Thromb Haemost 2013; 110(03): 501-514
DOI: 10.1160/TH13-03-0211
Review Article
Schattauer GmbH

Pathophysiological role of neutrophils in acute myocardial infarction

Federico Carbone
1   Department of Internal Medicine, University of Genoa, Genoa, Italy
,
Alessio Nencioni
1   Department of Internal Medicine, University of Genoa, Genoa, Italy
,
François Mach
2   Division of Cardiology, Foundation for Medical Researches, Faculty of Medicine, Department of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
,
Nicolas Vuilleumier
3   Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Switzerland
4   Department of Human Protein Science, Geneva Faculty of Medicine, Geneva, Switzerland
,
Fabrizio Montecucco
1   Department of Internal Medicine, University of Genoa, Genoa, Italy
2   Division of Cardiology, Foundation for Medical Researches, Faculty of Medicine, Department of Internal Medicine, Geneva University Hospital, Geneva, Switzerland
› Author Affiliations
Financial Support: This research was funded by EU FP7, Grant number 201668, AtheroRemo to Dr. F. Mach. This work was also supported by the Swiss National Science Foundation Grants to Dr. F. Mach (#310030-118245), Dr N. Vuilleumier (#310030-140736), and to Dr. Montecucco (#32003B-134963/1) and by the Italian Ministry of Health (GR-2008-1135635 to Dr. A. Nencioni).
Further Information

Publication History

Received: 12 March 2013

Accepted after major revision: 04 May 2013

Publication Date:
22 November 2017 (online)

Summary

The pathogenesis of acute myocardial infarction is known to be mediated by systemic, intraplaque and myocardial inflammatory processes. Among different immune cell subsets, compelling evidence now indicates a pivotal role for neutrophils in acute coronary syndromes. Neutrophils infiltrate coronary plaques and the infarcted myocardium and mediate tissue damage by releasing matrix-degrading enzymes and reactive oxygen species. In addition, neutrophils are also involved in post-infarction adverse cardiac remodelling and neointima formation after angioplasty. The promising results obtained in preclinical models with pharmacological approaches interfering with neutrophil recruitment or function have confirmed the pathophysiological relevance of these immune cells in acute coronary syndromes and prompted further studies of these therapeutic interventions. This narrative review will provide an update on the role of neutrophils in acute myocardial infarction and on the pharmacological means that were devised to prevent neutrophil-mediated tissue damage and to reduce post-ischaemic outcomes.

 
  • References

  • 1 Steinberg D. Atherogenesis in perspective: hypercholesterolemia and inflammation as partners in crime. Nat Med 2002; 08: 1211-1217.
  • 2 Shah PK. Molecular mechanisms of plaque instability. Curr Opin Lipidol 2007; 18: 492-499.
  • 3 Hulten EA. et al. Prognostic value of cardiac computed tomography angiography: a systematic review and meta-analysis. J Am Coll Cardiol 2011; 57: 1237-1247.
  • 4 Swirski FK, Nahrendorf M. Leukocyte behavior in atherosclerosis, myocardial infarction, and heart failure. Science 2013; 339: 161-166.
  • 5 Soehnlein O. Multiple roles for neutrophils in atherosclerosis. Circ Res 2012; 110: 875-888.
  • 6 Hoffman M. et al. Leukocytes and coronary heart disease. Atherosclerosis 2004; 172: 1-6.
  • 7 Coller BS. Leukocytosis and ischaemic vascular disease morbidity and mortality: is it time to intervene?. Arterioscler Thromb Vasc Biol 2005; 25: 658-670.
  • 8 Guasti L. et al. Neutrophils and clinical outcomes in patients with acute coronary syndromes and/or cardiac revascularisation. A systematic review on more than 34,000 subjects. Thromb Haemost 2011; 106: 591-599.
  • 9 Dinerman JL. et al. Increased neutrophil elastase release in unstable angina pectoris and acute myocardial infarction. J Am Coll Cardiol 1990; 15: 1559-1563.
  • 10 Bell D. et al. Inflammatory response, neutrophil activation, and free radical production after acute myocardial infarction: effect of thrombolytic treatment. Br Heart J 1990; 63: 82-87.
  • 11 Mazzone A. et al. Increased expression of neutrophil and monocyte adhesion molecules in unstable coronary artery disease. Circulation 1993; 88: 358-363.
  • 12 Takeshita S. et al. Systemic inflammatory responses in acute coronary syndrome: increased activity observed in polymorphonuclear leukocytes but not T lymphocytes. Atherosclerosis 1997; 135: 187-192.
  • 13 Leckie MJ. et al. Automated quantitation of peripheral blood neutrophil activation in patients with myocardial ischaemia. Int J Cardiol 2004; 95: 307-313.
  • 14 Avanzas P. et al. Markers of inflammation and multiple complex stenoses (pancoronary plaque vulnerability) in patients with non-ST segment elevation acute coronary syndromes. Heart 2004; 90: 847-852.
  • 15 Avanzas P. et al. Multiple complex stenoses, high neutrophil count and C-reactive protein levels in patients with chronic stable angina. Atherosclerosis 2004; 175: 151-157.
  • 16 Haumer M. et al. Association of neutrophils and future cardiovascular events in patients with peripheral artery disease. J Vasc Surg 2005; 41: 610-617.
  • 17 Grau AJ. et al. Leukocyte count as an independent predictor of recurrent ischaemic events. Stroke 2004; 35: 1147-1152.
  • 18 Gillum RF. et al. Counts of neutrophils, lymphocytes, and monocytes, cause-specific mortality and coronary heart disease: the NHANES-I epidemiologic follow-up study. Ann Epidemiol 2005; 15: 266-271.
  • 19 Horne BD. et al. Intermountain Heart Collaborative Study Group. Which white blood cell subtypes predict increased cardiovascular risk?. J Am Coll Cardiol 2005; 45: 1638-1643.
  • 20 Nijm J. et al. Circulating levels of proinflammatory cytokines and neutrophil-platelet aggregates in patients with coronary artery disease. Am J Cardiol 2005; 95: 452-456.
  • 21 Tsai JC. et al. Association of peripheral total and differential leukocyte counts with metabolic syndrome and risk of ischaemic cardiovascular diseases in patients with type 2 diabetes mellitus. Diabetes Metab Res Rev 2007; 23: 111-118.
  • 22 Zazula AD. et al. An assessment of neutrophils/lymphocytes ratio in patients suspected of acute coronary syndrome. Arq Bras Cardiol 2008; 90: 31-36.
  • 23 Papa A. et al. Predictive value of elevated neutrophil-lymphocyte ratio on cardiac mortality in patients with stable coronary artery disease. Clin Chim Acta 2008; 395: 27-31.
  • 24 Karabinos I. et al. Neutrophil count on admission predicts major in-hospital events in patients with a non-ST-segment elevation acute coronary syndrome. Clin Cardiol 2009; 32: 561-568.
  • 25 Poludasu S. et al. Neutrophil to lymphocyte ratio as a predictor of long-term mortality in African Americans undergoing percutaneous coronary intervention. Clin Cardiol 2009; 32: E6-E10.
  • 26 Setianto BY. et al. Circulating soluble CD40 ligand mediates the interaction between neutrophils and platelets in acute coronary syndrome. Heart Vessels 2010; 25: 282-287.
  • 27 Muhmmed Suliman MA. et al. Predictive value of neutrophil to lymphocyte ratio in outcomes of patients with acute coronary syndrome. Arch Med Res 2010; 41: 618-622.
  • 28 Husser O. et al. White blood cell subtypes after STEMI: temporal evolution, association with cardiovascular magnetic resonance--derived infarct size and impact on outcome. Inflammation 2011; 34: 73-84.
  • 29 Park BJ. et al. Relationship of neutrophil-lymphocyte ratio with arterial stiffness and coronary calcium score. Clin Chim Acta 2011; 412: 925-929.
  • 30 Hartaigh B. et al. Which leukocyte subsets predict cardiovascular mortality? From the LUdwigshafen RIsk and Cardiovascular Health (LURIC) Study. Atherosclerosis 2012; 224: 161-169.
  • 31 Arbel Y. et al. Neutrophil/lymphocyte ratio is related to the severity of coronary artery disease and clinical outcome in patients undergoing angiography. Atherosclerosis 2012; 225: 456-460.
  • 32 Kaya MG. et al. Prognostic value of neutrophil/lymphocyte ratio in patients with ST-elevated myocardial infarction undergoing primary coronary intervention: A prospective, multicenter study. Int J Cardiol. 2012. epub ahead of print
  • 33 Park JJ. et al. Prognostic value of neutrophil to lymphocyte ratio in patients presenting with ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention. Am J Cardiol 2013; 111: 636-642.
  • 34 Hopps E. et al. Pathophysiology of polymorphonuclear leukocyte in arterial hypertension. Clin Hemorheol Microcirc 2009; 41: 209-218.
  • 35 Shurtz-Swirski R. et al. Involvement of peripheral polymorphonuclear leukocytes in oxidative stress and inflammation in type 2 diabetic patients. Diabetes Care 2001; 24: 104-110.
  • 36 Rudolph TK, Rudolph V, Baldus S. Contribution of myeloperoxidase to smoking-dependent vascular inflammation. Proc Am Thorac Soc 2008; 05: 820-823.
  • 37 Soehnlein O. et al. Functional alterations of myeloid cell subsets in hyperlipidaemia: relevance for atherosclerosis. J Cell Mol Med 2009; 13: 4293-4303.
  • 38 Guasti L. et al. Cytokine production from peripheral blood mononuclear cells and polymorphonuclear leukocytes in patients studied for suspected obstructive sleep apnea. Sleep Breath 2011; 15: 3-11.
  • 39 Turkmen K. et al. The relationship between neutrophil-to-lymphocyte ratio and inflammation in end-stage renal disease patients. Ren Fail 2012; 34: 155-159.
  • 40 Mehta J. et al. Neutrophil function in ischaemic heart disease. Circulation 1989; 79: 549-556.
  • 41 De Servi S. et al. Clinical and angiographic correlates of leukocyte activation in unstable angina. J Am Coll Cardiol 1995; 26: 1146-1150.
  • 42 Amaro A. et al. Activity of leucocyte elastase in women with coronary artery disease documented using angiography. J Cardiovasc Risk 1995; 02: 149-153.
  • 43 Biasucci LM. et al. Delayed neutrophil apoptosis in patients with unstable angina: relation to C-reactive protein and recurrence of instability. Eur Heart J 2009; 30: 2220-2225.
  • 44 Koşar F. et al. Plasma leukocyte elastase concentration and coronary artery disease. Angiology 1998; 49: 193-201.
  • 45 Smith FB. et al. Tissue plasminogen activator and leucocyte elastase as predictors of cardiovascular events in subjects with angina pectoris: Edinburgh Artery Study. Eur Heart J 2000; 21: 1607-1613.
  • 46 Zhang R. et al. Association between myeloperoxidase levels and risk of coronary artery disease. J Am Med Assoc 2001; 286: 2136-2142.
  • 47 Garlichs CD. et al. Delay of neutrophil apoptosis in acute coronary syndromes. J Leukoc Biol 2004; 75: 828-835.
  • 48 Buffon A. et al. Widespread coronary inflammation in unstable angina. N Engl J Med 2002; 347: 5-12.
  • 49 Sánchez de Miguel L. et al. Nitric oxide production by neutrophils obtained from patients during acute coronary syndromes: expression of the nitric oxide synthase isoforms. J Am Coll Cardiol 2002; 39: 818-825.
  • 50 Smith C. et al. Increased Levels of Neutrophil-Activating Peptide-2 in Acute Coronary Syndromes. J Am Coll Cardiol 2006; 48: 1591-1599.
  • 51 Mocatta TJ. et al. Plasma concentrations of myeloperoxidase predict mortality after myocardial infarction. J Am Coll Cardiol 2007; 49: 1993-2000.
  • 52 Roman RM. et al. Prognostic value of myeloperoxidase in coronary artery disease: comparison of unstable and stable angina patients. Coron Artery Dis 2010; 21: 129-136.
  • 53 Naruko T. et al. Increased expression and plasma levels of myeloperoxidase are closely related to the presence of angiographically-detected complex lesion morphology in unstable angina. Heart 2010; 96: 1716-1722.
  • 54 Cojocaru IM. et al. Plasma myeloperoxidase levels in patients with acute ischaemic stroke. Rom J Intern Med 2010; 48: 101-104.
  • 55 Kafkas N. et al. Serum levels of gelatinase associated lipocalin as indicator of the inflammatory status in coronary artery disease. Int J Inflam 2012; 2012: 189797
  • 56 Salonen I. et al. Serum myeloperoxidase is independent of the risk factors of atherosclerosis. J Clin Invest 2012; 23: 251-258.
  • 57 Soehnlein O, Lindbom L. Phagocyte partnership during the onset and resolution of inflammation. Nat Rev Immunol 2010; 10: 427-439.
  • 58 Quercioli A. et al. Receptor activator of NF-κB ligand (RANKL) increases the release of neutrophil products associated with coronary vulnerability. Thromb Haemost 2012; 107: 124-139.
  • 59 Zernecke A. et al. Protective role of CXC receptor 4/CXC ligand 12 unveils the importance of neutrophils in atherosclerosis. Circ Res 2008; 102: 209-217.
  • 60 Drechsler M. et al. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation 2010; 122: 1837-1845.
  • 61 van Leeuwen M, Gijbels MJJ, Duijvestijn A. et al. Accumulation of myeloperoxidase-positive neutrophils in atherosclerotic lesions in LDLR-/- mice. Arterioscler Thromb Vasc Biol 2008; 28: 84-89.
  • 62 Hosokawa T. et al. Neutrophil infiltration and oxidant-production in human atherosclerotic carotid plaques. Histol Histopathol 2011; 26: 1-11.
  • 63 Rotzius P. et al. ApoE-/-/Lysozyme MEGFP/EGFP mice as a versatile model to study monocyte and neutrophil trafficking in atherosclerosis. Atherosclerosis 2009; 202: 111-118.
  • 64 Lansky AJ. et al. Gender and the Extent of Coronary Atherosclerosis, Plaque Composition, and Clinical Outcomes in Acute Coronary Syndromes. J Clin Invest 2012; 05: S62-S72.
  • 65 Virmani R. et al. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol 2000; 20: 1262-1275.
  • 66 Richardson PD. et al. Influence of plaque configuration and stress distribution on fissuring of coronary atherosclerotic plaques. Lancet 1989; 02: 941-944.
  • 67 Virmani R. et al. Pathology of the vulnerable plaque. J Am Coll Cardiol 2006; 47: C13-C18.
  • 68 Burke AP. et al. Coronary risk factors and plaque morphology in men with coronary disease who died suddenly. N Engl J Med 1997; 336: 1276-1282.
  • 69 Shah PK. Mechanisms of plaque vulnerability and rupture. J Am Coll Cardiol 2003; 41: 15S-22S.
  • 70 Naruko T. et al. Neutrophil infiltration of culprit lesions in acute coronary syndromes. Circulation 2002; 106: 2894-2900.
  • 71 Ionita MG. et al. High neutrophil numbers in human carotid atherosclerotic plaques are associated with characteristics of rupture-prone lesions. Arterioscler Thromb Vasc Biol 2010; 30: 1842-1848.
  • 72 Leclercq A. et al. Involvement of intraplaque hemorrhage in atherothrombosis evolution via neutrophil protease enrichment. J Leukoc Biol 2007; 82: 1420-1429.
  • 73 Borregaard N. et al. Neutrophil granules: a library of innate immunity proteins. Trends Immunol 2007; 28: 340-345.
  • 74 Galis ZS. et al. Targeted disruption of the matrix metalloproteinase-9 gene impairs smooth muscle cell migration and geometrical arterial remodelling. Circ Res 2002; 91: 852-859.
  • 75 Choi ET. et al. Matrix metalloproteinase-9 modulation by resident arterial cells is responsible for injury-induced accelerated atherosclerotic plaque development in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2005; 25: 1020-1025.
  • 76 Kuzuya M. et al. Effect of MMP-2 deficiency on atherosclerotic lesion formation in apoE-deficient mice. Arterioscler Thromb Vasc Biol 2006; 26: 1120-1125.
  • 77 Allan JA. et al. Binding of gelatinases A and B to type-I collagen and other matrix components. Biochem J 1995; 309: 299-306.
  • 78 Bäck M. et al. Matrix Metalloproteinases in Atherothrombosis. Prog Cardiovasc Dis 2010; 52: 410-428.
  • 79 Dorweiler B. et al. Subendothelial infiltration of neutrophil granulocytes and liberation of matrix-destabilising enzymes in an experimental model of human neo-intima. Thromb Haemost 2008; 99: 373-381.
  • 80 Laxton RC. et al. A role of matrix metalloproteinase-8 in atherosclerosis. Circ Res 2009; 105: 921-929.
  • 81 Dollery CM. et al. Neutrophil elastase in human atherosclerotic plaques: production by macrophages. Circulation 2003; 107: 2829-2836.
  • 82 Henriksen PA, Sallenave JM. Human neutrophil elastase: mediator and therapeutic target in atherosclerosis. Int J Biochem Cell Biol 2008; 40: 1095-1100.
  • 83 Moreno JA. et al. In vitro and in vivo evidence for the role of elastase shedding of CD163 in human atherothrombosis. Eur Heart J 2012; 33: 252-263.
  • 84 Rao NV. et al. Characterisation of proteinase-3 (PR-3), a neutrophil serine proteinase. Structural and functional properties. J Biol Chem 1991; 266: 9540-9548.
  • 85 Pezzato E. Proteinase-3 directly activates MMP-2 and degrades gelatin and Matrigel; differential inhibition by (-)epigallocatechin-3-gallate. J Leukoc Biol 2003; 74: 88-94.
  • 86 Kuckleburg CJ. et al. Proteinase 3 contributes to transendothelial migration of NB1-positive neutrophils. J Immunol 2012; 188: 2419-2426.
  • 87 Nauseef WM. How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev 2007; 219: 88-102.
  • 88 Prokopowicz Z. et al. Neutrophil Myeloperoxidase: Soldier and Statesman. Arch Immunol Ther Exp 2012; 60: 43-54.
  • 89 Daugherty A. et al. Myeloperoxidase, a catalyst for lipoprotein oxidation, is expressed in human atherosclerotic lesions. J Clin Invest 1994; 94: 437-444.
  • 90 Malle E. et al. Immunohistochemical evidence for the myeloperoxidase/H2O2/halide system in human atherosclerotic lesions: colocalisation of myeloperoxidase and hypochlorite-modified proteins. Eur J Biochem 2000; 267: 4495-4503.
  • 91 Hochholzer W. et al. Novel biomarkers in cardiovascular disease: Update 2010. Am Heart J 2010; 160: 583-594.
  • 92 Zhang R. et al. Myeloperoxidase functions as a major enzymatic catalyst for initiation of lipid peroxidation at sites of inflammation. J Biol Chem 2002; 277: 46116-46122.
  • 93 Podrez EA. et al. Macrophage scavenger receptor CD36 is the major receptor for LDL modified by monocyte-generated reactive nitrogen species. J Clin Invest 2000; 105: 1095-1108.
  • 94 Fu X. et al. Hypochlorous acid oxygenates the cysteine switch domain of pro-matrilysin (MMP-7). A mechanism for matrix metalloproteinase activation and atherosclerotic plaque rupture by myeloperoxidase. J Biol Chem 2001; 276: 41279-41287.
  • 95 Kebir El D. et al. Myeloperoxidase delays neutrophil apoptosis through CD11b/CD18 integrins and prolongs inflammation. Circ Res 2008; 103: 352-359.
  • 96 Penn MS. et al. Smooth muscle cell surface tissue factor pathway activation by oxidized low-density lipoprotein requires cellular lipid peroxidation. Blood 2000; 96: 3056-3063.
  • 97 Drechsler M. et al. Neutrophilic granulocytes - promiscuous accelerators of atherosclerosis. Thromb Haemost 2011; 106: 839-848.
  • 98 Ouyang W. et al. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity 2008; 28: 454-467.
  • 99 Pejnovic N. et al. Increased atherosclerotic lesions and Th17 in interleukin-18 deficient apolipoprotein E-knockout mice fed high-fat diet. Mol Immunol 2009; 47: 37-45.
  • 100 Smith E. et al. Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E-deficient mice. Circulation 2010; 121: 1746-1755.
  • 101 Liao YH. et al. Interleukin-17A contributes to myocardial ischaemia/reperfusion injury by regulating cardiomyocyte apoptosis and neutrophil infiltration. J Am Coll Cardiol 2012; 59: 420-429.
  • 102 Pelletier M. et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood 2010; 115: 335-343.
  • 103 Liuzzo G. et al. Interleukin-17 in atherosclerosis and cardiovascular disease: the good, the bad, and the unknown. Eur Heart J 2013; 34: 556-559.
  • 104 Lee A. et al. Inhibition of apoptosis and prolongation of neutrophil functional longevity by inflammatory mediators. J Leukoc Biol 1993; 54: 283-288.
  • 105 Yamashiro S. et al. Expression of CCR6 and CD83 by cytokine-activated human neutrophils. Blood 2000; 96: 3958-3963.
  • 106 Kumar V, Sharma A. Neutrophils: Cinderella of innate immune system. Int Immunopharmacol 2010; 10: 1325-1334.
  • 107 Iking-Konert C. et al. Transdifferentiation of polymorphonuclear neutrophils: acquisition of CD83 and other functional characteristics of dendritic cells. J Mol Med 2001; 79: 464-474.
  • 108 Abi Abdallah DS. et al. Mouse neutrophils are professional antigen-presenting cells programmed to instruct Th1 and Th17 T-cell differentiation. Int Immunol 2011; 23: 317-326.
  • 109 Farb A. et al. Coronary plaque erosion without rupture into a lipid core. A frequent cause of coronary thrombosis in sudden coronary death. Circulation 1996; 93: 1354-1363.
  • 110 Mazor R. et al. Primed polymorphonuclear leukocytes constitute a possible link between inflammation and oxidative stress in hyperlipidemic patients. Atherosclerosis 2008; 197: 937-943.
  • 111 Eiserich JP. et al. Formation of nitric oxide-derived inflammatory oxidants by myeloperoxidase in neutrophils. Nature 1998; 391: 393-397.
  • 112 Förstermann U. Nitric oxide and oxidative stress in vascular disease. Pflugers Arch 2010; 459: 923-939.
  • 113 Calcerrada P. et al. Nitric oxide-derived oxidants with a focus on peroxynitrite: molecular targets, cellular responses and therapeutic implications. Curr Pharm Des 2011; 17: 3905-3932.
  • 114 Hazell LJ. et al. Presence of hypochlorite-modified proteins in human atherosclerotic lesions. J Clin Invest 1996; 97: 1535-1544.
  • 115 Sugiyama S. et al. Hypochlorous acid, a macrophage product, induces endothelial apoptosis and tissue factor expression: involvement of myeloperoxidase-mediated oxidant in plaque erosion and thrombogenesis. Arterioscler Thromb Vasc Biol 2004; 24: 1309-1314.
  • 116 Rowe RG, Weiss SJ. Breaching the basement membrane: who, when and how?. Trends Cell Biol 2008; 18: 560-574.
  • 117 Montecucco F. et al. Anti-Apolipoprotein A-1 auto-antibodies are active mediators of atherosclerotic plaque vulnerability. Eur Heart J 2011; 32: 412-421.
  • 118 Soehnlein O. et al. Neutrophil secretion products pave the way for inflammatory monocytes. Blood 2008; 112: 1461-1471.
  • 119 Chertov O. et al. Identification of human neutrophil-derived cathepsin G and azurocidin/CAP37 as chemoattractants for mononuclear cells and neutrophils. J Exp Med 1997; 186: 739-747.
  • 120 Sun R. et al. Identification of neutrophil granule protein cathepsin G as a novel chemotactic agonist for the G protein-coupled formyl peptide receptor. J Immunol 2004; 173: 428-436.
  • 121 Ott I. et al. Increased neutrophil-platelet adhesion in patients with unstable angina. Circulation 1996; 94: 1239-1246.
  • 122 Adlbrecht C. et al. Active endothelin is an important vasoconstrictor in acute coronary thrombi. Thromb Haemost 2007; 97: 642-649.
  • 123 Badrnya S. et al. Platelets directly enhance neutrophil transmigration in response to oxidised low-density lipoprotein. Thromb Haemost 2012; 108: 719-729.
  • 124 Huo Y. et al. Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E. Nat Med 2003; 09: 61-67.
  • 125 Vanichakarn P. et al. Neutrophil CD40 enhances platelet-mediated inflammation. Thromb Res 2008; 122: 346-358.
  • 126 Li G. et al. CD40 Ligand Promotes Mac-1 Expression, Leukocyte Recruitment, and Neointima Formation after Vascular Injury. J Clin Invest 2008; 172: 1141-1152.
  • 127 Fuchs TA. et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA 2010; 107: 15880-15885.
  • 128 King III SB. et al. Revascularisation for Coronary Artery Disease: Stents Versus Bypass Surgery. Annu Rev Med 2010; 61: 199-213.
  • 129 Inoue T, Node K. Molecular basis of restenosis and novel issues of drug-eluting stents. Circ J 2009; 73: 615-621.
  • 130 Libby P. et al. A cascade model for restenosis. A special case of atherosclerosis progression. Circulation 1992; 86: III47-III52.
  • 131 Rogers C. et al. A mAb to the beta2-leukocyte integrin Mac-1 (CD11b/CD18) reduces intimal thickening after angioplasty or stent implantation in rabbits. Proc Natl Acad Sci USA 1998; 95: 10134-10139.
  • 132 Simon DI. et al. Decreased neointimal formation in Mac-1(-/-) mice reveals a role for inflammation in vascular repair after angioplasty. J Clin Invest 2000; 105: 293-300.
  • 133 Hayashi S. et al. Roles of P-selectin in inflammation, neointimal formation, and vascular remodelling in balloon-injured rat carotid arteries. Circulation 2000; 102: 1710-1717.
  • 134 Wang K. et al. Platelet, not endothelial, P-selectin is required for neointimal formation after vascular injury. Arterioscler Thromb Vasc Biol 2005; 25: 1584-1589.
  • 135 Hristov M. et al. Importance of CXC chemokine receptor 2 in the homing of human peripheral blood endothelial progenitor cells to sites of arterial injury. Circ Res 2007; 100: 590-597.
  • 136 Liehn EA. et al. Blockade of keratinocyte-derived chemokine inhibits endothelial recovery and enhances plaque formation after arterial injury in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 2004; 24: 1891-1896.
  • 137 Xing D. et al. Endothelial cells overexpressing interleukin-8 receptors reduce inflammatory and neointimal responses to arterial injury. Circulation 2012; 125: 1533-1541.
  • 138 Soehnlein O. et al. Neutrophil-derived cathelicidin protects from neointimal hyperplasia. Sci Transl Med 2011; 03: 103ra98.
  • 139 Frangogiannis NG. Regulation of the inflammatory response in cardiac repair. Circ Res 2012; 110: 159-173.
  • 140 Arslan F. et al. Myocardial ischaemia/reperfusion injury is mediated by leukocytic toll-like receptor-2 and reduced by systemic administration of a novel anti-toll-like receptor-2 antibody. Circulation 2010; 121: 80-90.
  • 141 Mann DL. The emerging role of innate immunity in the heart and vascular system: for whom the cell tolls. Circ Res 2011; 108: 1133-1145.
  • 142 Andrassy M. et al. High-mobility group box-1 in ischaemia-reperfusion injury of the heart. Circulation 2008; 117: 3216-3226.
  • 143 Hill JH, Ward PA. The phlogistic role of C3 leukotactic fragments in myocardial infarcts of rats. J Exp Med 1971; 133: 885-900.
  • 144 Meldrum DR. et al. Hydrogen peroxide induces tumor necrosis factor alpha-mediated cardiac injury by a P38 mitogen-activated protein kinase-dependent mechanism. Surgery 1998; 124: 291-296.
  • 145 Sellak H. et al. Reactive oxygen species rapidly increase endothelial ICAM-1 ability to bind neutrophils without detectable upregulation. Blood 1994; 83: 2669-2677.
  • 146 Gordon JW. et al. Multiple facets of NF-kB in the heart: to be or not to NF-kB. Circ Res 2011; 108: 1122-1132.
  • 147 Kleinbongard P. et al. TNFalpha in atherosclerosis, myocardial ischaemia/reperfusion and heart failure. Pharmacol Ther 2010; 127: 295-314.
  • 148 Bujak M. et al. Interleukin-1 receptor type I signalling critically regulates infarct healing and cardiac remodelling. Am J Pathol 2008; 173: 57-67.
  • 149 Frangogiannis NG. Chemokines in ischaemia and reperfusion. Thromb Haemost 2007; 97: 738-747.
  • 150 Dewald O. et al. Of mice and dogs: species-specific differences in the inflammatory response following myocardial infarction. Am J Pathol 2004; 164: 665-677.
  • 151 Briaud SA. et al. Leukocyte trafficking and myocardial reperfusion injury in ICAM-1/P-selectin-knockout mice. Am J Physiol Heart Circ Physiol 2001; 280: H60-H67.
  • 152 Metzler B. et al. Mouse model of myocardial remodelling after ischaemia: role of intercellular adhesion molecule-1. Cardiovasc Res 2001; 49: 399-407.
  • 153 Bournazou I. et al. Apoptotic human cells inhibit migration of granulocytes via release of lactoferrin. J Clin Invest 2009; 119: 20-32.
  • 154 Nahrendorf M. et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med 2007; 204: 3037-3047.
  • 155 Dutta P. et al. Myocardial infarction accelerates atherosclerosis. Nature 2012; 487: 325-329.
  • 156 Jordan J. The role of neutrophils in myocardial ischaemia-reperfusion injury. J Clin Invest 1999; 43: 860-878.
  • 157 Entman ML, Smith CW. Postreperfusion inflammation: a model for reaction to injury in cardiovascular disease. Cardiovasc Res 1994; 28: 1301-1311.
  • 158 Frame LH. et al. Early membrane damage during coronary reperfusion in dogs. Detection by radiolabeled anticardiac myosin (Fab’)2. J Clin Invest 1983; 72: 535-544.
  • 159 Farb A. et al. Myocardial infarct extension during reperfusion after coronary artery occlusion: pathologic evidence. J Am Coll Cardiol 1993; 21: 1245-1253.
  • 160 Rochitte CE. et al. Magnitude and time course of microvascular obstruction and tissue injury after acute myocardial infarction. Circulation 1998; 98: 1006-1014.
  • 161 Vinten-Johansen J. Involvement of neutrophils in the pathogenesis of lethal myocardial reperfusion injury. Cardiovasc Res 2004; 61: 481-497.
  • 162 Liehn EA. et al. Double-edged role of the CXCL12/CXCR4 axis in experimental myocardial infarction. J Am Coll Cardiol 2011; 58: 2415-2423.
  • 163 Walter DH. et al. Impaired CXCR4 signalling contributes to the reduced neovascularisation capacity of endothelial progenitor cells from patients with coronary artery disease. Circ Res 2005; 97: 1142-1151.
  • 164 Corsini A. et al. New insights into the pharmacodynamic and pharmacokinetic properties of statins. Pharmacol Ther 1999; 84: 413-428.
  • 165 Dunzendorfer S. et al. Mevalonate-Dependent Inhibition of Transendothelial Migration and Chemotaxis of Human Peripheral Blood Neutrophils by Pravastatin. Circ Res 1997; 81: 963-969.
  • 166 Kaneider NC. et al. Induction of apoptosis and inhibition of migration of inflammatory and vascular wall cells by cerivastatin. Atherosclerosis 2001; 158: 23-33.
  • 167 Chello M. et al. Simvastatin increases neutrophil apoptosis and reduces inflammatory reaction after coronary surgery. Ann Thorac Surg 2007; 83: 1374-1380.
  • 168 Sugano R. et al. Polymorphonuclear leukocytes may impair endothelial function: results of crossover randomized study of lipid-lowering therapies. Arterioscler Thromb Vasc Biol 2005; 25: 1262-1267.
  • 169 Eccles KA. et al. Simvastatin alters human endothelial cell adhesion molecule expression and inhibits leukocyte adhesion under flow. Atherosclerosis 2008; 200: 69-79.
  • 170 Walter T. et al. Effect of atorvastatin on cellular adhesion molecules on leukocytes in patients with normocholesterolemic coronary artery disease. In Vivo 2010; 24: 189-193.
  • 171 Kinsella A. et al. The impact of high-dose statin therapy on transendothelial neutrophil migration and serum cholesterol levels in healthy male volunteers. J Clin Invest 2011; 67: 1103-1108.
  • 172 Omi H. et al. Statins inhibit high glucose-mediated neutrophil-endothelial cell adhesion through decreasing surface expression of endothelial adhesion molecules by stimulating production of endothelial nitric oxide. J Clin Invest 2003; 65: 118-124.
  • 173 Okouchi M. et al. Cerivastatin ameliorates high insulin-enhanced neutrophil-endothelial cell adhesion and endothelial intercellular adhesion molecule-1 expression by inhibiting mitogen-activated protein kinase activation. J Clin Invest 2003; 17: 380-386.
  • 174 Bandoh T. et al. Antioxidative Potential of Fluvastatin via the Inhibition of Nicotinamide Adenine Dinucleotide Phosphate (NADPH) Oxidase Activity. J Clin Invest 2003; 26: 818-822.
  • 175 Kowalski J. et al. Neutrophil superoxide anion generation during atorvastatin and fluvastatin therapy used in coronary heart disease primary prevention. J Cardiovasc Pharmacol 2006; 48: 143-147.
  • 176 Schönbeck U, Libby P. Inflammation, immunity, and HMG-CoA reductase inhibitors: statins as antiinflammatory agents?. Circulation 2004; 109: II18-26.
  • 177 Alblas J. et al. Activation of Rhoa and ROCK are essential for detachment of migrating leukocytes. Mol Biol Cell 2001; 12: 2137-2145.
  • 178 Maher BM. et al. Statins alter neutrophil migration by modulating cellular Rho activity--a potential mechanism for statins-mediated pleotropic effects?. J Leukoc Biol 2009; 85: 186-193.
  • 179 Kaneider NC. et al. Rho-GTPase-dependent platelet-neutrophil interaction affected by HMG-CoA reductase inhibition with altered adenosine nucleotide release and function. Arterioscler Thromb Vasc Biol 2002; 22: 1029-1035.
  • 180 Nakamura K. et al. Statin prevents plaque disruption in apoE-knockout mouse model through pleiotropic effect on acute inflammation. Atherosclerosis 2009; 206: 355-361.
  • 181 Davidson MH, Toth PP. High-density lipoprotein metabolism: potential therapeutic targets. Am J Cardiol 2007; 100: n32-n40.
  • 182 Murphy AJ. et al. Neutrophil activation is attenuated by high-density lipoprotein and apolipoprotein A-I in in vitro and in vivo models of inflammation. Arterioscler Thromb Vasc Biol 2011; 31: 1333-1341.
  • 183 Kannan KB. et al. Free cholesterol alters lipid raft structure and function regulating neutrophil Ca2+ entry and respiratory burst: correlations with calcium channel raft trafficking. J Immunol 2007; 178: 5253-5261.
  • 184 Shao D. et al. Lipid rafts determine efficiency of NADPH oxidase activation in neutrophils. FEBS Lett 2003; 550: 101-106.
  • 185 Furlaneto CJ. et al. Apolipoproteins A-I and A-II downregulate neutrophil functions. Lipids 2002; 37: 925-928.
  • 186 Liao X-L. et al. Neutrophils activation can be diminished by apolipoprotein A-I. Life Sci 2005; 77: 325-335.
  • 187 Nicholls SJ. et al. Reconstituted high-density lipoproteins inhibit the acute pro-oxidant and proinflammatory vascular changes induced by a periarterial collar in normocholesterolemic rabbits. Circulation 2005; 111: 1543-1550.
  • 188 Puranik R. et al. Low dose apolipoprotein A-I rescues carotid arteries from inflammation in vivo. Atherosclerosis 2008; 196: 240-247.
  • 189 Woollard KJ. et al. Pathophysiological levels of soluble P-selectin mediate adhesion of leukocytes to the endothelium through Mac-1 activation. Circ Res 2008; 103: 1128-1138.
  • 190 Pierini LM. et al. Membrane lipid organisation is critical for human neutrophil polarisation. J Biol Chem 2003; 278: 10831-10841.
  • 191 Itoh S. et al. Redistribution of P-selectin glycoprotein ligand-1 (PSGL-1) in chemokine-treated neutrophils: a role of lipid microdomains. J Leukoc Biol 2007; 81: 1414-1421.
  • 192 Oh H. et al. Membrane cholesterol is a biomechanical regulator of neutrophil adhesion. Arterioscler Thromb Vasc Biol 2009; 29: 1290-1297.
  • 193 Yvan-Charvet L. et al. ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation. Science 2010; 328: 1689-1693.
  • 194 Murphy AJ. et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J Clin Invest 2011; 121: 4138-4149.
  • 195 Westerterp M. et al. Regulation of hematopoietic stem and progenitor cell mobilisation by cholesterol efflux pathways. Cell Stem Cell 2012; 11: 195-206.
  • 196 Murphy PM. et al. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol Rev 2000; 52: 145-176.
  • 197 Montecucco F. et al. Induction of neutrophil chemotaxis by leptin: crucial role for p38 and Src kinases. Ann NY Acad Sci 2006; 1069: 463-471.
  • 198 Braunersreuther V. et al. Chemokine CCL5/RANTES inhibition reduces myocardial reperfusion injury in atherosclerotic mice. J Mol Cell Cardiol 2010; 48: 789-798.
  • 199 Braunersreuther V. et al. A novel RANTES antagonist prevents progression of established atherosclerotic lesions in mice. Arterioscler Thromb Vasc Biol 2008; 28: 1090-1096.
  • 200 Mahler DA. et al. Efficacy and safety of a monoclonal antibody recognising interleukin-8 in COPD: a pilot study. Chest 2004; 126: 926-934.
  • 201 Montecucco F. et al. CC chemokine CCL5 plays a central role impacting infarct size and post-infarction heart failure in mice. Eur Heart J 2012; 33: 1964-1974.
  • 202 Montecucco F. et al. Single administration of the CXC chemokine-binding protein Evasin-3 during ischaemia prevents myocardial reperfusion injury in mice. Arterioscler Thromb Vasc Biol 2010; 30: 1371-1377.
  • 203 Widdowson KL. et al. Evaluation of potent and selective small-molecule antagonists for the CXCR2 chemokine receptor. J Med Chem 2004; 47: 1319-1321.
  • 204 Tarzami ST. et al. Opposing effects mediated by the chemokine receptor CXCR2 on myocardial ischaemia-reperfusion injury: recruitment of potentially damaging neutrophils and direct myocardial protection. Circulation 2003; 108: 2387-2392.
  • 205 Liehn EA. et al. Ccr1 deficiency reduces inflammatory remodelling and preserves left ventricular function after myocardial infarction. J Cell Mol Med 2008; 12: 496-506.
  • 206 Montecucco F. et al. Inhibition of nicotinamide phosphoribosyltransferase reduces neutrophil-mediated injury in myocardial infarction. Antioxid. Redox Signal 2013; 18: 630-641.
  • 207 Bauer I. et al. The NAD+-dependent histone deacetylase SIRT6 promotes cytokine production and migration in pancreatic cancer cells by regulating Ca2+ responses. J Biol Chem 2012; 287: 40924-40937.