to recipient cells. This study aimed to characterize the miRNA profile of synovial fibroblasts-derived small EVs (FS_OA_sEVs) and investigate their role in inflammation in chondrocytes. Chondrocytes were isolated from macroscopically preserved and lesioned OA cartilage (C_OAmin and C_OAmax) and synovial fibroblasts from OA synovium. Synovial fibroblasts-derived small EVs (FS_OA_sEVs) were characterized according to ISEV guidelines and used for miRNA profiling and bioinformatics analysis. miR-21-5p was identified as one of the most abundant, and its target genes, such as KLF6, were enriched in OA-related processes including inflammation. Treatment of C_OAmin chondrocytes with FS_OA_sEVs resulted in decreased expression of COL2A1 and ACAN and an increase in catabolic markers MMP-3 and MMP-13. Moreover, C-OAmin receiving FS_OA_sEVs exhibited increased levels of inflammatory markers and miR-21-5p expression, resembling chondrocytes’ phenotype from lesioned OA cartilage, whereas miR-21-5p inhibition reversed their expression of inflammatory markers and miR-21-5p. Compared to C_OA min, C_OAmax chondrocytes exhibited increased miR-21-5p and inflammatory markers expression and decreased KLF6 expression. miR-21-5p inhibition in C_OAmax led to KLF6 upregulation and suppression of inflammatory mediators, whereas co-treatment with siRNA against KLF6 negated this effect, confirming a potential direct regulatory relationship between miR-21-5p and KLF6. Our results provide novel insights into the FS_OA_sEV-mediated inflammation axis, highlighting FS_OA_sEV-derived miR-21-5p as a driver of OA progression via regulating inflammation in chondrocytes. Full article
Show Figures

Figure 1

32 pages, 4746 KiB  
Article
Loosening the Lid on Shoulder Osteoarthritis: How the Transcriptome and Metabolic Syndrome Correlate with End-Stage Disease
by Samuel J. Lynskey, Zihui Ling, Mark Ziemann, Stephen D. Gill, Sean L. McGee and Richard S. Page
Int. J. Mol. Sci. 2025, 26(7), 3145; https://doi.org/10.3390/ijms26073145 - 28 Mar 2025
Viewed by 1151
Abstract
Metabolic syndrome (MetS) associated with Osteoarthritis (OA) is an increasingly recognised entity. Whilst the degenerative pattern in cuff-tear arthropathy (CTA) has been well documented, the biological processes behind primary shoulder OA and CTA remain less understood. This study investigates transcriptomic differences in these [...] Read more.
Metabolic syndrome (MetS) associated with Osteoarthritis (OA) is an increasingly recognised entity. Whilst the degenerative pattern in cuff-tear arthropathy (CTA) has been well documented, the biological processes behind primary shoulder OA and CTA remain less understood. This study investigates transcriptomic differences in these conditions, alongside the impact of MetS in patients undergoing total shoulder replacement. In a multi-centre study, 20 OA patients undergoing total shoulder replacement were included based on specific treatment indications for OA and cuff-tear arthropathy as well as 25 patients undergoing rotator cuff repair (RCR) as a comparator group. Tissues from subchondral bone, capsule (OA and RCR), and synovium were biopsied, and RNA sequencing was performed using Illumina platforms. Differential gene expression was conducted using DESeq2, adjusting for demographic factors, followed by pathway enrichment using the mitch package. Gene expressions in CTA and primary OA was differentially affected. CTA showed mitochondrial dysfunction, GATD3A downregulation, and increased cartilage degradation, while primary OA was marked by upregulated inflammatory and catabolic pathways. The effect of MetS on these pathologies was further shown. MetS further disrupted WNT/β-catenin signalling in CTA, and in OA. Genes such as ACAN, PANX3, CLU, and VAT1L were upregulated, highlighting potential biomarkers for early OA detection. This transcriptomic analysis reveals key differences between end-stage CTA and primary glenohumeral OA. CTA shows heightened metabolic/protein synthesis activity with less immune-driven inflammation. Under MetS, mitochondrial dysfunction (including GATD3A downregulation) and altered Wnt/β-catenin signalling intensify cartilage and bone damage. In contrast, primary OA features strong complement activation, inflammatory gene expression, and collagen remodelling. MetS worsens both conditions via oxidative stress, advanced glycation end products, and ECM disruption—particularly, increased CS/DS degradation. These distinctions support targeted treatments, from antioxidants and Wnt modulators to aggrecanase inhibitors or clusterin augmentation. Addressing specific molecular disruptions, especially those amplified by MetS, may preserve shoulder function, delay surgical intervention, and improve long-term patient outcomes. Full article
Show Figures

Figure 1

39 pages, 2670 KiB  
Review
HFE-Related Hemochromatosis May Be a Primary Kupffer Cell Disease
by Elias Kouroumalis, Ioannis Tsomidis and Argyro Voumvouraki
Biomedicines 2025, 13(3), 683; https://doi.org/10.3390/biomedicines13030683 - 10 Mar 2025
Viewed by 1341
Abstract
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The [...] Read more.
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The most common genetic disease of iron deposition is classic hemochromatosis (HH) type 1, which is caused by mutations of HFE. Other rare forms of HH include type 2A with mutations at the gene hemojuvelin or type 2B with mutations in HAMP that encodes hepcidin. HH type 3, is caused by mutations of the gene that encodes transferrin receptor 2. Mutations of SLC40A1 which encodes ferroportin cause either HH type 4A or HH type 4B. In the present review, an overview of iron metabolism including absorption by enterocytes and regulation of iron by macrophages, liver sinusoidal endothelial cells (LSECs) and hepatocyte production of hepcidin is presented. Hereditary Hemochromatosis and the current pathogenetic model are analyzed. Finally, a new hypothesis based on published data was suggested. The Kupffer cell is the primary defect in HFE hemochromatosis (and possibly in types 2 and 3), while the hepcidin-relative deficiency, which is the common underlying abnormality in the three types of HH, is a secondary consequence. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Back to TopTop