Skip to main content
Log in

How neutrophil extracellular traps orchestrate the local immune response in gout

  • Review
  • Published:
Journal of Molecular Medicine Aims and scope Submit manuscript

Abstract

Neutrophil granulocytes possess a large arsenal of pro-inflammatory substances and mechanisms that empower them to drive local acute immune reactions to invading microorganisms or endogenous inflammatory triggers. The use of this armory needs to be tightly controlled to avoid chronic inflammation and collateral tissue damage. In gout, inflammation arises from precipitation of uric acid in the form of needle-shaped monosodium urate crystals. Inflammasome activation by these crystals in local immune cells results in a rapid and dramatic recruitment of neutrophils. This neutrophil influx is accompanied by the infamously intense clinical symptoms of inflammation during an acute gout attack. Neutrophilic inflammation however is equipped with a built-in safeguard; activated neutrophils form neutrophil extracellular traps (NETs). At the very high neutrophil densities that occur at the site of inflammation, NETs build aggregates that densely pack the monosodium urate (MSU) crystals and trap and degrade pro-inflammatory mediators by inherent proteases. Local removal of cytokines and chemokines by aggregated NETs explains how acute inflammation can stop in the consistent presence of the inflammatory trigger. Aggregated NETs resemble early stages of the typical large MSU deposits that constitute the pathognomonic structures of gout, tophi. Although tophi contribute to muscosceletal damage and mortality in patients with chronic gout, they can therefore be considered as a payoff that is necessary to silence the intense inflammatory response during acute gout.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1

Similar content being viewed by others

References

  1. Takei H, Araki A, Watanabe H, Ichinose A, Sendo F (1996) Rapid killing of human neutrophils by the potent activator phorbol 12-myristate 13-acetate (PMA) accompanied by changes different from typical apoptosis or necrosis. J Leukocyte Biol 59:229–240

    CAS  PubMed  Google Scholar 

  2. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A (2004) Neutrophil extracellular traps kill bacteria. Science 303:1532–1535

    Article  CAS  PubMed  Google Scholar 

  3. Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y (2010) PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med 207:1853–1862

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Wang Y, Li M, Stadler S, Correll S, Li P, Wang D, Hayama R, Leonelli L, Han H, Grigoryev SA et al (2009) Histone hypercitrullination mediates chromatin decondensation and neutrophil extracellular trap formation. J Cell Biol 184:205–213

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Papayannopoulos V, Metzler KD, Hakkim A, Zychlinsky A (2010) Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol 191:677–691

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Metzler KD, Fuchs TA, Nauseef WM, Reumaux D, Roesler J, Schulze I, Wahn V, Papayannopoulos V, Zychlinsky A (2011) Myeloperoxidase is required for neutrophil extracellular trap formation: implications for innate immunity. Blood 117:953–959

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A (2007) Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 176:231–241

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Akong-Moore K, Chow OA, von Kockritz-Blickwede M, Nizet V (2012) Influences of chloride and hypochlorite on neutrophil extracellular trap formation. PLoS One 7, e42984. doi:10.1371/journal.pone.0042984

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD et al (2007) Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 13:463–469

    Article  CAS  PubMed  Google Scholar 

  10. Yipp BG, Petri B, Salina D, Jenne CN, Scott BN, Zbytnuik LD, Pittman K, Asaduzzaman M, Wu K, Meijndert HC et al (2012) Infection-induced NETosis is a dynamic process involving neutrophil multitasking in vivo. Nat Med 18:1386–1393

    Article  CAS  PubMed  Google Scholar 

  11. Pilsczek FH, Salina D, Poon KK, Fahey C, Yipp BG, Sibley CD, Robbins SM, Green FH, Surette MG, Sugai M et al (2010) A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol 185:7413–7425

    Article  CAS  PubMed  Google Scholar 

  12. Patel S, Kumar S, Jyoti A, Srinag BS, Keshari RS, Saluja R, Verma A, Mitra K, Barthwal MK, Krishnamurthy H et al (2010) Nitric oxide donors release extracellular traps from human neutrophils by augmenting free radical generation. Nitric Oxide: Biol Chem / Off J Nitric Oxide Soc 22:226–234

    Article  CAS  Google Scholar 

  13. Neeli I, Khan SN, Radic M (2008) Histone deimination as a response to inflammatory stimuli in neutrophils. J Immunol 180:1895–1902

    Article  CAS  PubMed  Google Scholar 

  14. Gupta AK, Joshi MB, Philippova M, Erne P, Hasler P, Hahn S, Resink TJ (2010) Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis-mediated cell death. FEBS Lett 584:3193–3197

    Article  CAS  PubMed  Google Scholar 

  15. Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Gross WL, Werb Z, Grone HJ, Brinkmann V, Jenne DE (2009) Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med 15:623–625

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Behnen M, Leschczyk C, Moller S, Batel T, Klinger M, Solbach W, Laskay T (2014) Immobilized immune complexes induce neutrophil extracellular trap release by human neutrophil granulocytes via FcgammaRIIIB and Mac-1. J Immunol 193:1954–1965

    Article  CAS  PubMed  Google Scholar 

  17. Kingsbury SR, Conaghan PG, McDermott MF (2011) The role of the NLRP3 inflammasome in gout. J Inflamm Res 4:39–49

    CAS  PubMed Central  PubMed  Google Scholar 

  18. Saffarzadeh M, Juenemann C, Queisser MA, Lochnit G, Barreto G, Galuska SP, Lohmeyer J, Preissner KT (2012) Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One 7, e32366. doi:10.1371/journal.pone.0032366

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Caudrillier A, Kessenbrock K, Gilliss BM, Nguyen JX, Marques MB, Monestier M, Toy P, Werb Z, Looney MR (2012) Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J Clin Investig 122:2661–2671

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Luo L, Zhang S, Wang Y, Rahman M, Syk I, Zhang E, Thorlacius H (2014) Proinflammatory role of neutrophil extracellular traps in abdominal sepsis. Am J Physiol Lung Cell Mol Physiol 307:L586–L596

    Article  CAS  PubMed  Google Scholar 

  21. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD Jr, Wrobleski SK, Wakefield TW, Hartwig JH, Wagner DD (2010) Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 107:15880–15885

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Kienhofer D, Hahn J, Schubert I, Reinwald C, Ipseiz N, Lang SC, Borras EB, Amann K, Sjowall C, Barron AE et al (2014) No evidence of pathogenic involvement of cathelicidins in patient cohorts and mouse models of lupus and arthritis. PLoS One 9, e115474. doi:10.1371/journal.pone.0115474

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, Meller S, Chamilos G, Sebasigari R, Riccieri V et al (2011) Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med 3:73ra19

    Article  PubMed Central  PubMed  Google Scholar 

  24. Hakkim A, Furnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE, Zychlinsky A (2010) Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A 107:9813–9818

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Jacob CO, Eisenstein M, Dinauer MC, Ming W, Liu Q, John S, Quismorio FP Jr, Reiff A, Myones BL, Kaufman KM et al (2012) Lupus-associated causal mutation in neutrophil cytosolic factor 2 (NCF2) brings unique insights to the structure and function of NADPH oxidase. Proc Natl Acad Sci U S A 109:E59–E67

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Barton LL, Johnson CR (1986) Discoid lupus erythematosus and X-linked chronic granulomatous disease. Pediatr Dermatol 3:376–379

    Article  CAS  PubMed  Google Scholar 

  27. Cale CM, Morton L, Goldblatt D (2007) Cutaneous and other lupus-like symptoms in carriers of X-linked chronic granulomatous disease: incidence and autoimmune serology. Clin Exp Immunol 148:79–84

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Lee BW, Yap HK (1994) Polyarthritis resembling juvenile rheumatoid arthritis in a girl with chronic granulomatous disease. Arthritis Rheum 37:773–776

    Article  CAS  PubMed  Google Scholar 

  29. Marks DJ, Miyagi K, Rahman FZ, Novelli M, Bloom SL, Segal AW (2009) Inflammatory bowel disease in CGD reproduces the clinicopathological features of Crohn’s disease. Am J Gastroenterol 104:117–124

    Article  CAS  PubMed  Google Scholar 

  30. El Kebir D, Filep JG (2013) Modulation of neutrophil apoptosis and the resolution of inflammation through beta2 integrins. Front Immunol 4:60

    Article  PubMed Central  PubMed  Google Scholar 

  31. Serhan CN, Savill J (2005) Resolution of inflammation: the beginning programs the end. Nat Immunol 6:1191–1197

    Article  CAS  PubMed  Google Scholar 

  32. Schauer C, Janko C, Munoz LE, Zhao Y, Kienhofer D, Frey B, Lell M, Manger B, Rech J, Naschberger E et al (2014) Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med 20:511–517

    Article  CAS  PubMed  Google Scholar 

  33. Hultqvist M, Olofsson P, Holmberg J, Backstrom BT, Tordsson J, Holmdahl R (2004) Enhanced autoimmunity, arthritis, and encephalomyelitis in mice with a reduced oxidative burst due to a mutation in the Ncf1 gene. Proc Natl Acad Sci U S A 101:12646–12651

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  34. Olofsson P, Holmberg J, Tordsson J, Lu S, Akerstrom B, Holmdahl R (2003) Positional identification of Ncf1 as a gene that regulates arthritis severity in rats. Nat Genet 33:25–32

    Article  CAS  PubMed  Google Scholar 

  35. Campbell AM, Kashgarian M, Shlomchik MJ (2012) NADPH oxidase inhibits the pathogenesis of systemic lupus erythematosus. Sci Transl Med 4:157ra141

    Article  PubMed Central  PubMed  Google Scholar 

  36. Simchowitz L, Atkinson JP, Spilberg I (1982) Stimulation of the respiratory burst in human neutrophils by crystal phagocytosis. Arthritis Rheum 25:181–188

    Article  CAS  PubMed  Google Scholar 

  37. Chen L, Hsieh MS, Ho HC, Liu YH, Chou DT, Tsai SH (2004) Stimulation of inducible nitric oxide synthase by monosodium urate crystals in macrophages and expression of iNOS in gouty arthritis. Nitric Oxide: Biol Chem / Off J Nitric Oxide Soc 11:228–236

    Article  CAS  Google Scholar 

  38. Chu SC, Yang SF, Tzang BS, Hsieh YS, Lue KH, Lu KH (2010) Cathepsin B and cystatin C play an inflammatory role in gouty arthritis of the knee. Clin Chim Acta Int J Clin Chem 411:1788–1792

    Article  CAS  Google Scholar 

  39. Schorn C, Frey B, Lauber K, Janko C, Strysio M, Keppeler H, Gaipl US, Voll RE, Springer E, Munoz LE et al (2011) Sodium overload and water influx activate the NALP3 inflammasome. J Biol Chem 286:35–41

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Busso N, So A (2012) Microcrystals as DAMPs and their role in joint inflammation. Rheumatology 51:1154–1160

    Article  CAS  PubMed  Google Scholar 

  41. Ng G, Sharma K, Ward SM, Desrosiers MD, Stephens LA, Schoel WM, Li T, Lowell CA, Ling CC, Amrein MW et al (2008) Receptor-independent, direct membrane binding leads to cell-surface lipid sorting and Syk kinase activation in dendritic cells. Immunity 29:807–818

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  42. Meissner F, Molawi K, Zychlinsky A (2008) Superoxide dismutase 1 regulates caspase-1 and endotoxic shock. Nat Immunol 9:866–872

    Article  CAS  PubMed  Google Scholar 

  43. Meissner F, Seger RA, Moshous D, Fischer A, Reichenbach J, Zychlinsky A (2010) Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease. Blood 116:1570–1573

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  44. Gabelloni ML, Sabbione F, Jancic C, Fuxman Bass J, Keitelman I, Iula L, Oleastro M, Geffner JR, Trevani AS (2013) NADPH oxidase derived reactive oxygen species are involved in human neutrophil IL-1beta secretion but not in inflammasome activation. Eur J Immunol 43:3324–3335

    Article  CAS  PubMed  Google Scholar 

  45. van de Veerdonk FL, Smeekens SP, Joosten LAB, Kullberg BJ, Dinarello CA, van der Meer JWM, Netea MG (2010) Reactive oxygen species-independent activation of the IL-1β inflammasome in cells from patients with chronic granulomatous disease. Proc Natl Acad Sci U S A 107:3030–3033

    Article  PubMed Central  PubMed  Google Scholar 

  46. So A, Busso N (2014) The concept of the inflammasome and its rheumatologic implications. Joint Bone Spine: Rev Rhum 81:398–402

    Article  CAS  Google Scholar 

  47. Mankan AK, Dau T, Jenne D, Hornung V (2012) The NLRP3/ASC/Caspase-1 axis regulates IL-1beta processing in neutrophils. Eur J Immunol 42:710–715

    Article  CAS  PubMed  Google Scholar 

  48. Harijith A, Ebenezer DL, Natarajan V (2014) Reactive oxygen species at the crossroads of inflammasome and inflammation. Front Physiol 5:352

    Article  PubMed Central  PubMed  Google Scholar 

  49. Apostolidou E, Skendros P, Kambas K, Mitroulis I, Konstantinidis T, Chrysanthopoulou A, Nakos K, Tsironidou V, Koffa M, Boumpas DT et al (2014) Neutrophil extracellular traps regulate IL-1beta-mediated inflammation in familial Mediterranean fever. Ann Rheum Dis. doi:10.1136/annrheumdis-2014-205958

    Google Scholar 

  50. Joosten LA, Netea MG, Fantuzzi G, Koenders MI, Helsen MM, Sparrer H, Pham CT, van der Meer JW, Dinarello CA, van den Berg WB (2009) Inflammatory arthritis in caspase 1 gene-deficient mice: contribution of proteinase 3 to caspase 1-independent production of bioactive interleukin-1beta. Arthritis Rheum 60:3651–3662

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Joosten LA, Netea MG, Mylona E, Koenders MI, Malireddi RK, Oosting M, Stienstra R, van de Veerdonk FL, Stalenhoef AF, Giamarellos-Bourboulis EJ et al (2010) Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1beta production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum 62:3237–3248

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A et al (2014) The adaptor ASC has extracellular and ‘prionoid’ activities that propagate inflammation. Nat Immunol 15:727–737

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Terkeltaub R, Baird S, Sears P, Santiago R, Boisvert W (1998) 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 41:900–909

    Article  CAS  PubMed  Google Scholar 

  54. Scanu A, Oliviero F, Gruaz L, Sfriso P, Pozzuoli A, Frezzato F, Agostini C, Burger D, Punzi L (2010) High-density lipoproteins downregulate CCL2 production in human fibroblast-like synoviocytes stimulated by urate crystals. Arthritis Res Ther 12:R23

    Article  PubMed Central  PubMed  Google Scholar 

  55. Schorn C, Strysio M, Janko C, Munoz LE, Schett G, Herrmann M (2010) The uptake by blood-borne phagocytes of monosodium urate is dependent on heat-labile serum factor(s) and divalent cations. Autoimmunity 43:236–238

    Article  CAS  PubMed  Google Scholar 

  56. Mitroulis I, Kambas K, Chrysanthopoulou A, Skendros P, Apostolidou E, Kourtzelis I, Drosos GI, Boumpas DT, Ritis K (2011) Neutrophil extracellular trap formation is associated with IL-1beta and autophagy-related signaling in gout. PLoS One 6, e29318. doi:10.1371/journal.pone.0029318

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Shah K, Spear J, Nathanson LA, McCauley J, Edlow JA (2007) Does the presence of crystal arthritis rule out septic arthritis? J Emerg Med 32:23–26

    Article  PubMed  Google Scholar 

  58. Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, Park D, Woodson RI, Ostankovich M, Sharma P et al (2009) Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 461:282–286

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  59. Bournazou I, Pound JD, Duffin R, Bournazos S, Melville LA, Brown SB, Rossi AG, Gregory CD (2009) Apoptotic human cells inhibit migration of granulocytes via release of lactoferrin. J Clin Investig 119:20–32

    CAS  PubMed Central  PubMed  Google Scholar 

  60. Dalbeth N, Pool B, Gamble GD, Smith T, Callon KE, McQueen FM, Cornish J (2010) Cellular characterization of the gouty tophus: a quantitative analysis. Arthritis Rheum 62:1549–1556

    Article  CAS  PubMed  Google Scholar 

  61. Dalbeth N, House ME, Horne A, Taylor WJ (2013) Reduced creatinine clearance is associated with early development of subcutaneous tophi in people with gout. BMC Musculoskelet Disord 14:363

    Article  PubMed Central  PubMed  Google Scholar 

  62. Yu TF, Gutman AB (1967) Principles of current management of primary gout. Am J Med Sci 254:893–907

    Article  CAS  PubMed  Google Scholar 

  63. Yamanaka H, Togashi R, Hakoda M, Terai C, Kashiwazaki S, Dan T, Kamatani N (1998) Optimal range of serum urate concentrations to minimize risk of gouty attacks during anti-hyperuricemic treatment. Adv Exp Med Biol 431:13–18

    CAS  PubMed  Google Scholar 

  64. Tramontini N, Huber C, Liu-Bryan R, Terkeltaub RA, Kilgore KS (2004) Central role of complement membrane attack complex in monosodium urate crystal-induced neutrophilic rabbit knee synovitis. Arthritis Rheum 50:2633–2639

    Article  PubMed  Google Scholar 

  65. Terkeltaub RA, Dyer CA, Martin J, Curtiss LK (1991) Apolipoprotein (apo) E inhibits the capacity of monosodium urate crystals to stimulate neutrophils. Characterization of intraarticular apo E and demonstration of apo E binding to urate crystals in vivo. J Clin Investig 87:20–26

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  66. Landis RC, Yagnik DR, Florey O, Philippidis P, Emons V, Mason JC, Haskard DO (2002) Safe disposal of inflammatory monosodium urate monohydrate crystals by differentiated macrophages. Arthritis Rheum 46:3026–3033

    Article  CAS  PubMed  Google Scholar 

  67. You M, Fischer M, Deeg MA, Crabb DW (2002) Ethanol induces fatty acid synthesis pathways by activation of sterol regulatory element-binding protein (SREBP). J Biol Chem 277:29342–29347

    Article  CAS  PubMed  Google Scholar 

  68. Perez-Ruiz F, Martin I, Canteli B (2007) Ultrasonographic measurement of tophi as an outcome measure for chronic gout. J Rheumatol 34:1888–1893

    PubMed  Google Scholar 

  69. Puig JG, de Miguel E, Castillo MC, Rocha AL, Martinez MA, Torres RJ (2008) Asymptomatic hyperuricemia: impact of ultrasonography. Nucleosides, Nucleotides Nucleic Acids 27:592–595

    Article  CAS  PubMed  Google Scholar 

  70. Chhana A, Callon KE, Pool B, Naot D, Gamble GD, Dray M, Pitto R, Bentley J, McQueen FM, Cornish J et al (2013) The effects of monosodium urate monohydrate crystals on chondrocyte viability and function: implications for development of cartilage damage in gout. J Rheumatol 40:2067–2074

    Article  CAS  PubMed  Google Scholar 

  71. Chhana A, Callon KE, Pool B, Naot D, Watson M, Gamble GD, McQueen FM, Cornish J, Dalbeth N (2011) Monosodium urate monohydrate crystals inhibit osteoblast viability and function: implications for development of bone erosion in gout. Ann Rheum Dis 70:1684–1691

    Article  CAS  PubMed  Google Scholar 

  72. Barthelemy CR, Nakayama DA, Carrera GF, Lightfoot RW Jr, Wortmann RL (1984) Gouty arthritis: a prospective radiographic evaluation of sixty patients. Skelet Radiol 11:1–8

    Article  CAS  Google Scholar 

  73. Rajan A, Aati O, Kalluru R, Gamble GD, Horne A, Doyle AJ, McQueen FM, Dalbeth N (2013) Lack of change in urate deposition by dual-energy computed tomography among clinically stable patients with long-standing tophaceous gout: a prospective longitudinal study. Arthritis Res Ther 15:R160

    Article  PubMed Central  PubMed  Google Scholar 

  74. Scott JT, Holloway VP, Glass HI, Arnot RN (1969) Studies of uric acid pool size and turnover rate. Ann Rheum Dis 28:366–373

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Fiddis RW, Vlachos N, Calvert PD (1983) Studies of urate crystallisation in relation to gout. Ann Rheum Dis 42(Suppl 1):12–15

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  76. Liote F (2011) Treatment of hyperuricemia, gout and other crystalline arthritidies. Reumatismo 63:276–283

    CAS  Google Scholar 

  77. Biermann MHC, Araujo EG, Maueröder C, Lell M, Schett G, Manger B, Herrmann M, Rech J, Munoz LE (2014) Dual-energy CT in gout: an update exemplified by selected clinical cases. Gout Hyperuricemia 1:122–126

    Google Scholar 

  78. Araujo EG, Bayat S, Petsch C, Englbrecht M, Faustini F, Kleyer A, Hueber AJ, Cavallaro A, Lell M, Dalbeth N et al. (2015) Tophus resolution with pegloticase– a prospective dual energy computed tomography study. RMD Open in press

  79. Huang X, Du H, Gu J, Zhao D, Jiang L, Li X, Zuo X, Liu Y, Li Z, Li X et al (2014) An allopurinol-controlled, multicenter, randomized, double-blind, parallel between-group, comparative study of febuxostat in Chinese patients with gout and hyperuricemia. Int J Rheum Dis 17:679–686

    Article  CAS  PubMed  Google Scholar 

  80. Sarawate CA, Patel PA, Schumacher HR, Yang W, Brewer KK, Bakst AW (2006) Serum urate levels and gout flares: analysis from managed care data. J Clin Rheumat: Pract Rep Rheum Musculoskelet Dis 12:61–65

    Article  Google Scholar 

  81. Riedel AA, Nelson M, Joseph-Ridge N, Wallace K, MacDonald P, Becker M (2004) Compliance with allopurinol therapy among managed care enrollees with gout: a retrospective analysis of administrative claims. J Rheumatol 31:1575–1581

    PubMed  Google Scholar 

  82. Sundy JS, Ganson NJ, Kelly SJ, Scarlett EL, Rehrig CD, Huang W, Hershfield MS (2007) Pharmacokinetics and pharmacodynamics of intravenous PEGylated recombinant mammalian urate oxidase in patients with refractory gout. Arthritis Rheum 56:1021–1028

    Article  CAS  PubMed  Google Scholar 

  83. Zhang W, Doherty M, Bardin T, Pascual E, Barskova V, Conaghan P, Gerster J, Jacobs J, Leeb B, Liote F et al (2006) EULAR evidence based recommendations for gout. Part II: management. Report of a task force of the EULAR Standing Committee for International Clinical Studies Including Therapeutics (ESCISIT). Ann Rheum Dis 65:1312–1324

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  84. Schlesinger N (2014) Anti-interleukin-1 therapy in the management of gout. Curr Rheumatol Rep 16:398

    Article  PubMed  Google Scholar 

  85. Edwards NL, So A (2014) Emerging therapies for gout. Rheum Dis Clin North Am 40:375–387

    Article  PubMed  Google Scholar 

  86. de Luca A, Smeekens SP, Casagrande A, Iannitti R, Conway KL, Gresnigt MS, Begun J, Plantinga TS, Joosten LA, van der Meer JW et al (2014) IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc Natl Acad Sci U S A 111:3526–3531

    Article  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the doctoral training program GRK1660 and the priority program SPP 1468 Osteoimmunology IMMUNOBONE of the German Research Foundation (DFG) and the IMI funded research project BTCure.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Markus H Hoffmann.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maueröder, C., Kienhöfer, D., Hahn, J. et al. How neutrophil extracellular traps orchestrate the local immune response in gout. J Mol Med 93, 727–734 (2015). https://doi.org/10.1007/s00109-015-1295-x

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00109-015-1295-x

Keywords

Navigation