Thromb Haemost 2016; 115(03): 501-508
DOI: 10.1160/th15-07-0614
Theme Issue Article
Schattauer GmbH

Animal models for plaque rupture: a biomechanical assessment

Kim Van der Heiden*
1   Department of Biomedical Engineering, Thoraxcenter, Erasmus Medical Center, Rotterdam, The Netherlands
,
Ayla Hoogendoorn*
1   Department of Biomedical Engineering, Thoraxcenter, Erasmus Medical Center, Rotterdam, The Netherlands
,
Mat J. Daemen
2   Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
,
Frank J. H. Gijsen
1   Department of Biomedical Engineering, Thoraxcenter, Erasmus Medical Center, Rotterdam, The Netherlands
› Author Affiliations
Further Information

Publication History

Received: 31 July 2015

Accepted after minor revision: 22 October 2015

Publication Date:
20 March 2018 (online)

Summary

Rupture of atherosclerotic plaques is the main cause of acute cardiovascular events. Animal models of plaque rupture are rare but essential for testing new imaging modalities to enable diagnosis of the patient at risk. Moreover, they enable the design of new treatment strategies to prevent plaque rupture. Several animal models for the study of atherosclerosis are available. Plaque rupture in these models only occurs following severe surgical or pharmaceutical intervention. In the process of plaque rupture, composition, biology and mechanics each play a role, but the latter has been disregarded in many animal studies. The biomechanical environment for atherosclerotic plaques is comprised of two parts, the pressure-induced stress distribution, mainly - but not exclusively – influenced by plaque composition, and the strength distribution throughout the plaque, largely determined by the inflammatory state. This environment differs considerably between humans and most animals, resulting in suboptimal conditions for plaque rupture. In this review we describe the role of the biomechanical environment in plaque rupture and assess this environment in animal models that present with plaque rupture.

* These authors contributed equally.


 
  • References

  • 1 Jia H. et al. In vivo diagnosis of plaque erosion and calcified nodule in patients with acute coronary syndrome by intravascular optical coherence tomography. J Am Coll Cardiol 2013; 62: 1748-1758.
  • 2 Van der Wal AC. et al. Site of intimal rupture or erosion of thrombosed coronary atherosclerotic plaques is characterized by an inflammatory process irrespective of the dominant plaque morphology. Circulation 1994; 89: 36-44.
  • 3 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.
  • 4 Muller JE. et al. Circadian variation and triggers of onset of acute cardiovascular disease. Circulation 1989; 79: 733-743.
  • 5 Virmani R. et al. Pathology of the vulnerable plaque. J Am Coll Cardiol 2006; 47: C13-18.
  • 6 Libby P, Pasterkamp G. Requiem for the ‘vulnerable plaque’. Eur Heart J. 2015 Epub ahead of print.
  • 7 Mann JM, Davies MJ. Vulnerable plaque. Relation of characteristics to degree of stenosis in human coronary arteries. Circulation 1996; 94: 928-931.
  • 8 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.
  • 9 Davies MJ. Stability and Instability: Two Faces of Coronary Atherosclerosis: The Paul Dudley White Lecture 1995. Circulation 1996; 94: 2013-2020.
  • 10 Falk E. et al. Coronary Plaque Disruption. Circulation 1995; 92: 657-671.
  • 11 Schaar JA. et al. Terminology for high-risk and vulnerable coronary artery plaques. Report of a meeting on the vulnerable plaque, June 17 and 18, 2003, Santorini, Greece. Eur Heart J 2004; 25: 1077-1082.
  • 12 Stone GW. et al. A prospective natural-history study of coronary atherosclerosis. N Engl J Med 2011; 364: 226-235.
  • 13 Hofman A. et al. The Rotterdam Study: 2012 objectives and design update. Eur J Epidemiol 2011; 26: 657-686.
  • 14 Stary HC. Natural History and Histological Classification of Atherosclerotic Lesions: An Update. Arterioscler Thromb Vasc Biol 2000; 20: 1177-1178.
  • 15 Libby P, Hansson GK. Inflammation and immunity in diseases of the arterial tree: players and layers. Circ Res 2015; 116: 307-311.
  • 16 Koskinas KC. et al. Natural history of experimental coronary atherosclerosis and vascular remodeling in relation to endothelial shear stress: A serial, in vivo intravascular ultrasound study. Circulation 2010; 121: 2092-2101.
  • 17 Kubo T. et al. The Dynamic Nature of Coronary Artery Lesion Morphology Assessed by Serial Virtual Histology Intravascular Ultrasound Tissue Characterisation. J Am Coll Cardiol Elsevier Inc.; 2010; 55: 1590-1597.
  • 18 Lendon CL. et al. Atherosclerotic plaque caps are locally weakened when mac-rophages density is increased. Atherosclerosis 1991; 87: 87-90.
  • 19 Akyildiz AC. et al. Effects of intima stiffness and plaque morphology on peak cap stress. Biomed Eng Online 2011; 10: 25.
  • 20 Ohayon J. et al. Necrotic core thickness and positive arterial remodeling index: emergent biomechanical factors for evaluating the risk of plaque rupture. Am J Physiol Heart Circ Physiol 2008; 295: H717-727.
  • 21 Akyildiz AC. et al. The effects of plaque morphology and material properties on peak cap stress in human coronary arteries. Comput Methods Biomech Biomed Engin 2015; 1-9.
  • 22 Akyildiz AC. et al. Mechanical properties of human atherosclerotic intima tissue. J Biomech 2014; 47: 773-783.
  • 23 Walsh MT. et al. Uniaxial tensile testing approaches for characterisation of atherosclerotic plaques. J Biomech 2014; 47: 793-804.
  • 24 Cheng GC. et al. Distribution of circumferential stress in ruptured and stable atherosclerotic lesions. A structural analysis with histopathological correlation. Circulation 1993; 87: 1179-1187.
  • 25 Burleigh MC. et al. Collagen types I and III, collagen content, GAGs and mechanical strength of human atherosclerotic plaque caps: span-wise variations. Atherosclerosis 1992; 96: 71-81.
  • 26 Vengrenyuk Y. et al. Micro-CT based analysis of a new paradigm for vulnerable plaque rupture: cellular microcalcifications in fibrous caps. Mol Cell Biomech 2008; 5: 37-47.
  • 27 Gijsen FJH. et al. Carotid Plaque Morphological Classification Compared With Biomechanical Cap Stress: Implications for a Magnetic Resonance Imaging-Based Assessment. Stroke 2015; 46: 2124-2128.
  • 28 Getz GS, Reardon CA. Animal models of atherosclerosis. Arterioscler Thromb Vasc Biol 2012; 32: 1104-1115.
  • 29 Bentzon JF, Falk E. Atherosclerotic lesions in mouse and man: is it the same disease?. Curr Opin Lipidol 2010; 21: 434-440.
  • 30 Miller NE. Associations of high-density lipoprotein subclasses and apolipopro-teins with ischaemic heart disease and coronary atherosclerosis. Am Heart J 1987; 113: 589-597.
  • 31 Falk E. Pathogenesis of atherosclerosis. J Am Coll Cardiol 2006; 47: C7-12.
  • 32 Kolodgie FD. et al. Intraplaque haemorrhage and progression of coronary athe-roma. N Engl J Med 2003; 349: 2316-2325.
  • 33 Jackson CL. et al. Assessment of unstable atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2007; 27: 714-720.
  • 34 Hansson GK, Heistad DD. Two views on plaque rupture. Arterioscler Thromb Vasc Biol 2007; 27: 697.
  • 35 Schwartz SM. et al. Plaque rupture in humans and mice. Arterioscler Thromb Vasc Biol 2007; 27: 705-713.
  • 36 Bond AR, Jackson CL. The fat-fed apolipoprotein E knockout mouse brachio-cephalic artery in the study of atherosclerotic plaque rupture. J Biomed Biotech-nol 2011; 2011: 379069.
  • 37 Falk E. et al. Putative murine models of plaque rupture. Arterioscler Thromb Vasc Biol 2007; 27: 969-972.
  • 38 Jackson CL. Defining and defending murine models of plaque rupture. Arte-rioscler Thromb Vasc Biol 2007; 27: 973-977.
  • 39 Riou LM. et al. Effects of mechanical properties and atherosclerotic artery size on biomechanical plaque disruption - Mouse vs. human. J Biomech 2014; 47: 765-772.
  • 40 Campbell IC. et al. Biomechanical modeling and morphology analysis indicates plaque rupture due to mechanical failure unlikely in atherosclerosis-prone mice. Am J Physiol Heart Circ Physiol 2013; 304: H473-486.
  • 41 Hayenga HN. et al. Regional atherosclerotic plaque properties in ApoE-/-mice quantified by atomic force, immunofluorescence, and light microscopy. J Vasc Res 2011; 48: 495-504.
  • 42 Tracqui P. et al. Mapping elasticity moduli of atherosclerotic plaque in situ via atomic force microscopy. J Struct Biol 2011; 174: 115-123.
  • 43 Ohayon J. et al. Is arterial wall-strain stiffening an additional process responsible for atherosclerosis in coronary bifurcations?: an in vivo study based on dynamic CT and MRI. Am J Physiol Heart Circ Physiol 2011; 301: H1097-1106.
  • 44 Cardoso L. et al. Effect of tissue properties, shape and orientation of microcal-cifications on vulnerable cap stability using different hyperelastic constitutive models. J Biomech 2014; 47: 870-877.
  • 45 Von der Thüsen JH. et al. Induction of Atherosclerotic Plaque Rupture in Apoli-poprotein E-/- Mice After Adenovirus-Mediated Transfer of p53. Circulation 2002; 105: 2064-2070.
  • 46 Gough PJ. et al. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest 2006; 116: 59-69.
  • 47 Ma T. et al. Th17 cells and IL-17 are involved in the disruption of vulnerable plaques triggered by short-term combination stimulation in apolipoprotein E-knockout mice. Cell Mol Immunol 2013; 10: 338-348.
  • 48 Jin S. et al. Endogenous renovascular hypertension combined with low shear stress induces plaque rupture in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012; 32: 2372-2379.
  • 49 Teng Z. et al. Plaque haemorrhage in carotid artery disease: Pathogenesis, clinical and biomechanical considerations. J Biomech 2014; 47: 847-858.
  • 50 Johnson J. 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.
  • 51 Chen Y-C et al.. A novel mouse model of atherosclerotic plaque instability for drug testing and mechanistic/therapeutic discoveries using gene and microRNA expression profiling. Circ Res 2013; 113: 252-265.
  • 52 Van Herck JL. et al. Impaired fibrillin-1 function promotes features of plaque instability in apolipoprotein E-deficient mice. Circulation 2009; 120: 2478-2487.
  • 53 Van der Donckt C. et al. Elastin fragmentation in atherosclerotic mice leads to intraplaque neovascularisation, plaque rupture, myocardial infarction, stroke, and sudden death. Eur Heart J. 2014 ehu041.
  • 54 Roche-Molina M. et al. Induction of sustained hypercholesterolemia by single adeno-associated virus-mediated gene transfer of mutant hPCSK9. Arterioscler Thromb Vasc Biol 2015; 35: 50-59.
  • 55 Booth RF. et al. Rapid development of atherosclerotic lesions in the rabbit carotid artery induced by perivascular manipulation. Atherosclerosis 1989; 76: 257-268.
  • 56 Campbell DJ. et al. The effect of hypertension on the accumulation of lipids and the uptake of (3H)cholesterol by the aorta of normal-fed and cholesterol-fed rabbits. Atherosclerosis 1973; 18: 301-319.
  • 57 Friedman M, Byers SO. Aortic atherosclerosis intensification in rabbits by prior endothelial denudation. Arch Pathol 1965; 79: 345-356.
  • 58 Lehr HA. et al. Immunopathogenesis of atherosclerosis: endotoxin accelerates atherosclerosis in rabbits on hypercholesterolemic diet. Circulation 2001; 104: 914-920.
  • 59 Yin W. et al. A diet high in saturated fat and sucrose alters glucoregulation and induces aortic fatty streaks in New Zealand White rabbits. Int J Exp Diabetes Res 2002; 3: 179-184.
  • 60 Constantinides P, Chakravarti RN. Rabbit arterial thrombosis production by systemic procedures. Arch Pathol 1961; 72: 197-208.
  • 61 Johnstone MT. et al. In vivo magnetic resonance imaging of experimental thrombosis in a rabbit model. Arterioscler Thromb Vasc Biol 2001; 21: 1556-1560.
  • 62 Fang S-M et al.. Developing a novel rabbit model of atherosclerotic plaque rupture and thrombosis by cold-induced endothelial injury. J Biomed Sci 2009; 16: 39.
  • 63 Fan J. et al. Rabbit models for the study of human atherosclerosis: from patho-physiological mechanisms to translational medicine. Pharmacol Ther 2015; 146: 104-119.
  • 64 Shiomi M, Fan J. Unstable coronary plaques and cardiac events in myocardial infarction-prone Watanabe heritable hyperlipidemic rabbits: questions and quandaries. Curr Opin Lipidol 2008; 19: 631-636.
  • 65 Mahley RW. et al. Swine lipoproteins and atherosclerosis. Changes in the plasma lipoproteins and apoproteins induced by cholesterol feeding. Biochemistry 1975; 14: 2817-2823.
  • 66 Granada JF. et al. Porcine models of coronary atherosclerosis and vulnerable plaque for imaging and interventional research. EuroIntervention 2009; 5: 140-148.
  • 67 Rapacz J. et al. Lipoprotein mutations in pigs are associated with elevated plasma cholesterol and atherosclerosis. Science 1986; 234: 1573-1577.
  • 68 Prescott MF. et al. Development of complex atherosclerotic lesions in pigs with inherited hyper-LDL cholesterolemia bearing mutant alleles for apolipoprotein B. Am J Pathol 1991; 139: 139-147.
  • 69 Thim T. Human-like atherosclerosis in minipigs: a new model for detection and treatment of vulnerable plaques. Dan Med Bull 2010; 57: B4161.
  • 70 Gerrity RG. et al. Diabetes-induced accelerated atherosclerosis in swine. Diabetes 2001; 50: 1654-1665.
  • 71 Baker AB. et al. Regulation of heparanase expression in coronary artery disease in diabetic, hyperlipidemic swine. Atherosclerosis 2010; 213: 436-442.
  • 72 Mohler ER. et al. Site-specific atherogenic gene expression correlates with subsequent variable lesion development in coronary and peripheral vasculature. Arterioscler Thromb Vasc Biol 2008; 28: 850-855.
  • 73 Koskinas KC. et al. Thin-capped atheromata with reduced collagen content in pigs develop in coronary arterial regions exposed to persistently low endothelial shear stress. Arterioscler Thromb Vasc Biol 2013; 33: 1494-1504.
  • 74 Thim T. et al. Familial hypercholesterolaemic downsized pig with human-like coronary atherosclerosis: a model for preclinical studies. EuroIntervention 2010; 6: 261-268.
  • 75 Ishii A. et al. Swine model of carotid artery atherosclerosis: experimental induction by surgical partial ligation and dietary hypercholesterolemia. Am J Neur-oradiol 2006; 27: 1893-1899.
  • 76 Shi Z-S et al.. Vulnerable plaque in a Swine model of carotid atherosclerosis. Am J Neuroradiol 2009; 30: 469-472.
  • 77 Jiang X-B et al.. Overexpression of matrix metalloproteinase-9 is correlated with carotid intraplaque haemorrhage in a swine model. J Neurointerv Surg 2013; 5: 473-477.
  • 78 Soulez G. et al. Vulnerable carotid atherosclerotic plaque creation in a Swine model: evaluation of stenosis creation using absorbable and permanent suture in a diabetic dyslipidemic model. J Vasc Interv Radiol 2012; 23: 1700-1708 e4.
  • 79 Al-Mashhadi RH. et al. Abstract 17809: In Vivo High Resolution Isotropic 3D MRI of Coronary Atherosclerosis in Hypertensive Hypercholesterolemic Mini-pigs. Circulation 2014; 130: A17809.