Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Circ Res. Author manuscript; available in PMC 2013 Jan 20.
Published in final edited form as:
PMCID: PMC3357004
NIHMSID: NIHMS349282
PMID: 22267839

Genetic Basis of Atherosclerosis: Insights from Mice and Humans

Abstract

Atherosclerosis is a complex and heritable disease involving multiple cell types and the interactions of many different molecular pathways. The genetic and molecular mechanisms of atherosclerosis have in part been elucidated by mouse models; at least 100 different genes have been shown to influence atherosclerosis in mice. Importantly, unbiased genome-wide association studies have recently identified a number of novel loci robustly associated with atherosclerotic coronary artery disease (CAD). Here we review the genetic data elucidated from mouse models of atherosclerosis, as well as significant associations for human CAD. Furthermore, we discuss in greater detail some of these novel human CAD loci. The combination of mouse and human genetics has the potential to identify and validate novel genes that influence atherosclerosis, some of which may be candidates for new therapeutic approaches.

Keywords: CAD, Lipids, Mice, GWAS, Genome-wide

Introduction

The process of atherosclerosis has been the topic of substantial investigation over many decades. Atherosclerosis is a complex process which has a strong heritable component, and involves a number of cellular processes and molecular mechanisms. Exploiting genetic manipulation in mice and natural genetic variation in humans has the potential to inform our understanding of the pathophysiology of atherosclerosis and identify novel targets for therapeutic intervention. In this article, we first review the data involving deletion of specific genes in mice and the effect on atherosclerosis, as well as unbiased approaches to discovery of genetic loci associated with atherosclerosis in mice. Then we review the current status of human genetic variation and its association with coronary artery disease.

Genetics of atherosclerosis in mice

The most widely used in vivo model for human atherosclerosis is currently the mouse. Wild-type mice on a chow diet are not susceptible to atherosclerosis, and this has led investigators over the years to develop three primary mouse models for examining atherosclerosis: 1; The cholesterol-cholate diet is a high fat (15%) cholesterol (1.25%) and cholate (0.5% cholic acid) based diet, known as the “Paigen Diet”, and is primarily used to induce atherosclerosis in wild-type mice. The combination of cholesterol and cholate facilitates cholesterol and fat absorption, suppresses the conversion of cholesterol to bile acid, which reduces cholesterol clearance, resulting in increased plasma cholesterol.1 The cholate also affects gene expression of lipid, lipoprotein, and inflammatory genes, and the net result is increased atherosclerosis in mice.2,3 However, lesion sizes in mice maintained solely on the Paigen Diet remain small,4 and therefore this strategy has not been as extensively employed as the other two models. 2; Low density lipoprotein receptor knockout (KO) mice (Ldlr−/−) lack the primary receptor for LDL cholesterol uptake, causing a modest increase in plasma LDL cholesterol levels,5 however to induce atherogenesis Ldlr−/− mice must have their plasma cholesterol levels increased further through the feeding of a “Western type diet (WTD)”6 or crossing with another model such as the Apobec1 knockout mouse. 3; Apolipoprotein-E KO mice (Apoe−/−) lack the primary lipoprotein required for the uptake of lipoproteins through the hepatic receptors, leading to even greater plasma cholesterol increase than the Ldlr−/− mice, and consequently atherosclerosis can be observed in Apoe−/− mice maintained on a regular chow diet, with advanced disease observed when the mice are maintained on a WTD (Figure 1A).5,7,8

An external file that holds a picture, illustration, etc.
Object name is nihms349282f1.jpg
Phenotypic Progression of Atherosclerosis in mouse and humans

[A] Approximate progression of observable atherosclerosis in mouse models, noting that double knockouts (DKOs) on either the Ldlr−/− or Apoe−/− backgrounds can have decrease as well as increased atherosclerosis. [B] An approximate illustration of clinical characterization of atherosclerosis in humans.

Candidate gene approaches in mice

In a candidate gene loss-of-function approach, a KO mouse model for the specific gene is interrogated using the models outlined above. If the Paigen diet is used, no backcrossing is required, however generally double KO (dKO) mice are generated by crossing the candidate KO with either Ldlr−/− or Apoe−/− mice. Using these approaches, approximately 100 genes have been reported to robustly and significantly affect atherosclerosis in mice with at a significance threshold of p<0.01. Of these 100 genes, approximately 60% of initial studies were performed using the Apoe−/− background, 25% using the Ldlr−/−, 5% tested both backgrounds, and 10% using the Paigen diet. Of the 100 genes, deletions of 65 were shown to decrease atherosclerosis, while deletion of 35 increased atherosclerosis. The 100 genes generally fall into two categories, those that affect atherosclerosis by influencing plasma lipids (n=33, Table 1), and those that do not substantially influence plasma lipids and therefore presumably work through other pathways (n=67, Table 2).

Table 1

Genes affecting atherosclerosis in mice primarily via influencing plasma cholesterol levels1

Effect in the dKO on:
ChrMbCandidate
Gene 2
BackgroundAtheroscleroticPlasma
Cholesterol 3
Ref
171.63Fn1Apoe−/−DecreaseDecrease99
1132.95Mapkapk2Ldlr−/−DecreaseIncrease100
1158.36Soat1Apoe−/− and Ldlr−/−DecreaseDecrease101
1163.33Tnfsf4Ldlr−/−DecreaseIncrease31
1172.98Fcgr3Ldlr−/−DecreaseIncrease102

284.98AplnrApoe−/−DecreaseIncrease103
2126.42HdcApoe−/− with Paigen dietDecreaseIncrease104
2148.70Cst3Apoe−/−IncreaseIncrease105

310.01Fabp5Apoe−/−DecreaseDecrease106
310.20Fabp4Apoe−/−DecreaseDecrease107
396.63Pdzk1Apoe−/−IncreaseIncrease108
3107.54Csf1Apoe−/−DecreaseIncrease109

453.04Abca1Apoe−/− and Ldlr−/−Increase4Decrease110
498.69Angptl3Apoe−/−DecreaseDecrease111

517.29Cd36Apoe−/−Decrease5Decrease112
530.34Il6Apoe−/−IncreaseIncrease113
5125.76Scarb1Apoe−/−IncreaseIncrease114

65.21Pon2Paigen dietIncreaseDecrease115
617.26Cav1Apoe−/−DecreaseIncrease116
629.01LepLdlr−/−IncreaseIncrease117
6122.53Apobec1Ldlr−/−IncreaseIncrease118
6129.44Olr1Ldlr−/−DecreaseIncrease119

840.69Msr1Apoe−/−DecreaseIncrease120
8108.46LcatApoe−/− and Ldlr−/−DecreaseDecrease121

946.04Apoa1Ldlr−/−Decrease6Decrease122
968.50RoraPaigen dietIncreaseDecrease123
970.64LipcApoe−/−DecreaseIncrease124

1019.31Ifngr1Apoe−/−DecreaseIncrease125
1088.92Nr1h4Apoe−/−IncreaseIncrease126

15101.98Soat2Apoe−/−DecreaseIncrease127

1733.91Angptl4Apoe−/−DecreaseDecrease128

1875.10LipgApoe−/−DecreaseIncrease129

1934.37FasApoe−/−IncreaseDecrease130
1Only studies with a significant difference in atherosclerosis with a minimum significant level of p<0.01 are given when compared to controls. A number of gene names and symbols have changed several time and we have attempted to use the most up to date gene symbol according to the Mouse Genome Database (MGD) (http://www.informatics.jax.org).
2Full gene names can be obtained from the MGD. Multiple genes have been tested in both Apoe−/− and Ldlr−/−, we report the earliest study to show significance and in several of these studies both backgrounds were examined. Genes were identified by interrogating the MGD and NCBI.
3The threshold for a significant difference in plasma cholesterol was set to p<0.05.
4No changes in whole dKO mice, only in bone macrophage derived transplant experiments from the dKO mice.
5Large significant decrease in VLDL-TG.
6Female specific effect. Chr, chromosome, Mb, millions of bases from the acromere.

Table 2

Genes affecting atherosclerosis in mice without affecting plasma cholesterol levels

ChrMbCandidate
Gene1
BackgroundAtherosclerosis
in the dKO
Ref
140.28Il1r1Apoe−/− with Paigen dietDecrease 2131
1132.92Il10Ldlr−/−Increase132
1166.04SelpApoe−/−Decrease133

225.28Fut7Apoe−/−Decrease134
275.51Nfe2l2Apoe−/−Decrease135
2164.77Mmp9Apoe−/−Increase136

327.08Nceh1Apoe−/−Increase137
386.80Cd1d1Apoe−/−Decrease 2138
387.16Cd5lLdlr−/−Decrease139
395.30CtssLdlr−/−Decrease140
396.84Gja5Apoe−/−Increase 2141
3115.81Vcam1Ldlr−/−Decrease14
3151.46PtgfrLdlr−/−Decrease142

419.94TtpaApoe−/−Increase143
466.49Tlr4Apoe−/−Decrease144

523.87Nos3Apoe−/−Increase145
591.32Cxcl1Ldlr−/−Decrease146
592.78Cxcl10Apoe−/−Decrease147
5104.86Spp1Apoe−/−Decrease148
5112.77Hps4Paigen dietDecrease 2149
5134.70Ncf1Apoe−/−Decrease150

65.12Pon1Paigen dietIncrease151
6116.36Alox5Ldlr−/− and Paigen dietDecrease26
6125.50VwfLdlr−/−Decrease152

754.02Hps5Paigen dietDecrease 2149

823.76IkbkbLdlr−/−Increase153
846.48Tlr3Apoe−/−Increase 2154
897.30Cx3cl1Apoe−/−Decrease155
8108.04Hsd11b2Apoe−/−Increase156
8108.67Pla2g15Apoe−/−Increase157

97.34Mmp12Apoe−/−Decrease136
97.45Mmp3Apoe−/−Increase136
914.55Fut4Apoe−/−Decrease134
920.82Icam1Apoe−/−Decrease13
9108.24Gpx1Apoe−/−Increase158
9119.96Cx3cr1Apoe−/−Decrease159
9124.02Ccr2Apoe−/−Decrease160

1019.65BaxApoe−/− with Paigen dietIncrease161
10117.88IfngLdlr−/−Decrease162
10126.97Lrp1Ldlr−/−Increase 3163

1149.66Mapk9Apoe−/−Decrease164
1170.16Alox15Apoe−/−Decrease165
1170.27Cxcl16Ldlr−/−Increase166
1170.47Pfn1 (+/−)Ldlr−/−Decrease167
1178.73Nos2Apoe−/−Decrease168
1181.85Ccl2Ldlr−/−Decrease169
1187.61MpoLdlr−/−Increase170
1196.96Tbx21Ldlr−/−Decrease171
11104.47Itgb3Apoe−/− and Ldlr−/−Decrease172
11105.82Ace (+/−)Apoe−/−Decrease173
11106.52Pecam1Ldlr−/−Increase174

1216.54Lipin1Paigen dietIncrease175
1232.86Pik3cgApoe−/−Decrease176
12114.09Gpr132Ldlr−/−Decrease177

1447.99Lgals3Apoe−/−Decrease178
1456.38Ltb4r1Apoe−/−Decrease179
1466.59CluApoe−/−Decrease180

154.68C6Apoe−/−Decrease 234
1554.08Tnfrsf11bApoe−/−Increase181

1617.33Serpind1Apoe−/−Increase182

1712.57PlgApoe−/−Increase183
1729.23Cdkn1aApoe−/−Decrease184
1735.82Ddr1Ldlr−/−Decrease185

1835.02Egr1Apoe−/−Decrease186

1942.83Hps1Paigen dietIncrease 2149
1946.08Hps6Paigen dietDecrease 2149
X160.58Ace2Apoe−/−Increase187

See Table 1 for extended legend.

1Candidate genes are homozygous KO unless otherwise indicates by as heterozygous by ‘(+/−)’.
2Lipid data not reported.
3Conditional KO.

In a candidate gene gain-of-function approach, the generation of over-expression transgenic mice in the Apoe−/− or Ldlr−/− background has been applied. This approach has several advantages; it allows the examination of genes when knockouts are embryonic lethal, and it allows for the examination of human specific genes, such as cholesteryl ester transfer protein (CETP), which does not have a mouse ortholog.9 Additional methods for probing the role of specific genes in mouse atherosclerosis include bone-marrow transplantation to show the effects on atherosclerosis from hematopoetic-expressed genes (such as with Abcg110) antibody-inhibition studies,11 and small molecule pharmacologic studies12, however those studies are not reviewed here in the interest of space.

Many genes that were tested have no or minimal effect on atherosclerosis, or have not been replicated. On several occasions, genes have been reported to have an effect only under certain conditions, such as in the Icam1−/− mouse studies where atherosclerosis is affected on the Apoe−/− background,13 but not in the Ldlr−/− background.14

Unbiased mapping approaches in mice

In addition to testing specific genes for their effects on atherosclerosis, mice have also been used to identify atherosclerosis genes using unbiased mapping approaches specifically through the use of quantitative trait locus (QTL) analysis. Thus far, 43 significant mouse QTL have been mapped in mice for atherosclerosis (Table 3).15 QTL mapping typically involving an F2 generation from two progenitor strains still requires a pro-atherogenic model in order to map atherosclerosis loci. Of the 43 loci identified, 19 have been mapped using progenitor strains that had the Apoe−/− gene introduced, 12 were mapped using strains with Ldlr−/−, and an additional 12 QTL were mapped simply by maintaining the F2 mapping population on the Paigen diet.

Table 3

Mouse atherosclerosis quantitative trait loci (QTL) 1

ChrPeak location or
interval Chr (Mb)2
Model3QTL name 4RefProven GeneRef
1Paigen dietAth1188Tnfsf431
1Apoe−/−Ath30189
1105.4Apoe−/−QTL-#190
1166.3Apoe−/−QTL-#190
1176.6Apoe−/−Ath9191

2Paigen dietAth2192
2Apoe−/−Ath28193
2148.5Ldlr−/−Athla1194

3Apoe−/−Ath23193
3115.5Ldlr−/−Ascla434
3147.8Ldlr−/−Ascla334

4137.4 – 150.9Ldlr−/−Athsq1195
445.7 – 105.0Paigen diet, Apoe−/−Ath8196,197

5Apoe−/−Ath24193

6Ldlr−/−Artles25,26Alox529
6Ldlr−/−Ath3825
6Ldlr−/−Athsq2195
6116.6 – 128.5Ldlr−/−Ath3729

757.0Paigen dietAorls2198
7Paigen dietAth3199
7Apoe−/−Ath31189

965.3Apoe−/−Ath29189,197,200

1082.1Paigen dietAorls1198,201
10Apoe−/−Ath11191
1020.1Paigen dietAth20201
1067.7Paigen dietAth17201
1018.5Ldlr−/−Ascla234
1021.4Ldlr−/−Ascla134

11108.6Paigen dietAth19201

12Paigen dietAth21201
12Paigen dietAth6202204
1235.9Paigen dietAth18201
1214.3Ldlr−/−Ascla534Adam1733
1255.1Ldlr−/−Ascla634
12Apoe−/−Cath1205

13Apoe−/−Ath25193
13Apoe−/−Ath32189

1451.9 – 165.9Apoe−/−Ath13191

15Apoe−/−Ath22193
15Apoe−/−Ath33189

17Apoe−/−Ath26193, 189

18Apoe−/−Ath27193

1938.5Apoe−/−Ath16191
1Data sourced from the Mouse Genome Database (MGD) (http://www.informatics.jax.org).
2If positions are not listed this indicates they were not reported in the publication.
3QTL crosses performed with different progenitor inbred strains which may have had Ldlr−/− or Apoe−/− introduced into the backgrounds of the strains.
4QTL-# indicates that no QTL name has yet been assigned. Chr Chromosome, Mb millions of base pairs from acromere.

The nature of QTL analysis makes identifying causal genes extremely difficult, as the mapped loci (confidence intervals) typically include hundreds of genes with dozens of plausible candidates, even after applying advanced mapping strategies and bioinformatics.1624 Consequently plausible candidates for just a small number of atherosclerosis QTL have been successfully mapped. Artles1 was the first atherosclerosis QTL to be positionally cloned as the gene arachidonate 5-lipoxygenase (Alox5),25 and the atherosclerosis phenotype was confirmed in Alox5−/− mice,26 although other investigators did not see the same effect.27 Historically, many KO mice have been generated with the use of embryonic stem (ES) cells derived from the “129” strain, subsequently backcrossed to the more standardized “C57BL/6 (B6)” strain. However, no matter how many backcrosses are performed to B6, a significant proportion surrounding the locus of the KO gene will continue to remain from the 129 strain due to linkage, the so called “passenger gene region”.28 These genes will contain polymorphisms between the ES donor strain and the backcrossed recipient strain, and could affect the phenotype being examined leading to a false conclusion that the knocked-out gene is solely responsible for the QTL effect.28,29 As we summarize below, ALOX5 has not been convincingly proven in human genetic studies to be associated with CAD, however, CXCL12 which is significantly associated with human CAD, maps to the orthologous mouse chromosome (Chr) 6 at 117.1 million bases (Mb) from the acromere, less than 1Mb away from Alox5 (116.3Mb), and within the so called “passenger gene” region of the Alox5−/− strain. Consequently, polymorphisms between 129 alleles in Cxcl12 and B6 control mice may be a significant contributor to the Artles1 QTL.

A causal gene was also reported for Ath1, one of the first atherosclerosis QTL ever mapped in mice, which is located on distal mouse Chr1.30 The gene was identified as tumor necrosis factor (ligand) superfamily member 4 (Tnfsf4, also known as Ox40l). Tnfsf4−/− mice showed decreased atherosclerosis, and two human populations that were examined reported SNPs having marginal significance for MI (p<0.05).31 However, other investigators did not observe the significance of those SNPs to MI in their human populations,32 and as presented later, there is currently no genome-wide association locus reported near the human ortholog TNFSF4 at the syntenic human location (Chr1 at 173Mb). A third QTL, a disintegrin and metalloproteinase 17 (ADAM17) was proposed as the causal gene for an atherosclerosis QTL on mouse Chr 12, however the effect in Adam17−/− has not yet been reported.33,34

Mice have proven extremely powerful in identifying genes and loci that affect atherosclerosis. More than 100 gene deletion mouse models have revealed detailed biological processes leading to atherosclerosis, crucial insights into the nature of specific causal mechanisms affected by lipid metabolism, inflammation, and the pathogenesis of atherosclerosis itself. It is also interesting to note that many of these genes have shown large effect sizes, typically 20 – 50%. It is therefore likely that deletion of more than one of these genes would not have an additive effect. It is possible that many of the genes act in common pathways leading to atherosclerosis. These genes may be under a large selection pressure in humans preventing null mutations from arising in the general population which may partly explain why few of these genes have been robustly shown to affect CAD in humans, but nonetheless could be relevant to human CAD pathology.

A further point that might explain the large number of mouse atherosclerosis genes that have not been convincingly validated in humans could be the differences in pathogenesis and phenotyping of atherosclerosis between mouse and human (Figure 1). The most important clinical consequences of atherosclerosis in humans arise from lesions in the coronary, carotid, and cerebral arteries, whereas the focus in mice is on the aorta and proximal great vessels. The size, structure, hydrodynamics, embryonic origin and abundance of the smooth muscle cells, and the elasticity of the arteries differ considerably between mouse and humans.35 Furthermore, while plaque disruption and thrombosis is a major cause of cardiovascular events in humans, similar events have not been convincingly demonstrated in mice. Perhaps then it is not surprising that translation of the genetic discoveries in mice to human CAD susceptibly has been slow and unconvincing; but this has not necessarily been due solely to the limitations in the mouse model.

Human genetics has until recently been considerably underpowered to identify significant genetic associations with CAD. Indeed, as human studies continue to increase in power, more of the genes and loci identified in the mouse may be replicated in humans. Nonetheless, recent human genetic susceptibility loci, as we discuss below, have been identified in genes and loci that could not have been extrapolated from the accumulated knowledge of these mouse studies. This leads to the conclusion that current mouse model strategies for studying atherosclerosis, relevant to human genetics and biology, may need to be revised.

Genetics of atherosclerosis in humans

The relationship of genetic variation to atherosclerotic cardiovascular disease in humans has the potential to provide important information on the pathophysiology of atherosclerosis in humans. However, the ability to identify causal genes in human studies has until recently been limited by low sample sizes, and heterogeneity in predictive subclinical phenotypes (Figure 1B).36,37 The expectation is that there would be numerous genes underlying the susceptibility to atherosclerosis in the general population, and that each would have small effects, similar to all complex traits.38 There have been hundreds of candidate gene studies and multiple linkage studies. While some of these studies identified “significant” genetic associations with particular genes and polymorphisms, numerous other studies failed to replicate those associations, and specific replication studies have showed that most of those loci could not be replicated.39 More recently genome-wide association studies (GWAS) has unearthed multiple loci significantly associated with CAD, many of which have been replicated numerous times independently and across ethnic groups. Here, we briefly review the candidate gene and human linkage studies, and then focus primarily on the new developments related to GWAS of CAD.

Candidate gene studies in humans

Using the candidate gene approach, polymorphisms in specific genes with plausible biological relevance are genotyped to determine association with CAD. The candidate gene approach has revealed some loci with significant and convincing effects on atherosclerosis.40,41 However, overall the traditional candidate gene approach has yielded a slew of negative studies, or weak associations that could not be replicated. The candidate gene approach for CAD took a more high-throughput approach several years ago with the development of the cardiovascular gene-centric 50K SNP array.42 This array was designed specifically to examine variants in CAD candidate genes, and a recent meta-analysis involving > 30,000 cases and > 75,000 controls revealed several new genes highly significantly associated with CAD.

Linkage studies

Although traditional risk factors such as age, gender, and family history are still better predictors of CAD than genetic markers and genetic scores, twin-studies demonstrate a strong heritable component of genetic susceptibility to atherosclerosis. In rare instances atherosclerosis can appear as a Mendelian disorder, such as in familial hypercholesterolemia (FH), where mutations in the LDL-receptor significantly increase plasma LDL cholesterol and consequently atherosclerosis susceptibility.43 However, even in FH, there is wide a variation in atherosclerosis. Family based linkage studies have been deployed to identify atherosclerosis loci in an unbiased approach, and such linkage studies can be applied in two general ways; by examining multiple large families in a QTL type approach, or by examining a single very large family under a Mendelian inheritance hypothesis. As with the mouse QTL mapping, the linkage regions using either approach are typically too large to identify plausible causal genes.44 More than 40 loci have been identified using human linkage studies for different measures of human CAD (MI, acute coronary syndrome, coronary calcification, carotid intimal-medial thickness and peripheral arterial occlusive disease), although very few of the loci have been replicated between studies. In several instances, strong candidate genes were identified including; lipoprotein receptor-related protein 6 (LRP6) at 12q13.2,45 arachidonate 5-lipoxygenase-activating protein (ALOX5AP) at 13q12-13,46 and myocyte enhancer factor 2A (MEF2A) at 15q26.3. 47 Atherosclerosis screening in mouse models has not yet been reported for these genes, and the new GWAS loci discussed later map outside of the original confidence intervals which identified LRP6 and ALOX5AP, indicating these may be distinct loci. MEF2A is more controversial than the other two genes,44,48 and maps just 20Mb away from ADAMTS7 (discussed below) on chromosome 15 which may represent linkage of MEF2A to a broader region that includes ADAMTS7. Like mouse QTL analysis, family based linkage studies have been very successful in identifying loci, but have yielded a limited a number of convincing genes that affect atherosclerosis.

Genome-wide association studies (GWAS)

Human genome wide association studies (GWAS) examine millions of polymorphisms simultaneously to examine association to a phenotype in a large unrelated but ethnically similar population, and the phenotype is either treated as a continuous variable, or a case-control binary population comparison. As there are a large number of tests performed simultaneously, the currently accepted threshold for genome-wide significance is p<5×10−8.49 For GWAS of CAD the case-control approach has been the most commonly reported, and this has yielded multiple loci with highly significant associations to myocardial infarction (MI) and coronary artery disease (CAD) (Table 4). In 2007, this approach successfully identified a locus at chromosome 9p21.3 that was highly associated with risk of MI and CAD.5052 Concurrently, the Wellcome Trust Case Control Consortium (WTCCC)53 reported seven additional novel genome-wide significant loci for MI. Further studies and meta-analyses have added additional significant loci.50,5458 The most recent and extensive findings come from two global consortia, CARDIoGRAM59 and C4D,60 and bring the total number of genome-wide significant loci for MI/CAD to 34 (Table 4). The CARDIoGRAM consortium pooled the data from 14 different GWAS for a total of 140,000 patients, and reported 13 new loci that are genome-wide significant for CAD, while the C4D consortium examined 70,000 patients focusing on South Asians and Europeans and identified four further loci. Beyond the 34 current loci, there are many hundreds of other SNPs that are nominally significant (5×10−8>p<0.05). These may harbor important genes that will reach significance in larger meta-analyzes, and further information gained from expression-QTL and bioinformatics 16 may provide reason to investigate some of these nominally significant loci more urgently. Information from many of the genes derived from mouse atherosclerosis studies that have nominally significant human associations to CAD may also provide impetus to investigate those genes further.

Table 4

Genome wide significantly associated CAD loci in humans (p<5×10−8)

#SNPBandMbGenetic elements in region (+/− 50,000 from reported SNP)Most likely
Causal gene
Related
Associations
1rs112065101p32.355.50TMEM61, RP11-12C17.2, BSND, PCSK9, USP24PCSK9Lipids
2rs171140361p32.256.96PPAP2B, RP11-213P13.1PPAP2B
3rs5998391p13.3109.82SARS, CELSR2, PSRC1, MYBPHL, SORT1SORT1Lipids
4rs174656371q41222.82MIA3, RP11-378J18.6, RP11-378J18.5, AIDA
5rs4299376*2p2144.07DYNC2LI1, ABCG5, ABCG8, LRPPRCABCG5/8Lipids
6rs67258872q33.1203.75ICA1L, AC010900.1, WDR12, ALS2CR8, AC010900.2
7rs23063743q22.3138.12MRAS, ESYT3
8rs2706399*5q31.131.87IRF1, IL5, RAD50IL5
9rs125264536p24.112.93PHACTR1PHACTR1
10rs69039566p24.111.77C6orf105C6orf105
11rs176099406p21.3135.03ANKS1A, RP11-527B17.2ANKS1A
12rs121902876q23.2134.21RP3-323P13.2, RP4-662A9.2, TCF21TCF21
13rs37982206q25.3160.96LPAL2,AL591069.1, LPALPALipids
14rs109535417q22.3107.24DUS4L, BCAP29
15rs115569247q32.2129.66RP11-306G20.1, ZC3HC1, 5S_rRNA.114, KLHDC10
16rs17321515*8q24.13126.49TRIB1, RP11-136O12.2TRIB1Lipids
17rs49775749p21.322.10CDKN2A,CDKN2BCDKN2A,CDKN2BGlaucoma, Diabetes
18rs70254869q33124.42DAB2IPDAB2IP
19rs5794599q34.2136.15ABO, Y_RNA.342, LCN1P2, SURF6ABOLipids
20rs250508310p11.2330.34KIAA1462KIAA1462
21rs174604810q11.2144.78RP11-20J15.2, RP11-20J15.3CXCL12Inflammatory diseases
22rs141244410q23.3191.00CH25H, LIPALIPA
23rs1241340910q24.32104.72CNNM2, AL356608.1BP
24rs97481911q22.3103.66RP11-563P16.1, PDGFDPDGFD
25rs96418411q23.3116.65BUD13, AP006216.10, AP006216.11, ZNF259, APOA5, AP006216.5, APOA4, APOA1APOA5-A4-C3-A1Lipids
26rs318450412q24.12111.88SH2B3, ATXN2CKD, BP, Diabetes
27rs477314413q34110.96COL4A1, AL161773.1, COL4A2
28rs289581114q32.2100.13AL160313.1, HHIPL1, CYP46A1
29rs382580715q25.179.09AC022748.1, ADAMTS7, MORF4L1ADAMTS7
30rs21617217p13.32.13SMG6, AC090617.1, AC130689.5, AL450226.2
31rs1293658717p11.217.54PEMT, SMCR2, RAI1PEMT
32rs4652217q21.3246.99CALCOCO2, RP11-463M16.4, SNORA68.3, RP11-463M16.5, ATP5G1, UBE2Z
33rs112260819p13.211.16SMARCA4, AC011442.1, LDLRLDLRLipids
34rs998260121q22.1135.60AP000318.2, C21orf82

SNP: this indicates the most commonly reported SNP for the locus, however there may be multiple genome-wide significant SNPs in the locus in independent linkage disequilibrium spanning thousands of bases. Band: Chromosome, arm and chromatin band of the locus.

*SNP associations made in the IBC 50K SNP array where a lower threshold is required for genome-wide significance due to fewer tests being performed (p<1×10−6). Mb: indicates the location of the reported SNP in millions of bases from the proximal end of the chromosome, assembly: NCBIM37 2011. “Related Associations” indicates that the locus, typical the same SNP listed above, has been reported as genome-wide significant for a phenotype known to affect CAD. BP indicates blood pressure, CKD indicates chronic kidney disease.

Several of the CAD loci are significantly associated with LDL cholesterol levels (Table 4), while three quarters of all the CAD loci are completely novel and were not previously suspected to be involved in atherosclerosis or MI. Indeed the majority of the loci have not even been shown to be significantly associated with other CAD risk factors such as diabetes mellitus, hypertension, or BMI. Conversely, many lipid genes, inflammatory genes, and genes previously shown to affect other CAD risk factors have not yet been ‘validated’ by unbiased human GWAS studies, at least by the high bar of being genome-wide significant.42 Notably, several novel CAD loci are associated with lipids (SORT1, TRIB1, and LPA) and others are related to blood pressure and hypertension,61 suggesting that these risk factors lie in the causal pathway for these particular genes. However, it appears highly likely that the majority of CAD loci will remain unassociated with known risk factors. Furthermore, little is known about the biology of most of the genes at these loci and the mechanisms by which they may influence atherosclerosis or MI.

Examples of novel CAD loci identified by GWAS and implications for pathophysiology of atherosclerosis

9p21(CDKN2A-B/ANRIL/MTAP)

The 9p21 locus was the first CAD locus discovered using GWAS, and has been reliably reproduced, remaining the strongest association with CAD in the human genome.54 This locus has long been known to be involved in cancer susceptibility,62 however there are no associations with known risk factors of CAD such as hyperlipidemia or hypertension.56 The SNPs associated with CAD are all located in intergenic locations, rather than in coding or gene regulatory regions such as un-translated regions (UTRs) or promoter regions (Figure 2A). Consequently, identifying and characterizing the causal variant and gene has been challenging. Nonetheless, several studies have begun to characterize the functional aspects of this locus, focusing on the nearest genes: cyclin-dependent kinase inhibitor 2A (CDKN2A), the homologue CDKN2B,63,64 and the neighboring non-coding transcripts.

An external file that holds a picture, illustration, etc.
Object name is nihms349282f2.jpg
The 9p21 human CAD locus and the orthologous mouse region

Horizontal lines indicate the full contiguous length of the labeled transcript. [A] 400,000 bases near the 9p21 CAD locus. The “CAD risk interval” indicates the region containing the most GWSA SNPs for CAD. [B] 400,000 bases surrounding the orthologous mouse region of the CAD locus on human 9p21. A recent update of the mouse genome at this locus (Assembly NCBIM37, Gene build Jan 2011) indicates that there is a region lacking overlapping sequenced contigs (designated “Unknown sequence”) which may be as large as 50,000 bases. The sequenced contigs flanking this region are themselves small and may indicate a region of excessive repetitive DNA preventing the ability to confidently sequence the region.

The two primary genes CDKN2A and 2B are present on the same strand, and encode several different splice variants; CDKN2B encodes the p15 protein, while CDKN2A encodes the p16 protein as well as the p14/ARF protein (p19/ARK in mice), although due to alternative splicing p16 and p14 share no homology at the amino acid level.65 More recently, an additional antisense (relative to CDKN2B) non-coding RNA was identified and termed ANRIL. The first exon of ANRIL is in the promoter of p14 (CDKN2A) and overlaps with two exons of p15 (CDKN2B), and is now referred to as CDKN2B-antisense (CDKN2B-AS).66 Due to the increased attention at this locus, additional non-coding antisense transcripts on the ‘+ strand’ have been identified that overlap with these sequences. Furthermore the gene coding for methylthioadenosine phosphorylase (MTAP) has been shown to have multiple alternative transcripts, both coding and non-coding. Two of the non-coding MTAP transcripts have shared exons with ANRIL. Finally, there is also a new small protein-coding gene called C9ORF53, which encodes a 79 amino acid protein and remains largely uncharacterized.

Although it is still unclear which of the genes at this locus is specifically responsible for the CAD association and what the mechanism may be, functional studies have implicated enhancer activity for the neighboring genes.63,64 One study examining mRNA expression of the genes at the locus from peripheral blood mononuclear cells, with follow up in a much smaller sample size of atherosclerosis plaque tissue, suggested that transcript levels of ANRIL (CDKN2B-AS), but not CDKN2A, 2B, C9ORF53 and MTAP are associated with atherosclerosis.67 However, CDKN2B-AS/ANRIL has not yet been identified in mice which will make in vivo characterization of such an effect difficult. Indeed, mice and humans differ significantly at this region for two reasons, 1) there are significantly more transcripts identified for all the genes in the region in humans compared to mice, and 2) much of the region that is significantly associated with CAD in humans is either missing, or is not yet convincingly sequenced in mice (Figure 2B).

Despite this, a mouse strain with a targeted deletion of 70kb in the orthologous region on mouse Chr 4, where CDKN2B-AS/ANRIL should be (Chr4Δ70kb/Δ70kb), significantly affects cardiac expression of the neighboring genes and the proliferation properties of vascular cells. Cardiac expression of Cdkn2a and Cdkn2b is reduced in Chr4Δ70kb/Δ70kb mice, indicating that the locus may be a long range regulator for genes that affect CAD through cardiac and vascular cell proliferation, possibly via CDKN2A and CDKN2B.64 Studies using human cells of defined genotype showed that lymphoblastoid cells homozygous for the ‘protective’ haplotype showed binding of STAT1 to an enhancer in this region whereas cells homozygous for the ‘risk’ haplotype had no STAT1 binding. Interferon-γ stimulation and STAT1 binding was shown to regulate CDKN2B and CDKN2B-AS expression in human vascular endothelial cells. Thus genetic variation within enhancers at the 9p21 locus may influence vascular cells responses to inflammatory stimuli, resulting in effects on atherogenesis.68

ADAMTS7

The ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family of proteases is comprised of 19 discrete proteins that are zinc metalloproteases. These proteins are secreted and interact with, and degrade components of, the extracellular matrix. Some ADAMTS family members have partially defined biological functions. ADAMTS13 is the protease that cleaves von Willebrand factor, and mutations in ADAMTS13 cause thrombotic thrombocytopenic purpura. ADAMTS2, 3, and 14 regulate the maturation of procollagen into mature collagen via cleavage of an N-terminal preprotein region, and thus these proteins are collectively known as the procollagen-n-proteinases.69 Finally, many other ADAMTS proteins are known to bind and cleave numerous proteoglycans such as aggrecan, versican, and brevican.70

There are multiple lines of evidence linking the ADAMTS family of metalloproteases with the pathogenesis of atherosclerosis. The observation that multiple ADAMTS (ADAMTS1, 4, and 9) proteins can cleave versican, a proteoglycan both highly expressed in regions of the vasculature prone to plaque formation71 and associated with adult cardiovascular disease,72 suggests a role for these proteins in plaque formation. Multiple ADAMTS proteins are expressed in human carotid lesions.73 Specifically, ADAMTS1 localizes with vascular smooth muscle cells (VSMCs) within lesions and its expression is higher in migrating and proliferating VSMCs.74 ADAMTS4 localizes with macrophages in atherosclerotic lesions, and demonstrates higher levels of expression during plaque development in atherosclerotic mouse models.73 Additionally, a SNP in ADAMTS1 has been associated with a nearly two-fold increase in cardiovascular disease.75

In a recent GWAS meta-analysis involving patients with (n=12,393) and without (n=7,383) CAD, as ascertained by coronary angiographic phenotyping, the SNP most highly associated with CAD lies in an intron of the ADAMTS7 gene (p-value=5.0×1013).76 This finding was replicated by the CARDIoGRAM consortium.55 The linkage disequilibrium block that this SNP lies in is completely encompassed by the ADAMTS7 coding region, strongly implicating ADAMTS7 as the causal gene. ADAMTS7 can bind and cleave the cartilage oligomeric matrix protein (COMP), an interaction thought to play a role in the progression of arthritis.77 More recently, ADAMTS7 over-expression was shown to increase neointimal formation post-injury in a rat carotid artery balloon-injury model.78 Subsequent knock down of ADAMTS7 expression by topical application of siRNAs decreased post-injury neointimal formation. Over- and under-expression of ADAMTS7 also increased and decreased primary VSMC migration, respectively,78 while exogenous expression of COMP decreased both post-injury neointimal formation as well as primary VSMC migration. The statistical analyses in human cohorts combined with in vivo model observations suggest that ADAMTS7 is likely to play a causal role in atherogenesis, presumably through enhanced VSMC infiltration and migration in plaques. These results also make ADAMTS7 an exciting target for future studies and potential therapeutic intervention.

CXCL12 (chemokine (C-X-C motif) ligand 12)

Most of the CAD GWAS loci do not harbor classical inflammatory genes (Table 4). One exception is the 10q11 locus near the gene CXCL12, previously known as stromal cell derived factor 1 (SDF-1). The most significant SNPs at this locus are in fact in a gene desert located between two non-coding uncharacterized transcripts with CXCL12 being the nearest gene located ~100,000 bases away. CXCL12 is a complex gene with two common splice variants (α and β), and up to six additional coding variants. CXCL12 is known to have a major role in angiogenesis and is a key regulator of endothelial cell responses, as well as having anti-inflammatory properties by modulating the accumulation of leukocytes and bone marrow derived stem cells in inflamed tissue.79

Additional evidence that CXCL12 is the causal gene for atherosclerosis at this locus was recently reported in a clinical study examining two CAD populations. The risk allele for the SNP (rs1746048) was shown to be associated with increased atherosclerosis and higher CXCL12 mRNA transcript levels, particularly for the α-isoform in human liver and natural killer cells.80 In addition, the risk allele was reported to confer increased plasma protein levels of CXCL12,80 indicating that higher plasma CXCL12 might result in increased atherosclerosis. However, in a smaller human study, CXCL12 levels were lower in subjects with unstable angina.81 Additionally, knock-out mouse data (Cxcl12−/−) indicates that deficiency of CXCL12 is likely to be detrimental to health as it is required for B-cell lymphopoiesis, myelopoiesis, correct formation of the ventricular septum,82,83 and T-cell development.84 The Cxcl12-α variant in mice has also been shown to confer protection against myocardial ischemia/reperfusion injury.85 Additional studies to over-express Cxcl12 in mice would be a useful in clarifying the biological effects of increased CXCL12 but it is possible that differential tissue specific roles may be involved, that the mouse response may differ from human, and that acute vascular injury models may differ from chronic atherosclerosis models.

ABO (ABO blood group, transferase A, alpha 1–3-N-acetylgalactosaminyltransferase, transferase B, alpha 1–3-galactosyltransferase)

The ABO blood groups were associated with plasma cholesterol levels by the Framingham Study more than 35 years ago.86 The same study linked ABO blood groups with Intermittent claudication86 and various other studies at that time also drew links to ischemic heart disease,87,88 atherosclerosis obliterans,87,89 and venous thromboembolism.87,88 Despite these early significant discoveries, the mechanism regarding how different ABO groups affect lipids and cardiovascular disease remain unknown. GWAS studies have recently ‘rediscovered’ the ABO locus to be both genome-wide significant for LDL-C cholesterol,90 as well as with myocardial infarction (MI), whereby the O blood group affords protections from MI in patients with angiographic CAD.76 The specific mechanisms by which the ABO gene locus influences multiple cardiovascular risk factors and MI remain unclear. One reason for the lack of progress is that studying glycoconjugates is considerably more difficult than simply studying nucleic acids or proteins,91 and because there is a distinct lack of an appropriate in vivo animal and specifically mouse models for the human ABO locus.92

Conclusions & Summary

Our review of the mouse model literature reveals a large list of genes that have been shown to affect atherosclerosis in mice. However, very few of these have shown up in human genetic studies to be genome-wide significantly associated with CAD, although some of these mouse atherosclerosis genes may become significant with larger human genetic studies. The gene-centric cardiovascular genotyping array (IBC 50K CAD Consortium) was designed to include many CAD candidate genes that arose from mouse studies, and this approach has been successful in adding several unique associations to the current list of 34 genome-wide significant loci.42 Targeting more of the genes identified from mouse studies through gene-centric approaches and re-sequencing could identify yet more genes that affect human CAD. Nonetheless, currently there remains substantial “missing genetic heritability” for human CAD.93

Mouse QTL for atherosclerosis may have some overlap with the recent human GWAS loci; however most of these QTL remain large and even with novel bioinformatic approaches identifying candidate genes from these QTL remains difficult. Denser genotyping in mouse crosses provides little extra power due to the relatedness of the strains and the limited recombination events. The future ‘Collaborative Cross’, which is generating hundreds of densely genotyped recombinant-inbred strains, may help generate more finite QTL intervals, although this may prove difficult for atherosclerosis given that if the new strains are not prone to atherosclerosis each will have to be crossed to Apoe−/− or Ldlr−/− strains.9496

It is also possible that some of the associations of these genes with murine atherosclerosis are false positives, arising due to variants in nearby causal “passenger genes”, or to biology which is specific to the common mouse atherosclerosis models, predominantly Apoe−/− and Ldlr−/− background mice. Indeed the limitations of mouse models may be much more substantial and pernicious than previously recognized. It is apparent from recent human GWAS that there is almost no overlap with the rodent KO models, and that the rodent QTL have not been sufficiently specific to demonstrate overlap, even after at least 34 genome-wide significant loci for human CAD have been described. It is therefore important to illustrate the potential limitations of the commonly used mouse CAD models so that they can be avoided or minimized in future functional studies. The key issues include; (1) current models utilize extreme hypercholesterolemia to induce atherosclerosis – this may be a particularly non-physiological model for the study of non-lipid candidate genes which appear to be more abundant than lipid genes based on recent human GWAS. (2) there is a lack of homology between mouse and human for key genes of interest; for example, CETP is not expressed in mouse, the 9p21 locus is markedly different between human and mouse, and the ABO variants are substantially different between mouse and human.

Moving forward we can expect a more human-translational strategy with mouse studies focused on validating the recent human loci and genes. This will require reasonable genetic homology between mouse and human, but more importantly, there must be a renewed effort to apply new and improved mouse models, particularly with less extreme lipid disturbance for loci with no impact on lipids themselves.

The human heritability of CAD is estimated to be between 30–60%.97 The recent large-scale meta-analysis of human GWAS data reveals 34 significant loci, and the accumulation of these loci explains just 10% of the heritability of CAD.98 Many reasons have been provided as to why the detected human genetic variance for complex traits is so low; GWA studies only examine SNPs and neglect much of the other forms of genomic variation; deletions, insertions, inversions, duplications, copy number variants, as well as imprinting/epigenetics, epistasis (gene-by-gene and gene-by-environment), and that there is lack of statistical power to assess rare variants.93 However, even though only 10% of human CAD variation is currently explained, it is explained by mostly novel loci that have not yet been investigated. Each of these loci may contain therapeutically targetable genes that may go a long way to reducing CAD incidence, and certainly there remains a lot of work to be done to uncover the functional biology of these novel genes and the mechanisms by which they are related to atherosclerosis.

In terms of human studies, traditional risk factors such as age, sex, smoking, and a family history of premature coronary heart disease are still more predictive of CAD that human genetic markers and genetic scores. Aging in particular affects all metabolic processes and has a profound effect on atherosclerosis, and we do not yet fully understand the impact of aging at the cellular level. However, it is intriguing that some of the new GWAS loci contain genes involved in immunity and cell growth since aging is known to affect these crucial mechanisms. Future more detailed stratified meta-analyses and assessment of interaction effects may yield important gene-by-environment interactions, and help aid in deciphering the causal mechanism.

Overall, mouse models of atherosclerosis, in particular the candidate gene based approaches, have helped us discover and understand many intricacies of the disease and have provided some insight, albeit biased, into the biology of the disease. Recent human GWAS have elucidated a substantial number of novel loci and genes, indicating that we still have some way to go before we fully understand the genetic mechanisms and biological pathways causally involved in human atherosclerosis.

Acknowledgments

Sources of Funding: NHLBI and Leducq Fondation

List of nonstandard abbreviations

B6C57BL/6
C4DThe Coronary Artery Disease (C4D) Genetics Consortium Study
CARDIoGRAMThe Coronary Artery Disease Genome-Wide Replication and Meta-Analysis study
ChrChromosome
CKDChronic kidney disease.
dKODouble knock-out
ESEmbryonic stem
FHFamilial hypercholesterolemia
GWASGenome-wide association studies
GWSAGenome-wide significantly associated
KbKilo bases
MbMillion bases
MGDMouse Genome Database
NCBIM37National Center for Biotechnology Information mouse version 37
QTLQuantitative trait locus
UTRsUn-translated regions
VSMCsVascular smooth muscle cells
WTCCCWellcome Trust Case Control Consortium
WTDWestern type diet

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: None

Citations

1. Ando H, Tsuruoka S, Yamamoto H, Takamura T, Kaneko S, Fujimura A. Regulation of cholesterol 7alpha-hydroxylase mRNA expression in C57BL/6 mice fed an atherogenic diet. Atherosclerosis. 2005;178(2):265–269. [PubMed] [Google Scholar]
2. Xu G, Zhao L, Fuchs M. Differences between hepatic and biliary lipid metabolism and secretion in genetically gallstone-susceptible and gallstone-resistant mice. Chin Med J (Engl) 2002;115(9):1292–1295. [PubMed] [Google Scholar]
3. Khanuja B, Cheah YC, Hunt M, Nishina PM, Wang DQ, Chen HW, Billheimer JT, Carey MC, Paigen B. Lith1, a major gene affecting cholesterol gallstone formation among inbred strains of mice. Proc Natl Acad Sci U S A. 1995;92(17):7729–7733. [PMC free article] [PubMed] [Google Scholar]
4. Nishina PM, Verstuyft J, Paigen B. Synthetic low and high fat diets for the study of atherosclerosis in the mouse. J Lipid Res. 1990;31(5):859–869. [PubMed] [Google Scholar]
5. Getz GS, Reardon CA. Diet and murine atherosclerosis. Arterioscler Thromb Vasc Biol. 2006;26(2):242–249. [PubMed] [Google Scholar]
6. Ishibashi S, Brown MS, Goldstein JL, Gerard RD, Hammer RE, Herz J. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J Clin Invest. 1993;92(2):883–893. [PMC free article] [PubMed] [Google Scholar]
7. Plump AS, Smith JD, Hayek T, Aalto-Setala 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):343–353. [PubMed] [Google Scholar]
8. Plump AS, Breslow JL. Apolipoprotein E and the apolipoprotein E-deficient mouse. Annu Rev Nutr. 1995;15:495–518. [PubMed] [Google Scholar]
9. Jiang XC, Agellon LB, Walsh A, Breslow JL, Tall A. Dietary cholesterol increases transcription of the human cholesteryl ester transfer protein gene in transgenic mice. Dependence on natural flanking sequences. J Clin Invest. 1992;90(4):1290–1295. [PMC free article] [PubMed] [Google Scholar]
10. Baldan A, Pei L, Lee R, Tarr P, Tangirala RK, Weinstein MM, Frank J, Li AC, Tontonoz P, Edwards PA. Impaired development of atherosclerosis in hyperlipidemic Ldlr−/− and ApoE−/− mice transplanted with Abcg1−/− bone marrow. Arterioscler Thromb Vasc Biol. 2006;26(10):2301–2307. [PubMed] [Google Scholar]
11. Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998;394(6689):200–203. [PubMed] [Google Scholar]
12. Lieu HD, Withycombe SK, Walker Q, Rong JX, Walzem RL, Wong JS, Hamilton RL, Fisher EA, Young SG. Eliminating atherogenesis in mice by switching off hepatic lipoprotein secretion. Circulation. 2003;107(9):1315–1321. [PubMed] [Google Scholar]
13. Collins RG, Velji R, Guevara NV, Hicks MJ, Chan L, Beaudet AL. P-Selectin or intercellular adhesion molecule (ICAM)-1 deficiency substantially protects against atherosclerosis in apolipoprotein E-deficient mice. J Exp Med. 2000;191(1):189–194. [PMC free article] [PubMed] [Google Scholar]
14. Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, Davis V, Gutierrez-Ramos JC, Connelly PW, Milstone DS. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J Clin Invest. 2001;107(10):1255–1262. [PMC free article] [PubMed] [Google Scholar]
15. Wang X, Ishimori N, Korstanje R, Rollins J, Paigen B. Identifying novel genes for atherosclerosis through mouse-human comparative genetics. Am J Hum Genet. 2005;77(1):1–15. [PMC free article] [PubMed] [Google Scholar]
16. DiPetrillo K, Wang X, Stylianou IM, Paigen B. Bioinformatics toolbox for narrowing rodent quantitative trait loci. Trends Genet. 2005;21(12):683–692. [PubMed] [Google Scholar]
17. Stylianou IM, Affourtit JP, Shockley KR, Wilpan RY, Abdi FA, Bhardwaj S, Rollins J, Churchill GA, Paigen B. Applying gene expression, proteomics and single-nucleotide polymorphism analysis for complex trait gene identification. Genetics. 2008;178(3):1795–1805. [PMC free article] [PubMed] [Google Scholar]
18. Stylianou IM, Korstanje R, Li R, Sheehan S, Paigen B, Churchill GA. Quantitative trait locus analysis for obesity reveals multiple networks of interacting loci. Mamm Genome. 2006;17(1):22–36. [PubMed] [Google Scholar]
19. Stylianou IM, Langley SR, Walsh K, Chen Y, Revenu C, Paigen B. Differences in DBA/1J and DBA/2J reveal lipid QTL genes. J Lipid Res. 2008;49(11):2402–2413. [PMC free article] [PubMed] [Google Scholar]
20. Stylianou IM, Tsaih SW, DiPetrillo K, Ishimori N, Li R, Paigen B, Churchill G. Complex genetic architecture revealed by analysis of high-density lipoprotein cholesterol in chromosome substitution strains and F2 crosses. Genetics. 2006;174(2):999–1007. [PMC free article] [PubMed] [Google Scholar]
21. Su Z, Cox A, Shen Y, Stylianou IM, Paigen B. Farp2 and Stk25 are candidate genes for the HDL cholesterol locus on mouse chromosome 1. Arterioscler Thromb Vasc Biol. 2009;29(1):107–113. [PMC free article] [PubMed] [Google Scholar]
22. Su Z, Ishimori N, Chen Y, Leiter EH, Churchill GA, Paigen B, Stylianou IM. Four additional mouse crosses improve the lipid QTL landscape and identify Lipg as a QTL gene. J Lipid Res. 2009 [PMC free article] [PubMed] [Google Scholar]
23. Flint J, Valdar W, Shifman S, Mott R. Strategies for mapping and cloning quantitative trait genes in rodents. Nat Rev Genet. 2005;6(4):271–286. [PubMed] [Google Scholar]
24. Flint J, Mott R. Finding the molecular basis of quantitative traits: successes and pitfalls. Nat Rev Genet. 2001;2(6):437–445. [PubMed] [Google Scholar]
25. Mehrabian M, Wong J, Wang X, Jiang Z, Shi W, Fogelman AM, Lusis AJ. Genetic locus in mice that blocks development of atherosclerosis despite extreme hyperlipidemia. Circ Res. 2001;89(2):125–130. [PubMed] [Google Scholar]
26. Mehrabian M, Allayee H, Wong J, Shi W, Wang XP, Shaposhnik Z, Funk CD, Lusis AJ. Identification of 5-lipoxygenase as a major gene contributing to atherosclerosis susceptibility in mice. Circ Res. 2002;91(2):120–126. [PubMed] [Google Scholar]
27. Zhao L, Moos MP, Grabner R, Pedrono F, Fan J, Kaiser B, John N, Schmidt S, Spanbroek R, Lotzer K, Huang L, Cui J, Rader DJ, Evans JF, Habenicht AJ, Funk CD. The 5-lipoxygenase pathway promotes pathogenesis of hyperlipidemia-dependent aortic aneurysm. Nat Med. 2004;10(9):966–973. [PubMed] [Google Scholar]
28. Lusis AJ, Yu J, Wang SS. The problem of passenger genes in transgenic mice. Arterioscler Thromb Vasc Biol. 2007;27(10):2100–2103. [PubMed] [Google Scholar]
29. Ghazalpour A, Wang X, Lusis AJ, Mehrabian M. Complex inheritance of the 5-lipoxygenase locus influencing atherosclerosis in mice. Genetics. 2006;173(2):943–951. [PMC free article] [PubMed] [Google Scholar]
30. Paigen B, Mitchell D, Reue K, Morrow A, Lusis AJ, LeBoeuf RC. Ath-1, a gene determining atherosclerosis susceptibility and high density lipoprotein levels in mice. Proc Natl Acad Sci U S A. 1987;84(11):3763–3767. [PMC free article] [PubMed] [Google Scholar]
31. Wang X, Ria M, Kelmenson PM, Eriksson P, Higgins DC, Samnegard A, Petros C, Rollins J, Bennet AM, Wiman B, de Faire U, Wennberg C, Olsson PG, Ishii N, Sugamura K, Hamsten A, Forsman-Semb K, Lagercrantz J, Paigen B. Positional identification of TNFSF4, encoding OX40 ligand, as a gene that influences atherosclerosis susceptibility. Nat Genet. 2005;37(4):365–372. [PubMed] [Google Scholar]
32. Koch W, Hoppmann P, Mueller JC, Schomig A, Kastrati A. Lack of support for association between common variation in TNFSF4 and myocardial infarction in a German population. Nat Genet. 2008;40(12):1386–1387. author reply 1387–1388. [PubMed] [Google Scholar]
33. Holdt LM, Thiery J, Breslow JL, Teupser D. Increased ADAM17 mRNA expression and activity is associated with atherosclerosis resistance in LDL-receptor deficient mice. Arterioscler Thromb Vasc Biol. 2008;28(6):1097–1103. [PMC free article] [PubMed] [Google Scholar]
34. Teupser D, Tan M, Persky AD, Breslow JL. Atherosclerosis quantitative trait loci are sex- and lineage-dependent in an intercross of C57BL/6 and FVB/N low-density lipoprotein receptor−/− mice. Proc Natl Acad Sci U S A. 2006;103(1):123–128. [PMC free article] [PubMed] [Google Scholar]
35. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011;473(7347):317–325. [PubMed] [Google Scholar]
36. Newman AB, Naydeck BL, Sutton-Tyrrell K, Edmundowicz D, O'Leary D, Kronmal R, Burke GL, Kuller LH. Relationship between coronary artery calcification and other measures of subclinical cardiovascular disease in older adults. Arterioscler Thromb Vasc Biol. 2002;22(10):1674–1679. [PubMed] [Google Scholar]
37. Psaty BM, Furberg CD, Kuller LH, Bild DE, Rautaharju PM, Polak JF, Bovill E, Gottdiener JS. Traditional risk factors and subclinical disease measures as predictors of first myocardial infarction in older adults: the Cardiovascular Health Study. Arch Intern Med. 1999;159(12):1339–1347. [PubMed] [Google Scholar]
38. Hegele RA. Candidate genes, small effects, and the prediction of atherosclerosis. Crit Rev Clin Lab Sci. 1997;34(4):343–367. [PubMed] [Google Scholar]
39. Morgan TM, Krumholz HM, Lifton RP, Spertus JA. Nonvalidation of reported genetic risk factors for acute coronary syndrome in a large-scale replication study. JAMA. 2007;297(14):1551–1561. [PubMed] [Google Scholar]
40. Humphries SE, Morgan L. Genetic risk factors for stroke and carotid atherosclerosis: insights into pathophysiology from candidate gene approaches. Lancet Neurol. 2004;3(4):227–235. [PubMed] [Google Scholar]
41. Cohen JC, Boerwinkle E, Mosley TH, Jr, Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med. 2006;354(12):1264–1272. [PubMed] [Google Scholar]
42. Keating BJ, Tischfield S, Murray SS, Bhangale T, Price TS, Glessner JT, Galver L, Barrett JC, Grant SF, Farlow DN, Chandrupatla HR, Hansen M, Ajmal S, Papanicolaou GJ, Guo Y, Li M, Derohannessian S, de Bakker PI, Bailey SD, Montpetit A, Edmondson AC, Taylor K, Gai X, Wang SS, Fornage M, Shaikh T, Groop L, Boehnke M, Hall AS, Hattersley AT, Frackelton E, Patterson N, Chiang CW, Kim CE, Fabsitz RR, Ouwehand W, Price AL, Munroe P, Caulfield M, Drake T, Boerwinkle E, Reich D, Whitehead AS, Cappola TP, Samani NJ, Lusis AJ, Schadt E, Wilson JG, Koenig W, McCarthy MI, Kathiresan S, Gabriel SB, Hakonarson H, Anand SS, Reilly M, Engert JC, Nickerson DA, Rader DJ, Hirschhorn JN, Fitzgerald GA. Concept, design and implementation of a cardiovascular gene-centric 50 k SNP array for large-scale genomic association studies. PLoS ONE. 2008;3(10):e3583. [PMC free article] [PubMed] [Google Scholar]
43. Hobbs HH, Russell DW, Brown MS, Goldstein JL. The LDL receptor locus in familial hypercholesterolemia: mutational analysis of a membrane protein. Annu Rev Genet. 1990;24:133–170. [PubMed] [Google Scholar]
44. Chen Y, Rollins J, Paigen B, Wang X. Genetic and genomic insights into the molecular basis of atherosclerosis. Cell Metab. 2007;6(3):164–179. [PMC free article] [PubMed] [Google Scholar]
45. Mani A, Radhakrishnan J, Wang H, Mani MA, Nelson-Williams C, Carew KS, Mane S, Najmabadi H, Wu D, Lifton RP. LRP6 mutation in a family with early coronary disease and metabolic risk factors. Science. 2007;315(5816):1278–1282. [PMC free article] [PubMed] [Google Scholar]
46. Helgadottir A, Manolescu A, Thorleifsson G, Gretarsdottir S, Jonsdottir H, Thorsteinsdottir U, Samani NJ, Gudmundsson G, Grant SF, Thorgeirsson G, Sveinbjornsdottir S, Valdimarsson EM, Matthiasson SE, Johannsson H, Gudmundsdottir O, Gurney ME, Sainz J, Thorhallsdottir M, Andresdottir M, Frigge ML, Topol EJ, Kong A, Gudnason V, Hakonarson H, Gulcher JR, Stefansson K. The gene encoding 5-lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat Genet. 2004;36(3):233–239. [PubMed] [Google Scholar]
47. Wang L, Fan C, Topol SE, Topol EJ, Wang Q. Mutation of MEF2A in an inherited disorder with features of coronary artery disease. Science. 2003;302(5650):1578–1581. [PMC free article] [PubMed] [Google Scholar]
48. Altshuler D, Hirschhorn JN. MEF2A sequence variants and coronary artery disease: a change of heart? J Clin Invest. 2005;115(4):831–833. [PMC free article] [PubMed] [Google Scholar]
49. McCarthy MI, Abecasis GR, Cardon LR, Goldstein DB, Little J, Ioannidis JP, Hirschhorn JN. Genome-wide association studies for complex traits: consensus, uncertainty and challenges. Nat Rev Genet. 2008;9(5):356–369. [PubMed] [Google Scholar]
50. Samani NJ, Erdmann J, Hall AS, Hengstenberg C, Mangino M, Mayer B, Dixon RJ, Meitinger T, Braund P, Wichmann HE, Barrett JH, Konig IR, Stevens SE, Szymczak S, Tregouet DA, Iles MM, Pahlke F, Pollard H, Lieb W, Cambien F, Fischer M, Ouwehand W, Blankenberg S, Balmforth AJ, Baessler A, Ball SG, Strom TM, Braenne I, Gieger C, Deloukas P, Tobin MD, Ziegler A, Thompson JR, Schunkert H. Genomewide association analysis of coronary artery disease. N Engl J Med. 2007;357(5):443–453. [PMC free article] [PubMed] [Google Scholar]
51. McPherson R, Pertsemlidis A, Kavaslar N, Stewart A, Roberts R, Cox DR, Hinds DA, Pennacchio LA, Tybjaerg-Hansen A, Folsom AR, Boerwinkle E, Hobbs HH, Cohen JC. A common allele on chromosome 9 associated with coronary heart disease. Science. 2007;316(5830):1488–1491. [PMC free article] [PubMed] [Google Scholar]
52. Helgadottir A, Thorleifsson G, Manolescu A, Gretarsdottir S, Blondal T, Jonasdottir A, Sigurdsson A, Baker A, Palsson A, Masson G, Gudbjartsson DF, Magnusson KP, Andersen K, Levey AI, Backman VM, Matthiasdottir S, Jonsdottir T, Palsson S, Einarsdottir H, Gunnarsdottir S, Gylfason A, Vaccarino V, Hooper WC, Reilly MP, Granger CB, Austin H, Rader DJ, Shah SH, Quyyumi AA, Gulcher JR, Thorgeirsson G, Thorsteinsdottir U, Kong A, Stefansson K. A common variant on chromosome 9p21 affects the risk of myocardial infarction. Science. 2007;316(5830):1491–1493. [PubMed] [Google Scholar]
53. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447(7145):661–678. [PMC free article] [PubMed] [Google Scholar]
54. Kathiresan S, Voight BF, Purcell S, Musunuru K, Ardissino D, Mannucci PM, Anand S, Engert JC, Samani NJ, Schunkert H, Erdmann J, Reilly MP, Rader DJ, Morgan T, Spertus JA, Stoll M, Girelli D, McKeown PP, Patterson CC, Siscovick DS, O'Donnell CJ, Elosua R, Peltonen L, Salomaa V, Schwartz SM, Melander O, Altshuler D, Merlini PA, Berzuini C, Bernardinelli L, Peyvandi F, Tubaro M, Celli P, Ferrario M, Fetiveau R, Marziliano N, Casari G, Galli M, Ribichini F, Rossi M, Bernardi F, Zonzin P, Piazza A, Yee J, Friedlander Y, Marrugat J, Lucas G, Subirana I, Sala J, Ramos R, Meigs JB, Williams G, Nathan DM, MacRae CA, Havulinna AS, Berglund G, Hirschhorn JN, Asselta R, Duga S, Spreafico M, Daly MJ, Nemesh J, Korn JM, McCarroll SA, Surti A, Guiducci C, Gianniny L, Mirel D, Parkin M, Burtt N, Gabriel SB, Thompson JR, Braund PS, Wright BJ, Balmforth AJ, Ball SG, Hall AS, Linsel-Nitschke P, Lieb W, Ziegler A, Konig I, Hengstenberg C, Fischer M, Stark K, Grosshennig A, Preuss M, Wichmann HE, Schreiber S, Ouwehand W, Deloukas P, Scholz M, Cambien F, Li M, Chen Z, Wilensky R, Matthai W, Qasim A, Hakonarson HH, Devaney J, Burnett MS, Pichard AD, Kent KM, Satler L, Lindsay JM, Waksman R, Knouff CW, Waterworth DM, Walker MC, Mooser V, Epstein SE, Scheffold T, Berger K, Huge A, Martinelli N, Olivieri O, Corrocher R, McKeown P, Erdmann E, Konig IR, Holm H, Thorleifsson G, Thorsteinsdottir U, Stefansson K, Do R, Xie C, Siscovick D. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat Genet. 2009;41(3):334–341. [PMC free article] [PubMed] [Google Scholar]
55. Erdmann J, Grosshennig A, Braund PS, Konig IR, Hengstenberg C, Hall AS, Linsel-Nitschke P, Kathiresan S, Wright B, Tregouet DA, Cambien F, Bruse P, Aherrahrou Z, Wagner AK, Stark K, Schwartz SM, Salomaa V, Elosua R, Melander O, Voight BF, O'Donnell CJ, Peltonen L, Siscovick DS, Altshuler D, Merlini PA, Peyvandi F, Bernardinelli L, Ardissino D, Schillert A, Blankenberg S, Zeller T, Wild P, Schwarz DF, Tiret L, Perret C, Schreiber S, El Mokhtari NE, Schafer A, Marz W, Renner W, Bugert P, Kluter H, Schrezenmeir J, Rubin D, Ball SG, Balmforth AJ, Wichmann HE, Meitinger T, Fischer M, Meisinger C, Baumert J, Peters A, Ouwehand WH, Deloukas P, Thompson JR, Ziegler A, Samani NJ, Schunkert H. New susceptibility locus for coronary artery disease on chromosome 3q22.3. Nat Genet. 2009;41(3):280–282. [PMC free article] [PubMed] [Google Scholar]
56. Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P, Jonsdottir GM, Thorleifsson G, Helgadottir H, Steinthorsdottir V, Stefansson H, Williams C, Hui J, Beilby J, Warrington NM, James A, Palmer LJ, Koppelman GH, Heinzmann A, Krueger M, Boezen HM, Wheatley A, Altmuller J, Shin HD, Uh ST, Cheong HS, Jonsdottir B, Gislason D, Park CS, Rasmussen LM, Porsbjerg C, Hansen JW, Backer V, Werge T, Janson C, Jonsson UB, Ng MC, Chan J, So WY, Ma R, Shah SH, Granger CB, Quyyumi AA, Levey AI, Vaccarino V, Reilly MP, Rader DJ, Williams MJ, van Rij AM, Jones GT, Trabetti E, Malerba G, Pignatti PF, Boner A, Pescollderungg L, Girelli D, Olivieri O, Martinelli N, Ludviksson BR, Ludviksdottir D, Eyjolfsson GI, Arnar D, Thorgeirsson G, Deichmann K, Thompson PJ, Wjst M, Hall IP, Postma DS, Gislason T, Gulcher J, Kong A, Jonsdottir I, Thorsteinsdottir U, Stefansson K. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009;41(3):342–347. [PubMed] [Google Scholar]
57. Gretarsdottir S, Baas AF, Thorleifsson G, Holm H, den Heijer M, de Vries JP, Kranendonk SE, Zeebregts CJ, van Sterkenburg SM, Geelkerken RH, van Rij AM, Williams MJ, Boll AP, Kostic JP, Jonasdottir A, Walters GB, Masson G, Sulem P, Saemundsdottir J, Mouy M, Magnusson KP, Tromp G, Elmore JR, Sakalihasan N, Limet R, Defraigne JO, Ferrell RE, Ronkainen A, Ruigrok YM, Wijmenga C, Grobbee DE, Shah SH, Granger CB, Quyyumi AA, Vaccarino V, Patel RS, Zafari AM, Levey AI, Austin H, Girelli D, Pignatti PF, Olivieri O, Martinelli N, Malerba G, Trabetti E, Becker LC, Becker DM, Reilly MP, Rader DJ, Mueller T, Dieplinger B, Haltmayer M, Urbonavicius S, Lindblad B, Gottsater A, Gaetani E, Pola R, Wells P, Rodger M, Forgie M, Langlois N, Corral J, Vicente V, Fontcuberta J, Espana F, Grarup N, Jorgensen T, Witte DR, Hansen T, Pedersen O, Aben KK, de Graaf J, Holewijn S, Folkersen L, Franco-Cereceda A, Eriksson P, Collier DA, Stefansson H, Steinthorsdottir V, Rafnar T, Valdimarsson EM, Magnadottir HB, Sveinbjornsdottir S, Olafsson I, Magnusson MK, Palmason R, Haraldsdottir V, Andersen K, Onundarson PT, Thorgeirsson G, Kiemeney LA, Powell JT, Carey DJ, Kuivaniemi H, Lindholt JS, Jones GT, Kong A, Blankensteijn JD, Matthiasson SE, Thorsteinsdottir U, Stefansson K. Genome-wide association study identifies a sequence variant within the DAB2IP gene conferring susceptibility to abdominal aortic aneurysm. Nat Genet. 2010;42(8):692–697. [PMC free article] [PubMed] [Google Scholar]
58. Wang F, Xu CQ, He Q, Cai JP, Li XC, Wang D, Xiong X, Liao YH, Zeng QT, Yang YZ, Cheng X, Li C, Yang R, Wang CC, Wu G, Lu QL, Bai Y, Huang YF, Yin D, Yang Q, Wang XJ, Dai DP, Zhang RF, Wan J, Ren JH, Li SS, Zhao YY, Fu FF, Huang Y, Li QX, Shi SW, Lin N, Pan ZW, Li Y, Yu B, Wu YX, Ke YH, Lei J, Wang N, Luo CY, Ji LY, Gao LJ, Li L, Liu H, Huang EW, Cui J, Jia N, Ren X, Li H, Ke T, Zhang XQ, Liu JY, Liu MG, Xia H, Yang B, Shi LS, Xia YL, Tu X, Wang QK. Genome-wide association identifies a susceptibility locus for coronary artery disease in the Chinese Han population. Nat Genet. 2011;43(4):345–349. [PubMed] [Google Scholar]
59. Schunkert H, Konig IR, Kathiresan S, Reilly MP, Assimes TL, Holm H, Preuss M, Stewart AF, Barbalic M, Gieger C, Absher D, Aherrahrou Z, Allayee H, Altshuler D, Anand SS, Andersen K, Anderson JL, Ardissino D, Ball SG, Balmforth AJ, Barnes TA, Becker DM, Becker LC, Berger K, Bis JC, Boekholdt SM, Boerwinkle E, Braund PS, Brown MJ, Burnett MS, Buysschaert I, Carlquist JF, Chen L, Cichon S, Codd V, Davies RW, Dedoussis G, Dehghan A, Demissie S, Devaney JM, Diemert P, Do R, Doering A, Eifert S, Mokhtari NE, Ellis SG, Elosua R, Engert JC, Epstein SE, de Faire U, Fischer M, Folsom AR, Freyer J, Gigante B, Girelli D, Gretarsdottir S, Gudnason V, Gulcher JR, Halperin E, Hammond N, Hazen SL, Hofman A, Horne BD, Illig T, Iribarren C, Jones GT, Jukema JW, Kaiser MA, Kaplan LM, Kastelein JJ, Khaw KT, Knowles JW, Kolovou G, Kong A, Laaksonen R, Lambrechts D, Leander K, Lettre G, Li M, Lieb W, Loley C, Lotery AJ, Mannucci PM, Maouche S, Martinelli N, McKeown PP, Meisinger C, Meitinger T, Melander O, Merlini PA, Mooser V, Morgan T, Muhleisen TW, Muhlestein JB, Munzel T, Musunuru K, Nahrstaedt J, Nelson CP, Nothen MM, Olivieri O, Patel RS, Patterson CC, Peters A, Peyvandi F, Qu L, Quyyumi AA, Rader DJ, Rallidis LS, Rice C, Rosendaal FR, Rubin D, Salomaa V, Sampietro ML, Sandhu MS, Schadt E, Schafer A, Schillert A, Schreiber S, Schrezenmeir J, Schwartz SM, Siscovick DS, Sivananthan M, Sivapalaratnam S, Smith A, Smith TB, Snoep JD, Soranzo N, Spertus JA, Stark K, Stirrups K, Stoll M, Tang WH, Tennstedt S, Thorgeirsson G, Thorleifsson G, Tomaszewski M, Uitterlinden AG, van Rij AM, Voight BF, Wareham NJ, Wells GA, Wichmann HE, Wild PS, Willenborg C, Witteman JC, Wright BJ, Ye S, Zeller T, Ziegler A, Cambien F, Goodall AH, Cupples LA, Quertermous T, Marz W, Hengstenberg C, Blankenberg S, Ouwehand WH, Hall AS, Deloukas P, Thompson JR, Stefansson K, Roberts R, Thorsteinsdottir U, O'Donnell CJ, McPherson R, Erdmann J, Samani NJ. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat Genet. 2011;43(4):333–338. [PMC free article] [PubMed] [Google Scholar]
60. A genome-wide association study in Europeans and South Asians identifies five new loci for coronary artery disease. Nat Genet. 2011;43(4):339–344. [PubMed] [Google Scholar]
61. Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang SJ, Pihur V, Vollenweider P, O'Reilly PF, Amin N, Bragg-Gresham JL, Teumer A, Glazer NL, Launer L, Zhao JH, Aulchenko Y, Heath S, Sober S, Parsa A, Luan J, Arora P, Dehghan A, Zhang F, Lucas G, Hicks AA, Jackson AU, Peden JF, Tanaka T, Wild SH, Rudan I, Igl W, Milaneschi Y, Parker AN, Fava C, Chambers JC, Fox ER, Kumari M, Go MJ, van der Harst P, Kao WH, Sjogren M, Vinay DG, Alexander M, Tabara Y, Shaw-Hawkins S, Whincup PH, Liu Y, Shi G, Kuusisto J, Tayo B, Seielstad M, Sim X, Nguyen KD, Lehtimaki T, Matullo G, Wu Y, Gaunt TR, Onland-Moret NC, Cooper MN, Platou CG, Org E, Hardy R, Dahgam S, Palmen J, Vitart V, Braund PS, Kuznetsova T, Uiterwaal CS, Adeyemo A, Palmas W, Campbell H, Ludwig B, Tomaszewski M, Tzoulaki I, Palmer ND, Aspelund T, Garcia M, Chang YP, O'Connell JR, Steinle NI, Grobbee DE, Arking DE, Kardia SL, Morrison AC, Hernandez D, Najjar S, McArdle WL, Hadley D, Brown MJ, Connell JM, Hingorani AD, Day IN, Lawlor DA, Beilby JP, Lawrence RW, Clarke R, Hopewell JC, Ongen H, Dreisbach AW, Li Y, Young JH, Bis JC, Kahonen M, Viikari J, Adair LS, Lee NR, Chen MH, Olden M, Pattaro C, Bolton JA, Kottgen A, Bergmann S, Mooser V, Chaturvedi N, Frayling TM, Islam M, Jafar TH, Erdmann J, Kulkarni SR, Bornstein SR, Grassler J, Groop L, Voight BF, Kettunen J, Howard P, Taylor A, Guarrera S, Ricceri F, Emilsson V, Plump A, Barroso I, Khaw KT, Weder AB, Hunt SC, Sun YV, Bergman RN, Collins FS, Bonnycastle LL, Scott LJ, Stringham HM, Peltonen L, Perola M, Vartiainen E, Brand SM, Staessen JA, Wang TJ, Burton PR, Artigas MS, Dong Y, Snieder H, Wang X, Zhu H, Lohman KK, Rudock ME, Heckbert SR, Smith NL, Wiggins KL, Doumatey A, Shriner D, Veldre G, Viigimaa M, Kinra S, Prabhakaran D, Tripathy V, Langefeld CD, Rosengren A, Thelle DS, Corsi AM, Singleton A, Forrester T, Hilton G, McKenzie CA, Salako T, Iwai N, Kita Y, Ogihara T, Ohkubo T, Okamura T, Ueshima H, Umemura S, Eyheramendy S, Meitinger T, Wichmann HE, Cho YS, Kim HL, Lee JY, Scott J, Sehmi JS, Zhang W, Hedblad B, Nilsson P, Smith GD, Wong A, Narisu N, Stancakova A, Raffel LJ, Yao J, Kathiresan S, O'Donnell CJ, Schwartz SM, Ikram MA, Longstreth WT, Jr, Mosley TH, Seshadri S, Shrine NR, Wain LV, Morken MA, Swift AJ, Laitinen J, Prokopenko I, Zitting P, Cooper JA, Humphries SE, Danesh J, Rasheed A, Goel A, Hamsten A, Watkins H, Bakker SJ, van Gilst WH, Janipalli CS, Mani KR, Yajnik CS, Hofman A, Mattace-Raso FU, Oostra BA, Demirkan A, Isaacs A, Rivadeneira F, Lakatta EG, Orru M, Scuteri A, Ala-Korpela M, Kangas AJ, Lyytikainen LP, Soininen P, Tukiainen T, Wurtz P, Ong RT, Dorr M, Kroemer HK, Volker U, Volzke H, Galan P, Hercberg S, Lathrop M, Zelenika D, Deloukas P, Mangino M, Spector TD, Zhai G, Meschia JF, Nalls MA, Sharma P, Terzic J, Kumar MV, Denniff M, Zukowska-Szczechowska E, Wagenknecht LE, Fowkes FG, Charchar FJ, Schwarz PE, Hayward C, Guo X, Rotimi C, Bots ML, Brand E, Samani NJ, Polasek O, Talmud PJ, Nyberg F, Kuh D, Laan M, Hveem K, Palmer LJ, van der Schouw YT, Casas JP, Mohlke KL, Vineis P, Raitakari O, Ganesh SK, Wong TY, Tai ES, Cooper RS, Laakso M, Rao DC, Harris TB, Morris RW, Dominiczak AF, Kivimaki M, Marmot MG, Miki T, Saleheen D, Chandak GR, Coresh J, Navis G, Salomaa V, Han BG, Zhu X, Kooner JS, Melander O, Ridker PM, Bandinelli S, Gyllensten UB, Wright AF, Wilson JF, Ferrucci L, Farrall M, Tuomilehto J, Pramstaller PP, Elosua R, Soranzo N, Sijbrands EJ, Altshuler D, Loos RJ, Shuldiner AR, Gieger C, Meneton P, Uitterlinden AG, Wareham NJ, Gudnason V, Rotter JI, Rettig R, Uda M, Strachan DP, Witteman JC, Hartikainen AL, Beckmann JS, Boerwinkle E, Vasan RS, Boehnke M, Larson MG, Jarvelin MR, Psaty BM, Abecasis GR, Chakravarti A, Elliott P, van Duijn CM, Newton-Cheh C, Levy D, Caulfield MJ, Johnson T, Tang H, Knowles J, Hlatky M, Fortmann S, Assimes TL, Quertermous T, Go A, Iribarren C, Absher D, Risch N, Myers R, Sidney S, Ziegler A, Schillert A, Bickel C, Sinning C, Rupprecht HJ, Lackner K, Wild P, Schnabel R, Blankenberg S, Zeller T, Munzel T, Perret C, Cambien F, Tiret L, Nicaud V, Proust C, Uitterlinden A, van Duijn C, Whitteman J, Cupples LA, Demissie-Banjaw S, Ramachandran V, Smith A, Folsom A, Morrison A, Chen IY, Bis J, Volcik K, Rice K, Taylor KD, Marciante K, Smith N, Glazer N, Heckbert S, Harris T, Lumley T, Kong A, Thorleifsson G, Thorgeirsson G, Holm H, Gulcher JR, Stefansson K, Andersen K, Gretarsdottir S, Thorsteinsdottir U, Preuss M, Schreiber S, Konig IR, Lieb W, Hengstenberg C, Schunkert H, Fischer M, Grosshennig A, Medack A, Stark K, Linsel-Nitschke P, Bruse P, Aherrahrou Z, Peters A, Loley C, Willenborg C, Nahrstedt J, Freyer J, Gulde S, Doering A, Meisinger C, Klopp N, Illig T, Meinitzer A, Tomaschitz A, Halperin E, Dobnig H, Scharnagl H, Kleber M, Laaksonen R, Pilz S, Grammer TB, Stojakovic T, Renner W, Marz W, Bohm BO, Winkelmann BR, Winkler K, Hoffmann MM, Siscovick DS, Musunuru K, Barbalic M, Guiducci C, Burtt N, Gabriel SB, Stewart AF, Wells GA, Chen L, Jarinova O, Roberts R, McPherson R, Dandona S, Pichard AD, Rader DJ, Devaney J, Lindsay JM, Kent KM, Qu L, Satler L, Burnett MS, Li M, Reilly MP, Wilensky R, Waksman R, Epstein S, Matthai W, Knouff CW, Waterworth DM, Hakonarson HH, Walker MC, Hall AS, Balmforth AJ, Wright BJ, Nelson C, Thompson JR, Ball SG, Felix JF, Demissie S, Loehr LR, Rosamond WD, Folsom AR, Benjamin E, Aulchenko YS, Haritunians T, Couper D, Murabito J, Wang YA, Stricker BH, Gottdiener JS, Chang PP, Willerson JT, Boger CA, Fuchsberger C, Gao X, Yang Q, Schmidt H, Ketkar S, Pare G, Atkinson EJ, Lohman K, Cornelis MC, Probst-Hensch NM, Kronenberg F, Tonjes A, Eiriksdottir G, Launer LJ, Rampersaud E, Mitchell BD, Struchalin M, Cavalieri M, Giallauria F, Metter J, de Boer J, Siscovick D, Zillikens MC, Feitosa M, Province M, de Andrade M, Turner ST, Wild PS, Schnabel RB, Wilde S, Munzel TF, Leak TS, Koenig W, Zgaga L, Zemunik T, Kolcic I, Minelli C, Hu FB, Johansson A, Zaboli G, Ellinghaus D, Imboden M, Nitsch D, Brandstatter A, Kollerits B, Kedenko L, Magi R, Stumvoll M, Kovacs P, Boban M, Campbell S, Endlich K, Nauck M, Badola S, Curhan GC, Franke A, Rochat T, Paulweber B, Wang W, Schmidt R, Shlipak MG, Borecki I, Kramer BK, Gyllensten U, Hastie N, Heid IM, Fox CS, Felix SB, Watzinger N, Homuth G, Aragam J, Zweiker R, Lind L, Rodeheffer RJ, Greiser KH, Deckers JW, Stritzke J, Lackner KJ, Ingelsson E, Kullo I, Haerting J, Reffelmann T, Redfield MM, Werdan K, Mitchell GF, Arnett DK, Blettner M, Friedrich N, Benjamin EJ, Lord GM, Gale DP, Wass MN, Ahmadi KR, Beckmann J, Bilo HJ, Cook HT, Cotlarciuc I, Davey Smith G, de Silva R, Deng G, Devuyst O, Dikkeschei LD, Dimkovic N, Dockrell M, Dominiczak A, Ebrahim S, Eggermann T, Floege J, Forouhi NG, Gansevoort RT, Han X, Homan van der Heide JJ, Hepkema BG, Hernandez-Fuentes M, Hypponen E, de Jong PE, Kleefstra N, Lagou V, Lapsley M, Luttropp K, Marechal C, Nordfors L, Penninx BW, Perucha E, Pouta A, Roderick PJ, Ruokonen A, Sanna S, Schalling M, Schlessinger D, Schlieper G, Seelen MA, Smit JH, Stenvinkel P, Sternberg MJ, Swaminathan R, Ubink-Veltmaat LJ, Wallace C, Waterworth D, Zerres K, Waeber G, Maxwell PH, McCarthy MI, Lightstone L. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478(7367):103–109. [PMC free article] [PubMed] [Google Scholar]
62. Suzuki H, Zhou X, Yin J, Lei J, Jiang HY, Suzuki Y, Chan T, Hannon GJ, Mergner WJ, Abraham JM. Intragenic mutations of CDKN2B and CDKN2A in primary human esophageal cancers. Human Molecular Genetics. 1995;4(10):1883–1887. [PubMed] [Google Scholar]
63. Jarinova O, Stewart AF, Roberts R, Wells G, Lau P, Naing T, Buerki C, McLean BW, Cook RC, Parker JS, McPherson R. Functional analysis of the chromosome 9p21.3 coronary artery disease risk locus. Arterioscler Thromb Vasc Biol. 2009;29(10):1671–1677. [PubMed] [Google Scholar]
64. Visel A, Zhu Y, May D, Afzal V, Gong E, Attanasio C, Blow MJ, Cohen JC, Rubin EM, Pennacchio LA. Targeted deletion of the 9p21 non-coding coronary artery disease risk interval in mice. Nature. 2010;464(7287):409–412. [PMC free article] [PubMed] [Google Scholar]
65. Ruas M, Peters G. The p16INK4a/CDKN2A tumor suppressor and its relatives. Biochim Biophys Acta. 1998;1378(2):F115–F177. [PubMed] [Google Scholar]
66. Pasmant E, Laurendeau I, Heron D, Vidaud M, Vidaud D, Bieche I. Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: identification of ANRIL, an antisense noncoding RNA whose expression coclusters with ARF. Cancer Res. 2007;67(8):3963–3969. [PubMed] [Google Scholar]
67. Holdt LM, Beutner F, Scholz M, Gielen S, Gabel G, Bergert H, Schuler G, Thiery J, Teupser D. ANRIL expression is associated with atherosclerosis risk at chromosome 9p21. Arterioscler Thromb Vasc Biol. 2010;30(3):620–627. [PubMed] [Google Scholar]
68. Harismendy O, Notani D, Song X, Rahim NG, Tanasa B, Heintzman N, Ren B, Fu XD, Topol EJ, Rosenfeld MG, Frazer KA. 9p21 DNA variants associated with coronary artery disease impair interferon-gamma signalling response. Nature. 2011;470(7333):264–268. [PMC free article] [PubMed] [Google Scholar]
69. Salter RC, Ashlin TG, Kwan AP, Ramji DP. ADAMTS proteases: key roles in atherosclerosis? J Mol Med. 2010;88(12):1203–1211. [PubMed] [Google Scholar]
70. Porter S, Clark IM, Kevorkian L, Edwards DR. The ADAMTS metalloproteinases. Biochem J. 2005;386(Pt 1):15–27. [PMC free article] [PubMed] [Google Scholar]
71. Wight TN, Merrilees MJ. Proteoglycans in atherosclerosis and restenosis: key roles for versican. Circ Res. 2004;94(9):1158–1167. [PubMed] [Google Scholar]
72. Wight TN. Arterial remodeling in vascular disease: a key role for hyaluronan and versican. Front Biosci. 2008;13:4933–4937. [PubMed] [Google Scholar]
73. Wagsater D, Bjork H, Zhu C, Bjorkegren J, Valen G, Hamsten A, Eriksson P. ADAMTS-4 and -8 are inflammatory regulated enzymes expressed in macrophage-rich areas of human atherosclerotic plaques. Atherosclerosis. 2008;196(2):514–522. [PubMed] [Google Scholar]
74. Jonsson-Rylander AC, Nilsson T, Fritsche-Danielson R, Hammarstrom A, Behrendt M, Andersson JO, Lindgren K, Andersson AK, Wallbrandt P, Rosengren B, Brodin P, Thelin A, Westin A, Hurt-Camejo E, Lee-Sogaard CH. Role of ADAMTS-1 in atherosclerosis: remodeling of carotid artery, immunohistochemistry, and proteolysis of versican. Arterioscler Thromb Vasc Biol. 2005;25(1):180–185. [PubMed] [Google Scholar]
75. Sabatine MS, Ploughman L, Simonsen KL, Iakoubova OA, Kirchgessner TG, Ranade K, Tsuchihashi Z, Zerba KE, Long DU, Tong CH, Packard CJ, Pfeffer MA, Devlin JJ, Shepherd J, Campos H, Sacks FM, Braunwald E. Association between ADAMTS1 matrix metalloproteinase gene variation, coronary heart disease, and benefit of statin therapy. Arterioscler Thromb Vasc Biol. 2008;28(3):562–567. [PubMed] [Google Scholar]
76. Reilly MP, Li M, He J, Ferguson JF, Stylianou IM, Mehta NN, Burnett MS, Devaney JM, Knouff CW, Thompson JR, Horne BD, Stewart AF, Assimes TL, Wild PS, Allayee H, Nitschke PL, Patel RS, Martinelli N, Girelli D, Quyyumi AA, Anderson JL, Erdmann J, Hall AS, Schunkert H, Quertermous T, Blankenberg S, Hazen SL, Roberts R, Kathiresan S, Samani NJ, Epstein SE, Rader DJ, Qasim AN, DerOhannessian SL, Qu L, Cappola TP, Chen Z, Matthai W, Hakonarson HH, Wilensky R, Kent KM, Lindsay JM, Pichard AD, Satler L, Waksman R. Identification of ADAMTS7 as a novel locus for coronary atherosclerosis and association of ABO with myocardial infarction in the presence of coronary atherosclerosis: two genome-wide association studies. Lancet. 2011;377(9763):383–392. [PMC free article] [PubMed] [Google Scholar]
77. Liu CJ, Kong W, Ilalov K, Yu S, Xu K, Prazak L, Fajardo M, Sehgal B, Di Cesare PE. ADAMTS-7: a metalloproteinase that directly binds to and degrades cartilage oligomeric matrix protein. FASEB J. 2006;20(7):988–990. [PMC free article] [PubMed] [Google Scholar]
78. Wang L, Zheng J, Bai X, Liu B, Liu CJ, Xu Q, Zhu Y, Wang N, Kong W, Wang X. ADAMTS-7 mediates vascular smooth muscle cell migration and neointima formation in balloon-injured rat arteries. Circ Res. 2009;104(5):688–698. [PubMed] [Google Scholar]
79. Salcedo R, Oppenheim JJ. Role of chemokines in angiogenesis: CXCL12/SDF-1 and CXCR4 interaction, a key regulator of endothelial cell responses. Microcirculation. 2003;10(3–4):359–370. [PubMed] [Google Scholar]
80. Mehta NN, Li M, William D, Khera AV, Derohannessian S, Qu L, Ferguson JF, McLaughlin C, Shaikh LH, Shah R, Patel PN, Bradfield JP, He J, Stylianou IM, Hakonarson H, Rader DJ, Reilly MP. The novel atherosclerosis locus at 10q11 regulates plasma CXCL12 levels. Eur Heart J. 2011 [PMC free article] [PubMed] [Google Scholar]
81. Damas JK, Waehre T, Yndestad A, Ueland T, Muller F, Eiken HG, Holm AM, Halvorsen B, Froland SS, Gullestad L, Aukrust P. Stromal cell-derived factor-1alpha in unstable angina: potential antiinflammatory and matrix-stabilizing effects. Circulation. 2002;106(1):36–42. [PubMed] [Google Scholar]
82. Nagasawa T, Hirota S, Tachibana K, Takakura N, Nishikawa S, Kitamura Y, Yoshida N, Kikutani H, Kishimoto T. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996;382(6592):635–638. [PubMed] [Google Scholar]
83. Ma Q, Jones D, Borghesani PR, Segal RA, Nagasawa T, Kishimoto T, Bronson RT, Springer TA. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci U S A. 1998;95(16):9448–9453. [PMC free article] [PubMed] [Google Scholar]
84. Ara T, Itoi M, Kawabata K, Egawa T, Tokoyoda K, Sugiyama T, Fujii N, Amagai T, Nagasawa T. A role of CXC chemokine ligand 12/stromal cell-derived factor-1/pre-B cell growth stimulating factor and its receptor CXCR4 in fetal and adult T cell development in vivo. J Immunol. 2003;170(9):4649–4655. [PubMed] [Google Scholar]
85. Hu X, Dai S, Wu WJ, Tan W, Zhu X, Mu J, Guo Y, Bolli R, Rokosh G. Stromal cell derived factor-1 alpha confers protection against myocardial ischemia/reperfusion injury: role of the cardiac stromal cell derived factor-1 alpha CXCR4 axis. Circulation. 2007;116(6):654–663. [PMC free article] [PubMed] [Google Scholar]
86. Garrison RJ, Havlik RJ, Harris RB, Feinleib M, Kannel WB, Padgett SJ. ABO blood group and cardiovacular disease: the Framingham study. Atherosclerosis. 1976;25(2–3):311–318. [PubMed] [Google Scholar]
87. Morton NE. Genetic markers in atherosclerosis: a review. J Med Genet. 1976;13(2):81–90. [PMC free article] [PubMed] [Google Scholar]
88. Vogel F. Controversy in human genetics. ABO blood groups and disease. Am J Hum Genet. 1970;22(4):464–475. [PMC free article] [PubMed] [Google Scholar]
89. Weiss NS. ABO blood type and arteriosclerosis obliterans. Am J Hum Genet. 1972;24(1):65–70. [PMC free article] [PubMed] [Google Scholar]
90. Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M, Pirruccello JP, Ripatti S, Chasman DI, Willer CJ, Johansen CT, Fouchier SW, Isaacs A, Peloso GM, Barbalic M, Ricketts SL, Bis JC, Aulchenko YS, Thorleifsson G, Feitosa MF, Chambers J, Orho-Melander M, Melander O, Johnson T, Li X, Guo X, Li M, Shin Cho Y, Jin Go M, Jin Kim Y, Lee JY, Park T, Kim K, Sim X, Twee-Hee Ong R, Croteau-Chonka DC, Lange LA, Smith JD, Song K, Hua Zhao J, Yuan X, Luan J, Lamina C, Ziegler A, Zhang W, Zee RY, Wright AF, Witteman JC, Wilson JF, Willemsen G, Wichmann HE, Whitfield JB, Waterworth DM, Wareham NJ, Waeber G, Vollenweider P, Voight BF, Vitart V, Uitterlinden AG, Uda M, Tuomilehto J, Thompson JR, Tanaka T, Surakka I, Stringham HM, Spector TD, Soranzo N, Smit JH, Sinisalo J, Silander K, Sijbrands EJ, Scuteri A, Scott J, Schlessinger D, Sanna S, Salomaa V, Saharinen J, Sabatti C, Ruokonen A, Rudan I, Rose LM, Roberts R, Rieder M, Psaty BM, Pramstaller PP, Pichler I, Perola M, Penninx BW, Pedersen NL, Pattaro C, Parker AN, Pare G, Oostra BA, O'Donnell CJ, Nieminen MS, Nickerson DA, Montgomery GW, Meitinger T, McPherson R, McCarthy MI, McArdle W, Masson D, Martin NG, Marroni F, Mangino M, Magnusson PK, Lucas G, Luben R, Loos RJ, Lokki ML, Lettre G, Langenberg C, Launer LJ, Lakatta EG, Laaksonen R, Kyvik KO, Kronenberg F, Konig IR, Khaw KT, Kaprio J, Kaplan LM, Johansson A, Jarvelin MR, Janssens AC, Ingelsson E, Igl W, Kees Hovingh G, Hottenga JJ, Hofman A, Hicks AA, Hengstenberg C, Heid IM, Hayward C, Havulinna AS, Hastie ND, Harris TB, Haritunians T, Hall AS, Gyllensten U, Guiducci C, Groop LC, Gonzalez E, Gieger C, Freimer NB, Ferrucci L, Erdmann J, Elliott P, Ejebe KG, Doring A, Dominiczak AF, Demissie S, Deloukas P, de Geus EJ, de Faire U, Crawford G, Collins FS, Chen YD, Caulfield MJ, Campbell H, Burtt NP, Bonnycastle LL, Boomsma DI, Boekholdt SM, Bergman RN, Barroso I, Bandinelli S, Ballantyne CM, Assimes TL, Quertermous T, Altshuler D, Seielstad M, Wong TY, Tai ES, Feranil AB, Kuzawa CW, Adair LS, Taylor HA, Jr, Borecki IB, Gabriel SB, Wilson JG, Holm H, Thorsteinsdottir U, Gudnason V, Krauss RM, Mohlke KL, Ordovas JM, Munroe PB, Kooner JS, Tall AR, Hegele RA, Kastelein JJ, Schadt EE, Rotter JI, Boerwinkle E, Strachan DP, Mooser V, Stefansson K, Reilly MP, Samani NJ, Schunkert H, Cupples LA, Sandhu MS, Ridker PM, Rader DJ, van Duijn CM, Peltonen L, Abecasis GR, Boehnke M, Kathiresan S. Biological, clinical and population relevance of 95 loci for blood lipids. Nature. 2010;466(7307):707–713. [PMC free article] [PubMed] [Google Scholar]
91. Roseman S. Reflections on glycobiology. J Biol Chem. 2001;276(45):41527–41542. [PubMed] [Google Scholar]
92. Yamamoto M, Lin XH, Kominato Y, Hata Y, Noda R, Saitou N, Yamamoto F. Murine equivalent of the human histo-blood group ABO gene is a cis-AB gene and encodes a glycosyltransferase with both A and B transferase activity. J Biol Chem. 2001;276(17):13701–13708. [PubMed] [Google Scholar]
93. Eichler EE, Flint J, Gibson G, Kong A, Leal SM, Moore JH, Nadeau JH. Missing heritability and strategies for finding the underlying causes of complex disease. Nat Rev Genet. 2010;11(6):446–450. [PMC free article] [PubMed] [Google Scholar]
94. Churchill GA, Airey DC, Allayee H, Angel JM, Attie AD, Beatty J, Beavis WD, Belknap JK, Bennett B, Berrettini W, Bleich A, Bogue M, Broman KW, Buck KJ, Buckler E, Burmeister M, Chesler EJ, Cheverud JM, Clapcote S, Cook MN, Cox RD, Crabbe JC, Crusio WE, Darvasi A, Deschepper CF, Doerge RW, Farber CR, Forejt J, Gaile D, Garlow SJ, Geiger H, Gershenfeld H, Gordon T, Gu J, Gu W, de Haan G, Hayes NL, Heller C, Himmelbauer H, Hitzemann R, Hunter K, Hsu HC, Iraqi FA, Ivandic B, Jacob HJ, Jansen RC, Jepsen KJ, Johnson DK, Johnson TE, Kempermann G, Kendziorski C, Kotb M, Kooy RF, Llamas B, Lammert F, Lassalle JM, Lowenstein PR, Lu L, Lusis A, Manly KF, Marcucio R, Matthews D, Medrano JF, Miller DR, Mittleman G, Mock BA, Mogil JS, Montagutelli X, Morahan G, Morris DG, Mott R, Nadeau JH, Nagase H, Nowakowski RS, O'Hara BF, Osadchuk AV, Page GP, Paigen B, Paigen K, Palmer AA, Pan HJ, Peltonen-Palotie L, Peirce J, Pomp D, Pravenec M, Prows DR, Qi Z, Reeves RH, Roder J, Rosen GD, Schadt EE, Schalkwyk LC, Seltzer Z, Shimomura K, Shou S, Sillanpaa MJ, Siracusa LD, Snoeck HW, Spearow JL, Svenson K, Tarantino LM, Threadgill D, Toth LA, Valdar W, de Villena FP, Warden C, Whatley S, Williams RW, Wiltshire T, Yi N, Zhang D, Zhang M, Zou F. The Collaborative Cross, a community resource for the genetic analysis of complex traits. Nat Genet. 2004;36(11):1133–1137. [PubMed] [Google Scholar]
95. Durrant C, Tayem H, Yalcin B, Cleak J, Goodstadt L, de Villena FP, Mott R, Iraqi FA. Collaborative Cross mice and their power to map host susceptibility to Aspergillus fumigatus infection. Genome Res. 2011;21(8):1239–1248. [PMC free article] [PubMed] [Google Scholar]
96. Philip VM, Sokoloff G, Ackert-Bicknell CL, Striz M, Branstetter L, Beckmann MA, Spence JS, Jackson BL, Galloway LD, Barker P, Wymore AM, Hunsicker PR, Durtschi DC, Shaw GS, Shinpock S, Manly KF, Miller DR, Donohue KD, Culiat CT, Churchill GA, Lariviere WR, Palmer AA, O'Hara BF, Voy BH, Chesler EJ. Genetic analysis in the Collaborative Cross breeding population. Genome Res. 2011;21(8):1223–1238. [PMC free article] [PubMed] [Google Scholar]
97. Marenberg ME, Risch N, Berkman LF, Floderus B, de Faire U. Genetic susceptibility to death from coronary heart disease in a study of twins. N Engl J Med. 1994;330(15):1041–1046. [PubMed] [Google Scholar]
98. Schunkert H, Konig IR, Kathiresan S, Reilly MP, Assimes TL, Holm H, Preuss M, Stewart AF, Barbalic M, Gieger C, Absher D, Aherrahrou Z, Allayee H, Altshuler D, Anand SS, Andersen K, Anderson JL, Ardissino D, Ball SG, Balmforth AJ, Barnes TA, Becker DM, Becker LC, Berger K, Bis JC, Boekholdt SM, Boerwinkle E, Braund PS, Brown MJ, Burnett MS, Buysschaert I, Carlquist JF, Chen L, Cichon S, Codd V, Davies RW, Dedoussis G, Dehghan A, Demissie S, Devaney JM, Diemert P, Do R, Doering A, Eifert S, Mokhtari NE, Ellis SG, Elosua R, Engert JC, Epstein SE, de Faire U, Fischer M, Folsom AR, Freyer J, Gigante B, Girelli D, Gretarsdottir S, Gudnason V, Gulcher JR, Halperin E, Hammond N, Hazen SL, Hofman A, Horne BD, Illig T, Iribarren C, Jones GT, Jukema JW, Kaiser MA, Kaplan LM, Kastelein JJ, Khaw KT, Knowles JW, Kolovou G, Kong A, Laaksonen R, Lambrechts D, Leander K, Lettre G, Li M, Lieb W, Loley C, Lotery AJ, Mannucci PM, Maouche S, Martinelli N, McKeown PP, Meisinger C, Meitinger T, Melander O, Merlini PA, Mooser V, Morgan T, Muhleisen TW, Muhlestein JB, Munzel T, Musunuru K, Nahrstaedt J, Nelson CP, Nothen MM, Olivieri O, Patel RS, Patterson CC, Peters A, Peyvandi F, Qu L, Quyyumi AA, Rader DJ, Rallidis LS, Rice C, Rosendaal FR, Rubin D, Salomaa V, Sampietro ML, Sandhu MS, Schadt E, Schafer A, Schillert A, Schreiber S, Schrezenmeir J, Schwartz SM, Siscovick DS, Sivananthan M, Sivapalaratnam S, Smith A, Smith TB, Snoep JD, Soranzo N, Spertus JA, Stark K, Stirrups K, Stoll M, Tang WH, Tennstedt S, Thorgeirsson G, Thorleifsson G, Tomaszewski M, Uitterlinden AG, van Rij AM, Voight BF, Wareham NJ, Wells GA, Wichmann HE, Wild PS, Willenborg C, Witteman JC, Wright BJ, Ye S, Zeller T, Ziegler A, Cambien F, Goodall AH, Cupples LA, Quertermous T, Marz W, Hengstenberg C, Blankenberg S, Ouwehand WH, Hall AS, Deloukas P, Thompson JR, Stefansson K, Roberts R, Thorsteinsdottir U, O'Donnell CJ, McPherson R, Erdmann J, Samani NJ. Large-scale association analysis identifies 13 new susceptibility loci for coronary artery disease. Nat Genet. 2011 [PMC free article] [PubMed] [Google Scholar]
99. Tan MH, Sun Z, Opitz SL, Schmidt TE, Peters JH, George EL. Deletion of the alternatively spliced fibronectin EIIIA domain in mice reduces atherosclerosis. Blood. 2004;104(1):11–18. [PubMed] [Google Scholar]
100. Jagavelu K, Tietge UJ, Gaestel M, Drexler H, Schieffer B, Bavendiek U. Systemic deficiency of the MAP kinase-activated protein kinase 2 reduces atherosclerosis in hypercholesterolemic mice. Circ Res. 2007;101(11):1104–1112. [PubMed] [Google Scholar]
101. Yagyu H, Kitamine T, Osuga J, Tozawa R, Chen Z, Kaji Y, Oka T, Perrey S, Tamura Y, Ohashi K, Okazaki H, Yahagi N, Shionoiri F, Iizuka Y, Harada K, Shimano H, Yamashita H, Gotoda T, Yamada N, Ishibashi S. Absence of ACAT-1 attenuates atherosclerosis but causes dry eye and cutaneous xanthomatosis in mice with congenital hyperlipidemia. J Biol Chem. 2000;275(28):21324–21330. [PubMed] [Google Scholar]
102. Kelly JA, Griffin ME, Fava RA, Wood SG, Bessette KA, Miller ER, Huber SA, Binder CJ, Witztum JL, Morganelli PM. Inhibition of arterial lesion progression in CD16-deficient mice: evidence for altered immunity and the role of IL-10. Cardiovasc Res. 2010;85(1):224–231. [PMC free article] [PubMed] [Google Scholar]
103. Hashimoto T, Kihara M, Imai N, Yoshida S, Shimoyamada H, Yasuzaki H, Ishida J, Toya Y, Kiuchi Y, Hirawa N, Tamura K, Yazawa T, Kitamura H, Fukamizu A, Umemura S. Requirement of apelin-apelin receptor system for oxidative stress-linked atherosclerosis. Am J Pathol. 2007;171(5):1705–1712. [PMC free article] [PubMed] [Google Scholar]
104. Wang KY, Tanimoto A, Guo X, Yamada S, Shimajiri S, Murata Y, Ding Y, Tsutsui M, Kato S, Watanabe T, Ohtsu H, Hirano K, Kohno K, Sasaguri Y. Histamine deficiency decreases atherosclerosis and inflammatory response in apolipoprotein E knockout mice independently of serum cholesterol level. Arterioscler Thromb Vasc Biol. 2011;31(4):800–807. [PubMed] [Google Scholar]
105. Bengtsson E, To F, Hakansson K, Grubb A, Branen L, Nilsson J, Jovinge S. Lack of the cysteine protease inhibitor cystatin C promotes atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25(10):2151–2156. [PubMed] [Google Scholar]
106. Makowski L, Boord JB, Maeda K, Babaev VR, Uysal KT, Morgan MA, Parker RA, Suttles J, Fazio S, Hotamisligil GS, Linton MF. Lack of macrophage fatty-acid-binding protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis. Nat Med. 2001;7(6):699–705. [PMC free article] [PubMed] [Google Scholar]
107. Perrella MA, Pellacani A, Layne MD, Patel A, Zhao D, Schreiber BM, Storch J, Feinberg MW, Hsieh CM, Haber E, Lee ME. Absence of adipocyte fatty acid binding protein prevents the development of accelerated atherosclerosis in hypercholesterolemic mice. FASEB J. 2001;15(10):1774–1776. [PubMed] [Google Scholar]
108. Kocher O, Yesilaltay A, Shen CH, Zhang S, Daniels K, Pal R, Chen J, Krieger M. Influence of PDZK1 on lipoprotein metabolism and atherosclerosis. Biochim Biophys Acta. 2008;1782(5):310–316. [PMC free article] [PubMed] [Google Scholar]
109. Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, Miyata M. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and apolipoprotein E. Proc Natl Acad Sci U S A. 1995;92(18):8264–8268. [PMC free article] [PubMed] [Google Scholar]
110. Aiello RJ, Brees D, Bourassa PA, Royer L, Lindsey S, Coskran T, Haghpassand M, Francone OL. Increased atherosclerosis in hyperlipidemic mice with inactivation of ABCA1 in macrophages. Arterioscler Thromb Vasc Biol. 2002;22(4):630–637. [PubMed] [Google Scholar]
111. Ando Y, Shimizugawa T, Takeshita S, Ono M, Shimamura M, Koishi R, Furukawa H. A decreased expression of angiopoietin-like 3 is protective against atherosclerosis in apoE-deficient mice. J Lipid Res. 2003;44(6):1216–1223. [PubMed] [Google Scholar]
112. Febbraio M, Podrez EA, Smith JD, Hajjar DP, Hazen SL, Hoff HF, Sharma K, Silverstein RL. Targeted disruption of the class B scavenger receptor CD36 protects against atherosclerotic lesion development in mice. J Clin Invest. 2000;105(8):1049–1056. [PMC free article] [PubMed] [Google Scholar]
113. Schieffer B, Selle T, Hilfiker A, Hilfiker-Kleiner D, Grote K, Tietge UJ, Trautwein C, Luchtefeld M, Schmittkamp C, Heeneman S, Daemen MJ, Drexler H. Impact of interleukin-6 on plaque development and morphology in experimental atherosclerosis. Circulation. 2004;110(22):3493–3500. [PubMed] [Google Scholar]
114. Trigatti B, Rayburn H, Vinals M, Braun A, Miettinen H, Penman M, Hertz M, Schrenzel M, Amigo L, Rigotti A, Krieger M. Influence of the high density lipoprotein receptor SR-BI on reproductive and cardiovascular pathophysiology. Proc Natl Acad Sci U S A. 1999;96(16):9322–9327. [PMC free article] [PubMed] [Google Scholar]
115. Ng CJ, Bourquard N, Grijalva V, Hama S, Shih DM, Navab M, Fogelman AM, Lusis AJ, Young S, Reddy ST. Paraoxonase-2 deficiency aggravates atherosclerosis in mice despite lower apolipoprotein-B-containing lipoproteins: anti-atherogenic role for paraoxonase-2. J Biol Chem. 2006;281(40):29491–29500. [PubMed] [Google Scholar]
116. Frank PG, Lee H, Park DS, Tandon NN, Scherer PE, Lisanti MP. Genetic ablation of caveolin-1 confers protection against atherosclerosis. Arterioscler Thromb Vasc Biol. 2004;24(1):98–105. [PubMed] [Google Scholar]
117. Hasty AH, Shimano H, Osuga J, Namatame I, Takahashi A, Yahagi N, Perrey S, Iizuka Y, Tamura Y, Amemiya-Kudo M, Yoshikawa T, Okazaki H, Ohashi K, Harada K, Matsuzaka T, Sone H, Gotoda T, Nagai R, Ishibashi S, Yamada N. Severe hypercholesterolemia, hypertriglyceridemia, and atherosclerosis in mice lacking both leptin and the low density lipoprotein receptor. J Biol Chem. 2001;276(40):37402–37408. [PubMed] [Google Scholar]
118. Powell-Braxton L, Veniant M, Latvala RD, Hirano KI, Won WB, Ross J, Dybdal N, Zlot CH, Young SG, Davidson NO. A mouse model of human familial hypercholesterolemia: markedly elevated low density lipoprotein cholesterol levels and severe atherosclerosis on a low-fat chow diet. Nat Med. 1998;4(8):934–938. [PubMed] [Google Scholar]
119. Mehta JL, Sanada N, Hu CP, Chen J, Dandapat A, Sugawara F, Satoh H, Inoue K, Kawase Y, Jishage K, Suzuki H, Takeya M, Schnackenberg L, Beger R, Hermonat PL, Thomas M, Sawamura T. Deletion of LOX-1 reduces atherogenesis in LDLR knockout mice fed high cholesterol diet. Circ Res. 2007;100(11):1634–1642. [PubMed] [Google Scholar]
120. Suzuki H, Kurihara Y, Takeya M, Kamada N, Kataoka M, Jishage K, Ueda O, Sakaguchi H, Higashi T, Suzuki T, Takashima Y, Kawabe Y, Cynshi O, Wada Y, Honda M, Kurihara H, Aburatani H, Doi T, Matsumoto A, Azuma S, Noda T, Toyoda Y, Itakura H, Yazaki Y, Kodama T. A role for macrophage scavenger receptors in atherosclerosis and susceptibility to infection. Nature. 1997;386(6622):292–296. [PubMed] [Google Scholar]
121. Lambert G, Sakai N, Vaisman BL, Neufeld EB, Marteyn B, Chan CC, Paigen B, Lupia E, Thomas A, Striker LJ, Blanchette-Mackie J, Csako G, Brady JN, Costello R, Striker GE, Remaley AT, Brewer HB, Jr, Santamarina-Fojo S. Analysis of glomerulosclerosis and atherosclerosis in lecithin cholesterol acyltransferase-deficient mice. J Biol Chem. 2001;276(18):15090–15098. [PubMed] [Google Scholar]
122. Zabalawi M, Bhat S, Loughlin T, Thomas MJ, Alexander E, Cline M, Bullock B, Willingham M, Sorci-Thomas MG. Induction of fatal inflammation in LDL receptor and ApoA-I double-knockout mice fed dietary fat and cholesterol. Am J Pathol. 2003;163(3):1201–1213. [PMC free article] [PubMed] [Google Scholar]
123. Mamontova A, Seguret-Mace S, Esposito B, Chaniale C, Bouly M, Delhaye-Bouchaud N, Luc G, Staels B, Duverger N, Mariani J, Tedgui A. Severe atherosclerosis and hypoalphalipoproteinemia in the staggerer mouse, a mutant of the nuclear receptor RORalpha. Circulation. 1998;98(24):2738–2743. [PubMed] [Google Scholar]
124. Mezdour H, Jones R, Dengremont C, Castro G, Maeda N. Hepatic lipase deficiency increases plasma cholesterol but reduces susceptibility to atherosclerosis in apolipoprotein E-deficient mice. J Biol Chem. 1997;272(21):13570–13575. [PubMed] [Google Scholar]
125. Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C. IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest. 1997;99(11):2752–2761. [PMC free article] [PubMed] [Google Scholar]
126. Hanniman EA, Lambert G, McCarthy TC, Sinal CJ. Loss of functional farnesoid X receptor increases atherosclerotic lesions in apolipoprotein E-deficient mice. J Lipid Res. 2005;46(12):2595–2604. [PubMed] [Google Scholar]
127. Willner EL, Tow B, Buhman KK, Wilson M, Sanan DA, Rudel LL, Farese RV., Jr Deficiency of acyl CoA:cholesterol acyltransferase 2 prevents atherosclerosis in apolipoprotein E-deficient mice. Proc Natl Acad Sci U S A. 2003;100(3):1262–1267. [PMC free article] [PubMed] [Google Scholar]
128. Adachi H, Fujiwara Y, Kondo T, Nishikawa T, Ogawa R, Matsumura T, Ishii N, Nagai R, Miyata K, Tabata M, Motoshima H, Furukawa N, Tsuruzoe K, Kawashima J, Takeya M, Yamashita S, Koh GY, Nagy A, Suda T, Oike Y, Araki E. Angptl 4 deficiency improves lipid metabolism, suppresses foam cell formation and protects against atherosclerosis. Biochem Biophys Res Commun. 2009;379(4):806–811. [PubMed] [Google Scholar]
129. Ishida T, Choi SY, Kundu RK, Spin J, Yamashita T, Hirata K, Kojima Y, Yokoyama M, Cooper AD, Quertermous T. Endothelial lipase modulates susceptibility to atherosclerosis in apolipoprotein-E-deficient mice. J Biol Chem. 2004;279(43):45085–45092. [PubMed] [Google Scholar]
130. Feng X, Li H, Rumbin AA, Wang X, La Cava A, Brechtelsbauer K, Castellani LW, Witztum JL, Lusis AJ, Tsao BP. ApoE−/−Fas−/− C57BL/6 mice: a novel murine model simultaneously exhibits lupus nephritis, atherosclerosis, and osteopenia. J Lipid Res. 2007;48(4):794–805. [PubMed] [Google Scholar]
131. Chamberlain J, Francis S, Brookes Z, Shaw G, Graham D, Alp NJ, Dower S, Crossman DC. Interleukin-1 regulates multiple atherogenic mechanisms in response to fat feeding. PLoS ONE. 2009;4(4):e5073. [PMC free article] [PubMed] [Google Scholar]
132. Potteaux S, Esposito B, van Oostrom O, Brun V, Ardouin P, Groux H, Tedgui A, Mallat Z. Leukocyte-derived interleukin 10 is required for protection against atherosclerosis in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol. 2004;24(8):1474–1478. [PubMed] [Google Scholar]
133. Bourdillon MC, Randon J, Barek L, Zibara K, Covacho C, Poston RN, Chignier E, McGregor JL. Reduced atherosclerotic lesion size in P-selectin deficient apolipoprotein E-knockout mice fed a chow but not a fat diet. J Biomed Biotechnol. 2006;2006(2):49193. [PMC free article] [PubMed] [Google Scholar]
134. Homeister JW, Daugherty A, Lowe JB. Alpha(1,3)fucosyltransferases FucT-IV and FucT-VII control susceptibility to atherosclerosis in apolipoprotein E−/− mice. Arterioscler Thromb Vasc Biol. 2004;24(10):1897–1903. [PubMed] [Google Scholar]
135. Sussan TE, Jun J, Thimmulappa R, Bedja D, Antero M, Gabrielson KL, Polotsky VY, Biswal S. Disruption of Nrf2, a key inducer of antioxidant defenses, attenuates ApoE-mediated atherosclerosis in mice. PLoS ONE. 2008;3(11):e3791. [PMC free article] [PubMed] [Google Scholar]
136. Johnson JL, George SJ, Newby AC, Jackson CL. Divergent effects of matrix metalloproteinases 3, 7, 9, and 12 on atherosclerotic plaque stability in mouse brachiocephalic arteries. Proc Natl Acad Sci U S A. 2005;102(43):15575–15580. [PMC free article] [PubMed] [Google Scholar]
137. Sekiya M, Osuga J, Nagashima S, Ohshiro T, Igarashi M, Okazaki H, Takahashi M, Tazoe F, Wada T, Ohta K, Takanashi M, Kumagai M, Nishi M, Takase S, Yahagi N, Yagyu H, Ohashi K, Nagai R, Kadowaki T, Furukawa Y, Ishibashi S. Ablation of neutral cholesterol ester hydrolase 1 accelerates atherosclerosis. Cell Metab. 2009;10(3):219–228. [PubMed] [Google Scholar]
138. Tupin E, Nicoletti A, Elhage R, Rudling M, Ljunggren HG, Hansson GK, Berne GP. CD1d-dependent activation of NKT cells aggravates atherosclerosis. J Exp Med. 2004;199(3):417–422. [PMC free article] [PubMed] [Google Scholar]
139. Arai S, Shelton JM, Chen M, Bradley MN, Castrillo A, Bookout AL, Mak PA, Edwards PA, Mangelsdorf DJ, Tontonoz P, Miyazaki T. A role for the apoptosis inhibitory factor AIM/Spalpha/Api6 in atherosclerosis development. Cell Metab. 2005;1(3):201–213. [PubMed] [Google Scholar]
140. Sukhova GK, Zhang Y, Pan JH, Wada Y, Yamamoto T, Naito M, Kodama T, Tsimikas S, Witztum JL, Lu ML, Sakara Y, Chin MT, Libby P, Shi GP. Deficiency of cathepsin S reduces atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2003;111(6):897–906. [PMC free article] [PubMed] [Google Scholar]
141. Chadjichristos CE, Scheckenbach KE, van Veen TA, Richani Sarieddine MZ, de Wit C, Yang Z, Roth I, Bacchetta M, Viswambharan H, Foglia B, Dudez T, van Kempen MJ, Coenjaerts FE, Miquerol L, Deutsch U, Jongsma HJ, Chanson M, Kwak BR. Endothelial-specific deletion of connexin40 promotes atherosclerosis by increasing CD73-dependent leukocyte adhesion. Circulation. 2010;121(1):123–131. [PubMed] [Google Scholar]
142. Yu Y, Lucitt MB, Stubbe J, Cheng Y, Friis UG, Hansen PB, Jensen BL, Smyth EM, FitzGerald GA. Prostaglandin F2alpha elevates blood pressure and promotes atherosclerosis. Proc Natl Acad Sci U S A. 2009;106(19):7985–7990. [PMC free article] [PubMed] [Google Scholar]
143. Terasawa Y, Ladha Z, Leonard SW, Morrow JD, Newland D, Sanan D, Packer L, Traber MG, Farese RV., Jr Increased atherosclerosis in hyperlipidemic mice deficient in alpha -tocopherol transfer protein and vitamin E. Proc Natl Acad Sci U S A. 2000;97(25):13830–13834. [PMC free article] [PubMed] [Google Scholar]
144. Michelsen KS, Wong MH, Shah PK, Zhang W, Yano J, Doherty TM, Akira S, Rajavashisth TB, Arditi M. Lack of Toll-like receptor 4 or myeloid differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E. Proc Natl Acad Sci U S A. 2004;101(29):10679–10684. [PMC free article] [PubMed] [Google Scholar]
145. Knowles JW, Reddick RL, Jennette JC, Shesely EG, Smithies O, Maeda N. Enhanced atherosclerosis and kidney dysfunction in eNOS(−/−)Apoe(−/−) mice are ameliorated by enalapril treatment. J Clin Invest. 2000;105(4):451–458. [PMC free article] [PubMed] [Google Scholar]
146. Boisvert WA, Rose DM, Johnson KA, Fuentes ME, Lira SA, Curtiss LK, Terkeltaub RA. Up-regulated expression of the CXCR2 ligand KC/GRO-alpha in atherosclerotic lesions plays a central role in macrophage accumulation and lesion progression. Am J Pathol. 2006;168(4):1385–1395. [PMC free article] [PubMed] [Google Scholar]
147. Heller EA, Liu E, Tager AM, Yuan Q, Lin AY, Ahluwalia N, Jones K, Koehn SL, Lok VM, Aikawa E, Moore KJ, Luster AD, Gerszten RE. Chemokine CXCL10 promotes atherogenesis by modulating the local balance of effector and regulatory T cells. Circulation. 2006;113(19):2301–2312. [PubMed] [Google Scholar]
148. 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):1318–1331. [PMC free article] [PubMed] [Google Scholar]
149. Paigen B, Holmes PA, Novak EK, Swank RT. Analysis of atherosclerosis susceptibility in mice with genetic defects in platelet function. Arteriosclerosis. 1990;10(4):648–652. [PubMed] [Google Scholar]
150. Barry-Lane PA, Patterson C, van der Merwe M, Hu Z, Holland SM, Yeh ET, Runge MS. p47phox is required for atherosclerotic lesion progression in ApoE(−/−) mice. J Clin Invest. 2001;108(10):1513–1522. [PMC free article] [PubMed] [Google Scholar]
151. Shih DM, Gu L, Xia YR, Navab M, Li WF, Hama S, Castellani LW, Furlong CE, Costa LG, Fogelman AM, Lusis AJ. Mice lacking serum paraoxonase are susceptible to organophosphate toxicity and atherosclerosis. Nature. 1998;394(6690):284–287. [PubMed] [Google Scholar]
152. Methia N, Andre P, Denis CV, Economopoulos M, Wagner DD. Localized reduction of atherosclerosis in von Willebrand factor-deficient mice. Blood. 2001;98(5):1424–1428. [PubMed] [Google Scholar]
153. Kanters E, Pasparakis M, Gijbels MJ, Vergouwe MN, Partouns-Hendriks I, Fijneman RJ, Clausen BE, Forster I, Kockx MM, Rajewsky K, Kraal G, Hofker MH, de Winther MP. Inhibition of NF-kappaB activation in macrophages increases atherosclerosis in LDL receptor-deficient mice. J Clin Invest. 2003;112(8):1176–1185. [PMC free article] [PubMed] [Google Scholar]
154. Cole JE, Navin TJ, Cross AJ, Goddard ME, Alexopoulou L, Mitra AT, Davies AH, Flavell RA, Feldmann M, Monaco C. From the Cover: Unexpected protective role for Toll-like receptor 3 in the arterial wall. Proc Natl Acad Sci U S A. 2011;108(6):2372–2377. [PMC free article] [PubMed] [Google Scholar]
155. Saederup N, Chan L, Lira SA, Charo IF. Fractalkine deficiency markedly reduces macrophage accumulation and atherosclerotic lesion formation in CCR2−/− mice: evidence for independent chemokine functions in atherogenesis. Circulation. 2008;117(13):1642–1648. [PMC free article] [PubMed] [Google Scholar]
156. Deuchar GA, McLean D, Hadoke PW, Brownstein DG, Webb DJ, Mullins JJ, Chapman K, Seckl JR, Kotelevtsev YV. 11beta-hydroxysteroid dehydrogenase type 2 deficiency accelerates atherogenesis and causes proinflammatory changes in the endothelium in apoe−/− mice. Endocrinology. 2011;152(1):236–246. [PMC free article] [PubMed] [Google Scholar]
157. Taniyama Y, Fuse H, Satomi T, Tozawa R, Yasuhara Y, Shimakawa K, Shibata S, Hattori M, Nakata M, Taketomi S. Loss of lysophospholipase 3 increases atherosclerosis in apolipoprotein E-deficient mice. Biochem Biophys Res Commun. 2005;330(1):104–110. [PubMed] [Google Scholar]
158. Lewis P, Stefanovic N, Pete J, Calkin AC, Giunti S, Thallas-Bonke V, Jandeleit-Dahm KA, Allen TJ, Kola I, Cooper ME, de Haan JB. Lack of the antioxidant enzyme glutathione peroxidase-1 accelerates atherosclerosis in diabetic apolipoprotein E-deficient mice. Circulation. 2007;115(16):2178–2187. [PubMed] [Google Scholar]
159. Lesnik P, Haskell CA, Charo IF. Decreased atherosclerosis in CX3CR1−/− mice reveals a role for fractalkine in atherogenesis. J Clin Invest. 2003;111(3):333–340. [PMC free article] [PubMed] [Google Scholar]
160. Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2−/− mice reveals a role for chemokines in the initiation of atherosclerosis. Nature. 1998;394(6696):894–897. [PubMed] [Google Scholar]
161. Yamada S, Ding Y, Tanimoto A, Wang KY, Guo X, Li Z, Tasaki T, Nabesima A, Murata Y, Shimajiri S, Kohno K, Ichijo H, Sasaguri Y. Apoptosis Signal-Regulating Kinase 1 Deficiency Accelerates Hyperlipidemia-Induced Atheromatous Plaques via Suppression of Macrophage Apoptosis. Arterioscler Thromb Vasc Biol. 2011;31(7):1555–1564. [PubMed] [Google Scholar]
162. Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW, Lichtman AH. Influence of interferon-gamma on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. Arterioscler Thromb Vasc Biol. 2003;23(3):454–460. [PubMed] [Google Scholar]
163. Boucher P, Gotthardt M, Li WP, Anderson RG, Herz J. LRP: role in vascular wall integrity and protection from atherosclerosis. Science. 2003;300(5617):329–332. [PubMed] [Google Scholar]
164. Ricci R, Sumara G, Sumara I, Rozenberg I, Kurrer M, Akhmedov A, Hersberger M, Eriksson U, Eberli FR, Becher B, Boren J, Chen M, Cybulsky MI, Moore KJ, Freeman MW, Wagner EF, Matter CM, Luscher TF. Requirement of JNK2 for scavenger receptor A-mediated foam cell formation in atherogenesis. Science. 2004;306(5701):1558–1561. [PubMed] [Google Scholar]
165. Cyrus T, Witztum JL, Rader DJ, Tangirala R, Fazio S, Linton MF, Funk CD. Disruption of the 12/15-lipoxygenase gene diminishes atherosclerosis in apo E-deficient mice. J Clin Invest. 1999;103(11):1597–1604. [PMC free article] [PubMed] [Google Scholar]
166. Aslanian AM, Charo IF. Targeted disruption of the scavenger receptor and chemokine CXCL16 accelerates atherosclerosis. Circulation. 2006;114(6):583–590. [PubMed] [Google Scholar]
167. Romeo GR, Moulton KS, Kazlauskas A. Attenuated expression of profilin-1 confers protection from atherosclerosis in the LDL receptor null mouse. Circ Res. 2007;101(4):357–367. [PubMed] [Google Scholar]
168. Kuhlencordt PJ, Chen J, Han F, Astern J, Huang PL. Genetic deficiency of inducible nitric oxide synthase reduces atherosclerosis and lowers plasma lipid peroxides in apolipoprotein E-knockout mice. Circulation. 2001;103(25):3099–3104. [PubMed] [Google Scholar]
169. Gu L, Okada Y, Clinton SK, Gerard C, Sukhova GK, Libby P, Rollins BJ. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol Cell. 1998;2(2):275–281. [PubMed] [Google Scholar]
170. Brennan ML, Anderson MM, Shih DM, Qu XD, Wang X, Mehta AC, Lim LL, Shi W, Hazen SL, Jacob JS, Crowley JR, Heinecke JW, Lusis AJ. Increased atherosclerosis in myeloperoxidase-deficient mice. J Clin Invest. 2001;107(4):419–430. [PMC free article] [PubMed] [Google Scholar]
171. Buono C, Binder CJ, Stavrakis G, Witztum JL, Glimcher LH, Lichtman AH. T-bet deficiency reduces atherosclerosis and alters plaque antigen-specific immune responses. Proc Natl Acad Sci U S A. 2005;102(5):1596–1601. [PMC free article] [PubMed] [Google Scholar]
172. Weng S, Zemany L, Standley KN, Novack DV, La Regina M, Bernal-Mizrachi C, Coleman T, Semenkovich CF. Beta3 integrin deficiency promotes atherosclerosis and pulmonary inflammation in high-fat-fed, hyperlipidemic mice. Proc Natl Acad Sci U S A. 2003;100(11):6730–6735. [PMC free article] [PubMed] [Google Scholar]
173. Hayek T, Pavlotzky E, Hamoud S, Coleman R, Keidar S, Aviram M, Kaplan M. Tissue angiotensin-converting-enzyme (ACE) deficiency leads to a reduction in oxidative stress and in atherosclerosis: studies in ACE-knockout mice type 2. Arterioscler Thromb Vasc Biol. 2003;23(11):2090–2096. [PubMed] [Google Scholar]
174. Goel R, Schrank BR, Arora S, Boylan B, Fleming B, Miura H, Newman PJ, Molthen RC, Newman DK. Site-specific effects of PECAM-1 on atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2008;28(11):1996–2002. [PMC free article] [PubMed] [Google Scholar]
175. Reue K, Xu P, Wang XP, Slavin BG. Adipose tissue deficiency, glucose intolerance, and increased atherosclerosis result from mutation in the mouse fatty liver dystrophy (fld) gene. J Lipid Res. 2000;41(7):1067–1076. [PubMed] [Google Scholar]
176. Chang JD, Sukhova GK, Libby P, Schvartz E, Lichtenstein AH, Field SJ, Kennedy C, Madhavarapu S, Luo J, Wu D, Cantley LC. Deletion of the phosphoinositide 3-kinase p110gamma gene attenuates murine atherosclerosis. Proc Natl Acad Sci U S A. 2007;104(19):8077–8082. [PMC free article] [PubMed] [Google Scholar]
177. Parks BW, Lusis AJ, Kabarowski JH. Loss of the lysophosphatidylcholine effector, G2A, ameliorates aortic atherosclerosis in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol. 2006;26(12):2703–2709. [PubMed] [Google Scholar]
178. Nachtigal M, Ghaffar A, Mayer EP. Galectin-3 gene inactivation reduces atherosclerotic lesions and adventitial inflammation in ApoE-deficient mice. Am J Pathol. 2008;172(1):247–255. [PMC free article] [PubMed] [Google Scholar]
179. Subbarao K, Jala VR, Mathis S, Suttles J, Zacharias W, Ahamed J, Ali H, Tseng MT, Haribabu B. Role of leukotriene B4 receptors in the development of atherosclerosis: potential mechanisms. Arterioscler Thromb Vasc Biol. 2004;24(2):369–375. [PubMed] [Google Scholar]
180. !!! INVALID CITATION !!!
181. Bennett BJ, Scatena M, Kirk EA, Rattazzi M, Varon RM, Averill M, Schwartz SM, Giachelli CM, Rosenfeld ME. Osteoprotegerin inactivation accelerates advanced atherosclerotic lesion progression and calcification in older ApoE−/− mice. Arterioscler Thromb Vasc Biol. 2006;26(9):2117–2124. [PubMed] [Google Scholar]
182. Aihara K, Azuma H, Akaike M, Ikeda Y, Sata M, Takamori N, Yagi S, Iwase T, Sumitomo Y, Kawano H, Yamada T, Fukuda T, Matsumoto T, Sekine K, Sato T, Nakamichi Y, Yamamoto Y, Yoshimura K, Watanabe T, Nakamura T, Oomizu A, Tsukada M, Hayashi H, Sudo T, Kato S. Strain-dependent embryonic lethality and exaggerated vascular remodeling in heparin cofactor II-deficient mice. J Clin Invest. 2007;117(6):1514–1526. [PMC free article] [PubMed] [Google Scholar]
183. Xiao Q, Danton MJ, Witte DP, Kowala MC, Valentine MT, Bugge TH, Degen JL. Plasminogen deficiency accelerates vessel wall disease in mice predisposed to atherosclerosis. Proc Natl Acad Sci U S A. 1997;94(19):10335–10340. [PMC free article] [PubMed] [Google Scholar]
184. Merched AJ, Chan L. Absence of p21Waf1/Cip1/Sdi1 modulates macrophage differentiation and inflammatory response and protects against atherosclerosis. Circulation. 2004;110(25):3830–3841. [PubMed] [Google Scholar]
185. Franco C, Hou G, Ahmad PJ, Fu EY, Koh L, Vogel WF, Bendeck MP. Discoidin domain receptor 1 (ddr1) deletion decreases atherosclerosis by accelerating matrix accumulation and reducing inflammation in low-density lipoprotein receptor-deficient mice. Circ Res. 2008;102(10):1202–1211. [PubMed] [Google Scholar]
186. Harja E, Bucciarelli LG, Lu Y, Stern DM, Zou YS, Schmidt AM, Yan SF. Early growth response-1 promotes atherogenesis: mice deficient in early growth response-1 and apolipoprotein E display decreased atherosclerosis and vascular inflammation. Circ Res. 2004;94(3):333–339. [PubMed] [Google Scholar]
187. Thomas MC, Pickering RJ, Tsorotes D, Koitka A, Sheehy K, Bernardi S, Toffoli B, Nguyen-Huu TP, Head GA, Fu Y, Chin-Dusting J, Cooper ME, Tikellis C. Genetic Ace2 deficiency accentuates vascular inflammation and atherosclerosis in the ApoE knockout mouse. Circ Res. 2010;107(7):888–897. [PubMed] [Google Scholar]
188. Paigen B, Mitchell D, Holmes PA, Albee D. Genetic analysis of strains C57BL/6J and BALB/cJ for Ath-1, a gene determining atherosclerosis susceptibility in mice. Biochem Genet. 1987;25(11–12):881–892. [PubMed] [Google Scholar]
189. Wang SS, Schadt EE, Wang H, Wang X, Ingram-Drake L, Shi W, Drake TA, Lusis AJ. Identification of pathways for atherosclerosis in mice: integration of quantitative trait locus analysis and global gene expression data. Circ Res. 2007;101(3):e11–e30. [PubMed] [Google Scholar]
190. Tomita H, Zhilicheva S, Kim S, Maeda N. Aortic arch curvature and atherosclerosis have overlapping quantitative trait loci in a cross between 129S6/SvEvTac and C57BL/6J apolipoprotein E-null mice. Circ Res. 2010;106(6):1052–1060. [PMC free article] [PubMed] [Google Scholar]
191. Dansky HM, Shu P, Donavan M, Montagno J, Nagle DL, Smutko JS, Roy N, Whiteing S, Barrios J, McBride TJ, Smith JD, Duyk G, Breslow JL, Moore KJ. A phenotype-sensitizing Apoe-deficient genetic background reveals novel atherosclerosis predisposition loci in the mouse. Genetics. 2002;160(4):1599–1608. [PMC free article] [PubMed] [Google Scholar]
192. Paigen B, Nesbitt MN, Mitchell D, Albee D, LeBoeuf RC. Ath-2, a second gene determining atherosclerosis susceptibility and high density lipoprotein levels in mice. Genetics. 1989;122(1):163–168. [PMC free article] [PubMed] [Google Scholar]
193. Smith JD, Bhasin JM, Baglione J, Settle M, Xu Y, Barnard J. Atherosclerosis susceptibility loci identified from a strain intercross of apolipoprotein E-deficient mice via a high-density genome scan. Arterioscler Thromb Vasc Biol. 2006;26(3):597–603. [PubMed] [Google Scholar]
194. 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(1):204–210. [PMC free article] [PubMed] [Google Scholar]
195. Welch CL, Bretschger S, Latib N, Bezouevski M, Guo Y, Pleskac N, Liang CP, Barlow C, Dansky H, Breslow JL, Tall AR. Localization of atherosclerosis susceptibility loci to chromosomes 4 and 6 using the Ldlr knockout mouse model. Proc Natl Acad Sci U S A. 2001;98(14):7946–7951. [PMC free article] [PubMed] [Google Scholar]
196. Korstanje R, Eriksson P, Samnegard A, Olsson PG, Forsman-Semb K, Sen S, Churchill GA, Rollins J, Harris S, Hamsten A, Paigen B. Locating Ath8, a locus for murine atherosclerosis susceptibility and testing several of its candidate genes in mice and humans. Atherosclerosis. 2004;177(2):443–450. [PubMed] [Google Scholar]
197. Wang SS, Shi W, Wang X, Velky L, Greenlee S, Wang MT, Drake TA, Lusis AJ. Mapping, genetic isolation, and characterization of genetic loci that determine resistance to atherosclerosis in C3H mice. Arterioscler Thromb Vasc Biol. 2007;27(12):2671–2676. [PubMed] [Google Scholar]
198. Colinayo VV, Qiao JH, Wang X, Krass KL, Schadt E, Lusis AJ, Drake TA. Genetic loci for diet-induced atherosclerotic lesions and plasma lipids in mice. Mamm Genome. 2003;14(7):464–471. [PubMed] [Google Scholar]
199. Stewart-Phillips JL, Lough J, Skamene E. ATH-3, a new gene for atherosclerosis in the mouse. Clin Invest Med. 1989;12(2):121–126. [PubMed] [Google Scholar]
200. Su Z, Li Y, James JC, McDuffie M, Matsumoto AH, Helm GA, Weber JL, Lusis AJ, Shi W. Quantitative trait locus analysis of atherosclerosis in an intercross between C57BL/6 and C3H mice carrying the mutant apolipoprotein E gene. Genetics. 2006;172(3):1799–1807. [PMC free article] [PubMed] [Google Scholar]
201. Ishimori N, Li R, Kelmenson PM, Korstanje R, Walsh KA, Churchill GA, Forsman-Semb K, Paigen B. Quantitative trait loci analysis for plasma HDL-cholesterol concentrations and atherosclerosis susceptibility between inbred mouse strains C57BL/6J and 129S1/SvImJ. Arterioscler Thromb Vasc Biol. 2004;24(1):161–166. [PubMed] [Google Scholar]
202. Davis RC, Schadt EE, Cervino AC, Peterfy M, Lusis AJ. Ultrafine mapping of SNPs from mouse strains C57BL/6J, DBA/2J, and C57BLKS/J for loci contributing to diabetes and atherosclerosis susceptibility. Diabetes. 2005;54(4):1191–1199. [PubMed] [Google Scholar]
203. Purcell MK, Mu JL, Higgins DC, Elango R, Whitmore H, Harris S, Paigen B. Fine mapping of Ath6, a quantitative trait locus for atherosclerosis in mice. Mamm Genome. 2001;12(7):495–500. [PubMed] [Google Scholar]
204. 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(7):1328–1335. [PubMed] [Google Scholar]
205. Li Q, Li Y, Zhang Z, Gilbert TR, Matsumoto AH, Dobrin SE, Shi W. Quantitative trait locus analysis of carotid atherosclerosis in an intercross between C57BL/6 and C3H apolipoprotein E-deficient mice. Stroke. 2008;39(1):166–173. [PMC free article] [PubMed] [Google Scholar]
-