🎧 New: AI-Generated Podcasts Turn your study notes into engaging audio conversations. Learn more

Loading...
Loading...
Loading...
Loading...
Loading...
Loading...
Loading...

Full Transcript

Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 Contents lists available at ScienceDirect Seminars in Arthritis and Rheumatism journal homepage: www.elsevier.com/locate/semarthrit The biology of urate Robert T. Keenan Division of Rheumatology, Duke University School of Medicine, Durham 2771...

Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 Contents lists available at ScienceDirect Seminars in Arthritis and Rheumatism journal homepage: www.elsevier.com/locate/semarthrit The biology of urate Robert T. Keenan Division of Rheumatology, Duke University School of Medicine, Durham 27710, NC, USA A B S T R A C T Urate is the end-product of the purine metabolism in humans. The dominant source of urate is endogenous purines and the remainder comes through diet. Approximately two thirds of urate is eliminated via the kidney with the rest excreted in the feces. While the transporter BCRP, encoded by ABCG2, has been found to play a role in both the gut and kidney, SLC22A12 and SLC2A9 encoding URAT1 and GLUT9, respectively, are the two transporters best characterized. Only 8 12% of the filtered urate is excreted by the kidney. Renal elimination of urate depends substantially on specific transporters, including URAT1, GLUT9 and BCRP. Studies that have assessed the biologic effects of urate have produced highly variable results. Although there is a suggestion that urate may have anti-oxidant properties in some circumstances, the majority of evidence indicates that urate is pro-inflammatory. Hyperuricemia can result in the formation of monosodium urate (MSU) crystals that may be recognized as danger signals by the immune system. This immune response results in the activation of the NLRP3 inflammasome and ultimately in the production and release of interleukin-1b, and IL-18, that mediate both inflammation, pyroptotic cell death, and necroinflammation. It has also been demonstrated that soluble urate mediates effects on the kidney to induce hypertension and can induce long term epigenetic reprogramming in myeloid cells to induce “trained immunity.” Together, these sequelae of urate are thought to mediate most of the physiological effects of hyperuricemia and gout, illustrating this biologically active molecule is more than just an “end-product” of purine metabolism. © 2020 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license. (http://creativecommons.org/licenses/by-nc-nd/4.0/) Introduction The metabolic production of uric acid is ubiquitous amongst animals and other life forms. It is important to recognize that urate can have both beneficial and pathological roles in human physiology. Uric acid (C5H4N4O3) is a heterocyclic organic compound with a molecular weight of 168 Da. It is synthesized from the metabolism of the purine nucleic acids, adenine and guanine. In this process, adenosine monophosphate is converted to inosine and guanine monophosphate is converted to guanosine. These two nucleosides are then converted to hypoxanthine and guanine. Hypoxanthine is oxidized to form xanthine by xanthine-oxidase, and guanine is deaminated to form xanthine by guanine deaminase. Xanthine is again oxidized by xanthine oxidase to form uric acid. At physiologic pH, uric acid in the body exists mainly as urate [1]. The normal reference range for urate in human blood is 1.5 to 6.0 mg/dL in women and 2.5 7.0 mg/dL in men. When the level of urate is >6.8 mg/dL (405 mmol/l), the limit of solubility under physiological conditions, crystals of urate may form as monosodium urate (MSU) [1]. Multiple factors Publication of this supplement was supported by Horizon Therapeutics plc. E-mail address: [email protected] have been found to influence MSU crystal formation, including temperature, sodium and cation concentration, pH, mechanical stress, cartilage components, uric acid binding antibodies, and components of cartilage and synovial fluid [2,3] (Fig. 1). It is important to understand and appreciate that physiological conditions are not always at ideal clinically, and crystal formation and deposition can occur at levels between 6 and 7 mg/dL. Therefore, 6 mg/dL (360 mmol/l) should be seen as the limit of solubility in both female and male patients. Humans, some higher primates, and certain New World monkeys do not show any detectable uricase activity [4,5]. Urate is maintained in humans and higher primates because of the loss of uricase activity resulting from various mutations of its encoding gene during the Miocene epoch. At first glance, uric acid or urate seems to be just metabolic waste needing excretion, but some studies implicate otherwise. It has been suggested that there may be benefits to the absence of uricase and high urate levels. These may include antioxidant activity, better maintenance of blood pressure in times of low salt ingestion, and changes in fructose metabolism [4]. Some studies have demonstrated that uric acid is a major immunological adjuvant or ‘danger’ signal found in damaged cells, and that it alone could promote T cell activation [6]. Additionally, the uric acid danger signal may play a role in tumor immunity [7]. This https://doi.org/10.1016/j.semarthrit.2020.04.007 0049-0172/© 2020 The Author(s). Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license. (http://creativecommons.org/licenses/by-nc-nd/4.0/) R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 Clinical manifestation Disease progression Risk factor or cause Asymptomatic state No disease Normouricaemia Hyperuricaemia • Genetic factors (e.g. SNPs in urate transporter genes) • Environmental factors (e.g. diet and BMI) • Impaired kidney function • Increasing age • Male sex • Medications • High cell turnover states, etc. Symptomatic disease MSU crystal deposition • Reduced solubility of urate • Increased nucleation of MSU crystals • Growth of MSU crystals Recurrent gout flares Acute inflammatory response to deposited MSU crystals S3 Symptomatic disease with complications Chronic gouty arthritis and tophaceous gout Chronic granulomatous inflammatory response to deposited crystals Fig. 1. Disease progression and risk factors. potential role of uric acid will be further discussed in the section addressing pain and Inflammation associated with MSU crystals. is a substantial contributor to extra-renal elimination of urate. Importantly, ABCG2 polymorphisms have been found to be associated with early onset gout in patients less than 30 years old [20,21]. Elimination of urate Definition of hyperuricemia The production and catabolism of purines are relatively constant between 300 and 400 mg per day and about two-thirds of the urate produced by these products is eliminated via the kidney, and the rest is eliminated by the gastrointestinal tract [1,8,9]. In healthy, fully functioning nephrons, this process is responsive to serum urate levels resulting in increased renal excretion with rises in serum urate. Urate is almost completely unbound by plasma proteins, therefore nearly all urate undergoes ultrafiltration by glomeruli. Subsequent to filtration by the glomeruli, urate enters the S1 segment of the proximal tubule where it undergoes resorption, resulting in a net reclamation of between 90% and 98%. At the distal proximal tubule, secretion occurs in which resorbed urate is transported into the tubule lumen resulting in approximately 10% of the filtered urate being excreted in the urine and 90% is reabsorbed [1,10,11]. Three transporters, URAT1, GLUT9, and BCRP, have been reported to play important roles in the elimination and regulation of urate [1,12 16] (Fig. 2). URAT1 is the most important apical urate exchanger in the proximal tubule and it plays a key role in the physiology of urate homeostasis. The URAT1 protein is encoded by the SLC22A12 gene. GLUT9 (SLC2A9), and is responsible for the basolateral transport of urate in the proximal tubule. ABCG2 is one of three human adenosine triphosphate (ATP) binding cassette (ABC) transporters, and it is involved in the cellular export of a larger number of chemically and structurally diverse compounds. It has been shown to play a significant role in renal and gastrointestinal urate excretion. Additional studies have shown the importance of organic anion transporters (OAT) in both the retention and elimination of urate from the kidney. Interestingly, recent studies have shown a potential uricosuric effect of a sodium-glucose transporter 2 (SGLT2) inhibitor which raises the possibility of urate transport across the SGLT2 [17]. Almost six decades ago, Sorensen reported that in healthy individuals the gut is responsible for 20 30% of the urate that is eliminated daily [18]. Despite the role of the gut being recognized for its role in urate homeostasis for over 50 years, the mechanism in extra-renal elimination of urate is still not completely understood. A study in rats showed that significant amounts of externally administered and endogenous urate were recovered in the intestinal lumen with minimal biliary excretion [8]. Additional experiments showed that the efflux transporter, ABCG2 contributes to the intestinal excretion of urate [19]. Although still under appreciated, these results support the view that direct intestinal secretion Hyperuricemia is defined as a serum urate concentration >6.8 mg/ dL (the in vitro solubility limit of MSU). Gout typically emerges in individuals with serum urate >6.8 mg/dL (360 mmol/L) and its prevalence rises with increases in serum urate above this threshold [22,23]. Guidelines recommend lowering serum urate to <6 mg/dL (360 mmol/l), the level clinically seen as the limit of solubility, as deposition can occur at levels around 6.8 mg/dL in vivo as noted above. The guidelines also note some patients should be treated more aggressively to target serum urate levels below 5 mg/dL (mmol/ L) (i.e., patients with higher urate burden and disease severity, often manifested by the presence of clinically evident tophi, recurrent flares, and significant disability) [24]. Development of hyperuricemia Dietary factors Dietary factors have been demonstrated to play a significant role in the development of hyperuricemia [9,25]. Excess consumption of alcohol and purine-rich foods (e.g., meats, seafood, some vegetables, and animal protein) have been implicated in the development of hyperuricemia since urate is the end product of purine degradation. Results from the Third National Health and Nutrition Examination Survey (NHANES III) indicated that higher levels of meat and seafood consumption were associated with higher serum urate levels, but that total protein intake was not [26]. Dairy consumption was inversely associated with serum urate concentrations. Sir William Osler implicated sugar intake as a risk for gout [27]. Over a century later, NHANES data indicated an increase in urate of 0.33 mg/dL in participants drinking 1 3.9 sugar-sweetened servings per day vs participants drinking none after adjustment for diet including total energy intake, age, sex, medications, hypertension, and estimated glomerular filtration rate (eGFR) [28]. Results from the Cross-Sectional Analysis from the Brazilian Longitudinal Study of Adult Health also indicated that consumption of soft drinks and dietary fructose is positively associated with hyperuricemia and higher urate levels in Brazilian adults [29]. Other studies addressing this issue have confirmed this relationship, but the association was weak S4 R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 Fig. 2. Urate transporters. [30 32]. The relationship makes physiological sense given the metabolism of fructose compared to sucrose results in the generation of additional substrates (AMP) for uric acid generation, but prospective studies are needed to improve our understanding of the role of fructose in the development of hyperuricemia and gout [33]. Although social and medical dogma dictate the importance of diet in urate formation and subsequently gout (particularly gout flares), a recent meta-analysis has challenged the absolute role purine intake plays in hyperuricemia. In a study of almost 17,000 individuals of European descent, Major and colleagues did find an association between dietary intake of alcohol, meats, and soft drinks with hyperuricemia, and an inverse relationship with skim milk, eggs, cheese and non-citrus fruits [34]. These findings were consistent with other diet and hyperuricemia studies, but interestingly, they found the role of diet contributing to no more than 0.3% of the variance in serum urate levels. Conversely, they found 23.9% of the variance in serum urate levels was explained by genome wide single nucleotide variation. This study not only illuminates the role diet and environmental factors play in serum urate levels, but also the factor we cannot control, genetics. UBE2Q2, IGF1R, NFAT5, MAF, HLF, ACVR1B-ACVRL1 and B3GNT4. These include genes that encode transporters, transcriptional factors, signaling receptors, and enzymes [25,40,41]. It is important to note that not all individuals with hyperuricemia develop gout (i.e., symptomatic hyperuricemia) and that some people with high urate levels remain asymptomatic [42]. Genetic factors have been implicated in the transition from hyperuricemia to gout. A multistage genome-wide association study (GWAS) resulted in the discovery of three loci,17q23.2 (rs11653176, BCAS3), 9p24.2 (rs12236871, RFX3) and 11p15.5 (rs179785, KCNQ1) containing inflammatory candidate genes that appear to be related to progression from hyperuricemia to inflammatory gout [43]. A second GWAS demonstrated three loci (CNTN5, MIR302F, and ZNF724) associated with mechanisms of gout development, which differ from the known gout risk loci that elevate serum urate levels. CNTN5 is a member of the contactin family and it is associated with inflammatory diseases including ankylosing spondylitis and Behçet’s disease. The microRNA MIR302F also appears to be involved in cancer; and, the exact function of ZNF724 is not known [44]. Genetic factors Anti-inflammatory and pro-inflammatory actions of urate Multiple genetic factors contribute to the development of hyperuricemia. Genome-wide association studies have identified multiple loci associated with the development of hyperuricemia. Polymorphisms and mutations in the genes SLC22A12, SLC2A9, and ABCG2 encoding, the previously described transporters (URAT1, GLUT9, and BCRP, respectively) have been implicated in the development of hyperuricemia and gout [35 38]. Other important genes implicated in hyperuricemia and gout include: SLC22A11, encoding OAT4; SLC17A1, encoding NPT1, SLC17A3, encoding NPT4; PDZK1, encoding PDZ domain containing 1 scaffolding protein involved in assembly of urate transportasome; and GCKR, encoding glucokinase regulator (regulatory protein inhibiting glucokinase in the liver and pancreas) [39]. Other loci that have been associated with variations in serum urate concentrations include regions in or near TRIM46, INHBB, SFMBT1, TMEM171, VEGFA, BAZ1B, PRKAG2, STC1, HNF4G, A1CF, ATXN2, It has been suggested that urate acts as a free radical scavenger, and is considered one of the most important natural anti-oxidants in humans [45,46]. The anti-oxidant effects of urate prompted exploration of neuroprotective effects of urate. Multiple beneficial actions have been suggested on the basis of results from studies in animals and administration of urate also appears to improve cognitive function in patients with intellectual disability [47,48]. The mechanism by which urate positively impacts neurological function includes suppression of oxygen radical accumulation, stabilization of calcium homeostasis, preservation of mitochondrial function, and protection of neurons from glutamate-associated toxicity. Urate has been posited as neuroprotective in several diseases including stroke, Parkinson’s disease (PD), multiple sclerosis (MS) and Alzheimer’s disease (AD). The presence of high circulating levels of urate has been related with lower severity of neurological damage R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 and lower volume of cerebral infarction in patients with stroke [49]. Urate administration during the course of acute ischemic stroke is associated with smaller cerebral ischemia [47]. The URICO-ITCUS study, a randomized, double-blinded, placebo-controlled phase 2b/3 trial evaluating 411 patients with acute stroke showed adding a 1000 mg intravenous uric acid to thrombolytic therapy did not increase the proportion of patients achieving excellent outcomes (defined as a modified Rankin Scale score of 0-1, or 2 if premorbid score was 2) [50]. However, further analysis of this study showed the uric acid group had less early ischemic worsening compared to placebo, especially in patients with good collateral circulation [51]. High serum urate levels have been associated with reduced risk for development of PD and slower progression in patients diagnosed with PD and multiple sclerosis [52 55]. Epidemiologic studies suggest hyperuricemia may have a protective effect against Alzheimer’s disease [56]. Further support for this observation was demonstrated by a prospective study following patients with mild dementia [57]. The author’s found patients in the lowest serum urate quintile had a significantly greater progression of cognitive decline, compared with those in the highest quintile over a 36-month period. In a murine MS model, there was a dose dependent relationship between high uric acid levels and progression of disease [58]. However, attempts to slow MS progression by administering inosine to raise urate levels have not been effective and this treatment also increased the risk for renal complications [59,60]. In contrast, a recent meta-analysis showed inconsistent effects and may differ depending whether dementia is of the vascular or AD type [61]. Other studies have seen similar results regarding the impact of hyperuricemia, specifically on vascular dementia [62]. To muddle the impact of urate on dementia, urate lowering therapy (ULT) seemed to decrease the risk of dementia in gout patients [63]. Contrary to the data supporting urate’s anti-inflammatory effects, urate has been found to be a pro-oxidant by forming radicals in reactions with other oxidants, and these radicals seem to target predominantly lipids (e.g., low density lipoproteins) [46]. Urate may also interfere with normal endothelial function by inhibition of nitric oxide (NO) function under conditions of oxidative stress [64]. Additionally, urate has been shown to increase in NADPH oxidase activity and reactive oxygen species (ROS) production in mature adipocytes. The stimulation of NADPH oxidase-dependent ROS by urate has been shown to activate mitogen-activated protein kinase p38 and ERK1/2, decrease NO bioavailability, and increase protein nitrosylation and lipid oxidation [65]. Facilitating the counterargument to urate being anti-inflammatory, it has been noted that the anti-inflammatory actions of urate are modest. A study in rats indicated that urate was unlikely to play an important role in protecting neurons by quenching oxygen radicals [66]. Other studies have suggested that urate may not have substantial anti-oxidant effects. Oxidative stress has been assessed by measurement of F2-Isoprostanes (F2-IsoPs) and protein carbonyls (PC); and lowering urate levels to 1 mg/dL in patients with refractory gout by administration of pegloticase did not change concentrations of either F2-IsoP or PC. The authors of this study concluded that urate is not a major factor controlling oxidative stress in vivo [67]. Additional studies have either failed to demonstrate a neuroprotective effect of urate in humans and/or suggested adverse actions. A longitudinal study in 1598 healthy elderly people showed that the risk of dementia, particularly vascular or mixed dementia, may be increased with high serum urate levels [64]. However, a systematic review and meta-analysis indicated inconsistent relationships between all types of dementia and serum urate levels [68]. Evidence related to a potential anti-oxidant and organ-protective effect of urate, and thus a potential risk of urate lowering, are mixed and the limited prospective data available provides no clear support. Conversely, the pro-inflammatory effects of urate may outweigh the potential anti-inflammatory effects, particularly when above the limit S5 of solubility (>6.8 mg/dL). Hyperuricemia and its pro-inflammatory effect has been linked to adverse metabolic, cardiovascular, and renal effects. Consequences of hyperuricemia Metabolic effects Higher serum urate levels have been shown to be associated with increased risk for the development of diabetes [37,69] and the metabolic syndrome [70 72]. High serum urate is also associated with elevated body mass index, hypercholesterolemia, hypertriglyceridemia, increased fasting plasma glucose, and insulin resistance [73 75]. This consistent evidence has illuminated the dose effect of urate on the risk for metabolic syndrome, and subsequently may contribute to the adverse cardiovascular effects of elevated urate levels [76,77]. Cardiovascular effects There are well-known adverse cardiovascular effects of elevated serum urate levels [78]. Results from multiple observational studies have demonstrated significant associations between hyperuricemia and hypertension [79], cardiovascular and cerebrovascular events [80 84], and heart failure [85]. It has been speculated that elevated serum urate may contribute to cardiovascular disease via multiple mechanisms, including impaired NO production/endothelial dysfunction, increased vascular stiffness, elevated oxidative stress, endothelial cell apoptosis, and vascular, cardiac, and renal fibrosis [86 91]. A small number of prospective interventional studies have provided evidence that lower urate levels may decrease cardiovascular risk. It has been shown that administration of allopurinol in patients with chronic kidney disease decreased C-reactive protein, slowed the progression of renal disease, and reduced the risk for cardiovascular events [92]. It has also been demonstrated that treatment with febuxostat prevents arterial stiffening as measured by pulse wave velocity in patients with severe chronic refractory tophaceous gout [93]. Additionally, there is some evidence pegloticase and subsequent urate lowering effects decreases blood pressure in patients with chronic refractory gout, although further studies are needed to further support this [94]. Hypertension Results from multiple analyses have demonstrated a strong association between elevated urate levels and hypertension. A meta-analysis that included 25 studies (97,824 participants) indicated that hyperuricemia was associated with a significantly elevated risk for incident hypertension [95]. Similar results were reported in an earlier meta-analysis of 18 prospective cohort studies representing data from 55,607 participants [79]. Results from several studies, including adolescents and adults, have demonstrated that lowering serum urate with xanthine oxidase inhibitors and uricosurics in patients with either asymptomatic hyperuricemia or gout also decreases blood pressure [94,96 100]. Conversely, a recent study evaluating overweight and obese hypertensive patients with probenecid and allopurinol did not show any significant change in endothelial function or mean blood pressure [101]. It is important to recognize this patient population was not significantly hyperuricemic and whose mean serum urate was 6.1 mg/dL + 0.9 mg/ dL, thus possibly impacting the results in comparison study populations with a higher baseline serum urate level. Renal effects Elevated urate levels have been associated with a large number of changes in renal structure and function. Urate significantly increases toll-like receptor (TLR) 4, NOD-like receptor family pyrin domain containing 3 (NALP3), caspase-1, interleukin (IL) 1b, and S6 R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 intercellular adhesion molecule-1 expression in the human primary renal proximal tubule epithelial cells [102]. Urate also decreases the expression of E-cadherin in epithelial cells resulting in a loss of cellto-cell contact among renal tubular cells and decreased secretion of substances (e.g., NO) that increase renal blood flow [103]. Elevated urate can also lead to endothelial dysfunction, vascular smooth muscle cell proliferation, and increased IL-6 synthesis, all of which may contribute to the progression of chronic kidney disease (CKD) [104]. Patients with CKD stages 3 5 have higher serum urate levels that are associated with elevated systolic blood pressure, higher C-reactive protein levels, lower glomerular filtration rate, and lower flow-mediated dilatation [105]. Elevated serum urate has also been correlated with worsening renal function irrespective of age, gender, race, diabetes, hypertension, alcohol use, smoking, lipids, and baseline renal function [106]. A retrospective study including 12,751 patients with CKD stage 2, 3, and 4 and serum urate levels > 7 mg/dL over an 8-year period evaluated the impact of urate lowering on progression of CKD [107]. Of the 2690 patients on urate lowering therapy, 42% achieved a target serum urate of < 6 mg/dL. Those with CKD stage 2 and 3 achieving a target of < 6 mg/dL were 30% more likely to see an improvement in eGFR, while no difference was seen in patients with CKD stage 4. The evidence from in vitro, ex vivo, epidemiological and even some clinical studies have shown an effect of urate lowering on kidney function, but prospective evidence of intervention and slowing the progression of chronic kidney injury is limited. In a randomized, double-blinded, placebo-controlled trial, Kimura and colleagues evaluated the effects of febuxostat and lowering serum urate on the slope of the eGFR in asymptomatic hyperuricemia patient with stage 3 CKD [108]. After two years of follow-up, the study found no significant difference between the febuxostat group and placebo group, but it is noteworthy the placebo group only had a 1% decline in eGFR. This trial and similar trials have only included low risk patients that may not have had significant progression regardless, as noted by the placebo group [109]. Additional studies need to include higher-risk cohorts to have the statistical power and better evaluate the impact of lowering serum urate on kidney function decline. A subsequent study aimed to compare allopurinol and febuxostat versus standard of care, evaluating CKD stage 3 patients who were hyperuricemic (defined as > 7 mg/dL) [110]. Forty patients received allopurinol with a mean dose of 164 mg, 30 patients received febuxostat with a mean dose of 55 mg, and 71 patients received standard treatment. Over the course of the observation period that ranged from 24 to 87 months, the febuxostat group had significantly lower mean serum urate compared to both allopurinol and control, maintained a higher mean eGFR, and had a longer renal survival time free from disease progression. Although this study contradicts the findings of the aforementioned study, it begs the question of whether or not treating to target in both the febuxostat and allopurinol groups would have resulted in positive results in both groups. Fig. 3. Sequence of monosodium urate crystallization Adapted from Martillo MA et al. Curr Rheumatol Rep. 2014; 16:400. ]2]. Formation of MSU crystals, inflammation, and organ damage The process of MSU crystallization is similar to that for the formation of other crystals, and is thought to depend on both urate concentration and other factors (Fig. 3). It has been suggested that MSU remains in solution until an event that changes solubility (e.g., increased concentration and decreased temperature). This results in clustering of MSU molecules while still in solution. These clusters aggregate into crystal nuclei, providing the basis for additional crystal formation and growth. Local factors may promote nucleation and crystal growth (e.g., low pH, elevated calcium levels, organic molecules originating from cartilage or the synovium, albumin, antibodies, sodium ions) [2,3]. MSU crystal formation Pain and inflammation associated with MSU crystals Although the presence of hyperuricemia is essential for the formation of MSU crystals, only a fraction (2 6%) of hyperuricemic patients develop clinical gout. This may result from a variable biologic response to elevated urate or inadequate detection of the disease. Conversely, some patients have normal serum urate (6.0 mg/dL) at the time of an acute gout attack, indicating that the relationship between serum urate level and acute MSU crystallization is complex [2,111]. Urate levels decrease during an acute attack [112], possibly secondary to the uricosuric effects of inflammatory mediators and hormone production, such IL-6 and adrenocorticotropic hormone, respectively. MSU crystals activate a specific inflammatory cascade in leukocytes leading to production of IL-1 and IL-18 [113 117]. The innate immune system recognizes and responds to pathogens and to endogenous molecules released by host cells via detection of pathogenassociated and danger-associated molecular patterns (PAMPS and DAMPs respectively). The inflammation MSU crystals induce has been revealed to be more complex than initially appreciated. MSU crystals cause the release of reactive oxygen species and reactive nitrogen species are released from fibroblasts and fibroblast-like synoviocytes causing cell death [118]. In vitro studies have also shown R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 that MSU crystals (in addition to calcium pyrophosphate, calcium oxalate, and cysteine crystals) can cause direct cytotoxicity, inflammation, and inflammation-driven cytotoxicity. This crystal-induced inflammation is referred to as necroinflammation [118]. Necrotic cells release stimulatory molecules such as DAMPs, IL-1a, and other alarmins fueling the recruitment of innate immune cells such as neutrophils and macrophages into the synovium and surrounding soft tissue [119]. This response is mediated by pattern-recognition receptors that include TLRs, intracellular RIG-like helicases, and NOD-like receptors (NLRs). NLRs respond to PAMPs/DAMPs through the formation of the NLRP3 inflammasome. These are multimeric cytoplasmic protein complexes which act as molecular platforms for the activation of inflammatory caspases following stimulation by foreign agonists. A typical inflammasome is composed of an NLR, an adaptor protein such as apoptosis-associated specklike protein containing a CARD and an effector caspase that activates proinflammatory cytokines, in particular IL-1b. MSU crystals (and uric acid) may act as a ‘danger’ signal to cells in a similar way to microbial pathogens and result in the activation of inflammasomes [118,120,121]. Caspase-1 cleaves pro-IL-1b and pro-IL-8 to generate IL-1b and IL-18, permitting the release of these cytokines into the extracellular space [114,122]. Once secreted, IL-1b leads to production of other chemokines and cytokines, resulting in the recruitment of leukocytes. Intracellular IL-1b signals for inflammation via activation of nuclear factor-kB (NFkB) and other inflammatory molecules [123,124]. While IL-1b is recognized as the chief cytokine in gout, both IL-1a and IL-8 may also play important roles in initiating inflammatory cascades in this disease [125]. Pain associated with MSU crystal deposition is mediated by several inflammatory substances that are formed after cell injury by crystals [126]. These molecules (prostaglandins, bradykinin, cytokines, substance P, transient receptor potential ankyrin 1) exert their effects through different receptor subtypes present in both peripheral sensory neurons and the spinal cord [127]. An important issue in considering the pain associated with gout is the variability in timing, extent, and duration of acute inflammatory responses in patients with this disease. Advanced imaging methods (i.e., high resolution ultrasound and highly S7 specific dual energy computed tomography) have shown that patients with MSU crystal deposits may remain asymptomatic and a large number of factors, such as long-chain fatty acids, complement, kinins, microRNA, IL-37, and neutrophil microvesicles may influence clinical expression of MSU crystal accumulation (Fig. 4) [128]. MSU crystals and inflammation in specific organ systems Joints: acute versus chronic joint pathology There is a close relationship between MSU crystal deposition and joint damage. In people with advanced gout, bone erosion is a frequent finding on plain radiography. In addition, other structural damage including joint space narrowing and features of new bone formation, such as bone sclerosis and spur formation, are frequently observed [129]. Factors contributing to joint damage include receptor activator of NFkB ligand (RANKL) expressing osteoclasts and T cells present within gouty tophus tissues, and infiltrating cells express inflammatory cytokines such as IL-1, IL-6, and tumor necrosis factora [130]. MSU crystals may also interact with articular tissues to influence the development of structural joint damage [131]. It has also been shown that MSU crystals decrease osteocyte viability and, through interactions with macrophages, promote a shift in osteocyte function that increases bone resorption and inflammation [132]. MSU crystals also may cause the death of chondrocytes via autophagy and/ or endoplasmic reticulum stress [133]. Cardiovascular system Silent deposits of MSU crystals are associated with more severe coronary calcification [134]. Dual energy computed tomography has demonstrated MSU deposition in the aortas and coronary arteries of patients with gout [135]. However, an epidemiologic study indicated that the presence of MSU crystals did not seem to result in further elevation of the already increased cardiovascular risk in hyperuricemic patients [136]. Fig. 4. Inflammatory responses to urate crystal deposits Adapted from Terkeltaub R. BMC Medicine 2017;15:158. [128]. S8 R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 Kidney Results from an in vitro study supported the conclusion that oxidative stress generated by MSU crystals promotes renal apoptosis through the mitochondrial caspase-dependent apoptosis pathway [137]. Another in vitro study indicated that MSU crystals are cytotoxic to renal cells and this cytotoxicity involves necroptosis as a form of regulated cell death in vitro and in vivo [138]. Results from in vitro studies of human embryonic kidney cells have shown that MSU crystals promote expression of reactive oxygen species, inducible nitric oxide synthase, and cyclo-oxgenase 2. In addition, enhanced caspase expression stimulated by MSU crystals resulted in renal cell apoptosis [137]. It has also been shown that incubation of MSU with human renal mesangial cells (HRMCs) increases intercellular adhesion molecule-1 expression and subsequent cell cell adhesion between HRMCs and monocytic cells. Infiltration of monocytes into the glomerular mesangium contributes to the development of glomerulonephritis [139]. Resolution of acute crystal induced inflammation Although chronic low grade inflammation is thought to be incited by the presence of MSU crystals, acute crystal induced inflammation, the gout flare, can begin to resolve in a few days, and typically resolves in less than 2 3 weeks without intervention. That said, without a counter response from the immune system itself, a cytokine storm could result [118]. The immune system is able to dampen the inflammation of a gout flare despite the nidus of the flare, the MSU crystal, still being present. Just as the inflammatory process is complex, so is its resolution. There are multiple factors and mediators in the involvement of the dampening of MSU crystal induced inflammation. The switching of pro-inflammatory to anti-inflammatory macrophages have been surmised to play a role [140]. Also, typical anti-inflammatory mediators of the immune system such as transforming growth factor beta (TGF-b), IL-10, IL-37, and melanocortin are implicated in acute gouty inflammation resolution [141 144]. Schauer and colleagues have shown that once the inflammatory process approaches crescendo, specifically, an abundance of neutrophils recruited to the site of inflammation, neutrophil extracellular traps (NETs) form, dampening of the inflammatory response and facilitating flare resolution [145]. Analysis of tophi have revealed extracellular DNA to be in direct contact with MSU crystals. The ingestion of MSU crystals by neutrophils causes cell death and rapid extrusion of cellular DNA results in NET formation, or NETosis. As the number of neutrophils are recruited, the NETs increase and cluster the crystals, forming aggregated NETs (aggNETs) [144]. This packaging of crystals and tophi formation is thought to be the tipping point from inflammation to resolution of a gout attack [144]. Functional studies using NETs have also shown their ability to quickly absorb, trap, cleave and degrade inflammatory mediators, thereby removing the fuel to the fire [145]. Conclusions The balance of urate formation and excretion is driven by multiple enzymatic pathways and genetic studies have suggested that multiple genetic polymorphisms may influence uric acid excretion. Urate has both pro- and anti-inflammatory effects. However, elevated serum levels of soluble urate have been shown to play a role in many disease states including gout and articular degenerative disorders as well as cardiovascular inflammation, atherosclerosis, and renal disease. Development of MSU crystals is influenced by urate concentration and a number of additional factors that may be targets for achieving and maintaining urate homeostasis. Urate, and its effect on our biology is complex, even paradoxical, yet this relatively underappreciated “endproduct” of purine metabolism still deserves more attention. Disclosure Robert T. Keenan has been or is a consultant with Horizon Therapeutics, Selecta Biosciences, Atom Biosciences, Dyve Biosciences, Sobi; and, has received research support from Sobi and Selecta Biosciences. References [1] Maiuolo J, Oppedisano F, Gratteri S, Muscoli C, Mollace V. Regulation of uric acid metabolism and excretion. Int J Cardiol 2016;213:8–14. [2] Martillo MA, Nazzal L, Crittenden DB. The crystallization of monosodium urate. Curr Rheumatol Rep 2014;16:400. [3] Chhana A, Pool B, Callon KE, Tay ML, Musson D, et al. Monosodium urate crystals reduce osteocyte viability and indirectly promote a shift in osteocyte function towards a proinflammatory and proresorptive state. Arthritis Res Ther 2018;20:208.  n-Vicente J. Uric acid and evolution. Rheumatology [4] Alvarez-Lario B, Macarro 2010;49:2010–5. [5] Johnson RJ, Titte S, Cade JR, Rideout BA, Oliver WJ. Uric acid, evolution and primitive cultures. Semin Nephrol 2005;25:3–8. [6] Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature 2003;425(6957):516–21. [7] Hu DE, Moore AM, Thomsen LL, Brindle KM. Uric acid promotes tumor immune rejection. Cancer Res. 2004;65(15):5059–62. [8] Hosomi A, Nakanishi T, Fujita T, Tamai I. Extra-renal elimination of uric acid via intestinal efflux transporter BCRP/ABCG2. PLoS One 2012;7:e30456. [9] Lee SJ, Terkeltaub RA, Kavanaugh A. Recent developments in diet and gout. Curr Opin Rheumatol 2006;18:193–8. [10] Lipkowitz MS. Regulation of uric acid excretion by the kidney. Curr Rheumatol Rep 2012;14:179–88. [11] Marangella M. Uric acid elimination in the urine. Pathophysiological implications. Contrib Nephrol. 2005;147:132–48. [12] Sakurai H. Urate transporters in the genomic era. Curr Opin Nephrol Hypertens 2013;22:545–50. mençon B, Lu € scher BP, Fine M, et al. Expression, purification, and structural [13] Cle insights for the human uric acid transporter, GLUT9, using the Xenopus laevis oocytes system. PLoS One 2014;9:e108852. [14] Wong K, Briddon SJ, Holliday ND, Kerr ID. Plasma membrane dynamics and tetrameric organisation of ABCG2 transporters in mammalian cells revealed by single particle imaging techniques. Biochim Biophys Acta 2016;1863:19–29. [15] Matsuo H, Nakayama A, Sakiyama M, et al. ABCG2 dysfunction causes hyperuricemia due to both renal urate underexcretion and renal urate overload. Sci Rep 2014;4:3755. [16] George RL, Keenan RT. Genetics of hyperuricemia and gout: implications for the present and future. Curr Rheumatol Rep 2013;15(2):309. [17] Li J, Badve S, Zhou ZL, Oh R, Lee M, Perkovic V, de Zeeuw D, Mahaffey K, Fulcer G, Matthews D, Neal B. The effects of canagliflozin on uric acid and gout in patients with type 2 diabetes in the CANVAS program. poster presentation at the 79th scientific session of the American Diabetes Association; June 7 11; 2019. [18] Sorensen LB. Role of the intestinal tract in the elimination of uric acid. Arthritis Rheum 1965;8(5):694–706. [19] Matsuo H, Takada T, Ichida K, Nakayama A, Ikebuchi Y, et al. Common defects of ABCG2, a high-capacity urate exporter, cause gout: a function-based genetic analysis in a Japanese population. Sci Transl Med 2009;1(5) 5ra11. doi: 10.1126/ scitranslmed.3000237. [20] Matsuo H, Ichida K, Takada T, Nakayama A, Nakashima H, et al. Common dysfunctional variants in ABCG2 are a major cause of early-onset gout. Sci Rep 2013;31:2014. [21] Zaidi F, Narang RK, Phipps-Green A, Gamble GG, Tausche AK, et al. Systematic genetic analysis of early-onset gout: ABCG2 is the only associated locus. Rheumatology 2020 pil:kez685 [Epub ahead of print}. [22] Hyndman D, Liu S, Miner JN. Urate handling in the human body. Curr Rheumatol Rep 2016;18:34. [23] Choi HK, Mount DB, Reginato AM. Pathogenesis of gout. Ann Intern Med 2005;143:499–516. [24] Khanna D, Fitzgerald JD, Khanna PP, et al. 2012 American College of Rheumatology guidelines for management of gout, part 1: systematic nonpharmacologic and pharmacologic therapeutic approaches to hyperuricemia. Arthritis Care Res. 2012;64:1431–46. [25] MacFarlane LA, Kim SC. Gout: a review of nonmodifiable and modifiable risk factors. Rheum Dis Clin N Am 2014;40:581–604. [26] Choi HK, Liu S, Curhan G. Intake of purine-rich foods, protein, and dairy products and relationship to serum levels of uric acid: the Third National Health and Nutrition Examination Survey. Arthritis Rheum 2005;52:283–9. [27] Osler W. The principles and practice of medicine. 4th ed. New York: D. Appleton and Company; 1901. [28] Choi JW, Ford ES, Gao X, Choi HK. Sugar-sweetened soft drinks, diet soft drinks, and serum uric acid level: the Third National Health and Nutrition Examination Survey. Arthritis Rheum 2008;59:109–16. [29] Siqueira JH, Mill JG, Velasquez-Melendez G, et al. Sugar-sweetened soft drinks and fructose consumption are associated with hyperuricemia: cross-sectional analysis from the Brazilian Longitudinal Study of Adult Health (ELSA-Brasil). Nutrients 2018;10:E981. R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 [30] Jamnik J, Rehman S, Blanco Mejia S, et al. Fructose intake and risk of gout and hyperuricemia: a systematic review and meta-analysis of prospective cohort studies. BMJ Open 2016;6:e013191. [31] Ebrahimpour-Koujan S, Saneei P, Larijani B, Esmaillzadeh A. Consumption of sugar sweetened beverages and dietary fructose in relation to risk of gout and hyperuricemia: a systematic review and meta-analysis. Crit Rev Food Sci Nutr 2018:1–10. [32] Ayoub-Charette S, Liu Q, Khan TA, Au-Yeung F, Blanco Mejia S, et al. Important food sources of fructose-containing sugars and incident gout: a systematic review and meta-analysis of prospective cohort studies. BMJ Open 2019;9: e024171. [33] Pillinger MH, Keenan RT. Update on the Management of Hyperuricemia and Gout. Bull NYU Hosp Jt Dis 2008;66(3):231–9. [34] Major TJ, Topless RK, Dalbeth N, Merriman TR. Evaluation of the diet wide contribution to serum urate levels: meta analysis of population based cohorts. BMJ 2018;363:k3951. [35] Major TJ, Dalbeth N, Stahl EA, Merriman TR. An update on the genetics of hyperuricaemia and gout. Nat Rev Rheumatol 2018;1:341–53. [36] Okada Y, Sim X, Go MJ, et al. Meta-analysis identifies multiple loci associated with kidney function-related traits in east Asian populations. Nat Genet 2012;44:904–9. [37] Dehghan A, van Hoek M, Sijbrands EJ, Hofman A, Witteman JC. High serum uric acid as a novel risk factor for type 2 diabetes. Diabetes Care 2008;31:361–2. [38] Anzai N, Jutabha P, Amonpatumrat-Takahashi S, Sakurai H. Recent advances in renal urate transport: characterization of candidate transporters indicated by genome-wide association studies. Clin Exp Nephrol 2012;16:89–95. [39] Tai V, Merriman TR, Dalbeth N. Genetic advances in gout: potential applications in clinical practice. Curr Opin Rheumatol 2019;31(2):144–51. [40] Merriman TR. An update on the genetic architecture of hyperuricemia and gout. Arthritis Res Ther 2015;17:98. €ttgen A, Albrecht E, Teumer A, et al. Genome-wide association analyses iden[41] Ko tify 18 new loci associated with serum urate concentrations. Nat Genet 2013;45:145–54. [42] Benn CL, Dua P, Gurrell R, Loudon P, Pike A, et al. Physiology of hyperuricemia and urate-lowering treatments. Front Med 2018;5:160. [43] Li C, Li Z, Liu S, Wang C, Han L, et al. Genome wide association analysis identifies three new risk loci for gout arthritis in Han Chinese. Nat Commun 2015;6:7041. [44] Kawamura Y, Nakaoka H, Nakayama A, et al. Genome-wide association study revealed novel loci which aggravate asymptomatic hyperuricaemia into gout. Ann Rheum Dis 2019;78:1430–7. [45] 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;78:6858–62. [46] Sautin YY, Johnson RJ. Uric acid: the oxidant-antioxidant paradox. Nucleosides Nucleotides Nucleic Acids 2008;27:608–19. [47] De Giorgi A, Fabbian F, Pala M, et al. Uric acid: friend or foe? Uric acid and cognitive function "Gout kills more wise men than simple". Eur Rev Med Pharmacol Sci 2015;19:640–6. [48] Vogel W, Broverman DM, Draguns JG. The role of glutamic acid in cognitive behaviors. Psychol Bull 1966;65:367–82. [49] Chamorro A, Planas AM, Muner DS, Deulofeu R. Uric acid administration for neuroprotection in patients with acute brain ischemia. Med Hypotheses 2004;62:173–6. [50] Chamorro A, Amaro S, Castellanos M, Segura T, Arenillas J, et al. Safety and efficacy of uric acid in patients with acute stroke (URICO-ICTUS): a randomised double-blind phase 2b/3 trial. Lancet Neurol 2014;13(5):453–60.  A, Liull L, Rudilosso S, et al. Uric acid therapy prevents [51] Amaro S, Laredo C, Remu early ischemic stroke progression a tertiary analysis of the URICO-ICTUS Trial (Efficacy study of combined treatment with uric acid and r-tPA in acute ischemic stroke. Stroke 2016;47(11):2874–6. [52] Liu CY, Xu Y, Cui LY, et al. Correlations between serum uric acid level and disease activity, intrathecal inflammation reactivity in patients with multiple sclerosis. Chin Med Sci J 2012;27:88–91. [53] Chen X, Wu G, Schwarzschild MA. Urate in Parkinson's disease: more than a biomarker? Curr Neurol Neurosci Rep 2012;12:367–75. rrez F, Iglesias F, et al. Serum uric acid levels in multiple scle[54] Guerrero AL, Gutie rosis patients inversely correlate with disability. Neurol Sci 2011;32:347–50. [55] Schlesinger I, Schlesinger N. Uric acid in Parkinson's disease. Mov Disord 2008;23:1653–7. [56] Kim TS, Pae CU, Yoon SJ, et al. Decreased plasma antioxidants in patients with Alzheimer’s disease. Int J Geriatr Psychiatry 2006;21(4):344–8. [57] Irizarry MC, Raman R, Schwarzschild MA, et al. Plasma urate and progression of mild cognitive impairment. Neurodegener Dis 2009;6(1 2):23–8. [58] Hooper DC, Spitsin S, Kean RB, et al. Uric acid, a natural scavenger of peroxynitrite, in experimental allergic encephalomyelitis and multiple sclerosis. Proc Natl Acad Sci USA 1998;95:675–80. ~ oz García D, Midaglia L, Martinez Vilela J, et al. Associated Inosine to inter[59] Mun feron: results of a clinical trial in multiple sclerosis. Acta Neurol Scand 2015;131:405–10. [60] Gonsette RE, Sindic C, D'hooghe MB, et al. Boosting endogenous neuroprotection in multiple sclerosis: the ASsociation of Inosine and Interferon beta in relapsingremitting Multiple Sclerosis (ASIIMS) trial. Mult Scler 2010;16:455–62. [61] Tana C, Ticinesi A, Prati B, Nouvenne A, Meschi T. Uric acid and cognitive function in older individuals. Nutrients 2018;10(8).  A, Bardin T, Perez-Ruiz F, Debette S, Richette P. Uric acid [62] Latourte A, Soumare and incident dementia over 12 years of follow-up: a population-based cohort study. Ann Rheum Dis 2018;77:328–35. S9 [63] Singh JA, Cleveland JD. Comparative effectiveness of allopurinol versus febuxostat for preventing incident dementia in older adults: a propensity-matched analysis. Arthritis Res Ther 2018;20(1):167. [64] Pasalic D, Marinkovic N, Feher-Turkovic L. Uric acid as one of the important factors in multifactorial disorders facts and controversies. Biochem Med 2012;22:63–75. [65] Sautin YY, Nakagawa T, Zharikov S, Johnson RJ. Adverse effects of the classic antioxidant uric acid in adipocytes: NADPH oxidase-mediated oxidative/nitrosative stress. Am J Physiol Cell Physiol 2007;293:C584–96. [66] Liu D, Yun Y, Yang D, et al. What is the biological function of uric acid? an antioxidant for neural protection or a biomarker for cell death. Dis Markers 2019;2019:4081962. [67] Hershfield MS, Roberts 2nd LJ, Ganson NJ, et al. Treating gout with pegloticase, a PEGylated urate oxidase, provides insight into the importance of uric acid as an antioxidant in vivo. Proc Natl Acad Sci USA 2010;107:14351–6. [68] Khan AA, Quinn TJ, Hewitt J, Fan Y, Dawson J. Serum uric acid level and association with cognitive impairment and dementia: systematic review and metaanalysis. Age 2016;38:16. [69] Johnson RJ, Nakagawa T, Sanchez-Lozada LG, et al. Sugar, uric acid, and the etiology of diabetes and obesity. Diabetes 2013;62:3307–15. [70] Sumiyoshi H, Ohyama Y, Imai K, Kurabayashi M, Saito Y, Nakamura T. Association of uric acid with incident metabolic syndrome in a Japanese general population. Int Heart J 2019;60:830–5. [71] Billiet L, Doaty S, Katz JD, Velasquez MT. Review of hyperuricemia as new marker for metabolic syndrome. ISRN Rheumatol 2014:852954. [72] Choi HK, Ford ES, Li C, Curhan G. Prevalence of the metabolic syndrome in patients with gout: the Third National Health and Nutrition Examination Survey. Arthritis Rheum 2007;57:109–15. [73] Kushiyama A, Tanaka K, Hara S, Kawazu S. Linking uric acid metabolism to diabetic complications. World J Diabetes 2014;5:787–95. [74] Lippi G, Montagnana M, Luca Salvagno G, Targher G, Cesare Guidi G. Epidemiological association between uric acid concentration in plasma, lipoprotein(a), and the traditional lipid profile. Clin Cardiol 2010;33:E76–80. [75] Mangge H, Zelzer S, Puerstner P, et al. Uric acid best predicts metabolically unhealthy obesity with increased cardiovascular risk in youth and adults. Obesity 2013;21:E71–7. [76] Liu CW, Chang WC, Lee CC, Chen KH, Wu YW, Hwang JJ. Hyperuricemia Is associated with a higher prevalence of metabolic syndrome in military individuals. Mil Med 2018;183:e391–5. [77] Liu CW, Chen KH, Tseng CK, Chang WC, Wu YW, Hwang JJ. The dose-response effects of uric acid on the prevalence of metabolic syndrome and electrocardiographic left ventricular hypertrophy in healthy individuals. Nutr Metab Cardiovasc Dis 2019;29:30–8. [78] Feig DI, Kang DH, Johnson RJ. Uric acid and cardiovascular risk. N Engl J Med 2008;359:1811–21. [79] Grayson PC, Kim SY, LaValley M, Choi HK. Hyperuricemia and incident hypertension: a systematic review and meta-analysis. Arthritis Care Res 2011;63:102–10. [80] Kim SY, Guevara JP, Kim KM, Choi HK, Heitjan DF, Albert DA. Hyperuricemia and coronary heart disease: a systematic review and meta-analysis. Arthritis Care Res 2010;62:170–80. [81] Strasak A, Ruttmann E, Brant L, et al. Serum uric acid and risk of cardiovascular mortality: a prospective long-term study of 83,683 Austrian men. Clin Chem 2008;54:273–84. [82] Kivity S, Kopel E, Maor E, Abu-Bachar F, Segev S, Sidi Y, Olchovsky D. Association of serum uric acid and cardiovascular disease in healthy adults. Am J Cardiol 2013;111:1146–51. [83] Hannawi S, AlSalmi I, Moller I, Naredo E. Uric acid is independent cardiovascular risk factor, as manifested by increased carotid intima-media thickness in rheumatoid arthritis patients. Clin Rheumatol 2017;36:1897–902. [84] Pagidipati NJ, Clare RM, Keenan RT, Chiswell K, Roe MT, Hess CN. Association of gout with long-term cardiovascular outcomes among patients with obstructive coronary artery disease. J Am Heart Assoc 2018;7(16):e009328. [85] Duan X, Ling F. Is uric acid itself a player or a bystander in the pathophysiology of chronic heart failure? Med Hypotheses 2008;70:578–81. [86] Aroor AR, McKarns S, Demarco VG, Jia G, Sowers JR. Maladaptive immune and inflammatory pathways lead to cardiovascular insulin resistance. Metabolism 2013;62:1543–52. [87] Puddu P, Puddu GM, Cravero E, Vizioli L, Muscari A. Relationships among hyperuricemia, endothelial dysfunction and cardiovascular disease: molecular mechanisms and clinical implications. J Cardiol 2012;59:235–42.  M, Pavasini R, Ferrari R. Uric acid and coronary [88] Biscaglia S, Ceconi C, Malagu artery disease: an elusive link deserving further attention. Int J Cardiol 2016;13:28–32. [89] Li P, Zhang L, Zhang M, Zhou C, Lin N. Uric acid enhances PKC-dependent eNOS phosphorylation and mediates cellular ER stress: a mechanism for uric acidinduced endothelial dysfunction. Int J Mol Med 2016;37:989–97. [90] Chaudhary K, Malhotra K, Sowers J, Aroor A. Uric Acid key ingredient in the recipe for cardiorenal metabolic syndrome. Cardiorenal Med 2013;3:208–20. [91] Ndrepepa G. Uric acid and cardiovascular disease. Clin Chim Acta 2018;484:150–63. [92] Goicoechea M, de Vinuesa SG, Verdalles U, et al. Effect of allopurinol in chronic kidney disease progression and cardiovascular risk. Clin J Am Soc Nephrol 2010;5:1388–93. [93] Tausche AK, Christoph M, Forkmann M, et al. As compared to allopurinol, uratelowering therapy with febuxostat has superior effects on oxidative stress and S10 [94] [95] [96] [97] [98] [99] [100] [101] [102] [103] [104] [105] [106] [107] [108] [109] [110] [111] [112] [113] [114] [115] [116] [117] [118] [119] [120] R.T. Keenan / Seminars in Arthritis and Rheumatism 50 (2020) S2 S10 pulse wave velocity in patients with severe chronic tophaceous gout. Rheumatol Int 2014;34:101–9. Johnson RJ, Choi HK, Yeo AE, Lipsky PE. Pegloticase treatment significantly decreases blood pressure in patients with chronic gout. Hypertension 2019;74:95–101. Wang J, Qin T, Chen J, Li Y, Wang L, Huang H, Li J. Hyperuricemia and risk of incident hypertension: a systematic review and meta-analysis of observational studies. PLoS One 2014;9:e114259. Kim HA, Seo YI, Song YW. Four-week effects of allopurinol and febuxostat treatments on blood pressure and serum creatinine level in gouty men. J Korean Med Sci 2014;29:1077–81. Liu P, Wang H, Zhang F, Chen Y, Wang D, Wang Y. The effects of allopurinol on the carotid intima-media thickness in patients with type 2 diabetes and asymptomatic hyperuricemia: a three-year randomized parallel-controlled study. Intern Med 2015;54:2129–37. Gunawardhana L, McLean L, Punzi HA, et al. effect of febuxostat on ambulatory blood pressure in subjects with hyperuricemia and hypertension: a phase 2 randomized placebo-controlled study. J Am Heart Assoc 2017;6:e006683. Hosoya T, Ishikawa T, Ogawa Y, Sakamoto R, Ohashi T. Multicenter, open-label study of long-term topiroxostat (FYX-051) administration in Japanese hyperuricemic patients with or without gout. Clin Drug Investig 2018;38:1135–43. Soletsky B, Feig DL. Uric acid reduction rectifies prehypertension in obese adolescents. Hypertension 2012;60(5):1148–56. Borgi L, McMullan C, Wohlhueter A, Curhan GC, Fisher ND, Forman JP. Effect of uric acid lowering agents on endothelial function: a randomized double-blind, placebo-controlled trial. Hypertension 2017;69(2):243–8. Xiao J, Zhang XL, Fu C, et al. Soluble uric acid increases NALP3 inflammasome and interleukin-1b expression in human primary renal proximal tubule epithelial cells through the Toll-like receptor 4-mediated pathway. Int J Mol Med 2015;35:1347–54. Ryoo JH, Choi JM, Oh CM, Kim MG. The association between uric acid and chronic kidney disease in Korean men: a 4-year follow-up study. J Korean Med Sci 2013;28:855–60. Siu YP, Leung KT, Tong MK, Kwan TH. Use of allopurinol in slowing the progression of renal disease through its ability to lower serum uric acid level. Am J Kidney Dis 2006;47:51–9. Kanbay M, Yilmaz MI, Sonmez A, et al. Serum uric acid level and endothelial dysfunction in patients with nondiabetic chronic kidney disease. Am J Nephrol 2011;33:298–304. Weiner DE, Tighiouart H, Elsayed EF, Griffith JL, Salem DN, Levey AS. Uric acid and incident kidney disease in the community. J Am Soc Nephrol 2008;19:1204–11. Levy G, Shi JM, Cheetham TC, Rashid N. Urate lowering therapy in moderate to severe chronic kidney disease. Perm J 2018;22:17–142. Kimura K, osova T, Uchida S, Inaba M, Makino H, et al. Febuxostat therapy for patients with stage 3 CKD and asymptomatic hyperuricemia: a randomized trial. Am J Kidney Dis 2018;72(6):798–810. Tiku A, Badvesv, Johnson DW. Urate-lowering therapy for preventing kidney disease progression: are we there yet? Am J Kidney Dis 2018;72(6):776–8. Lee JW, Lee KH. Comparison of renoprotective effects of febuxostat and allopurinol in hyperuricemic patients with chronic kidney disease. Int Urol Nephrol 2019;51:467–73. Schlesinger N, Norquist JM, Watson DJ. Serum urate during acute gout. J Rheumatol 2009;36:1287–9. Urano W, Yamanaka H, Tsutani H, et al. The inflammatory process in the mechanism of decreased serum uric acid concentrations during acute gouty arthritis. J Rheumatol 2002;29:1950–3. trilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid Martinon F, Pe crystals activate the NALP3 inflammasome. Nature 2006;440:237–41. Cronstein BN, Sunkureddi P. Mechanistic aspects of inflammation and clinical management of inflammation in acute gouty arthritis. J Clin Rheumatol 2013;19:19–29. Kingsbury SR, Conaghan PG, McDermott MF. The role of the NLRP3 inflammasome in gout. J Inflamm Res 2011;4:39–49. Cavalcanti NG, Marques CD, Lins E Lins TU, et al. Cytokine profile in gout: inflammation driven by IL-6 and IL-18? Immunol Investig 2016;45:383–95. Inokuchi T, Moriwaki Y, Tsutsui H, et al. Plasma interleukin (IL)-18 (interferongamma-inducing factor) and other inflammatory cytokines in patients with gouty arthritis and monosodium urate monohydrate crystal-induced secretion of IL-18. Cytokine 2006;33:21–7. Desai J, Steiger S, Anders HJ. Molecular pathophysiology of gout. Trends Mol Med 2017;23:756–68. Franklin BS, Mangan MS, Latz E. Crystal formation in inflammation. Annu Rev Immunol 2016;34:173–202. Liu-Bryan R, Scott P, Sydlaske A, Rose DM, Terkeltaub R. Innate immunity conferred by Toll-like receptors 2 and 4 and myeloid differentiation factor 88 [121] [122] [123] [124] [125] [126] [127] [128] [129] [130] [131] [132] [133] [134] [135] [136] [137] [138] [139] [140] [141] [142] [143] [144] [145] expression is pivotal to monosodium urate monohydrate crystal-in

Use Quizgecko on...
Browser
Browser