Skip to main content

Cardiac and vascular phenotypes in the apolipoprotein E-deficient mouse

Abstract

Cardiovascular death is frequently associated with atherosclerosis, a chronic multifactorial disease and a leading cause of death worldwide. Genetically engineered mouse models have proven useful for the study of the mechanisms underlying cardiovascular diseases. The apolipoprotein E-deficient mouse has been the most widely used animal model of atherosclerosis because it rapidly develops severe hypercholesterolemia and spontaneous atherosclerotic lesions similar to those observed in humans. In this review, we provide an overview of the cardiac and vascular phenotypes and discuss the interplay among nitric oxide, reactive oxygen species, aging and diet in the impairment of cardiovascular function in this mouse model.

Introduction

Over the last two decades, the mouse has become a very important experimental animal in studies of physiology and a growing number of pathophysiologies due to its easy breeding, short generation time and the availability of inbred strains. The limitations of the small size of the mouse for studying cardiac and vascular hemodynamics and function have been overcome in our and other laboratories due to advances in surgical techniques, as in Borst et al. [1], who described detailed procedures for the analysis of cardiac functional parameters in the mouse.

As a consequence of the progressive advancement of molecular biology techniques, it is possible to knockout and restore endogenous genes or add exogenous genes into the mouse, allowing the development of mouse models for human diseases. Two decades ago, the first gene-targeted murine model of atherosclerosis was created by the inactivation of the apolipoprotein E (apoE) gene by homologous recombination [2, 3]. Among the genetically engineered models, the apoE-deficient (apoE-/-) mouse is considered to be one of the most relevant models because it develops spontaneous hypercholesterolemia and arterial lesions similar to those observed in humans.

In the apoE-/- mouse, most of the plasma cholesterol (PC) is found in the atherogenic lipoprotein fractions, namely the very-low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL) and low-density lipoprotein (LDL) fractions, and this profile is aggravated by a Western-type diet. On a chow diet, apoE-/- mice exhibit increased total PC (~8-fold), triglycerides (1.7-fold), VLDL+IDL (18-fold) and LDL (14-fold) compared to C57BL/6J (C57) mice. When fed a Western-type diet, a dramatic increase in the proportions of these lipids is observed in total PC (~14-fold), particularly in the VLDL+IDL lipoprotein fraction (~30-fold) [2, 4–10].

The combination of the availability of mouse models of atherosclerosis and the technology to perform hemodynamic measurements in mice has enabled the study of the effects of hypercholesterolemia and/or atherosclerosis on cardiac vascular function. In this review, we focus on studies from our laboratory and from other investigators regarding the vascular and cardiac phenotypes in the apoE-/- mouse. We also discuss the underlying mechanisms that have been shown to contribute to cardiac and vascular dysfunction in this mouse model.

Vascular phenotypes in apoe-/- mice

Initiation and progression of atherosclerosis

The tendency to consider atherosclerosis as the effect of dyslipidemia or inflammation alone has been replaced by a new concept that this disease results from lipid disorders, enhanced oxidative stress and inflammation [11–18]. The development of atherosclerosis in conditions of hypercholesterolemia is initiated by the oxidation of lipoproteins such as LDL (oxLDL) in the subendothelial space and dysfunction of endothelial cells. In parallel endothelial cells express vascular and intercellular adhesion molecules (VCAM-1 and ICAM-1). Consequently, monocytes move into the subendothelial space where they undergo differentiation into macrophages under the influence of stimulating factors. These cells express scavenger receptors that recognize oxLDL and promote their phagocytosis. The degradation of oxLDL in lysosomes is slow, facilitating the formation of foam cells. The pivotal stage of atherogenesis seems to be antigen presentation by macrophages to T lymphocytes that recognize oxLDL and locally release proinflammatory cytokines such as interleukins (IL-1, 6, 12 and 18) and chemokines such as tumor necrosis factor alpha (TNFα) and monocyte chemoattractant protein-1 (MCP-1), thereby enhancing the inflammatory cascades. The accumulation and transformation of macrophages into large foam cells form fatty streaks that are characteristics of early atherosclerosis, which progresses into mature atherosclerotic plaques (fibrous caps) in the intima.

The mechanisms and progression of atherosclerosis in the apoE-/- mice have been described before [19–22]. In this model, atherosclerotic lesions are observed throughout the macrovasculature, with the most prevalent sites located in the aortic root, aortic arch, common carotid, superior mesenteric artery and renal and pulmonary arteries. Histopathological studies of the progression of the lesions in the apoE-/- mouse fed a chow diet revealed that the first signs of lesions appear at 6-8 weeks of age, as indicated by the attachment of monocytes to the endothelial cells and a disruption of the subendothelial elastic lamina [21, 22]. Lesions containing foam cells and smooth muscle cells are observed at 8-10 weeks and fibrous plaques appear at 15-20 weeks [21, 22]. The acceleration and severity of atherosclerotic lesions at all stages can be induced by a Western-type diet.

Hemodynamics: endothelial dysfunction

Recently, we reviewed the potential mechanisms underlying endothelial dysfunction in the apoE-/- mouse and how endothelial dysfunction is influenced by aging, gender and diet [23]. In brief, the conducting vessels of apoE-/- mice show an impaired endothelial nitric oxide (NO)-dependent relaxation response to acetylcholine (ACh), which is associated with plaque formation [6, 24, 25] and aggravated by a Western-type diet [10, 26–28] and by aging [24, 29]. As illustrated in Figure 1, the main potential mechanism underlying the endothelial dysfunction of apoE-/- mice may be the endothelial NO synthase (eNOS) pathway, due to the uncoupling of eNOS in the endothelium. This uncoupling decreases the bioavailability of NO, increases production of superoxide anion (•O2-) and generates peroxynitrite (•ONOO-), which is associated with the activation of the endothelin-1 (ET-1) system [6, 28, 30–33]. In resistance vessels, which rarely develop atherosclerotic lesions, impaired endothelial NO-dependent relaxation responses to ACh have also been observed, primarily in female apoE-/- mice [34, 35]. This effect appears to be mediated by hydrogen peroxide (H2O2) or an endothelium-derived hyperpolarizing factor (EDHF)-like principle agent, increased NADPH oxidase-derived •O2- and ET-1.

Figure 1
figure 1

A hypothetical scheme illustrating the role of ROS and decreased NO bioavailability in vascular dysfunction in conditions of hypercholesterolemia and atherosclerosis in the apoE-/- mouse. Abbreviations: ADMA, asymmetric dimethyl-L-arginine; BH4, tetrahydrobiopterin; eNOS, endothelial nitric oxide synthase; NADPH, nicotinamide adenine dinucleotide phosphate; NO, nitric oxide; •O2-, superoxide anion; •ONOO-, peroxynitrite; PARP, poly-ADP-ribose polymerase; PGI2, prostacyclin; ROS, reactive oxygen species.

Local oxidation of circulating lipoproteins and their incorporation within the vascular endothelium, associated with oxidative processes, plays a pivotal role in the pathogenesis of endothelial dysfunction in the apoE-/- mouse [6, 10]. The endothelial dysfunction promotes the entry and retention of LDL particles in the artery wall [36, 37] enhancing the progression of lesions and creating a vicious cycle. This idea is corroborated by the finding that in vitro treatment of aortas with oxLDL mimicked the endothelial NO dysfunction that was observed in apoE-/- mice [10]. The finding that gene transfer of human paraoxonase 1, which destroys lipid peroxides, into apoE-/- mice decreased the oxLDL content of the plaques and restored endothelial function [29] corroborates this point of view. Others have shown that treatment of apoE-/- mice with large empty phospholipid vesicles, which accelerates the reverse pathway of lipid transport from peripheral tissues to the liver, restored endothelium-dependent relaxation, leukocyte adherence, and endothelial expression of VCAM-1 to normal or nearly normal levels [38].

Hemodynamics: arterial blood pressure

Genetically engineered mouse models have been used to investigate hemodynamics in several cardiovascular diseases, including hypercholesterolemia and atherosclerosis. Several studies, in which blood pressure (BP) was recorded acutely, have shown that BP levels in young-adult (up to 30-week-old) hypercholesterolemic apoE-/- mice fed a standard chow diet or a Western-type diet are similar to those of control mice [7, 31, 32, 39–45]. Conversely, Yang et al. [46], observed normal resting values of BP in young apoE-/- mice but high levels in adult apoE-/- mice, compared with age-matched control mice. However, continuous and prolonged recordings of BP have revealed that apoE-/- mice exhibit elevated mean 24-hour BP levels, total abolition of BP circadian cycles and increased BP variability in the very-low-frequency band, which can be improved by the restoration of vagal and NOS-mediated regulation with statins [47].

The contribution of high BP to the development of atherosclerosis, and vice-versa, is being actively debated because data involving concurrent measurements of BP and atherosclerosis in mouse models have revealed an association in some studies and a lack of association in others between hypertension and atherosclerosis [48]. One of the first studies showed that subcutaneous infusions of norepinephrine induced increases in BP and in atherosclerotic lesion size in apoE-/- mice [33]. Considering that angiotensin II (Ang II) accelerates the development of atherosclerosis in both LDL receptor-/- and apoE-/- mice [33, 49, 50] and that hypercholesterolemia has been associated with activation of the renin-angiotensin system [51, 52], the apoE-/- mouse became an exciting model for evaluating the hemodynamic effects of Ang II in hypercholesterolemic mice (see a hypothetical mechanism in Figure 1). Currently, the suggested mechanisms by which Ang II promotes atherosclerosis include increased oxidative stress, the production of monocyte chemoattractants, and the diapedesis of monocytes [33]. Previous studies of chronic infusions of Ang II in apoE-/- mice have led to three conclusions: (a) Ang II-induced atherosclerosis was not associated with an increase in BP [53]; (b) Ang II-induced hypertension was not the direct cause of the profound increase in atherosclerotic lesions [33] because this increase in BP was similar in magnitude to that observed in wild-type control mice [54]; and (c) low doses of Ang II that did not affect BP increased aortic atherosclerotic lesions [55, 56]. Likewise, it has been shown that pharmacological blockade of the Ang II AT1 receptor in apoE-/- mice or genetic disruption of the AT1 receptor (apoE-/-/AT1-/- mice) reduces the inflammatory and atherosclerotic lesion process irrespective of BP [32, 57]. Additionally, long-term Ang (1-7) treatment has been shown to produce vasoprotective and atheroprotective actions in the apoE-/- mouse without direct effects on BP [45]. Conversely, it has been shown that AT1 receptor antagonist treatment lowered BP but had no effect on atherosclerosis [58]. Thus, it appears that the actions of Ang peptides on the pathogenesis of atherosclerosis and on BP occur through independent mechanisms. In our and other laboratories, this issue has been addressed by activating the renin-angiotensin system through partial clipping (stenosis) of the left renal artery (known as two kidney-one clip, or 2K1C, hypertension) in young apoE-/- mice. In animals with concurrent hypercholesterolemia and activation of the endogenous renin-angiotensin system, the level of arterial hypertension was similar to that observed in normocholesterolemic wild-type C57 mice with high Ang II [41, 42]. However, an increase in the thickness of the aortic wall is noted to some extent in the hypertensive C57 mouse and especially in the hypercholesterolemic apoE-/- mouse, and the concurrence of both factors (hypercholesterolemia and hypertension) is observed to exert an additive effect on the aortic wall thickness in the apoE-/- mouse [42]. Therefore, it appears that the development of arterial hypertension in atherosclerotic apoE-/- mice is associated with other concurrent factors, particularly aging, imbalance in NOS/reactive oxygen species (ROS) production and Ang II (see schematic illustration in Figure 1).

Vascular remodeling

The vessel wall of a plaque-prone region expands to protect the lumen from the expanding plaque (positive remodeling) until the plaque grows so extensively that the outer lumen can no longer compensate and the plaque begins to encroach upon the lumen (negative remodeling) [59, 60]. In apoE-/- mice, positive remodeling has been reported in aortas at both early and late stages of atherosclerosis [42, 59, 61–63]. However, this observation may not apply for all large vessels as, in carotid arteries, positive remodeling has been reported at the early stages [60], but negative remodeling has been reported in aged animals [59, 60]. A possible explanation for this discrepancy is that an excessive accumulation of lipids along the internal elastic lamina and within the media results in the death of medial cells and loss of the normal capacity to remodel in the carotid artery [59]. An additional and reasonable factor influencing or determining this difference between the carotid artery and the aorta in aged animals is that the animals were fed a regular diet in the study reporting positive remodeling [60], whereas the animals were fed a Western-type diet in the study reporting negative remodeling [59]. In addition to the lack of compensatory remodeling in the carotid arteries [59], we have observed that aged apoE-/- mice show an inward (negative) remodeling in the coronary arteries (unpublished data).

Interplay among lipid and lipoprotein oxidation, DNA damage and inflammation

Oxidative stress and inflammation are intimately linked to the evolution of atherosclerosis. Oxidative stress originated primarily from ROS in mitochondria can damage cells or components of cells by initiating chemical chain reactions, such as lipid peroxidation, leading to the formation of oxLDL and oxidizing proteins or DNA [64, 65]. LDL is oxidized in a slow process upon contact with ROS in the sub-intimal space by all major cells of the arterial wall via different mechanisms. ROS induce the oxidation of cholesterol and polyunsaturated fatty acids in LDL. Consequently, lipid peroxidation results in the formation of several products, such as oxysterols and aldehydes, which react with lysine residues of apolipoprotein B (apoB), generating oxLDL [14]. In addition to their metabolic deviation, oxLDL exhibits a large variety of biological and atherogenic properties involved in the activation of inflammatory, mitogenic or proapoptotic pathways, contributing to the progression of atherosclerosis [14, 66].

Another cellular target of ROS is DNA; oxidative DNA lesions have been described ranging from base modifications to single- and double-strand breaks [67]. Consequently, excessive oxidative stress-induced DNA damage, which was originally described as the limited ability of a cell to divide, also appears to contribute to the pathogenesis of age-associated vascular disorders [65, 68]. Recent studies in our laboratory [61] revealed vascular senescence in the aortas of atherosclerotic aged apoE-/- mice but not in non-atherosclerotic aged wild-type C57 mice. This result suggests that in the aorta the occurrence of vascular senescence in aging may be related to the development of vascular disease, and this phenomenon may be the cause of the endothelial dysfunction observed in this murine model. Based on the observation that the induction of senescence in human aortic endothelial cells by the inhibition of telomere function results in decreased eNOS activity [69, 70], studies with the atherosclerotic apoE-/- mouse may provide an opportunity to evaluate and understand the interaction of vascular endothelial dysfunction and vascular senescence. We postulated that the renin-angiotensin system plays a dual role, contributing additively with ROS to DNA damage and additively with aging to arterial hypertension.

Oxidative damage to DNA in atherosclerosis also modifies other biochemical and regulatory pathways such as the overexpression of poly(ADP-ribose) polymerase (PARP-1) [71, 72]. PARP-1 is a nuclear enzyme that recognizes and is activated by DNA strand breaks and is thus involved in DNA damage response pathways; simultaneously, PARP-1 may induce cell dysfunction or necrosis by depleting cellular energy pools [73]. Pacher et al. [74] reported that the activation of PARP-1 is associated with hypertension and aging but not with atherosclerosis. In contrast, other investigators [71, 75] have shown that functional alterations in the endothelium of the apoE-/- mouse are dependent on the activation of PARP-1 in endothelial cells. This activation leads to proinflammatory gene expression and thus contributes to the vascular senescence and endothelial dysfunction that are observed in atherosclerosis. Moreover, pharmacological blockade and genetic manipulations have demonstrated that endogenous PARP-1 is required for atherogenesis in the apoE-/- mouse and functions by increasing the expression of adhesion molecules upon endothelial activation, enhancing inflammation, and inducing features of plaque vulnerability. Thus, inhibition of PARP1 may represent a promising therapeutic target in atherosclerosis [73]. Figure 1 illustrates the interplay among ROS, oxidative damage to DNA, the renin-angiotensin system, vascular senescence and atherosclerosis in the apoE-/- mouse.

Atherosclerosis is a chronic disease, and whether it is initiated and/or aggravated by dyslipidemia depends on the contribution of oxidative stress and proinflammatory mediators. Thus, the impact of these contributors on the development of atherosclerotic lesions may reveal new treatment options, independent of serum lipids reduction. Recent studies in apoE-/- mice have demonstrated a reduction in atherosclerosis development using an immunodeficiency strategy [20], low dose of the phytochemical tetrahydrocannabinol [76], antioxidant N-Acetylcysteine [77], PARP inhibition [73] or mononuclear cell therapy [63].

In relation to cell therapy, there is consistent evidence [78] that the atherosclerotic process initiated by endothelial death results in the subsequent replacement of the affected cells by endothelial progenitor cells; in addition, the evidence indicates that the cellular repair of ongoing vascular injury is mediated by progenitor cells. Indeed, treatment with spleen-derived mononuclear cells increases vascular NOS activity and restores endothelium-dependent relaxation in the aortas of apoE-/- mice [79]. Moreover, mononuclear cell therapy in apoE-/- mice results in the homing of endothelial progenitor cells, a decrease in oxidative stress and an upregulation of eNOS protein expression [63], which indicates that cell therapy is a promising tool for the restoration of endothelial function and the prevention of atherosclerosis development, independent of a reduction in serum lipids.

Cardiac phenotypes in apoe-/- mice

Resting HR

The mouse has a high resting heart rate (HR), which is related to its high metabolism. Hemodynamic measurements have shown that the resting HR in the conscious wild-type C57 mouse ranges from 520 to 650 bpm [42, 80, 81]. Similar values have been observed with acute measurements in conscious apoE-/- mice [32, 41, 42, 46, 80, 82, 83]. Interestingly, some studies have shown that HR is preserved even when the BP is high in apoE-/- mice fed either a standard chow diet [46] or a Western-type diet [84]. However, others have evaluated HR by continuous recording over 24 hours and observed a significantly increased mean HR and a total abolition of its circadian cycles in the apoE-/- mouse compared with wild-type control mice [47]. Moreover, apoE-/- mice exhibited decreased HR variability associated with abnormal neurohumoral control of HR and a defective parasympathetic drive to the heart, which were further exacerbated under a high-fat diet regimen [47, 71]. It has also been observed that chronic treatment of apoE-/- mice with a statin reduced their HRs to control levels and restored their circadian variations, despite persistent elevations in PC levels [47]. Although further studies are needed to clarify the mechanisms involved in the abnormal cardiac rhythm in the apoE-/- mouse, there is evidence that the imbalance in NO/•O2- production may play a pivotal role [47, 71].

Cardiac hypertrophy

Cardiac hypertrophy is also an important parameter to evaluate because it can be related to cardiac dysfunction (see the illustration in Figure 2). In studies with apoE-/- mice in our laboratory, we regularly evaluate cardiac hypertrophy, which can be determined by measuring the cardiac weight-to-body weight ratio [42, 81]. In apoE-/- mice fed a normal diet, the cardiac weight has been reported as normal at different ages in several studies [42, 46, 61, 83, 85], but others have observed myocardial hypertrophy in adult [46] and aged [80, 85] animals. However, there are some controversies regarding the possible factors determining cardiac hypertrophy in aged animals. For example, it has been reported that the animals were normotensive and that the cardiac hypertrophy could be related to an elevated blood velocity and wave reflections close to the heart [80]. On the contrary, others reported both hypertension and myocardial hypertrophy in aged apoE-/- mice [46]. In this case, cardiac hypertrophy could be the consequence of an increased afterload, as has been observed in non-atherosclerotic hypertensive mice [42, 81]. Importantly, the cardiac hypertrophy in response to the aortic coarctation-induced pressure overload is higher in apoE-/- mice than in C57 mice [83], leading to the hypothesis that apolipoprotein E may play an important role in modulating cardiac hypertrophy, even in wild-type mice. On a Western-type diet, apoE-/- mice show cardiac hypertrophy that is worsened with aging [86]. Thus, despite certain controversies, studies have provided important evidence that the apoE-/- mouse develops cardiac hypertrophy, which appears to be influenced by several factors, including increased aortic stiffness, arterial hypertension, and a Western-type diet combined with the aging process (see the schematic illustration in Figure 2).

Figure 2
figure 2

A schematic illustration describing cardiac phenotypes and hypothetical factors that influence cardiac dysfunction in the apoE-/- mouse. Differences in line thickness indicate the relative contribution of that factor.

Cardiac function

Technical progress has resulted in the ability to perform arterial and cardiac catheterization; in vivo imaging in the mouse is then utilized, with the aim of understanding cardiac function and dysfunction. Recently, Borst et al. [1] published an extraordinary review about methods employed for the induction and analysis of myocardial infarction in mice. However, murine models of atherosclerosis have coronary arteries that are relatively resistant to the consequences of atherosclerosis and usually do not exhibit myocardial infarction or other cardinal features of human coronary heart disease or reduced lifespan [22, 87]. Hemodynamic analysis in young apoE-/- mice fed a standard chow diet shows normal left ventricular end diastolic pressure and systolic pressure, cardiac output, fractional shortening, and other cardiac functional parameters [87, 88]. Some studies show that feeding young animals an atherogenic diet does not seem to affect cardiac function [25], and these animals rarely show occlusive coronary artery disease and myocardial infarction under this diet [89]. However, others have reported changes in the left ventricular geometry, elevated cardiac outflow velocities, decreased distensibility of the ascending aorta and increased pulse-wave velocity, even when apoE-/- mice are fed a regular chow diet [80, 90, 91]. In aged apoE-/- mice, it has been reported that increased cardiac afterload due to increased aortic stiffening appears to contribute to a reduced cardiac reserve, indicating a reduced cardiac functional reserve in this animal [92].

Interestingly, while recently measuring the diameter of the aorta through contrast angiography, we incidentally observed aortic regurgitation associated with a larger (~3-fold) valvular thickness in elderly apoE-/- mice [61]. Aortic regurgitation was observed in the elderly (hypercholesterolemic and atherosclerotic) apoE-/- mouse but neither in the young (hypercholesterolemic) apoE-/- mouse nor in the elderly C57 control mouse (normocholesterolemic), indicating that this cardiac dysfunction is dependent on the concurrence of hypercholesterolemia and aging [61]. In that study, the authors showed that the aortic valve damage was characterized by diffuse acellularity and myxoid thickening of the spongy layer, as well as valve fibrosis with dense collagen formation in the apoE-/- mouse.

Among the animal models of heart failure, the major limitation of the apoE-/- mouse is the infrequency of coronary plaques and thrombosis, two common complications of human atherosclerosis, as recently reviewed [93]. Cardiac function has also been evaluated by inter-breeding apoE-/- and renin-angiotensinogen transgenic (R+A+) mice; in this animal model, heart failure develops with aging, as indicated by the decreased ejection fraction and increased lung weight in aged mice [94].

Conclusion

Since its creation two decades ago, the apoE-/- mouse, which spontaneously develops hypercholesterolemia and vascular atherosclerotic lesions on a regular chow diet, has greatly contributed to the understanding of the atherosclerosis disease process. Excess production of ROS responding to internal or environmental stress triggers several signaling steps culminating with lipid oxidation, DNA damage, senescence and production of proinflammatory substances. The renin-angiotensin system also appears to play an important role, contributing additively with ROS to changes in vascular function. Long-term recordings of HR and BP show evidence that apoE-/- mice exhibit hypertension and tachycardia and abolition of their circadian cycles, primarily when under the influence of aging and a Western-type diet. Studies also provide evidence of impaired cardiac function in apoE-/- mice, which is related to and/or aggravated by aging and a Western-type diet. Therefore, the studies in apoE-/- mice have contributed new insights into the mechanisms underlying the cardiac and vascular dysfunction that occur in this chronic inflammatory disease.

References

  1. Borst O, Ochmann C, Schönberger T, Jacoby C, Stellos K, Seizer P, Flögel U, Lang F, Gawaz M: Methods employed for induction and analysis of experimental myocardial infarction in mice. Cell Physiol Biochem. 2011, 28: 1-12. 10.1159/000331708.

    Article  CAS  PubMed  Google Scholar 

  2. Plump AS, Smith JD, Hayek T, Aalto-Setälä K, Walsh A, Verstuyft JG, Rubin EM, Breslow JL: Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell. 1992, 71 (2): 343-53. 10.1016/0092-8674(92)90362-G.

    Article  CAS  PubMed  Google Scholar 

  3. Piedrahita JA, Zhang SH, Hagaman JR, Oliver PM, Maeda N: Generation of mice carrying a mutant apolipoprotein E gene inactivated by gene targeting in embryonic stem cells. Proc Natl Acad Sci USA. 1992, 89 (10): 4471-5. 10.1073/pnas.89.10.4471.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Zhang SH, Reddick RL, Burkey B, Maeda N: Diet-induced atherosclerosis in mice heterozygous and homozygous for apolipoprotein E gene disruption. J Clin Invest. 1994, 94 (3): 937-45. 10.1172/JCI117460.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Bourassa PA, Milos PM, Gaynor BJ, Breslow JL, Aiello RJ: Estrogen reduces atherosclerotic lesion development in apolipoprotein E-deficient mice. Proc Natl Acad Sci USA. 1996, 93 (19): 10022-7. 10.1073/pnas.93.19.10022.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  6. Bonthu S, Heistad DD, Chappell DA, Lamping KG, Faraci FM: Atherosclerosis, vascular remodeling, and impairment of endothelium-dependent relaxation in genetically altered hyperlipidemic mice. Arterioscler Thromb Vasc Biol. 1997, 17 (11): 2333-40. 10.1161/01.ATV.17.11.2333.

    Article  CAS  PubMed  Google Scholar 

  7. d'Uscio LV, Barton M, Shaw S, Lüscher TF: Chronic ET(A) receptor blockade prevents endothelial dysfunction of small arteries in apolipoprotein E-deficient mice. Cardiovasc Res. 2002, 53 (2): 487-95. 10.1016/S0008-6363(01)00469-2.

    Article  PubMed  Google Scholar 

  8. Surra JC, Guillén N, Arbonés-Mainar JM, Barranquero C, Navarro MA, Arnal C, Orman I, Segovia JC, Osada J: Sex as a profound modifier of atherosclerotic lesion development in apolipoprotein E-deficient mice with different genetic backgrounds. J Atheroscler Thromb. 2010, 17 (7): 712-21. 10.5551/jat.3541.

    Article  CAS  PubMed  Google Scholar 

  9. Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R: ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb. 1994, 14 (1): 133-40. 10.1161/01.ATV.14.1.133.

    Article  CAS  PubMed  Google Scholar 

  10. Jiang F, Gibson AP, Dusting GJ: Endothelial dysfunction induced by oxidized low-density lipoproteins in isolated mouse aorta: a comparison with apolipoprotein-E deficient mice. Eur J Pharmacol. 2001, 424 (2): 141-9. 10.1016/S0014-2999(01)01140-2.

    Article  CAS  PubMed  Google Scholar 

  11. Ross R: The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature. 1993, 362: 801-9. 10.1038/362801a0.

    Article  CAS  PubMed  Google Scholar 

  12. Hansson GK: Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005, 352: 1685-95. 10.1056/NEJMra043430.

    Article  CAS  PubMed  Google Scholar 

  13. Stoll G, Bendszus M: Inflammation and atherosclerosis: novel insights into plaque formation and destabilization. Stroke. 2006, 37 (7): 1923-32. 10.1161/01.STR.0000226901.34927.10.

    Article  CAS  PubMed  Google Scholar 

  14. Negre-Salvayre A, Coatrieux C, Ingueneau C, Salvayre R: Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br J Pharmacol. 2008, 153 (1): 6-20. 10.1038/sj.bjp.0707395.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Getz GS: Overview of murine atherosclerosis series. Curr Drug Targets. 2007, 8: 1144-49. 10.2174/138945007782403865.

    Article  CAS  PubMed  Google Scholar 

  16. Weber C, Zernecke A, Libby P: The multifaceted contributions of leukocyte subsets to atherosclerosis: lessons from mouse models. Nat Rev Immunol. 2008, 8: 802-15. 10.1038/nri2415.

    Article  CAS  PubMed  Google Scholar 

  17. Steinberg D: Atherogenesis in perspective: hypercholesterolemia and inflammation as partners in crime. Nat Med. 2002, 8: 1211-7. 10.1038/nm1102-1211.

    Article  CAS  PubMed  Google Scholar 

  18. Jawień J: New insights into immunological aspects of atherosclerosis. Pol Arch Med Wewn. 2008, 118 (3): 127-31.

    PubMed  Google Scholar 

  19. Shemesh S, Kamari Y, Shaish A, Olteanu S, Kandel-Kfir M, Almog T, Grosskopf I, Apte RN, Harats D: Interleukin-1 receptor type-1 in non-hematopoietic cells is the target for the pro-atherogenic effects of interleukin-1 in apoE-deficient mice. Atherosclerosis. 2012,

    Google Scholar 

  20. Zhou X, Nicoletti A, Elhage R, Hansson GK: Transfer of CD4(+) T cells aggravates atherosclerosis in immunodeficient apolipoprotein E knockout mice. Circulation. 2000, 102: 2919-22.

    Article  CAS  PubMed  Google Scholar 

  21. Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R: ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb. 1994, 14 (1): 133-40. 10.1161/01.ATV.14.1.133.

    Article  CAS  PubMed  Google Scholar 

  22. Coleman R, Hayek T, Keidar S, Aviram M: A mouse model for human atherosclerosis: long-term histopathological study of lesion development in the aortic arch of apolipoprotein E-deficient (E0) mice. Acta Histochem. 2006, 108 (6): 415-24. 10.1016/j.acthis.2006.07.002.

    Article  CAS  PubMed  Google Scholar 

  23. Meyrelles SS, Peotta VA, Pereira TM, Vasquez EC: Endothelial dysfunction in the apolipoprotein E-deficient mouse: insights into the influence of diet, gender and aging. Lipids Health Dis. 2011, 10: 211-10.1186/1476-511X-10-211.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Crauwels HM, Van Hove CE, Holvoet P, Herman AG, Bult H: Plaque-associated endothelial dysfunction in apolipoprotein E-deficient mice on a regular diet. Effect of human apolipoprotein AI. Cardiovasc Res. 2003, 59 (1): 189-99. 10.1016/S0008-6363(03)00353-5.

    Article  CAS  PubMed  Google Scholar 

  25. Johansson ME, Hägg U, Wikström J, Wickman A, Bergström G, Gan LM: Haemodynamically significant plaque formation and regional endothelial dysfunction in cholesterol-fed ApoE-/- mice. Clin Sci (Lond). 2005, 108 (6): 531-8. 10.1042/CS20040322.

    Article  CAS  Google Scholar 

  26. Deckert V, Lizard G, Duverger N, Athias A, Palleau V, Emmanuel F, Moisant M, Gambert P, Lallemant C, Lagrost L: Impairment of endothelium-dependent arterial relaxation by high-fat feeding in ApoE-deficient mice: toward normalization by human ApoA-I expression. Circulation. 1999, 100 (11): 1230-5.

    Article  CAS  PubMed  Google Scholar 

  27. Yaghoubi M, Oliver-Krasinski J, Cayatte AJ, Cohen RA: Decreased sensitivity to nitric oxide in the aorta of severely hypercholesterolemic apolipoprotein E-deficient mice. J Cardiovasc Pharmacol. 2000, 36 (6): 751-7. 10.1097/00005344-200012000-00010.

    Article  CAS  PubMed  Google Scholar 

  28. d'Uscio LV, Baker TA, Mantilla CB, Smith L, Weiler D, Sieck GC, Katusic ZS: Mechanism of endothelial dysfunction in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2001, 21 (6): 1017-22. 10.1161/01.ATV.21.6.1017.

    Article  PubMed  Google Scholar 

  29. Guns PJ, Van Assche T, Verreth W, Fransen P, Mackness B, Mackness M, Holvoet P, Bult H: Paraoxonase 1 gene transfer lowers vascular oxidative stress and improves vasomotor function in apolipoprotein E-deficient mice with pre-existing atherosclerosis. Br J Pharmacol. 2008, 153 (3): 508-16. 10.1038/sj.bjp.0707585.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Laursen JB, Somers M, Kurz S, McCann L, Warnholtz A, Freeman BA, Tarpey M, Fukai T, Harrison DG: Endothelial regulation of vasomotion in apoE-deficient mice: implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation. 2001, 103 (9): 1282-8.

    Article  CAS  PubMed  Google Scholar 

  31. Barton M, Haudenschild CC, d'Uscio LV, Shaw S, Münter K, Lüscher TF: Endothelin ETA receptor blockade restores NO-mediated endothelial function and inhibits atherosclerosis in apolipoprotein E-deficient mice. Proc Natl Acad Sci USA. 1998, 95 (24): 14367-72. 10.1073/pnas.95.24.14367.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  32. Wassmann S, Czech T, van Eickels M, Fleming I, Böhm M, Nickenig G: Inhibition of diet-induced atherosclerosis and endothelial dysfunction in apolipoprotein E/angiotensin II type 1A receptor double-knockout mice. Circulation. 2004, 110 (19): 3062-7. 10.1161/01.CIR.0000137970.47771.AF.

    Article  CAS  PubMed  Google Scholar 

  33. Weiss D, Kools JJ, Taylor WR: Angiotensin II-induced hypertension accelerates the development of atherosclerosis in apoE-deficient mice. Circulation. 2001, 103 (3): 448-54.

    Article  CAS  PubMed  Google Scholar 

  34. Cola MS, Gava AL, Meyrelles SS, Vasquez EC: Endothelial dysfunction of resistance vessels in female apolipoprotein E-deficient mice. Lipids Health Dis. 2010, 9: 51-10.1186/1476-511X-9-51.

    Article  PubMed Central  PubMed  Google Scholar 

  35. Kitayama J, Faraci FM, Lentz SR, Heistad DD: Cerebral vascular dysfunction during hypercholesterolemia. Stroke. 2007, 38 (7): 2136-41. 10.1161/STROKEAHA.107.481879.

    Article  CAS  PubMed  Google Scholar 

  36. Tabas I, Williams KJ, Boren J: Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation. 2007, 116: 1832-44. 10.1161/CIRCULATIONAHA.106.676890.

    Article  CAS  PubMed  Google Scholar 

  37. Libby P, Ridker PM, Hansson GK: Progress and challenges in translating the biology of atherosclerosis. Nature. 2011, 473: 317-25. 10.1038/nature10146.

    Article  CAS  PubMed  Google Scholar 

  38. Williams KJ, Scalia R, Mazany KD, Rodrigueza WV, Lefer AM: Rapid restoration of normal endothelial functions in genetically hyperlipidemic mice by a synthetic mediator of reverse lipid transport. Arterioscler Thromb Vasc Biol. 2000, 20 (4): 1033-9. 10.1161/01.ATV.20.4.1033.

    Article  CAS  PubMed  Google Scholar 

  39. Meyrelles SS, Chapleau MW: Impaired carotid occlusion reflex in apoE deficient mice and its reversal by gene transfer to carotid sinus. J Hypertens. 2000, 18 (Suppl 4): S202-

    Google Scholar 

  40. Gervais M, Pons S, Nicoletti A, Cosson C, Giudicelli JF, Richer C: Fluvastatin prevents renal dysfunction and vascular NO deficit in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2003, 23 (2): 183-9. 10.1161/01.ATV.0000051404.84665.49.

    Article  CAS  PubMed  Google Scholar 

  41. Arruda RM, Peotta VA, Meyrelles SS, Vasquez EC: Evaluation of vascular function in apolipoprotein E knockout mice with angiotensin-dependent renovascular hypertension. Hypertension. 2005, 46 (4): 932-6. 10.1161/01.HYP.0000182154.61862.52.

    Article  CAS  PubMed  Google Scholar 

  42. Nogueira BV, Peotta VA, Meyrelles SS, Vasquez EC: Evaluation of aortic remodeling in apolipoprotein E-deficient mice and renovascular hypertensive mice. Arch Med Res. 2007, 38 (8): 816-21. 10.1016/j.arcmed.2007.06.005.

    Article  CAS  PubMed  Google Scholar 

  43. Stapleton PA, Goodwill AG, James ME, Frisbee JC: Altered mechanisms of endothelium-dependent dilation in skeletal muscle arterioles with genetic hypercholesterolemia. Am J Physiol Regul Integr Comp Physiol. 2007, 293 (3): R1110-9. 10.1152/ajpregu.00410.2007.

    Article  CAS  PubMed  Google Scholar 

  44. Custodis F, Baumhäkel M, Schlimmer N, List F, Gensch C, Böhm M, Laufs U: Heart rate reduction by ivabradine reduces oxidative stress, improves endothelial function, and prevents atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2008, 117 (18): 2377-87. 10.1161/CIRCULATIONAHA.107.746537.

    Article  CAS  PubMed  Google Scholar 

  45. Tesanovic S, Vinh A, Gaspari TA, Casley D, Widdop RE: Vasoprotective and atheroprotective effects of angiotensin (1-7) in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2010, 30 (8): 1606-13. 10.1161/ATVBAHA.110.204453.

    Article  CAS  PubMed  Google Scholar 

  46. Yang R, Powell-Braxton L, Ogaoawara AK, Dybdal N, Bunting S, Ohneda O, Jin H: Hypertension and endothelial dysfunction in apolipoprotein E knockout mice. Arterioscler Thromb Vasc Biol. 1999, 19 (11): 2762-8. 10.1161/01.ATV.19.11.2762.

    Article  CAS  PubMed  Google Scholar 

  47. Pelat M, Dessy C, Massion P, Desager JP, Feron O, Balligand JL: Rosuvastatin decreases caveolin-1 and improves nitric oxide-dependent heart rate and blood pressure variability in apolipoprotein E-/- mice in vivo. Circulation. 2003, 107 (19): 2480-6. 10.1161/01.CIR.0000065601.83526.3E.

    Article  CAS  PubMed  Google Scholar 

  48. Lu H, Cassis LA, Daugherty A: Atherosclerosis and arterial blood pressure in mice. Curr Drug Targets. 2007, 8 (11): 181-9.

    Article  Google Scholar 

  49. Daugherty A, Manning MW, Cassis LA: Antagonism of AT2 receptors augments angiotensin II-induced abdominal aortic aneurysms and atherosclerosis. Br J Pharmacol. 2001, 134 (4): 865-70. 10.1038/sj.bjp.0704331.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Bruemmer D, Collins AR, Noh G, Wang W, Territo M, Arias-Magallona S, Fishbein MC, Blaschke F, Kintscher U, Graf K, Law RE, Hsueh WA: Angiotensin II-accelerated atherosclerosis and aneurysm formation is attenuated in osteopontin-deficient mice. J Clin Invest. 2003, 112 (9): 1318-31.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  51. Ferrario CM, Smith R, Levy P, Strawn W: The hypertension-lipid connection: insights into the relation between angiotensin II and cholesterol in atherogenesis. Am J Med Sci. 2002, 323 (1): 17-24. 10.1097/00000441-200201000-00004.

    Article  PubMed  Google Scholar 

  52. Daugherty A, Rateri DL, Lu H, Inagami T, Cassis LA: Hypercholesterolemia stimulates angiotensin peptide synthesis and contributes to atherosclerosis through the AT1A receptor. Circulation. 2004, 110 (25): 3849-57. 10.1161/01.CIR.0000150540.54220.C4.

    Article  CAS  PubMed  Google Scholar 

  53. Daugherty A, Manning MW, Cassis LA: Angiotensin II promotes atherosclerotic lesions and aneurysms in apolipoprotein E-deficient mice. J Clin Invest. 2000, 105: 1605-12. 10.1172/JCI7818.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Vinh A, Widdop RE, Drummond GR, Gaspari TA: Chronic angiotensin IV treatment reverses endothelial dysfunction in ApoE-deficient mice. Cardiovasc Res. 2008, 77 (1): 178-87.

    Article  CAS  PubMed  Google Scholar 

  55. Ayabe N, Babaev VR, Tang Y, Tanizawa T, Fogo AB, Linton MF, Ichikawa I, Fazio S, Kon V: Transiently heightened angiotensin II has distinct effects on atherosclerosis and aneurysm formation in hyperlipidemic mice. Atherosclerosis. 2006, 184 (2): 312-21. 10.1016/j.atherosclerosis.2005.05.016.

    Article  CAS  PubMed  Google Scholar 

  56. Nobuhiko A, Suganuma E, Babaev VR, Fogo A, Swift LL, Linton MF, Fazio S, Ichikawa I, Kon V: Angiotensin II amplifies macrophage-driven atherosclerosis. Arterioscler Thromb Vasc Biol. 2004, 24 (11): 2143-8. 10.1161/01.ATV.0000145607.03879.e0.

    Article  CAS  PubMed  Google Scholar 

  57. Cheng XW, Song H, Sasaki T, Hu L, Inoue A, Bando YK, Shi GP, Kuzuya M, Okumura K, Murohara T: Angiotensin type 1 receptor blocker reduces intimal neovascularization and plaque growth in apolipoprotein E-deficient mice. Hypertension. 2011, 57 (5): 981-9. 10.1161/HYPERTENSIONAHA.110.168385.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  58. Zhou Y, Chen R, Catanzaro SE, Hu L, Dansky HM, Catanzaro DF: Differential effects of angiotensin II on atherogenesis at the aortic sinus and descending aorta of apolipoprotein E-deficient mice. Am J Hypertens. 2005, 18 (4 Pt 1): 486-92.

    Article  CAS  PubMed  Google Scholar 

  59. Seo HS, Lombardi DM, Polinsky P, Powell-Braxton L, Bunting S, Schwartz SM, Rosenfeld ME: Peripheral vascular stenosis in apolipoprotein E-deficient mice: potential roles of lipid deposition, medial atrophy, and adventitial inflammation. Arterioscler Thromb Vasc Biol. 1997, 17: 3593-3601. 10.1161/01.ATV.17.12.3593.

    Article  CAS  PubMed  Google Scholar 

  60. Walker M, Campbell BR, Azer K, Tong C, Fang K, Cook JJ, Forrest MJ, Kempadoo K, Wright SD, Saltzman JS, MacIntyre E, Hargreaves R: A novel 3-dimensional micro-ultrasound approach to automated measurement of carotid arterial plaque volume as a biomarker for experimental atherosclerosis. Atherosclerosis. 2009, 204 (1): 55-65. 10.1016/j.atherosclerosis.2008.09.013.

    Article  CAS  PubMed  Google Scholar 

  61. Pereira TM, Nogueira BV, Lima LC, Porto ML, Arruda JA, Vasquez EC, Meyrelles SS: Cardiac and vascular changes in elderly atherosclerotic mice: the influence of gender. Lipids Health Dis. 2010, 9: 87-10.1186/1476-511X-9-87.

    Article  PubMed Central  PubMed  Google Scholar 

  62. Bentzon JF, Pasterkamp G, Falk E: Expansive remodeling is a response of the plaque-related vessel wall in aortic roots of ApoE-deficient mice: an experiment of nature. Arterioscler Thromb Vasc Biol. 2003, 23: 257-62. 10.1161/01.ATV.0000051387.70962.79.

    Article  CAS  PubMed  Google Scholar 

  63. Porto ML, Lima LC, Pereira TM, Nogueira BV, Tonini CL, Campagnaro BP, Meyrelles SS, Vasquez EC: Mononuclear cell therapy attenuates atherosclerosis in apoE KO mice. Lipids Health Dis. 2011, 10: 155-10.1186/1476-511X-10-155.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  64. Pashkow FJ: Oxidative stress and inflammation in heart disease: do antioxidants have a role in treatment and/or prevention?. Int J Inflam. 2011, 2011: 514623-

    Article  PubMed Central  PubMed  Google Scholar 

  65. Minamino T, Komuro I: Vascular cell senescence: contribution to atherosclerosis. Circ Res. 2007, 100 (1): 15-26. 10.1161/01.RES.0000256837.40544.4a.

    Article  CAS  PubMed  Google Scholar 

  66. Uchida K, Itakura K, Kawakishi S, Hiai H, Toyokuni S, Stadtman ER: Characterization of epitopes recognized by 4-hydroxy-2-nonenal specific antibodies. Arch Biochem Biophys. 1995, 324: 241-8. 10.1006/abbi.1995.0036.

    Article  CAS  PubMed  Google Scholar 

  67. Hasty P, Campisi J, Hoeijmakers J, van Steeg H, Vijg J: Aging and genome maintenance: lessons from the mouse?. Science. 2003, 299: 1355-9. 10.1126/science.1079161.

    Article  CAS  PubMed  Google Scholar 

  68. Ota H, Eto M, Ogawa S, Iijima K, Akishita M, Ouchi Y: SIRT1/eNOS axis as a potential target against vascular senescence, dysfunction and atherosclerosis. J Atheroscler Thromb. 2010, 17 (5): 431-5. 10.5551/jat.3525.

    Article  CAS  PubMed  Google Scholar 

  69. Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I: Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002, 105 (13): 1541-4. 10.1161/01.CIR.0000013836.85741.17.

    Article  CAS  PubMed  Google Scholar 

  70. Matsushita H, Chang E, Glassford AJ, Cooke JP, Chiu CP, Tsao PS: eNOS activity is reduced in senescent human endothelial cells: Preservation by hTERT immortalization. Circ Res. 2001, 89 (9): 793-8. 10.1161/hh2101.098443.

    Article  CAS  PubMed  Google Scholar 

  71. Hans CP, Feng Y, Naura AS, Zerfaoui M, Rezk BM, Xia H, Kaye AD, Matrougui K, Lazartigues E, Boulares AH: Protective effects of PARP-1 knockout on dyslipidemia-induced autonomic and vascular dysfunction in ApoE mice: effects on eNOS and oxidative stress. PLoS One. 2009, 4 (10): e7430-10.1371/journal.pone.0007430.

    Article  PubMed Central  PubMed  Google Scholar 

  72. Martinet W, Knaapen MW, De Meyer GR, Herman AG, Kockx MM: Elevated levels of oxidative DNA damage and DNA repair enzymes in human atherosclerotic plaques. Circulation. 2002, 106 (8): 927-32. 10.1161/01.CIR.0000026393.47805.21.

    Article  CAS  PubMed  Google Scholar 

  73. von Lukowicz T, Hassa PO, Lohmann C, Borén J, Braunersreuther V, Mach F, Odermatt B, Gersbach M, Camici GG, Stähli BE, Tanner FC, Hottiger MO, Lüscher TF, Matter CM: PARP1 is required for adhesion molecule expression in atherogenesis. Cardiovasc Res. 2008, 78 (1): 158-66. 10.1093/cvr/cvm110.

    Article  CAS  PubMed  Google Scholar 

  74. Pacher P, Mabley JG, Soriano FG, Liaudet L, Szabó C: Activation of poly(ADP-ribose) polymerase contributes to the endothelial dysfunction associated with hypertension and aging. Int J Mol Med. 2002, 9 (6): 659-64.

    CAS  PubMed  Google Scholar 

  75. Benkö R, Pacher P, Vaslin A, Kollai M, Szabó C: Restoration of the endothelial function in the aortic rings of apolipoprotein E deficient mice by pharmacological inhibition of the nuclear enzyme poly(ADP-ribose) polymerase. Life Sci. 2004, 75 (10): 1255-61. 10.1016/j.lfs.2004.04.007.

    Article  PubMed  Google Scholar 

  76. Steffens S, Veillard NR, Arnaud C, Pelli G, Burger F, Staub C, Karsak M, Zimmer A, Frossard JL, Mach F: Low dose oral cannabinoid therapy reduces progression of atherosclerosis in mice. Nature. 2005, 434: 782-6. 10.1038/nature03389.

    Article  CAS  PubMed  Google Scholar 

  77. Shimada K, Murayama T, Yokode M, Kita T, Uzui H, Ueda T, Lee JD, Kishimoto C: N-acetylcysteine reduces the severity of atherosclerosis in apolipoprotein E-deficient mice by reducing superoxide production. Circ J. 2009, 73 (7): 1337-41. 10.1253/circj.CJ-08-1148.

    Article  CAS  PubMed  Google Scholar 

  78. Xu Q: The impact of progenitor cells in atherosclerosis. Nat Clin Pract Cardiovasc Med. 2006, 3 (2): 94-101. 10.1038/ncpcardio0396.

    Article  CAS  PubMed  Google Scholar 

  79. Wassmann S, Werner N, Czech T, Nickenig G: Improvement of endothelial function by systemic transfusion of vascular progenitor cells. Circ Res. 2006, 99 (8): e74-83. 10.1161/01.RES.0000246095.90247.d4.

    Article  CAS  PubMed  Google Scholar 

  80. Hartley CJ, Reddy AK, Madala S, Martin-McNulty B, Vergona R, Sullivan ME, Halks-Miller M, Taffet GE, Michael LH, Entman ML, Wang YX: Hemodynamic changes in apolipoprotein E-knockout mice. Am J Physiol Heart Circ Physiol. 2000, 279 (5): H2326-34.

    CAS  PubMed  Google Scholar 

  81. Peotta VA, Gava AL, Vasquez EC, Meyrelles SS: Evaluation of baroreflex control of heart rate in renovascular hypertensive mice. Can J Physiol Pharmacol. 2007, 85 (8): 761-6. 10.1139/Y07-067.

    Article  CAS  PubMed  Google Scholar 

  82. Braun A, Trigatti BL, Post MJ, Sato K, Simons M, Edelberg JM, Rosenberg RD, Schrenzel M, Krieger M: Loss of SR-BI expression leads to the early onset of occlusive atherosclerotic coronary artery disease, spontaneous myocardial infarctions, severe cardiac dysfunction, and premature death in apolipoprotein E-deficient mice. Circ Res. 2002, 90 (3): 270-6. 10.1161/hh0302.104462.

    Article  CAS  PubMed  Google Scholar 

  83. Wu JH, Hagaman J, Kim S, Reddick RL, Maeda N: Aortic constriction exacerbates atherosclerosis and induces cardiac dysfunction in mice lacking apolipoprotein E. Arterioscler Thromb Vasc Biol. 2002, 22 (3): 469-75. 10.1161/hq0302.105287.

    Article  CAS  PubMed  Google Scholar 

  84. Kuhlencordt PJ, Gyurko R, Han F, Scherrer-Crosbie M, Aretz TH, Hajjar R, Picard MH, Huang PL: Accelerated atherosclerosis, aortic aneurysm formation, and ischemic heart disease in apolipoprotein E/endothelial nitric oxide synthase double-knockout mice. Circulation. 2001, 104 (4): 448-54. 10.1161/hc2901.091399.

    Article  CAS  PubMed  Google Scholar 

  85. Wang YX: Cardiovascular functional phenotypes and pharmacological responses in apolipoprotein E deficient mice. Neurobiol Aging. 2005, 26 (3): 309-16. 10.1016/j.neurobiolaging.2004.06.018.

    Article  CAS  PubMed  Google Scholar 

  86. Qin YW, Ye P, He JQ, Sheng L, Wang LY, Du J: Simvastatin inhibited cardiac hypertrophy and fibrosis in apolipoprotein E-deficient mice fed a "Western-style diet" by increasing PPAR α and γ expression and reducing TC, MMP-9, and Cat S levels. Acta Pharmacol Sin. 2010, 31 (10): 1350-8. 10.1038/aps.2010.109.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  87. Braun A, Rigotti A, Trigatti BL: Myocardial infarction following atherosclerosis in murine models. Curr Drug Targets. 2008, 9 (3): 217-23. 10.2174/138945008783755566.

    Article  CAS  PubMed  Google Scholar 

  88. French CJ, Zaman AK, Sobel BE: Cardiac fibrosis and diastolic dysfunction after myocardial infarction in apolipoprotein E knockout mice. Coron Artery Dis. 2009, 20 (7): 479-82. 10.1097/MCA.0b013e3283319b6c.

    Article  PubMed  Google Scholar 

  89. Calara F, Silvestre M, Casanada F, Yuan N, Napoli C, Palinski W: Spontaneous plaque rupture and secondary thrombosis in apolipoprotein E-deficient and LDL receptor-deficient mice. J Pathol. 2001, 195 (2): 257-63. 10.1002/path.915.

    Article  CAS  PubMed  Google Scholar 

  90. Onozuka H, Fujii S, Mikami T, Yamada S, Ishimori N, Shimizu T, Furumoto T, Nakai Y, Komuro K, Nishihara K, Okamoto H, Kitabatake A: In vivo echocardiographic detection of cardiovascular lesions in apolipoprotein E-knockout mice using a novel high-frequency high-speed echocardiography technique. Circ J. 2002, 66 (3): 272-6. 10.1253/circj.66.272.

    Article  PubMed  Google Scholar 

  91. Wang YX, Halks-Miller M, Vergona R, Sullivan ME, Fitch R, Mallari C, Martin-McNulty B, da Cunha V, Freay A, Rubanyi GM, Kauser K: Increased aortic stiffness assessed by pulse wave velocity in apolipoprotein E-deficient mice. Am J Physiol Heart Circ Physiol. 2000, 278 (2): H428-34.

    CAS  PubMed  Google Scholar 

  92. Vincelette J, Martin-McNulty B, Vergona R, Sullivan ME, Wang YX: Reduced cardiac functional reserve in apolipoprotein E knockout mice. Transl Res. 2006, 148 (1): 30-6. 10.1016/j.lab.2006.03.007.

    Article  CAS  PubMed  Google Scholar 

  93. Zaragoza C, Gomez-Guerrero C, Martin-Ventura JL, Blanco-Colio L, Lavin B, Mallavia B, Tarin C, Mas S, Ortiz A, Egido J: Animal models of cardiovascular diseases. J Biomed Biotechnol. 2011, 2011: 497841-

    Article  PubMed Central  PubMed  Google Scholar 

  94. Sabharwal R, Weiss RM, Stauss HM, Sigmund CD, Abboud FM, Chapleau MW: ApoE-/-R+A+ mice with hypercholesterolemia, hypertension, and decreased baroreflex sensitivity spontaneously develop heart failure with age. FASEB J. 2007, 21: 753.14-

    Google Scholar 

Download references

Acknowledgements

ECV and SSM are supported by the National Council for the Development of Science and Technology (CNPq, Ref. 302113/2008-8 Grant) and the State Agency for the Development of Science and Technology (FAPES, Ref. FAPES/PRONEX 012/2009).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Elisardo C Vasquez.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

ECV, VAP, ALG, TMCP and SSM equally conceived and prepared this review. All of the authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Vasquez, E.C., Peotta, V.A., Gava, A.L. et al. Cardiac and vascular phenotypes in the apolipoprotein E-deficient mouse. J Biomed Sci 19, 22 (2012). https://doi.org/10.1186/1423-0127-19-22

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1423-0127-19-22

Keywords