Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Proc Natl Acad Sci U S A. 2011 Feb 1; 108(5): 2088–2093.
Published online 2011 Jan 18. doi: 10.1073/pnas.1012743108
PMCID: PMC3033282
PMID: 21245324

Uric acid is a danger signal of increasing risk for osteoarthritis through inflammasome activation

Associated Data

Supplementary Materials

Abstract

Uric acid (UA) is known to activate the NLRP3 (Nacht, leucine-rich repeat and pyrin domain containing protein 3) inflammasome. When activated, the NLRP3 (also known as NALP3) inflammasome leads to the production of IL-18 and IL-1β. In this cohort of subjects with knee osteoarthritis (OA), synovial fluid uric acid was strongly correlated with synovial fluid IL-18 and IL-1β. Synovial fluid uric acid and IL-18 were strongly and positively associated with OA severity as measured by both radiograph and bone scintigraphy, and synovial fluid IL-1β was associated with OA severity but only by radiograph. Furthermore, synovial fluid IL-18 was associated with a 3-y change in OA severity, on the basis of the radiograph. We conclude that synovial fluid uric acid is a marker of knee OA severity. The correlation of synovial fluid uric acid with the two cytokines (IL-18 and IL-1β) known to be produced by uric acid-activated inflammasomes and the association of synovial fluid IL-18 with OA progression, lend strong support to the potential involvement of the innate immune system in OA pathology and OA progression.

Keywords: arthritis, inflammation, interleukin-18, interleukin-1β, tumor necrosis factor alpha

Uric acid (UA) is constitutively present in normal cells, increased in concentration when cells are injured, and released from dying cells (1). On the basis of a theory proposed by Matzinger, the products of cell stress and tissue damage may represent “danger signals” that function as endogenous adjuvants recognized by the immune system (2). Matzinger proposed that immunity is controlled by an internal conversation between tissues and the cells of the immune system (3). This proposal introduced a new immunological model of an immune system capable of sensing cellular stress and tissue damage (4). Shi subsequently identified uric acid as one of these principal endogenous danger signals released from injured cells and mediating the immune response to antigens associated with injured cells (1). The molecular mechanism of this innate immune response to uric acid was further shown to be the result of the activation of the NALP3 inflammasome, a cytosolic, multiprotein complex that mediates caspase activation by uric acid crystals, leading to the production of the active forms of IL-1β and IL-18 (5). Recently, Kono et al. demonstrated in an in vivo hepatoxicity mouse model that uric acid is a physiological regulator of the inflammation induced by tissue injury (6). These data form the basis for our hypothesis that synovial fluid uric acid is a factor regulating tissue inflammation, disease severity, and progression in osteoarthritis (OA).

Uric acid is best known for its role in gout. When uric acid concentrations exceed the limit of solubility (∼6.8 mg/dL or even lower under conditions of low pH or temperature), crystal formation can ensue, which is capable of activating the NALP3 inflammasome (5) and triggering the acute severe attacks of joint inflammation characteristic of gout (7). Several studies have previously posited an association of uric acid and OA. These include a study of hip replacement patients wherein elevated serum uric acid concentrations were associated with the presence of multijoint hand OA (8). A second study noted the apparent colocalization of gout attacks and radiographic OA at a multitude of joint sites (big toe, midfoot, knee, and distal finger joints) and suggested that OA may facilitate the localized deposition of gout (monosodium urate or MSU) crystals (9). To our knowledge, no previous study has assessed synovial fluid uric acid concentration with regard to radiographic and scintigraphic OA features and severity. In the Prediction of Osteoarthritis Progression (POP) study, we had a unique resource of paired serum and synovial fluid samples with which to evaluate the potential biological association and pathogenic role for uric acid in OA.

Results

Although this cohort was recruited on the basis of knee OA and absence of gout history, a surprising total of 39% of this cohort had serum uric acid concentrations above their respective reference ranges of 7.2 mg/dL for men and 6.0 mg/dL for women (10). The synovial fluid analyses were limited to 69 study participants (49 women and 20 men) with sufficient synovial fluid volumes for uric acid analyses. The mean ± SD age of this subset was 64.5 ± 10.1 y (68.9 ± 7.17 y for men, and 62.7 ± 10.6 y for women). The mean ± SD body mass index was 32.4 ± 7.1 kg/m2 and was similar between the sexes. Knee OA ranged from 1 to 4 in severity (23.1, 14.6, 49.2, and 13.1% for each Kellgren-Lawrence grade).

Uric acid was quantified in both knees of 63 participants and single knees of 6 participants, and their paired sera. The mean serum uric acid concentrations were significantly higher than their paired synovial fluid uric acid concentrations (P < 0.0001) (Fig. 1). Of the 132 synovial fluid samples, matched serum exceeded synovial fluid uric acid concentrations for 113 (85.6%), whereas only 19 (14.4%) had greater synovial fluid than serum uric acid concentrations. The difference between the serum and synovial fluid ranged from +8.27 to −4.12 mg/dL (median difference +1.15 mg/dL). As serum uric acid levels increased, the difference between the serum and synovial fluid uric acid also increased significantly (r2 = 0.1978, P < 0.0001) (Fig. 2A). Serum uric acid correlated strongly only with synovial fluid uric acid (P < 0.0001), whereas synovial fluid UA, IL-18, and IL-1β correlated strongly with each other (P < 0.0001) (Fig. 2 BD). A statistically significant correlation was found between the serum-synovial fluid UA gradient and synovial fluid IL-18 (r2 = 0.176, P < 0.0001) as well as synovial fluid IL-1β (r2 = 0.236, P = 0.0005) (Fig. S1). Additionally, a statistically significant correlation was also found between synovial fluid TNFα and synovial fluid IL-18 (r2 = 0.241, P < 0.0001) as well as synovial fluid IL-1β (r2 = 0.061, P = 0.002) (Fig. S2). No correlation was found for synovial fluid chondroitin sulfate (CS), allantoin, or pyrophosphate (PPi) with each other or with any of the other synovial fluid measures (UA, IL-18, IL-1β, TNFα) or with the serum-synovial fluid UA gradient (Figs. S3S5).

An external file that holds a picture, illustration, etc.
Object name is pnas.1012743108fig01.jpg

Blood and synovial fluid uric acid concentrations for paired samples. Mean serum (n = 69) uric acid concentrations were significantly higher than their paired synovial fluid uric acid concentrations for left (n = 67) and right (n = 65) knees (P < 0.0001). Mean synovial fluid uric acid concentrations from left and right knees were not significantly different (P = 0.22) (paired t test). Error bars represent the SD. The numeric values depicted on the bars are mean ± SD (concentration range).

An external file that holds a picture, illustration, etc.
Object name is pnas.1012743108fig02.jpg

Synovial fluid uric acid and cytokine correlations. (A) Bland-Altman analysis showed a significant association of increased uric acid gradient between the blood and synovial fluid with increasing serum uric acid concentration (R2 = 0.20, P < 0.0001). (BD) Strong correlations were found between synovial fluid uric acid, IL-18, and IL1β. Linear regression was used to provide unadjusted r2 and general estimating equations (GEEs), with adjustments for age, sex, BMI, and chondrocalcinosis, were used for P values with the following results: (B) IL-18 and UA (r2 = 0.41, P < 0.0001); (C) IL-1β and UA (r2 = 0.34, P < 0.0001); and (D) IL-18 and IL-1β (r2 = 0.69, P < 0.0001).

The mean ± SD (range) synovial fluid concentrations of the cytokines were as follows: IL-18 127.1 ± 66.5 pg/mL (8.8–299.5), n = 130; IL-1β 0.375 ± 0.446 pg/mL (0–1.589), n = 48; and TNFα 3.79 ± 2.21 pg/mL (0.76–16.33), n = 123. The mean ± SD (range) synovial fluid concentration of CS was 60 ± 24 μg/mL (7.4–141), n = 118. The mean ± SD (range) synovial fluid concentration of allantoin was 0.37 ± 0.31 μg/mL (0.03–1.52), n = 111. The mean ± SD (range) synovial fluid concentration of PPi was 2.78 ± 0.63 μmol/L (0.91–6.76), n = 123. Knee dominance had no effect on synovial fluid analyte concentration and all our analyses were knee, not individual based, controlling for within-subject correlation.

Although synovial fluid is known to be an ultrafiltrate of plasma to which other components are added from the joint, only synovial fluid, and not serum uric acid concentrations, correlated significantly with severity of OA by two methods of measuring varying aspects of this disease process, namely, knee X-rays and late-phase knee bone scans. X-rays provide a static measure of disease severity, whereas late-phase bone scans provide a dynamic measure of bone turnover, which has been shown to be associated with radiographic features and clinical symptoms of OA (11, 12). In models adjusting for age, sex, and body mass index (BMI), baseline synovial fluid uric acid, IL-18, and IL-1β all correlated independently with X-ray features of OA represented by osteophyte scores, whereas IL-18, IL-1β, and TNFα also correlated with joint space narrowing (Table 1). Synovial fluid uric acid, IL-18, and TNFα also correlated strongly with late-phase bone scan scores (Table 1). IL-18 additionally correlated with knee pain (Table 1).

Table 1.

Associations of analytes with knee OA severity and progression of knee OA using individual models (adjusted for age, sex, BMI, and chondrocalcinosis)

Parameter estimate (95% CIs) P value*
Baseline knee OA severity status
Change (Δ) in knee OA severity status
Bone scanJSNOSTPainΔBone scanΔJSNΔOSTΔPain
SF Uric Acid0.12 (0.05/0.19) 0.0010.06 (−0.003/0.12) 0.060.14 (0.07/0.21) <0.00010.02 (−0.03/0.07) 0.410.04 (−0.10/0.18) 0.580.001 (−0.03/0.03) 0.97−0.02 (−0.18/0.13) 0.77−0.07 (−0.15/0.0001) 0.05
SF ln IL-180.57 (0.32/0.82) <0.00010.21 (0.01/0.40) 0.040.45 (0.18/0.73) 0.0010.14 (0.02/0.25) 0.020.23 (−0.14/0.59) 0.220.04 (−0.05 /0.13) 0.400.46 (0.06/0.86) 0.02−0.02 (−0.21/0.17) 0.84
SF ln IL-1β0.09 (−0.002/0.19) 0.060.16 (0.004/0.31) 0.0450.25 (0.11/0.40) 0.0010.05 (−0.04/0.13) 0.280.10 (−0.24/0.45) 0.550.04 (−0.02/0.09) 0.18−0.37 (−0.89/0.15) 0.16−0.02 (−0.18/0.13) 0.78
SF ln TNFα0.44 (0.31/0.78) <0.00010.16 (0.04/0.45) 0.020.29 (−0.04/0.62) 0.080.12 (−0.01/0.26) 0.070.04 (−0.57/0.65) 0.900.06 (−0.05/0.16) 0.300.70 (0.11/1.29) 0.02−0.02 (−0.28/0.24) 0.87

JSN, joint space narrowing; OST, osteophyte; BMI, body mass index; Δ, change over 3 y in knee OA status; SF, synovial fluid; ln, natural log transformed.

*P values adjusted by GLM (generalized linear modeling to control for within-subject correlation of knee data).

Additional adjustment for baseline OA status.

Three-year follow-up bone scans, radiographs, and pain scores were available for 37, 31, and 38 knees, respectively. Models were run using this 3-y follow-up subset to determine whether the baseline concentrations of synovial fluid measures were associated with changes in knee OA severity and pain over time (Table 1). Adjusting for age, sex, body mass index, and baseline OA severity, baseline IL-18 and TNFα were associated with a 3-y change in osteophyte scores.

Discussion

Although still of uncertain etiology, there are many clinical factors that contribute to the risk and progression of OA, including obesity, joint malalignment, trauma, age, and female sex (13). The strong association demonstrated here, between synovial fluid uric acid and OA severity, quantified radiographically and scintigraphically, demonstrates that uric acid is a marker of disease severity and raises the strong possibility that uric acid may be a factor promoting the pathological process of OA through activation of the inflammasome. This was supported further by a strong association of synovial fluid IL-18 and IL-1β with OA severity and longitudinal data demonstrating an association of baseline synovial fluid IL-18 with radiographic OA progression over 3 y.

Chondrocytes appear normally to be in a state of autophagy thought necessary for maintaining cellular integrity, function, and survival (14). The majority of chondrocytes in OA cartilage, however, are severely degenerated (15) and many are undergoing cell death or “chondroptosis” (16). It was recently demonstrated that OA cartilage is associated with a reduction and loss of regulators of the autophagy pathway (ULK, Beclin-1, and LC3 expression) and an increase in cell death (characterized by increased expression of PARP p85) (14). The extent to which uric acid is released during autophagy is currently unknown, but cell death could result in local supersaturation of uric acid as cytolysis generates large quantities of purines from RNA and DNA (17). As suggested by Matzinger's theories, the release of uric acid could provide an inflammatory danger signal responsible for activating a chronic immune inflammatory response. This response to uric acid has recently been further elucidated as being the result of activation of the NALP3 inflammasome with subsequent production of the active forms of IL-18 and IL-1β (5). The mean concentration of IL-18 we measured in synovial fluid was 1.4 times higher than previously reported for OA synovial fluid, 3.5 times higher than previously reported for normal synovial fluid, but still 2 to 3 times lower than typically seen in rheumatoid arthritis (RA) (18, 19). IL-1β is not typically detected in normal synovial fluid (20, 21) and is very low (3.91 ± 4.66 pg/mL) or undetectable (22) in OA synovial fluid. Our measured concentrations of synovial fluid IL-1β were similarly low or undetectable despite the use of a high sensitivity assay. Because the production of mature active IL-18 and IL-1β requires inflammasome activation (23), these results (high concentrations of synovial fluid IL-18, with correlation between synovial fluid UA and synovial fluid IL-18 and IL-1β) provide strong evidence of a potential role for uric acid in stimulating the production of these cytokines in OA, most likely from resident macrophages via inflammasome activation.

It is already believed that macrophages play a role in osteoarthritis. It was recently demonstrated that both inflammatory and destructive responses generated by osteoarthritis synovium are dependent largely on macrophages. These effects are cytokine driven through a combination of IL-1 and TNFα (24). In gout, monocytes and immature macrophages act to stimulate an acute gout attack triggered as TLR2 and TLR4, which in conjunction with the cytosolic TLR adapter protein myeloid differentiation factor 88 (MyD88), induce the ingestion of MSU crystals by phagocytes and activate the production of proinflammatory molecules, TNFα, IL-1β, IL-6, IL-18, and IL-8, and endothelial activation (5, 2529). We hypothesize a similar but subacute process in the OA joint.

IL-18 is a proinflammatory cytokine that plays a unique and influential role in both the innate and adaptive immune responses (30, 31). IL-18 has been shown to increase production of the potent inflammatory effector TNFα in a dose-dependent manner, with even small amounts of IL-18 (1 pg/mL) inducing TNFα production (3133). Indeed, the principal effect of IL-18 in the RA joint seems to be the direct promotion of TNFα production via binding with macrophage IL-18R (33). That synovial fluid TNFα was expressed in response to IL-18 stimulation in this study is further supported by the study of Martinon who observed a delayed release of TNFα in response to uric acid and speculated that it was in response to inflammasome activation (5). The strong correlation we found between synovial fluid IL-18 and TNFα is consistent with these reports. The concentration of synovial fluid TNFα we found in our subjects was similar to previous reports for OA synovial fluid and approximately sixfold lower than has been reported for RA (34, 35). Although we found a strong correlation of TNFα with uric acid concentration and OA severity, the lower levels of TNFα when compared with RA, along with the correspondingly lower IL-18, suggest a less acute inflammatory process driven by IL-18 as part of an innate immune response.

Although the concentration of IL-1β we found was not particularly high, its correlation with OA severity is not surprising. IL-1β action is under tight regulation with the most important control being the balance between the expression of the IL-1RI active receptor and the IL-1RII inactive or decoy receptor (36, 37). It is well established that in the OA joint there is imbalance between the expression of these two receptors with an up-regulation of the active receptor (IL-1RI) and little or no expression of the decoy receptor (IL-1RII), thereby significantly increasing the potency of even small amounts of IL-1β (3739).

To our knowledge, no previous study reported an association of IL-18 with pain in OA. Recent animal studies have revealed a role for IL-18 in pain. In a study of mechanisms of antigen-induced inflammation in mice, IL-18 was shown to play a significant role in hypernociception via the production of IFN-γ, ET-1, and PGE2 (40). In a recent tibial fracture model in rats, IL-18 up-regulation via inflammasome activation was shown to play a contributory role in complex regional pain syndrome (41). Our data would suggest a similar role for IL-18 in humans.

One recent study of cartilage degeneration in the ankle revealed that MSU surface crystal deposits correlated strongly with cartilage lesions in the talus (42). This study suggested that MSU crystals may contribute both to the initiation and propagation of cartilage degradation. Muehleman proposed that metabolic changes and cartilage fibrillation or fissures enhance the nucleation of MSU crystals, which in turn contribute to mechanically induced damage (42). A process similar to the one that occurs in the ankle likely occurs in the knee, characterized by loss of proteoglycan from the cartilage into the synovial fluid, facilitating the nucleation of uric acid MSU crystals that could precipitate onto the surface of cartilage and act as a nidus for inflammation and mechanical degradation (43). In this scenario, the amount of uric acid in the synovial fluid sufficient for contributing to the cartilage degradation process is less than the concentration typically associated with clinical gout and gout flares (>6.8 mg/dL). Accordingly, the amount of uric acid in the synovial fluid and not in the serum would correlate with radiographic and scintigraphic measures of OA severity, as demonstrated in this study. Unlike previous reports that found no increase in serum uric acid for those with knee OA, serum uric acid concentrations for many of our subjects were higher than established reference ranges (8, 10, 44). Synovial fluid uric acid was also higher in our study compared with a previous report (45).

One limitation of this study is that we were unable to assess the presence or absence of MSU crystals in the joint because all of the synovial fluid samples had been centrifuged and stored as supernatants. Crystalline uric acid, as little as 1.0 mg/dL, and not soluble uric acid, is thought capable of activating dendritic cells and macrophages and priming the T cell response (1, 5, 46). Interestingly, the median drop we found in soluble uric acid from serum to synovial fluid was 1.15 mg/dL. We wonder whether the serum/synovial fluid gradient represents precipitated uric acid. Shi proposed that a chemical phase transition could be the key event that transformed the normal autologous uric acid component into a danger signal. However, as suggested in Muehlemann's study of MSU crystals and cartilage degradation in the ankle, uric acid in the synovial fluid likely adopts both forms—crystalline and soluble—depending on the chemical environment (42). Laurent and Burt et al. both reported, and we confirmed (summarized in Materials and Methods), that uric acid in vitro was less soluble with increasing amounts of chondroitin sulfate (47, 48), a common constituent of OA synovial fluid. We found no correlation between synovial fluid CS and uric acid, or CS and the serum-synovial fluid UA gradient. However, other components of OA synovial fluid have been reported to facilitate uric acid crystal formation (43, 48, 49), so CS may not act alone and therefore would not alone correlate with inflammasome-activated cytokines. For several decades the variation in the protein coat of MSU crystals has been suggested to regulate immune responses to them. Terkeltaub showed that the inflammatory response to MSU crystals could be inhibited by apo B lipoprotein (50). Kanevets et al. have recently demonstrated substantial amounts of uric acid-binding antibodies in unimmunized mice (51), and Ig-coated MSU crystals have been observed in gout specimens (summarized in Kanevets, ref. 51). Uric acid-binding antibodies facilitated urate crystal formation but did not block uric acid-mediated activation of dendritic cells (51). Further work is indicated to evaluate for the presence of uric acid-binding antibodies in OA synovial fluids as another possible potentiator of crystal formation.

Soluble uric acid is also suggested to exert an antiinflammatory effect, possibly due to its role as an antioxidant, on distant sites of inflammation (46, 52). The action of various reactive oxygen species upon uric acid leads to multiple oxidation products with the most abundant being allantoin (53). The synovial fluid allantoin concentrations we observed were similar to those previously reported in RA synovial fluid (54), and just as in that study, as well as other studies of plasma in healthy adults (55), we found no correlation between allantoin and uric acid concentrations. More importantly, we found no correlation between allantoin and the difference in uric acid concentrations from serum to synovial fluid. Thus, oxidative consumption of uric acid within the joint would likely not account for the lower concentrations of uric acid in the joint fluid relative to the serum.

Calcium pyrophosphate dihydrate (CPPD) crystals have also been shown to activate the NALP3 inflammasome although the amount of CPPD required for activation is considerably higher (10 times) than for MSU (5). CPPD is considered the etiologic agent in pseudogout as well as chondrocalcinosis (56). When CPPD is found in OA synovial fluid in conjunction with MSU, the concentration of CPPD is always very low (57). In our study we would have excluded subjects with pseudogout on the basis of exclusion of individuals with gout-like attacks, and controlled for the presence of radiographic chondrocalcinosis (CC) during the analysis. CPPD crystals are thought to form when excess inorganic pyrophosphate (PPi) complexes with calcium to form crystals (58). The concentration of synovial fluid PPi we measured was similar to what has been previously reported for OA synovial fluid (59) and was not associated with any of the synovial fluid cytokines or OA severity. These data decrease the likelihood that CPPD is the source of inflammasome activation in our subjects.

In summary, we compared serum and synovial fluid uric acid levels in patients with knee OA but with no clinical evidence or self-report of gout. We found a significant difference in uric acid levels between matched sera and synovial fluid despite the fact that synovial fluid is a dialysate of blood. Synovial fluid uric acid correlated with radiographic and scintigraphic measure of OA severity and synovial fluid IL-18 and IL-1β, two cytokines known to be produced by uric acid-activated inflammasomes. The precarious balance between soluble uric acid and MSU crystals within the joint, the prevalence of serum uric acid concentrations in the crystal-forming range in this knee cohort, and possibly uric acid released from injured or dying chondrocytes, may, in the presence of nucleating agents such as nonaggregated proteoglycans, collagen, and chondroitin sulfate (43, 4749), tip the balance toward MSU crystallization and the proinflammatory state. A model integrating these concepts is proposed in Fig. 3. These results lend strong support to the potential involvement of the innate immune system in OA pathology. It will be important to better understand the physical chemical properties and forms of uric acid in the OA joint and to further validate synovial fluid uric acid as a risk factor for OA progression as this would have important clinical implications given the availability of well-established pharmacological means of systemically lowering uric acid.

An external file that holds a picture, illustration, etc.
Object name is pnas.1012743108fig03.jpg

Schematic depicting the interaction of OA and uric acid. According to this model, osteoarthritis leads to the release of nucleating agents for urate crystallization. The development of osteoarthritis also leads to increased cell death and the release of uric acid. Taken together with uric acid diffused into the joint fluid from the blood, these factors could contribute to constitutive subacute inflammation and progression of osteoarthritic joints via inflammasome activation.

Materials and Methods

Study Population.

A total of 159 participants (118 female, 41 male) were enrolled in the National Institutes of Health sponsored POP study. A total of 138 participants returned for follow-up 3 y later to assess progression of knee OA. The study was conducted from January 2003 until March 2008 and was approved by the Duke University Institutional Review Board with informed consent being obtained from all subjects. Participants met radiographic criteria for knee OA on the basis of a Kellgren-Lawrence grade of 1–4 (60) and American College of Rheumatology criteria (61) for symptomatic OA of at least one knee. OA exclusion criteria included: exposure to a corticosteroid (either parenteral or oral) within 3 mo before the study evaluation; knee arthroscopic surgery within 6 mo before the study evaluation; known history of avascular necrosis, inflammatory arthritis, Paget's disease, joint infection, periarticular fracture, neuropathic arthropathy, pseudogout, or reactive arthritis. None of the participants had a history of gout in the knee, and none were taking traditional gout medications (allopurinol or colchicine) or anticoagulants. One patient had a remote history of symptoms suggestive of podagra.

Radiographic Imaging.

Posteroanterior fixed-flexion knee radiographs were obtained with the SynaFlexer lower limb positioning frame (Synarc) and were read by the consensus of two graders, blinded to the results of any of the other measures. In addition to Kellgren-Lawrence grade (60), knee radiographs were scored for individual radiographic features of OA of the weight-bearing tibiofemoral joint for joint space narrowing (JSN) and osteophyte (OST) scored semiquantitatively (0–3 scale) using the standardized OARSI atlas (62). Maximum possible scores were 6 for JSN and 12 for OST. Intrarater reliability of the X-ray readings by intraclass correlation coefficients were 0.69 for KL grade, 0.84 for JSN, and 0.81 for OST as described previously (11).

Scintigraphic Imaging.

Scintigraphic images of the knee were obtained at 2.5 h (late phase) after injection of 99mTc methylene diphosphonate. Four views of the knee (anteroposterior, mediolateral, lateromedial, and posteroanterior) were obtained to precisely localize the site of uptake within each knee. The intensity of bone scintigraphic radiotracer uptake was scored semiquantitatively (0–3: 0 = normal, 1 = mild, 2 = moderate, and 3 = intense) by consensus of an experienced nuclear medicine physician (REC) and a senior nuclear medicine resident blinded to the other data. Each compartment of the tibiofemoral knee joint was scored (medial and lateral) for a maximum possible score of 6. A random 20% of the knee bone scans were rescored, blinded to the original scores; intraclass correlation coefficients ranged from 0.86 to 0.89 for the lateral and medial knee compartments as previously described (11).

Serum and Synovial Fluid Samples.

Serum and knee synovial fluid samples were obtained within 10 min of each other after a 2-h fast in the midafternoon. All synovial fluid samples were centrifuged and cell free supernatants, along with sera, were frozen at −80 °C. Uric acid concentrations were measured on baseline sera of all 159 participants. Sufficient volumes of synovial fluid were available for quantification of uric acid from 132 knees representing 69 individuals. A total of 97 of the synovial fluid samples were obtained directly (neat, no dilution); 35 synovial fluid samples were collected by small volume (10 mL) lavage with the precise correction factor to account for the dilutional effects of lavage determined by the urea correction method as previously described (63). Of these 132 synovial fluid samples, remaining sample volumes were adequate for measurement of IL-18 from 130 knees, IL-1β from 48 knees, chondroitin sulfate (CS) from 118 knees, TNFα and PPi from 123 knees, and allantoin from 111 knees. See SI Materials and Methods for all biochemical analyses of these samples.

Pain Scores.

Knee symptoms were ascertained by the NHANES I criterion (64) of pain, aching, or stiffness on most days of any 1 month in the last year; for subjects answering yes, symptoms were quantified as mild, moderate, or severe, yielding a total score of 0–3 for each knee.

Statistical Analysis.

To meet assumptions of normality by the D'Agostino and Pearson omnibus test (GraphPad Software), all synovial fluid analytes, other than uric acid, were natural log transformed. The proportion of individuals with serum uric acid concentrations greater than a well-established reference range (>7.2 mg/dL for males and >6 mg/dL for females) (10) was calculated for the total sample (n = 159) and the subsample (n = 69) of individuals with paired sera and synovial fluid. The values were compared by paired t test. When lavaged synovial fluid samples that had been corrected for dilution were analyzed separately from neat samples, no effect of dilution was seen, so those results were pooled together. Descriptive statistics and univariate analyses were performed using Graphpad Prism software. Relationships between synovial fluid analytes and OA were analyzed using the GenMOD procedure, to control for within-subject correlation of knee data, with the addition of a repeated statement to control for age, sex, BMI, and chondrocalcinosis (GLM, SAS Enterprise Guide). This procedure is equivalent to using general estimating equations (GEE). Because r2 values are not provided with GLM/GEE, linear regressions were also performed to provide unadjusted r2 values (Graphpad Prism).

Supplementary Material

Supporting Information:

Acknowledgments

Special thanks to Dr. Catherine Hammet-Stabler for assistance with enzymatic uric acid measurements and Drs. David Pisetsky and Robert Terkeltaub for editorial suggestions. This work was supported by National Institutes of Health/National Institute of Arthritis and Musculoskeletal and Skin Diseases Grants R01 AR048769, P01 AR50245, U01 AR050898 and National Institute on Aging Claude D. Pepper Older Americans Independence Centers Grant 5P30 AG028716.

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission. J.T. is a guest editor invited by the Editorial Board.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1012743108/-/DCSupplemental.

References

1. Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425:516–521. [PubMed] [Google Scholar]
2. Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994;12:991–1045. [PubMed] [Google Scholar]
3. Matzinger P. The danger model: A renewed sense of self. Science. 2002;296:301–305. [PubMed] [Google Scholar]
4. Pulendran B. Immune activation: Death, danger and dendritic cells. Curr Biol. 2004;14:R30–R32. [PubMed] [Google Scholar]
5. Martinon F, Pétrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241. [PubMed] [Google Scholar]
6. Kono H, Chen C-J, Ontiveros F, Rock KL. Uric acid promotes an acute inflammatory response to sterile cell death in mice. J Clin Invest. 2010;120:1939–1949. [PMC free article] [PubMed] [Google Scholar]
7. Schumacher HR., Jr The pathogenesis of gout. Cleve Clin J Med. 2008;75(Suppl 5):S2–S4. [PubMed] [Google Scholar]
8. Sun Y, et al. Serum uric acid and patterns of radiographic osteoarthritis—the Ulm Osteoarthritis Study. Scand J Rheumatol. 2000;29:380–386. [PubMed] [Google Scholar]
9. Roddy E, Zhang W, Doherty M. Are joints affected by gout also affected by osteoarthritis? Ann Rheum Dis. 2007;66:1374–1377. [PMC free article] [PubMed] [Google Scholar]
10. Painter PC, Cope JY, Smith JL. Reference information for the clinical laboratory. In: Burtis CA, Ashwood ER, editors. Tietz Textbook of Clinical Chemistry. 3rd Ed. Philadelphia: WB Saunders; 1999. p. 1838. [Google Scholar]
11. Kraus VB, et al. Association of bone scintigraphic abnormalities with knee malalignment and pain. Ann Rheum Dis. 2009;68:1673–1679. [PMC free article] [PubMed] [Google Scholar]
12. Kim HR, So Y, Moon SG, Lee IS, Lee SH. Clinical value of (99m)Tc-methylene diphosphonate (MDP) bone single photon emission computed tomography (SPECT) in patients with knee osteoarthritis. Osteoarthritis Cartilage. 2008;16:212–218. [PubMed] [Google Scholar]
13. Kraus VB, Vail TP, Worrell T, McDaniel G. A comparative assessment of alignment angle of the knee by radiographic and physical examination methods. Arthritis Rheum. 2005;52:1730–1735. [PubMed] [Google Scholar]
14. Caramés B, Taniguchi N, Otsuki S, Blanco FJ, Lotz M. Autophagy is a protective mechanism in normal cartilage, and its aging-related loss is linked with cell death and osteoarthritis. Arthritis Rheum. 2010;62:791–801. [PMC free article] [PubMed] [Google Scholar]
15. Aigner T, Söder S, Gebhard PM, McAlinden A, Haag J. Mechanisms of disease: Role of chondrocytes in the pathogenesis of osteoarthritis—structure, chaos and senescence. Nat Clin Pract Rheumatol. 2007;3:391–399. [PubMed] [Google Scholar]
16. Roach HI, Aigner T, Söder S, Haag J, Welkerling H. Pathobiology of osteoarthritis: Pathomechanisms and potential therapeutic targets. Curr Drug Targets. 2007;8:271–282. [PubMed] [Google Scholar]
17. Shi Y. Caught red-handed: Uric acid is an agent of inflammation. J Clin Invest. 2010;120:1809–1811. [PMC free article] [PubMed] [Google Scholar]
18. Shao XT, et al. Expression of interleukin-18, IL-18BP, and IL-18R in serum, synovial fluid, and synovial tissue in patients with rheumatoid arthritis. Clin Exp Med. 2009;9:215–221. [PubMed] [Google Scholar]
19. Bokarewa M, Hultgren O. Is interleukin-18 useful for monitoring rheumatoid arthritis? Scand J Rheumatol. 2005;34:433–436. [PubMed] [Google Scholar]
20. Higuchi H, et al. Changes in biochemical parameters after anterior cruciate ligament injury. Int Orthop. 2006;30:43–47. [PMC free article] [PubMed] [Google Scholar]
21. Kahle P, et al. Determination of cytokines in synovial fluids: Correlation with diagnosis and histomorphological characteristics of synovial tissue. Ann Rheum Dis. 1992;51:731–734. [PMC free article] [PubMed] [Google Scholar]
22. Scanzello CR, et al. Local cytokine profiles in knee osteoarthritis: Elevated synovial fluid interleukin-15 differentiates early from end-stage disease. Osteoarthritis Cartilage. 2009;17:1040–1048. [PubMed] [Google Scholar]
23. Bryant C, Fitzgerald KA. Molecular mechanisms involved in inflammasome activation. Trends Cell Biol. 2009;19:455–464. [PubMed] [Google Scholar]
24. Bondeson J, Wainwright SD, Lauder S, Amos N, Hughes CE. The role of synovial macrophages and macrophage-produced cytokines in driving aggrecanases, matrix metalloproteinases, and other destructive and inflammatory responses in osteoarthritis. Arthritis Res Ther. 2006;8:R187. [PMC free article] [PubMed] [Google Scholar]
25. Liu-Bryan RP, Scott P, Sydlaske A, Rose DM, Terkeltaub R. Innate immunity conferred by Toll-like receptors 2 and 4 and myeloid differentiation factor 88 expression is pivotal to monosodium urate monohydrate crystal-induced inflammation. Arthritis Rheum. 2005;52:2936–2946. [PubMed] [Google Scholar]
26. di Giovine FS, Malawista SE, Thornton E, Duff GW. Urate crystals stimulate production of tumor necrosis factor alpha from human blood monocytes and synovial cells. Cytokine mRNA and protein kinetics, and cellular distribution. J Clin Invest. 1991;87:1375–1381. [PMC free article] [PubMed] [Google Scholar]
27. Di Giovine FS, Malawista SE, Nuki G, Duff GW. Interleukin 1 (IL 1) as a mediator of crystal arthritis. Stimulation of T cell and synovial fibroblast mitogenesis by urate crystal-induced IL 1. J Immunol. 1987;138:3213–3218. [PubMed] [Google Scholar]
28. Guerne PA, Terkeltaub R, Zuraw B, Lotz M. Inflammatory microcrystals stimulate interleukin-6 production and secretion by human monocytes and synoviocytes. Arthritis Rheum. 1989;32:1443–1452. [PubMed] [Google Scholar]
29. Onello E, Traynor-Kaplan A, Sklar L, Terkeltaub R. Mechanism of neutrophil activation by an unopsonized inflammatory particulate. Monosodium urate crystals induce pertussis toxin-insensitive hydrolysis of phosphatidylinositol 4,5-bisphosphate. J Immunol. 1991;146:4289–4294. [PubMed] [Google Scholar]
30. Matsui K, Tsutsui H, Nakanishi K. Pathophysiological roles for IL-18 in inflammatory arthritis. Expert Opin Ther Targets. 2003;7:701–724. [PubMed] [Google Scholar]
31. Liew FY, Wei X-Q, McInnes IB. Role of interleukin 18 in rheumatoid arthritis. Ann Rheum Dis. 2003;62(Suppl 2):ii48–ii50. [PMC free article] [PubMed] [Google Scholar]
32. Dai S-M, Matsuno H, Nakamura H, Nishioka K, Yudoh K. Interleukin-18 enhances monocyte tumor necrosis factor α and interleukin-1β production induced by direct contact with T lymphocytes: Implications in rheumatoid arthritis. Arthritis Rheum. 2004;50:432–443. [PubMed] [Google Scholar]
33. Gracie JA, et al. A proinflammatory role for IL-18 in rheumatoid arthritis. J Clin Invest. 1999;104:1393–1401. [PMC free article] [PubMed] [Google Scholar]
34. Hriuchi T, et al. The increase of parathyroid hormone-related peptide and cytokine levels in synovial fluid of elderly rheumatoid arthritis and osteoarthritis. Endocr J. 1999;45:643–649. [PubMed] [Google Scholar]
35. Pozgan U, et al. Expression and activity profiling of selected cysteine cathepsins and matrix metalloproteinases in synovial fluids from patients with rheumatoid arthritis and osteoarthritis. Biol Chem. 2010;391:571–579. [PubMed] [Google Scholar]
36. Colotta F, et al. Interleukin-1 type II receptor: A decoy target for IL-1 that is regulated by IL-4. Science. 1993;261:472–475. [PubMed] [Google Scholar]
37. Martel-Pelletier J, et al. The interleukin-1 receptor in normal and osteoarthritic human articular chondrocytes. Identification as the type I receptor and analysis of binding kinetics and biologic function. Arthritis Rheum. 1992;35:530–540. [PubMed] [Google Scholar]
38. Wang J, Verdonk P, Elewaut D, Veys EM, Verbruggen G. Homeostasis of the extracellular matrix of normal and osteoarthritic human articular cartilage chondrocytes in vitro. Osteoarthritis Cartilage. 2003;11:801–809. [PubMed] [Google Scholar]
39. Attur MG, et al. Reversal of autocrine and paracrine effects of interleukin 1 (IL-1) in human arthritis by type II IL-1 decoy receptor. Potential for pharmacological intervention. J Biol Chem. 2000;275:40307–40315. [PubMed] [Google Scholar]
40. Verri WA, Jr, et al. Antigen-induced inflammatory mechanical hypernociception in mice is mediated by IL-18. Brain Behav Immun. 2007;21:535–543. [PubMed] [Google Scholar]
41. Li WW, et al. The NALP1 inflammasome controls cytokine production and nociception in a rat fracture model of complex regional pain syndrome. Pain. 2009;147:277–286. [PMC free article] [PubMed] [Google Scholar]
42. Muehleman C, et al. Association between crystals and cartilage degeneration in the ankle. J Rheumatol. 2008;35:1108–1117. [PMC free article] [PubMed] [Google Scholar]
43. McGill NW, Dieppe PA. The role of serum and synovial fluid components in the promotion of urate crystal formation. J Rheumatol. 1991;18:1042–1045. [PubMed] [Google Scholar]
44. Hart DJ, Doyle DV, Spector TD. Association between metabolic factors and knee osteoarthritis in women: The Chingford Study. J Rheumatol. 1995;22:1118–1123. [PubMed] [Google Scholar]
45. Regan EA, Bowler RP, Crapo JD. Joint fluid antioxidants are decreased in osteoarthritic joints compared to joints with macroscopically intact cartilage and subacute injury. Osteoarthritis Cartilage. 2008;16:515–521. [PubMed] [Google Scholar]
46. Zare F, et al. Uric acid, a nucleic acid degradation product, down-regulates dsRNA-triggered arthritis. J Leukoc Biol. 2006;79:482–488. [PubMed] [Google Scholar]
47. Laurent TC. Solubility of sodium urate in the presence of chondroitin-4-sulphate. Nature. 1964;202:1334. [PubMed] [Google Scholar]
48. Burt HM, Dutt YC. Growth of monosodium urate monohydrate crystals: effect of cartilage and synovial fluid components on in vitro growth rates. Ann Rheum Dis. 1986;45:858–864. [PMC free article] [PubMed] [Google Scholar]
49. Choi HK, Mount DB, Reginato AM, American College of Physicians. American Physiological Society Pathogenesis of gout. Ann Intern Med. 2005;143:499–516. [PubMed] [Google Scholar]
50. Terkeltaub R, Curtiss LK, Tenner AJ, Ginsberg MH. Lipoproteins containing apoprotein B are a major regulator of neutrophil responses to monosodium urate crystals. J Clin Invest. 1984;73:1719–1730. [PMC free article] [PubMed] [Google Scholar]
51. Kanevets U, Sharma K, Dresser K, Shi Y. A role of IgM antibodies in monosodium urate crystal formation and associated adjuvanticity. J Immunol. 2009;182:1912–1918. [PMC free article] [PubMed] [Google Scholar]
52. Ames BN, Cathcart R, Schwiers E, Hochstein P. Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: A hypothesis. Proc Natl Acad Sci USA. 1981;78:6858–6862. [PMC free article] [PubMed] [Google Scholar]
53. Kaur H, Halliwell B. Action of biologically-relevant oxidizing species upon uric acid. Identification of uric acid oxidation products. Chem Biol Interact. 1990;73:235–247. [PubMed] [Google Scholar]
54. Grootveld M, Halliwell B. Measurement of allantoin and uric acid in human body fluids. A potential index of free-radical reactions in vivo? Biochem J. 1987;243:803–808. [PMC free article] [PubMed] [Google Scholar]
55. Benzie IFF, Chung W-Y, Tomlinson B. Simultaneous measurement of allantoin and urate in plasma: Analytical evaluation and potential clinical application in oxidant:antioxidant balance studies. Clin Chem. 1999;45:901–904. [PubMed] [Google Scholar]
56. Richette P, Bardin T, Doherty M. An update on the epidemiology of calcium pyrophosphate dihydrate crystal deposition disease. Rheumatology (Oxford) 2009;48:711–715. [PubMed] [Google Scholar]
57. Robier C, Neubauer M, Quehenberger F, Ranier F. Ann Rheum Dis. 2010. Coincidence of calcium pyrophosphate and monosodium urate crystals in the synovial fluid of patients with gout determined by the cytocentrifugation technique. October 21 [ePub ahead of print] [PubMed] [Google Scholar]
58. Rosenthal AK. Calcium crystal deposition and osteoarthritis. Rheum Dis Clin North Am. 2006;32:401–412. vii. [PubMed] [Google Scholar]
59. Hearn PR, Russell RGG. Formation of calcium pyrophosphate crystals in vitro: Implications for calcium pyrophosphate crystal deposition disease (pseudogout) Ann Rheum Dis. 1980;39:222–227. [PMC free article] [PubMed] [Google Scholar]
60. Kellgren JH, Lawrence JS. Radiological assessment of osteo-arthrosis. Ann Rheum Dis. 1957;16:494–502. [PMC free article] [PubMed] [Google Scholar]
61. Altman RD. Criteria for classification of clinical osteoarthritis. J Rheumatol Suppl. 1991;27:10–12. [PubMed] [Google Scholar]
62. Altman RD, Hochberg M, Murphy WA, Jr, Wolfe F, Lequesne M. Atlas of individual radiographic features in osteoarthritis. Osteoarthritis Cartilage. 1995;(3Suppl A):3–70. [PubMed] [Google Scholar]
63. Kraus VB, et al. Urea as a passive transport marker for arthritis biomarker studies. Arthritis Rheum. 2002;46:420–427. [PubMed] [Google Scholar]
64. Davis MA, Ettinger WH, Neuhaus JM. Obesity and osteoarthritis of the knee: Evidence from the National Health and Nutrition Examination Survey (NHANES I) Semin Arthritis Rheum. 1990;20(3, Suppl 1):34–41. [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

-