1
|
Morici L, Allémann E, Rodríguez-Nogales C, Jordan O. Cartilage-targeted drug nanocarriers for osteoarthritis therapy. Int J Pharm 2024; 666:124843. [PMID: 39424088 DOI: 10.1016/j.ijpharm.2024.124843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Osteoarthritis (OA) is a joint disease common worldwide. Currently, no disease-modifying osteoarthritis drugs (DMOADs) have successfully passed clinical trials, often due to a lack of cartilage penetration. Thus, targeting the extracellular matrix (ECM) is a major priority. The design of cartilage-targeting drug delivery systems (DDSs) for intra-articular administration requires consideration of the physicochemical properties of articular cartilage, such as its porosity and negative fixed charge. Various positively charged biomaterials such as polyaminoacids, proteins, polymers, and lipids can be used as DDSs to enhance cartilage penetration. Cationic nanocarriers interact electrostatically with anionic glycosaminoglycans of the ECM, ensuring passive cartilage-targeting penetration and prolonged retention. Active targeting strategies involve DDSs surface decoration using antibodies or peptides with a strong affinity for collagen II and chondrocytes in the cartilage. This review presents all the relevant bio-physicochemical properties of healthy and OA cartilages, as well as state-of-the-art intra-articular cartilage-targeted DDSs, intending to better understand the recent advances in the application of cartilage-targeting delivery systems for OA therapy.
Collapse
Affiliation(s)
- Luca Morici
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Carlos Rodríguez-Nogales
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland.
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1206 Geneva, Switzerland.
| |
Collapse
|
2
|
Zou L, Yang K, Yu Y, Wang C, Zhao J, Lu C, He D. Analysis of joint protein expression profile in anterior disc displacement of TMJ with or without OA. Oral Dis 2024; 30:4463-4482. [PMID: 38251222 DOI: 10.1111/odi.14871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/09/2023] [Accepted: 01/07/2024] [Indexed: 01/23/2024]
Abstract
OBJECTIVE Anterior disc displacement (ADD) is a common clinical issue and may cause osteoarthritis (OA). However, the research of protein changes in synovial fluid as disease development marker and potential treatment clue is still insufficient. MATERIALS AND METHODS We conducted the high-resolution mass spectrometry (MS) of synovial fluid collected from 60 patients with normal disk position to ADD and ADD with osteoarthritis (OA). The proteins with significant changes among the 3 groups were analyzed by biological information and further validated by in primary rat condyle chondrocytes and OA animal model. RESULTS FGL2, THBS4, TNC, FN1, OMD etc. were significantly increased in ADD without OA (p < 0.05), which reflected the active extracellular matrix and collagen metabolism. FGFR1, FBLN2, GRB2 etc. were significantly increased in ADD with OA group (p < 0.05), which revealed an association with apoptosis and ferroptosis. Proteins such as P4HB, CBLN4, FHL1, VIM continuously increase in the whole disease progress (p < 0.05). Both the in vitro and in vivo results are consistent with protein changes detected in MS profile. CONCLUSION This study firstly provides the expression changes of proteins from normal disc condyle relationship toward ADD with OA, which can be selected and studied further as disease progress marker and potential treatment targets.
Collapse
Affiliation(s)
- Luxiang Zou
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| | - Kaiwen Yang
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| | - Yeke Yu
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| | - Chuyao Wang
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| | - Jieyun Zhao
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| | - Chuan Lu
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| | - Dongmei He
- Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- National Clinical Research Center of Stomatology, Shanghai, China
| |
Collapse
|
3
|
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 2024:S1063-4584(24)01397-9. [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] [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
|
4
|
Arruda AL, Katsoula G, Chen S, Reimann E, Kreitmaier P, Zeggini E. The Genetics and Functional Genomics of Osteoarthritis. Annu Rev Genomics Hum Genet 2024; 25:239-257. [PMID: 39190913 DOI: 10.1146/annurev-genom-010423-095636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Osteoarthritis is the most prevalent whole-joint degenerative disorder, and is characterized by the degradation of articular cartilage and the underlying bone structures. Almost 600 million people are affected by osteoarthritis worldwide. No curative treatments are available, and management strategies focus mostly on pain relief. Here, we provide a comprehensive overview of the available human genetic and functional genomics studies for osteoarthritis to date and delineate how these studies have helped shed light on disease etiopathology. We highlight genetic discoveries from genome-wide association studies and provide a detailed overview of molecular-level investigations in osteoarthritis tissues, including methylation-, transcriptomics-, and proteomics-level analyses. We review how functional genomics data from different molecular levels have helped to prioritize effector genes that can be used as drug targets or drug-repurposing opportunities. Finally, we discuss future directions with the potential to drive a step change in osteoarthritis research.
Collapse
Affiliation(s)
- Ana Luiza Arruda
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- Munich School for Data Science, Helmholtz Munich, Neuherberg, Germany
| | - Georgia Katsoula
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Shibo Chen
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
| | - Ene Reimann
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Peter Kreitmaier
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| |
Collapse
|
5
|
Rydén M, Sjögren A, Önnerfjord P, Turkiewicz A, Tjörnstrand J, Englund M, Ali N. Exploring the Early Molecular Pathogenesis of Osteoarthritis Using Differential Network Analysis of Human Synovial Fluid. Mol Cell Proteomics 2024; 23:100785. [PMID: 38750696 PMCID: PMC11252953 DOI: 10.1016/j.mcpro.2024.100785] [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: 08/15/2023] [Revised: 04/17/2024] [Accepted: 05/11/2024] [Indexed: 06/23/2024] Open
Abstract
The molecular mechanisms that drive the onset and development of osteoarthritis (OA) remain largely unknown. In this exploratory study, we used a proteomic platform (SOMAscan assay) to measure the relative abundance of more than 6000 proteins in synovial fluid (SF) from knees of human donors with healthy or mildly degenerated tissues, and knees with late-stage OA from patients undergoing knee replacement surgery. Using a linear mixed effects model, we estimated the differential abundance of 6251 proteins between the three groups. We found 583 proteins upregulated in the late-stage OA, including MMP1, collagenase 3 and interleukin-6. Further, we selected 760 proteins (800 aptamers) based on absolute fold changes between the healthy and mild degeneration groups. To those, we applied Gaussian Graphical Models (GGMs) to analyze the conditional dependence of proteins and to identify key proteins and subnetworks involved in early OA pathogenesis. After regularization and stability selection, we identified 102 proteins involved in GGM networks. Notably, network complexity was lost in the protein graph for mild degeneration when compared to controls, suggesting a disruption in the regular protein interplay. Furthermore, among our main findings were several downregulated (in mild degeneration versus healthy) proteins with unique interactions in the healthy group, one of which, SLCO5A1, has not previously been associated with OA. Our results suggest that this protein is important for healthy joint function. Further, our data suggests that SF proteomics, combined with GGMs, can reveal novel insights into the molecular pathogenesis and identification of biomarker candidates for early-stage OA.
Collapse
Affiliation(s)
- Martin Rydén
- Clinical Epidemiology Unit, Department of Clinical Sciences Lund, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Amanda Sjögren
- Clinical Epidemiology Unit, Department of Clinical Sciences Lund, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden.
| | - Patrik Önnerfjord
- Department of Clinical Sciences Lund, Rheumatology, Rheumatology and Molecular Skeletal Biology, Faculty of Medicine, Lund University, Lund, Sweden
| | - Aleksandra Turkiewicz
- Clinical Epidemiology Unit, Department of Clinical Sciences Lund, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jon Tjörnstrand
- Department of Orthopaedics, Skåne University Hospital, Lund, Sweden
| | - Martin Englund
- Clinical Epidemiology Unit, Department of Clinical Sciences Lund, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Neserin Ali
- Clinical Epidemiology Unit, Department of Clinical Sciences Lund, Orthopedics, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Kraus VB, Sun S, Reed A, Soderblom EJ, Moseley MA, Zhou K, Jain V, Arden N, Li YJ. An osteoarthritis pathophysiological continuum revealed by molecular biomarkers. SCIENCE ADVANCES 2024; 10:eadj6814. [PMID: 38669329 PMCID: PMC11051665 DOI: 10.1126/sciadv.adj6814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
We aimed to identify serum biomarkers that predict knee osteoarthritis (OA) before the appearance of radiographic abnormalities in a cohort of 200 women. As few as six serum peptides, corresponding to six proteins, reached AUC 77% probability to distinguish those who developed OA from age-matched individuals who did not develop OA up to 8 years later. Prediction based on these blood biomarkers was superior to traditional prediction based on age and BMI (AUC 51%) or knee pain (AUC 57%). These results identify a prolonged molecular derangement of joint tissue before the onset of radiographic OA abnormalities consistent with an unresolved acute phase response. Among all 24 protein biomarkers predicting incident knee OA, the majority (58%) also predicted knee OA progression, revealing the existence of a pathophysiological "OA continuum" based on considerable similarity in the molecular pathophysiology of the progression to incident OA and the progression of established 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
| | - Shuming Sun
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Alexander Reed
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Erik J. Soderblom
- Duke Proteomics and Metabolomics Core Facility, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - M. Arthur Moseley
- Duke Proteomics and Metabolomics Core Facility, Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| | - Kaile Zhou
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Vaibhav Jain
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Nigel Arden
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, UK
| | - Yi-Ju Li
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| |
Collapse
|
7
|
Sowislok A, Busch A, Kaschani F, Kaiser M, Jäger M. Differences in the Synovial Fluid Proteome of Septic and Aseptic Implant Failure. Antibiotics (Basel) 2024; 13:346. [PMID: 38667022 PMCID: PMC11047638 DOI: 10.3390/antibiotics13040346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/25/2024] [Accepted: 04/07/2024] [Indexed: 04/29/2024] Open
Abstract
Implant loosening is a severe complication after total joint replacement. Here, differential diagnosis between septic and aseptic cases is crucial for further surgical treatment, but low-grade periprosthetic joint infections (PJIs) in particular remain a challenge. In this study, we analyzed the synovial fluid proteome of 21 patients undergoing revision surgery for septic (eight cases) or aseptic (thirteen cases) implant failure using LC-MS/MS to identify potential new biomarkers as future diagnostic tools. Staphylococci were found in four cases, Streptococci in two cases, Serratia marcescens and Cutibacterium acnes in one case. Proteomic analysis of the synovial fluid resulted in the identification of 515 different proteins based on at least two peptides. A statistical comparison revealed 37 differentially abundant proteins (p < 0.05), of which 17 proteins (46%) showed a higher abundance in the septic group. The proteins with the highest fold change included the known marker proteins c-reactive protein (7.57-fold) and the calprotectin components protein S100-A8 (4.41-fold) and protein S100-A9 (3.1-fold). However, the protein with the highest fold change was leucine-rich alpha-2-glycoprotein 1 (LRG1) (9.07-fold), a currently discussed new biomarker for inflammatory diseases. Elevated LRG1 levels could facilitate the diagnosis of PJI in the future, but their significance needs to be further investigated.
Collapse
Affiliation(s)
- Andrea Sowislok
- Chair of Orthopedics and Trauma Surgery, University of Duisburg-Essen, 45147 Essen, Germany;
| | - André Busch
- Department of Orthopedics, Trauma and Reconstructive Surgery, Katholisches Klinikum Essen Philippus, 45355 Essen, Germany;
| | - Farnusch Kaschani
- Analytics Core Facility Essen (ACE), ZMB, Chemical Biology, University of Duisburg-Essen, 45141 Essen, Germany;
| | - Markus Kaiser
- Chemical Biology, Faculty of Biology, University of Duisburg-Essen, 45141 Essen, Germany;
| | - Marcus Jäger
- Chair of Orthopedics and Trauma Surgery, University of Duisburg-Essen, 45147 Essen, Germany;
- Department of Orthopedics, Trauma and Reconstructive Surgery, Katholisches Klinikum Essen Philippus, 45355 Essen, Germany;
- Department of Orthopedics, Trauma and Reconstructive Surgery, St. Marien Hospital Mülheim a. d. Ruhr, 45468 Mülheim, Germany
| |
Collapse
|
8
|
Rai MF, Collins KH, Lang A, Maerz T, Geurts J, Ruiz-Romero C, June RK, Ramos Y, Rice SJ, Ali SA, Pastrello C, Jurisica I, Thomas Appleton C, Rockel JS, Kapoor M. Three decades of advancements in osteoarthritis research: insights from transcriptomic, proteomic, and metabolomic studies. Osteoarthritis Cartilage 2024; 32:385-397. [PMID: 38049029 DOI: 10.1016/j.joca.2023.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a complex disease involving contributions from both local joint tissues and systemic sources. Patient characteristics, encompassing sociodemographic and clinical variables, are intricately linked with OA rendering its understanding challenging. Technological advancements have allowed for a comprehensive analysis of transcripts, proteomes and metabolomes in OA tissues/fluids through omic analyses. The objective of this review is to highlight the advancements achieved by omic studies in enhancing our understanding of OA pathogenesis over the last three decades. DESIGN We conducted an extensive literature search focusing on transcriptomics, proteomics and metabolomics within the context of OA. Specifically, we explore how these technologies have identified individual transcripts, proteins, and metabolites, as well as distinctive endotype signatures from various body tissues or fluids of OA patients, including insights at the single-cell level, to advance our understanding of this highly complex disease. RESULTS Omic studies reveal the description of numerous individual molecules and molecular patterns within OA-associated tissues and fluids. This includes the identification of specific cell (sub)types and associated pathways that contribute to disease mechanisms. However, there remains a necessity to further advance these technologies to delineate the spatial organization of cellular subtypes and molecular patterns within OA-afflicted tissues. CONCLUSIONS Leveraging a multi-omics approach that integrates datasets from diverse molecular detection technologies, combined with patients' clinical and sociodemographic features, and molecular and regulatory networks, holds promise for identifying unique patient endophenotypes. This holistic approach can illuminate the heterogeneity among OA patients and, in turn, facilitate the development of tailored therapeutic interventions.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Kelsey H Collins
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Annemarie Lang
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jeroen Geurts
- Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica, INIBIC -Hospital Universitario A Coruña, SERGAS, Spain
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, USA
| | - Yolande Ramos
- Dept. Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Sarah J Rice
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shabana Amanda Ali
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada; Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - C Thomas Appleton
- Department of Medicine, University of Western Ontario, London, ON, Canada
| | - Jason S Rockel
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, UHN, Toronto, ON, Canada.
| |
Collapse
|
9
|
Struglics A. Biomarkers have to make sense. Osteoarthritis Cartilage 2024; 32:232-233. [PMID: 38128875 DOI: 10.1016/j.joca.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Affiliation(s)
- André Struglics
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, BMC C12, Klinikgatan 28, SE-221 84 Lund, Sweden.
| |
Collapse
|
10
|
Kraus VB, Reed A, Soderblom EJ, Golightly YM, Nelson AE, Li YJ. Serum proteomic biomarkers diagnostic of knee osteoarthritis. Osteoarthritis Cartilage 2024; 32:329-337. [PMID: 37734705 PMCID: PMC10925913 DOI: 10.1016/j.joca.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/03/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
OBJECTIVE To better understand the pathogenesis of knee osteoarthritis (OA) through identification of serum diagnostics. DESIGN We conducted multiple reaction monitoring mass spectrometry analysis of 107 peptides in baseline sera of two cohorts: the Foundation for National Institutes of Health (NIH) (n = 596 Kellgren-Lawrence (KL) grade 1-3 knee OA participants); and the Johnston County Osteoarthritis Project (n = 127 multi-joint controls free of radiographic OA of the hands, hips, knees (bilateral KL=0), and spine). Data were split into (70%) training and (30%) testing sets. Diagnostic peptide and clinical data predictors were selected by random forest (RF); selection was based on association (p < 0.05) with OA status in multivariable logistic regression models. Model performance was based on area under the curve (AUC) of receiver operating characteristic (ROC) and precision-recall (PR) curves. RESULTS RF selected 23 peptides (19 proteins) and body mass index (BMI) as diagnostic of OA. BMI weakly diagnosed OA (ROC-AUC 0.57, PR-AUC 0.812) and only symptomatic OA cases. ACTG was the strongest univariable predictor (ROC-AUC 0.705, PR-AUC 0.897). The final model (8 serum peptides) was highly diagnostic (ROC-AUC 0.833, 95% confidence interval [CI] 0.751, 0.905; PR-AUC 0.929, 95% CI 0.876, 0.973) in the testing set and equally diagnostic of non-symptomatic and symptomatic cases (AUCs 0.830-0.835), and not significantly improved with addition of BMI. The STRING database predicted multiple high confidence interactions of the 19 diagnostic OA proteins. CONCLUSIONS No more than 8 serum protein biomarkers were required to discriminate knee OA from non-OA. These biomarkers lend strong support to the involvement and cross-talk of complement and coagulation pathways in the development of OA.
Collapse
Affiliation(s)
- Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University, Durham, NC, United States; Department of Medicine, Duke University, Durham, NC, United States.
| | - Alexander Reed
- Duke Molecular Physiology Institute, Duke University, Durham, NC, United States
| | - Erik J Soderblom
- Duke Proteomics and Metabolomics Core Facility, Center for Genomic and Computational Biology, Duke University, Durham, NC, United States
| | - Yvonne M Golightly
- College of Allied Health Professions, University of Nebraska Medical Center, Omaha, NE, United States; Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Amanda E Nelson
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yi-Ju Li
- Duke Molecular Physiology Institute, Duke University, Durham, NC, United States; Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, United States
| |
Collapse
|
11
|
Zhang Y, Li Z, Chen C, Wei W, Li Z, Huang H, Zhou H, He W, Xia J, Li B, Yang Y. S100A12 is involved in the pathology of osteoarthritis by promoting M1 macrophage polarization via the NF-κB pathway. Connect Tissue Res 2024; 65:133-145. [PMID: 38492210 DOI: 10.1080/03008207.2024.2310852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/23/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease that affects millions worldwide. Synovitis and macrophage polarization are important factors in the development of OA. However, the specific components of synovial fluid (SF) responsible for promoting macrophage polarization remain unclear. METHODS Semi-quantitative antibody arrays were used to outline the proteome of SF. Differential expression analysis and GO/KEGG were performed on the obtained data. Immunohistochemistry and ELISA were used to investigate the relationship between SF S100A12 levels and synovitis levels in clinalclinical samples. In vitro cell experiments were conducted to investigate the effect of S100A12 on macrophage polarization. Public databases were utilized to predict and construct an S100A12-centered lncRNA-miRNA-mRNA competing endogenous RNA network, which was preliminarily validated using GEO datasets. RESULTS The study outlines the protein profile in OA and non-OA SF. The results showed that the S100A12 level was significantly increased in OA SF and inflammatory chondrocytes. The OA synovium had more severe synovitis and higher levels of S100A12 than non-OA synovium. Exogenous S100A12 upregulated the levels of M1 markers and phosphorylated p65 and promoted p65 nuclear translocation, while pretreatment with BAY 11-7082 reversed these changes. It was also discovered that LINC00894 was upregulated in OA and significantly correlated with S100A12, potentially regulating S100A12 expression by acting as a miRNA sponge. CONCLUSIONS This study demonstrated that S100A12 promotes M1 macrophage polarization through the NF-κB pathway, and found that LINC00894 has the potential to regulate the expression of S100A12 as a therapeutic approach.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zihua Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cheng Chen
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wang Wei
- The First Department of Orthopaedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No.157, Xiwu Road, Xi'an, Shaanxi, China
| | - Zhendong Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haichao Zhou
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenbao He
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang Xia
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bing Li
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunfeng Yang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Al Turkestani N, Li T, Bianchi J, Gurgel M, Prieto J, Shah H, Benavides E, Soki F, Mishina Y, Fontana M, Rao A, Zhu H, Cevidanes L. A comprehensive patient-specific prediction model for temporomandibular joint osteoarthritis progression. Proc Natl Acad Sci U S A 2024; 121:e2306132121. [PMID: 38346188 PMCID: PMC10895339 DOI: 10.1073/pnas.2306132121] [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: 04/17/2023] [Accepted: 01/03/2024] [Indexed: 02/15/2024] Open
Abstract
Temporomandibular joint osteoarthritis (TMJ OA) is a prevalent degenerative disease characterized by chronic pain and impaired jaw function. The complexity of TMJ OA has hindered the development of prognostic tools, posing a significant challenge in timely, patient-specific management. Addressing this gap, our research employs a comprehensive, multidimensional approach to advance TMJ OA prognostication. We conducted a prospective study with 106 subjects, 74 of whom were followed up after 2 to 3 y of conservative treatment. Central to our methodology is the development of an innovative, open-source predictive modeling framework, the Ensemble via Hierarchical Predictions through Nested cross-validation tool (EHPN). This framework synergistically integrates 18 feature selection, statistical, and machine learning methods to yield an accuracy of 0.87, with an area under the ROC curve of 0.72 and an F1 score of 0.82. Our study, beyond technical advancements, emphasizes the global impact of TMJ OA, recognizing its unique demographic occurrence. We highlight key factors influencing TMJ OA progression. Using SHAP analysis, we identified personalized prognostic predictors: lower values of headache, lower back pain, restless sleep, condyle high gray level-GL-run emphasis, articular fossa GL nonuniformity, and long-run low GL emphasis; and higher values of superior joint space, mouth opening, saliva Vascular-endothelium-growth-factor, Matrix-metalloproteinase-7, serum Epithelial-neutrophil-activating-peptide, and age indicate recovery likelihood. Our multidimensional and multimodal EHPN tool enhances clinicians' decision-making, offering a transformative translational infrastructure. The EHPN model stands as a significant contribution to precision medicine, offering a paradigm shift in the management of temporomandibular disorders and potentially influencing broader applications in personalized healthcare.
Collapse
Affiliation(s)
- Najla Al Turkestani
- Department of Restorative Dentistry, Faculty of Dentistry, King Abdulaziz University, Jeddah21589, Saudi Arabia
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Tengfei Li
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Jonas Bianchi
- Department of Orthodontics, University of the Pacific, Arthur A. Dugoni School of Dentistry, San Francisco, CA94103
| | - Marcela Gurgel
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Juan Prieto
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Hina Shah
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Erika Benavides
- Department of Periodontics & Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Fabiana Soki
- Department of Periodontics & Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Margherita Fontana
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Arvind Rao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI48109
- Department of Computational Medicine & Bioinformatics, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| | - Hongtu Zhu
- Department of Radiology and Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Lucia Cevidanes
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
13
|
Fan X, Sun AR, Young RSE, Afara IO, Hamilton BR, Ong LJY, Crawford R, Prasadam I. Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications. Bone Res 2024; 12:7. [PMID: 38311627 PMCID: PMC10838951 DOI: 10.1038/s41413-023-00304-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 02/06/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.
Collapse
Affiliation(s)
- Xiwei Fan
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia Rujia Sun
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Reuben S E Young
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, QLD, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Isaac O Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- School of Electrical Engineering and Computer Science, Faculty of Engineering, Architecture and Information Technology, University of Queensland, Brisbane, QLD, Australia
| | - Brett R Hamilton
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD, Australia
| | - Louis Jun Ye Ong
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ross Crawford
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia.
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
14
|
Zhang Y, Li G, Wang J, Zhou F, Ren X, Su J. Small Joint Organoids 3D Bioprinting: Construction Strategy and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2302506. [PMID: 37814373 DOI: 10.1002/smll.202302506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 09/28/2023] [Indexed: 10/11/2023]
Abstract
Osteoarthritis (OA) is a chronic disease that causes pain and disability in adults, affecting ≈300 million people worldwide. It is caused by damage to cartilage, including cellular inflammation and destruction of the extracellular matrix (ECM), leading to limited self-repairing ability due to the lack of blood vessels and nerves in the cartilage tissue. Organoid technology has emerged as a promising approach for cartilage repair, but constructing joint organoids with their complex structures and special mechanisms is still challenging. To overcome these boundaries, 3D bioprinting technology allows for the precise design of physiologically relevant joint organoids, including shape, structure, mechanical properties, cellular arrangement, and biological cues to mimic natural joint tissue. In this review, the authors will introduce the biological structure of joint tissues, summarize key procedures in 3D bioprinting for cartilage repair, and propose strategies for constructing joint organoids using 3D bioprinting. The authors also discuss the challenges of using joint organoids' approaches and perspectives on their future applications, opening opportunities to model joint tissues and response to joint disease treatment.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Trauma Orthopedics, Zhongye Hospital, Shanghai, 200941, China
| | - Jian Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Fengjin Zhou
- Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China
| | - Xiaoxiang Ren
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Musculoskeletal Organoid Research Center, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
15
|
Antoniadis A, Wegrzyn J, Omoumi P, Loisay L, Hügle T, Geurts J. Elevated secretion of pro-collagen I-alpha and vascular endothelial growth factor as biomarkers of acetabular labrum degeneration and calcification in hip osteoarthritis: An explant study. J Orthop Translat 2024; 44:19-25. [PMID: 38179125 PMCID: PMC10765489 DOI: 10.1016/j.jot.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/29/2023] [Accepted: 08/23/2023] [Indexed: 01/06/2024] Open
Abstract
Background Hip osteoarthritis (OA) involves structural degeneration of different joint compartments, including femoral head cartilage, periarticular ligaments and the acetabular labrum. However, the molecular mechanisms underlying labrum degeneration in hip OA remain poorly understood. Aim To assess secretion of putative biomarkers for OA from explanted human labrum tissues under basal and inflammatory conditions and to determine whether these could differentiate between OA and calcification status compared to fracture controls. Methods Intact labrum specimens were collected from patients undergoing joint arthroplasty for primary hip OA (n = 15, mean age 70) or non-OA femoral neck fracture (n = 5, mean age 64). Tissues were dissected in equal-sized samples and explanted for one week. To mimic activation of inflammatory signaling by endogenous damage-associated molecular patterns (DAMP) tissue were stimulated with a toll-like receptor 4 (TLR4) agonist (1 μg/mL LPS). The involvement of transforming growth factor-beta (TGF-beta) signaling was evaluated by treatment with a TGF-beta type 1 receptor inhibitor (10 μM SB-505124). Secretion of aggrecan (ACAN), pro-collagen-I alpha (Pro-Col-Iα), cartilage oligomeric matrix protein (COMP), interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) was assessed by enzyme-linked immunosorbent assay (ELISA). Labrum calcification was evaluated by 3D whole mount fluorescent microscopy of ethyl cinnamate-based optically cleared tissues stained with Alcian blue/Alizarin red. Results Whole mount microscopy revealed non-OA fracture controls were non-calcified, whereas six OA labra (40%) were partially calcified or ossified. Basal secretion of Pro-Col-Iα and VEGF was increased four-fold in OA versus non-OA labra. Pro-Col-Iα levels were correlated with those of VEGF (r = 0.65) and COMP (r = 0.54). Stimulation of DAMP signaling through TLR4 affected secretion of IL-6, VEGF, COMP and Pro-Col-Iα, with distinct responses between non-OA and OA tissues. Inhibition of TGF-beta signaling specifically reduced elevated secretion of Pro-Col- Iα and VEGF in calcified OA labrum. Conclusions Secretion of the putative OA biomarkers Pro-Col-Iα and VEGF is elevated in degenerated human acetabular labrum and may serve as indicators of OA and calcification status. Secretion of both factors was partially regulated by TGF-beta signaling in calcified OA labrum tissues.The Translational potential of this article:Our findings suggest that a biomarker panel consisting of Pro-Col-Iα/VEGF/COMP may be valuable for assessing subradiographic labrum degeneration and calcification in hip OA. Targeting TGF-beta signaling may offer a means to reduce vascular invasion and fibrosis in acetabular labrum tissue.
Collapse
Affiliation(s)
- Alexander Antoniadis
- Department of Orthopedics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Julien Wegrzyn
- Department of Orthopedics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Patrick Omoumi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Léa Loisay
- Department of Rheumatology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Thomas Hügle
- Department of Rheumatology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Jeroen Geurts
- Department of Rheumatology, Lausanne University Hospital and University of Lausanne, Switzerland
| |
Collapse
|
16
|
Paz-González R, Turkiewicz A, Ali N, Ruiz-Romero C, Blanco FJ, Englund M, Önnerfjord P. Proteomic profiling of human menisci from mild joint degeneration and end-stage osteoarthritis versus healthy controls. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100417. [PMID: 38098679 PMCID: PMC10720269 DOI: 10.1016/j.ocarto.2023.100417] [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: 06/02/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Objective To gain new insight into the molecular changes of the meniscus by comparing the proteome profiles of healthy controls with mild degeneration and end-stage osteoarthritis (OA). Method We obtained tissue plugs from lateral and medial menisci of 37 individuals (central part of the posterior horn) classified as healthy (n = 12), mild signs of joint damage (n = 13) and end-stage OA (n = 12). The protein profile was analysed by nano-liquid chromatography-mass spectrometry using data-independent acquisition and quantified by Spectronaut. Linear-mixed effects modelling was applied to extract the between-group comparisons. Results A similar protein profile was observed for the mild group as compared to healthy controls while the most different group was end-stage OA mainly for the medial compartment. When a pattern of gradual change in protein levels from healthy to end-stage OA was required, a 42-proteins panel was identified, suggesting a potential role in OA development. The levels of QSOX1 were lower and G6PD higher in the mild group following the proposed protein abundance pattern. Qualitative protein changes suggest lower levels of CYTL1 as a potential biomarker of early joint degradation. Conclusion For future targeted proteomic approaches, we propose a candidate panel of 42 proteins based on gradually altered meniscal posterior horn protein abundance patterns associated with joint degradation.
Collapse
Affiliation(s)
- Rocío Paz-González
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica. INIBIC-Hospital Universitario A Coruña, SERGAS, 15006, A Coruña, Spain
| | - Aleksandra Turkiewicz
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Neserin Ali
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica. INIBIC-Hospital Universitario A Coruña, SERGAS, 15006, A Coruña, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Francisco J. Blanco
- Grupo de Investigación de Reumatología (GIR), Unidad de Proteómica. INIBIC-Hospital Universitario A Coruña, SERGAS, 15006, A Coruña, Spain
- Grupo de Reumatología y Salud, Departamento de Fisioterapia y Medicina. Centro de investigaciones Avanzadas (CICA), Universidad de A Coruña, A Coruña, Spain
| | - Martin Englund
- Clinical Epidemiology Unit, Orthopedics, Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Zhang Y, Xie J, Wen S, Cao P, Xiao W, Zhu J, Li S, Wang Z, Cen H, Zhu Z, Ding C, Ruan G. Evaluating the causal effect of circulating proteome on the risk of osteoarthritis-related traits. Ann Rheum Dis 2023; 82:1606-1617. [PMID: 37595989 DOI: 10.1136/ard-2023-224459] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/02/2023] [Indexed: 08/20/2023]
Abstract
OBJECTIVES This study aims to identify circulating proteins that are causally associated with osteoarthritis (OA)-related traits through Mendelian randomisation (MR)-based analytical framework. METHODS Large-scale two-sample MR was employed to estimate the effects of thousands of plasma proteins on 12 OA-related traits. Additional analyses including Bayesian colocalisation, Steiger filtering analysis, assessment of protein-altering variants and mapping expression quantitative trait loci to protein quantitative trait loci were performed to investigate the reliability of the MR findings; protein-protein interaction, pathway enrichment analysis and evaluation of drug targets were conducted to deepen the understanding and identify potential therapeutic targets of OA. RESULTS Dozens of circulating proteins were identified to have putatively causal effects on OA-related traits, and a majority of these proteins were either drug targets or considered druggable. CONCLUSIONS Through MR analysis, we have identified numerous plasma proteins associated with OA-related traits, shedding light on protein-mediated mechanisms and offering promising therapeutic targets for OA.
Collapse
Affiliation(s)
- Yan Zhang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingyu Xie
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Simin Wen
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Peihua Cao
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wende Xiao
- Department of orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jianwei Zhu
- Department of orthopedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shengfa Li
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiqiang Wang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Han Cen
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhaohua Zhu
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Guangfeng Ruan
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Clinical Research Centre, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
18
|
Jiang P, Liang D, Wang H, Zhou R, Che X, Cong L, Li P, Wang C, Li W, Wei X, Li P. TMT quantitative proteomics reveals key proteins relevant to microRNA-1-mediated regulation in osteoarthritis. Proteome Sci 2023; 21:21. [PMID: 37993861 PMCID: PMC10664301 DOI: 10.1186/s12953-023-00223-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023] Open
Abstract
Osteoarthritis (OA) is the second-commonest arthritis, but pathogenic and regulatory mechanisms underlying OA remain incompletely understood. Here, we aimed to identify the mechanisms associated with microRNA-1 (miR-1) treatment of OA in rodent OA models using a proteomic approach. First, N = 18 Sprague Dawley (SD) rats underwent sham surgery (n = 6) or ACL transection (n = 12), followed at an interval of one week by randomization of the ACL transection group to intra-articular administration of either 50 µL placebo (control group) or miR-1 agomir, a mimic of endogenous miR-1 (experimental group). After allowing for eight weeks of remodeling, articular cartilage tissue was harvested and immunohistochemically stained for the presence of MMP-13. Second, N = 30 Col2a1-cre-ERT2 /GFPf1/fl -RFP-miR-1 transgenic mice were randomized to intra-articular administration of either placebo (control group, N = 15) or tamoxifen, an inducer of miR-1 expression (experimental group, N = 15), before undergoing surgical disruption of the medial meniscus (DMM) after an interval of five days. After allowing for eight weeks of remodeling, articular cartilage tissue was harvested and underwent differential proteomic analysis. Specifically, tandem mass tagging (TMT) quantitative proteomic analysis was employed to identify inter-group differentially-expressed proteins (DEP), and selected DEPs were validated using real-time quantitative polymerase chain reaction (RT-qPCR) technology. Immunohistochemically-detected MMP-13 expression was significantly lower in the experimental rat group, and proteomic analyses of mouse tissue homogenate demonstrated that of 3526 identified proteins, 345 were differentially expressed (relative up- and down-regulation) in the experimental group. Proteins Fn1, P4ha1, P4ha2, Acan, F2, Col3a1, Fga, Rps29, Rpl34, and Fgg were the *top ten most-connected proteins, implying that miR-1 may regulate an expression network involving these proteins. Of these ten proteins, three were selected for further validation by RT-qPCR: the transcript of Fn1, known to be associated with OA, exhibited relative upregulation in the experimental group, whereas the transcripts of P4ha1 and Acan exhibited relative downregulation. These proteins may thus represent key miR-1 targets during OA-regulatory mechanisms, and may provide additional insights regarding therapeutic mechanisms of miR-1 in context of OA.
Collapse
Affiliation(s)
- Pinpin Jiang
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Dan Liang
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Hang Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Raorao Zhou
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xianda Che
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Linlin Cong
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Penghua Li
- Department of Laboratory Medicine, Fenyang Hospital Affiliated to Shanxi Medical University, Fenyang, China
| | - Chunfang Wang
- Laboratory Animal Center of Shanxi Medical University, Taiyuan, China
| | - Wenjin Li
- Department of Stomatology, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaochun Wei
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Pengcui Li
- Department of Orthopaedic Surgery, the Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Bone and Soft Tissue Injury, Second Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
19
|
Copp G, Robb KP, Viswanathan S. Culture-expanded mesenchymal stromal cell therapy: does it work in knee osteoarthritis? A pathway to clinical success. Cell Mol Immunol 2023; 20:626-650. [PMID: 37095295 PMCID: PMC10229578 DOI: 10.1038/s41423-023-01020-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative multifactorial disease with concomitant structural, inflammatory, and metabolic changes that fluctuate in a temporal and patient-specific manner. This complexity has contributed to refractory responses to various treatments. MSCs have shown promise as multimodal therapeutics in mitigating OA symptoms and disease progression. Here, we evaluated 15 randomized controlled clinical trials (RCTs) and 11 nonrandomized RCTs using culture-expanded MSCs in the treatment of knee OA, and we found net positive effects of MSCs on mitigating pain and symptoms (improving function in 12/15 RCTs relative to baseline and in 11/15 RCTs relative to control groups at study endpoints) and on cartilage protection and/or repair (18/21 clinical studies). We examined MSC dose, tissue of origin, and autologous vs. allogeneic origins as well as patient clinical phenotype, endotype, age, sex and level of OA severity as key parameters in parsing MSC clinical effectiveness. The relatively small sample size of 610 patients limited the drawing of definitive conclusions. Nonetheless, we noted trends toward moderate to higher doses of MSCs in select OA patient clinical phenotypes mitigating pain and leading to structural improvements or cartilage preservation. Evidence from preclinical studies is supportive of MSC anti-inflammatory and immunomodulatory effects, but additional investigations on immunomodulatory, chondroprotective and other clinical mechanisms of action are needed. We hypothesize that MSC basal immunomodulatory "fitness" correlates with OA treatment efficacy, but this hypothesis needs to be validated in future studies. We conclude with a roadmap articulating the need to match an OA patient subset defined by molecular endotype and clinical phenotype with basally immunomodulatory "fit" or engineered-to-be-fit-for-OA MSCs in well-designed, data-intensive clinical trials to advance the field.
Collapse
Affiliation(s)
- Griffin Copp
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Kevin P Robb
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada.
- Krembil Research Institute, University Health Network, Toronto, ON, Canada.
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.
- Department of Medicine, Division of Hematology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
20
|
Liu D, Xiao WF, Li YS. The Diagnostic and Prognostic Value of Synovial Fluid Analysis in Joint Diseases. Methods Mol Biol 2023; 2695:295-308. [PMID: 37450127 DOI: 10.1007/978-1-0716-3346-5_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Liquid biopsy is an emergent test method for the diagnosis and prognosis in the clinic. Joint fluid, also known as synovial fluid, contains a variety of bioactive constituents that can be selectively detected and further evaluated in a convenient fashion. Therefore, synovial fluid analysis functions as a specific form of liquid biopsy and plays a vital role in numerous joint diseases. In spite of the component analysis of aspirated synovial fluid beingconsidered as the gold standard for diagnosis of joint infections, biopsy of joint fluid benefits the initial diagnosis and long-term prognosis of degenerative, inflammatory, autoimmune, traumatic, congenital, and even neoplastic joint diseases. The convenience and accuracy for disease evaluation are significantly elevated as a result of the combination of synovial fluid analysis and other novel clinical technologies. In this review, we shed light on the latent role of synovial fluid in the diagnosis and prognosis of articular diseases and proposed future prospects for relevant research in this field.
Collapse
Affiliation(s)
- Di Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen-Feng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Sheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Beier F. The impact of omics research on our understanding of osteoarthritis and future treatments. Curr Opin Rheumatol 2023; 35:55-60. [PMID: 36350386 DOI: 10.1097/bor.0000000000000919] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE OF REVIEW To review recent studies using 'Omics' approaches (genomics, proteomics, metabolomics, single cell analyses) in patient populations and animal models of osteoarthritis (OA), with the goal of identifying disease-modifying mechanisms that could serve as therapeutic and diagnostic targets. RECENT FINDINGS The number of genes, pathways and molecules with potential roles in OA pathogenesis has grown substantially over the last 18 months. Studies have expanded from their traditional focus on cartilage and gene expression to other joint tissues, proteins and metabolites. Single cell approaches provide unprecedented resolution and exciting insights into the heterogeneity of cellular activities in OA. Functional validation and investigation of underlying mechanisms in animal models of OA, in particular genetically engineered mice, link Omics findings to pathophysiology and potential therapeutic applications. SUMMARY Although great progress has been made in the use of Omics approaches to OA, in both animal models and patient samples, much work remains to be done. In addition to filling gaps in data sets not yet existing, integration of data from the various approaches, mechanistic investigations, and linkage of Omics data to patient stratification remain significant challenges.
Collapse
Affiliation(s)
- Frank Beier
- Department of Physiology and Pharmacology, Western University Bone and Joint Institute, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
22
|
Struglics A, Larsson S, Lohmander LS, Swärd P. Technical performance of a proximity extension assay inflammation biomarker panel with synovial fluid. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100293. [DOI: 10.1016/j.ocarto.2022.100293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023] Open
|
23
|
Towards Precision Medicine for Osteoarthritis: Focus on the Synovial Fluid Proteome. Int J Mol Sci 2022; 23:ijms23179731. [PMID: 36077129 PMCID: PMC9455979 DOI: 10.3390/ijms23179731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/29/2022] Open
Abstract
Osteoarthritis (OA) is a joint degenerative disease that most affects old age. The study of proteomics in synovial fluid (SF) has the task of providing additional elements to diagnose and predict the progress of OA. This review aims to identify the most significant biomarkers in the study of OA and to stimulate their routine use. Some of the major components of the ECM, such as proteoglycan aggrecan and decorin, were found considerably reduced in OA. Some biomarkers have proved useful for staging the temporality of OA: Periostin was found to be increased in early OA, while CRTA1 and MMPs were found to be increased in late OA. In its natural attempt at tissue regeneration, Collagen III was found to be increased in early OA while decreased in late OA. Some molecules studied in other areas, such as ZHX3 (oncological marker), LYVE1, and VEGF (lymph and angiogenesis markers), also have been found to be altered in OA. It also has been recorded that alteration of the hormonal pathway, using a dosage of PPAR-γ and RETN, can influence the evolution of OA. IL-1, one of the most investigated biomarkers in OA-SF, is not as reliable as a target of OA in recent studies. The study of biomarkers in SF appears to be, in combination with the clinical and radiological aspects, an additional weapon to address the diagnosis and staging of OA. Therefore, it can guide us more appropriately towards the indication of arthroplasty in patients with OA.
Collapse
|