Skip main navigation

Do Glucose and Lipids Exert Independent Effects on Atherosclerotic Lesion Initiation or Progression to Advanced Plaques?

Originally publishedhttps://doi.org/10.1161/01.RES.0000259589.34348.74Circulation Research. 2007;100:769–781

    Abstract

    It is becoming increasingly clear that suboptimal blood glucose control results in adverse effects on large blood vessels, thereby accelerating atherosclerosis and cardiovascular disease, manifested as myocardial infarction, stroke, and peripheral vascular disease. Cardiovascular disease is accelerated by both type 1 and type 2 diabetes. In type 1 diabetes, hyperglycemia generally occurs in the absence of elevated blood lipid levels, whereas type 2 diabetes is frequently associated with dyslipidemia. In this review article, we discuss hyperglycemia versus hyperlipidemia as culprits in diabetes-accelerated atherosclerosis and cardiovascular disease, with emphasis on studies in mouse models and isolated vascular cells. Recent studies on LDL receptor–deficient mice that are hyperglycemic, but exhibit no marked dyslipidemia compared with nondiabetic controls, show that diabetes in the absence of diabetes-induced hyperlipidemia is associated with an accelerated formation of atherosclerotic lesions, similar to what is seen in fat-fed nondiabetic mice. These effects of diabetes are masked in severely dyslipidemic mice, suggesting that the effects of glucose and lipids on lesion initiation might be mediated by similar mechanisms. Recent evidence from isolated endothelial cells demonstrates that glucose and lipids can induce endothelial dysfunction through similar intracellular mechanisms. Analogous effects of glucose and lipids are also seen in macrophages. Furthermore, glucose exerts many of its cellular effects through lipid mediators. We propose that diabetes without associated dyslipidemia accelerates atherosclerosis by mechanisms that can also be activated by hyperlipidemia.

    Atherosclerosis is the principal cause of mortality in people with type 1 and type 2 diabetes. Stroke, coronary heart disease, and peripheral arterial disease are more common, and occur at an earlier age, in patients with diabetes than in the general population.1–2 Unfortunately, the cellular and molecular mechanisms whereby diabetes accelerates cardiovascular disease and atherosclerosis are still poorly understood. Nor is it known whether type 1 diabetes and type 2 diabetes accelerate cardiovascular disease through similar mechanisms. Dyslipidemia plays an important role in patients with type 2 diabetes3–6 but does not appear to fully explain the increase in cardiovascular events.7 Type 1 diabetes is associated with more subtle lipid abnormalities,8 and it is unknown to what extent these lipid abnormalities contribute to cardiovascular end points.

    It is becoming increasingly clear that suboptimal blood glucose control may accelerate cardiovascular disease, at least in patients with type 1 diabetes.9 The Diabetes Control and Complications Trial (DCCT) involved young patients with type 1 diabetes assigned to conventional or intensive insulin therapy between 1983 and 1993. At the end of the DCCT, there had been few cardiovascular events, and there was no significant difference between the groups.10 However, the progression of carotid intima/media thickening, as a measure of atherosclerosis, was significantly reduced in patients on the intensive insulin regimen compared with patients on conventional therapy.11 Most of these patients were then followed in the Epidemiology of Diabetes Interventions and Complications study, during which glycemic control in the 2 groups was similar. Interestingly, intensive insulin therapy during the DCCT reduced the risk of cardiovascular events by 57% more than 10 years later.9 This effect was significantly associated with improved glycemic control during the DCCT phase of the study. The mechanisms underlying the long-lasting effects of improved blood glucose control are unclear. It is possible that hyperglycemia leads to long-lasting modifications of the arterial wall, for example, by stimulating advanced glycation endproduct (AGE) modification of proteins.12 It is also possible that increased lesion initiation results in more cardiovascular events many years later. The role of glycemic control in cardiovascular end points associated with type 2 diabetes is less clear, based on intervention studies.13 It is possible that in type 2 diabetes, dyslipidemia is more important as a factor driving cardiovascular disease.

    This review article focuses on lipids versus glucose as cardiovascular risk factors associated with diabetes and discusses findings indicating that glucose may exert primarily “lipid-like” effects in vascular cells.

    Type 1 Diabetes Can Accelerate Lesion Initiation Through Mechanisms Independent of Diabetes-Induced Hypercholesterolemia

    Type 1 diabetes accelerates lesion initiation in several animal models and in humans. In some mouse and swine models, type 1 diabetes increases formation of lesions without associated increases in plasma lipid levels.14–19 However, in most mouse models, the extent of atherosclerosis in diabetic mice increases in association with a marked increase in plasma lipoproteins.19 Recently, we developed a mouse model that allows dissection of the relative contributions of diabetes versus diabetes-induced dyslipidemia. This model is a LDL receptor (LDLR)-deficient mouse (C57BL/6 background) that expresses a lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) transgene under control of the insulin promoter.17 The GP protein is seen as “self” and does not result in pathologies in the mouse. Instead, the significance of the expressed GP transgene is that diabetes can be induced at will by LCMV injection, which causes a T cell–mediated destruction of the GP-expressing β-cells, similar to the autoimmune process leading to type 1 diabetes in humans. Diabetic LDLR−/−;GP mice fed a semipurified low-fat diet do not develop marked diabetes-induced dyslipidemia but show accelerated lesion initiation compared with nondiabetic controls. Formation of these lesions is normalized by intensive insulin therapy following development of overt diabetes, showing that the effects on lesion initiation are caused by diabetes—most likely hyperglycemia in the setting of the basal hyperlipidemia in LDLR−/− mice.17 Cross-sections of the brachiocephalic artery showed no lesions in nondiabetic mice, whereas fatty streak lesions were present in diabetic mice, as illustrated in Figure 1 (left). Lesions in the diabetic mice consisted primarily of macrophages.17 Further evidence in support of the hypothesis that hyperglycemia can stimulate lesion initiation comes from a recent study by Goldberg and colleagues, in which human aldose reductase transgenic mice20–21 were crossed with LDLR-deficient mice.18 Aldose reductase converts glucose to sorbitol, which is then further converted to fructose in the polyol pathway, implicated in many of the complications of diabetes.18 Interestingly, diabetic human aldose reductase transgenic LDLR-deficient mice showed larger lesions compared with diabetic controls, but aldose reductase has no effect in nondiabetic mice.18 The mechanism or cell type responsible for this effect is not yet known, but aortas from diabetic mice expressing human aldose reductase had increased mRNA levels of 2 markers of reactive oxygen species: glucose 6 phosphate dehydrogenase and glutathione peroxidase.18 The selective effect of the aldose reductase transgene in diabetic mice argues for a role of glucose conversion through the polyol pathway in diabetes-accelerated lesion initiation. However, in addition to acting on glucose, aldose reductase can also act on lipids.22 Thus, further studies are needed to definitively demonstrate that the accelerating effect of diabetes on lesion initiation is attributable to hyperglycemia.

    Figure 1. Diabetes accelerates lesion initiation and progression. Schematic illustration of the effects of diabetes on initiation of lesions of atherosclerosis (left) and progression of more advanced lesions (right). Results from both mice and human subjects indicate that diabetes accelerates monocyte and macrophage (light blue) accumulation in early fatty streak lesions, most likely by enhancing endothelial (dark blue) cell adhesion molecule expression. The increased accumulation of hyaluronic acid (turquoise) in the medial layer (brown) of the artery might also contribute to diabetes-accelerated lesion initiation. In more advanced lesions, diabetes stimulates necrotic core formation (indicated by fragmented macrophages) and intraplaque hemorrhage (red cells surrounded by macrophages) through mechanisms that remain to be identified.

    There is also evidence from human postmortem studies that type 1 diabetes accelerates lesion initiation, defined as increased fatty streaks or intima/media thickness, in the absence of diabetes-induced dyslipidemia in youths and children.23–25

    Together, these findings show that type 1 diabetes can accelerate lesion initiation in humans and animal models without an associated increase in blood lipid levels. Hyperglycemia may play a role in accelerating lesion initiation, at least within the normal to high plasma cholesterol levels seen in humans. However, animal studies have clearly shown that hyperglycemia alone is insufficient for lesion initiation when total cholesterol levels are very low (<3 mmol/L; 100 mg/dL).14,16–17 Thus, diabetes-induced lesion initiation requires low basal levels of circulating lipids.

    Severe Dyslipidemia Masks the Effects of Diabetes on Lesion Initiation

    In diabetic animals with severe dyslipidemia, lesion initiation is primarily driven by hyperlipidemia, which appears to overshadow the atherogenic effect of other risk factors, such as hyperglycemia or AGEs.17,26 Thus, when plasma cholesterol levels are dramatically elevated (more than ≈16 mmol/L; 600 mg/dL), the effects of diabetes on lesion initiation are masked. We hypothesize that high lipid levels mask the effects of diabetes because glucose and lipids use similar mechanisms to induce lesion formation. Several Western-type diet–fed mouse models show no additional effect of diabetes beyond that of hyperlipidemia.19 In the work of Reaven et al, LDLR-deficient mice were fed a high-fat diet for 6 months.26 Although streptozotocin-diabetic LDLR−/− mice were hyperglycemic and showed higher levels of AGEs, they had similar plasma cholesterol levels as controls (both ≈1000 mg/dL) and did not demonstrate increased aortic lesion area. Furthermore, in the LDLR−/−;GP model, diabetes accelerates lesion initiation when the mice are fed a low-fat diet, but when fed a high-fat diet resulting in blood cholesterol levels greater than 1000 mg/dL, lesion initiation is driven by plasma lipid levels.17

    Together, these studies indicate that severe hyperlipidemia overrides the effects of diabetes on lesion initiation. However, cholesterol levels greater than 600 mg/dL are rarely seen in humans, even in individuals with familial hypercholesterolemia, and although these findings are of scientific interest, their clinical significance is unclear. Importantly, lesion initiation does not necessarily result in clinical symptoms, and it has been suggested that macrophage-rich fatty streak lesions regress unless a fibrous cap of smooth muscle exists to encapsulate the macrophages.27 Many animal studies are restricted to these early lesions.

    Cellular Mechanisms Involved in Diabetes-Accelerated Lesion Initiation

    Monocyte–Endothelial Adhesion

    It is generally believed that infiltration of monocytes into the subendothelial space, and subsequent accumulation of lipid-loaded macrophages, causes lesion initiation. In young human subjects, accumulation of lipid-loaded macrophages is often seen in areas with intimal smooth muscle–rich masses.27–28 In many small animal models, on the other hand, macrophage accumulation occurs in areas without preexisting intimal thickening. Recruitment of monocytes is regulated by endothelial adhesion molecules and their corresponding monocyte ligands. The most studied endothelial adhesion molecules include E-selectin, P-selectin, vascular cell adhesion molecule-1 (VCAM-1), and intracellular adhesion molecule-1, but there are many more.29

    Studies in animals and humans indicate that diabetes causes an increased endothelial expression of adhesion molecules. Thus, diabetic rabbits showed more endothelial VCAM-1 compared with nondiabetic rabbits.30 Hyperglycemia is likely to exert an acute stimulatory effect on endothelial adhesion molecule expression in vivo, because infusion of high (25 mmol/L) glucose leads to upregulation of endothelial P-selectin and intracellular adhesion molecule-1 immunoreactivity in rat mesenteric venules, accompanied by increased leukocyte adhesion.31 A study of a small group of human subjects with type 1 or type 2 diabetes showed increased endothelial VCAM-1 immunoreactivity compared with nondiabetic controls.32 Interestingly, RAGE (receptor for AGEs) functions as an endothelial adhesion receptor by binding leukocyte β2 integrins.33 Expression of RAGE is increased in endothelial cells from patients with type 2 diabetes.34 Induction of RAGE might therefore serve as an additional pathway to increase monocyte–endothelial adhesion in diabetes.

    In addition, monocytes from humans with poorly controlled type 1 or 2 diabetes exhibit increased adherence to endothelial monolayers,35 suggesting that a combination of increased endothelial adhesion molecule expression and increased ability of monocytes to adhere to these adhesion molecules contribute to the increased lesion initiation associated with diabetes.

    Subendothelial Macrophage Accumulation and Activation

    Endothelial adhesion of monocytes is required, but is not sufficient, for formation of macrophage-rich fatty streaks.36 Macrophage accumulation is increased in lesions from patients with type 1 and type 2 diabetes.37 A similarly increased macrophage accumulation has been observed in LDLR−/−;GP mice with and without diabetes-induced dyslipidemia.17,38 The effects of diabetes on lesion initiation is shown schematically in Figure 1 (left).

    There is strong evidence that a mechanism whereby hyperlipidemia induces lesion initiation is through retention of lipoproteins by proteoglycans within the artery wall.39–40 Although diabetes does not consistently result in increased proteoglycan levels in the arterial wall, hyaluronic acid is increased in the media of arteries from patients with type 1 diabetes compared with controls.41 The LDLR−/−;GP mouse model shows similar increases in medial glycosaminoglycan accumulation in diabetic mice.17 This increased glycosaminoglycan staining is likely attributable, at least in part, to accumulation of hyaluronic acid (our unpublished observations, 2004). Hyaluronic acid (a glycosaminoglycan) binds to the proteoglycan versican,42 which in turn can trap LDL.43 This is interesting because smooth muscle cell (SMC)-targeted overexpression of one of the hyaluronic acid–synthesizing enzymes (hyaluronic acid synthetase 2) causes lesion initiation in nondiabetic mice44 and thus mimics the effects of diabetes. Furthermore, hyaluronic acid can contribute to monocyte binding through interaction with the monocyte hyaluronic acid receptor CD44.45–46 Accordingly, mice deficient in CD44 show reduced lesion initiation and macrophage recruitment.47 Therefore, hyaluronic acid may be a player in type 1 diabetes–stimulated lesion initiation, as illustrated in Figure 1.

    The Table summarizes the time course of lesion initiation and progression in the LDLR-deficient mouse fed low-fat and high-fat diets. As discussed above, accumulation of medial glycosaminoglycans and intimal macrophages are the first events in lesion initiation in this model, both of which are stimulated by diabetes without accompanied diabetes-induced dyslipidemia in low-fat fed mice. However, it should be noted that both of these processes are stimulated by a high-fat diet also (Table). Thus, the effects of diabetes appear to mimic those of hyperlipidemia in this model.

    Table 1. Time Course of Changes in Brachiocephalic Artery Plaque Morphology in LDLR−/− Mice Fed Low- and High-Fat Semipurified Diets

    Characteristics 12 Weeks 16 Weeks 30 Weeks
    Unless otherwise stated, results are shown as frequencies for lesion characteristics (mean±SEM percentage of brachiocephalic artery cross-sections scored positive for a given feature). The results are partly unpublished (F. Johansson, K.E. Bornfeldt, 2006) and partly summarized from Renard et al,17 Lamharzi et al,38 and MacDougall et al.76
    *Processes stimulated by diabetes.
    High-fat diet (1.25% cholesterol, 40% fat)
        Cross-sectional area (×1000 μm2) 18±5* 146±35 171±25
        Mac-2–positive area (×1000 μm2) 15±4* 16±2 14±3
        Medial glycosaminoglycans 75±11 99±1 100±0
        Intimal collagen 44±17 99±1 100±0
        Necrotic core 24±16 77±7 100±0
        Lateral macrophages 8±4 66±11 70±26
        Intraplaque hemorrhage 0±0* 1±1 0±0
        Cholesterol clefts 5±5 46±14 70±31
        Intimal calcification 7±7 10±6 64±45
    Low-fat diet (0% cholesterol, 11% fat)
        Cross-sectional area (×1000 μm2) 1±1* ND 109±25
        Mac-2–positive area (×1000 μm2) 1±1* ND 9±2
        Medial glycosaminoglycans 26±8* ND 99±1
        Intimal collagen 0±0 ND 97±3
        Necrotic core 0±0 ND 74±11
        Lateral macrophages 0±0 ND 77±11
        Intraplaque hemorrhage 0±0 ND 1±1
        Cholesterol clefts 0±0 ND 64±14
        Intimal calcification 0±0 ND 30±10

    Does Diabetes Directly Affect Lesion Progression?

    Several steps characterize the conversion of the fatty streak lesion to an advanced, clinically significant lesion. These steps include the encapsulation of the macrophage-rich mass by SMCs to form a fibrous cap, and the death of macrophages to form one or several necrotic cores. Advanced plaques also frequently show signs of calcification and intraplaque hemorrhage. Rupture of thin-cap fibroatheromas and subsequent thrombosis cause most of the clinical events in humans.48 It is becoming increasingly clear that progression of the different features of advanced plaques are driven by distinct factors and mechanisms. Furthermore, increased lesion size shows no clear correlation with morphological features (Table). We discuss regulation of progression of a few features often observed in clinically relevant lesions and how these may be regulated by diabetes. However, very little is known about the effects of diabetes on advanced plaques. No animal or human studies have been reported in which direct effects of diabetes on advanced lesions were investigated without confounding effects on lesion initiation. It is therefore not known whether diabetes accelerates cardiovascular events merely by accelerating lesion initiation, or whether it has direct effects on the processes governing progression to advanced plaques.

    Progression of Fibrous Cap and Necrotic Cores

    Formation of a fibrous cap with proliferating SMCs is accelerated in diabetic swine, perhaps as a result of accelerated lesion initiation.49 Diabetic BALB/c mice show increased lesion initiation, which is also accompanied by SMC accumulation.14 Although formation of a fibrous cap plays an important role in early lesion progression, it is generally believed that thinning of this cap causes plaque rupture and clinical symptoms.48 It has recently been shown that forced SMC apoptosis in apolipoprotein E–null (apoE−/−) mice results in thinner fibrous caps and larger necrotic cores.50 It is not known whether diabetes specifically affects cap thinning in advanced plaques. On the contrary, apoptosis has been found to be reduced in SMCs derived from patients with diabetes51 and in SMCs exposed to elevated glucose.52–53

    It is clearer that diabetes can result in increased necrotic core formation (Figure 1, right). In postmortem studies, atherosclerotic lesions from patients with type 1 or type 2 diabetes were found to have larger necrotic cores than lesions from nondiabetic subjects.37,54 Necrotic cores form as macrophages undergo apoptosis or necrosis, possibly in combination with a decreased phagocytic capacity of surviving macrophages, resulting in accumulation of cell debris and free lipid. Accumulation of free unesterified cholesterol has been proposed to mediate macrophage death and thereby necrotic core formation.55–56 Diabetes has been associated with increased expression of receptors associated with uptake of modified LDL, such as CD36, scavenger receptor A, and lectin-like oxidized LDL receptor (LOX-1); and reduced levels of the reverse cholesterol transporters ATP-binding cassette A1 (ABCA1) and ABCG1, all of which could accelerate accumulation of intracellular cholesterol and potentially necrotic core formation.57–61 However, further studies are needed to investigate whether the ability of macrophages in advanced atherosclerotic plaques to esterify free cholesterol to the less harmful cholesteryl ester is impaired, and whether this mechanism is affected by diabetes. It should be noted that macrophage apoptosis might affect early and advanced lesions differentially. Thus, reduced macrophage apoptosis results in increased formation of lesions in LDLR−/− mice.62 More relevant to type 2 diabetes are findings showing that macrophages lacking the insulin receptor (thereby being insulin resistant) are more susceptible to modified LDL–induced apoptosis and increased necrotic core formation in LDLR-deficient mice.63 This topic will be covered in detail in a forthcoming article in this thematic review series.

    Progression of Intraplaque Hemorrhage, Plaque Rupture, and Arterial Occlusion

    Cardiovascular events, most likely caused by plaque rupture, occur at an earlier age, and with an increased frequency, in patients with diabetes. Rupture of thin cap fibroatheromas appear to account for a majority of thrombotic events in patients with diabetes, as it does in patients without diabetes.37 In the mouse, rupture or fissuring of advanced plaques in the brachiocephalic artery and aorta is likely to result in intraplaque hemorrhage.64 Diabetes results in increased intraplaque hemorrhage, both in humans65 and fat-fed LDLR-deficient mice17 (Figure 1, right). In addition to disruption of the fibrous cap or the macrophage-rich shoulder regions, intraplaque erythrocytes can originate from leaking vasa vasorum. In humans, intraplaque hemorrhage is often seen deep in the lesion and appears to result primarily from disrupted microvessels.66 Mice also develop increased networks of microvessels in response to atherosclerosis, at least in the aorta.67 In the smaller brachiocephalic artery, intraplaque hemorrhage is often seen in the plaque shoulder regions and most likely originates from the lumen as result of plaque disruption. It has been proposed that intraplaque hemorrhage can contribute to necrotic core formation by providing a large amount of cholesterol derived from erythrocyte cell membranes.66

    The mechanism whereby diabetes promotes intraplaque hemorrhage is unknown. However, it is interesting to speculate that diabetes might increase protease activity, which in turn would destabilize the fibrous cap or extracellular matrix surrounding the macrophage-rich shoulder regions. Recent studies in apoE−/− mice demonstrate that macrophage-targeted expression of matrix metalloproteinase 9 (MMP-9), or an active form of MMP-9, causes intraplaque hemorrhage.64,68 A recent study identified genes differentially expressed in stable and unstable areas of human advanced lesions. Several proteases were upregulated in unstable areas compared with stable areas from the same lesion, including MMP-9.69 Aortas from diabetic rats70 and internal mammary artery from patients with type 2 diabetes71 exhibit increased MMP-9 activity. Monocytes from patients with type 1 diabetes also show higher expression of MMP-9 than those of control subjects.72 Furthermore, advanced atherosclerotic plaques from patients with diabetes show increased MMP-9 expression.73 It is possible that hyperglycemia is directly responsible for the increased MMP-9 activity, as elevated glucose can stimulate MMP-9 expression in human macrophages and endothelial cells.74 Together, these studies suggest that the increased intraplaque hemorrhage associated with diabetes could be attributable to an increased activity of proteases, such as MMP-9.

    Whereas many of the features of lesion initiation and progression discussed above can be addressed using mouse models, currently there are no ideal mouse models available for studies of arterial occlusion caused by thrombosis. Thus, whereas advanced atherosclerotic plaques in LDLR-deficient and apoE-deficient mice show many of the signs of advanced human lesions, and even undergo plaque rupture,64 thrombotic occlusion and cardiovascular events are extremely rare in the mouse. Clearly, better animal models are required for mechanistic studies addressing the effects of diabetes on atherothrombosis.

    Progression of Intimal Calcification

    Contrary to calcification of the medial layer,75 calcification of advanced lesions does not appear to be stimulated by diabetes in humans.37 Based on results from LDLR−/− mice, intimal calcification occurs late in lesion progression (Table).76 Thus, there is no evidence that intimal calcification is directly stimulated by diabetes in lesions of a similar stage as those of nondiabetic controls.

    Genetic Factors Regulating Effects of Glucose and Lipids on Lesion Initiation and Progression to Advanced Plaques

    It is clear from epidemiological and family studies that cardiovascular disease associated with type 2 diabetes has a genetic basis.77–78 However, the identification of specific genetic factors controlling cardiovascular disease in diabetes has been a daunting task. This is because both atherosclerosis and type 2 diabetes are multigenic disorders with contributions from environmental factors. In addition, each disorder proceeds through stages, the genetic components of which may vary in importance during disease evolution. Finally, the regulation of lipid and glucose homeostases are intertwined, making it difficult to separate influences of lipid and glucose metabolism on lesion initiation and progression. The discovery of specific genes involved in cardiovascular disease associated with diabetes is being performed in humans and mice using candidate gene and whole genome scanning approaches.

    Candidate Genes

    Association studies have been performed to examine relationships between gene polymorphisms and incidence or severity of sub-clinical and advanced atherosclerosis among individuals with and without insulin resistance or type 2 diabetes. Lipid metabolism genes already known to predispose to atherosclerosis, and glucose metabolism genes known to predispose to insulin resistance and diabetes, have provided the initial list of candidate genes. Increased prevalence and severity of cardiovascular diseases were seen among type 2 diabetic individuals with specific alleles of lipoprotein lipase,79 cholesteryl ester transfer protein,80–81 apoA-IV,82 and peroxisome proliferator-activated receptor (PPAR)-α,83 with protection seen for an allele of PPAR-γ2.84

    Interestingly, few genes directly related to glucose metabolism have been connected to cardiovascular disease associated with diabetes. In one case, a common polymorphism controlling the basal and insulin stimulated expression of phosphoenolpyruvate carboxykinase was associated with carotid intimal media thickness in a small study of a Canadian Oji-Cree population.85 In another case, single nucleotide polymorphisms identified for tyrosine phosphatase-1B, which acts as a negative regulator of insulin and leptin signaling,86 associated with scores for coronary calcified plaques among a group of type 2 diabetic subjects.87

    Glycoxidation and lipid oxidation are thought to contribute to atherosclerosis and, therefore, it may be expected that alleles for genes that protect cells from oxidation damage may retard cardiovascular disease associated with diabetes. Common variants in the paraoxonase 1 gene88 and the endothelial nitric oxide synthase gene89 were shown to modulate risk of coronary heart disease among subjects with type 2 diabetes. The important contribution to atherosclerosis of RAGE has been shown in several studies of diabetic and nondiabetic mice.90 Functional polymorphisms have been identified in the promoter of the human gene coding for RAGE, and the −374-AA genotype has been associated with protection against several diabetic complications.91–92 Studies of aldose reductase genotype variations93 in association with intima media thickness would be worthwhile, given the recent observation that elevations of aldose reductase can promote atherosclerosis in diabetic mice.18

    Mouse models have also contributed to our knowledge about genetic factors controlling accelerated atherosclerosis as a function of plasma glucose levels.19 For instance, simply deleting the LDLR gene ensures responsiveness to diabetes with respect to accelerating atherosclerosis17–18 as compared with lack of accelerated atherosclerosis in wild-type C57BL/6 mice.14 Deficiency in lipocalin-type prostaglandin D2 synthase results in increased insulin resistance and formation of accelerated early atherosclerosis.94 Furthermore, the combination of a human apoB transgene and lipoprotein lipase deficiency with streptozotocin-induced diabetes results in hyperlipidemia and more atherosclerosis.95 These studies point to several potential candidate genes involved in diabetes-accelerated atherosclerosis.

    An important consideration in the mouse is the role of background strain. In most cases, diabetes-accelerated atherosclerosis has been studied in genetically engineered mutants of C57BL/6. Although C57BL/6 mice are susceptible to atherosclerosis, as induced by special diets and gene mutations, compared with most strains,96 accompanying increases in plasma lipids characteristic of this strain may obscure subsequent insults to the vasculature resulting from hyperglycemia. It may be more useful to study mice more resistant to atherosclerosis, such as BALB/c, DBA/2, and NOD. For instance, diabetic BALB/c, but not C57BL/6, mice show increased lesion formation compared with their nondiabetic littermates.14 Furthermore, C57BL/6 mice carrying obesity/diabetes mutations fail to exhibit increased lesion formation as compared with wild-type controls.97 Thus, it is crucial that studies of diabetic complications are conducted in multiple genetic backgrounds.

    Overall, only a few genetic studies in humans implicate specific lipid and glucose homeostasis genes in cardiovascular disease associated with type 2 diabetes. Such studies are limited by sample size and the strength of gene effects on vascular disease as seen in the diabetic state. However, the combination of studies demonstrates that specific genetic factors are important in determining risk for cardiovascular disease associated with type 2 diabetes. To date, there are a paucity of studies of the association between glucose and lipid homeostasis genes and cardiovascular disease associated with type 1 diabetes.

    Genome Scanning

    The work outlined above argues that diabetes-accelerated atherosclerosis is a multigenic disorder influenced by distinct contributions from many genes. Genome-scanning approaches may allow the simultaneous discovery of multiple loci responsible for atherosclerosis in human subjects or animals with diabetes.

    Although many genome-scanning studies in humans have been applied to glucose homeostasis, obesity, insulin resistance, and diabetes, few have included quantitative traits associated with cardiovascular disease. The calcium-activated neutral protease 10 gene located on chromosome 2p was among the first genes linked to type 2 diabetes susceptibility using genome scanning.98 In this population of Mexican Americans, association of variation in calcium-activated neutral protease 10 was predictive for carotid intima/media thickness and measures of insulin resistance. In a recent study of families, which included type 2 diabetic patients, the incidence and severity of calcified plaques (coronary and/or carotid) was linked to multiple chromosomes.99 Using a subset of these families enriched for diabetic patients, the strongest evidence for linkage was with genetic markers on chromosome 3p. This linkage was not independent from traits of the metabolic syndrome (increased blood glucose, body weight, and dyslipidemia). Thus, genome scanning is identifying chromosomes harboring genes predisposing to atherosclerosis and diabetes. These studies support the concept that common genetic factors may predispose to both type 2 diabetes and atherosclerosis.100–101

    Genetic linkage studies in mice are providing a more rapid analysis of phenotype to genotype relationships as well as identifying traits that are colinked. Several studies have addressed whether distinct patterns of glucose versus lipid metabolism contribute to atherosclerosis. In a cross between PERA and C57BL/6 mice lacking the LDLR, quantitative trait loci for plasma lipids, body weight, insulin, and aortic root atherosclerosis were identified.102 The chromosome 4 quantitative trait loci contributed to variations in lipids and body weight with significant genotype effects on extent of atherosclerosis, suggesting that a common genetic mediator exists for these combined traits.

    In contrast to the work above, there are 2 genome-scanning studies in mice in which traits for plasma glucose concentration and atherosclerosis lesion size are not correlated. A quantitative trait locus analysis was conducted in an F2 cross between C57BL/6/J and C57BLKS/J mice both carrying the deficiency of leptin receptor (db/db) to test the relationship between atherosclerosis and type 2–like diabetes. Separate genetic factors were identified for atherosclerosis (chromosome 12) and elevated glucose levels (chromosomes 8 and 17), supporting the idea that distinct genetic factors control each of these traits.103 The separation of glucose and atherosclerosis was also seen in a study of 2 congenic strains of C57BL/6 carrying loci derived from CAST/Ei (CAST).104 Two regions of chromosome 2 alleles from CAST mice were introgressed into the C57BL/6 background and subsequently bred to LDLR−/− mice to create hyperlipidemic backgrounds for the CAST alleles. Mice fed chow and Western diets were studied for measures of glucose and lipid homeostasis and atherosclerosis. The MOB5 congenic was insulin resistant and showed atherosclerosis comparable to LDLR−/− mice. However, plasma glucose levels were lowest for this strain, illustrating that elevated plasma glucose concentrations are not sufficient for promoting atherosclerosis in fat-fed mice with plasma cholesterol levels of ≈1000 mg/dL. In contrast, although the MOB6 congenic showed comparable glucose levels to the LDLR−/− strain, atherosclerosis was 2-fold reduced in this strain.104 Plasma lipids and lipoprotein profiles were nearly identical between MOB6 and MOB5 strains. This study illustrates that individual traits associated with the metabolic syndrome can be separated and that insulin resistance was more important for promoting atherosclerotic lesions than elevated plasma glucose levels.

    Overall, diabetes or insulin resistance may amplify the risk associated with genetic susceptibility to atherosclerosis already present in individuals. It is still not clear whether diabetes susceptibility genes provide additional or unique genetic elements affecting atherosclerosis and cardiovascular disease. It remains a major challenge to dissociate such genes involved in lipid and glucose homeostasis.

    Do Glucose and Lipids Exert Independent Effects in Cultured Endothelial Cells or Macrophages?

    If elevated glucose indeed exerts biological effects that are distinct from those of elevated lipids, then one would expect to see different biological responses, or at least intracellular signaling events, to elevated glucose and lipids in vascular cells involved in atherosclerosis, such as endothelial cells and macrophages. This, however, is generally not the case. In many instances, the effects of glucose can be mimicked by long-chain fatty acids common in circulation (such as palmitate, oleate, and linoleate), or by oxidized phospholipids, which are present in modified LDL and are believed to play an important role in atherosclerosis.

    In macrovascular endothelial cells, elevated levels of glucose have been shown to induce VCAM-1 expression, potentially explaining the effects of diabetes on macrophage accumulation in lesions of atherosclerosis.105 However, a stimulatory effect of glucose is not always seen, and experiments by other groups have not found increased VCAM-1 expression in human endothelial cells exposed to elevated glucose.106–107 These discrepancies may be attributable to differences in sources of cells, incubation time, and concentration of glucose; differences in posttranslational modification of proteins and lipids by glucose; presence or absence of insulin; or other factors related to the experimental design. Interestingly, serum samples from patients with diabetes did induce increased VCAM-1 expression under conditions in which glucose alone had no effect,106 suggesting that hyperglycemia alone might not explain the effects of diabetes on VCAM-1 expression. These results are consistent with in vivo studies, in which hyperglycemic mice do not develop early lesions unless plasma lipid levels are above a certain threshold. AGE interaction with RAGE has also been shown to induce VCAM-1 in human endothelial cells,107 and injection of AGEs can induce VCAM-1 expression in rabbits in vivo.108 However, other studies have not observed VCAM-1 expression in AGE-stimulated endothelial cells,109 potentially because of the presence of different RAGE splice variants.110 Similarly, palmitate alone has been shown to induce VCAM-1 expression in macrovascular endothelial cells by some111 but not by others.112 High glucose has also been shown to increase monocyte adhesion to endothelial cells though a mechanism that involves increased interleukin (IL)-8 production.113–114 This effect of glucose is mimicked by minimally modified LDL or by specific oxidized phospholipids.115–117

    In human macrophages, elevated glucose has been shown to increase expression of the scavenger receptor and fatty acid transporter CD36,118 an effect that is mimicked by oleate and linoleate.119–120 However, a similar effect of glucose on CD36 has not been found in peritoneal macrophages from mice.121 It is possible that the different responsiveness of human and mouse macrophages to glucose could be attributable to the lower expression of aldose reductase in mouse macrophages, because increasing expression of aldose reductase in mouse macrophages results in increased CD36 expression.18 Other receptors involved in the uptake of modified LDL have also been demonstrated to be induced by glucose.57–58 In fact, glucose alone appears able to promote cholesterol accumulation in a macrophage cell line.122 Accordingly, monocytes from people with diabetes express elevated levels of CD36 and increased uptake in vitro of modified LDL.35,59 In addition, elevated glucose stimulates cellular lipid loading by reducing levels of ABCG1, which is involved in cholesterol efflux from macrophages.61 Oleate and linoleate result in a similar reduction of ABCG1 levels in macrophages.123

    Both type 1and type 2 diabetes are associated with augmented inflammation, as indicated by increased secretion of proinflammatory cytokines from monocytes from these patients.124–125 Fatty acids and glucose can stimulate secretion of cytokines from macrophages. Elevated glucose levels induce secretion of IL-12 in mouse peritoneal macrophages, as well as elevate mRNA levels for IL-1β, IL-6, IL-12, and tumor necrosis factor-α.126 Saturated fatty acids, such as palmitate, induce tumor necrosis factor-α in a macrophage cell line.127 Together, these results suggest that the effects of elevated glucose levels in isolated endothelial cells and macrophages are often mimicked by lipids.

    Elevated Glucose and Lipids Can Induce Similar Intracellular Signals in Endothelial Cells

    One intracellular mechanism whereby glucose can mediate biological effects has been identified by Brownlee and colleagues. This group has shown that many of the effects of elevated glucose on bovine aortic endothelial cells are attributable to increased mitochondrial production of superoxide and subsequent inhibition of the glycolytic enzyme glyceraldehyde-3 phosphate dehydrogenase (GAPDH).128–129 Inhibition of GAPDH activity by elevated glucose leads to accumulation of upstream glycolytic intermediates, such as glyceraldehyde 3-phosphate, which in turn can result in increased activity of the aldose reductase-dependent sorbitol pathway, increased DAG formation and protein kinase C activation, increased flux through the hexosamine pathway, and increased levels of intracellular AGEs.128 Interestingly, the same group has recently shown that elevated levels of oleate mimic many of these effects of elevated glucose in bovine aortic endothelial cells.130 These “glucose-like” effects of fatty acids are shown schematically in Figure 2A. It is not yet known whether this phenomenon can be generalized to different cell types or species. In addition to fatty acids, minimally oxidized LDL and oxidized phospholipids also appear to mediate their effects in endothelial cells through increased superoxide generation.116

    Figure 2. Glucose and fatty acids stimulate many of the same intracellular pathways in vascular cells. A, “Glucose-like” effects of glucose and fatty acids. Both elevated glucose and fatty acids can stimulate superoxide formation in endothelial cells, resulting in similar downstream effects. B, “Lipid-like” effects of glucose. Many of the biological effects of glucose are mediated via bioactive lipids, including DAG, sphingolipids, and arachidonic acid metabolites. AA indicates arachidonic acid; AR, aldose reductase; COX, cyclooxygenase; LOX, lipoxygenase; S1P, sphingosine 1-phosphate.

    Many of the Signaling Pathways Induced by Glucose Are Mediated by Bioactive Lipids

    Interestingly, many of the intracellular signaling pathways shown to be induced by elevated glucose are mediated by bioactive lipids. This provides a plethora of interactions between glucose and lipids. We discuss 3 of these lipid mediators of glucose action: DAG, sphingosine 1-phosphate, and arachidonic acid metabolites, as summarized in Figure 2B.

    It is well established that elevated glucose causes increased DAG synthesis and mass in tissues and in isolated cells.131–133 In the aorta of diabetic dogs, increased DAG levels are sustained for several years.132 Fatty acyl–coenzyme A (acyl-CoA) synthesis is required for DAG synthesis. Accordingly, it has been shown that chronic elevation of glucose increases acyl-CoA levels in rat skeletal muscle.134 How does glucose increase fatty acyl-CoA and DAG mass? King and colleagues have shown that the increased DAG mass occurs partly by de novo synthesis from glucose, probably via glyceraldehyde 3-phosphate and phosphatidic acid.133 The increased DAG is likely to explain the ability of glucose to activate DAG-sensitive protein kinase C isoforms, as well as downstream effects, such as decreased nitric oxide generation.133 One mechanism whereby glucose may increase DAG levels has been identified by Brownlee, as discussed above.128 In addition, Ruderman and colleagues have proposed a malonyl-CoA/long-chain acyl-CoA model that operates in several different cell types, including endothelial cells. Elevated glucose leads to increased levels of malonyl-CoA, which inhibits fatty acid β oxidation by inhibiting mitochondrial fatty acyl-CoA uptake, thereby channeling increased levels of acyl-CoAs into DAG and triacylglycerol.135 Together, these studies indicate that elevated glucose causes increased DAG synthesis, probably by several different mechanisms that all require fatty acyl-CoA.

    It has recently been shown that elevated levels of glucose can activate the sphingosine kinase pathway in aortic endothelial cells and that diabetes leads to a similar activation in the rat aorta.136 The glucose-induced increase in sphingosine kinase activity was shown to be attributable to selective activation of the sphingosine kinase 1 isoform. By using a dominant-negative mutant of sphingosine kinase 1, Wang and colleagues demonstrated that glucose-induced adhesion of monocytic cells was abolished in the absence of a functional sphingosine kinase 1.136 Activation of sphingosine kinase results in generation of sphingosine-1 phosphate (S1P). Interestingly, S1P was recently shown to inhibit monocyte adherence to aortas from diabetic NOD mice through activation of the S1P1 receptor.137 The overall biological effect of S1P is likely to depend on S1P receptor subtypes expressed under different conditions. Thus, in addition to DAG, sphingolipids appear to mediate some of the effects of elevated glucose, at least in endothelial cells.

    Elevated glucose levels have also been shown to regulate several arachidonic acid metabolites. In the context of elevated glucose, arachidonic acid metabolism by cyclooxygenases to prostanoids, and by lipoxygenases to hydroxyeicosatetraenoic acids (HETE), are most relevant.138 Elevated glucose levels induce cyclooxygenase-2 expression in human aortic endothelial cells, and this is associated with an increased release of thromboxane B2139 and prostaglandin E2.140 It has recently been shown that a thromboxane A2 receptor antagonist protects against endothelial dysfunction and diabetes-accelerated lesion initiation in apoE-deficient mice.141 Elevated glucose also induces expression of 12/15 lipoxygenase and increases levels of the 12/15 lipoxygenase product 12(S)HETE in aortic endothelial cells.142–143 Interestingly, overexpression of 15-lipoxygenase in the endothelium of LDLR-deficient mice results in larger fatty streaks compared with LDLR−/− controls.144 Accordingly, 12(S)-HETE has been found to stimulate monocyte adhesion to cultured endothelial cells.145 Minimally oxidized LDL and oxidized phospholipids also appear to mediate their effects in endothelial cells through increased arachidonic acid release and subsequent stimulation of the lipoxygenase pathway.115,117 In this context, it is interesting that elevated glucose can cause oxidation of LDL38 and that reactive carbonyl species, AGEs, and advanced lipoxidation end products (ALEs) also might result in modification of lipids in lipoproteins.146

    Conclusions and Perspective

    It is clear that type 1 diabetes, without marked changes in plasma lipid levels, can accelerate atherosclerosis and cardiovascular disease in humans and experimental animal models. Elevated glucose might play an important role in this effect, but glucose is likely to act in concert with basal lipids levels. Thus, hyperglycemia alone appears insufficient to accelerate atherosclerosis, at least in various animal models. Similarly, when plasma lipid levels are extremely high, the effect of hyperglycemia appears to be overpowered, perhaps because glucose and lipids act, at least in part, by the same mechanisms to accelerate atherosclerosis. We propose that there is a significant amount of cross-talk between signaling events induced by glucose and lipids.

    In the past 10 years, much has been learned regarding how diabetes accelerates lesion initiation, but little is still known about how diabetes affects more advanced lesions. Given the fact that different phases of lesion progression are regulated by different mechanisms, it is unlikely that diabetes accelerates cardiovascular events merely by inducing an accelerated formation of lesions of atherosclerosis. Studies on advanced lesions will be necessary to further our knowledge on the cellular and molecular mechanisms whereby diabetes leads to cardiovascular events.

    Note Added In Proof

    A recent study by Shi et al (J Clin Invest. 2006;116:3015–3025) shows that fatty acids stimulate cytokine production primarily through a Toll-like receptor 4 (TLR4)-dependent mechanism in macrophages. It will be important to evaluate the role of this TLR4 pathway versus intracellular effects of fatty acids in vascular cells.

    Original received October 10, 2006; revision received January 5, 2007; accepted January 24, 2007.

    We regret that, because of space limitations, we could not discuss all the important contributions to this.

    Sources of Funding

    Supported by NIH grants HL62887 and HL076719 to (K.E.B.) and HL52848 (to R.C.L.).

    Disclosures

    None.

    Footnotes

    Correspondence to K.E. Bornfeldt, Department of Pathology, 1959 NE Pacific St, University of Washington, Seattle, WA 98195-7470. E-mail

    References

    • 1 Ruderman NB, Haudenschild C. Diabetes as an atherogenic factor. Prog Cardiovasc Dis. 1984; 26: 373–412.CrossrefMedlineGoogle Scholar
    • 2 Laing SP, Swerdlow AJ, Slater SD, Burden AC, Morris A, Waugh NR, Gatling W, Bingley PJ, Patterson CC. Mortality from heart disease in a cohort of 23,000 patients with insulin-treated diabetes. Diabetologia. 2003; 46: 760–765.CrossrefMedlineGoogle Scholar
    • 3 Yki-Järvinen H. Management of type 2 diabetes mellitus and cardiovascular risk. Drugs. 2001; 60: 975–983.Google Scholar
    • 4 Steiner G. Treating lipid abnormalities in patients with type 2 diabetes mellitus. Am J Cardiol. 2001; 88: 12 Suppl 1, 37–40.Google Scholar
    • 5 Robins SJ. Targeting low high-density lipoprotein cholesterol for therapy: lessons from the Veterans Affairs High-density Lipoprotein Intervention Trial. Am J Cardiol. 2001; 88 (suppl 1): 19–23.Google Scholar
    • 6 Huang ES, Meigs JB, Singer DE. The effect of interventions to prevent cardiovascular disease in patients with type 2 diabetes mellitus. Am J Med. 2001; 111: 633–642.CrossrefMedlineGoogle Scholar
    • 7 Gotto AM. Lipid management in diabetic patients: lessons from prevention trials. Am J Med. 2002; 112 (suppl 8A): 19S–26S.CrossrefMedlineGoogle Scholar
    • 8 Ginsberg HN. Lipoprotein physiology in nondiabetic and diabetic states. Relationship to atherogenesis. Diabetes Care. 1991; 14: 839–855.CrossrefMedlineGoogle Scholar
    • 9 Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B; Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med. 2005; 353: 2643–2653.CrossrefMedlineGoogle Scholar
    • 10 DCCT; The Diabetes Control and Complications Trial Research Group. The effects of intensive treatment of diabetes on the development and progression of long-term complications of insulin-dependent diabetes mellitus. N Engl J Med. 1993; 329: 977–986.CrossrefMedlineGoogle Scholar
    • 11 Nathan DM, Lachin J, Cleary P, Orchard T, Brillon DJ, Backlund JY, O’Leary DH, Genuth S; Diabetes Control and Complications Trial; Epidemiology of Diabetes Interventions and Complications Research Group. Intensive diabetes therapy and carotid intima-media thickness in type 1 diabetes mellitus. N Engl J Med. 2003; 348: 2294–2303.CrossrefMedlineGoogle Scholar
    • 12 Yan SF, Ramasamy R, Bucciarelli LG, Wendt T, Lee LK, Hudson BI, Stern DM, Lalla E, DU Yan S, Rong LL, Naka Y, Schmidt AM. RAGE and its ligands: a lasting memory in diabetic complications? Diab Vasc Dis Res. 2004; 1: 10–20.CrossrefMedlineGoogle Scholar
    • 13 UKPDS Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS33). Lancet. 1998; 352: 837–853.CrossrefMedlineGoogle Scholar
    • 14 Kunjathoor VV, Wilson DL, LeBoeuf RC. Increased atherosclerosis in streptozotocin-induced diabetic mice. J Clin Invest. 1996; 97: 1767–1773.CrossrefMedlineGoogle Scholar
    • 15 Simionescu M, Popov D, Sima A, Hasu M, Costache G, Faitar S, Vulpanovici A, Stancu C, Stern D, Simionescu N. Pathobiochemistry of combined diabetes and atherosclerosis studied on a novel animal model. The hyperlipemic-hyperglycemic hamster. Am J Pathol. 1996; 148: 997–1014.MedlineGoogle Scholar
    • 16 Gerrity RG, Natarajan R, Nadler JL, Kimsey T. Diabetes-induced accelerated atherosclerosis in swine. Diabetes. 2001; 50: 1654–1665.CrossrefMedlineGoogle Scholar
    • 17 Renard CB, Kramer F, Johansson F, Lamharzi N, Tannock LR, von Herrath MG, Chait A, Bornfeldt KE. Diabetes and diabetes-associated lipid abnormalities have distinct effects on initiation and progression of atherosclerotic lesions. J Clin Invest. 2004; 114: 659–668.CrossrefMedlineGoogle Scholar
    • 18 Vikramadithyan RK, Hu Y, Noh HL, Liang CP, Hallam K, Tall AR, Ramasamy R, Goldberg IJ. Human aldose reductase expression accelerates diabetic atherosclerosis in transgenic mice. J Clin Invest. 2005; 115: 2434–2443.CrossrefMedlineGoogle Scholar
    • 19 Goldberg IJ, Dansky HM. Diabetic vascular disease: an experimental objective. Arterioscler Thromb Vasc Biol. 2006; 26: 1693–1701.LinkGoogle Scholar
    • 20 Lee AY, Chung SK, Chung SS. Demonstration that polyol accumulation is responsible for diabetic cataract by the use of transgenic mice expressing the aldose reductase gene in the lens. Proc Natl Acad Sci U S A. 1995; 92: 2780–2784.CrossrefMedlineGoogle Scholar
    • 21 Yamaoka T, Nishimura C, Yamashita K, Itakura M, Yamada T, Fujimoto J, Kokai Y. Acute onset of diabetic pathological changes in transgenic mice with human aldose reductase cDNA. Diabetologia. 1995; 38: 255–261.CrossrefMedlineGoogle Scholar
    • 22 Srivastava SK, Ramana KV, Bhatnagar A. Role of aldose reductase and oxidative damage in diabetes and the consequent potential for therapeutic options. Endocr Rev. 2005; 26: 380–392.CrossrefMedlineGoogle Scholar
    • 23 McGill HC Jr, McMahan CA, Zieske AW, Malcom GT, Tracy RE, Strong JP. Effects of nonlipid risk factors on atherosclerosis in youth with a favorable lipoprotein profile. Circulation. 2001; 103: 1546–1550.CrossrefMedlineGoogle Scholar
    • 24 McGill HC Jr, McMahan CA, Malcom GT, Oalmann MC, Strong JP. Relation of glycohemoglobin and adiposity to atherosclerosis in youth. Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Arterioscler Thromb Vasc Biol. 1995; 15: 431–440.CrossrefMedlineGoogle Scholar
    • 25 Järvisalo MJ, Putto-Laurila A, Jartti L, Lehtimäki T, Solakivi T, Rönnemaa T, Raitakari OT. Carotid artery intima-media thickness in children with type 1 diabetes. Diabetes. 2002; 51: 493–498.CrossrefMedlineGoogle Scholar
    • 26 Reaven P, Merat S, Casanada F, Sutphin M, Palinski W. Effect of streptozotocin-induced hyperglycemia on lipid profiles, formation of advanced glycation endproducts in lesions, and extent of atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol. 1997; 17: 2250–2256.CrossrefMedlineGoogle Scholar
    • 27 Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death. Arterioscler Thromb Vasc Biol. 2000; 20: 1262–1275.CrossrefMedlineGoogle Scholar
    • 28 Stary HC, Chandler AB, Dinsmore RE, Fuster V, Glagov S, Insull W Jr, Rosenfeld ME, Schwartz CJ, Wagner WD, Wissler RW. A definition of advanced types of atherosclerotic lesions and a histological classification of atherosclerosis. A report from the Committee on Vascular Lesions of the Council of Atherosclerosis, American Heart Association. Circulation. 1995; 92: 1355–1374.CrossrefMedlineGoogle Scholar
    • 29 Quehenberger O. Thematic review series: the immune system and atherogenesis. Molecular mechanisms regulating monocyte recruitment in atherosclerosis. J Lipid Res. 2005; 46: 1582–1590.CrossrefMedlineGoogle Scholar
    • 30 Richardson M, Hadcock SJ, DeReske M, Cybulsky MI. Increased expression in vivo of VCAM-1 and E-selectin by the aortic endothelium of normolipemic and hyperlipemic diabetic rabbits. Arterioscler Thromb. 1994; 14: 760–769.CrossrefMedlineGoogle Scholar
    • 31 Booth G, Stalker TJ, Lefer AM, Scalia R. Mechanisms of amelioration of glucose-induced endothelial dysfunction following inhibition of protein kinase C in vivo. Diabetes. 2002; 51: 1556–1564.CrossrefMedlineGoogle Scholar
    • 32 Ribau JC, Hadcock SJ, Teoh K, DeReske M, Richardson M. Endothelial adhesion molecule expression is enhanced in the aorta and internal mammary artery of diabetic patients. J Surg Res. 1999; 85: 225–233.CrossrefMedlineGoogle Scholar
    • 33 Chavakis T, Bierhaus A, Al-Fakhri N, Schneider D, Witte S, Linn T, Nagashima M, Morser J, Arnold B, Preissner KT, Nawroth PP. The pattern recognition receptor (RAGE) is a counterreceptor for leukocyte integrins: a novel pathway for inflammatory cell recruitment. J Exp Med. 2003; 198: 1507–1515.CrossrefMedlineGoogle Scholar
    • 34 Feng L, Matsumoto C, Schwartz A, Schmidt AM, Stern DM, Pile-Spellman J. Chronic vascular inflammation in patients with type 2 diabetes: endothelial biopsy and RT-PCR analysis. Diabetes Care. 2005; 28: 379–384.CrossrefMedlineGoogle Scholar
    • 35 Cipolletta C, Ryan KE, Hanna EV, Trimble ER. Activation of peripheral blood CD14+ monocytes occurs in diabetes. Diabetes. 2005; 54: 2779–2786.CrossrefMedlineGoogle Scholar
    • 36 Steinberg D. Thematic review series: the pathogenesis of atherosclerosis: an interpretive history of the cholesterol controversy. Part III: Mechanistically defining the role of hyperlipidemia. J Lipid Res. 2005; 46: 2037–2051.CrossrefMedlineGoogle Scholar
    • 37 Burke AP, Kolodgie FD, Zieske A, Fowler DR, Weber DK, Varghese PJ, Farb A, Virmani R. Morphologic findings of coronary atherosclerotic plaques in diabetics: a postmortem study. Arterioscler Thromb Vasc Biol. 2004; 24: 1266–1271.LinkGoogle Scholar
    • 38 Lamharzi N, Renard CB, Kramer F, Pennathur S, Heinecke JW, Chait A, Bornfeldt KE. Hyperlipidemia in concert with hyperglycemia stimulates the proliferation of macrophages in atherosclerotic lesions: potential role of glucose-oxidized LDL. Diabetes. 2004; 53: 3217–3225.CrossrefMedlineGoogle Scholar
    • 39 Williams KJ, Tabas I. The response-to-retention hypothesis of early atherogenesis. Arterioscler Thromb Vasc Biol. 1995; 15: 551–561.CrossrefMedlineGoogle Scholar
    • 40 Gustafsson M, Boren J. Mechanism of lipoprotein retention by the extracellular matrix. Curr Opin Lipidol. 2004; 15: 505–514.CrossrefMedlineGoogle Scholar
    • 41 Heickendorff L, Ledet T, Rasmussen LM. Glycosaminoglycans in the human aorta in diabetes mellitus: a study of tunica media from areas with and without atherosclerotic plaque. Diabetologia. 1994; 37: 286–292.CrossrefMedlineGoogle Scholar
    • 42 LeBaron RG, Zimmermann DR, Ruoslahti E. Hyaluronate binding properties of versican. J Biol Chem. 1992; 267: 10003–10010.CrossrefMedlineGoogle Scholar
    • 43 Olin KL, Potter-Perigo S, Barrett PH, Wight TN, Chait A. Lipoprotein lipase enhances the binding of native and oxidized low density lipoproteins to versican and biglycan synthesized by cultured arterial smooth muscle cells. J Biol Chem. 1999; 274: 34629–34636.CrossrefMedlineGoogle Scholar
    • 44 Chai S, Chai Q, Danielsen CC, Hjorth P, Nyengaard JR, Ledet T, Yamaguchi Y, Rasmussen LM, Wogensen L. Overexpression of hyaluronan in the tunica media promotes the development of atherosclerosis. Circ Res. 2005; 96: 583–591.LinkGoogle Scholar
    • 45 Wilkinson TS, Bressler SL, Evanko SP, Braun KR, Wight TN. Overexpression of hyaluronan synthases alters vascular smooth muscle cell phenotype and promotes monocyte adhesion. J Cell Physiol. 2006; 206: 378–385.CrossrefMedlineGoogle Scholar
    • 46 McKee CM, Penno MB, Cowman M, Burdick MD, Strieter RM, Bao C, Noble PW. Hyaluronan (HA) fragments induce chemokine gene expression in alveolar macrophages. The role of HA size and CD44. J Clin Invest. 1996; 98: 2403–2413.CrossrefMedlineGoogle Scholar
    • 47 Cuff CA, Kothapalli D, Azonobi I, Chun S, Zhang Y, Belkin R, Yeh C, Secreto A, Assoian RK, Rader DJ, Pure E. The adhesion receptor CD44 promotes atherosclerosis by mediating inflammatory cell recruitment and vascular cell activation. J Clin Invest. 2001; 108: 1031–1040.CrossrefMedlineGoogle Scholar
    • 48 Virmani R, Burke AP, Farb A, Kolodgie FD. Pathology of the vulnerable plaque. J Am Coll Cardiol. 2006; 47: (8 suppl): C13–C18.CrossrefMedlineGoogle Scholar
    • 49 Suzuki LA, Poot M, Gerrity RG, Bornfeldt KE. Diabetes accelerates smooth muscle accumulation in lesions of atherosclerosis: lack of direct growth-promoting effects of high glucose levels. Diabetes. 2001; 50: 851–860.CrossrefMedlineGoogle Scholar
    • 50 Clarke MC, Figg N, Maguire JJ, Davenport AP, Goddard M, Littlewood TD, Bennett MR Apoptosis of vascular smooth muscle cells induces features of plaque vulnerability in atherosclerosis. Nat Med. 2006; 12: 1075–1080.CrossrefMedlineGoogle Scholar
    • 51 Ruiz E, Gordillo-Moscoso A, Padilla E, Redondo S, Rodriguez E, Reguillo F, Briones AM, van Breemen C, Okon E, Tejerina T. Human vascular smooth muscle cells from diabetic patients are resistant to induced apoptosis due to high Bcl-2 expression. Diabetes. 2006; 55: 1243–1251.CrossrefMedlineGoogle Scholar
    • 52 Hall JL, Matter CM, Wang X, Gibbons GH. Hyperglycemia inhibits vascular smooth muscle cell apoptosis through a protein kinase C-dependent pathway. Circ Res. 2000; 87: 574–580.CrossrefMedlineGoogle Scholar
    • 53 Li H, Telemaque S, Miller RE, Marsh JD. High glucose inhibits apoptosis induced by serum deprivation in vascular smooth muscle cells via upregulation of Bcl-2 and Bcl-xl. Diabetes. 2005; 54: 540–545.CrossrefMedlineGoogle Scholar
    • 54 Virmani R, Burke AP, Kolodgie F. Morphological characteristics of coronary atherosclerosis in diabetes mellitus. Can J Cardiol. 2006; 22 (suppl B): 81B–84B.CrossrefMedlineGoogle Scholar
    • 55 Tabas I. Free cholesterol-induced cytotoxicity: a possible contributing factor to macrophage foam cell necrosis in advanced atherosclerotic lesions. Trends Cardiovasc Med. 1997; 7: 256–263.CrossrefMedlineGoogle Scholar
    • 56 Feng B, Zhang D, Kuriakose G, Devlin CM, Kockx M, Tabas I. Niemann-Pick C heterozygosity confers resistance to lesional necrosis and macrophage apoptosis in murine atherosclerosis. Proc Natl Acad Sci U S A. 2003; 100: 10423–10428.CrossrefMedlineGoogle Scholar
    • 57 Fukuhara-Takaki K, Sakai M, Sakamoto Y, Takeya M, Horiuchi S. Expression of class A scavenger receptor is enhanced by high glucose in vitro and under diabetic conditions in vivo: one mechanism for an increased rate of atherosclerosis in diabetes. J Biol Chem. 2005; 280: 3355–3364.CrossrefMedlineGoogle Scholar
    • 58 Li L, Sawamura T, Renier G. Glucose enhances human macrophage LOX-1 expression: role for LOX-1 in glucose-induced macrophage foam cell formation. Circ Res. 2004; 94: 892–901.LinkGoogle Scholar
    • 59 Sampson MJ, Davies IR, Braschi S, Ivory K, Hughes DA. Increased expression of a scavenger receptor (CD36) in monocytes from subjects with Type 2 diabetes. Atherosclerosis. 2003; 167: 129–134.CrossrefMedlineGoogle Scholar
    • 60 Passarelli M, Tang C, McDonald TO, O’Brien KD, Gerrity RG, Heinecke JW, Oram JF. Advanced glycation end product precursors impair ABCA1-dependent cholesterol removal from cells. Diabetes. 2005; 54: 2198–2205.CrossrefMedlineGoogle Scholar
    • 61 Mauldin JP, Srinivasan S, Mulya A, Gebre A, Parks JS, Daugherty A, Hedrick CC. Reduction in ABCG1 in type 2 diabetic mice increases macrophage foam cell formation. J Biol Chem. 2006; 281: 21216–21224.CrossrefMedlineGoogle Scholar
    • 62 Liu J, Thewke DP, Su YR, Linton MF, Fazio S, Sinensky MS. Reduced macrophage apoptosis is associated with accelerated atherosclerosis in low-density lipoprotein receptor-null mice. Arterioscler Thromb Vasc Biol. 2005; 25: 174–179.LinkGoogle Scholar
    • 63 Han S, Liang CP, DeVries-Seimon T, Ranalletta M, Welch CL, Collins-Fletcher K, Accili D, Tabas I, Tall AR. Macrophage insulin receptor deficiency increases ER stress-induced apoptosis and necrotic core formation in advanced atherosclerotic lesions. Cell Metab. 2006; 3: 257–266.CrossrefMedlineGoogle Scholar
    • 64 Gough PJ, Gomez IG, Wille PT, Raines EW. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest. 2006; 116: 59–69.CrossrefMedlineGoogle Scholar
    • 65 Moreno PR, Fuster V. New aspects in the pathogenesis of diabetic atherothrombosis. J Am Coll Cardiol. 2004; 44: 2293–2300.CrossrefMedlineGoogle Scholar
    • 66 Kolodgie FD, Gold HK, Burke AP, Fowler DR, Kruth HS, Weber DK, Farb A, Guerrero LJ, Hayase M, Kutys R, Narula J, Finn AV, Virmani R. Intraplaque hemorrhage and progression of coronary atheroma. N Engl J Med. 2003; 349: 2316–2325.CrossrefMedlineGoogle Scholar
    • 67 Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, Sylvin E, Lo KM, Gillies S, Javaherian K, Folkman J. Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis. Proc Natl Acad Sci U S A. 2003; 100: 4736–4741.CrossrefMedlineGoogle Scholar
    • 68 de Nooijer R, Verkleij CJ, von der Thusen JH, Jukema JW, van der Wall EE, van Berkel TJ, Baker AH, Biessen EA. 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.LinkGoogle Scholar
    • 69 Papaspyridonos M, Smith A, Burnand KG, Taylor P, Padayachee S, Suckling KE, James CH, Greaves DR, Patel L. Novel candidate genes in unstable areas of human atherosclerotic plaques. Arterioscler Thromb Vasc Biol. 2006; 26: 1837–1844.LinkGoogle Scholar
    • 70 Uemura S, Matsushita H, Li W, Glassford AJ, Asagami T, Lee KH, Harrison DG, Tsao PS. Diabetes mellitus enhances vascular matrix metalloproteinase activity: role of oxidative stress. Circ Res. 2001; 88: 1291–1298.CrossrefMedlineGoogle Scholar
    • 71 Chung AW, Hsiang YN, Matzke LA, McManus BM, van Breemen C, Okon EB. Reduced expression of vascular endothelial growth factor paralleled with the increased angiostatin expression resulting from the upregulated activities of matrix metalloproteinase-2 and -9 in human type 2 diabetic arterial vasculature. Circ Res. 2006; 99: 140–148.LinkGoogle Scholar
    • 72 Xue M, Thompson PJ, Clifton-Bligh R, Fulcher G, Gallery ED, Jackson C. Leukocyte matrix metalloproteinase-9 is elevated and contributes to lymphocyte activation in type I diabetes. Int J Biochem Cell Biol. 2005; 37: 2406–2416.CrossrefMedlineGoogle Scholar
    • 73 Marfella R, D’Amico M, Esposito K, Baldi A, Di Filippo C, Siniscalchi M, Sasso FC, Portoghese M, Cirillo F, Cacciapuoti F, Carbonara O, Crescenzi B, Baldi F, Ceriello A, Nicoletti GF, D’Andrea F, Verza M, Coppola L, Rossi F, Giugliano D. The ubiquitin-proteasome system and inflammatory activity in diabetic atherosclerotic plaques: effects of rosiglitazone treatment. Diabetes. 2006; 55: 622–632.CrossrefMedlineGoogle Scholar
    • 74 Death AK, Fisher EJ, McGrath KC, Yue DK. High glucose alters matrix metalloproteinase expression in two key vascular cells: potential impact on atherosclerosis in diabetes. Atherosclerosis. 2003: 168; 263–269.CrossrefMedlineGoogle Scholar
    • 75 Everhart JE, Pettitt DJ, Knowler WC, Rose FA, Bennett PH. Medial artery calcifications and its association with mortality and complications of diabetes. Diabetologia. 1988; 31: 16–23.CrossrefMedlineGoogle Scholar
    • 76 MacDougall ED, Kramer F, Polinsky P, Barnhart S, Askari B, Johansson F, Varon R, Rosenfeld ME, Oka K, Chan L, Schwartz SM, Bornfeldt KE. Aggressive very low-density lipoprotein (VLDL) and LDL lowering by gene transfer of the VLDL receptor combined with a low-fat diet regimen induces regression and reduces macrophage content in advanced atherosclerotic lesions in LDL receptor-deficient mice. Am J Pathol. 2006; 168: 2064–2073.CrossrefMedlineGoogle Scholar
    • 77 Fumeron F, Reis AF, Velho G. Genetics of macrovascular complications in diabetes. Curr Diab Rep. 2006; 6: 162–168.CrossrefMedlineGoogle Scholar
    • 78 Lange LA, Bowden DW, Langefeld CD, Wagenknecht LE, Carr JJ, Rich SS, Riley WA, Freedman BI. Heritability of carotid artery intima-medial thickness in type 2 diabetes. Stroke. 2002; 33: 1876–1881.LinkGoogle Scholar
    • 79 Socquard E, Durlach A, Clavel C, Nazeyrollas P, Durlach V. Association of HindIII and PvuII genetic polymorphisms of lipoprotein lipase with lipid metabolism and macrovascular events in type 2 diabetic patients. Diabetes Metab. 2006; 32: 262–269.CrossrefMedlineGoogle Scholar
    • 80 Durlach A, Clavel C, Girard-Globa A, Durlach V. Sex-dependent association of a genetic polymorphism of cholesteryl ester transfer protein with high-density lipoprotein cholesterol and macrovascular pathology in type II diabetic patients. J Clin Endocrinol Metab. 1999; 84: 3656–3659.CrossrefMedlineGoogle Scholar
    • 81 Kawasaki I, Tahara H, Emoto M, Shoji T, Nishizawa Y. Relationship between TaqIB cholesteryl ester transfer protein gene polymorphism and macrovascular complications in Japanese patients with type 2 diabetes. Diabetes. 2002; 51: 871–874.CrossrefMedlineGoogle Scholar
    • 82 Rewers M, Kamboh MI, Hoag S, Shetterly SM, Ferrell RE, Hamman RF. ApoA-IV polymorphism associated with myocardial infarction in obese NIDDM patients. The San Luis Valley Diabetes Study. Diabetes. 1994; 43: 1485–1489.CrossrefMedlineGoogle Scholar
    • 83 Gouni-Berthold I, Giannakidou E, Muller-Wieland D, Faust M, Kotzka J, Berthold HK, Krone W. Association between the PPARα L162V polymorphism, plasma lipoprotein levels, and atherosclerotic disease in patients with diabetes mellitus type 2 and in nondiabetic controls. Am Heart J. 2004; 147: 1117–1124.CrossrefMedlineGoogle Scholar
    • 84 Temelkova-Kurktschiev T, Hanefeld M, Chinetti G, Zawadzki C, Haulon S, Kubaszek A, Koehler C, Leonhardt W, Staels B, Laakso M. Ala12Ala genotype of the peroxisome proliferator-activated receptor gamma2 protects against atherosclerosis. J Clin Endocrinol Metab. 2004; 89: 4238–4242.CrossrefMedlineGoogle Scholar
    • 85 Hegele RA, Al-Shali KZ, House AA, Hanley AJ, Harris SB, Mamakeesick M, Fenster A, Zinman B, Cao H, Spence JD. Disparate associations of a functional promoter polymorphism in PCK1 with carotid wall ultrasound traits. Stroke. 2005; 36: 2566–2570.LinkGoogle Scholar
    • 86 Haj FG, Zabolotny JM, Kim YB, Kahn BB, Neel BG. Liver-specific protein-tyrosine phosphatase 1B (PTP1B) re-expression alters glucose homeostasis of PTP1B-/- mice. J Biol Chem. 2005; 280: 15038–15046.CrossrefMedlineGoogle Scholar
    • 87 Burdon KP, Bento JL, Langefeld CD, Campbell JK, Carr JJ, Wagenknecht LM, Herrington DM, Freedman BI, Rich SS, Bowden DW. Association of protein tyrosine phosphatase-N1 polymorphisms with coronary calcified plaque in the Diabetes Heart Study. Diabetes. 2006; 55: 651–658.CrossrefMedlineGoogle Scholar
    • 88 Pfohl M, Koch M, Enderle MD, Kuhn R, Fullhase J, Karsch KR, Haring HU. Paraoxonase 192 Gln/Arg gene polymorphism, coronary artery disease, and myocardial infarction in type 2 diabetes. Diabetes. 1999; 48: 623–627.CrossrefMedlineGoogle Scholar
    • 89 Zhang C, Lopez-Ridaura R, Hunter DJ, Rifai N, Hu FB. Common variants of the endothelial nitric oxide synthase gene and the risk of coronary heart disease among U.S. diabetic men. Diabetes. 2006; 55: 2140–2147.CrossrefMedlineGoogle Scholar
    • 90 Bucciarelli LG, Wendt T, Qu W, Lu Y, Lalla E, Rong LL, Goova MT, Moser B, Kislinger T, Lee DC, Kashyap Y, Stern DM, Schmidt AM. RAGE blockade stabilizes established atherosclerosis in diabetic apolipoprotein E-null mice. Circulation. 2002; 106: 2827–2835.LinkGoogle Scholar
    • 91 dos Santos KG, Canani LH, Gross JL, Tschiedel B, Pires Souto KE, Roisenberg I. The -374A allele of the receptor for advanced glycation end products gene is associated with a decreased risk of ischemic heart disease in African-Brazilians with type 2 diabetes. Mol Genet Metab. 2005; 85: 149–156.CrossrefMedlineGoogle Scholar
    • 92 Pettersson-Fernholm K, Forsblom C, Hudson BI, Perola M, Grant PJ, Groop PH; Finn-Diane Study Group. The functional -374 T/A RAGE gene polymorphism is associated with proteinuria and cardiovascular disease in type 1 diabetic patients. Diabetes. 2003; 52: 891–894.CrossrefMedlineGoogle Scholar
    • 93 Demaine AG. Polymorphisms of the aldose reductase gene and susceptibility to diabetic microvascular complications. Curr Med Chem. 2003; 10: 1389–1398.CrossrefMedlineGoogle Scholar
    • 94 Ragolia L, Palaia T, Hall CE, Maesaka JK, Eguchi N, Urade Y. Accelerated glucose intolerance, nephropathy, and atherosclerosis in prostaglandin D2 synthase knock-out mice. J Biol Chem. 2005; 280: 29946–29955.CrossrefMedlineGoogle Scholar
    • 95 Kako Y, Masse M, Huang LS, Tall AR, Goldberg IJ. Lipoprotein lipase deficiency and CETP in streptozotocin-treated apoB-expressing mice. J Lipid Res. 2002; 43: 872–877.CrossrefMedlineGoogle Scholar
    • 96 Paigen B. Genetics of responsiveness to high-fat and high-cholesterol diets in the mouse. Am J Clin Nutr. 1995; 62: 458S–462S.CrossrefMedlineGoogle Scholar
    • 97 Nishina PM, Naggert JK, Verstuyft J, Paigen B. Atherosclerosis in genetically obese mice: the mutants obese, diabetes, fat, tubby, and lethal yellow. Metabolism. 1994; 43: 554–558.CrossrefMedlineGoogle Scholar
    • 98 Goodarzi MO, Taylor KD, Guo X, Quinones MJ, Cui J, Li Y, Saad MF, Yang H, Hsueh WA, Hodis HN, Rotter JI. Association of the diabetes gene calpain-10 with subclinical atherosclerosis: the Mexican-Am Coronary Artery Disease Study. Diabetes. 2005; 54: 1228–1232.CrossrefMedlineGoogle Scholar
    • 99 Bowden DW, Rudock M, Ziegler J, Lehtinen AB, Xu J, Wagenknecht LE, Herrington D, Rich SS, Freedman BI, Carr JJ, Langefeld CD. Coincident linkage of type 2 diabetes, metabolic syndrome, and measures of cardiovascular disease in a genome scan of the diabetes heart study. Diabetes. 2006; 55: 1985–1994.CrossrefMedlineGoogle Scholar
    • 100 Federici M, Lauro R. Review article: diabetes and atherosclerosis-running on a common road. Aliment Pharmacol Ther. 2005; 22 (suppl 2): 11–15.CrossrefGoogle Scholar
    • 101 Mitchell BD, Imumorin IG. Genetic determinants of diabetes and atherosclerosis. Curr Atheroscler Rep. 2002; 4: 193–198.CrossrefMedlineGoogle Scholar
    • 102 Seidelmann SB, De Luca C, Leibel RL, Breslow JL, Tall AR, Welch CL. Quantitative trait locus mapping of genetic modifiers of metabolic syndrome and atherosclerosis in low-density lipoprotein receptor-deficient mice: identification of a locus for metabolic syndrome and increased atherosclerosis on chromosome 4. Arterioscler Thromb Vasc Biol. 2005; 25: 204–210.LinkGoogle Scholar
    • 103 Mu JL, Naggert JK, Svenson KL, Collin GB, Kim JH, McFarland C, Nishina PM, Levine DM, Williams KJ, Paigen B. Quantitative trait loci analysis for the differences in susceptibility to atherosclerosis and diabetes between inbred mouse strains C57BL/6J and C57BLKS/J. J Lipid Res. 1999; 40: 1328–1335.CrossrefMedlineGoogle Scholar
    • 104 Estrada-Smith D, Collins AR, Wang X, Crockett C, Castellani L, Lusis AJ, Davis RC. Impact of chromosome 2 obesity loci on cardiovascular complications of insulin resistance in LDL receptor-deficient C57BL/6 mice. Diabetes. 2006; 55: 2265–2271.CrossrefMedlineGoogle Scholar
    • 105 Kouroedov A, Eto M, Joch H, Volpe M, Luscher TF, Cosentino F. Selective inhibition of protein kinase Cβ2 prevents acute effects of high glucose on vascular cell adhesion molecule-1 expression in human endothelial cells. Circulation. 2004; 110: 91–96.LinkGoogle Scholar
    • 106 Rasmussen LM, Schmitz O, Ledet T. Increased expression of vascular cell adhesion molecule-1 (VCAM-1) in cultured endothelial cells exposed to serum from type 1 diabetic patients: no effects of high glucose concentrations. Scand J Clin Lab Invest. 2002; 62: 485–493.CrossrefMedlineGoogle Scholar
    • 107 Schmidt AM, Hori O, Chen JX, Li JF, Crandall J, Zhang J, Cao R, Yan SD, Brett J, Stern D. Advanced glycation endproducts interacting with their endothelial receptor induce expression of vascular cell adhesion molecule-1 (VCAM-1) in cultured human endothelial cells and in mice. A potential mechanism for the accelerated vasculopathy of diabetes. J Clin Invest. 1995; 96: 1395–1403.CrossrefMedlineGoogle Scholar
    • 108 Vlassara H, Fuh H, Donnelly T, Cybulsky M. Advanced glycation endproducts promote adhesion molecule (VCAM-1, ICAM-1) expression and atheroma formation in normal rabbits. Mol Med. 1995; 1: 447–456.CrossrefMedlineGoogle Scholar
    • 109 Valencia JV, Mone M, Zhang J, Weetall M, Buxton FP, Hughes TE. Divergent pathways of gene expression are activated by the RAGE ligands S100b and AGE-BSA. Diabetes. 2004; 53: 743–751.CrossrefMedlineGoogle Scholar
    • 110 Lieuw-a-Fa ML, Schalkwijk CG, Engelse M, van Hinsbergh VW. Interaction of Nε(carboxymethyl)lysine- and methylglyoxal-modified albumin with endothelial cells and macrophages. Splice variants of RAGE may limit the responsiveness of human endothelial cells to AGEs. Thromb Haemost. 2006; 95: 320–328.CrossrefMedlineGoogle Scholar
    • 111 Cacicedo JM, Yagihashi N, Keaney JF Jr, Ruderman NB, Ido Y. AMPK inhibits fatty acid-induced increases in NF-κB transactivation in cultured human umbilical vein endothelial cells. Biochem Biophys Res Commun. 2004; 324: 1204–1209.CrossrefMedlineGoogle Scholar
    • 112 Chen W, Jump DB, Grant MB, Esselman WJ, Busik JV. Dyslipidemia, but not hyperglycemia, induces inflammatory adhesion molecules in human retinal vascular endothelial cells. Invest Ophthalmol Vis Sci. 2003; 44: 5016–5022.CrossrefMedlineGoogle Scholar
    • 113 Srinivasan S, Yeh M, Danziger EC, Hatley ME, Riggan AE, Leitinger N, Berliner JA, Hedrick CC. Glucose regulates monocyte adhesion through endothelial production of interleukin-8. Circ Res. 2003; 92: 371–377.LinkGoogle Scholar
    • 114 Srinivasan S, Bolick DT, Hatley ME, Natarajan R, Reilly KB, Yeh M, Chrestensen C, Sturgill TW, Hedrick CC. Glucose regulates interleukin-8 production in aortic endothelial cells through activation of the p38 mitogen-activated protein kinase pathway in diabetes. J Biol Chem. 2004; 279: 31930–31936.CrossrefMedlineGoogle Scholar
    • 115 Honda HM, Leitinger N, Frankel M, Goldhaber JI, Natarajan R, Nadler JL, Weiss JN, Berliner JA. Induction of monocyte binding to endothelial cells by MM-LDL: role of lipoxygenase metabolites. Arterioscler Thromb Vasc Biol. 1999; 19: 680–686.CrossrefMedlineGoogle Scholar
    • 116 Gharavi NM, Baker NA, Mouillesseaux KP, Yeung W, Honda HM, Hsieh X, Yeh M, Smart EJ, Berliner JA. Role of endothelial nitric oxide synthase in the regulation of SREBP activation by oxidized phospholipids. Circ Res. 2006; 98: 768–776.LinkGoogle Scholar
    • 117 Huber J, Furnkranz A, Bochkov VN, Patricia MK, Lee H, Hedrick CC, Berliner JA, Binder BR, Leitinger N. Specific monocyte adhesion to endothelial cells induced by oxidized phospholipids involves activation of cPLA2 and lipoxygenase. J Lipid Res. 2006; 47: 1054–1062.CrossrefMedlineGoogle Scholar
    • 118 Griffin E, Re A, Hamel N, Fu C, Bush H, McCaffrey T, Asch AS. A link between diabetes and atherosclerosis: glucose regulates expression of CD36 at the level of translation. Nat Med. 2001; 7: 840–846.CrossrefMedlineGoogle Scholar
    • 119 Svensson L, Camejo G, Cabre A, Vallve JC, Pedreno J, Noren K, Wiklund O, Hulten LM. Fatty acids modulate the effect of darglitazone on macrophage CD36 expression. Eur J Clin Invest. 2003; 33: 464–471.CrossrefMedlineGoogle Scholar
    • 120 Vallve JC, Uliaque K, Girona J, Cabre A, Ribalta J, Heras M, Masana L. Unsaturated fatty acids and their oxidation products stimulate CD36 gene expression in human macrophages. Atherosclerosis. 2002; 164: 45–56.CrossrefMedlineGoogle Scholar
    • 121 Liang CP, Han S, Okamoto H, Carnemolla R, Tabas I, Accili D, Tall AR. Increased CD36 protein as a response to defective insulin signaling in macrophages. J Clin Invest. 2004; 113: 764–773.CrossrefMedlineGoogle Scholar
    • 122 Hayek T, Hussein K, Aviram M, Coleman R, Keidar S, Pavoltzky E, Kaplan M. Macrophage foam-cell formation in streptozotocin-induced diabetic mice: stimulatory effect of glucose. Atherosclerosis. 2005; 183: 25–33.CrossrefMedlineGoogle Scholar
    • 123 Uehara Y, Miura SI, von Eckardstein A, Abe S, Fujii A, Matsuo Y, Rust S, Lorkowski S, Assmann G, Yamada T, Saku K. Unsaturated fatty acids suppress the expression of the ATP-binding cassette transporter G1 (ABCG1) and ABCA1 genes via an LXR/RXR responsive element. Atherosclerosis. In press.Google Scholar
    • 124 Devaraj S, Jialal I. Low-density lipoprotein postsecretory modification, monocyte function, and circulating adhesion molecules in type 2 diabetic patients with and without macrovascular complications: the effect of α-tocopherol supplementation. Circulation. 2000; 102: 191–196.CrossrefMedlineGoogle Scholar
    • 125 Devaraj S, Glaser N, Griffen S, Wang-Polagruto J, Miguelino E, Jialal I. Increased monocytic activity and biomarkers of inflammation in patients with type 1 diabetes. Diabetes. 2006; 55: 774–779.CrossrefMedlineGoogle Scholar
    • 126 Wen Y, Gu J, Li SL, Reddy MA, Natarajan R, Nadler JL. Elevated glucose and diabetes promote interleukin-12 cytokine gene expression in mouse macrophages. Endocrinology. 2006; 147: 2518–2525.CrossrefMedlineGoogle Scholar
    • 127 Suganami T, Nishida J, Ogawa Y. A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor α. Arterioscler Thromb Vasc Biol. 2005; 25: 2062–2068.LinkGoogle Scholar
    • 128 Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005; 54: 1615–1625.CrossrefMedlineGoogle Scholar
    • 129 Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, Szabo C, Brownlee M. Inhibition of GAPDH activity by poly(ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest. 2003; 112: 1049–1057.CrossrefMedlineGoogle Scholar
    • 130 Du X, Edelstein D, Obici S, Higham N, Zou MH, Brownlee M. Insulin resistance reduces arterial prostacyclin synthase and eNOS activities by increasing endothelial fatty acid oxidation. J Clin Invest. 2006; 116: 1071–1080.CrossrefMedlineGoogle Scholar
    • 131 Inoguchi T, Battan R, Handler E, Sportsman JR, Heath W, King GL. Preferential elevation of protein kinase C isoform βII and diacylglycerol levels in the aorta and heart of diabetic rats: differential reversibility to glycemic control by islet cell transplantation. Proc Natl Acad Sci U S A. 1992; 89: 11059–11063.CrossrefMedlineGoogle Scholar
    • 132 Xia P, Inoguchi T, Kern TS, Engerman RL, Oates PJ, King GL. Characterization of the mechanism for the chronic activation of diacylglycerol-protein kinase C pathway in diabetes and hypergalactosemia. Diabetes. 1994; 43: 1122–1129.CrossrefMedlineGoogle Scholar
    • 133 Rask-Madsen C, King GL. Proatherosclerotic mechanisms involving protein kinase C in diabetes and insulin resistance. Arterioscler Thromb Vasc Biol. 2005; 25: 487–496.LinkGoogle Scholar
    • 134 Laybutt DR, Schmitz-Peiffer C, Saha AK, Ruderman NB, Biden TJ, Kraegen EW. Muscle lipid accumulation and protein kinase C activation in the insulin-resistant chronically glucose-infused rat. Am J Physiol. 1999; 277: E1070–E1076.MedlineGoogle Scholar
    • 135 Roduit R, Nolan C, Alarcon C, Moore P, Barbeau A, Delghingaro-Augusto V, Przybykowski E, Morin J, Masse F, Massie B, Ruderman N, Rhodes C, Poitout V, Prentki M. A role for the malonyl-CoA/long-chain acyl-CoA pathway of lipid signaling in the regulation of insulin secretion in response to both fuel and nonfuel stimuli. Diabetes. 2004; 53: 1007–1019.CrossrefMedlineGoogle Scholar
    • 136 Wang L, Xing XP, Holmes A, Wadham C, Gamble JR, Vadas MA, Xia P. Activation of the sphingosine kinase-signaling pathway by high glucose mediates the proinflammatory phenotype of endothelial cells. Circ Res. 2005; 97: 891–899.LinkGoogle Scholar
    • 137 Whetzel AM, Bolick DT, Srinivasan S, Macdonald TL, Morris MA, Ley K, Hedrick CC. Sphingosine-1 phosphate prevents monocyte/endothelial interactions in type 1 diabetic NOD mice through activation of the S1P1 receptor. Circ Res. 2006; 99: 731–739.LinkGoogle Scholar
    • 138 Natarajan R, Nadler JL. Lipid inflammatory mediators in diabetic vascular disease. Arterioscler Thromb Vasc Biol. 2004; 24: 1542–1548.LinkGoogle Scholar
    • 139 Cosentino F, Eto M, De Paolis P, van der Loo B, Bachschmid M, Ullrich V, Kouroedov A, Delli Gatti C, Joch H, Volpe M, Luscher TF. High glucose causes upregulation of cyclooxygenase-2 and alters prostanoid profile in human endothelial cells: role of protein kinase C and reactive oxygen species. Circulation. 2003; 107: 1017–1023.LinkGoogle Scholar
    • 140 Sheu ML, Ho FM, Yang RS, Chao KF, Lin WW, Lin-Shiau SY, Liu SH. High glucose induces human endothelial cell apoptosis through a phosphoinositide 3-kinase-regulated cyclooxygenase-2 pathway. Arterioscler Thromb Vasc Biol. 2005; 25: 539–545.LinkGoogle Scholar
    • 141 Zuccollo A, Shi C, Mastroianni R, Maitland-Toolan KA, Weisbrod RM, Zang M, Xu S, Jiang B, Oliver-Krasinski JM, Cayatte AJ, Corda S, Lavielle G, Verbeuren TJ, Cohen RA. The thromboxane A2 receptor antagonist S18886 prevents enhanced atherogenesis caused by diabetes mellitus. Circulation. 2005; 112: 3001–3008.LinkGoogle Scholar
    • 142 Brown ML, Jakubowski JA, Leventis LL, Deykin D. Elevated glucose alters eicosanoid release from porcine aortic endothelial cells. J Clin Invest. 1988; 82: 2136–2141.CrossrefMedlineGoogle Scholar
    • 143 Patricia MK, Natarajan R, Dooley AN, Hernandez F, Gu JL, Berliner JA, Rossi JJ, Nadler JL, Meidell RS, Hedrick CC. Adenoviral delivery of a leukocyte-type 12 lipoxygenase ribozyme inhibits effects of glucose and platelet-derived growth factor in vascular endothelial and smooth muscle cells. Circ Res. 2001; 88: 659–665.CrossrefMedlineGoogle Scholar
    • 144 Harats D, Shaish A, George J, Mulkins M, Kurihara H, Levkovitz H, Sigal E. Overexpression of 15-lipoxygenase in vascular endothelium accelerates early atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2000; 20: 2100–2105.CrossrefMedlineGoogle Scholar
    • 145 Patricia MK, Kim JA, Harper CM, Shih PT, Berliner JA, Natarajan R, Nadler JL, Hedrick CC. Lipoxygenase products increase monocyte adhesion to human aortic endothelial cells. Arterioscler Thromb Vasc Biol. 1999; 19: 2615–2622.CrossrefMedlineGoogle Scholar
    • 146 Baynes JW, Thorpe SR. Glycoxidation and lipoxidation in atherogenesis. Free Radic Biol Med. 2000; 28: 1708–1716.CrossrefMedlineGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.