Thromb Haemost 2009; 101(06): 1006-1011
DOI: 10.1160/TH08-07-0469
Theme Issue Article
Schattauer GmbH

Vulnerable atherosclerotic plaque metalloproteinases and foam cell phenotypes

Andrew C. Newby
1   University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
,
Sarah J. George
1   University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
,
Yasmin Ismail
1   University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
,
Jason L. Johnson
1   University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
,
Graciela B. Sala-Newby
1   University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
,
Anita C. Thomas
1   University of Bristol, Bristol Heart Institute, Bristol Royal Infirmary, Bristol, UK
› Author Affiliations
Financial support: The authors’ work is supported by the British Heart Foundation and the European Vascular Genomics Network.
Further Information

Publication History

Received: 23 July 2008

Accepted after minor revision: 15 January 2008

Publication Date:
24 November 2017 (online)

Summary

Plaque rupture underlies most myocardial infarctions. Plaques vulnerable to rupture have thin fibrous caps, an excess of macrophages over vascular smooth muscle cells, large lipid cores, and depletion of collagen and other matrix proteins form the cap and lipid core. Production of matrix metalloproteinases from macrophages is prominent in human plaques, and studies in genetically modified mice imply a causative role for metallopro teinases in plaque vulnerability. Recent in-vitro studies on human monocyte-derived macrophages and on foam-cell macrophages generated in vivo suggest the existence of several macrophage phenotypes with distinct patterns of metalloproteinase expression. These phenotypes could play differing roles in cap, core and aneurysm formation.

 
  • References

  • 1 Virmani R, Burke AP, Farb A. et al. Pathology of the vulnerable plaque. J Am Coll Cardiol 2006; 47 8 Supplement 1 C13-C18.
  • 2 Dollery CM, Libby P. Atherosclerosis and proteinase activation. Cardiovasc Res 2006; 69: 625-635.
  • 3 Baigent C, Keech A, Kearney PM. et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005; 366: 1267-1278.
  • 4 Crisby M, Nordin-Fredriksson G, Shah PK. et al. Pravastatin treatment increases collagen content and decreases lipid content, inflammation, metalloproteinases, and cell death in human carotid plaques: Implications for plaque stabilization. Circulation 2001; 103: 926-933.
  • 5 Underhill HR, Yuan C, Zhao XQ. et al. Effect of rosuvastatin therapy on carotid plaque morphology and composition in moderately hypercholesterolemic patients: a high-resolution magnetic resonance imaging trial. Am Heart J 2008; 155: 584e1-8.
  • 6 Van Mieghem CA, McFadden EP, de Feyter PJ. et al. Noninvasive detection of subclinical coronary atherosclerosis coupled with assessment of changes in plaque characteristics using novel invasive imaging modalities: the Integrated Biomarker and Imaging Study (IBIS). J Am Coll Cardiol 2006; 47: 1134-1142.
  • 7 Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res 2006; 69: 562-573.
  • 8 Rajagopalan S, Meng XP, Ramasamy S. et al. Reactive oxygen species produced by macrophage-derived foam cells regulate the activity of vascular matrix metalloproteinases in vitro. J Clin Invest 1996; 98: 2572-2579.
  • 9 Shimizu K, Shichiri M, Libby P, Lee RT, Mitchell RN. Th2-predominant inflammation and blockade of IFN-gamma signaling induce aneurysms in allografted aortas. J Clin Invest 2004; 114: 300-308.
  • 10 Newby AC. Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc Res 2006; 69: 614-624.
  • 11 Clarke MC, Figg N, Maguire JJ. et al. Apoptosis of vascular smooth muscle cells induces features of plaque vulnerability in atherosclerosis. Nat Med 2006; 12: 1075-1080.
  • 12 Johnson JL, Carson K, Williams HM. et al. Plaque rupture after short periods of fat-feeding in the apolipo-protein E knockout mouse: Model characterisation, and effects of pravastatin treatment. Circulation 2005; 111: 1422-1430.
  • 13 Lemaitre V, Soloway PD, D’Armiento J. Increased medial degradation with pseudo-aneurysm formation in apolipoprotein E-knockout mice deficient in tissue inhibitor of metalloproteinases-1. Circulation 2003; 107: 333-338.
  • 14 Silence J, Collen D, Lijnen HR. Reduced athero-sclerotic plaque but enhanced aneurysm formation in mice with inactivation of the tissue inhibitor of metalloproteinase-1 (TIMP-1) gene. Circ Res 2002; 90: 897-903.
  • 15 Johnson JL, Baker AH, Oka K. et al. Suppression of atherosclerotic plaque progression and instability by tissue inhibitor of metalloproteinase-2: Involvement of macrophage migration and apoptosis. Circulation 2006; 113: 2435-2444.
  • 16 Rouis M, Adamy C, Duverger N. et al. Adenovirus-mediate overexpression of tissue inhibitor of metalloproteinase-1 reduces atherosclerotic lesions in apolipo-protein E deficient mice. Circulation 1999; 100: 533-540.
  • 17 Johnson JL, Fritsche-Danielson R, Behrendt M. et al. Effect of broad-spectrum matrix metalloproteinase inhibition on atherosclerotic plaque stability. Cardiovasc Res 2006; 71: 586-595.
  • 18 Lemaitre V, O’Byrne TK, Borczuk AC. et al. ApoE knockout mice expressing human matrix metalloproteinase-1 in macrophages have less advanced atherosclerosis. J Clin Invest 2001; 107: 1227-1234.
  • 19 Gough PJ, Gomez IG, Wille PT. et al. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest 2006; 116: 59-69.
  • 20 de Nooijer R, Verkleij CJN, von der Thusen JH. et al. Lesional overexpression of matrix metalloproteinase-9 promotes intraplaque hemorrhage in advanced lesions but not at earlier stages of atherogenesis. Arterioscler Thromb Vasc Biol 2006; 26: 340-346.
  • 21 Silence J, Lupu F, Collen D. et al. Persistence of atherosclerotic plaque but reduced aneurysm formation in mice with stromelysin-1 (MMP-3) gene inactivation. Arterioscler Thromb Vasc Biol 2001; 21: 1440-1445.
  • 22 Johnson JL, George SJ, Newby AC. et al. Divergent effects of matrix metalloproteinases 3, 7, 9, and 12 on atherosclerotic plaque stability in mouse brachiocephalic arteries. Proc Natl Acad Sci USA 2005; 102: 15575-15580.
  • 23 Luttun A, Lutgens E, Manderveld A. et al. Loss of matrix metalloproteinase-9 or matrix metalloproteinase-12 protects apolipoprotein E-deficient mice against atherosclerotic media destruction but differentially affects plaque growth. Circulation 2004; 109: 1408-1414.
  • 24 Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol 2006; 6: 508-519.
  • 25 Tedgui A, Mallat Z. Cytokines in atherosclerosis: Pathogenic and regulatory pathways. Physiol Rev 2006; 86: 515-581.
  • 26 Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003; 19: 71-82.
  • 27 Libby P, Nahrendorf M, Pittet MJ. et al. Diversity of denizens of the atherosclerotic plaque: not all monocytes are created equal. Circulation 2008; 117: 3168-3170.
  • 28 Newby AC. Metalloproteinase Expression in Monocytes and Macrophages and its Relationship to Atherosclerotic Plaque Instability. Arterioscler Thromb Vasc Biol. 2008 in print.
  • 29 Brand K, Page S, Rogler G. et al. Activated transcription factor nuclear factor-kappa B is present in the atherosclerotic lesion. J Clin Invest 1996; 97: 1715-1722.
  • 30 Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol 2005; 5: 953-964.
  • 31 Bouhlel MA, Derudas B, Rigamonti E. et al. PPAR-gamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab 2007; 6: 137-143.
  • 32 Chase A, Bond M, Crook MF. et al. Role of nuclear factor-κB activation in metalloproteinase-1, -3 and -9 secretion by human macrophages in vitro and rabbit foam cells produced in vivo. Arterioscler Thromb Vasc Biol 2002; 22: 765-771.
  • 33 Johnson JL, Sala-Newby GB, Ismail Y. et al. Low tissue inhibitor of metalloproteinases 3 and high matrix metalloproteinase 14 levels defines a subpopulation of highly invasive foam-cell macrophages. Arterioscler Thromb Vasc Biol 2008; 28: 1647-1653.
  • 34 Thomas AC, Sala-Newby GB, Ismail Y. et al. Genomics of foam cells and nonfoamy macrophages from rabbits identifies arginase-I as a differential regulator of nitric oxide production. Arterioscler Thromb Vasc Biol 2007; 27: 571-577.
  • 35 Kuzuya M, Nakamura K, Sasaki T. et al. Effect of MMP-2 deficiency on atherosclerotic lesion formation in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 2006; 26: 1120-1125.
  • 36 Liang J, Liu E, Yu Y. et al. Macrophage metalloelastase accelerates the progression of atherosclerosis in transgenic rabbits. Circulation 2006; 113: 1993-2001.
  • 37 Deguchi J-O, Aikawa E, Libby P. et al. Matrix metalloproteinase-13/Collagenase-3 deletion promotes collagen accumulation and organization in mouse atherosclerotic plaques. Circulation 2005; 112: 2708-2715.
  • 38 Schneider F, Sukhova GK, Aikawa M. et al. Matrix-metalloproteinase-14 deficiency in bone-marrow-derived cells promotes collagen accumulation in mouse atherosclerotic plaques. Circulation 2008; 117: 931-939.