Skip to main content
Erschienen in: Seminars in Immunopathology 4/2013

01.07.2013 | Review

Neutrophils, IL-1β, and gout: is there a link?

verfasst von: Ioannis Mitroulis, Konstantinos Kambas, Konstantinos Ritis

Erschienen in: Seminars in Immunopathology | Ausgabe 4/2013

Einloggen, um Zugang zu erhalten

Abstract

Gout is a prototype crystal-induced inflammatory disorder, characterized by neutrophil infiltration into inflamed joints. The identification of the role of NLRP3 inflammasome in the recognition of monosodium urate crystals and the subsequent release of IL-1β was a milestone in the elucidation of the pathogenesis of this disorder. IL-1β signaling is considered nowadays as the initiatory event that induces gouty inflammation and promotes the recruitment of vast numbers of neutrophils at the sites of inflammation. Crystal-induced neutrophil activation results in apoptosis inhibition, degranulation, superoxide production, cytokine release and, as recently described, formation of neutrophil extracellular traps, further amplifying the inflammatory process. Finally, neutrophil apoptosis and uptake of apoptotic material by macrophages drive the resolution of acute inflammation. In this review, we discuss the recent experimental data regarding the crosstalk between IL-1β and neutrophils in the pathogenesis of acute gout.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
2.
Zurück zum Zitat Annemans L, Spaepen E, Gaskin M et al (2008) Gout in the UK and Germany: prevalence, comorbidities and management in general practice 2000–2005. Ann Rheum Dis 67:960–966PubMedCrossRef Annemans L, Spaepen E, Gaskin M et al (2008) Gout in the UK and Germany: prevalence, comorbidities and management in general practice 2000–2005. Ann Rheum Dis 67:960–966PubMedCrossRef
3.
Zurück zum Zitat Campion EW, Glynn RJ, DeLabry LO (1987) Asymptomatic hyperuricemia. Risks and consequences in the Normative Aging Study. Am J Med 82:421–426PubMedCrossRef Campion EW, Glynn RJ, DeLabry LO (1987) Asymptomatic hyperuricemia. Risks and consequences in the Normative Aging Study. Am J Med 82:421–426PubMedCrossRef
4.
Zurück zum Zitat Terkeltaub R (2010) Update on gout: new therapeutic strategies and options. Nat Rev Rheumatol 6:30–38PubMedCrossRef Terkeltaub R (2010) Update on gout: new therapeutic strategies and options. Nat Rev Rheumatol 6:30–38PubMedCrossRef
5.
Zurück zum Zitat Agudelo CA, Schumacher HR (1973) The synovitis of acute gouty arthritis. A light and electron microscopic study. Hum Pathol 4:265–279PubMedCrossRef Agudelo CA, Schumacher HR (1973) The synovitis of acute gouty arthritis. A light and electron microscopic study. Hum Pathol 4:265–279PubMedCrossRef
6.
Zurück zum Zitat Duncan H, Bluhm GB, Riddle JM et al (1968) Synovial urate crystals in acute gouty arthritis. A study of their origin and significance. Clin Orthop Relat Res 59:277–285PubMedCrossRef Duncan H, Bluhm GB, Riddle JM et al (1968) Synovial urate crystals in acute gouty arthritis. A study of their origin and significance. Clin Orthop Relat Res 59:277–285PubMedCrossRef
7.
Zurück zum Zitat Malawista SE, de Boisfleury AC, Naccache PH (2011) Inflammatory gout: observations over a half-century. FASEB J 25:4073–4078PubMedCrossRef Malawista SE, de Boisfleury AC, Naccache PH (2011) Inflammatory gout: observations over a half-century. FASEB J 25:4073–4078PubMedCrossRef
8.
Zurück zum Zitat Mitroulis I, Skendros P, Ritis K (2010) Targeting IL-1beta in disease; the expanding role of NLRP3 inflammasome. Eur J Intern Med 21:157–163PubMedCrossRef Mitroulis I, Skendros P, Ritis K (2010) Targeting IL-1beta in disease; the expanding role of NLRP3 inflammasome. Eur J Intern Med 21:157–163PubMedCrossRef
9.
Zurück zum Zitat Masters SL, Simon A, Aksentijevich I et al (2009) Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease. Annu Rev Immunol 27:621–668PubMedCrossRef Masters SL, Simon A, Aksentijevich I et al (2009) Horror autoinflammaticus: the molecular pathophysiology of autoinflammatory disease. Annu Rev Immunol 27:621–668PubMedCrossRef
10.
Zurück zum Zitat Dinarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27:519–550PubMedCrossRef Dinarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27:519–550PubMedCrossRef
11.
Zurück zum Zitat Leemans JC, Cassel SL, Sutterwala FS (2011) Sensing damage by the NLRP3 inflammasome. Immunol Rev 243:152–162PubMedCrossRef Leemans JC, Cassel SL, Sutterwala FS (2011) Sensing damage by the NLRP3 inflammasome. Immunol Rev 243:152–162PubMedCrossRef
13.
Zurück zum Zitat Martinon F, Mayor A, Tschopp J (2009) The inflammasomes: guardians of the body. Annu Rev Immunol 27:229–265PubMedCrossRef Martinon F, Mayor A, Tschopp J (2009) The inflammasomes: guardians of the body. Annu Rev Immunol 27:229–265PubMedCrossRef
14.
Zurück zum Zitat Wang D, Zhang S, Li L et al (2010) Structural insights into the assembly and activation of IL-1β with its receptors. Nat Immunol 11:905–911PubMedCrossRef Wang D, Zhang S, Li L et al (2010) Structural insights into the assembly and activation of IL-1β with its receptors. Nat Immunol 11:905–911PubMedCrossRef
15.
Zurück zum Zitat Gabay C, Lamacchia C, Palmer G (2010) IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol 6:232–241PubMedCrossRef Gabay C, Lamacchia C, Palmer G (2010) IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol 6:232–241PubMedCrossRef
16.
Zurück zum Zitat Arend WP, Welgus HG, Thompson RC et al (1990) Biological properties of recombinant human monocyte-derived interleukin 1 receptor antagonist. J Clin Invest 85:1694–1697PubMedCrossRef Arend WP, Welgus HG, Thompson RC et al (1990) Biological properties of recombinant human monocyte-derived interleukin 1 receptor antagonist. J Clin Invest 85:1694–1697PubMedCrossRef
17.
Zurück zum Zitat Colotta F, Dower SK, Sims JE et al (1994) The type II 'decoy' receptor: a novel regulatory pathway for interleukin 1. Immunol Today 15:562–566PubMedCrossRef Colotta F, Dower SK, Sims JE et al (1994) The type II 'decoy' receptor: a novel regulatory pathway for interleukin 1. Immunol Today 15:562–566PubMedCrossRef
18.
Zurück zum Zitat Garlanda C, Anders HJ, Mantovani A (2009) TIR8/SIGIRR: an IL-1R/TLR family member with regulatory functions in inflammation and T cell polarization. Trends Immunol 30:439–446PubMedCrossRef Garlanda C, Anders HJ, Mantovani A (2009) TIR8/SIGIRR: an IL-1R/TLR family member with regulatory functions in inflammation and T cell polarization. Trends Immunol 30:439–446PubMedCrossRef
19.
Zurück zum Zitat Smith DE, Hanna R, Friend D et al (2003) The soluble form of IL-1 receptor accessory protein enhances the ability of soluble type II IL-1 receptor to inhibit IL-1 action. Immunity 18:87–96PubMedCrossRef Smith DE, Hanna R, Friend D et al (2003) The soluble form of IL-1 receptor accessory protein enhances the ability of soluble type II IL-1 receptor to inhibit IL-1 action. Immunity 18:87–96PubMedCrossRef
20.
Zurück zum Zitat Duff GW, Atkins E, Malawista SE (1983) The fever of gout: urate crystals activate endogenous pyrogen production from human and rabbit mononuclear phagocytes. Trans Assoc Am Physicians 96:234–245PubMed Duff GW, Atkins E, Malawista SE (1983) The fever of gout: urate crystals activate endogenous pyrogen production from human and rabbit mononuclear phagocytes. Trans Assoc Am Physicians 96:234–245PubMed
21.
Zurück zum Zitat di Giovine FS, Malawista SE, Thornton E (1991) 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 87:1375–1381PubMedCrossRef di Giovine FS, Malawista SE, Thornton E (1991) 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 87:1375–1381PubMedCrossRef
22.
Zurück zum Zitat Terkeltaub R, Zachariae C, Santoro D et al (1991) Monocyte-derived neutrophil chemotactic factor/interleukin-8 is a potential mediator of crystal-induced inflammation. Arthritis Rheum 34:894–903PubMedCrossRef Terkeltaub R, Zachariae C, Santoro D et al (1991) Monocyte-derived neutrophil chemotactic factor/interleukin-8 is a potential mediator of crystal-induced inflammation. Arthritis Rheum 34:894–903PubMedCrossRef
23.
Zurück zum Zitat Guerne PA, Terkeltaub R, Zuraw B et al (1989) Inflammatory microcrystals stimulate interleukin-6 production and secretion by human monocytes and synoviocytes. Arthritis Rheum 32:1443–1452PubMedCrossRef Guerne PA, Terkeltaub R, Zuraw B et al (1989) Inflammatory microcrystals stimulate interleukin-6 production and secretion by human monocytes and synoviocytes. Arthritis Rheum 32:1443–1452PubMedCrossRef
24.
Zurück zum Zitat Yagnik DR, Hillyer P, Marshall D et al (2000) Noninflammatory phagocytosis of monosodium urate monohydrate crystals by mouse macrophages. Implications for the control of joint inflammation in gout. Arthritis Rheum 43:1779–1789PubMedCrossRef Yagnik DR, Hillyer P, Marshall D et al (2000) Noninflammatory phagocytosis of monosodium urate monohydrate crystals by mouse macrophages. Implications for the control of joint inflammation in gout. Arthritis Rheum 43:1779–1789PubMedCrossRef
25.
Zurück zum Zitat Landis RC, Yagnik DR, Florey O et al (2002) Safe disposal of inflammatory monosodium urate monohydrate crystals by differentiated macrophages. Arthritis Rheum 46:3026–3033PubMedCrossRef Landis RC, Yagnik DR, Florey O et al (2002) Safe disposal of inflammatory monosodium urate monohydrate crystals by differentiated macrophages. Arthritis Rheum 46:3026–3033PubMedCrossRef
26.
Zurück zum Zitat Martin WJ, Walton M, Harper J (2009) Resident macrophages initiating and driving inflammation in a monosodium urate monohydrate crystal-induced murine peritoneal model of acute gout. Arthritis Rheum 60:281–289PubMedCrossRef Martin WJ, Walton M, Harper J (2009) Resident macrophages initiating and driving inflammation in a monosodium urate monohydrate crystal-induced murine peritoneal model of acute gout. Arthritis Rheum 60:281–289PubMedCrossRef
27.
Zurück zum Zitat Roberge CJ, Grassi J, De Medicis R et al (1991) Crystal–neutrophil interactions lead to interleukin-1 synthesis. Agents Actions 34:38–41PubMedCrossRef Roberge CJ, Grassi J, De Medicis R et al (1991) Crystal–neutrophil interactions lead to interleukin-1 synthesis. Agents Actions 34:38–41PubMedCrossRef
28.
Zurück zum Zitat Roberge CJ, de Medicis R, Dayer JM et al (1994) Crystal-induced neutrophil activation. V. Differential production of biologically active IL-1 and IL-1 receptor antagonist. J Immunol 152:5485–5494PubMed Roberge CJ, de Medicis R, Dayer JM et al (1994) Crystal-induced neutrophil activation. V. Differential production of biologically active IL-1 and IL-1 receptor antagonist. J Immunol 152:5485–5494PubMed
29.
Zurück zum Zitat Liu-Bryan R, Scott P, Sydlaske A et al (2005) Innate immunity conferred by toll-like receptors 2 and 4 and myeloid differentiation factor 88 expression is pivotal to monosodium urate monohydrate crystal-induced inflammation. Arthritis Rheum 52:2936–2946PubMedCrossRef Liu-Bryan R, Scott P, Sydlaske A et al (2005) Innate immunity conferred by toll-like receptors 2 and 4 and myeloid differentiation factor 88 expression is pivotal to monosodium urate monohydrate crystal-induced inflammation. Arthritis Rheum 52:2936–2946PubMedCrossRef
30.
Zurück zum Zitat Scott P, Ma H, Viriyakosol S et al (2006) Engagement of CD14 mediates the inflammatory potential of monosodium urate crystals. J Immunol 177:6370–6378PubMed Scott P, Ma H, Viriyakosol S et al (2006) Engagement of CD14 mediates the inflammatory potential of monosodium urate crystals. J Immunol 177:6370–6378PubMed
31.
Zurück zum Zitat Chen CJ, Shi Y, Hearn A et al (2006) MyD88-dependent IL-1 receptor signaling is essential for gouty inflammation stimulated by monosodium urate crystals. J Clin Invest 116:2262–2271PubMedCrossRef Chen CJ, Shi Y, Hearn A et al (2006) MyD88-dependent IL-1 receptor signaling is essential for gouty inflammation stimulated by monosodium urate crystals. J Clin Invest 116:2262–2271PubMedCrossRef
32.
Zurück zum Zitat Joosten LA, Netea MG, Mylona E et al (2010) Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum 62:3237–3248PubMedCrossRef Joosten LA, Netea MG, Mylona E et al (2010) Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum 62:3237–3248PubMedCrossRef
33.
Zurück zum Zitat Mylona EE, Mouktaroudi M, Crisan TO et al (2012) Enhanced interleukin-1β production of PBMCs from patients with gout after stimulation with Toll-like receptor-2 ligands and urate crystals. Arthritis Res Ther 14:R158PubMedCrossRef Mylona EE, Mouktaroudi M, Crisan TO et al (2012) Enhanced interleukin-1β production of PBMCs from patients with gout after stimulation with Toll-like receptor-2 ligands and urate crystals. Arthritis Res Ther 14:R158PubMedCrossRef
34.
Zurück zum Zitat Martinon F, Petrilli V, Mayor A et al (2006) Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440:237–241PubMedCrossRef Martinon F, Petrilli V, Mayor A et al (2006) Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440:237–241PubMedCrossRef
35.
Zurück zum Zitat Hornung V, Bauernfeind F, Halle A et al (2008) Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 9:847–856PubMedCrossRef Hornung V, Bauernfeind F, Halle A et al (2008) Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat Immunol 9:847–856PubMedCrossRef
36.
Zurück zum Zitat Amaral FA, Costa VV, Tavares LD et al (2012) NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. Arthritis Rheum 64:474–484PubMedCrossRef Amaral FA, Costa VV, Tavares LD et al (2012) NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. Arthritis Rheum 64:474–484PubMedCrossRef
37.
Zurück zum Zitat Scanu A, Oliviero F, Ramonda R et al (2012) Cytokine levels in human synovial fluid during the different stages of acute gout: role of transforming growth factor β1 in the resolution phase. Ann Rheum Dis 71:621–624PubMedCrossRef Scanu A, Oliviero F, Ramonda R et al (2012) Cytokine levels in human synovial fluid during the different stages of acute gout: role of transforming growth factor β1 in the resolution phase. Ann Rheum Dis 71:621–624PubMedCrossRef
38.
Zurück zum Zitat Torres R, Macdonald L, Croll SD et al (2009) Hyperalgesia, synovitis and multiple biomarkers of inflammation are suppressed by interleukin 1 inhibition in a novel animal model of gouty arthritis. Ann Rheum Dis 68:1602–1608PubMedCrossRef Torres R, Macdonald L, Croll SD et al (2009) Hyperalgesia, synovitis and multiple biomarkers of inflammation are suppressed by interleukin 1 inhibition in a novel animal model of gouty arthritis. Ann Rheum Dis 68:1602–1608PubMedCrossRef
39.
Zurück zum Zitat Schlesinger N, Alten RE, Bardin T et al (2012) Canakinumab for acute gouty arthritis in patients with limited treatment options: results from two randomised, multicentre, active-controlled, double-blind trials and their initial extensions. Ann Rheum Dis 71(11):1839–1849. doi:10.1136/annrheumdis-2011-200908 PubMedCrossRef Schlesinger N, Alten RE, Bardin T et al (2012) Canakinumab for acute gouty arthritis in patients with limited treatment options: results from two randomised, multicentre, active-controlled, double-blind trials and their initial extensions. Ann Rheum Dis 71(11):1839–1849. doi:10.​1136/​annrheumdis-2011-200908 PubMedCrossRef
40.
Zurück zum Zitat Schumacher HR Jr, Sundy JS, Terkeltaub R et al (2012) 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 64:876–884PubMedCrossRef Schumacher HR Jr, Sundy JS, Terkeltaub R et al (2012) 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 64:876–884PubMedCrossRef
41.
Zurück zum Zitat McGonagle D, Tan AL, Shankaranarayana S et al (2007) Management of treatment resistant inflammation of acute on chronic tophaceous gout with anakinra. Ann Rheum Dis 66:1683–1684PubMedCrossRef McGonagle D, Tan AL, Shankaranarayana S et al (2007) Management of treatment resistant inflammation of acute on chronic tophaceous gout with anakinra. Ann Rheum Dis 66:1683–1684PubMedCrossRef
42.
Zurück zum Zitat Dalbeth N, Pool B, Gamble GD et al (2010) Cellular characterization of the gouty tophus: a quantitative analysis. Arthritis Rheum 62:1549–1556PubMedCrossRef Dalbeth N, Pool B, Gamble GD et al (2010) Cellular characterization of the gouty tophus: a quantitative analysis. Arthritis Rheum 62:1549–1556PubMedCrossRef
43.
Zurück zum Zitat Phelps P, McCarty DJ Jr (1966) Crystal-induced inflammation in canine joints. II. Importance of polymorphonuclear leukocytes. J Exp Med 124:115–126PubMedCrossRef Phelps P, McCarty DJ Jr (1966) Crystal-induced inflammation in canine joints. II. Importance of polymorphonuclear leukocytes. J Exp Med 124:115–126PubMedCrossRef
44.
Zurück zum Zitat Chang YH, Garalla EJ (1968) Suppression of urate crystal-induced canine joint inflammation by heterologous anti-polymorphonuclear leukocyte serum. Arthritis Rheum 11:145–150PubMedCrossRef Chang YH, Garalla EJ (1968) Suppression of urate crystal-induced canine joint inflammation by heterologous anti-polymorphonuclear leukocyte serum. Arthritis Rheum 11:145–150PubMedCrossRef
45.
Zurück zum Zitat Martin WJ, Harper JL (2010) Innate inflammation and resolution in acute gout. Immunol Cell Biol 88:15–19PubMedCrossRef Martin WJ, Harper JL (2010) Innate inflammation and resolution in acute gout. Immunol Cell Biol 88:15–19PubMedCrossRef
46.
Zurück zum Zitat Nathan C (2006) Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6:173–182PubMedCrossRef Nathan C (2006) Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6:173–182PubMedCrossRef
47.
Zurück zum Zitat Amulic B, Cazalet C, Hayes GL et al (2012) Neutrophil function: from mechanisms to disease. Annu Rev Immunol 30:459–489PubMedCrossRef Amulic B, Cazalet C, Hayes GL et al (2012) Neutrophil function: from mechanisms to disease. Annu Rev Immunol 30:459–489PubMedCrossRef
49.
Zurück zum Zitat Williams MR, Azcutia V, Newton G et al (2011) Emerging mechanisms of neutrophil recruitment across endothelium. Trends Immunol 32:461–469PubMedCrossRef Williams MR, Azcutia V, Newton G et al (2011) Emerging mechanisms of neutrophil recruitment across endothelium. Trends Immunol 32:461–469PubMedCrossRef
50.
Zurück zum Zitat Chavakis T (2012) Leucocyte recruitment in inflammation and novel endogenous negative regulators thereof. Eur J Clin Invest 42:686–691PubMedCrossRef Chavakis T (2012) Leucocyte recruitment in inflammation and novel endogenous negative regulators thereof. Eur J Clin Invest 42:686–691PubMedCrossRef
51.
Zurück zum Zitat Pekin TJ Jr, Zvaifler NJ (1964) Hemolytic complement in synovial fluid. J Clin Invest 43:1372–1382PubMedCrossRef Pekin TJ Jr, Zvaifler NJ (1964) Hemolytic complement in synovial fluid. J Clin Invest 43:1372–1382PubMedCrossRef
52.
Zurück zum Zitat Hunder GG, McDuffie FC, Mullen BJ (1977) Activation of complement components C3 and factor B in synovial fluids. J Lab Clin Med 89:160–171PubMed Hunder GG, McDuffie FC, Mullen BJ (1977) Activation of complement components C3 and factor B in synovial fluids. J Lab Clin Med 89:160–171PubMed
53.
Zurück zum Zitat Hasselbacher P (1979) Immunoelectrophoretic assay for synovial fluid C3 with correction for synovial fluid globulin. Arthritis Rheum 22:243–250PubMedCrossRef Hasselbacher P (1979) Immunoelectrophoretic assay for synovial fluid C3 with correction for synovial fluid globulin. Arthritis Rheum 22:243–250PubMedCrossRef
54.
Zurück zum Zitat Giclas PC, Ginsberg MH, Cooper NR (1979) Immunoglobulin G independent activation of the classical complement pathway by monosodium urate crystals. J Clin Invest 63:759–764PubMedCrossRef Giclas PC, Ginsberg MH, Cooper NR (1979) Immunoglobulin G independent activation of the classical complement pathway by monosodium urate crystals. J Clin Invest 63:759–764PubMedCrossRef
55.
Zurück zum Zitat Naff GB, Byers PH (1973) Complement as a mediator of inflammation in acute gouty arthritis.I. Studies on the reaction between human serum complement and sodium urate crystals. J Lab Clin Med 81:747–760PubMed Naff GB, Byers PH (1973) Complement as a mediator of inflammation in acute gouty arthritis.I. Studies on the reaction between human serum complement and sodium urate crystals. J Lab Clin Med 81:747–760PubMed
56.
Zurück zum Zitat Fields TR, Abramson SB, Weissmann G et al (1983) Activation of the alternative pathway of complement by monosodium urate crystals. Clin Immunol Immunopathol 26:249–257PubMedCrossRef Fields TR, Abramson SB, Weissmann G et al (1983) Activation of the alternative pathway of complement by monosodium urate crystals. Clin Immunol Immunopathol 26:249–257PubMedCrossRef
57.
Zurück zum Zitat Doherty M, Whicher JT, Dieppe PA (1983) Activation of the alternative pathway of complement by monosodium urate monohydrate crystals and other inflammatory particles. Ann Rheum Dis 42:285–291PubMedCrossRef Doherty M, Whicher JT, Dieppe PA (1983) Activation of the alternative pathway of complement by monosodium urate monohydrate crystals and other inflammatory particles. Ann Rheum Dis 42:285–291PubMedCrossRef
58.
Zurück zum Zitat Russell IJ, Mansen C, Kolb LM et al (1982) Activation of the fifth component of human complement (C5) induced by monosodium urate crystals: C5 convertase assembly on the crystal surface. Clin Immunol Immunopathol 24:239–250PubMedCrossRef Russell IJ, Mansen C, Kolb LM et al (1982) Activation of the fifth component of human complement (C5) induced by monosodium urate crystals: C5 convertase assembly on the crystal surface. Clin Immunol Immunopathol 24:239–250PubMedCrossRef
59.
Zurück zum Zitat Tramontini N, Huber C, Liu-Bryan R et al (2004) Central role of complement membrane attack complex in monosodium urate crystal-induced neutrophilic rabbit knee synovitis. Arthritis Rheum 50:2633–2639PubMedCrossRef Tramontini N, Huber C, Liu-Bryan R et al (2004) Central role of complement membrane attack complex in monosodium urate crystal-induced neutrophilic rabbit knee synovitis. Arthritis Rheum 50:2633–2639PubMedCrossRef
60.
Zurück zum Zitat Nishimura A, Akahoshi T, Takahashi M et al (1997) Attenuation of monosodium urate crystal-induced arthritis in rabbits by a neutralizing antibody against interleukin-8. J Leukoc Biol 62:444–449PubMed Nishimura A, Akahoshi T, Takahashi M et al (1997) Attenuation of monosodium urate crystal-induced arthritis in rabbits by a neutralizing antibody against interleukin-8. J Leukoc Biol 62:444–449PubMed
61.
Zurück zum Zitat Terkeltaub R, Baird S, Sears P et al (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 41:900–909PubMedCrossRef Terkeltaub R, Baird S, Sears P et al (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 41:900–909PubMedCrossRef
62.
Zurück zum Zitat Ryckman C, McColl SR, Vandal K et al (2003) 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 48:2310–2320PubMedCrossRef Ryckman C, McColl SR, Vandal K et al (2003) 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 48:2310–2320PubMedCrossRef
63.
Zurück zum Zitat Cybulsky MI, Chan MK, Movat HZ (1988) Acute inflammation and microthrombosis induced by endotoxin, interleukin-1, and tumour necrosis factor and their implication in Gram-negative infection. Lab Invest 58:365–378PubMed Cybulsky MI, Chan MK, Movat HZ (1988) Acute inflammation and microthrombosis induced by endotoxin, interleukin-1, and tumour necrosis factor and their implication in Gram-negative infection. Lab Invest 58:365–378PubMed
64.
Zurück zum Zitat Chapman PT, Yarwood H, Harrison AA et al (1997) Endothelial activation in monosodium urate monohydrate crystal-induced inflammation: in vitro and in vivo studies on the roles of tumor necrosis factor a and interleukin-1. Arthritis Rheum 40:955–965PubMedCrossRef Chapman PT, Yarwood H, Harrison AA et al (1997) Endothelial activation in monosodium urate monohydrate crystal-induced inflammation: in vitro and in vivo studies on the roles of tumor necrosis factor a and interleukin-1. Arthritis Rheum 40:955–965PubMedCrossRef
65.
Zurück zum Zitat Chapman PT, Jamar F, Harrison AA et al (1994) Noninvasive imaging of E-selectin expression by activated endothelium in urate crystal-induced arthritis. Arthritis Rheum 37:1752–1756PubMedCrossRef Chapman PT, Jamar F, Harrison AA et al (1994) Noninvasive imaging of E-selectin expression by activated endothelium in urate crystal-induced arthritis. Arthritis Rheum 37:1752–1756PubMedCrossRef
66.
Zurück zum Zitat Chapman PT, Jamar F, Harrison AA et al (1996) Characterization of E-selectin expression, leukocyte traffic and clinical sequelae in urate crystal-induced inflammation: an insight into gout. Br J Rheumatol 35:323–334PubMedCrossRef Chapman PT, Jamar F, Harrison AA et al (1996) Characterization of E-selectin expression, leukocyte traffic and clinical sequelae in urate crystal-induced inflammation: an insight into gout. Br J Rheumatol 35:323–334PubMedCrossRef
67.
Zurück zum Zitat Schumacher HR, Phelps P (1971) Sequential changes in human polymorphonuclear leukocytes after urate crystal phagocytosis. An electron microscopic study. Arthritis Rheum 14:513–526PubMedCrossRef Schumacher HR, Phelps P (1971) Sequential changes in human polymorphonuclear leukocytes after urate crystal phagocytosis. An electron microscopic study. Arthritis Rheum 14:513–526PubMedCrossRef
68.
Zurück zum Zitat Cherian PV, Schumacher HR Jr (1986) Immunochemical and ultrastructural characterization of serum proteins associated with monosodium urate crystals (MSU) in synovial fluid cells from patients with gout. Ultrastruct Pathol 10:209–219PubMedCrossRef Cherian PV, Schumacher HR Jr (1986) Immunochemical and ultrastructural characterization of serum proteins associated with monosodium urate crystals (MSU) in synovial fluid cells from patients with gout. Ultrastruct Pathol 10:209–219PubMedCrossRef
69.
Zurück zum Zitat Brandt KD (1974) The effect of synovial hyaluronate on the ingestion of monosodium urate crystals by leukocytes. Clin Chim Acta 55:307–315PubMedCrossRef Brandt KD (1974) The effect of synovial hyaluronate on the ingestion of monosodium urate crystals by leukocytes. Clin Chim Acta 55:307–315PubMedCrossRef
70.
Zurück zum Zitat Barabe F, Gilbert C, Liao N et al (1998) Crystal-induced neutrophil activation VI. Involvement of FcgammaRIIIB (CD16) and CD11b in response to inflammatory microcrystals. FASEB J 12:209–220PubMed Barabe F, Gilbert C, Liao N et al (1998) Crystal-induced neutrophil activation VI. Involvement of FcgammaRIIIB (CD16) and CD11b in response to inflammatory microcrystals. FASEB J 12:209–220PubMed
71.
Zurück zum Zitat Popa-Nita O, Marois L, Paré G et al (2008) Crystal-induced neutrophil activation: X. Proinflammatory role of the tyrosine kinase Tec. Arthritis Rheum 58:1866–1876PubMedCrossRef Popa-Nita O, Marois L, Paré G et al (2008) Crystal-induced neutrophil activation: X. Proinflammatory role of the tyrosine kinase Tec. Arthritis Rheum 58:1866–1876PubMedCrossRef
72.
Zurück zum Zitat Desaulniers P, Fernandes M, Gilbert C et al (2001) Crystal-induced neutrophil activation. VII. Involvement of Syk in the responses to monosodium urate crystals. J Leukoc Biol 70:659–668PubMed Desaulniers P, Fernandes M, Gilbert C et al (2001) Crystal-induced neutrophil activation. VII. Involvement of Syk in the responses to monosodium urate crystals. J Leukoc Biol 70:659–668PubMed
73.
Zurück zum Zitat Popa-Nita O, Proulx S, Paré G et al (2009) Crystal-induced neutrophil activation: XI. Implication and novel roles of classical protein kinase C. J Immunol 183:2104–2114PubMedCrossRef Popa-Nita O, Proulx S, Paré G et al (2009) Crystal-induced neutrophil activation: XI. Implication and novel roles of classical protein kinase C. J Immunol 183:2104–2114PubMedCrossRef
74.
Zurück zum Zitat Popa-Nita O, Rollet-Labelle E, Thibault N et al (2007) Crystal-induced neutrophil activation. IX. Syk-dependent activation of class Ia phosphatidylinositol 3-kinase. J Leukoc Biol 82:763–773PubMedCrossRef Popa-Nita O, Rollet-Labelle E, Thibault N et al (2007) Crystal-induced neutrophil activation. IX. Syk-dependent activation of class Ia phosphatidylinositol 3-kinase. J Leukoc Biol 82:763–773PubMedCrossRef
75.
Zurück zum Zitat Mitroulis I, Kambas K, Chrysanthopoulou A et al (2011) Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One 6:e29318PubMedCrossRef Mitroulis I, Kambas K, Chrysanthopoulou A et al (2011) Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One 6:e29318PubMedCrossRef
76.
Zurück zum Zitat Mantovani A, Cassatella MA, Costantini C et al (2011) Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol 11:519–531PubMedCrossRef Mantovani A, Cassatella MA, Costantini C et al (2011) Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol 11:519–531PubMedCrossRef
77.
Zurück zum Zitat Kaplan MJ, Radic M (2012) Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol 189:2689–2695PubMedCrossRef Kaplan MJ, Radic M (2012) Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol 189:2689–2695PubMedCrossRef
78.
Zurück zum Zitat Popa-Nita O, Naccache PH (2010) Crystal-induced neutrophil activation. Immunol Cell Biol 88:32–40PubMedCrossRef Popa-Nita O, Naccache PH (2010) Crystal-induced neutrophil activation. Immunol Cell Biol 88:32–40PubMedCrossRef
79.
Zurück zum Zitat Regan E, Flannelly J, Bowler R et al (2005) Extracellular superoxide dismutase and oxidant damage in osteoarthritis. Arthritis Rheum 52:3479–3491PubMedCrossRef Regan E, Flannelly J, Bowler R et al (2005) Extracellular superoxide dismutase and oxidant damage in osteoarthritis. Arthritis Rheum 52:3479–3491PubMedCrossRef
80.
Zurück zum Zitat Ross AD, Banda NK, Muggli M et al (2004) Enhancement of collagen-induced arthritis in mice genetically deficient in extracellular superoxide dismutase. Arthritis Rheum 50:3702–3711PubMedCrossRef Ross AD, Banda NK, Muggli M et al (2004) Enhancement of collagen-induced arthritis in mice genetically deficient in extracellular superoxide dismutase. Arthritis Rheum 50:3702–3711PubMedCrossRef
81.
Zurück zum Zitat Chia EW, Grainger R, Harper JL (2008) Colchicine suppresses neutrophil superoxide production in a murine model of gouty arthritis: a rationale for use of low-dose colchicine. Br J Pharmacol 153:1288–1295PubMedCrossRef Chia EW, Grainger R, Harper JL (2008) Colchicine suppresses neutrophil superoxide production in a murine model of gouty arthritis: a rationale for use of low-dose colchicine. Br J Pharmacol 153:1288–1295PubMedCrossRef
82.
Zurück zum Zitat Naccache PH, Grimard M, Roberge CJ et al (1991) Crystal-induced neutrophil activation. I. Initiation and modulation of calcium mobilization and superoxide production by microcrystals. Arthritis Rheum 34:333–342PubMedCrossRef Naccache PH, Grimard M, Roberge CJ et al (1991) Crystal-induced neutrophil activation. I. Initiation and modulation of calcium mobilization and superoxide production by microcrystals. Arthritis Rheum 34:333–342PubMedCrossRef
83.
Zurück zum Zitat Abramson S, Hoffstein ST, Weissmann G (1982) Superoxide anion generation by human neutrophils exposed to monosodium urate. Arthritis Rheum 25:174–180PubMedCrossRef Abramson S, Hoffstein ST, Weissmann G (1982) Superoxide anion generation by human neutrophils exposed to monosodium urate. Arthritis Rheum 25:174–180PubMedCrossRef
84.
Zurück zum Zitat Martin WJ, Grainger R, Harrison A et al (2010) Differences in MSU-induced superoxide responses by neutrophils from gout subjects compared to healthy controls and a role for environmental inflammatory cytokines and hyperuricemia in neutrophil function and survival. J Rheumatol 37:1228–1235PubMedCrossRef Martin WJ, Grainger R, Harrison A et al (2010) Differences in MSU-induced superoxide responses by neutrophils from gout subjects compared to healthy controls and a role for environmental inflammatory cytokines and hyperuricemia in neutrophil function and survival. J Rheumatol 37:1228–1235PubMedCrossRef
85.
Zurück zum Zitat Torres R, Macdonald L, Croll SD et al (2009) Hyperalgesia, synovitis and multiple biomarkers of inflammation are suppressed by interleukin 1 inhibition in a novel animal model of gouty arthritis. Ann Rheum Dis 68:1602–1608PubMedCrossRef Torres R, Macdonald L, Croll SD et al (2009) Hyperalgesia, synovitis and multiple biomarkers of inflammation are suppressed by interleukin 1 inhibition in a novel animal model of gouty arthritis. Ann Rheum Dis 68:1602–1608PubMedCrossRef
86.
Zurück zum Zitat Foell D, Wittkowski H, Vogl T et al (2007) S100 proteins expressed in phagocytes: a novel group of damage-associated molecular pattern molecules. J Leukoc Biol 81:28–37PubMedCrossRef Foell D, Wittkowski H, Vogl T et al (2007) S100 proteins expressed in phagocytes: a novel group of damage-associated molecular pattern molecules. J Leukoc Biol 81:28–37PubMedCrossRef
87.
Zurück zum Zitat Ryckman C, Gilbert C, de Médicis R et al (2004) Monosodium urate monohydrate crystals induce the release of the proinflammatory protein S100A8/A9 from neutrophils. J Leukoc Biol 76:433–40PubMedCrossRef Ryckman C, Gilbert C, de Médicis R et al (2004) Monosodium urate monohydrate crystals induce the release of the proinflammatory protein S100A8/A9 from neutrophils. J Leukoc Biol 76:433–40PubMedCrossRef
88.
Zurück zum Zitat Liu R, Aupperle K, Terkeltaub R (2001) Src family protein tyrosine kinase signaling mediates monosodium urate crystal-induced IL-8 expression by monocytic THP-1 cells. J Leukoc Biol 70:961–968PubMed Liu R, Aupperle K, Terkeltaub R (2001) Src family protein tyrosine kinase signaling mediates monosodium urate crystal-induced IL-8 expression by monocytic THP-1 cells. J Leukoc Biol 70:961–968PubMed
89.
Zurück zum Zitat Hachicha M, Naccache PH, McColl SR (1995) Inflammatory microcrystals differentially regulate the secretion of macrophage inflammatory protein 1 and interleukin 8 by human neutrophils: a possible mechanism of neutrophil recruitment to sites of inflammation in synovitis. J Exp Med 182:2019–2025PubMedCrossRef Hachicha M, Naccache PH, McColl SR (1995) Inflammatory microcrystals differentially regulate the secretion of macrophage inflammatory protein 1 and interleukin 8 by human neutrophils: a possible mechanism of neutrophil recruitment to sites of inflammation in synovitis. J Exp Med 182:2019–2025PubMedCrossRef
90.
Zurück zum Zitat Schlesinger N, Thiele RG (2010) The pathogenesis of bone erosions in gouty arthritis. Ann Rheum Dis 69:1907–1912PubMedCrossRef Schlesinger N, Thiele RG (2010) The pathogenesis of bone erosions in gouty arthritis. Ann Rheum Dis 69:1907–1912PubMedCrossRef
91.
Zurück zum Zitat Braun T, Zwerina J (2011) Positive regulators of osteoclastogenesis and bone resorption in rheumatoid arthritis. Arthritis Res Ther 13:235PubMedCrossRef Braun T, Zwerina J (2011) Positive regulators of osteoclastogenesis and bone resorption in rheumatoid arthritis. Arthritis Res Ther 13:235PubMedCrossRef
92.
Zurück zum Zitat Poubelle PE, Chakravarti A, Fernandes MJ et al (2007) Differential expression of RANK, RANK-L, and osteoprotegerin by synovial fluid neutrophils from patients with rheumatoid arthritis and by healthy human blood neutrophils. Arthritis Res Ther 9:R25PubMedCrossRef Poubelle PE, Chakravarti A, Fernandes MJ et al (2007) Differential expression of RANK, RANK-L, and osteoprotegerin by synovial fluid neutrophils from patients with rheumatoid arthritis and by healthy human blood neutrophils. Arthritis Res Ther 9:R25PubMedCrossRef
93.
Zurück zum Zitat Allaeys I, Rusu D, Picard S et al (2011) Osteoblast retraction induced by adherent neutrophils promotes osteoclast bone resorption: implication for altered bone remodeling in chronic gout. Lab Invest 91:905–920PubMedCrossRef Allaeys I, Rusu D, Picard S et al (2011) Osteoblast retraction induced by adherent neutrophils promotes osteoclast bone resorption: implication for altered bone remodeling in chronic gout. Lab Invest 91:905–920PubMedCrossRef
94.
Zurück zum Zitat Bratton DL, Henson PM (2011) Neutrophil clearance: when the party is over, clean-up begins. Trends Immunol 32:350–357PubMedCrossRef Bratton DL, Henson PM (2011) Neutrophil clearance: when the party is over, clean-up begins. Trends Immunol 32:350–357PubMedCrossRef
95.
Zurück zum Zitat Scarlatti F, Granata R, Meijer AJ et al (2009) Does autophagy have a license to kill mammalian cells? Cell Death Differ 16:12–20PubMedCrossRef Scarlatti F, Granata R, Meijer AJ et al (2009) Does autophagy have a license to kill mammalian cells? Cell Death Differ 16:12–20PubMedCrossRef
96.
Zurück zum Zitat Mitroulis I, Kourtzelis I, Kambas K et al (2010) Regulation of the autophagic machinery in human neutrophils. Eur J Immunol 40:1461–1472PubMedCrossRef Mitroulis I, Kourtzelis I, Kambas K et al (2010) Regulation of the autophagic machinery in human neutrophils. Eur J Immunol 40:1461–1472PubMedCrossRef
97.
98.
Zurück zum Zitat Labbé K, Saleh M (2008) Cell death in the host response to infection. Cell Death Differ 15:1339–1349PubMedCrossRef Labbé K, Saleh M (2008) Cell death in the host response to infection. Cell Death Differ 15:1339–1349PubMedCrossRef
99.
Zurück zum Zitat Tudan C, Fong D, Duronio V et al (2000) The inhibition of spontaneous and tumor necrosis factor-alpha induced neutrophil apoptosis by crystals of calcium pyrophosphate dihydrate and monosodium urate monohydrate. J Rheumatol 27:2463–2472PubMed Tudan C, Fong D, Duronio V et al (2000) The inhibition of spontaneous and tumor necrosis factor-alpha induced neutrophil apoptosis by crystals of calcium pyrophosphate dihydrate and monosodium urate monohydrate. J Rheumatol 27:2463–2472PubMed
100.
Zurück zum Zitat Akahoshi T, Nagaoka T, Namai R et al (1997) Prevention of neutrophil apoptosis by monosodium urate crystals. Rheumatol Int 16:231–235PubMedCrossRef Akahoshi T, Nagaoka T, Namai R et al (1997) Prevention of neutrophil apoptosis by monosodium urate crystals. Rheumatol Int 16:231–235PubMedCrossRef
101.
Zurück zum Zitat Rose DM, Sydlaske AD, Agha-Babakhani A et al (2006) Transglutaminase limits murine peritoneal acute gout-like inflammation by regulating macrophage clearance of apoptotic neutrophils. Arthritis Rheum 54:3363–3371PubMedCrossRef Rose DM, Sydlaske AD, Agha-Babakhani A et al (2006) Transglutaminase limits murine peritoneal acute gout-like inflammation by regulating macrophage clearance of apoptotic neutrophils. Arthritis Rheum 54:3363–3371PubMedCrossRef
102.
Zurück zum Zitat Fadok VA, Bratton DL, Konowal A et al (1998) Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest 101:890–898PubMedCrossRef Fadok VA, Bratton DL, Konowal A et al (1998) Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J Clin Invest 101:890–898PubMedCrossRef
103.
Zurück zum Zitat Steiger S, Harper JL (2012) Neutrophil cannibalism triggers TGF-β1 production and self-regulation of neutrophil inflammatory function in MSU crystal-induced inflammation. Arthritis Rheum. doi:10.1002/art.37822 Steiger S, Harper JL (2012) Neutrophil cannibalism triggers TGF-β1 production and self-regulation of neutrophil inflammatory function in MSU crystal-induced inflammation. Arthritis Rheum. doi:10.​1002/​art.​37822
104.
Zurück zum Zitat Shirahama T, Cohen AS (1974) Ultrastructural evidence for leakage of lysosomal contents after phagocytosis of monosodium urate crystals. A mechanism of gouty inflammation. Am J Pathol 76:501–520PubMed Shirahama T, Cohen AS (1974) Ultrastructural evidence for leakage of lysosomal contents after phagocytosis of monosodium urate crystals. A mechanism of gouty inflammation. Am J Pathol 76:501–520PubMed
105.
Zurück zum Zitat Remijsen Q, Vanden Berghe T, Wirawan E et al (2011) Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res 21:290–304PubMedCrossRef Remijsen Q, Vanden Berghe T, Wirawan E et al (2011) Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res 21:290–304PubMedCrossRef
106.
Zurück zum Zitat Kambas K, Mitroulis I, Apostolidou E et al (2012) Autophagy mediates the delivery of thrombogenic tissue factor to neutrophil extracellular traps in human sepsis. PLoS One 7:e45427PubMedCrossRef Kambas K, Mitroulis I, Apostolidou E et al (2012) Autophagy mediates the delivery of thrombogenic tissue factor to neutrophil extracellular traps in human sepsis. PLoS One 7:e45427PubMedCrossRef
107.
Zurück zum Zitat Guma M, Ronacher L, Liu-Bryan R et al (2009) Caspase 1-independent activation of interleukin-1beta in neutrophil-predominant inflammation. Arthritis Rheum 60:3642–3650PubMedCrossRef Guma M, Ronacher L, Liu-Bryan R et al (2009) Caspase 1-independent activation of interleukin-1beta in neutrophil-predominant inflammation. Arthritis Rheum 60:3642–3650PubMedCrossRef
108.
Zurück zum Zitat Joosten LA, Netea MG, Fantuzzi G et al (2009) Inflammatory arthritis in caspase 1 gene-deficient mice: contribution of proteinase 3 to caspase 1-independent production of bioactive interleukin-1beta. Arthritis Rheum 60:3651–3662PubMedCrossRef Joosten LA, Netea MG, Fantuzzi G et al (2009) Inflammatory arthritis in caspase 1 gene-deficient mice: contribution of proteinase 3 to caspase 1-independent production of bioactive interleukin-1beta. Arthritis Rheum 60:3651–3662PubMedCrossRef
109.
Zurück zum Zitat Coeshott C, Ohnemus C, Pilyavskaya A et al (1999) Converting enzyme-independent release of tumor necrosis factor α and IL-1β from a stimulated human monocytic cell line in the presence of activated neutrophils or purified proteinase 3. Proc Natl Acad Sci U S A 96:6261–6266PubMedCrossRef Coeshott C, Ohnemus C, Pilyavskaya A et al (1999) Converting enzyme-independent release of tumor necrosis factor α and IL-1β from a stimulated human monocytic cell line in the presence of activated neutrophils or purified proteinase 3. Proc Natl Acad Sci U S A 96:6261–6266PubMedCrossRef
110.
Zurück zum Zitat Mankan AK, Dau T, Jenne D et al (2012) The NLRP3/ASC/caspase-1 axis regulates IL-1β processing in neutrophils. Eur J Immunol 42:710–715PubMedCrossRef Mankan AK, Dau T, Jenne D et al (2012) The NLRP3/ASC/caspase-1 axis regulates IL-1β processing in neutrophils. Eur J Immunol 42:710–715PubMedCrossRef
111.
Zurück zum Zitat Watson RWG, Rotstein OD, Parodo J et al (1998) The IL-1b-converting enzyme (caspase-1) inhibits apoptosis of inflammatory neutrophils through activation of IL-1b. J Immunol 161:957–962PubMed Watson RWG, Rotstein OD, Parodo J et al (1998) The IL-1b-converting enzyme (caspase-1) inhibits apoptosis of inflammatory neutrophils through activation of IL-1b. J Immunol 161:957–962PubMed
112.
Zurück zum Zitat Schorn C, Janko C, Latzko M et al (2012) Monosodium urate crystals induce extracellular DNA traps in neutrophils, eosinophils, and basophils but not in mononuclear cells. Front Immun 3:277 Schorn C, Janko C, Latzko M et al (2012) Monosodium urate crystals induce extracellular DNA traps in neutrophils, eosinophils, and basophils but not in mononuclear cells. Front Immun 3:277
113.
Zurück zum Zitat Schorn C, Janko C, Krenn V et al (2012) Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front Immunol 3:376PubMed Schorn C, Janko C, Krenn V et al (2012) Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front Immunol 3:376PubMed
114.
Zurück zum Zitat Clark SR, Ma AC, Tavener SA et al (2007) Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 13:463–469PubMedCrossRef Clark SR, Ma AC, Tavener SA et al (2007) Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 13:463–469PubMedCrossRef
115.
Zurück zum Zitat Tillack K, Breiden P, Martin R et al (2012) T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. J Immunol 188:3150–3159PubMedCrossRef Tillack K, Breiden P, Martin R et al (2012) T lymphocyte priming by neutrophil extracellular traps links innate and adaptive immune responses. J Immunol 188:3150–3159PubMedCrossRef
116.
Zurück zum Zitat Sangaletti S, Tripodo C, Chiodoni C et al (2012) Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells towards ANCA induction and associated autoimmunity. Blood 120:3007–3018PubMedCrossRef Sangaletti S, Tripodo C, Chiodoni C et al (2012) Neutrophil extracellular traps mediate transfer of cytoplasmic neutrophil antigens to myeloid dendritic cells towards ANCA induction and associated autoimmunity. Blood 120:3007–3018PubMedCrossRef
117.
Zurück zum Zitat Saffarzadeh M, Juenemann C, Queisser MA et al (2012) Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One 7:e32366PubMedCrossRef Saffarzadeh M, Juenemann C, Queisser MA et al (2012) Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One 7:e32366PubMedCrossRef
Metadaten
Titel
Neutrophils, IL-1β, and gout: is there a link?
verfasst von
Ioannis Mitroulis
Konstantinos Kambas
Konstantinos Ritis
Publikationsdatum
01.07.2013
Verlag
Springer-Verlag
Erschienen in
Seminars in Immunopathology / Ausgabe 4/2013
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-013-0361-0

Weitere Artikel der Ausgabe 4/2013

Seminars in Immunopathology 4/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.