Loading

JSM Arthritis

Gouty Arthritis: New Targets for Future Therapies

Mini Review | Open Access | Volume 1 | Issue 2

  • 1. Departament of Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Brazil
+ Show More - Show Less
Corresponding Authors
Flavio Almeida Amaral, Departament of Bioquímica e Imunologia, Instituto de Ciências Biológicas,Universidade Federal de Minas Gerais, Av AntônioCarlos, 6627, Bloco O4, Sala 202 - Belo Horizonte/Minas Gerais/Brazil
Abstract

Gout is a very painful arthritis that affects millions of people around the world. In the last decade, various mechanisms associated to uric acid crystals-induced joint inflammation have been described, including details related to the activation of the cytoplasmic sensor NLRP3 inflammasome that eventually leads to the maturation and release the cytokine IL-1β. The identification of these signalling pathways opens new therapeutic opportunities for the treatment of gout and other related diseases. Here, we review the main inflammatory characteristics of gout, exploring the main intracellular signalling molecules and cell types involved in this disease and point out potential targets to improve the therapeutic options for joint inflammation in gout.

Keywords

•    Gout
•    Inflammasome
•    IL-1β
•    Pain
•    Neutrophil

Citation

Batista NV, Teixeira MM, Amaral FA (2016) Gouty Arthritis: New Targets for Future Therapies. JSM Arthritis 1(2): 1010.

INTRODUCTION

Gout is the most common form of arthritis worldwide [1]. It is a consequence of the deposition of monosodium urate (MSU) crystals in the joints, mainly in individuals with chronic hyperuricemia. In these individuals, there may be oversaturation of urate and precipitation of crystals predominantly in peripheral joints and surrounding tissues. Acute gout attacks are extremely painful and lead to joint disability, although with self-limited inflammation. However, continuing and prolonged deposition of MSU crystals can result in irreversible joint damage, bone erosion, development of disfiguring subcutaneous tophi and permanent disability [2]. Moreover, gout is associated with several other conditions that affect longevity and well-being of patients, including the metabolic syndrome [3], and cardiovascular [4-6], and renal diseases [7], evidencing the complexity and severity of this inflammatory condition.

Multiple risk factors are associated with the incidence of gout, including sustained hyperuricemia due to genetic factors [8], or medication [9], that impair renal excretion of uric acid; purine-rich diet and alcohol consumption [10], age and sex, with higher prevalence in elderly individuals and men [11], comorbid conditions, such as obesity and chronic renal disorder [9], and local tissue characteristics, exemplified by the increased propensity for nucleation and growth of MSU crystals in osteoarthritis joints [12]. Here, we focus our attention on the main mechanisms associated to joint inflammation in gout, exploring the strategies for experimental studies and the new possibilities for the treatment of acute gout.

Pathogenesis of acute gout: mechanisms based on IL-1β production

The deposition of MSU crystals in the joints and surrounding tissues is the initial event for the gout attack, since the crystals are rapidly recognized by innate immune cells culminating in the release of several inflammatory mediators and chemoattractant molecules. MSU crystals-induced inflammation is driven mainly by the release of the mature form of the cytokine interleukin (IL)- 1β by synoviocytes (fibroblasts and macrophages) [13]. In in vivo conditions, these cells store pro-IL-1β (inactive form) in their cytoplasm and the active form is processed and released after the phagocytosis of MSU crystals. Importantly, the investigation of MSU crystals-induced IL-1β release in in vitro studies normally needs previously de novo synthesis of pro-IL-1β by the cells through Pattern Recognition Receptors (PRR) activation, such as lipopolysaccharides from Gram-negative bacteria binding to Toll-like receptor (TLR)-4 (priming process). Recently, we demonstrated that the cytokines TNF-α and MIF have important contribution to MSU crystals-induced inflammation through the production of pro-IL-1β both in vitro and in vivo [14,15]. Furthermore, the production of mature form of IL-1β by human fibroblasts was dependent on previous priming with serum amyloid A protein upon MSU crystals stimulation [16].

The most important machinery for the cleavage of pro-IL-1β is the assembly of the cytoplasmic macromolecular complex termed inflammasome. In gout, the oligomerization of the inflammasome NLRP3 (nucleotide binding domain and leucine rich repeat containing proteins – prototype NLRP3) occurs after the phagocytosis of MSU crystals, culminating in the recruitment of the adaptor protein ASC (apoptosis-associated speck-like protein containing CARD), which lead to the cleavage and activation of caspase-1 and subsequently the cleavage of pro-IL-1β [17,18]. Experimentally, macrophages obtained from mice deficient in components of NLRP3 inflammasome (NLRP3, ASC, or caspase-1) were unable to process and release active IL-1β in response to MSU crystals [19]. In addition, the injection of MSU crystals in different compartments (peritoneal and joint cavities) in these knockout mice caused reduced inflammatory responses, including decreased IL-1β release, neutrophil recruitment, tissue damage and pain when compared to wild-type mice [19,20]. Regarding the importance of NLRP3 in the human context, recent studies suggest that NLRP3 mutations contribute to the development of auto inflammatory diseases, including hereditary periodic fever syndrome. NLRP3 polymorphisms could be related to more susceptibility for gouty arthritis or even be genetic markers for gout. In accordance, there is an association between the variant rs3806268 in NLRP3 gene and the risk of primary gout in a Chinese population [21]. However, other studies have not shown any association between genetic variants in NLRP3 polymorphisms and the susceptibility to gouty arthritis [22,23].

The precise mechanisms for NLRP3 activation by crystallized structures, including MSU crystals, are still under investigation. However, it has been proposed that NLRP3 is a sensor for common cellular changes downstream of triggering events (phagocytosis of MSU crystals) such as reactive oxygen species (ROS) production [24], potassium efflux [25], and release of cathepsin B from the lysosome [26]. Once released, IL-1β stimulates the synthesis of several other inflammatory molecules, including chemokines responsible for neutrophil recruitment [27,28]. Experimentally, the depletion of resident macrophages decreased the accumulation of neutrophils in murine peritoneal cavity following MSU crystals stimulation [30]. Alternative to NLRP3/caspase-1-dependent mechanism, neutrophil-derived serine proteases (cathepsin G, elastase, and proteinase 3) or mast cell-derived serine proteases (granzyme A and chymase) can cleave pro-IL-1β into a secreted and biologically active form of IL-1β [31-33]. Mast cells contribute to the early stages of MSU crystals-induced inflammation. Depletion of endogenous mast cells in mice has been found to significantly inhibits neutrophil influx in MSU crystals-induced peritonitis [34]. Furthermore, the presence of resident mast cells is associated to increased acute tissue swelling following intra-articular injection of MSU crystals in mice in a mechanism dependent on IL-1β production [35]. However, although mast cells are able to produce IL-1β in a NLRP3-dependent way through lipopolysaccharide stimulation [36], is not clear if the production of this cytokine in gout is also NLRP3-dependent or occurs through specific proteases [33].

Neutrophils quickly migrate to the joint following stimulation of MSU crystals in a mechanism dependent on the production of IL-1β. Neutrophil swarm to the joint occurs through the production of several chemoattractants, including CXCR1/2 binding chemokines, leukotriene B4, platelet-activating factor and calcium-binding proteins (S100A8 and S100A9) [37-42]. The importance of neutrophils to the pathogenesis of acute gout has been demonstrated experimentally by depletion of neutrophils or targeting their chemoattractants agents, resulting in reduced tissue inflammation [43-45]. Moreover, needle-shaped MSU crystals are found inside neutrophils [46]. Once migrated to the joint, neutrophils could contribute to the additional production of IL-1β [47-49]. However, the production of IL-1β by these cells seems not to be entirely dependent on inflammasome/ caspase-1 activation, since other proteases compensated the loss of caspase-1 in vivo [49]. Recently, it was demonstrated in vitro that murine neutrophils can indeed signal via the NLRP3 inflammasome, although only under soluble agonist’s stimulation and not by particulate/crystalline molecules [50]. This last finding has been validated in vivo, once neutrophils did not significantly contribute to alum-induced IL-1β production in mice. On the other hand, it was proposed that resident peritoneal macrophages, and not infiltrating neutrophils or monocytes, are the cell population responsible for IL-1β release at the initial stages of experimental gouty inflammation [30]. Thus, neutrophils contribute to acute joint dysfunction and damage, although further studies are needed to stablish the contribution of neutrophils as a relevant source of IL-1β and for chronic joint dysfunction and damage.

Experimental models of gout: basic research and pre-clinical studies

Experimental animal models are extremely useful to complement human investigations. In the context of gout, there are some models with important pathophysiological characteristics of gout, such as the activation of NLRP3 inflammasome for IL-1β production, recruitment of neutrophils, joint swelling and pain. Investigators have used distinct compartments in animals to inject MSU crystals, including air pouches, peritoneum, and joint. These compartments may yield different responses and may be studied indifferent species, although the majority of experiments have been conducted in mice.

Considering that gout attacks occurs in the joints, the closest models of gout consist on the injection of MSU crystals direct into the joint, either into tibiotarsal joint (ankle) [51], or into tibiofemoral (knee) joints [14,20,52,53]. In these models, there is a possibility to recover cells, especially neutrophils, from the joints and also the quantification of inflammatory molecules in surrounding tissue or even into the synovial cavity (although more difficult to obtain). Moreover, the clinical analysis can be performed through histopathology and joint pain bynociception assays, such as thermal (Hargreaves’ test) or mechanical (von Frey filaments) tests [54]. However, the uses of other compartments (air pouch and peritoneum) are also useful, mainly considering the higher exudate volume recovered for the analysis of inflammatory parameters.

In addition to the identification of new mechanisms associated to gouty arthritis, these proof-of-concept models are very useful for the studies of new targets for the treatment of gout, varying from NLRP3 inhibitors to blockers of neutrophil recruitment to the joint. In this regard, basic researchers need to improve the quality of these models. For instance, in vivo experimental systems only model acute aspects of gout, while the most aggressive form of the disease occurs when it becomes chronic, leading to permanent tissue damage and pain. Since mice produce uricase, an enzyme that cleaves uric acid, its blockage or the use of animal deficient of uricase is useful for the development of chronic models of gout. However, uricase-deficient mouse, although have hyperuricemia and its related nephropathy, dies before 4 weeks of age, unless they were treated with allopurinol [55]. In addition, gout is a disease related to metabolic syndrome, associated to inappropriate diet and life styles for long period. Thus, conditioning animals to the metabolic syndrome could represent more reliable models for the study of gout.

Perspectives for future therapies for gout

The current treatment for gout consist in decreasing joint inflammation and pain during acute flares and reducing hyperuricemia mainly during inter critical periods of gout. The therapeutic choices for the treatment of acute gout attacks include non-steroidal anti-inflammatory drugs (NSAIDs), colchicine and corticosteroids [56]. In the inter critical period, strategies aim to reduce or maintain serum uric acid levels below the saturation point, to dissolve existing crystals or prevent further crystal formation. The most common drugs for urate-lowering therapy are xanthine oxidase inhibitors, which reduce endogenous production of uric acid through inhibiting the conversion of hypoxanthine to xanthine and further xanthine to uric acid (allopurinol and febuxostat). Furthermore, uricosuric agents (benzbromarone, probenecid and sulfinpyrazone) prevent reuptake of uric acid at the proximal renal tubule and increase its renal excretion [57]. For most patients, the current therapies for acute gout is effective. However, many patients with chronic gout remaining unresponsive or do not tolerate these therapies. This is due to different factors including age, comorbidities, potential drug-drug interactions and risks of upper gastrointestinal ulceration and acute renal failure. Therefore, the development of alternative treatment strategies is highly necessary.

As mentioned, the NLRP3/caspase-1/IL-1β axis has a major role during the initial events of acute gout attacks, as evidenced experimentally using cell cultures, animal models or through clinical studies. Thus, it is reasonable to speculate that targeting those molecules may be useful for the treatment of gout when standard drugs are contraindicated or impractical. Among them, the most investigated target has been the neutralization of IL-1β with biologic drugs [58-63]. A number of therapeutic biological IL-1β inhibitors have been developed tested in different clinical trials and are currently clinically available. These include the dimeric fusion protein rilonacept (Arcalyst®) and a human monoclonal antibody called canakinumab (Ilaris®), which neutralise IL-1β directly. In addition, recombinant anakinra (Kineret®), an IL-1 receptor antagonist developed for the treatment of Rheumatoid Arthritis [64,65], is another option. Canakinumab has been recently licenced for the treatment of gout in Europe, but not in the USA. However, rilonacept, a drug initially used for treatment of Cryopyrin-Associated Periodic Syndromes (CAPS), syndromes associated with NLRP3 mutations, was recently rejected for gout treatment for FDA approval due to safety concerns [66].

Numerous promising inhibitors of NLRP3 inflammasome activation have been characterized. Although they could be useful for the treatment of gout, most studies still need further investigation before use. Probenecid is a uricosuric drug primarily used for the treatment of gout and other diseases associated with hyperuricemia [67]. Although this drug increases the rate of renal excretion of uric acid, it was discovered that probenecid also inhibits pannexin 1 channels (important for potassium efflux and consequently NLRP3 activation) [68]. MCC950, a diarylsulfonylurea-containing compound has been demonstrated to impair caspase-1-dependent processing of IL-1β through inhibiting both canonical and non-canonical activation of the NLRP3 inflammasome [69]. It was demonstrated that MCC950 can block ATP-, nigericin-, MSU crystals- and silica-induced IL-1β release from bone-marrow-derived macrophages, human monocyte-derived macrophages and peripheral blood mononuclear cells [70].

Parthenolide and Bay 11-7082, both NF-κB pathway inhibitors, have also been described as inhibitors of ATP-, nigericin- and MSU crystals-induced NLRP3 inflammasome activation in human THP-1 macrophages. Parthenolide can inhibit caspase-1 activation in response to NLRP3 stimulation or directly block NLRP3 by inhibiting its ATPase activity, a pathway required ATP-induced NLRP3 activation [71]. β-Hydroxybutyrate (BHB), a ketone body produced during fasting state, inhibits caspase-1 activation and IL-1β release in the presence of a large number of NLRP3 agonists, including ATP, nigericin, MSU crystals, and silica particles. In vivo, BHB attenuates caspase-1 activation and IL-1β secretion in a mouse model of urate crystal-induced peritonitis [72]. Finally, it was demonstrated that omega-3 fatty acid, through G protein-coupled receptor 120 (GPR120) and GPR40, also prevents NLRP3 inflammasome-dependent inflammation. LPS-primed BMDMs incubated with docosahexaenoic acid had decreased caspase-1 cleavage and IL-1β secretion following by MSU crystals, alum, ATP and nigericin stimulation. In vivo, omega-3 fatty acids reduced NLRP3-dependent high fat diet-induced type 2 diabetes [73]. Thus, there are clear evidences of these molecules in the inhibition of MSU crystals-induced NLPR3 activation, although further studies are necessary to establish the efficacy of these therapeutic strategies in the gout context.

The identification of endogenous mechanisms that control NLRP3 inflammasome activation or its protein synthesis open new window for the development of new targets for the treatment of gout and other related diseases. MicroRNAs (miRNAs) are noncoding RNAs responsible for post-transcriptional regulation of gene expression and have been intensively studied for therapeutic purposes [74]. The miRNA-223 has been showed to suppress NLRP3 inflammasome activity by control its protein expression through a conserved binding site within the 3’-UTR of NLRP3 transcript [75]. Although the latter study did not use MSU crystals as stimuli, the over expression of miRNA-223 in macrophages decreased the secretion of IL-1β following the activation of NLPR3 by ATP and nigericin [75]. Interestingly, the injection of miRNA-223 mimics reduced brain oedema and improved neurological functions in NLRP3-dependent rat model of intracerebral haemorrhage [76]. Downstream of NLRP3 activation, caspase-1 is also a target for miRNA. miRNA-135b can directly prevents caspase-1 protein expression in vitro and in vivo using a NLRP3-dependent model of smoking in mice [77]. Interestingly, the production of miR-135b in the cigarette-exposed mice is dependent on IL-1 receptor activation, demonstrating a negative feedback role of miRNA-135b to control lung inflammation.

CONCLUSION

Recent studies have clearly established canonical intracellular signalling pathways activated by MSU crystals in gout, especially mechanisms of activation of the NLRP3 inflammasome platform and subsequent maturation and release of IL-1β. This knowledge has opened new opportunities for the development of novel treatments for gout. However, it is still necessary that these are validated in gouty patients and, especially, whether they may be beneficial in chronic gout. Meanwhile, further information about the function of different cell types in gouty arthritis, their contribution to the initial and resolving phases of inflammation, and the improvement of current experimental models of gout certainly help to translate pre-clinical findings in to novel medications for patients.

ACKNOWLEDGEMENTS

This work was supported by grants from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG) e Coordenadoria de Aperfeiçoamento de Pessoal de Nível Superior (CAPES)

REFERENCES

1. Roddy E, Choi HK. Epidemiology of gout. Rheum Dis Clin North Am. 2014; 40: 155-175.

2. Dalbeth N, Haskard DO. Mechanisms of inflammation in gout. Rheumatology (Oxford). 2005; 44: 1090-1096.

3. Puig JG, Martinez MA. Hyperuricemia, gout and the metabolic syndrome. Curr Opin Rheumatol. 2008; 20: 187-191.

4. De Vera MA, Rahman MM, Bhole V, Kopec JA, Choi HK. Independent impact of gout on the risk of acute myocardial infarction among elderly women: a population-based study. Ann Rheum Dis. 2010; 69: 1162-1164.

5. Krishnan E, Baker JF, Furst DE, Schumacher HR. Gout and the risk of acute myocardial infarction. Arthritis Rheum. 2006; 54: 2688-2696.

6. Kuo CF, Yu KH, See LC, Chou IJ, Ko YS, Chang HC, et al. Risk of myocardial infarction among patients with gout: a nationwide population-based study. Rheumatology (Oxford). 2013; 52: 111-117.

7. Yu KH, Kuo CF, Luo SF, See LC, Chou IJ, Chang HC, et al. Risk of end-stage renal disease associated with gout: a nationwide population study. Arthritis Res Ther. 2012; 14: 83.

8. Sebesta I. Genetic disorders resulting in hyper- or hypouricemia. Adv Chronic Kidney Dis. 2012; 19: 398-403.

9. Pascual E, Perdiguero M. Gout, diuretics and the kidney. Ann Rheum Dis. 2006; 65: 981-982.

10. Fam AG. Gout: excess calories, purines, and alcohol intake and beyond. Response to a urate-lowering diet. J Rheumatol. 2005; 32: 773-777.

11. Kuo CF, Grainge MJ, Zhang W, Doherty M. Global epidemiology of gout: prevalence, incidence and risk factors. Nat Rev Rheumatol. 2015; 11: 649-662.

12. Roddy E, Zhang W, Doherty M. Are joints affected by gout also affected by osteoarthritis? Ann Rheum Dis. 2007; 66: 1374-1377.

13. Iwanaga T, Shikichi M, Kitamura H, Yanase H, Nozawa-Inoue K. Morphology and functional roles of synoviocytes in the joint. Arch Histol Cytol. 2000; 63: 17-31.

14. Amaral FA, Bastos LF, Oliveira TH, Dias AC, Oliveira VL, Tavares LD, et al. Transmembrane TNF-alpha is sufficient for articular inflammation and hypernociception in a mouse model of gout. Eur J Immunol. 2016; 46: 204-211.

15. Galvao I, Dias AC, Tavares LD, Rodrigues IP, Queiroz-Junior CM, Costa VV, et al. Macrophage migration inhibitory factor drives neutrophil accumulation by facilitating IL-1beta production in a murine model of acute gout. J Leukoc Biol. 2016; 99: 1035-1043.

16. Migita K, Koga T, Satomura K, Izumi M, Torigoshi T, Maeda Y, et al. Serum amyloid A triggers the mosodium urate -mediated mature interleukin-1beta production from human synovial fibroblasts. Arthritis Res Ther. 2012; 14: 119.

17. Leemans JC, Cassel SL, Sutterwala FS. Sensing damage by the NLRP3 inflammasome. Immunol Rev. 2011; 243: 152-162.

18. Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140: 821-832.

19. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006; 440: 237-241.

20. Amaral FA, Costa VV, Tavares LD, Sachs D, Coelho FM, Fagundes CT, et al. NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. Arthritis Rheum. 2012; 64: 474-484.

21. Deng J, Lin W, Chen Y, Wang X, Yin Z, Yao C, et al. rs3806268 of NLRP3 gene polymorphism is associated with the development of primary gout. Int J Clin Exp Pathol. 2015; 8: 13747-13752.

22. Meng DM, Zhou YJ, Wang L, Ren W, Cui LL, Han L, et al. Polymorphisms in the NLRP3 gene and risk of primary gouty arthritis. Mol Med Rep. 2013; 7: 1761-1766.

23. Wang LF, Ding YJ, Zhao Q, Zhang XL. Investigation on the association between NLRP3 gene polymorphisms and susceptibility to primary gout. Genet Mol Res. 2015; 14: 16410-16414.

24. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010; 11: 136-140.

25. Petrilli V, Papin S, Dostert C, Mayor A, Martinon F, Tschopp J. Activation of the NALP3 inflammasome is triggered by low intracellular potassium concentration. Cell Death Differ. 2007; 14: 1583-1589.

26. Hornung V, Bauernfeind F, Halle A, Samstad EO, Kono H, Rock KL, et al. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol. 2008; 9: 847-856.

27. Duff GW, Atkins E, Malawista SE. The fever of gout: urate crystals activate endogenous pyrogen production from human and rabbit mononuclear phagocytes. Trans Assoc Am Physicians. 1983; 96: 234- 245.

28. 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.

29. Terkeltaub R, Zachariae C, Santoro D, Martin J, Peveri P, Matsushima K. Monocyte-derived neutrophil chemotactic factor/interleukin-8 is a potential mediator of crystal-induced inflammation. Arthritis Rheum. 1991; 34: 894-903.

30. Martin WJ, Walton M, Harper J. Resident macrophages initiating and driving inflammation in a monosodium urate monohydrate crystal-induced murine peritoneal model of acute gout. Arthritis Rheum. 2009; 60: 281-289.

31. Black R, Kronheim S, Sleath P, Greenstreet T, Virca GD, March C, et al. The proteolytic activation of interleukin-1 beta. Agents Actions Suppl. 1991; 35: 85-89.

32. Hazuda DJ, Strickler J, Kueppers F, Simon PL, Young PR. Processing of precursor interleukin 1 beta and inflammatory disease. J Biol Chem. 1990; 265: 6318-6322.

33. Afonina IS, Muller C, Martin SJ, Beyaert R. Proteolytic Processing of Interleukin-1 Family Cytokines: Variations on a Common Theme. Immunity. 2015; 42: 991-1004.

34. Getting SJ, Flower RJ, Parente L, de Medicis R, Lussier A, Woliztky BA, et al. Molecular determinants of monosodium urate crystal-induced murine peritonitis: a role for endogenous mast cells and a distinct requirement for endothelial-derived selectins. J Pharmacol Exp Ther. 1997; 283: 123-130

35. Reber LL, Marichal T, Sokolove J, Starkl P, Gaudenzio N, Iwakura Y, et al. Contribution of mast cell-derived interleukin-1beta to uric acid crystal-induced acute arthritis in mice. Arthritis Rheumatol. 2014; 66: 2881-2891.

36. Nakamura Y, Kambe N, Saito M, Nishikomori R, Kim YG, Murakami M, et al. Mast cells mediate neutrophil recruitment and vascular leakage through the NLRP3 inflammasome in histamine-independent urticaria. J Exp Med. 2009; 206: 1037-1046.

37. Ryckman C, McColl SR, Vandal K, de Medicis R, Lussier A, Poubelle PE, et al. Role of S100A8 and S100A9 in neutrophil recruitment in response to monosodium urate monohydrate crystals in the air-pouch model of acute gouty arthritis. Arthritis Rheum. 2003; 48: 2310-2320.

38. Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW. Emerging mechanisms of neutrophil recruitment across endothelium. Trends Immunol. 2011; 32: 461-469.

39. Terkeltaub R, Baird S, Sears P, Santiago R, Boisvert W. The murine homolog of the interleukin-8 receptor CXCR-2 is essential for the occurrence of neutrophilic inflammation in the air pouch model of acute urate crystal-induced gouty synovitis. Arthritis Rheum. 1998; 41: 900-909.

40. Fujiwara K, Ohkawara S, Takagi K, Yoshinaga M, Matsukawa A. Involvement of CXC chemokine growth-related oncogene-alpha in monosodium urate crystal-induced arthritis in rabbits. Lab Invest. 2002; 82: 1297-1304.

41. Nishimura A, Akahoshi T, Takahashi M, Takagishi K, Itoman M, Kondo H, et al. Attenuation of monosodium urate crystal-induced arthritis in rabbits by a neutralizing antibody against interleukin-8. J Leukoc Biol. 1997; 62: 444-449.

42. Miguelez R, Palacios I, Navarro F, Gutierrez S, Sanchez-Pernaute O, Egido J, et al. Anti-inflammatory effect of a PAF receptor antagonist and a new molecule with antiproteinase activity in an experimental model of acute urate crystal arthritis. J Lipid Mediat Cell Signal. 1996; 13: 35-49.

43. Phelps P, Mc Carty DJ. Crystal-induced inflammation in canine joints. II. Importance of polymorphonuclear leukocytes. J Exp Med. 1966; 124: 115-126.

44. Chang YH, Garalla EJ. Suppression of urate crystal-induced canine joint inflammation by heterologous anti-polymorphonuclear leukocyte serum. Arthritis Rheum. 1968; 11: 145-150.

45. Becker MA. Clinical aspects of monosodium urate monohydrate crystal deposition disease (gout). Rheum Dis Clin North Am. 1988; 14: 377-394.

46. Agudelo CA, Schumacher HR. The synovitis of acute gouty arthritis. A light and electron microscopic study. Hum Pathol. 1973; 4: 265-279.

47. Roberge CJ, Grassi J, De Medicis R, Frobert Y, Lussier A, Naccache PH, et al. Crystal-neutrophil interactions lead to interleukin-1 synthesis. Agents Actions. 1991; 34: 38-41.

48. Roberge CJ, de Medicis R, Dayer JM, Rola-Pleszczynski M, Naccache PH, Poubelle PE. Crystal-induced neutrophil activation. V. Differential production of biologically active IL-1 and IL-1 receptor antagonist. J Immunol. 1994; 152: 5485-5494.

49. Guma M, Ronacher L, Liu-Bryan R, Takai S, Karin M, Corr M. Caspase 1-independent activation of interleukin-1beta in neutrophil-predominant inflammation. Arthritis Rheum. 2009; 60: 3642-3650.

50. Chen KW, Bezbradica JS, Gross CJ, Wall AA, Sweet MJ, Stow JL, et al. The murine neutrophil NLRP3 inflammasome is activated by soluble but not particulate or crystalline agonists. Eur J Immunol. 2016; 46: 1004-1010.

51. Torres R, Macdonald L, Croll SD, Reinhardt J, Dore A, Stevens S, et al. Hyperalgesia, synovitis and multiple biomarkers of inflammation are suppressed by interleukin 1 inhibition in a novel animal model of gouty arthritis. Ann Rheum Dis. 2009; 68: 1602-1608.

52. Galvao I, Dias AC, Tavares LD, Rodrigues IP, Queiroz-Junior CM, Costa VV, et al. Macrophage migration inhibitory factor drives neutrophil accumulation by facilitating IL-1beta production in a murine model of acute gout. J Leukoc Biol. 2016; 1035-1043.

53. Vieira AT, Macia L, Galvao I, Martins FS, Canesso MC, Amaral FA, et al. A Role for Gut Microbiota and the Metabolite-Sensing Receptor GPR43 in a Murine Model of Gout. Arthritis Rheumatol. 2015; 67: 1646-1656.

54. Amaral FA, Boff D, Teixeira MM. In Vivo Models to Study Chemokine Biology. Methods Enzymol. 2016; 570: 261-280.

55. Wu X, Wakamiya M, Vaishnav S, Geske R, Montgomery C, Jones P, et al. Hyperuricemia and urate nephropathy in urate oxidase-deficient mice. Proc Natl Acad Sci USA. 1994; 91: 742-746.

56. Terkeltaub R. Update on gout: new therapeutic strategies and options. Nat Rev Rheumatol. 2010; 6: 30-38.

57. Rees F, Hui M, Doherty M. Optimizing current treatment of gout. Nat Rev Rheumatol. 2014; 10: 271-283.

58. So A, De Smedt T, Revaz S, Tschopp J. A pilot study of IL-1 inhibition by anakinra in acute gout. Arthritis Res Ther. 2007; 9: 28.

59. Chen K, Fields T, Mancuso CA, Bass AR, Vasanth L. Anakinra’s efficacy is variable in refractory gout: report of ten cases. Semin Arthritis Rheum. 2010; 40: 210-214.

60. Schlesinger N, De Meulemeester M, Pikhlak A, Yucel AE, Richard D, Murphy V, et al. Canakinumab relieves symptoms of acute flares and improves health-related quality of life in patients with difficult-to-treat Gouty Arthritis by suppressing inflammation: results of a randomized, dose-ranging study. Arthritis Res Ther. 2011; 13: 53.

61. So A, De Meulemeester M, Pikhlak A, Yucel AE, Richard D, Murphy V, et al. Canakinumab for the treatment of acute flares in difficult-to-treat gouty arthritis: Results of a multicenter, phase II, dose-ranging study. Arthritis Rheum. 2010; 62: 3064-3076.

62. Schumacher HR, Evans RR, Saag KG, Clower J, Jennings W, Weinstein SP, et al. Rilonacept (interleukin-1 trap) for prevention of gout flares during initiation of uric acid-lowering therapy: results from a phase III randomized, double-blind, placebo-controlled, confirmatory efficacy study. Arthritis Care Res (Hoboken). 2012; 64: 1462-1470.

63. Schumacher HR, Sundy JS, Terkeltaub R, Knapp HR, Mellis SJ, Stahl N, et al. Rilonacept (interleukin-1 trap) in the prevention of acute gout flares during initiation of urate-lowering therapy: results of a phase II randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2012; 64: 876-884.

64. Mitroulis I, Skendros P, Ritis K. Targeting IL-1beta in disease; the expanding role of NLRP3 inflammasome. Eur J Intern Med. 2010; 21: 157-163.

65. Dubois EA, Rissmann R, Cohen AF. Rilonacept and canakinumab. Br J Clin Pharmacol. 2011; 71: 639-641.

66. Kotz J. The gout pipeline crystallizes. Nat Rev Drug Discov. 2012; 11: 425-426.

67. Robinson PC, Dalbeth N. Advances in pharmacotherapy for the treatment of gout. Expert Opin Pharmacother. 2015; 16: 533-546.

68. Silverman W, Locovei S, Dahl G. Probenecid, a gout remedy, inhibits pannexin 1 channels. Am J Physiol Cell Physiol. 2008; 295: 761-767.

69. Perregaux DG, McNiff P, Laliberte R, Hawryluk N, Peurano H, Stam E, et al. Identification and characterization of a novel class of interleukin-1 post-translational processing inhibitors. J Pharmacol Exp Ther. 2001; 299: 187-197.

70. Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015; 21: 248-255.

71. Juliana C, Fernandes-Alnemri T, Wu J, Datta P, Solorzano L, Yu JW, et al. Anti-inflammatory compounds parthenolide and Bay 11-7082 are direct inhibitors of the inflammasome. J Biol Chem. 2010; 285: 9792- 9802.

72. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015; 21: 263-269.

73. Yan Y, Jiang W, Spinetti T, Tardivel A, Castillo R, Bourquin C, et al. Omega-3 fatty acids prevent inflammation and metabolic disorder through inhibition of NLRP3 inflammasome activation. Immunity. 2013; 38: 1154-1163.

74. O’Connell RM, Rao DS, Chaudhuri AA, Baltimore D. Physiological and pathological roles for microRNAs in the immune system. Nat Rev Immunol. 2010; 10: 111-122.

75. Bauernfeind F, Rieger A, Schildberg FA, Knolle PA, Schmid-Burgk JL, Hornung V. NLRP3 inflammasome activity is negatively controlled by miR-223. J Immunol. 2012; 189: 4175-4181.

76. Yang Z, Zhong L, Xian R, Yuan B. MicroRNA-223 regulates inflammation and brain injury via feedback to NLRP3 inflammasome after intracerebral hemorrhage. Mol Immunol. 2015; 65: 267-276.

77. Halappanavar S, Nikota J, Wu D, Williams A, Yauk CL, Stampfli M. IL-1 receptor regulates microRNA-135b expression in a negative feedback mechanism during cigarette smoke-induced inflammation. J Immunol. 2013; 190: 3679-3686.

Batista NV, Teixeira MM, Amaral FA (2016) Gouty Arthritis: New Targets for Future Therapies. JSM Arthritis 1(2): 1010

Received : 21 Jun 2016
Accepted : 18 Jul 2016
Published : 20 Jul 2016
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Cancer Biology and Research
ISSN : 2373-9436
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X