Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Rev Rheumatol ; 20(5): 272-289, 2024 May.
Article in English | MEDLINE | ID: mdl-38605249

ABSTRACT

Up to 50% of individuals develop post-traumatic osteoarthritis (PTOA) within 10 years following knee-joint injuries such as anterior cruciate ligament rupture or acute meniscal tear. Lower-extremity PTOA prevalence is estimated to account for ≥12% of all symptomatic osteoarthritis (OA), or approximately 5.6 million cases in the USA. With knowledge of the inciting event, it might be possible to 'catch PTOA in the act' with sensitive imaging and soluble biomarkers and thereby prevent OA sequelae by early intervention. Existing biomarker data in the joint-injury literature can provide insights into the pathogenesis and early risk trajectory related to PTOA and can help to elucidate a research agenda for preventing or slowing the onset of PTOA. Non-traumatic OA and PTOA have many clinical, radiological and genetic similarities, and efforts to understand early risk trajectories in PTOA might therefore contribute to the identification and classification of early non-traumatic OA, which is the most prevalent form of OA.


Subject(s)
Biomarkers , Knee Injuries , Humans , Biomarkers/metabolism , Knee Injuries/complications , Knee Injuries/prevention & control , Osteoarthritis, Knee/prevention & control , Osteoarthritis, Knee/etiology , Anterior Cruciate Ligament Injuries/complications , Osteoarthritis/prevention & control , Osteoarthritis/etiology
2.
Osteoarthr Cartil Open ; 6(1): 100425, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38116469

ABSTRACT

Objective: To further validate a serum proteomics panel for predicting radiographic (structural) knee OA progression. Design: Serum peptides were targeted by multiple-reaction-monitoring mass spectrometry in the New York University cohort (n â€‹= â€‹104). Knee OA progression was defined as joint space narrowing ≥1 in the tibiofemoral compartment of one knee per study participant over a 24-month follow-up. The discriminative ability of an 11-peptide panel was evaluated by multivariable logistic regression and area under the receiver operating characteristic curve (AUC), without and with demographic characteristics of age, sex, and body mass index. The association of each peptide with OA progression was assessed by odds ratios (OR) in multivariable logistic regression models adjusted for demographics. Results: The cohort included 46 (44%) knee OA progressors. The panel of 11 peptides alone yielded AUC â€‹= â€‹0.66 (95% CI [0.55, 0.77]) for discriminating progressors from non-progressors; demographic traits alone yielded AUC â€‹= â€‹0.66 (95% CI [0.55, 0.77]). Together the 11 peptides and demographics yielded AUC â€‹= â€‹0.72 (95% CI [0.62, 0.83]). CRAC1 had the highest odds for predicting OA progression (OR 2.014, 95% CI [0.996, 4.296], p â€‹= â€‹0.058). Conclusions: We evaluated a parsimonious serum proteomic panel and found it to be a good discriminator of knee radiographic OA progression from non-progression. Since these biomarkers are quantifiable in serum, they could be deployed relatively easily to provide a simple, cost-effective strategy for identifying and monitoring individuals at high risk of knee OA progression.

3.
Arthritis Rheumatol ; 75(7): 1120-1131, 2023 07.
Article in English | MEDLINE | ID: mdl-36704903

ABSTRACT

OBJECTIVE: To evaluate the association of dipeptidylpeptidase 4 (DPP-4; also known as CD26) with cellular senescence of human cartilage and progression of knee osteoarthritis (OA). METHODS: Articular cartilage sections and chondrocytes were acquired from 35 individuals undergoing total knee replacement for OA to evaluate the following: 1) the association between OA severity and established senescence markers (senescence-associated ß-galactosidase activity and p16), which was quantified using immunohistochemistry and flow cytometry (n = 19 samples); 2) the coexpression of DPP-4 with established senescence markers, which was assessed using flow cytometry; and 3) expression levels of anabolic and catabolic genes, senescence-related genes, and senescence-associated secretory phenotypes in DPP-4+ and DPP-4- cells, which were isolated using fluorescence-activated cell sorting or magnetic-activated cell sorting (n = 16 samples). The concentration of soluble DPP-4 was measured in samples of synovial fluid and samples of plasma from the Prediction of Osteoarthritis Progression cohort and then evaluated for association with the severity of radiographic knee OA at baseline (n = 65 samples) and the progression of structural radiographic OA (n = 57 samples) over a 3-year period. RESULTS: DPP-4 expression was associated with higher senescence-associated ß-galactosidase activity, p16 expression, senescence-related gene and catabolic gene (ADAMTS5, MMP13, IL6, and IL8) expression, higher senescence-associated secretory phenotype secretion, and lower anabolic gene (COL2A1 and ACAN) expression in primary chondrocytes. Synovial fluid DPP-4 concentration was associated with radiographic OA progression (odds ratio 105.32; P = 0.015), proteases (synovial fluid matrix metalloproteinase 1 and matrix metalloproteinase 3), aggrecan degradation (synovial fluid sulfated glycosaminoglycan), indicators of activated macrophages (synovial fluid CD14 and CD163), and inflammation (synovial fluid interleukin-6). CONCLUSION: Our study identifies DPP-4 as a key surface marker in senescent chondrocytes and a predictor of radiographic OA progression.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Humans , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/metabolism , Chondrocytes/metabolism , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/metabolism , Cellular Senescence , Interleukin-6/metabolism , beta-Galactosidase/metabolism
4.
Oxid Med Cell Longev ; 2022: 5503575, 2022.
Article in English | MEDLINE | ID: mdl-35251476

ABSTRACT

Cellular senescence is characterized by cell cycle arrest and senescence-associated secretory phenotypes. Cellular senescence can be caused by various stress stimuli such as DNA damage, oxidative stress, and telomere attrition and is related to several chronic diseases, including atherosclerosis, Alzheimer's disease, and osteoarthritis. Chromobox homolog 4 (CBX4) has been shown to alleviate cellular senescence in human mesenchymal stem cells and is considered a possible target for senomorphic treatment. Here, we explored whether CBX4 expression is associated with replicative senescence in WI-38 fibroblasts, a classic human senescence model system. We also examined whether and how regulation of CBX4 modifies the senescence phenotype and functions as an antisenescence target in WI-38. During the serial culture of the WI-38 primary fibroblast cell line to a senescent state, we found increased expression of senescence markers, including senescence ß-galactosidase (SA-ßgal) activity, protein expression of p16, p21, and DPP4, and decreased proliferation marker EdU; moreover, CBX4 protein expression declined. With knockdown of CBX4, SA-ßgal activity and p16 protein expression increased, and EdU decreased. With the activation of CBX4, SA-ßgal activity, p16, and DPP4 protein decreased. In addition, CBX4 knockdown increased, while CBX4 activation decreased, gene expression of both CDKN2A (encoding the p16 protein) and DPP4. Genes related to DNA damage and cell cycle pathways were regulated by CBX4. These results demonstrate that CBX4 can regulate replicative senescence in a manner consistent with a senomorphic agent.


Subject(s)
Cellular Senescence/genetics , Fibroblasts/metabolism , Ligases/metabolism , Polycomb-Group Proteins/metabolism , Signal Transduction/genetics , Biomarkers/metabolism , Cell Cycle Checkpoints/genetics , Cell Line , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Damage/genetics , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Gene Expression Regulation , Gene Knockdown Techniques/methods , Humans , Ligases/genetics , Oxidative Stress/genetics , Phenotype , Polycomb-Group Proteins/genetics , Transduction, Genetic/methods , beta-Galactosidase/metabolism
5.
Front Immunol ; 12: 758386, 2021.
Article in English | MEDLINE | ID: mdl-34691080

ABSTRACT

Objectives: To identify plasma extracellular vesicles (EVs) associated with radiographic knee osteoarthritis (OA) progression. Methods: EVs of small (SEV), medium (MEV) and large (LEV) sizes from plasma of OA participants (n=30) and healthy controls (HCs, n=22) were profiled for surface markers and cytokine cargo by high-resolution flow cytometry. The concentrations of cytokines within (endo-) and outside (exo-) EVs were quantified by multiplex ELISA. EV associations with knee radiographic OA (rOA) progression were assessed by multivariable linear regression (adjusted for baseline clinical variables of age, gender, BMI and OA severity) and receiver operating characteristic (ROC) curve analysis. Results: Based on integrated mean fluorescence intensity (iMFI), baseline plasma MEVs carrying CD56 (corresponding to natural killer cells) predicted rOA progression with highest area under the ROC curve (AUC) 0.714 among surface markers. Baseline iMFI of TNF-α in LEVs, MEVs and SEVs, and the total endo-EV TNF-α concentration, predicted rOA progression with AUCs 0.688, 0.821, 0.821, 0.665, respectively. In contrast, baseline plasma exo-EV TNF-α (the concentration in the same unit of plasma after EV depletion) did not predict rOA progression (AUC 0.478). Baseline endo-EV IFN-γ and exo-EV IL-6 concentrations were also associated with rOA progression, but had low discriminant capacity (AUCs 0.558 and 0.518, respectively). Conclusions: Plasma EVs carry pro-inflammatory cargo that predict risk of knee rOA progression. These findings suggest that EV-associated TNF-α may be pathogenic in OA. The sequestration of pathogenic TNF-α within EVs may provide an explanation for the lack of success of systemic TNF-α inhibitors in OA trials to date.


Subject(s)
Extracellular Vesicles/immunology , Osteoarthritis, Knee/immunology , Tumor Necrosis Factor-alpha/immunology , Aged , Female , Humans , Male , Middle Aged , Osteoarthritis, Knee/pathology
6.
Arthritis Rheumatol ; 73(1): 89-99, 2021 01.
Article in English | MEDLINE | ID: mdl-32783329

ABSTRACT

OBJECTIVE: To evaluate the role of immune cells and their effector cytokines in the pathogenesis and progression of knee osteoarthritis (OA) in matched OA synovial fluid (SF) and synovial tissue samples. METHODS: Cells from matched samples of synovial tissue and SF acquired from individuals undergoing total knee replacement for OA (n = 39) were characterized for immune cell-associated surface markers and intracellular cytokine expression using polychromatic flow cytometry. Additional individuals with radiographic knee OA (Kellgren/Lawrence severity grades ≥1) who had available etarfolatide (inflammatory cell) imaging (n = 26) or baseline and 3-year data on progression of radiographic knee OA (n = 85) were also assessed. SF cytokine concentrations in all cohorts were evaluated for associations with synovial tissue and SF cell phenotypes and severity of radiographic knee OA. RESULTS: Macrophages (predominant in the synovial tissue, 53% of total cells) and neutrophils (predominant in the SF, 26% of total cells) were the major immune cell populations identified in the OA knee joints, exhibiting expression of or association with transforming growth factor ß1 (TGFß1) and elastase, respectively, in the SF. Expression levels of TGFß1 and elastase were significantly associated with severity of radiographic knee OA. Baseline SF concentrations of TGFß1 and elastase along with radiographic knee OA severity scores were predictive of knee OA progression, with areas under the receiver operating characteristic curves of 0.810 (for TGFß1), 0.806 (for elastase), and 0.846 (for both TGFß1 and elastase combined), with greater stability of prediction when both markers were utilized. CONCLUSION: Our findings demonstrate the hitherto underappreciated role of neutrophils in the sterile inflammatory process and progression of OA. Two soluble mediators, SF elastase and TGFß1, are strong predictors of knee OA progression, reflecting a synergistic role of neutrophil and macrophage populations in the pathogenesis and worsening of OA that could potentially be utilized to identify patients who may have a greater risk of more rapid disease progression.


Subject(s)
Cytokines/immunology , Macrophages/immunology , Neutrophils/immunology , Osteoarthritis, Knee/immunology , Adult , Aged , Aged, 80 and over , Arthroplasty, Replacement, Knee , Disease Progression , Female , Flow Cytometry , Humans , Leukocyte Elastase/immunology , Male , Middle Aged , Osteoarthritis/immunology , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/surgery , Synovial Fluid/cytology , Synovial Fluid/immunology , Synovial Membrane/immunology , Synovial Membrane/pathology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/immunology
7.
Sci Adv ; 5(10): eaax3203, 2019 10.
Article in English | MEDLINE | ID: mdl-31633025

ABSTRACT

Unlike highly regenerative animals, such as axolotls, humans are believed to be unable to counteract cumulative damage, such as repetitive joint use and injury that lead to the breakdown of cartilage and the development of osteoarthritis. Turnover of insoluble collagen has been suggested to be very limited in human adult cartilage. The goal of this study was to explore protein turnover in articular cartilage from human lower limb joints. Analyzing molecular clocks in the form of nonenzymatically deamidated proteins, we unmasked a position-dependent gradient (distal high, proximal low) of protein turnover, indicative of a gradient of tissue anabolism reflecting innate tissue repair capacity in human lower limb cartilages that is associated with expression of limb-regenerative microRNAs. This association shows a potential link to a capacity, albeit limited, for regeneration that might be exploited to enhance joint repair and establish a basis for human limb regeneration.


Subject(s)
Cartilage, Articular/metabolism , Extremities/physiology , Cartilage Oligomeric Matrix Protein/metabolism , Collagen/metabolism , Databases, Factual , Fibronectins/metabolism , Half-Life , Humans , Mass Spectrometry , MicroRNAs/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Proteome/analysis , Regeneration
8.
Arthritis Res Ther ; 21(1): 146, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31196179

ABSTRACT

BACKGROUND: To identify a synovial fluid (SF) biomarker profile characteristic of individuals with an inflammatory osteoarthritis (OA) endotype. METHODS: A total of 48 knees (of 25 participants) were characterized for an extensive array of SF biomarkers quantified by Rules Based Medicine using the high-sensitivity multiplex immunoassay, Myriad Human InflammationMAP® 1.0, which included 47 different cytokines, chemokines, and growth factors related to inflammation. Multivariable regression with generalized estimating equations (GEE) and false discovery rate (FDR) correction was used to assess associations of SF RBM biomarkers with etarfolatide imaging scores reflecting synovial inflammation; radiographic knee OA severity (based on Kellgren-Lawrence (KL) grade, joint space narrowing, and osteophyte scores); knee joint symptoms; and SF biomarkers associated with activated macrophages and knee OA progression including CD14 and CD163 (shed by activated macrophages) and elastase (shed by activated neutrophils). RESULTS: Significant associations of SF biomarkers meeting FDR < 0.05 included soluble (s)VCAM-1 and MMP-3 with synovial inflammation (FDR-adjusted p = 0.025 and 1.06 × 10-7); sVCAM-1, sICAM-1, TIMP-1, and VEGF with radiographic OA severity (p = 1.85 × 10-5 to 3.97 × 10-4); and VEGF, MMP-3, TIMP-1, sICAM-1, sVCAM-1, and MCP-1 with OA symptoms (p = 2.72 × 10-5 to 0.050). All these SF biomarkers were highly correlated with macrophage markers CD163 and CD14 in SF (r = 0.43 to 0.90, FDR < 0.05); all but MCP-1 were also highly correlated with neutrophil elastase in SF (r = 0.62 to 0.89, FDR < 0.05). CONCLUSIONS: A subset of six SF biomarkers was related to synovial inflammation in OA, as well as radiographic and symptom severity. These six OA-related SF biomarkers were specifically linked to indicators of activated macrophages and neutrophils. These results attest to an inflammatory OA endotype that may serve as the basis for therapeutic targeting of a subset of individuals at high risk for knee OA progression. TRIAL REGISTRATION: Written informed consent was received from participants prior to inclusion in the study; the study was registered at ClinicalTrials.gov ( NCT01237405 ) on November 9, 2010, prior to enrollment of the first participant.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , Neutrophils/metabolism , Osteoarthritis, Knee/metabolism , Synovial Fluid/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Cytokines/metabolism , Disease Progression , Female , Humans , Immunoassay , Inflammation/diagnosis , Macrophages/pathology , Male , Middle Aged , Neutrophils/pathology , Osteoarthritis, Knee/diagnosis , Radiography , Severity of Illness Index , Synovial Fluid/cytology
9.
Rheumatology (Oxford) ; 56(1): 156-164, 2017 01.
Article in English | MEDLINE | ID: mdl-28028161

ABSTRACT

OBJECTIVE: Alkaptonuria (AKU) is a rare autosomal recessive disease resulting from a single enzyme deficiency in tyrosine metabolism. As a result, homogentisic acid cannot be metabolized, causing systemic increases. Over time, homogentisic acid polymerizes and deposits in collagenous tissues, leading to ochronosis. Typically, this occurs in joint cartilages, leading to an early onset, rapidly progressing osteoarthropathy. The aim of this study was to examine tissue turnover in cartilage affected by ochronosis and its role in disease initiation and progression. METHODS: With informed patient consent, hip and knee cartilages were obtained at surgery for arthropathy due to AKU (n = 6; 2 knees/4 hips) and OA (n = 12; 5 knees/7 hips); healthy non-arthritic (non-OA n = 6; 1 knee/5 hips) cartilages were obtained as waste from trauma surgery. We measured cartilage concentrations (normalized to dry weight) of racemized aspartate, GAG, COMP and deamidated COMP (D-COMP). Unpaired AKU, OA and non-OA samples were compared by non-parametric Mann-Whitney U test. RESULTS: Despite more extractable total protein being obtained from AKU cartilage than from OA or non-OA cartilage, there was significantly less extractable GAG, COMP and D-COMP in AKU samples compared with OA and non-OA comparators. Racemized Asx (aspartate and asparagine) was significantly enriched in AKU cartilage compared with in OA cartilage. CONCLUSIONS: These novel data represent the first examination of cartilage matrix components in a sample of patients with AKU, representing almost 10% of the known UK alkaptonuric population. Compared with OA and non-OA, AKU cartilage demonstrates a very low turnover state and has low levels of extractable matrix proteins.


Subject(s)
Aging/metabolism , Alkaptonuria/metabolism , Cartilage, Articular/metabolism , Joint Diseases/metabolism , Ochronosis/metabolism , Adult , Aged , Aged, 80 and over , Aspartic Acid/metabolism , Biomarkers/metabolism , Cartilage Oligomeric Matrix Protein/metabolism , Case-Control Studies , Female , Glycosaminoglycans/metabolism , Hip Joint , Humans , Knee Joint , Male , Middle Aged , Osteoarthritis, Hip/metabolism , Osteoarthritis, Knee/metabolism , Young Adult
10.
J Proteome Res ; 15(2): 374-88, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26632656

ABSTRACT

Articular cartilage consists of chondrocytes and two major components, a collagen-rich framework and highly abundant proteoglycans. Most prior studies defining the zonal distribution of cartilage have extracted proteins with guanidine-HCl. However, an unextracted collagen-rich residual is left after extraction. In addition, the high abundance of anionic polysaccharide molecules extracted from cartilage adversely affects the chromatographic separation. In this study, we established a method for removing chondrocytes from cartilage sections with minimal extracellular matrix protein loss. The addition of surfactant to guanidine-HCl extraction buffer improved protein solubility. Ultrafiltration removed interference from polysaccharides and salts. Almost four-times more collagen peptides were extracted by the in situ trypsin digestion method. However, as expected, proteoglycans were more abundant within the guanidine-HCl extraction. These different methods were used to extract cartilage sections from different cartilage layers (superficial, intermediate, and deep), joint types (knee and hip), and disease states (healthy and osteoarthritic), and the extractions were evaluated by quantitative and qualitative proteomic analyses. The results of this study led to the identifications of the potential biomarkers of osteoarthritis (OA), OA progression, and the joint specific biomarkers.


Subject(s)
Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Proteome/metabolism , Proteomics/methods , Biomarkers/metabolism , Chondrocytes/metabolism , Chromatography/methods , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/isolation & purification , Extracellular Matrix Proteins/metabolism , Guanidines/chemistry , Hip Joint/metabolism , Humans , Knee Joint/metabolism , Mass Spectrometry/methods , Proteoglycans/metabolism , Proteome/chemistry , Proteome/isolation & purification , Reproducibility of Results , Surface-Active Agents/chemistry
11.
Clin Chim Acta ; 451(Pt B): 170-4, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26415819

ABSTRACT

BACKGROUND: While acute trauma is a major cause of osteoarthritis, its etiology is poorly understood. We sought to determine whether xanthine oxidase (XO), a major producer of reactive oxygen species, plays a role in the early events of acute joint injury. METHODS: We analyzed synovial fluid from 23 subjects with recent severe acute knee injury. As a control we evaluated SF from 23 individuals with no or minimal knee osteoarthritis. We measured XO activity, reactive oxygen+reactive nitrogen species (ROS+RNS), protein oxidative damage (carbonyl), the type II collagen synthesis marker procollagen II c-propeptide (CPII) and the type II collagen degradation marker collagen type II telopeptide (CTx-II). We also measured the proinflammatory cytokine IL-6. RESULTS: XO and ROS+RNS were higher (p=0.02 and p=0.001 respectively) in acute injury than control and were strongly positively associated (r=0.62, p=0.004). Carbonyl was higher in acute injury than control (p=0.0002) and was positively correlated with XO (r=0.68, p=0.0007) as well as with ROS+RNS (r=0.71, p=0.004). CPII was higher in acute injury than control (p<0.0001) and was negatively correlated with XO (r=-0.49, p=0.017). While CTxII was not significantly higher in acute injury than control, it was positively correlated with CPII (r=0.71, p=0.0002). IL-6 was higher in acute injury than control (p<0.0001). CONCLUSIONS: These results are consistent with a potentially injurious effect of XO activity in acute joint injury characterized by excess free radical production and oxidative damage. These effects are associated with an inhibition of type II collagen production that may impede the ability of the injured joint to repair.


Subject(s)
Osteoarthritis, Knee/metabolism , Synovial Fluid/metabolism , Xanthine Oxidase/metabolism , Adult , Aged , Aged, 80 and over , Collagen Type II/antagonists & inhibitors , Collagen Type II/biosynthesis , Female , Humans , Male , Middle Aged , Osteoarthritis, Knee/pathology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Young Adult
12.
Matrix Biol ; 39: 56-66, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25179675

ABSTRACT

Our friend and colleague, Dr. Dick Heinegård, contributed greatly to the understanding of joint tissue biochemistry, the discovery and validation of arthritis-related biomarkers and the establishment of methodology for proteomic studies in osteoarthritis (OA). To date, discovery of OA-related biomarkers has focused on cartilage, synovial fluid and serum. Methods, such as affinity depletion and hyaluronidase treatment have facilitated proteomics discovery research from these sources. Osteoarthritis usually involves multiple joints; this characteristic makes it easier to detect OA with a systemic biomarker but makes it hard to delineate abnormalities of individual affected joints. Although the abundance of cartilage proteins in urine may generally be lower than other tissue/sample sources, the protein composition of urine is much less complex and its collection is non-invasive thereby facilitating the development of patient friendly biomarkers. To date however, relatively few proteomics studies have been conducted in OA urine. Proteomics strategies have identified many proteins that may relate to pathological mechanisms of OA. Further targeted approaches to validate the role of these proteins in OA are needed. Herein we summarize recent proteomic studies related to joint tissues and the cohorts used; a clear understanding of the cohorts is important for this work as we expect that the decisive discoveries of OA-related biomarkers rely on comprehensive phenotyping of healthy non-OA and OA subjects. Besides the common phenotyping criteria that include, gender, age, and body mass index (BMI), it is essential to collect data on symptoms and signs of OA outside the index joints and to bolster this with objective imaging data whenever possible to gain the most precise appreciation of the total burden of disease. Proteomic studies on systemic biospecimens, such as serum and urine, rely on comprehensive phenotyping data to unravel the true meaning of the proteomic results.


Subject(s)
Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Proteome/metabolism , Animals , Biomarkers/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Humans , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...