The Role of NLRP3 Inflammasome in Pericarditis: Potential for Therapeutic Approaches

Adolfo G Mauro, Aldo Bonaventura, Alessandra Vecchié, Eleonora Mezzaroma, Salvatore Carbone, Pratyush Narayan, Nicola Potere, Antonio Cannatà, John F Paolini, Rossana Bussani, Fabrizio Montecucco, Gianfranco Sinagra, Benjamin W Van Tassel, Antonio Abbate, Stefano Toldo, Adolfo G Mauro, Aldo Bonaventura, Alessandra Vecchié, Eleonora Mezzaroma, Salvatore Carbone, Pratyush Narayan, Nicola Potere, Antonio Cannatà, John F Paolini, Rossana Bussani, Fabrizio Montecucco, Gianfranco Sinagra, Benjamin W Van Tassel, Antonio Abbate, Stefano Toldo

Abstract

Human samples of patients with chronic pericarditis and appropriate control subjects were stained for the inflammasome components. A mouse model of pericarditis was developed through the intrapericardial injection of zymosan A. Different inflammasome blockers were tested in the mouse model. Patients with pericarditis presented an intensification of the inflammasome activation compared with control subjects. The experimental model showed the pathological features of pericarditis. Among inflammasome blockers, NLRP3 inflammasome inhibitor, anakinra, and interleukin-1 trap were found to significantly improve pericardial alterations. Colchicine partially improved the pericardial inflammation. An intense activation of the inflammasome in pericarditis was demonstrated both in humans and in mice.

Keywords: IL, interleukin; IL-1 trap; IL-1α; IL-1β; NLRP3 inflammasome; NLRP3inh, NLRP3 inflammasome inhibitor; anakinra; colchicine; pericarditis.

Conflict of interest statement

This investigator-initiated study was supported by funding from Kiniksa Pharmaceuticals Ltd. Dr. Abbate has served as a consultant for AstraZeneca, Janssen, Kiniksa Pharmaceuticals, Merck, Novartis, Olatec, and Serpin Pharma. Dr. Toldo has received research support from Kiniksa, Serpin Pharma, and Olatec. Drs. Bonaventura and Vecchié have received travel grant support from Kiniksa Pharmaceuticals to attend the 2019 American Heart Association Scientific Sessions (Philadelphia, Pennsylvania). All other authors have reported that they have no relationships relevant to the contents of this paper to disclose.

© 2021 Published by Elsevier on behalf of the American College of Cardiology Foundation.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Schematic Representation of the Experimental Mouse Model of Acute Pericarditis Under general anesthesia, 1 mg of zymosan A (in 50 μl of sterile NaCl 0.9%) was injected in the pericardial sac of the mouse on day 0. Sham control procedures were performed injecting an equal volume of sterile NaCl 0.9% in the pericardial sac. Selected drugs were administered after the surgery was completed until day 7. On day 7, mice underwent echocardiography and were euthanized. IL = interleukin; NLRP3 = NACHT, leucine-rich repeat, and pyrin domain-containing protein 3; NSAID = nonsteroidal anti-inflammatory drug.
Figure 2
Figure 2
Upregulation of NLRP3 Inflammasome Components in Human Pericardial Specimens of Patients Affected by Pericarditis (A) Immunohistochemistry reveals upregulation of the NLRP3 protein following pericarditis in human samples (original magnification 40×). (B) Caspase-1 upregulation detected by immunohistochemical staining (original magnification 40×). C) Immunofluorescence staining of ASC (apoptosis-associated speck-like protein containing a COOH-terminus caspase activation and recruitment domain) (red) showing aggregates in the pericardium. Counterstaining of the nuclei achieved with DAPI (4′,6-diamidino-2-phenylindole) (blue) (original magnification 40×). Abbreviations as in Figure 1.
Figure 3
Figure 3
Assessment of Pericarditis in Mice (A) Pericardial effusion was measured at 7 days by transthoracic echocardiography as echo-free space between the 2 layers of the pericardium. Instillation of zymosan A produces a significant increase in pericardial fluid compared with sham control subjects injected with saline. (B) Pericardial thickening was measured at day 7 by histological assessment of hematoxylin and eosin–stained heart slides. Zymosan A increased significantly the thickness of the visceral pericardial layer compared with sham (original magnification 40×). (C) Immunofluorescence staining for ASC (red) and cardiac α-actin (green) in the pericardium of mice with experimental pericarditis. DAPI was used to counterstain the nuclei (blue) (original magnification 40×). Abbreviations as in Figure 2.
Figure 4
Figure 4
Assessment of IL-1α and IL-1β in Mice (A) IL-1α and IL-1β messenger RNA (mRNA) relative expression in the parietal pericardium was measured at 3 days by real-time polymerase chain reaction. Mice treated with intrapericardial instillation of zymosan A showed a significant increase in mRNA expression of both IL-1α and IL-1β compared with sham control subjects injected with saline. (B) Pericardial expression of IL-1α was measured at day 3 by immunofluorescence staining. Zymosan A–treated mice had a higher expression of IL-1α compared with sham. IL-1α positive cells are stained in red, while DAPI was used to counterstain the nuclei (blue). The myocardium appears green by natural autofluorescence. Original magnification 40×. (C) Immunofluorescence staining for IL-1β (red) in the pericardium of mice with experimental pericarditis demonstrated a higher expression of this cytokine in zymosan A–treated mice compared with sham control subjects. IL-1β is stained in red, while DAPI was used to counterstain the nuclei (blue). The myocardium appears green by natural autofluorescence. Original magnification 40×. Abbreviations as in Figures 1 and 2.
Figure 5
Figure 5
Effects of Ibuprofen Treatment in the Experimental Model of Pericarditis in Mice (A) Pericardial effusion was measured at 7 days following daily treatment with ibuprofen (100 mg/kg). Daily injection with ibuprofen significantly reduced fluid accumulation. (B) Treatment with ibuprofen did not reduce pericardial thickness. (C) ASC (apoptosis-associated speck-like protein containing a COOH-terminus caspase activation and recruitment domain) aggregates were not reduced by treatment with ibuprofen. ∗p < 0.05, ∗∗∗p < 0.001 vs. sham-treated mice; ###p < 0.001 vs. vehicle-treated mice.
Figure 6
Figure 6
Inhibition of the NLRP3 Inflammasome in Mice With Acute Pericarditis (A) Pericardial effusion measured at 7 days following daily treatment with colchicine (100 μg/kg) and NLRP3 inhibitor 16673-34-0 (100 mg/kg). Colchicine and 16673-34-0 significantly reduced pericardial effusion in mice with pericarditis. (B) Treatment with the NLRP3 inhibitor 16673-34-0, but not colchicine, showed a significant reduction of the pericardial thickness following acute pericarditis in mice compared with the vehicle-treated group. (C) ASC aggregates in the pericardium of mice with acute pericarditis were significantly reduced by colchicine and 16673-34-0. ∗p < 0.05, ∗∗∗p < 0.001 vs. sham-treated mice; ##p < 0.01, ###p < 0.001 vs. vehicle-treated mice. Abbreviations as in Figures 1 and 2.
Figure 7
Figure 7
IL-1 Blockade in Experimental Acute Pericarditis Pericardial effusion, thickness, and formation of ASC aggregates were measured 7 days after twice daily treatment with anakinra (100 mg/kg) or IL-1 trap. Anakinra significantly reduced (A) pericardial fluid accumulation, (B) thickening of the pericardium, and (C) ASC aggregation following acute pericarditis in mice. A dose response of IL-1 trap was tested in mice subjected to experimental pericarditis. Treatment with IL-1 trap every 48 h significantly reduced (D) pericardial effusion, (E) pericardial thickness, and (F) inflammasome formation with each dose tested. ∗p < 0.05, ∗∗∗p < 0.001 vs. sham-treated mice; #p < 0.05, ##p < 0.01, ###p < 0.001 vs. vehicle-treated mice. Abbreviations as in Figures 1 and 2.

References

    1. Adler Y., Charron P., Imazio M. 2015 ESC Guidelines for the diagnosis and management of pericardial diseases: the Task Force for the Diagnosis and Management of Pericardial Diseases of the European Society of Cardiology (ESC)Endorsed by: The European Association for Cardio-Thoracic Surgery (EACTS) Eur Heart J. 2015;36:2921–2964.
    1. Imazio M., Gaita F., LeWinter M. Evaluation and treatment of pericarditis: a systematic review. JAMA. 2015;314:1498–1506.
    1. Bonaventura A., Montecucco F. Inflammation and pericarditis: are neutrophils actors behind the scenes? J Cell Physiol. 2019;234:5390–5398.
    1. Buckley L.F., Viscusi M.M., Van Tassell B.W., Abbate A. Interleukin-1 blockade for the treatment of pericarditis. Eur Heart J Cardiovasc Pharmacother. 2018;4:46–53.
    1. Toldo S., Abbate A. The NLRP3 inflammasome in acute myocardial infarction. Nat Rev Cardiol. 2018;15:203–214.
    1. Mauro A.G., Bonaventura A., Mezzaroma E., Quader M., Toldo S. NLRP3 Inflammasome in Acute Myocardial Infarction. J Cardiovasc Pharmacol. 2019;74:175–187.
    1. Dinarello C.A. Interleukin-1. Cytokine Growth Factor Rev. 1997;8:253–265.
    1. Bertheloot D., Latz E. HMGB1, IL-1alpha, IL-33 and S100 proteins: dual-function alarmins. Cell Mol Immunol. 2017;14:43–64.
    1. Chiabrando J.G., Bonaventura A., Vecchie A. Management of acute and recurrent pericarditis: JACC State-of-the-Art Review. J Am Coll Cardiol. 2020;75:76–92.
    1. Bayes-Genis A., Adler Y., de Luna A.B., Imazio M. Colchicine in pericarditis. Eur Heart J. 2017;38:1706–1709.
    1. Brucato A., Imazio M., Gattorno M. Effect of anakinra on recurrent pericarditis among patients with colchicine resistance and corticosteroid dependence: the AIRTRIP randomized clinical trial. JAMA. 2016;316:1906–1912.
    1. Toldo S., Das A., Mezzaroma E. Induction of microRNA-21 with exogenous hydrogen sulfide attenuates myocardial ischemic and inflammatory injury in mice. Circ Cardiovasc Genet. 2014;7:311–320.
    1. Kumar H., Kumagai Y., Tsuchida T. Involvement of the NLRP3 inflammasome in innate and humoral adaptive immune responses to fungal beta-glucan. J Immunol. 2009;183:8061–8067.
    1. Monroe L.L., Armstrong M.G., Zhang X. Zymosan-induced peritonitis: effects on cardiac function, temperature regulation, translocation of bacteria, and role of dectin-1. Shock. 2016;46:723–730.
    1. Marchetti C., Swartzwelter B., Koenders M.I. NLRP3 inflammasome inhibitor OLT1177 suppresses joint inflammation in murine models of acute arthritis. Arthritis Res Ther. 2018;20:169.
    1. Marchetti C., Chojnacki J., Toldo S. A novel pharmacologic inhibitor of the NLRP3 inflammasome limits myocardial injury after ischemia-reperfusion in the mouse. J Cardiovasc Pharmacol. 2014;63:316–322.
    1. Marchetti C., Toldo S., Chojnacki J. Pharmacologic inhibition of the NLRP3 inflammasome preserves cardiac function after ischemic and nonischemic injury in the mouse. J Cardiovasc Pharmacol. 2015;66:1–8.
    1. Toldo S., Marchetti C., Mauro A.G. Inhibition of the NLRP3 inflammasome limits the inflammatory injury following myocardial ischemia-reperfusion in the mouse. Int J Cardiol. 2016;209:215–220.
    1. Van Tassell B.W., Varma A., Salloum F.N. Interleukin-1 trap attenuates cardiac remodeling after experimental acute myocardial infarction in mice. J Cardiovasc Pharmacol. 2010;55:117–122.
    1. Smilde B.J., Woudstra L., Fong Hing G. Colchicine aggravates coxsackievirus B3 infection in mice. Int J Cardiol. 2016;216:58–65.
    1. Wolach B., Gotfried M., Jedeikin A., Lishner M., Brossi A., Ravid M. Colchicine analogues: effect on amyloidogenesis in a murine model and, in vitro, on polymorphonuclear leukocytes. Eur J Clin Invest. 1992;22:630–634.
    1. Toldo S., Mezzaroma E., O'Brien L. Interleukin-18 mediates interleukin-1-induced cardiac dysfunction. Am J Physiol Heart Circ Physiol. 2014;306:H1025–H1031.
    1. Masumoto J., Taniguchi S., Ayukawa K. ASC, a novel 22-kDa protein, aggregates during apoptosis of human promyelocytic leukemia HL-60 cells. J Biol Chem. 1999;274:33835–33838.
    1. Toldo S., Mezzaroma E., Mauro A.G., Salloum F., Van Tassell B.W., Abbate A. The inflammasome in myocardial injury and cardiac remodeling. Antioxid Redox Signal. 2015;22:1146–1161.
    1. Mezzaroma E., Toldo S., Farkas D. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc Natl Acad Sci U S A. 2011;108:19725–19730.
    1. Kron J., Mauro A.G., Bonaventura A. Inflammasome formation in granulomas in cardiac sarcoidosis. Circ Arrhythm Electrophysiol. 2019;12
    1. Abbate A., Santini D., Biondi-Zoccai G.G. Cyclo-oxygenase-2 (COX-2) expression at the site of recent myocardial infarction: friend or foe? Heart. 2004;90:440–443.
    1. Signa S., D'Alessandro M., Consolini R. Failure of anti Interleukin-1 beta monoclonal antibody in the treatment of recurrent pericarditis in two children. Pediatr Rheumatol Online J. 2020;18:51.
    1. Sato M., Sano H., Iwaki D. Direct binding of Toll-like receptor 2 to zymosan, and zymosan-induced NF-kappa B activation and TNF-alpha secretion are down-regulated by lung collectin surfactant protein A. J Immunol. 2003;171:417–425.
    1. Pagé P.L., Plumb V.J., Okumura K., Waldo A.L. A new animal model of atrial flutter. J Am Coll Cardiol. 1986;8:872–879.
    1. Leak L.V., Ferrans V.J., Cohen S.R., Eidbo E.E., Jones M. Animal model of acute pericarditis and its progression to pericardial fibrosis and adhesions: ultrastructural studies. Am J Anat. 1987;180:373–390.
    1. Franchi L., Nunez G. The Nlrp3 inflammasome is critical for aluminium hydroxide-mediated IL-1beta secretion but dispensable for adjuvant activity. Eur J Immunol. 2008;38:2085–2089.
    1. Kojima A., Sakaue T., Okazaki M. A simple mouse model of pericardial adhesions. J Cardiothorac Surg. 2019;14:124.
    1. Imazio M., Spodick D.H., Brucato A., Trinchero R., Adler Y. Controversial issues in the management of pericardial diseases. Circulation. 2010;121:916–928.
    1. Vecchie A., Chiabrando J.G., Dell M.S. Clinical presentation and outcomes of acute pericarditis in a large urban hospital in the United States of America. Chest. 2020;158:2556–2557.
    1. Wohlford G.F., Buckley L.F., Vecchie A. Acute effects of interleukin-1 blockade using anakinra in patients with acute pericarditis. J Cardiovasc Pharmacol. 2020;76:50–52.
    1. Abbate A., Toldo S., Marchetti C., Kron J., Van Tassell B.W., Dinarello C.A. Interleukin-1 and the inflammasome as therapeutic targets in cardiovascular disease. Circ Res. 2020;126:1260–1280.
    1. Abbate A., Dinarello C.A. Anti-inflammatory therapies in acute coronary syndromes: is IL-1 blockade a solution? Eur Heart J. 2015;36:337–339.
    1. Buckley L.F., Abbate A. Interleukin-1 blockade in cardiovascular diseases: a clinical update. Eur Heart J. 2018;39:2063–2069.
    1. Dinarello C.A. The IL-1 family of cytokines and receptors in rheumatic diseases. Nat Rev Rheumatol. 2019;15:610–632.
    1. Klein A.L., Imazio M., Cremer P. RHAPSODY Investigators. Phase 3 Trial of Interleukin-1 Trap Rilonacept in recurrent pericarditis. N Engl J Med. 2021;384:31–41.
    1. Paschke S., Weidner A.F., Paust T., Marti O., Beil M., Ben-Chetrit E. Technical advance: Inhibition of neutrophil chemotaxis by colchicine is modulated through viscoelastic properties of subcellular compartments. J Leukoc Biol. 2013;94:1091–1096.
    1. Martinon F., Petrilli V., Mayor A., Tardivel A., Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241.
    1. Marques-da-Silva C., Chaves M.M., Castro N.G., Coutinho-Silva R., Guimaraes M.Z. Colchicine inhibits cationic dye uptake induced by ATP in P2X2 and P2X7 receptor-expressing cells: implications for its therapeutic action. Br J Pharmacol. 2011;163:912–926.
    1. Camacho-Lovillo M., Mendez-Santos A. Successful treatment of idiopathic recurrent pericarditis with interleukin-1 receptor antagonist (anakinra) Pediatr Cardiol. 2013;34:1293–1294.
    1. D'Elia E., Brucato A., Pedrotti P. Successful treatment of subacute constrictive pericarditis with interleukin-1beta receptor antagonist (anakinra) Clin Exp Rheumatol. 2015;33:294–295.
    1. Lazaros G., Vasileiou P., Danias P. Effusive-constrictive pericarditis successfully treated with anakinra. Clin Exp Rheumatol. 2015;33:945.
    1. Picco P., Brisca G., Traverso F., Loy A., Gattorno M., Martini A. Successful treatment of idiopathic recurrent pericarditis in children with interleukin-1beta receptor antagonist (anakinra): an unrecognized autoinflammatory disease? Arthritis Rheum. 2009;60:264–268.
    1. Scardapane A., Brucato A., Chiarelli F., Breda L. Efficacy of an interleukin-1beta receptor antagonist (anakinra) in idiopathic recurrent pericarditis. Pediatr Cardiol. 2013;34:1989–1991.
    1. Schatz A., Trankle C., Yassen A. Resolution of pericardial constriction with Anakinra in a patient with effusive-constrictive pericarditis secondary to rheumatoid arthritis. Int J Cardiol. 2016;223:215–216.
    1. Vassilopoulos D., Lazaros G., Tsioufis C., Vasileiou P., Stefanadis C., Pectasides D. Successful treatment of adult patients with idiopathic recurrent pericarditis with an interleukin-1 receptor antagonist (anakinra) Int J Cardiol. 2012;160:66–68.

Source: PubMed

3
S'abonner