Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Feb 10.
Published in final edited form as: Arterioscler Thromb Vasc Biol. 2007 Jul 26;27(10):2068–2078. doi: 10.1161/01.ATV.0000282199.66398.8c

Genetic Susceptibility to Peripheral Arterial Disease: A Dark Corner in Vascular Biology

Joshua W Knowles 1, Themistocles L Assimes 1, Jun Li 1, Thomas Quertermous 1, John P Cooke 1
PMCID: PMC4321902  NIHMSID: NIHMS151807  PMID: 17656669

Abstract

Peripheral arterial disease (PAD) is characterized by reduced blood flow to the limbs, usually as a consequence of atherosclerosis, and affects ≈12 million Americans. It is a common cause of cardiovascular morbidity and an independent predictor of cardiovascular mortality. Similar to other atherosclerotic diseases, such as coronary artery disease, PAD is the result of the complex interplay between injurious environmental stimuli and genetic predisposing factors of the host. Genetic susceptibility to PAD is likely contributed by sequence variants in multiple genes, each with modest effects. Although many of these variants probably alter susceptibility both to PAD and to coronary artery disease, it is likely that there exists a set of variants specifically to alter susceptibility to PAD. Despite the prevalence of PAD and its high societal burden, relatively little is known about such genetic variants. This review summarizes our limited present knowledge and gives an overview of recent, more powerful approaches to elucidating the genetic basis of PAD. We discuss the advantages and limitations of genetic studies and highlight the need for collaborative networks of PAD investigators for shedding light on this dark corner of vascular biology.

Keywords: peripheral vascular disease, genetics, epidemiology, atherosclerosis

Epidemiology, Diagnosis, and Established Risk Factors of PAD

Peripheral arterial disease (PAD) is a disease characterized by reduced blood flow to the lower extremities most often because of atherosclerosis. The prevalence of PAD varies substantially with age and certain risk factors. In population-based studies of adults, estimates of the prevalence of PAD range from 3% to 19%.19 In high-risk groups, such as subjects over the age of 50 years with a history of diabetes or smoking or subjects over the age of 70 years, the prevalence of PAD is >25%.10 In North America and Europe, PAD affects an estimated 27 million adults.8,11

PAD is a leading cause of morbidity through functional decline, intermittent claudication, and critical leg ischemia.10,12,13 PAD is also a significant predictor of cardiovascular mortality. Patients with PAD are twice as likely to have prevalent CAD as those without disease.1,2 Both asymptomatic and symptomatic PAD are associated with increased risk of stroke, myocardial infarction, and death.5,6,1416 In particular, the risk of cardiovascular death over a 10-year period is increased 6-fold in patients with PAD.14

Office-based diagnosis of PAD is made by medical history, physical examination, and the measurement of ankle blood pressures using a hand-held Doppler ultrasound device. The ankle brachial index (ABI) is defined as the ratio of ankle pressure to the brachial systolic pressure. The reference range of ABI is 0.91 to 1.3 (systolic pressure is normally slightly higher at the ankle than the arm because of pulse-wave reflections along the length of the limb arteries).17 An ABI <0.9 is consistent with a diagnosis of PAD, and the lower the ABI, the more hemodynamically severe the disease (ie, 0.90 is mild, 0.70 is moderate, and ≤0.5 is severe PAD). Individuals with an ABI >1.30 likely have noncompliant vessels secondary to pathological processes involving vascular fibrosis and calcification; these individuals tend to evidence greater coronary artery calcium.18 Some have suggested a tighter range for normal ABI, because individuals with an ABI between 0.90 and 1.00 manifest more ischemic leg pain,19 subclinical atherosclerosis,18,20,21 and are at greater risk for major adverse cardiovascular events.22,23 Although the ABI is an excellent screening test for the presence of PAD, it correlates only modestly with functional limitation.24,25 For example, a severe reduction in ABI (eg, an ABI of <0.5) is typically observed in a patient with resting pain or tissue loss. However, a similar ABI might be discovered in an individual with modest symptomatology (eg, calf discomfort after walking 2 blocks). These differences may be because of heterogeneity in collateral formation or skeletal muscle adaptation. Because most patients are asymptomatic or have atypical symptoms12 and because ABI measurements are often not performed or reimbursed, PAD frequently goes undiagnosed and untreated.3,810,16 The 2005 American College of Cardiology/American Heart Association Practice Guidelines for the Management of Patients with PAD recommend that all high-risk individuals have ABIs measured at least once.17

Atherosclerosis is a result of various injurious exposures that lead to endothelial dysfunction followed by chronic inflammatory infiltration in the arterial wall.2631 Not surprisingly, there is substantial overlap between the pathogenesis of PAD and other forms of systemic atherosclerotic diseases, like coronary artery disease (CAD) and carotid disease.9 PAD and CAD share several risk factors, including diabetes, smoking, hypertension, insulin resistance, low high-density lipoprotein levels, elevated low-density lipoprotein levels, and advanced age. Approximately 70% of the cases of PAD (as defined by ABI) can be attributed to these risk factors,4 somewhat higher than the corresponding estimates for CAD. Convincing evidence exists that some of these risk factors have proportionately greater effects on the development of PAD than CAD. For instance, diabetes mellitus (DM) and smoking are particularly strong risk factors for PAD.22,32,33 Hyperhomocysteinemia may also play a greater role in PAD than in CAD.34 However, even in the presence of traditional risk factors, the progression of PAD seems to be highly variable, which suggests the presence of other determinants of disease, at least some of which are probably inherited and, thus, genetic in nature.

Despite the prevalence and the high societal cost attributed to PAD,35 genetic susceptibility to PAD remains a “dark corner” in vascular biology. Our understanding of the genetic basis of PAD is limited and, so far, based on studies that examine a small number of genes in a small number of samples.28,3638 However, the completion of the human genome draft sequence,39 the cataloging of common human DNA variation by the International HapMap Project,40,41 and the technology for large-scale data collection have provided new opportunities to carry out more powerful studies that will hopefully more definitively identify genetic determinants of PAD.41 Currently, there are no genetic tests available that reliably identify the subset of subjects carrying inherited risk factors for developing PAD. Such tests could potentially be developed if genetic determinants of PAD are uncovered.

In this review, we first summarize and critically appraise the results of studies that support the role of genetics in the pathogenesis of atherosclerotic PAD. We do not discuss syndromic forms of PAD, such as those seen in Buerger's disease, Ehlers-Danlos syndrome, or systemic vasculitides, which are pathologically distinct from “garden-variety” atherosclerotic PAD and are likely to have different genetic contributions. We then summarize the results of existing studies of novel genetic determinants of PAD, highlighting the basic principles, strengths, and weaknesses of various study designs. Thereafter, we conclude by describing novel, more comprehensive, and hopefully more successful approaches to the discovery of genetic determinants of PAD.

The Role of Genetics in PAD

An important first step in the field of complex trait genetics is to estimate the contribution of inherited factors to the development of the disease. This estimate can be obtained through various types of familial aggregation studies.4247

Three family studies to date have reported estimates of the heritability of PAD.4547 In all 3 of the studies, the ABI was used as a surrogate for the presence and severity of PAD. In the first report from the National Heart, Lung, and Blood Institute twin study,45 ≈48% of the overall variability in the ABI could be attributed to additive genetic effects after adjusting for risk factors of atherosclerosis. However, inconsistent with this observation was the lack of a statistically significant higher concordance rate for an ABI <0.9 in monozygotic twins (33%) compared with dizygotic twins (31%). Of note, the authors of this study acknowledged that the estimation of genetic effects may have been biased by selective mortality and loss to follow-up of participants at high risk for PAD. Furthermore, twin studies are also known to overestimate genetic effects because of the presence of a higher degree of shared environmental factors compare with nontwin studies.48,49 A second study of hypertensive nontwin sibs participating in the Genetic Epidemiology Network of Arteriopathy Study46 reported an overall lower estimate of the heritability of ABI. In non-Hispanic whites, this estimate was 35.7% (P<0.001) before adjustment for atherosclerosis risk factors and 21.2% (P=0.006) after adjustment of risk factors. Estimates of heritability before and after adjustment for covariates were similar in African Americans (35.7%, P<0.001 and 19.5%, P=0.002) participating in the Genetic Epidemiology Network of Arteriopathy Study. In the third and final family study, investigators from the Framingham Offspring Study47 estimated a heritability of ABI similar to the one reported in the Genetic Epidemiology Network of Arteriopathy cohort. Using variance component analysis, this estimate was 27% before adjustment for covariates and 21% after adjustment for covariates (both P values <0.0001).

A significant limitation of the heritability studies mentioned above is that a large majority of subjects had ABIs in the borderline or reference range (>0.9).4547 In this range, there is either no correlation of the ABI with the degree of lower extremity atherosclerosis or it is very poor.16,21 Thus, heritability studies of ABI to date may reflect more the degree of genetic influence of ABI in the reference range, which may differ significantly from the degree of the genetic influence on PAD. Consistent with this possibility is the fact that established strong predictors of clinically significant PAD explain a relatively small proportion of the total variance of ABI in the 2 family studies that reported these estimates.45,47

Evidence quite suggestive for a genetic basis of PAD comes from a study of Valentine et al43 where they showed that both premature, symptomatic CAD and occult lower-extremity vascular disease are more common in families of probands with premature PAD, indicating a strong familial aggregation of vascular disease. However, this study was not able to quantify the degree of familial aggregation that was independent of that related to smoking and other established risk factors of atherosclerosis.

A report from the National Heart, Lung, and Blood Institute Multi-Ethnic Study of Atherosclerosis on the determinants of PAD provides some indirect evidence for the role of genes in the development of PAD.44 In this report, self-reported race/ethnic group predicted the presence of PAD (defined as an ABI <0.9) independent of all other “established” and “novel” cardiovascular disease risk factors of atherosclerosis with the lowest risk in Hispanics and Chinese and the highest risk in African Americans. These results suggest the presence of determinants of PAD that correlate with race, some of which could be genetic in nature. Several other studies have reported greater prevalence of PAD in African American individuals.5052

Collectively, family studies to date do suggest that PAD is heritable. However, what remains to be quantified accurately is the degree of genetic influence on the development of PAD independent of established risk factors of atherosclerosis that also clusters in families, such as smoking, diabetes, hyperlipidemia, and hypertension. What is clearer is that the genetic susceptibility to diseases that result from atherosclerosis is significant and cannot be attributed to a single gene.28,5355 Rather, susceptibility is likely a result of a combination of alleles in multiple genes, each with a modest effect on risk.

Existing Work From Linkage and Case-Control Association Studies of PAD

Traditional linkage studies examine the relationship between the sharing of DNA markers (microsatellite markers or single nucleotide polymorphisms [SNPs]) identity-by-descent and the sharing of phenotype between relatives within families. The conventional likelihood ratio–based test statistic is referred to as the log-odds score. Genome-wide linkage analysis using microsatellite markers often scans markers at a density of 3 to 10 centimorgans. A log-odds score >3 is considered statistically significant.56 Because of the limited number of recombination events within a pedigree, the resolution of linkage study may be quite low.57,58 Once linkage with a marker is detected, other more closely spaced markers in the same region are genotyped in a process referred to as fine mapping. The marker with the strongest signal is usually near 1 or more genes that may be causal for the phenotype under study. Traditionally, linkage studies have been very successful in identifying susceptibility genes with very large effects most commonly seen in diseases that follow Mendelian patterns of inheritance.59,60 However, they have proven to be more difficult to apply to the study of common and complex traits such as PAD.6164

Two linkage studies relating to PAD have been reported to date. In the first study, investigators from deCODE genotyped ≈1000 microsatellite markers in 272 Icelandic patients and 612 relatives (from 116 families)65 and identified a linkage peak on chromosome 1p. During fine-scale mapping of this region, a peak log-odds score of 3.9 was obtained at the marker D1S2895. However, the same linkage signal has not yet been reported in an independent sample. If confirmed, further fine mapping of this region will be needed to identify the causal genetic variant. The second study used ABI as the phenotype and only 350 microsatellites.46 In this study of 1310 African Americans and 796 non-Hispanic whites, no convincing evidence of linkage was reported (highest multi-point log-odds score of 2). However, as mentioned above, a limitation of this study was the high proportion of ABIs in the reference range.

In contrast to linkage analysis, association studies explore differences in allele frequencies between unrelated individuals with and without a particular phenotype/disease. Carefully designed association studies may achieve greater power and finer mapping resolution than family based linkage studies in detecting susceptibility loci with modest effects.30,64,66,67 For many common diseases, association studies are also much more practical to conduct than family based studies. The 2 basic association study designs are the cohort design and the case-control design, with the latter design having been more widely adopted because of its efficiency. To date, there have only been a small number of association studies of PAD examining a total of <20 genes. These studies are summarized in Table 1. All of these studies have used the case-control design with sample sizes ranging from ≈100 to 1300 subjects (median number of case subjects: <200; median number of control subjects: <450). In most of these studies, only 1 polymorphism per gene was genotyped. Collectively, these studies have not convincingly uncovered any novel genetic determinants of PAD.30,3638,57,6671

TABLE 1.

Examples of Existing Case-Control Studies to Identify PAD Susceptibility Genes

Gene HUGO Name Polymorphism (as Described) Case Subjects, No. Control Subjects, No. Ethnicity (as Described) Odds Ratio P Reference
Angiotensin-converting enzyme ACE Insertion/deletion 98 240 German NS 140
Angiotensin-converting enzyme ACE Insertion/deletion 45 316 Chinese NS 141
Angiotensin-converting enzyme ACE Insertion/deletion 522 522 White, Austrian NS 142
Chemokine, CC motif, receptor 5 CCR5 Δ32 76 172 Italian NS 143
Chemokine, CX3C motif, receptor 1 CX3CR1 V249I, T280 mol/L 522 522 White, Austrian NS 144
Cytochrome B α subunit (p22 phox) CYBA C242T 324 295 Austrian NS 145
Factor II F2 G20210A 336 300 Austrian NS 146
Factor II F2 G20210A 184 330 White, French 4.3 0.02 147
Factor II F2 G20210A 433 433 Austrian NS 148
Factor V F5 G1691A (Leiden) 336 300 Austrian NS 146
Factor V F5 G1691A (Leiden) 184 330 White, French NS 147
Factor V F5 G1691A (Leiden) 433 433 Austrian NS 148
Factor VII F7 R353Q 88 423 Scottish NS 149
Factor XIII, A subunit F13A1 V34L 873 523 Austrian NS 150
Fibrinogen FGB G-455A 88 423 Scottish NS 149
Fibrinogen FGB T1689G 104 663 Scottish NS 151
Fibrinogen FGB Bcl I digestion 121 126 Scottish 7.6 0.03 152
Haptoglobin HP Hp1, Hp2 141 1000 White, Belgian 1.82 <0.001 153
Hemochromatosis HFE C282Y, H63D 522 522 White, Austrian NS 154
Hepatic lipase LIPC G-250A 241 241 White, Austrian 1.69 0.02 155
Intracellular adhesion molecule 1 ICAM1 K469E 75 227 White, Italian 3.5 0.004 156
Integrin β3 (platelet glycoprotein IIIa) ITGB3 PLA1/A2 815 518 Austrian NS 157
Integrin β3 (platelet glycoprotein IIIa) ITGB3 P1A 104 663 Scottish 0.49 <0.05 151
Interleukin 6 IL6 G-174C 84 183 White, Italian 4.6 0.001 158
Methylenetetrahydrofolate reductase MTHFR C677T 51 51 Swiss NS 159
Methylenetetrahydrofolate reductase MTHFR C677T 433 433 Austrian NS 148
Plasminogen activator inhibitor 1 PAI1 HindIII 88 423 Scottish NS 149
Purinergic receptor P2Y P2RY12 H2 haplotype 184 330 White, French 2.3 0.002 160

NS indicates not specified.

Limitations of Studies to Date in Search of Novel Genetic Determinants of PAD

Many genetic studies to date, including those focusing on the genetic determinants of PAD, suffer from a number of weaknesses. These weaknesses include, but are not limited to, a lack of statistical power and confounding because of population stratification.64,72 Below, we briefly discuss the various reasons behind these design weaknesses, as well as reiterate solutions proposed by experts in the field of genetic epidemiology of complex traits both in general terms and as they relate to the studies of PAD published to date.73 We also make some of our own recommendations for studies of PAD.

Statistical Power

The most important root cause, by far, for the lack of reproducibility of most genetic association studies to date of complex traits, including PAD, is a lack of statistical power to detect the positive association described in either the original report or in subsequent replication studies. A lack of power in the setting of a very low pretest probability of an association between a given genetic variant and a phenotype, as well as a P value that is only marginally significant by “traditional” standards (ie, ≈0.05 or ≈0.01), virtually guarantees that the original report is a false positive.74 False-negative findings may also occur because of low power.

Critical factors influencing power include sample size, the prevalence of the exposure in the general population, and the odds ratio that one wishes to detect. It is now clear that, for complex diseases like PAD, sample size needs to be large enough to provide adequate power to detect a genotypic odds ratio as low as 1.2. Typically, this would require genotyping thousands of subjects.66

Linkage Disequilibrium

Linkage disequilibrium (LD) refers to the correlation between alleles at 1 polymorphic site with those at another polymorphic site nearby, and results from a polymorphism arising in a genomic region characterized by a unique combination of alleles in other polymorphic sites. This unique combination of closely linked polymorphisms is frequently referred to as a haplotype.75 LD between SNPs in a region is gradually reduced over time by chromosomal recombinations generally proportional to the distance in base pairs separating 2 polymorphic sites and is also related to the demographic history of the population.57,76,77 Through large-scale characterization of genetic variants in multiple race/ethnic groups, patterns of LD have been significantly refined.40,78,79 These efforts have confirmed the presence of considerably different patterns of LD between major racial/ethnic groups, with the lowest LD between SNPs present in populations of African descent.77

As a consequence of LD, the allele at 1 locus of a given region can often predict the allele of ≥1 neighboring loci.80 Many of the positive genetic association studies of PAD to date have genotyped only 1 SNP and have not explored the strength of the association in other SNPs in high LD with the positive SNP. Doing so could reveal even stronger and more convincing associations. On the other hand, it is possible that some of the negative reports to date are falsely negative because the chosen SNPs are in low LD with nongenotyped causal SNPs in the candidate genes. Based on LD patterns from HapMap data,80 as well as other publicly available resources (such as the SeattleSNPs Program in Genomic Applications), it is now possible to select a minimal set of SNPs (referred to as “tag” SNPs) that best represents all of the common variations in a region encoding a gene.40,77,8183

Misclassification

Misclassification of the exposure of interest in genetic association studies is a direct consequence of genotyping errors and can be nondifferential or differential. The former is a result of errors that are randomly distributed between case and control subjects, whereas the latter is a result of errors that occur preferentially in either between case and control subjects (eg, when the genotyping conditions are different in case subjects compared with control subjects). Nondifferential misclassification increases the rate of false-negatives by introducing noise and eroding study power but can be overcome by larger sample sizes. However, even an error rate of 1% can have a profound effect on power, especially if the allele in question is rare. Differential misclassification, on the other hand, leads to false-positive associations that are only exaggerated with larger sample sizes. To prevent differential misclassification because of systematic genotyping error, samples should be genotyped in random order. In large-scale studies, it is recommended that researchers routinely perform quality control assays by duplicating assays for some samples (in a blinded fashion) to assess reproducibility, using “no template controls” to detect DNA contamination, and including “positive control” DNAs with known genotypes. With good laboratory practice and many of the newer technologies, >99% reproducibility and <1% no-call rates can be achieved.

Incomplete characterization of the phenotype may also dilute power, which is usually nondifferential (ie, random with respect to genotype). Nondifferential misclassification is a common consequence of not using a “gold-standard” tool to characterize the phenotype of interest. For example, although the ABI has a high sensitivity and specificity for detecting occlusive lower extremity disease (as defined by at least one 50% stenosis by angiography), its performance is significantly worse in detecting subocclusive disease. Therefore, a considerable number of subjects with subocclusive but significant disease may be misclassified as control subjects. Perhaps the best noninvasive tool to characterize PAD in the context of genetic association studies will turn out to be an imaging procedure, such as an MRI or multislice CT that can accurately quantify the degree of atherosclerosis in the lower extremities even in the absence of calcification. Such imaging protocols are still under development but should be validated in the near future. In the meantime, we recommend using the traditional cutoff value ABI of 0.9 to identify case subjects to be included in genetic association studies of PAD despite the possibility of misclassification. For reasons outlined earlier in this review, we advise against the use of ABI as a continuous measure as a surrogate PAD phenotype for genetic association studies, particularly in populations with a low prevalence of disease.

Population Stratification/Substructure

Study design weaknesses that relate to statistical power can, in general, all be overcome by increasing sample size. In contrast, bias from confounding cannot be overcome by larger sample sizes and must be addressed by either matching case subjects to control subjects on the potential confounder in the recruitment phase of a study or by statistically adjusting the odds ratio for the potential confounder in the analysis phase.84

A variable can only be a confounder if it is correlated both with outcome and the exposure of interest. In the field of complex trait genetics, the potential confounder that has deservedly received the most attention is race/ethnic group. Statistical geneticists often refer to this type of confounding as “population stratification” or “population substructure.” The potential for population stratification in properly designed association studies has been a subject of debate.8588

Population stratification is possible because many phenotypes, including PAD, have significantly different rates across the major ancestral populations,44 and the reasons for these differences are not strictly genetic in nature. Similarly, allele frequencies for many SNPs and patterns of LD differ substantially across major ancestral populations.67,89,90 Therefore, if the race/ethnic mix is not similar in case and control subjects, false associations may arise as a result of confounding. Under certain circumstances, population stratification can also mask true associations. As a simple surrogate, self-reported race/ethnicity seems to correlate very well with the observed clusters of SNP frequencies specific to major populations.91 Therefore, in studies of subjects who identify solely with 1 of these major groups matching case and control subjects on self-reported race/ethnicity or performing a stratified analysis probably eliminates a vast majority of the potential confounding. However, the potential for significant residual confounding because of population stratification increases when studying recently admixed complex populations with crude race/ethnicity labels,86,92,93 such as African American or Latino populations.94 In African American populations, the range of European ancestry varies widely. Similarly, the degree of black ancestry in Latino populations can vary substantially.85 Because many studies (especially in the United States) involve a significant number of subjects from genetically admixed populations, the degree of admixture within case subjects and within control subjects can vary substantially by chance.86,95,96 Even a modest difference in population substructure between case and control subjects may have profound effects on association outcomes. Fortunately, analytical methods have been developed to effectively deal with residual population stratification using ancestry informative markers. Ancestry informative markers are SNPs that possess large differences (Δ) in allele frequencies among major ancestral populations. Ancestry informative markers can be used to control for population substructure using the “genomic control,”97,98 “structured association” methods,99103 or methods based on principle components analysis.104 In general, between 50 and several hundred well-spaced ancestry informative markers are needed to estimate the individual proportions of the major ancestral populations that exist in an admixed individual.86,105,106

Future Directions

An Approach to PAD Genetic Studies: Candidate Gene Studies

Identification of PAD susceptibility genes could have a significant impact on diagnosis and treatment of this under-recognized and often-untreated disease. Candidate gene studies can play a valuable role, as long as they are designed with certain principles in mind.107,108 Studies should include large sample sets that are accurately phenotyped with respect to PAD status, medications, physical activity,109111 and medical history. A detailed survey of the presence and duration of all well-established risk factors of atherosclerosis, such as smoking, hyperlipidemia, blood pressure, and diabetes, is crucial to adjust for exposure. Blood should be collected and used for DNA purification and other plasma measurements.

Candidate genes for PAD should be genotyped using platforms with very high accuracy and reproducibility. “Tag SNP” approaches are now standard, including SNPs to adequately cover variation in subjects from all race/ethnic groups in the study.90 It is also reasonable to consider genotyping additional nonsynonymous coding SNPs in candidate genes regardless of their frequency,67,112 because it is more likely a priori that exonic SNPs will produce functional effects that will be associated with disease status. Ideally, study power should be adequate to detect small effects. Should promising associations arise, associations must be replicated in ≥1 and preferably 2 other independent populations.

Genome-Wide Association Studies

Although a candidate gene approach is a valuable first step in trying to identify PAD susceptibility genes, even with well-informed choices of candidate genes, this approach is limited to a priori hypotheses. A logical next step is to perform a more comprehensive and unbiased scan of variations in all genes or in the entire genome, in what is commonly referred to as a genome-wide association (GWA) study.

The GWA design calls for the use of high throughput genotyping platforms to genotype as many SNPs in the genome as possible irrespective of the location of these SNPs relative to genes. In this respect, the GWA design is similar to linkage studies, because no previous information on gene function is necessary to select SNPs for genotyping. However, the GWA design is unlike linkage because it maintains the ability to detect modest genotypic effects expected in complex traits and, furthermore, localizes initial genetic signals to a much smaller genomic region compared with linkage. In the last few years, the GWA design has become analytically feasible primarily as a result of large-scale genetic variation characterization efforts by the International HapMap project and Perlegen in a large number of racially diverse subjects.79,113 These efforts have produced genotypes, frequencies, and assay information on >6 million SNPs of the estimated 11 to 15 million SNPs in the human genome with a minor allele frequency of >1%. Two companies, Affymetrix and Illumina, have developed commercially available high-throughput and highly parallel genotyping technology that allow for 650 000 to >1 million SNPs to be genotyped in a single individual at a fraction of the cost per genotype compared with other reliable platforms. To decrease costs without a substantial loss of power, 2-stage designs114116 can be used. In this approach, a GWA scan is initially performed in a subset of samples (stage 1), and a portion of the markers, those showing tentative association, are genotyped in the remaining samples. Importantly for PAD, GWA studies can be performed to look for variations associated with dichotomous variables (PAD versus no PAD) or continuous variables (potentially using CT or MRI measurements).

Reports of successful localization of common genetic variants using a GWA design influencing complex traits, such as macular degeneration,117 obesity,118,119 inflammatory bowel disease,120 rheumatoid arthritis, bipolar disorder, myocardial infarction,121,122 DM1 and DM2,123128 have recently been published. Although the studies differed in size and genotyping platforms used, many of the major findings have been convincingly reproduced. Thus, they collectively represent an important proof of principle: the GWA approach can identify common variants that contribute to common disease.64,129

Nevertheless, applying the GWA approach to complex diseases (like PAD) requires careful consideration of study design. The results of GWA studies in complex diseases, like myocardial infarction and DM2, highlight some of the necessary study characteristics, including large sample sizes (often requiring collaboration between multiple smaller studies), due consideration of population structure, and replication in multiple cohorts. The large numbers of association tests that result from GWA scans require that an even lower probability value threshold be used to identify promising associations between variants and disease. Early simulations suggest that a P value threshold of ≈5×10–7 to 5×10–8 is necessary in the setting of GWA studies.66,128 Again, sample sizes need to be adequate in GWA studies to maintain power to detect the modest effects expected in complex traits.96 For instance, in the recent studies in DM2 and CAD, many of the susceptibility variants identified were associated with odds ratios (or hazard ratios) of ≈1.15 to 1.3.122,124,125,127 Similarly, a recent GWA study identified a variant responsible for ≈1% of the variance in body mass index.119 The GWA studies in myocardial infarction and DM2 ultimately required thousands of subjects, although the initial scans can be performed in much fewer subjects.122 These studies serve as guides when considering this approach in PAD.

This field is still in its infancy, and numerous new approaches toward study design and analysis are being developed. For instance, the analysis of copy number variation130,131 may prove increasingly important. Furthermore, techniques are being developed to merge data sets from different genotyping platforms, improve power through weighting schemes based on limited previous knowledge,132 speed processing time of association tests,133 and analyze sophisticated gene/gene134,135 or gene/environment interactions.136 Already, methods have been developed to increase coverage of the genome through imputation of genomic data based on LD structure123 or to analyze continuous data at the extremes of phenotype.137139 New recommendations have also been published by working groups within the American Heart Association and National Human Genome Research Institute to guide study design.64,129

Although these studies have been spectacularly successful so far, it is an open question whether we will ultimately be able to identify most of the genetic variation that accounts for common diseases using this approach. It may turn out that rare variants account for much of the genetic susceptibility to common, complex diseases. These variants may not yield to this approach and might have to wait for whole genome sequencing for identification.

Summary and Call for PAD Networks

In summary, PAD is a common and complex trait of which the development is probably modified to some degree by uncharacterized genetic determinants that are independent of traditional risk factors of atherosclerosis. Based on limited studies to date, the heritability of PAD seems to be modest. More family studies of PAD are necessary to better estimate the relative importance of genetic and nongenetic determinants of disease. Carefully designed association studies of well-chosen candidate genes may identify some of these uncharacterized genetic determinants, but GWA studies are expected to be more revealing. Given the complex nature of the disease, future progress also depends crucially on close collaboration within the community of PAD investigators. Coordinated efforts by a large network of investigators are needed to develop standardized diagnostic instruments and to collect accurate phenotypic information in samples of sufficient size to perform adequately powered candidate gene and GWA studies. Within our own case-control association study of genetic determinants of PAD (http://cvmedicine.stanford.edu/genepad/index.html), we have begun to see this spirit of camaraderie play a positive role. This type of interaction will also facilitate independent replication of promising results and the subsequent elucidation of the biological mechanisms responsible for the various associations observed.

Acknowledgments

We thank Dr Hua Tang, Stanford Department of Genetics, for helpful comments.

Sources of Funding

This study was supported by grants from the National Institutes of Health (RO1 HL63685 and RO1 HL075774) and a National Heart, Lung, and Blood Institute National Research Service Award T32 HL07708, as well as by National Institutes of Health grant M01 RR 00070 (General Clinical Research Center, Stanford University School of Medicine). J.W.K. was supported by postdoctoral fellowship grants from Stanford University and the American Heart Association (0625154Y).

Footnotes

Disclosures

None.

References

  • 1.Zheng ZJ, Rosamond WD, Chambless LE, Nieto FJ, Barnes RW, Hutchinson RG, Tyroler HA, Heiss G. Lower Extremity arterial disease assessed by ankle-brachial index in a middle-aged population of African Americans and whites: the Atherosclerosis Risk in Communities (ARIC) Study. Am J Prev Med. 2005;29:42–49. doi: 10.1016/j.amepre.2005.07.019. [DOI] [PubMed] [Google Scholar]
  • 2.Zheng ZJ, Sharrett AR, Chambless LE, Rosamond WD, Nieto FJ, Sheps DS, Dobs A, Evans GW, Heiss G. Associations of ankle-brachial index with clinical coronary heart disease, stroke and preclinical carotid and popliteal atherosclerosis: the Atherosclerosis Risk in Communities (ARIC) Study. Atherosclerosis. 1997;131:115–125. doi: 10.1016/s0021-9150(97)06089-9. [DOI] [PubMed] [Google Scholar]
  • 3.Meijer WT, Hoes AW, Rutgers D, Bots ML, Hofman A, Grobbee DE. Peripheral arterial disease in the elderly: the Rotterdam Study. Arterioscler Thromb Vasc Biol. 1998;18:185–192. doi: 10.1161/01.atv.18.2.185. [DOI] [PubMed] [Google Scholar]
  • 4.Meijer WT, Grobbee DE, Hunink MG, Hofman A, Hoes AW. Determinants of peripheral arterial disease in the elderly: the Rotterdam study. Arch Intern Med. 2000;160:2934–2938. doi: 10.1001/archinte.160.19.2934. [DOI] [PubMed] [Google Scholar]
  • 5.Hooi JD, Kester AD, Stoffers HE, Rinkens PE, Knottnerus JA, van Ree JW. Asymptomatic peripheral arterial occlusive disease predicted cardiovascular morbidity and mortality in a 7-year follow-up study. J Clin Epidemiol. 2004;57:294–300. doi: 10.1016/j.jclinepi.2003.09.003. [DOI] [PubMed] [Google Scholar]
  • 6.Hooi JD, Stoffers HE, Kester AD, Rinkens PE, Kaiser V, van Ree JW, Knottnerus JA. Risk factors and cardiovascular diseases associated with asymptomatic peripheral arterial occlusive disease. The Limburg PAOD Study. Peripheral Arterial Occlusive Disease. Scand J Prim Health Care. 1998;16:177–182. doi: 10.1080/028134398750003142. [DOI] [PubMed] [Google Scholar]
  • 7.Ouriel K. Peripheral arterial disease. Lancet. 2001;358:1257–1264. doi: 10.1016/S0140-6736(01)06351-6. [DOI] [PubMed] [Google Scholar]
  • 8.Belch JJ, Topol EJ, Agnelli G, Bertrand M, Califf RM, Clement DL, Creager MA, Easton JD, Gavin JR, 3rd, Greenland P, Hankey G, Hanrath P, Hirsch AT, Meyer J, Smith SC, Sullivan F, Weber MA. Critical issues in peripheral arterial disease detection and management: a call to action. Arch Intern Med. 2003;163:884–892. doi: 10.1001/archinte.163.8.884. [DOI] [PubMed] [Google Scholar]
  • 9.Hiatt WR. Medical treatment of peripheral arterial disease and claudication. N Engl J Med. 2001;344:1608–1621. doi: 10.1056/NEJM200105243442108. [DOI] [PubMed] [Google Scholar]
  • 10.Hirsch AT, Criqui MH, Treat-Jacobson D, Regensteiner JG, Creager MA, Olin JW, Krook SH, Hunninghake DB, Comerota AJ, Walsh ME, McDermott MM, Hiatt WR. Peripheral arterial disease detection, awareness, and treatment in primary care. JAMA. 2001;286:1317–1324. doi: 10.1001/jama.286.11.1317. [DOI] [PubMed] [Google Scholar]
  • 11.Weitz JI, Byrne J, Clagett GP, Farkouh ME, Porter JM, Sackett DL, Strandness DE, Jr, Taylor LM. Diagnosis and treatment of chronic arterial insufficiency of the lower extremities: a critical review. Circulation. 1996;94:3026–3049. doi: 10.1161/01.cir.94.11.3026. [DOI] [PubMed] [Google Scholar]
  • 12.McDermott MM, Greenland P, Liu K, Guralnik JM, Criqui MH, Dolan NC, Chan C, Celic L, Pearce WH, Schneider JR, Sharma L, Clark E, Gibson D, Martin GJ. Leg symptoms in peripheral arterial disease: associated clinical characteristics and functional impairment. JAMA. 2001;286:1599–1606. doi: 10.1001/jama.286.13.1599. [DOI] [PubMed] [Google Scholar]
  • 13.McDermott MM, Liu K, Greenland P, Guralnik JM, Criqui MH, Chan C, Pearce WH, Schneider JR, Ferrucci L, Celic L, Taylor LM, Vonesh E, Martin GJ, Clark E. Functional decline in peripheral arterial disease: associations with the ankle brachial index and leg symptoms. JAMA. 2004;292:453–461. doi: 10.1001/jama.292.4.453. [DOI] [PubMed] [Google Scholar]
  • 14.Criqui MH, Langer RD, Fronek A, Feigelson HS, Klauber MR, McCann TJ, Browner D. Mortality over a period of 10 years in patients with peripheral arterial disease. N Engl J Med. 1992;326:381–386. doi: 10.1056/NEJM199202063260605. [DOI] [PubMed] [Google Scholar]
  • 15.Newman AB, Shemanski L, Manolio TA, Cushman M, Mittelmark M, Polak JF, Powe NR, Siscovick D. Ankle-arm index as a predictor of cardiovascular disease and mortality in the Cardiovascular Health Study. The Cardiovascular Health Study Group. Arterioscler Thromb Vasc Biol. 1999;19:538–545. doi: 10.1161/01.atv.19.3.538. [DOI] [PubMed] [Google Scholar]
  • 16.Mohler ER., 3rd Peripheral arterial disease: identification and implications. Arch Intern Med. 2003;163:2306–2314. doi: 10.1001/archinte.163.19.2306. [DOI] [PubMed] [Google Scholar]
  • 17.Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL, Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks D, Stanley JC, Taylor LM, Jr, White CJ, White J, White RA, Antman EM, Smith SC, Jr, Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Halperin JL, Hiratzka LF, Hunt SA, Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B. ACC/AHA 2005 guidelines for the management of patients with peripheral arterial disease (lower extremity, renal, mesenteric, and abdominal aortic): executive summary a collaborative report from the American Association for Vascular Surgery/Society for Vascular Surgery, Society for Cardiovascular Angiography and Interventions, Society for Vascular Medicine and Biology, Society of Interventional Radiology, and the ACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Peripheral Arterial Disease) endorsed by the American Association of Cardiovascular and Pulmonary Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular Nursing; TransAtlantic Inter-Society Consensus; and Vascular Disease Foundation. J Am Coll Cardiol. 2006;47:1239–1312. doi: 10.1016/j.jacc.2005.10.009. [DOI] [PubMed] [Google Scholar]
  • 18.Allison MA, Laughlin GA, Barrett-Connor E, Langer R. Association between the ankle-brachial index and future coronary calcium (the Rancho Bernardo study). Am J Cardiol. 2006;97:181–186. doi: 10.1016/j.amjcard.2005.08.019. [DOI] [PubMed] [Google Scholar]
  • 19.Wang JC, Criqui MH, Denenberg JO, McDermott MM, Golomb BA, Fronek A. Exertional leg pain in patients with and without peripheral arterial disease. Circulation. 2005;112:3501–3508. doi: 10.1161/CIRCULATIONAHA.105.548099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Allison MA, Laughlin GA, Barrett-Connor E. Association between the ankle-brachial index and carotid intimal medial thickness in the Rancho Bernardo Study. Am J Cardiol. 2006;98:1105–1109. doi: 10.1016/j.amjcard.2006.05.037. [DOI] [PubMed] [Google Scholar]
  • 21.McDermott MM, Liu K, Criqui MH, Ruth K, Goff D, Saad MF, Wu C, Homma S, Sharrett AR. Ankle-brachial index and subclinical cardiac and carotid disease: the multi-ethnic study of atherosclerosis. Am J Epidemiol. 2005;162:33–41. doi: 10.1093/aje/kwi167. [DOI] [PubMed] [Google Scholar]
  • 22.Newman AB, Siscovick DS, Manolio TA, Polak J, Fried LP, Borhani NO, Wolfson SK. Ankle-arm index as a marker of atherosclerosis in the Cardiovascular Health Study. Cardiovascular Heart Study (CHS) Collaborative Research Group. Circulation. 1993;88:837–845. doi: 10.1161/01.cir.88.3.837. [DOI] [PubMed] [Google Scholar]
  • 23.O'Hare AM, Katz R, Shlipak MG, Cushman M, Newman AB. Mortality and cardiovascular risk across the ankle-arm index spectrum: results from the Cardiovascular Health Study. Circulation. 2006;113:388–393. doi: 10.1161/CIRCULATIONAHA.105.570903. [DOI] [PubMed] [Google Scholar]
  • 24.Szuba A, Oka RK, Harada R, Cooke JP. Limb hemodynamics are not predictive of functional capacity in patients with PAD. Vasc Med. 2006;11:155–163. doi: 10.1177/1358863x06074828. [DOI] [PubMed] [Google Scholar]
  • 25.Long J, Modrall JG, Parker BJ, Swann A, Welborn MB, 3rd, Anthony T. Correlation between ankle-brachial index, symptoms, and health-related quality of life in patients with peripheral vascular disease. J Vasc Surg. 2004;39:723–727. doi: 10.1016/j.jvs.2003.12.006. [DOI] [PubMed] [Google Scholar]
  • 26.Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature. 1993;362:801–809. doi: 10.1038/362801a0. [DOI] [PubMed] [Google Scholar]
  • 27.Glass CK, Witztum JL. Atherosclerosis. The road ahead. Cell. 2001;104:503–516. doi: 10.1016/s0092-8674(01)00238-0. [DOI] [PubMed] [Google Scholar]
  • 28.Lusis AJ. Atherosclerosis. Nature. 2000;407:233–241. doi: 10.1038/35025203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sing CF, Stengard JH, Kardia SL. Genes, environment, and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2003;23:1190–1196. doi: 10.1161/01.ATV.0000075081.51227.86. [DOI] [PubMed] [Google Scholar]
  • 30.Boerwinkle E, Ellsworth DL, Hallman DM, Biddinger A. Genetic analysis of atherosclerosis: a research paradigm for the common chronic diseases. Hum Mol Genet. 1996;5:1405–1410. doi: 10.1093/hmg/5.supplement_1.1405. [DOI] [PubMed] [Google Scholar]
  • 31.Ellsworth DL, Sholinsky P, Jaquish C, Fabsitz RR, Manolio TA. Coronary heart disease. At the interface of molecular genetics and preventive medicine. Am J Prev Med. 1999;16:122–133. doi: 10.1016/s0749-3797(98)00138-x. [DOI] [PubMed] [Google Scholar]
  • 32.Kennedy M, Solomon C, Manolio TA, Criqui MH, Newman AB, Polak JF, Burke GL, Enright P, Cushman M. Risk factors for declining ankle-brachial index in men and women 65 years or older: the Cardiovascular Health Study. Arch Intern Med. 2005;165:1896–1902. doi: 10.1001/archinte.165.16.1896. [DOI] [PubMed] [Google Scholar]
  • 33.Halperin JL, Fuster V. Meeting the challenge of peripheral arterial disease. Arch Intern Med. 2003;163:877–878. doi: 10.1001/archinte.163.8.877. [DOI] [PubMed] [Google Scholar]
  • 34.Clarke R, Daly L, Robinson K, Naughten E, Cahalane S, Fowler B, Graham I. Hyperhomocysteinemia: an independent risk factor for vascular disease. N Engl J Med. 1991;324:1149–1155. doi: 10.1056/NEJM199104253241701. [DOI] [PubMed] [Google Scholar]
  • 35.Migliaccio-Walle K, Caro JJ, Ishak KJ, O'Brien JA. Costs and medical care consequences associated with the diagnosis of peripheral arterial disease. Pharmacoeconomics. 2005;23:733–742. doi: 10.2165/00019053-200523070-00007. [DOI] [PubMed] [Google Scholar]
  • 36.Peyser PA. Genetic epidemiology of coronary artery disease. Epidemiol Rev. 1997;19:80–90. doi: 10.1093/oxfordjournals.epirev.a017949. [DOI] [PubMed] [Google Scholar]
  • 37.Winkelmann BR, Hager J, Kraus WE, Merlini P, Keavney B, Grant PJ, Muhlestein JB, Granger CB. Genetics of coronary heart disease: current knowledge and research principles. Am Heart J. 2000;140:S11–S26. doi: 10.1067/mhj.2000.109636. [DOI] [PubMed] [Google Scholar]
  • 38.Winkelmann BR, Hager J. Genetic variation in coronary heart disease and myocardial infarction: methodological overview and clinical evidence. Pharmacogenomics. 2000;1:73–94. doi: 10.1517/14622416.1.1.73. [DOI] [PubMed] [Google Scholar]
  • 39.Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, Funke R, Gage D, Harris K, Heaford A, Howland J, Kann L, Lehoczky J, LeVine R, McEwan P, McKernan K, Meldrim J, Mesirov JP, Miranda C, Morris W, Naylor J, Raymond C, Rosetti M, Santos R, Sheridan A, Sougnez C, Stange-Thomann N, Stojanovic N, Subramanian A, Wyman D, Rogers J, Sulston J, Ainscough R, Beck S, Bentley D, Burton J, Clee C, Carter N, Coulson A, Deadman R, Deloukas P, Dunham A, Dunham I, Durbin R, French L, Grafham D, Gregory S, Hubbard T, Humphray S, Hunt A, Jones M, Lloyd C, McMurray A, Matthews L, Mercer S, Milne S, Mullikin JC, Mungall A, Plumb R, Ross M, Shownkeen R, Sims S, Waterston RH, Wilson RK, Hillier LW, McPherson JD, Marra MA, Mardis ER, Fulton LA, Chinwalla AT, Pepin KH, Gish WR, Chissoe SL, Wendl MC, Delehaunty KD, Miner TL, Delehaunty A, Kramer JB, Cook LL, Fulton RS, Johnson DL, Minx PJ, Clifton SW, Hawkins T, Branscomb E, Predki P, Richardson P, Wenning S, Slezak T, Doggett N, Cheng JF, Olsen A, Lucas S, Elkin C, Uberbacher E, Frazier M, Gibbs RA, Muzny DM, Scherer SE, Bouck JB, Sodergren EJ, Worley KC, Rives CM, Gorrell JH, Metzker ML, Naylor SL, Kucherlapati RS, Nelson DL, Weinstock GM, Sakaki Y, Fujiyama A, Hattori M, Yada T, Toyoda A, Itoh T, Kawagoe C, Watanabe H, Totoki Y, Taylor T, Weissenbach J, Heilig R, Saurin W, Artiguenave F, Brottier P, Bruls T, Pelletier E, Robert C, Wincker P, Smith DR, Doucette-Stamm L, Rubenfield M, Weinstock K, Lee HM, Dubois J, Rosenthal A, Platzer M, Nyakatura G, Taudien S, Rump A, Yang H, Yu J, Wang J, Huang G, Gu J, Hood L, Rowen L, Madan A, Qin S, Davis RW, Federspiel NA, Abola AP, Proctor MJ, Myers RM, Schmutz J, Dickson M, Grimwood J, Cox DR, Olson MV, Kaul R, Shimizu N, Kawasaki K, Minoshima S, Evans GA, Athanasiou M, Schultz R, Roe BA, Chen F, Pan H, Ramser J, Lehrach H, Reinhardt R, McCombie WR, de la Bastide M, Dedhia N, Blocker H, Hornischer K, Nordsiek G, Agarwala R, Aravind L, Bailey JA, Bateman A, Batzoglou S, Birney E, Bork P, Brown DG, Burge CB, Cerutti L, Chen HC, Church D, Clamp M, Copley RR, Doerks T, Eddy SR, Eichler EE, Furey TS, Galagan J, Gilbert JG, Harmon C, Hayashizaki Y, Haussler D, Hermjakob H, Hokamp K, Jang W, Johnson LS, Jones TA, Kasif S, Kaspryzk A, Kennedy S, Kent WJ, Kitts P, Koonin EV, Korf I, Kulp D, Lancet D, Lowe TM, McLysaght A, Mikkelsen T, Moran JV, Mulder N, Pollara VJ, Ponting CP, Schuler G, Schultz J, Slater G, Smit AF, Stupka E, Szustakowki J, Thierry-Mieg D, Wagner L, Wallis J, Wheeler R, Williams A, Wolf YI, Wolfe KH, Yang SP, Yeh RF, Collins F, Guyer MS, Peterson J, Felsenfeld A, Wetterstrand KA, Patrinos A, Morgan MJ. Initial sequencing and analysis of the human genome. Nature. 2001;409:860–921. doi: 10.1038/35057062. [DOI] [PubMed] [Google Scholar]
  • 40.Altshuler D, Brooks LD, Chakravarti A, Collins FS, Daly MJ, Donnelly P. A haplotype map of the human genome. Nature. 2005;437:1299–1320. doi: 10.1038/nature04226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.The International HapMap Project. Nature. 2003;426:789–796. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
  • 42.Khoury MJ, Beaty TH, Cohen BH. Fundamentals of Genetic Epidemiology. Oxford University Press; New York, NY: 1993. [Google Scholar]
  • 43.Valentine RJ, Verstraete R, Clagett GP, Cohen JC. Premature cardiovascular disease is common in relatives of patients with premature peripheral atherosclerosis. Arch Intern Med. 2000;160:1343–1348. doi: 10.1001/archinte.160.9.1343. [DOI] [PubMed] [Google Scholar]
  • 44.Allison MA, Criqui MH, McClelland RL, Scott JM, McDermott MM, Liu K, Folsom AR, Bertoni AG, Sharrett AR, Homma S, Kori S. The effect of novel cardiovascular risk factors on the ethnic-specific odds for peripheral arterial disease in the Multi-Ethnic Study of Atherosclerosis (MESA). J Am Coll Cardiol. 2006;48:1190–1197. doi: 10.1016/j.jacc.2006.05.049. [DOI] [PubMed] [Google Scholar]
  • 45.Carmelli D, Fabsitz RR, Swan GE, Reed T, Miller B, Wolf PA. Contribution of genetic and environmental influences to ankle-brachial blood pressure index in the NHLBI Twin Study. National Heart, Lung, and Blood Institute. Am J Epidemiol. 2000;151:452–458. doi: 10.1093/oxfordjournals.aje.a010230. [DOI] [PubMed] [Google Scholar]
  • 46.Kullo IJ, Turner ST, Kardia SL, Mosley TH, Jr, Boerwinkle E, Andrade MD. A genome-wide linkage scan for ankle-brachial index in African Am and non-Hispanic white subjects participating in the GENOA study. Atherosclerosis. 2006;187:433–438. doi: 10.1016/j.atherosclerosis.2005.10.003. [DOI] [PubMed] [Google Scholar]
  • 47.Murabito JM, Guo CY, Fox CS, D'Agostino RB. Heritability of the ankle-brachial index: the Framingham Offspring study. Am J Epidemiol. 2006;164:963–968. doi: 10.1093/aje/kwj295. [DOI] [PubMed] [Google Scholar]
  • 48.Risch N. The genetic epidemiology of cancer: interpreting family and twin studies and their implications for molecular genetic approaches. Cancer Epidemiol Biomarkers Prev. 2001;10:733–741. [PubMed] [Google Scholar]
  • 49.Falconer DS, Mackay TFC. Introduction to Quantitative Genetics. Longmans Green; Harlow, Essex, United Kingdom: 1996. [Google Scholar]
  • 50.Criqui MH, Vargas V, Denenberg JO, Ho E, Allison M, Langer RD, Gamst A, Bundens WP, Fronek A. Ethnicity and peripheral arterial disease: the San Diego Population Study. Circulation. 2005;112:2703–2707. doi: 10.1161/CIRCULATIONAHA.105.546507. [DOI] [PubMed] [Google Scholar]
  • 51.Collins TC, Petersen NJ, Suarez-Almazor M, Ashton CM. The prevalence of peripheral arterial disease in a racially diverse population. Arch Intern Med. 2003;163:1469–1474. doi: 10.1001/archinte.163.12.1469. [DOI] [PubMed] [Google Scholar]
  • 52.Selvin E, Erlinger TP. Prevalence of and risk factors for peripheral arterial disease in the United States: results from the National Health and Nutrition Examination Survey, 1999–2000. Circulation. 2004;110:738–743. doi: 10.1161/01.CIR.0000137913.26087.F0. [DOI] [PubMed] [Google Scholar]
  • 53.Sing CF, Moll PP. Genetics of variability of CHD risk. Int J Epidemiol. 1989;18:S183–S195. [PubMed] [Google Scholar]
  • 54.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;330:1041–1046. doi: 10.1056/NEJM199404143301503. [DOI] [PubMed] [Google Scholar]
  • 55.Lohmueller KE, Pearce CL, Pike M, Lander ES, Hirschhorn JN. Meta-analysis of genetic association studies supports a contribution of common variants to susceptibility to common disease. Nat Genet. 2003;33:177–182. doi: 10.1038/ng1071. [DOI] [PubMed] [Google Scholar]
  • 56.Lander E, Kruglyak L. Genetic dissection of complex traits: guidelines for interpreting and reporting linkage results. Nat Genet. 1995;11:241–247. doi: 10.1038/ng1195-241. [DOI] [PubMed] [Google Scholar]
  • 57.Hauser ER, Pericak-Vance MA. Genetic analysis for common complex disease. Am Heart J. 2000;140:S36–44. doi: 10.1067/mhj.2000.108946. [DOI] [PubMed] [Google Scholar]
  • 58.Mackay TF. The genetic architecture of quantitative traits. Annu Rev Genet. 2001;35:303–339. doi: 10.1146/annurev.genet.35.102401.090633. [DOI] [PubMed] [Google Scholar]
  • 59.Brooks-Wilson A, Marcil M, Clee SM, Zhang LH, Roomp K, van Dam M, Yu L, Brewer C, Collins JA, Molhuizen HO, Loubser O, Ouelette BF, Fichter K, Ashbourne-Excoffon KJ, Sensen CW, Scherer S, Mott S, Denis M, Martindale D, Frohlich J, Morgan K, Koop B, Pimstone S, Kastelein JJ, Genest J, Hayden MR. Mutations in ABC1 in Tangier disease and familial high-density lipoprotein deficiency. Nat Genet. 1999;22:336–345. doi: 10.1038/11905. [DOI] [PubMed] [Google Scholar]
  • 60.Lander ES, Schork NJ. Genetic dissection of complex traits. Science. 1994;265:2037–2048. doi: 10.1126/science.8091226. [DOI] [PubMed] [Google Scholar]
  • 61.Pankow JS, Heiss G, Evans GW, Sholinsky P, Province MA, Coon H, Ellison RC, Miller MB, Qaqish B. Familial aggregation and genome-wide linkage analysis of carotid artery plaque: the NHLBI family heart study. Hum Hered. 2004;57:80–89. doi: 10.1159/000077545. [DOI] [PubMed] [Google Scholar]
  • 62.Broeckel U, Hengstenberg C, Mayer B, Holmer S, Martin LJ, Comuzzie AG, Blangero J, Nurnberg P, Reis A, Riegger GA, Jacob HJ, Schunkert H. A comprehensive linkage analysis for myocardial infarction and its related risk factors. Nat Genet. 2002;30:210–214. doi: 10.1038/ng827. [DOI] [PubMed] [Google Scholar]
  • 63.Jeunemaitre X, Soubrier F, Kotelevtsev YV, Lifton RP, Williams CS, Charru A, Hunt SC, Hopkins PN, Williams RR, Lalouel JM, et al. Molecular basis of human hypertension: role of angiotensinogen. Cell. 1992;71:169–180. doi: 10.1016/0092-8674(92)90275-h. [DOI] [PubMed] [Google Scholar]
  • 64.Arnett DK, Baird AE, Barkley RA, Basson CT, Boerwinkle E, Ganesh SK, Herrington DM, Hong Y, Jaquish C, McDermott DA, O'Donnell CJ. Relevance of genetics and genomics for prevention and treatment of cardiovascular disease: a scientific statement from the American Heart Association Council on Epidemiology and Prevention, the Stroke Council, and the Functional Genomics and Translational Biology Interdisciplinary Working Group. Circulation. 2007;115:2878–2901. doi: 10.1161/CIRCULATIONAHA.107.183679. [DOI] [PubMed] [Google Scholar]
  • 65.Gudmundsson G, Matthiasson SE, Arason H, Johannsson H, Runarsson F, Bjarnason H, Helgadottir K, Thorisdottir S, Ingadottir G, Lindpaintner K, Sainz J, Gudnason V, Frigge ML, Kong A, Gulcher JR, Stefansson K. Localization of a gene for peripheral arterial occlusive disease to chromosome 1p31. Am J Hum Genet. 2002;70:586–592. doi: 10.1086/339251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Risch N, Merikangas K. The future of genetic studies of complex human diseases. Science. 1996;273:1516–1517. doi: 10.1126/science.273.5281.1516. [DOI] [PubMed] [Google Scholar]
  • 67.Botstein D, Risch N. Discovering genotypes underlying human phenotypes: past successes for mendelian disease, future approaches for complex disease. Nat Genet. 2003;33(suppl):228–237. doi: 10.1038/ng1090. [DOI] [PubMed] [Google Scholar]
  • 68.Boerwinkle E, Hixson JE, Hanis CL. Peeking under the peaks: following up genome-wide linkage analyses. Circulation. 2000;102:1877–1878. doi: 10.1161/01.cir.102.16.1877. [DOI] [PubMed] [Google Scholar]
  • 69.Collins FS, Guyer MS, Charkravarti A. Variations on a theme: cataloging human DNA sequence variation. Science. 1997;278:1580–1581. doi: 10.1126/science.278.5343.1580. [DOI] [PubMed] [Google Scholar]
  • 70.Kraus WE. Genetic approaches for the investigation of genes associated with coronary heart disease. Am Heart J. 2000;140:S27–S35. doi: 10.1067/mhj.2000.109380. [DOI] [PubMed] [Google Scholar]
  • 71.Peltonen L, McKusick VA. Genomics and medicine. Dissecting human disease in the postgenomic era. Science. 2001;291:1224–1229. doi: 10.1126/science.291.5507.1224. [DOI] [PubMed] [Google Scholar]
  • 72.Hirschhorn JN, Lohmueller K, Byrne E, Hirschhorn K. A comprehensive review of genetic association studies. Genet Med. 2002;4:45–61. doi: 10.1097/00125817-200203000-00002. [DOI] [PubMed] [Google Scholar]
  • 73.Newton-Cheh C, Hirschhorn JN. Genetic association studies of complex traits: design and analysis issues. Mutat Res. 2005;573:54–69. doi: 10.1016/j.mrfmmm.2005.01.006. [DOI] [PubMed] [Google Scholar]
  • 74.Wacholder S, Chanock S, Garcia-Closas M, El Ghormli L, Rothman N. Assessing the probability that a positive report is false: an approach for molecular epidemiology studies. J Natl Cancer Inst. 2004;96:434–442. doi: 10.1093/jnci/djh075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mueller JC. Linkage disequilibrium for different scales and applications. Brief Bioinform. 2004;5:355–364. doi: 10.1093/bib/5.4.355. [DOI] [PubMed] [Google Scholar]
  • 76.Goldstein DB. Islands of linkage disequilibrium. Nat Genet. 2001;29:109–111. doi: 10.1038/ng1001-109. [DOI] [PubMed] [Google Scholar]
  • 77.Crawford DC, Carlson CS, Rieder MJ, Carrington DP, Yi Q, Smith JD, Eberle MA, Kruglyak L, Nickerson DA. Haplotype diversity across 100 candidate genes for inflammation, lipid metabolism, and blood pressure regulation in two populations. Am J Hum Genet. 2004;74:610–622. doi: 10.1086/382227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.The EUCLID Study Group Randomised placebo-controlled trial of lisinopril in normotensive patients with insulin-dependent diabetes and normoalbuminuria or microalbuminuria. Lancet. 1997;349:1787–1792. [see comments] [PubMed] [Google Scholar]
  • 79.Hinds DA, Stuve LL, Nilsen GB, Halperin E, Eskin E, Ballinger DG, Frazer KA, Cox DR. Whole-genome patterns of common DNA variation in three human populations. Science. 2005;307:1072–1079. doi: 10.1126/science.1105436. [DOI] [PubMed] [Google Scholar]
  • 80.Gabriel SB, Schaffner SF, Nguyen H, Moore JM, Roy J, Blumenstiel B, Higgins J, DeFelice M, Lochner A, Faggart M, Liu-Cordero SN, Rotimi C, Adeyemo A, Cooper R, Ward R, Lander ES, Daly MJ, Altshuler D. The structure of haplotype blocks in the human genome. Science. 2002;296:2225–2229. doi: 10.1126/science.1069424. [DOI] [PubMed] [Google Scholar]
  • 81.Carlson CS, Eberle MA, Rieder MJ, Yi Q, Kruglyak L, Nickerson DA. Selecting a maximally informative set of single-nucleotide polymorphisms for association analyses using linkage disequilibrium. Am J Hum Genet. 2004;74:106–120. doi: 10.1086/381000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Johnson GC, Esposito L, Barratt BJ, Smith AN, Heward J, Di Genova G, Ueda H, Cordell HJ, Eaves IA, Dudbridge F, Twells RC, Payne F, Hughes W, Nutland S, Stevens H, Carr P, Tuomilehto-Wolf E, Tuomilehto J, Gough SC, Clayton DG, Todd JA. Haplotype tagging for the identification of common disease genes. Nat Genet. 2001;29:233–237. doi: 10.1038/ng1001-233. [DOI] [PubMed] [Google Scholar]
  • 83.Patil N, Berno AJ, Hinds DA, Barrett WA, Doshi JM, Hacker CR, Kautzer CR, Lee DH, Marjoribanks C, McDonough DP, Nguyen BT, Norris MC, Sheehan JB, Shen N, Stern D, Stokowski RP, Thomas DJ, Trulson MO, Vyas KR, Frazer KA, Fodor SP, Cox DR. Blocks of limited haplotype diversity revealed by high-resolution scanning of human chromosome 21. Science. 2001;294:1719–1723. doi: 10.1126/science.1065573. [DOI] [PubMed] [Google Scholar]
  • 84.Rothman KJ, Greenland S. Modern Epidemiology. Lippincott-Raven; Philadelphia, PA: 1998. [Google Scholar]
  • 85.Choudhry S, Coyle NE, Tang H, Salari K, Lind D, Clark SL, Tsai HJ, Naqvi M, Phong A, Ung N, Matallana H, Avila PC, Casal J, Torres A, Nazario S, Castro R, Battle NC, Perez-Stable EJ, Kwok PY, Sheppard D, Shriver MD, Rodriguez-Cintron W, Risch N, Ziv E, Burchard EG. Population stratification confounds genetic association studies among Latinos. Hum Genet. 2006;118:652–664. doi: 10.1007/s00439-005-0071-3. [DOI] [PubMed] [Google Scholar]
  • 86.Marchini J, Cardon LR, Phillips MS, Donnelly P. The effects of human population structure on large genetic association studies. Nat Genet. 2004;36:512–517. doi: 10.1038/ng1337. [DOI] [PubMed] [Google Scholar]
  • 87.Wang Y, Localio R, Rebbeck TR. Evaluating bias due to population stratification in case-control association studies of admixed populations. Genet Epidemiol. 2004;27:14–20. doi: 10.1002/gepi.20003. [DOI] [PubMed] [Google Scholar]
  • 88.Wang Y, Localio R, Rebbeck TR. Evaluating bias due to population stratification in epidemiologic studies of gene-gene or gene-environment interactions. Cancer Epidemiol Biomarkers Prev. 2006;15:124–132. doi: 10.1158/1055-9965.EPI-05-0304. [DOI] [PubMed] [Google Scholar]
  • 89.Halushka MK, Fan JB, Bentley K, Hsie L, Shen N, Weder A, Cooper R, Lipshutz R, Chakravarti A. Patterns of single-nucleotide polymorphisms in candidate genes for blood-pressure homeostasis. Nat Genet. 1999;22:239–247. doi: 10.1038/10297. [DOI] [PubMed] [Google Scholar]
  • 90.Cargill M, Altshuler D, Ireland J, Sklar P, Ardlie K, Patil N, Lane CR, Lim EP, Kalayanaraman N, Nemesh J, Ziaugra L, Friedland L, Rolfe A, Warrington J, Lipshutz R, Daley GQ, Lander ES. Characterization of single-nucleotide polymorphisms in coding regions of human genes. Nat Genet. 1999;22:231–238. doi: 10.1038/10290. [DOI] [PubMed] [Google Scholar]
  • 91.Tang H, Quertermous T, Rodriguez B, Kardia SL, Zhu X, Brown A, Pankow JS, Province MA, Hunt SC, Boerwinkle E, Schork NJ, Risch NJ. Genetic structure, self-identified race/ethnicity, and confounding in case-control association studies. Am J Hum Genet. 2005;76:268–275. doi: 10.1086/427888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Campbell CD, Ogburn EL, Lunetta KL, Lyon HN, Freedman ML, Groop LC, Altshuler D, Ardlie KG, Hirschhorn JN. Demonstrating stratification in a European Am population. Nat Genet. 2005;37:868–872. doi: 10.1038/ng1607. [DOI] [PubMed] [Google Scholar]
  • 93.Freedman ML, Reich D, Penney KL, McDonald GJ, Mignault AA, Patterson N, Gabriel SB, Topol EJ, Smoller JW, Pato CN, Pato MT, Petryshen TL, Kolonel LN, Lander ES, Sklar P, Henderson B, Hirschhorn JN, Altshuler D. Assessing the impact of population stratification on genetic association studies. Nat Genet. 2004;36:388–393. doi: 10.1038/ng1333. [DOI] [PubMed] [Google Scholar]
  • 94.Burchard EG, Borrell LN, Choudhry S, Naqvi M, Tsai HJ, Rodriguez-Santana JR, Chapela R, Rogers SD, Mei R, Rodriguez-Cintron W, Arena JF, Kittles R, Perez-Stable EJ, Ziv E, Risch N. Latino populations: a unique opportunity for the study of race, genetics, and social environment in epidemiological research. Am J Public Health. 2005;95:2161–2168. doi: 10.2105/AJPH.2005.068668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Risch N. Dissecting racial and ethnic differences. N Engl J Med. 2006;354:408–411. doi: 10.1056/NEJMe058265. [DOI] [PubMed] [Google Scholar]
  • 96.Hirschhorn JN, Daly MJ. Genome-wide association studies for common diseases and complex traits. Nat Rev Genet. 2005;6:95–108. doi: 10.1038/nrg1521. [DOI] [PubMed] [Google Scholar]
  • 97.Bacanu SA, Devlin B, Roeder K. The power of genomic control. Am J Hum Genet. 2000;66:1933–1944. doi: 10.1086/302929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Devlin B, Roeder K. Genomic control for association studies. Biometrics. 1999;55:997–1004. doi: 10.1111/j.0006-341x.1999.00997.x. [DOI] [PubMed] [Google Scholar]
  • 99.Pritchard JK, Donnelly P. Case-control studies of association in structured or admixed populations. Theor Popul Biol. 2001;60:227–237. doi: 10.1006/tpbi.2001.1543. [DOI] [PubMed] [Google Scholar]
  • 100.Pritchard JK, Stephens M, Donnelly P. Inference of population structure using multilocus genotype data. Genetics. 2000;155:945–959. doi: 10.1093/genetics/155.2.945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Purcell S, Sham P. Properties of structured association approaches to detecting population stratification. Hum Hered. 2004;58:93–107. doi: 10.1159/000083030. [DOI] [PubMed] [Google Scholar]
  • 102.Satten GA, Flanders WD, Yang Q. Accounting for unmeasured population substructure in case-control studies of genetic association using a novel latent-class model. Am J Hum Genet. 2001;68:466–477. doi: 10.1086/318195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Tang H, Peng J, Wang P, Risch NJ. Estimation of individual admixture: analytical and study design considerations. Genet Epidemiol. 2005;28:289–301. doi: 10.1002/gepi.20064. [DOI] [PubMed] [Google Scholar]
  • 104.Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–909. doi: 10.1038/ng1847. [DOI] [PubMed] [Google Scholar]
  • 105.Hao K, Li C, Rosenow C, Wong WH. Detect and adjust for population stratification in population-based association study using genomic control markers: an application of Affymetrix Genechip Human Mapping 10K array. Eur J Hum Genet. 2004;12:1001–1006. doi: 10.1038/sj.ejhg.5201273. [DOI] [PubMed] [Google Scholar]
  • 106.Reiner AP, Ziv E, Lind DL, Nievergelt CM, Schork NJ, Cummings SR, Phong A, Burchard EG, Harris TB, Psaty BM, Kwok PY. Population structure, admixture, and aging-related phenotypes in African American adults: the Cardiovascular Health Study. Am J Hum Genet. 2005;76:463–477. doi: 10.1086/428654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.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;297:1551–1561. doi: 10.1001/jama.297.14.1551. [DOI] [PubMed] [Google Scholar]
  • 108.McCarthy JJ, Parker A, Salem R, Moliterno DJ, Wang Q, Plow EF, Rao S, Shen G, Rogers WJ, Newby LK, Cannata R, Glatt K, Topol EJ. Large scale association analysis for identification of genes underlying premature coronary heart disease: cumulative perspective from analysis of 111 candidate genes. J Med Genet. 2004;41:334–341. doi: 10.1136/jmg.2003.016584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Regensteiner JG, Steiner JF, Hiatt WR. Exercise training improves functional status in patients with peripheral arterial disease. J Vasc Surg. 1996;23:104–115. doi: 10.1016/s0741-5214(05)80040-0. [DOI] [PubMed] [Google Scholar]
  • 110.Paffenbarger RS, Jr, Hyde RT, Wing AL, Hsieh CC. Physical activity, all-cause mortality, and longevity of college alumni. N Engl J Med. 1986;314:605–613. doi: 10.1056/NEJM198603063141003. [DOI] [PubMed] [Google Scholar]
  • 111.Myers J, Gullestad L, Bellin D, Ross H, Vagelos R, Fowler M. Physical activity patterns and exercise performance in cardiac transplant recipients. J Cardiopulm Rehabil. 2003;23:100–106. doi: 10.1097/00008483-200303000-00006. [DOI] [PubMed] [Google Scholar]
  • 112.Tabor HK, Risch NJ, Myers RM. Opinion: candidate-gene approaches for studying complex genetic traits: practical considerations. Nat Rev Genet. 2002;3:391–397. doi: 10.1038/nrg796. [DOI] [PubMed] [Google Scholar]
  • 113.HapMap The International HapMap Project. Nature. 2003;426:789–796. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
  • 114.Skol AD, Scott LJ, Abecasis GR, Boehnke M. Joint analysis is more efficient than replication-based analysis for two-stage genome-wide association studies. Nat Genet. 2006;38:209–213. doi: 10.1038/ng1706. [DOI] [PubMed] [Google Scholar]
  • 115.Wang H, Thomas DC, Pe'er I, Stram DO. Optimal two-stage genotyping designs for genome-wide association scans. Genet Epidemiol. 2006;30:356–368. doi: 10.1002/gepi.20150. [DOI] [PubMed] [Google Scholar]
  • 116.Skol AD, Scott LJ, Abecasis GR. Boehnke M Optimal designs for two-stage genome-wide association studies. Genet Epidemiol. 2007 doi: 10.1002/gepi.20240. [DOI] [PubMed] [Google Scholar]
  • 117.Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, Haynes C, Henning AK, SanGiovanni JP, Mane SM, Mayne ST, Bracken MB, Ferris FL, Ott J, Barnstable C, Hoh J. Complement factor H polymorphism in age-related macular degeneration. Science. 2005;308:385–389. doi: 10.1126/science.1109557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Herbert A, Gerry NP, McQueen MB, Heid IM, Pfeufer A, Illig T, Wichmann HE, Meitinger T, Hunter D, Hu FB, Colditz G, Hinney A, Hebebrand J, Koberwitz K, Zhu X, Cooper R, Ardlie K, Lyon H, Hirschhorn JN, Laird NM, Lenburg ME, Lange C, Christman MF. A common genetic variant is associated with adult and childhood obesity. Science. 2006;312:279–283. doi: 10.1126/science.1124779. [DOI] [PubMed] [Google Scholar]
  • 119.Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, Perry JR, Elliott KS, Lango H, Rayner NW, Shields B, Harries LW, Barrett JC, Ellard S, Groves CJ, Knight B, Patch AM, Ness AR, Ebrahim S, Lawlor DA, Ring SM, Ben-Shlomo Y, Jarvelin MR, Sovio U, Bennett AJ, Melzer D, Ferrucci L, Loos RJ, Barroso I, Wareham NJ, Karpe F, Owen KR, Cardon LR, Walker M, Hitman GA, Palmer CN, Doney AS, Morris AD, Davey-Smith G, Hattersley AT, McCarthy MI. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316:889–894. doi: 10.1126/science.1141634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, Steinhart AH, Abraham C, Regueiro M, Griffiths A, Dassopoulos T, Bitton A, Yang H, Targan S, Datta LW, Kistner EO, Schumm LP, Lee AT, Gregersen PK, Barmada MM, Rotter JI, Nicolae DL, Cho JH. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314:1461–1463. doi: 10.1126/science.1135245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Helgadottir A, Thorleifsson G, Manolescu A, Gretarsdottir S, Blondal T, Jonasdottir A, Jonasdottir A, Sigurdsson A, Baker A, Palsson A, Masson G, Gudbjartsson D, 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;316:1491–1493. doi: 10.1126/science.1142842. [DOI] [PubMed] [Google Scholar]
  • 122.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;316:1488–1491. doi: 10.1126/science.1142447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Scott LJ, Mohlke KL, Bonnycastle LL, Willer CJ, Li Y, Duren WL, Erdos MR, Stringham HM, Chines PS, Jackson AU, Prokunina-Olsson L, Ding CJ, Swift AJ, Narisu N, Hu T, Pruim R, Xiao R, Li XY, Conneely KN, Riebow NL, Sprau AG, Tong M, White PP, Hetrick KN, Barnhart MW, Bark CW, Goldstein JL, Watkins L, Xiang F, Saramies J, Buchanan TA, Watanabe RM, Valle TT, Kinnunen L, Abecasis GR, Pugh EW, Doheny KF, Bergman RN, Tuomilehto J, Collins FS, Boehnke M. A genome-wide association study of type 2 diabetes in Finns detects multiple susceptibility variants. Science. 2007;316:1341–1345. doi: 10.1126/science.1142382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Steinthorsdottir V, Thorleifsson G, Reynisdottir I, Benediktsson R, Jonsdottir T, Walters GB, Styrkarsdottir U, Gretarsdottir S, Emilsson V, Ghosh S, Baker A, Snorradottir S, Bjarnason H, Ng MC, Hansen T, Bagger Y, Wilensky RL, Reilly MP, Adeyemo A, Chen Y, Zhou J, Gudnason V, Chen G, Huang H, Lashley K, Doumatey A, So WY, Ma RC, Andersen G, Borch-Johnsen K, Jorgensen T, van Vliet-Ostaptchouk JV, Hofker MH, Wijmenga C, Christiansen C, Rader DJ, Rotimi C, Gurney M, Chan JC, Pedersen O, Sigurdsson G, Gulcher JR, Thorsteinsdottir U, Kong A, Stefansson K. A variant in CDKAL1 influences insulin response and risk of type 2 diabetes. Nat Genet. 2007;39:770–775. doi: 10.1038/ng2043. [DOI] [PubMed] [Google Scholar]
  • 125.Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ, Kathiresan S, Hirschhorn JN, Daly MJ, Hughes TE, Groop L, Altshuler D, Almgren P, Florez JC, Meyer J, Ardlie K, Bengtsson K, Isomaa B, Lettre G, Lindblad U, Lyon HN, Melander O, Newton-Cheh C, Nilsson P, Orho-Melander M, Rastam L, Speliotes EK, Taskinen MR, Tuomi T, Guiducci C, Berglund A, Carlson J, Gianniny L, Hackett R, Hall L, Holmkvist J, Laurila E, Sjogren M, Sterner M, Surti A, Svensson M, Svensson M, Tewhey R, Blumenstiel B, Parkin M, Defelice M, Barry R, Brodeur W, Camarata J, Chia N, Fava M, Gibbons J, Handsaker B, Healy C, Nguyen K, Gates C, Sougnez C, Gage D, Nizzari M, Gabriel SB, Chirn GW, Ma Q, Parikh H, Richardson D, Ricke D, Purcell S. Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science. 2007;316:1331–1336. doi: 10.1126/science.1142358. [DOI] [PubMed] [Google Scholar]
  • 126.Zeggini E, Weedon MN, Lindgren CM, Frayling TM, Elliott KS, Lango H, Timpson NJ, Perry JR, Rayner NW, Freathy RM, Barrett JC, Shields B, Morris AP, Ellard S, Groves CJ, Harries LW, Marchini JL, Owen KR, Knight B, Cardon LR, Walker M, Hitman GA, Morris AD, Doney AS, for the Wellcome Trust Case Control Consortium Replication of genome-wide association signals in U.K. samples reveals risk loci for type 2 diabetes. Science. 2007;316:1336–1341. doi: 10.1126/science.1142364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Sladek R, Rocheleau G, Rung J, Dina C, Shen L, Serre D, Boutin P, Vincent D, Belisle A, Hadjadj S, Balkau B, Heude B, Charpentier G, Hudson TJ, Montpetit A, Pshezhetsky AV, Prentki M, Posner BI, Balding DJ, Meyre D, Polychronakos C, Froguel P. A genome-wide association study identifies novel risk loci for type 2 diabetes. Nature. 2007;445:828–830. doi: 10.1038/nature05616. [DOI] [PubMed] [Google Scholar]
  • 128.Wellcome Trust Case Control Consortium Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–678. doi: 10.1038/nature05911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Chanock SJ, Manolio T, Boehnke M, Boerwinkle E, Hunter DJ, Thomas G, Hirschhorn JN, Abecasis G, Altshuler D, Bailey-Wilson JE, Brooks LD, Cardon LR, Daly M, Donnelly P, Fraumeni JF, Jr, Freimer NB, Gerhard DS, Gunter C, Guttmacher AE, Guyer MS, Harris EL, Hoh J, Hoover R, Kong CA, Merikangas KR, Morton CC, Palmer LJ, Phimister EG, Rice JP, Roberts J, Rotimi C, Tucker MA, Vogan KJ, Wacholder S, Wijsman EM, Winn DM, Collins FS. Replicating genotype-phenotype associations. Nature. 2007;447:655–660. doi: 10.1038/447655a. [DOI] [PubMed] [Google Scholar]
  • 130.Fiegler H, Redon R, Andrews D, Scott C, Andrews R, Carder C, Clark R, Dovey O, Ellis P, Feuk L, French L, Hunt P, Kalaitzopoulos D, Larkin J, Montgomery L, Perry GH, Plumb BW, Porter K, Rigby RE, Rigler D, Valsesia A, Langford C, Humphray SJ, Scherer SW, Lee C, Hurles ME, Carter NP. Accurate and reliable high-throughput detection of copy number variation in the human genome. Genome Res. 2006;16:1566–1574. doi: 10.1101/gr.5630906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Redon R, Ishikawa S, Fitch KR, Feuk L, Perry GH, Andrews TD, Fiegler H, Shapero MH, Carson AR, Chen W, Cho EK, Dallaire S, Freeman JL, Gonzalez JR, Gratacos M, Huang J, Kalaitzopoulos D, Komura D, MacDonald JR, Marshall CR, Mei R, Montgomery L, Nishimura K, Okamura K, Shen F, Somerville MJ, Tchinda J, Valsesia A, Woodwark C, Yang F, Zhang J, Zerjal T, Zhang J, Armengol L, Conrad DF, Estivill X, Tyler-Smith C, Carter NP, Aburatani H, Lee C, Jones KW, Scherer SW, Hurles ME. Global variation in copy number in the human genome. Nature. 2006;444:444–454. doi: 10.1038/nature05329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Roeder K, Devlin B, Wasserman L. Improving power in genome-wide association studies: weights tip the scale. Genet Epidemiol. 2007 doi: 10.1002/gepi.20237. [DOI] [PubMed] [Google Scholar]
  • 133.Kimmel G, Shamir R. A fast method for computing high-significance disease association in large population-based studies. Am J Hum Genet. 2006;79:481–492. doi: 10.1086/507317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Musani SK, Shriner D, Liu N, Feng R, Coffey CS, Yi N, Tiwari HK, Allison DB. Detection of gene x gene interactions in genome-wide association studies of human population data. Hum Hered. 2007;63:67–84. doi: 10.1159/000099179. [DOI] [PubMed] [Google Scholar]
  • 135.Pickrell J, Clerget-Darpoux F, Bourgain C. Power of genome-wide association studies in the presence of interacting loci. Genet Epidemiol. 2007 doi: 10.1002/gepi.20238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Van Steen K, Lange C. PBAT: a comprehensive software package for genome-wide association analysis of complex family-based studies. Hum Genomics. 2005;2:67–69. doi: 10.1186/1479-7364-2-1-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Wallace C, Chapman JM, Clayton DG. Improved power offered by a score test for linkage disequilibrium mapping of quantitative-trait loci by selective genotyping. Am J Hum Genet. 2006;78:498–504. doi: 10.1086/500562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Chen Z, Zheng G, Ghosh K, Li Z. Linkage disequilibrium mapping of quantitative-trait Loci by selective genotyping. Am J Hum Genet. 2005;77:661–669. doi: 10.1086/491658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Slatkin M. Disequilibrium mapping of a quantitative-trait locus in an expanding population. Am J Hum Genet. 1999;64:1764–1772. doi: 10.1086/302413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Taute BM, Handschug K, Taute R, Seifert H, Glaser C, Podhaisky H. Angiotensin-converting enzyme gene insertion/deletion polymorphism and peripheral arterial occlusive disease. Vasa. 1998;27:149–153. [PubMed] [Google Scholar]
  • 141.Tseng CH, Tseng CP. Lack of association between angiotensin-converting enzyme gene polymorphism and peripheral vascular disease in type 2 diabetic patients in Taiwan. Circ J. 2002;66:1014–1018. doi: 10.1253/circj.66.1014. [DOI] [PubMed] [Google Scholar]
  • 142.Renner W, Pabst E, Paulweber B, Malaimare L, Iglseder B, Wascher TC, Pilger E. The angiotensin-converting-enzyme insertion/deletion polymorphism is not a risk factor for peripheral arterial disease. Atherosclerosis. 2002;165:175–178. doi: 10.1016/s0021-9150(02)00207-1. [DOI] [PubMed] [Google Scholar]
  • 143.Ghilardi G, Biondi ML, Battaglioli L, Zambon A, Guagnellini E, Scorza R. Genetic risk factor characterizes abdominal aortic aneurysm from arterial occlusive disease in human beings: CCR5 Delta 32 deletion. J Vasc Surg. 2004;40:995–1000. doi: 10.1016/j.jvs.2004.08.014. [DOI] [PubMed] [Google Scholar]
  • 144.Gugl A, Renner W, Seinost G, Brodmann M, Pabst E, Wascher TC, Paulweber B, Iglseder B, Pilger E. Two polymorphisms in the fracalkine receptor CX3CR1 are not associated with peripheral arterial disease. Atherosclerosis. 2003;166:339–343. doi: 10.1016/s0021-9150(02)00362-3. [DOI] [PubMed] [Google Scholar]
  • 145.Renner W, Schallmoser K, Gallippi P, Krauss C, Toplak H, Wascher TC, Pilger E. C242T polymorphism of the p22 phox gene is not associated with peripheral arterial occlusive disease. Atherosclerosis. 2000;152:175–179. doi: 10.1016/s0021-9150(99)00448-7. [DOI] [PubMed] [Google Scholar]
  • 146.Renner W, Koppel H, Brodmann M, Pabst E, Schallmoser K, Toplak H, Wascher TC, Pilger E. Factor II G20210A and factor V G1691A gene mutations and peripheral arterial occlusive disease. Thromb Haemost. 2000;83:20–22. [PubMed] [Google Scholar]
  • 147.Reny JL, Alhenc-Gelas M, Fontana P, Bissery A, Julia PL, Fiessinger JN, Aiach M, Emmerich J. The factor II G20210A gene polymorphism, but not factor V Arg506Gln, is associated with peripheral arterial disease: results of a case-control study. J Thromb Haemost. 2004;2:1334–1340. doi: 10.1111/j.1538-7836.2004.00809.x. [DOI] [PubMed] [Google Scholar]
  • 148.Mueller T, Marschon R, Dieplinger B, Haidinger D, Gegenhuber A, Poelz W, Webersinke G, Haltmayer M. Factor V Leiden, prothrombin G20210A, and methylenetetrahydrofolate reductase C677T mutations are not associated with chronic limb ischemia: the Linz Peripheral Arterial Disease (LIPAD) study. J Vasc Surg. 2005;41:808–815. doi: 10.1016/j.jvs.2005.01.039. [DOI] [PubMed] [Google Scholar]
  • 149.Lee AJ, Fowkes FG, Lowe GD, Connor JM, Rumley A. Fibrinogen, factor VII and plasminogenactivatorinhibitor (PAI)-1 genotypes and the risk of coronary and peripheral atherosclerosis: Edinburgh Artery Study. Thromb Haemost. 1999;81:553–560. [PubMed] [Google Scholar]
  • 150.Renner W, Brodmann M, Pabst E, Stanger O, Wascher TC, Pilger E. The V34L polymorphism of factor XIII and peripheral arterial disease. Int Angiol. 2002;21:53–57. [PubMed] [Google Scholar]
  • 151.Smith FB, Connor JM, Lee AJ, Cooke A, Lowe GD, Rumley A, Fowkes FG. Relationship of the platelet glycoprotein PlA and fibrinogen T/G+1689 polymorphisms with peripheral arterial disease and ischaemic heart disease. Thromb Res. 2003;112:209–216. doi: 10.1016/j.thromres.2003.11.010. [DOI] [PubMed] [Google Scholar]
  • 152.Fowkes FG, Connor JM, Smith FB, Wood J, Donnan PT, Lowe GD. Fibrinogen genotype and risk of peripheral atherosclerosis. Lancet. 1992;339:693–696. doi: 10.1016/0140-6736(92)90596-u. [DOI] [PubMed] [Google Scholar]
  • 153.Delanghe J, Langlois M, Duprez D, De Buyzere M, Clement D. Haptoglobin polymorphism and peripheral arterial occlusive disease. Atherosclerosis. 1999;145:287–292. doi: 10.1016/s0021-9150(99)00079-9. [DOI] [PubMed] [Google Scholar]
  • 154.Koppel H, Krippl P, Gasser R, Wascher TC, Paulweber B, Pilger E, Renner W. Hemochromatosis gene (HFE) polymorphisms are not associated with peripheral arterial disease. Thromb Haemost. 2004;91:1258–1259. [PubMed] [Google Scholar]
  • 155.Eller P, Schgoer W, Mueller T, Tancevski I, Wehinger A, Ulmer H, Foeger B, Haltmayer M, Ritsch A, Patsch JR. Hepatic lipase polymorphism and increased risk of peripheral arterial disease. J Intern Med. 2005;258:344–348. doi: 10.1111/j.1365-2796.2005.01549.x. [DOI] [PubMed] [Google Scholar]
  • 156.Gaetani E, Flex A, Pola R, Papaleo P, De Martini D, Pola E, Aloi F, Flore R, Serricchio M, Gasbarrini A, Pola P. The K469E polymorphism of the intercellularadhesionmolecule1 (ICAM-1) gene is a risk factor for peripheral arterial occlusive disease. Blood Coagul Fibrinolysis. 2002;13:483–488. doi: 10.1097/00001721-200209000-00002. [DOI] [PubMed] [Google Scholar]
  • 157.Renner W, Brodmann M, Winkler M, Washer TC, Pilger E. The PLA1/A2 polymorphism of platelet glycoprotein IIIa is not associated with peripheral arterial disease. Thromb Haemost. 2001;85:745–746. [PubMed] [Google Scholar]
  • 158.Flex A, Gaetani E, Pola R, Santoliquido A, Aloi F, Papaleo P, Dal Lago A, Pola E, Serricchio M, Tondi P, Pola P. The –174 G/C polymorphism of the interleukin (IL)-6 gene promoter is associated with peripheral artery occlusive disease. Eur J Vasc Endovasc Surg. 2002;24:264–268. doi: 10.1053/ejvs.2002.1711. [DOI] [PubMed] [Google Scholar]
  • 159.Stricker H, Soldati G, Balmelli T, Mombelli G. Homocysteine, vitamins and gene mutations in peripheral arterial disease. Blood Coagul Fibrinolysis. 2001;12:469–475. doi: 10.1097/00001721-200109000-00007. [DOI] [PubMed] [Google Scholar]
  • 160.Fontana P, Gaussem P, Aiach M, Fiessinger JN, Emmerich J, Reny JL. P2Y12 H2 haplotype is associated with peripheral arterial disease: a case-control study. Circulation. 2003;108:2971–2973. doi: 10.1161/01.CIR.0000106904.80795.35. [DOI] [PubMed] [Google Scholar]

RESOURCES