Skip to main content
Top
Published in: Inflammation Research 3/2017

01-03-2017 | Original Research Paper

Macrophage-derived IL-1β enhances monosodium urate crystal-triggered NET formation

Authors: Payel Sil, Haley Wicklum, Chandler Surell, Balázs Rada

Published in: Inflammation Research | Issue 3/2017

Login to get access

Abstract

Objective and design

Arthritic gout is caused by joint inflammation triggered by the damaging effects of monosodium uric acid (MSU) crystal accumulation in the synovial space. Neutrophils play a major role in mediating joint inflammation in gout. Along with neutrophils, other immune cells, such as macrophages, are present in inflamed joints and contribute to gout pathogenesis. Neutrophils form neutrophil extracellular traps (NETs) in response to MSU crystals. In the presence of MSU crystals, macrophages release IL-1β, a cytokine crucial to initiate gout pathogenesis and neutrophil recruitment. Our research investigated interactions between human macrophages and neutrophils in an in vitro model system and asked how macrophages affect NET formation stimulated by MSU crystals.

Materials or subjects

Human neutrophils and PBMCs were isolated from peripheral blood of healthy volunteers. PBMCs were differentiated into macrophages in vitro using human M-CSF.

Treatment

Human neutrophils were pretreated with macrophage-conditioned media, neutrophil-conditioned media, recombinant human IL-1β or anakinra prior to stimulation by MSU crystals.

Method

Interaction of neutrophils with MSU crystals was evaluated by live imaging using confocal microscopy. The presence of myeloperoxidase (MPO) and neutrophil elastase (NE) was measured by ELISA. NET formation was quantitated by Sytox Orange-based extracellular DNA release assay and NE-DNA ELISA. AggNET formation was assessed by macroscopic evaluation.

Results

We found that crystal- and cell-free supernatants of macrophages stimulated with MSU crystals promote MSU crystal-stimulated NET formation in human neutrophils. This observation was confirmed by additional assays measuring the release of MPO, NE, and the enzymatic activity of NE. MSU crystal-induced NET formation remained unchanged when neutrophil supernatants were tested. IL-1β is a crucial cytokine orchestrating the onset of inflammation in gout and is known to be released in large amounts from macrophages following MSU crystal stimulation. We found that recombinant IL-1β strongly promoted MSU crystal-induced NET formation in human neutrophils. Interestingly, IL-1β alone did not induce any NET release. We also found that clinical grade anakinra, an IL-1 receptor blocker, strongly reduced the NETosis-enhancing effect of macrophage supernatants indicating that IL-1β is mainly responsible for this effect.

Conclusions

Macrophage-derived IL-1β enhances MSU crystal-induced NET release in neutrophils. We identified a new mechanism by which macrophages and IL-1β affect neutrophil functions, and could contribute to the inflammatory conditions present in gout. Our results also revealed a new anti-inflammatory mechanism of anakinra.
Literature
1.
go back to reference Busso N, So A. Microcrystals as DAMPs and their role in joint inflammation. Rheumatology (Oxford). 2012;51:1154–60.CrossRef Busso N, So A. Microcrystals as DAMPs and their role in joint inflammation. Rheumatology (Oxford). 2012;51:1154–60.CrossRef
3.
go back to reference Mitroulis I, Kambas K, Ritis K. Neutrophils, IL-1beta, and gout: is there a link? Semin Immunopathol. 2013;35:501–12.CrossRefPubMed Mitroulis I, Kambas K, Ritis K. Neutrophils, IL-1beta, and gout: is there a link? Semin Immunopathol. 2013;35:501–12.CrossRefPubMed
4.
go back to reference Pope RM, Tschopp J. The role of interleukin-1 and the inflammasome in gout: implications for therapy. Arthritis Rheum. 2007;56:3183–8.CrossRefPubMed Pope RM, Tschopp J. The role of interleukin-1 and the inflammasome in gout: implications for therapy. Arthritis Rheum. 2007;56:3183–8.CrossRefPubMed
5.
go back to reference Schauer C, Janko C, Munoz LE, Zhao Y, Kienhofer D, Frey B, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20:511–7.CrossRefPubMed Schauer C, Janko C, Munoz LE, Zhao Y, Kienhofer D, Frey B, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20:511–7.CrossRefPubMed
6.
go back to reference Schorn C, Janko C, Krenn V, Zhao Y, Munoz LE, Schett G, et al. Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front Immunol. 2012;3:376.PubMedPubMedCentral Schorn C, Janko C, Krenn V, Zhao Y, Munoz LE, Schett G, et al. Bonding the foe—NETting neutrophils immobilize the pro-inflammatory monosodium urate crystals. Front Immunol. 2012;3:376.PubMedPubMedCentral
7.
go back to reference Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–41.CrossRefPubMed Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–41.CrossRefPubMed
8.
go back to reference Oliveira SH, Canetti C, Ribeiro RA, Cunha FQ. Neutrophil migration induced by IL-1beta depends upon LTB4 released by macrophages and upon TNF-alpha and IL-1beta released by mast cells. Inflammation. 2008;31:36–46.CrossRefPubMed Oliveira SH, Canetti C, Ribeiro RA, Cunha FQ. Neutrophil migration induced by IL-1beta depends upon LTB4 released by macrophages and upon TNF-alpha and IL-1beta released by mast cells. Inflammation. 2008;31:36–46.CrossRefPubMed
9.
go back to reference Schett G, Dayer JM, Manger B. Interleukin-1 function and role in rheumatic disease. Nat Rev Rheumatol. 2016;12:14–24.CrossRefPubMed Schett G, Dayer JM, Manger B. Interleukin-1 function and role in rheumatic disease. Nat Rev Rheumatol. 2016;12:14–24.CrossRefPubMed
11.
go back to reference Schlesinger N. Treatment of chronic gouty arthritis: it is not just about urate-lowering therapy. Semin Arthritis Rheum. 2012;42:155–65.CrossRefPubMed Schlesinger N. Treatment of chronic gouty arthritis: it is not just about urate-lowering therapy. Semin Arthritis Rheum. 2012;42:155–65.CrossRefPubMed
12.
go back to reference 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;64:474–84.CrossRefPubMed 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;64:474–84.CrossRefPubMed
13.
go back to reference Rada B, Park JJ, Sil P, Geiszt M, Leto TL. NLRP3 inflammasome activation and interleukin-1beta release in macrophages require calcium but are independent of calcium-activated NADPH oxidases. Inflamm Res. 2014;63:821–30.CrossRefPubMedPubMedCentral Rada B, Park JJ, Sil P, Geiszt M, Leto TL. NLRP3 inflammasome activation and interleukin-1beta release in macrophages require calcium but are independent of calcium-activated NADPH oxidases. Inflamm Res. 2014;63:821–30.CrossRefPubMedPubMedCentral
14.
go back to reference Meissner F, Seger RA, Moshous D, Fischer A, Reichenbach J, Zychlinsky A. Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease. Blood. 2010;116:1570–3.CrossRefPubMedPubMedCentral Meissner F, Seger RA, Moshous D, Fischer A, Reichenbach J, Zychlinsky A. Inflammasome activation in NADPH oxidase defective mononuclear phagocytes from patients with chronic granulomatous disease. Blood. 2010;116:1570–3.CrossRefPubMedPubMedCentral
15.
go back to reference van Bruggen R, Koker MY, Jansen M, van Houdt M, Roos D, Kuijpers TW, et al. Human NLRP3 inflammasome activation is No 1–4 independent. Blood. 2010;115:5398–400.CrossRefPubMed van Bruggen R, Koker MY, Jansen M, van Houdt M, Roos D, Kuijpers TW, et al. Human NLRP3 inflammasome activation is No 1–4 independent. Blood. 2010;115:5398–400.CrossRefPubMed
18.
go back to reference Mitroulis I, Kambas K, Chrysanthopoulou A, Skendros P, Apostolidou E, Kourtzelis I, et al. Neutrophil extracellular trap formation is associated with IL-1beta and autophagy-related signaling in gout. PLoS One. 2011;6:e29318.CrossRefPubMedPubMedCentral Mitroulis I, Kambas K, Chrysanthopoulou A, Skendros P, Apostolidou E, Kourtzelis I, et al. Neutrophil extracellular trap formation is associated with IL-1beta and autophagy-related signaling in gout. PLoS One. 2011;6:e29318.CrossRefPubMedPubMedCentral
19.
go back to reference Naccache PH, Grimard M, Roberge CJ, Gilbert C, Lussier A, de Medicis R, et al. Crystal-induced neutrophil activation. I. Initiation and modulation of calcium mobilization and superoxide production by microcrystals. Arthritis Rheum. 1991;34:333–42.CrossRefPubMed Naccache PH, Grimard M, Roberge CJ, Gilbert C, Lussier A, de Medicis R, et al. Crystal-induced neutrophil activation. I. Initiation and modulation of calcium mobilization and superoxide production by microcrystals. Arthritis Rheum. 1991;34:333–42.CrossRefPubMed
20.
go back to reference Popa-Nita O, Naccache PH. Crystal-induced neutrophil activation. Immunol Cell Biol. 2010;88:32–40.CrossRefPubMed Popa-Nita O, Naccache PH. Crystal-induced neutrophil activation. Immunol Cell Biol. 2010;88:32–40.CrossRefPubMed
21.
go back to reference Pang L, Hayes CP, Buac K, Yoo DG, Rada B. Pseudogout-associated inflammatory calcium pyrophosphate dihydrate microcrystals induce formation of neutrophil extracellular traps. J Immunol. 2013;190:6488–500.CrossRefPubMed Pang L, Hayes CP, Buac K, Yoo DG, Rada B. Pseudogout-associated inflammatory calcium pyrophosphate dihydrate microcrystals induce formation of neutrophil extracellular traps. J Immunol. 2013;190:6488–500.CrossRefPubMed
22.
go back to reference Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5.CrossRefPubMed Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–5.CrossRefPubMed
23.
go back to reference Desai J, Kumar SV, Mulay SR, Konrad L, Romoli S, Schauer C, et al. PMA and crystal-induced neutrophil extracellular trap formation involves RIPK1-RIPK3-MLKL signaling. Eur J Immunol. 2016;46:223–9.CrossRefPubMed Desai J, Kumar SV, Mulay SR, Konrad L, Romoli S, Schauer C, et al. PMA and crystal-induced neutrophil extracellular trap formation involves RIPK1-RIPK3-MLKL signaling. Eur J Immunol. 2016;46:223–9.CrossRefPubMed
25.
go back to reference Yoo DG, Floyd M, Winn M, Moskowitz SM, Rada B. NET formation induced by Pseudomonas aeruginosa cystic fibrosis isolates measured as release of myeloperoxidase-DNA and neutrophil elastase-DNA complexes. Immunol Lett. 2014;160:186–94.CrossRefPubMed Yoo DG, Floyd M, Winn M, Moskowitz SM, Rada B. NET formation induced by Pseudomonas aeruginosa cystic fibrosis isolates measured as release of myeloperoxidase-DNA and neutrophil elastase-DNA complexes. Immunol Lett. 2014;160:186–94.CrossRefPubMed
26.
go back to reference Yoo DG, Winn M, Pang L, Moskowitz SM, Malech HL, Leto TL, et al. Release of cystic fibrosis airway inflammatory markers from Pseudomonas aeruginosa-stimulated human neutrophils involves NADPH oxidase-dependent extracellular DNA trap formation. Journal of immunology. 2014;192:4728–38.CrossRef Yoo DG, Winn M, Pang L, Moskowitz SM, Malech HL, Leto TL, et al. Release of cystic fibrosis airway inflammatory markers from Pseudomonas aeruginosa-stimulated human neutrophils involves NADPH oxidase-dependent extracellular DNA trap formation. Journal of immunology. 2014;192:4728–38.CrossRef
27.
go back to reference Sil P, Yoo DG, Floyd M, Gingerich A, Rada B. High throughput measurement of extracellular DNA release and quantitative NET formation in human neutrophils in vitro. J Vis Exp. 2016;(112). doi:10.3791/52779. Sil P, Yoo DG, Floyd M, Gingerich A, Rada B. High throughput measurement of extracellular DNA release and quantitative NET formation in human neutrophils in vitro. J Vis Exp. 2016;(112). doi:10.​3791/​52779.
28.
go back to reference Schorn C, Janko C, Latzko M, Chaurio R, Schett G, Herrmann M. Monosodium urate crystals induce extracellular DNA traps in neutrophils, eosinophils, and basophils but not in mononuclear cells. Front Immunol. 2012;3:277.PubMedPubMedCentral Schorn C, Janko C, Latzko M, Chaurio R, Schett G, Herrmann M. Monosodium urate crystals induce extracellular DNA traps in neutrophils, eosinophils, and basophils but not in mononuclear cells. Front Immunol. 2012;3:277.PubMedPubMedCentral
29.
go back to reference Busso N, Ea HK. The mechanisms of inflammation in gout and pseudogout (CPP-induced arthritis). Reumatismo. 2011;63:230–7. Busso N, Ea HK. The mechanisms of inflammation in gout and pseudogout (CPP-induced arthritis). Reumatismo. 2011;63:230–7.
30.
go back to reference Farrera C, Fadeel B. Macrophage clearance of neutrophil extracellular traps is a silent process. J Immunol. 2013;191:2647–56.CrossRefPubMed Farrera C, Fadeel B. Macrophage clearance of neutrophil extracellular traps is a silent process. J Immunol. 2013;191:2647–56.CrossRefPubMed
31.
go back to reference Nakazawa D, Shida H, Kusunoki Y, Miyoshi A, Nishio S, Tomaru U, et al. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun. 2016;67:19–28.CrossRefPubMed Nakazawa D, Shida H, Kusunoki Y, Miyoshi A, Nishio S, Tomaru U, et al. The responses of macrophages in interaction with neutrophils that undergo NETosis. J Autoimmun. 2016;67:19–28.CrossRefPubMed
32.
go back to reference Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316–20.CrossRefPubMedPubMedCentral Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science. 2015;349:316–20.CrossRefPubMedPubMedCentral
33.
go back to reference Seibenhener ML, Babu JR, Geetha T, Wong HC, Krishna NR, Wooten MW. Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol Cell Biol. 2004;24:8055–68.CrossRefPubMedPubMedCentral Seibenhener ML, Babu JR, Geetha T, Wong HC, Krishna NR, Wooten MW. Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol Cell Biol. 2004;24:8055–68.CrossRefPubMedPubMedCentral
34.
go back to reference Chenevier-Gobeaux C, Lemarechal H, Bonnefont-Rousselot D, Poiraudeau S, Ekindjian OG, Borderie D. Superoxide production and NADPH oxidase expression in human rheumatoid synovial cells: regulation by interleukin-1beta and tumour necrosis factor-alpha. Inflamm Res. 2006;55:483–90.CrossRefPubMed Chenevier-Gobeaux C, Lemarechal H, Bonnefont-Rousselot D, Poiraudeau S, Ekindjian OG, Borderie D. Superoxide production and NADPH oxidase expression in human rheumatoid synovial cells: regulation by interleukin-1beta and tumour necrosis factor-alpha. Inflamm Res. 2006;55:483–90.CrossRefPubMed
35.
go back to reference Yan B, Han P, Pan L, Lu W, Xiong J, Zhang M, et al. IL-1beta and reactive oxygen species differentially regulate neutrophil directional migration and Basal random motility in a zebrafish injury-induced inflammation model. J Immunol. 2014;192:5998–6008.CrossRefPubMed Yan B, Han P, Pan L, Lu W, Xiong J, Zhang M, et al. IL-1beta and reactive oxygen species differentially regulate neutrophil directional migration and Basal random motility in a zebrafish injury-induced inflammation model. J Immunol. 2014;192:5998–6008.CrossRefPubMed
36.
go back to reference Singh A, Zarember KA, Kuhns DB, Gallin JI. Impaired priming and activation of the neutrophil NADPH oxidase in patients with IRAK4 or NEMO deficiency. J Immunol. 2009;182:6410–7.CrossRefPubMedPubMedCentral Singh A, Zarember KA, Kuhns DB, Gallin JI. Impaired priming and activation of the neutrophil NADPH oxidase in patients with IRAK4 or NEMO deficiency. J Immunol. 2009;182:6410–7.CrossRefPubMedPubMedCentral
37.
go back to reference Perretti M, Appleton I, Parente L, Flower RJ. Pharmacology of interleukin-1-induced neutrophil migration. Agents Actions 1993;38 Spec No:C64–5. Perretti M, Appleton I, Parente L, Flower RJ. Pharmacology of interleukin-1-induced neutrophil migration. Agents Actions 1993;38 Spec No:C64–5.
38.
go back to reference Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol. 2007;5:577–82.CrossRefPubMed Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol. 2007;5:577–82.CrossRefPubMed
40.
go back to reference Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218.CrossRefPubMed Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218.CrossRefPubMed
41.
go back to reference Semerad CL, Liu F, Gregory AD, Stumpf K, Link DC. G-CSF is an essential regulator of neutrophil trafficking from the bone marrow to the blood. Immunity. 2002;17:413–23.CrossRefPubMed Semerad CL, Liu F, Gregory AD, Stumpf K, Link DC. G-CSF is an essential regulator of neutrophil trafficking from the bone marrow to the blood. Immunity. 2002;17:413–23.CrossRefPubMed
42.
go back to reference 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;68:1602–8.CrossRefPubMed 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;68:1602–8.CrossRefPubMed
Metadata
Title
Macrophage-derived IL-1β enhances monosodium urate crystal-triggered NET formation
Authors
Payel Sil
Haley Wicklum
Chandler Surell
Balázs Rada
Publication date
01-03-2017
Publisher
Springer International Publishing
Published in
Inflammation Research / Issue 3/2017
Print ISSN: 1023-3830
Electronic ISSN: 1420-908X
DOI
https://doi.org/10.1007/s00011-016-1008-0

Other articles of this Issue 3/2017

Inflammation Research 3/2017 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine