Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Apr 1.
Published in final edited form as: Am J Hypertens. 2010 Oct 14;24(4):392–400. doi: 10.1038/ajh.2010.218

Recent Findings in the Genetics of Blood Pressure and Hypertension Traits

Nora Franceschini 1, Alexander P Reiner 2, Gerardo Heiss 1
PMCID: PMC3110743  NIHMSID: NIHMS294745  PMID: 20948529

Abstract

We provide an overview of ongoing discovery efforts in the genetics of blood pressure (BP) and hypertension (HTN) traits. Two large genome-wide association meta-analyses of individuals of European descent were recently published, revealing ~13 new loci for BP traits. Only two of these loci harbor genes in a pathway known to affect BP (CYP17A1 and NPPA/NPPB). Functional variants in these loci are still unknown. Few genome-wide association studies (GWAS) of complex diseases have been published from non-European populations. The study of populations with different evolutionary history and linkage disequilibrium (LD) structure, such as individuals of African ancestry, may provide an opportunity to further narrow these regions to identify the causal gene(s). Several collaborative efforts toward discovery of low-frequency variants and copy number variation for BP traits are currently underway. As evidence for new loci for complex diseases accumulates the assessment of the epidemiologic architecture of these variants in populations assumes higher priority. The impact of public health–relevant contexts such as diet, physical activity, psychosocial factors, and aging has not been examined for most common variants associated with BP.

Keywords: blood pressure, genes, genome-wide association, hypertension


Hypertension (HTN) is a leading risk factor for cardiovascular disease morbidity and mortality, and renal disease.1,2 HTN affects 24% of the US adult population,3 with a disproportionate burden in minorities, particularly African Americans.4 The regulation of blood pressure (BP) is complex and multiple genes likely interact to influence BP, modulated by environmental contexts such as diet, physical activity, psychosocial factors, and aging. Although some of the pathways of BP control have been well described in humans and experimental models,58 identifying genes that contribute to the distribution of BP in populations and the underlying biology has proved challenging (reviewed in refs. 9,10).

The extensive and important work in functional variant discovery using genome-wide linkage and candidate gene approaches will not be reviewed here and is summarized elsewhere.11,12 Recent discovery efforts have used hypothesis-free association scans of the genome (genome-wide association studies, GWAS) to identify new loci for HTN susceptibility and BP distribution. So far, two large meta-analyses of GWAS in individuals of European ancestry have been published.13,14 Several collaborations to study BP traits are currently ongoing with expected publications of novel genetic findings in the next several months. Loci identified may provide new insights into the biology of BP regulation, and suggest targets for therapy and for population level risk reduction.

Here, we summarize the recent genetic findings of BP traits, while attempting to provide some perspective on the discovery efforts and impact on public health and clinical care. We review recently published GWAS of HTN and BP traits and outline other strategies of gene discovery focused on BP such as low-frequency variants and copy number variations. Finally, we discuss recent efforts toward generalization of these discoveries to individuals of different ancestry. Because the field is rapidly evolving, this review offers a snap shot of the current state of the knowledge of the genetics of BP traits.

OVERVIEW OF GENE DISCOVERY FOR BP TRAITS AND HTN

Common variants

GWAS findings in individuals of European ancestry

GWAS use dense sets of genetic markers to investigate associations of common single-nucleotide polymorphism (SNP) with complex traits. The analyses rely on the linkage disequilibrium (LD) or correlation patterns of typed (or imputed) SNPs with functional variants and, therefore, identified SNPs are usually proxies of untyped functional variants.15 At a genome-wide level, >80% of common SNPs (defined as having a minor allele frequency of at least 5%) can be interrogated by genotyping in the range of 500,000 to 1 million SNPs, depending on the population. To adjust for multiple testing and to decrease type I error (false-positive rates) stringent P values are used. Consortia and collaborative studies have enabled meta-analyses of multiple GWAS based on large samples (usually in the range of tens of thousands to >100,000 individuals) and statistical power sufficient to identify new common variants. Most published studies to date have targeted individuals of European ancestry. GWAS strategy for discovery has been successful in identifying new genes and loci for complex traits, although these findings explain a small proportion of the trait variability.16 Because this method relies on proxies of causal variants, the proportion of variation explained by common genetic variants is small, but may be underestimated.16,17 The heritability of BP is estimated as 30–40% in populations,9 but gene discovery has proved challenging for BP traits.18,19

Two large GWAS meta-analyses of individuals of European descent were recently published,13,14 revealing ~13 new loci for BP traits (Tables 1 and 2). These analyses were cross-sectional, included cohort and case–control studies, and involved individuals with a broad range of age and HTN prevalence (Table 1). The main genetic analyses assessed systolic (SBP) and diastolic (DBP) BP as quantitative traits, for which statistical power is generally greater than categorical outcomes such as HTN. Minimal covariate adjustments were performed: age, sex, and body mass index. For individuals using BP-lowering medications, a constant was added to measured BP. The study by Levy et al. had a discovery sample of over 29,000 participants from the Cohorts for Heart and Aging Research in Genome Epidemiology (CHARGE Consortium).14 Replication was done in 34,000 individuals participants of the Global Blood Pressure Genetics (Global BPgen) Consortium. Global BPgen recruited 13 cohort studies and 4 nonhypertensive control groups from case–control studies for the discovery phase.13 Global BPgen used the CHARGE Consortium samples and additional 70,000 samples of European individuals for in silico replication, and de novo genotyping was performed in 13,000 Asians (Table 1).

Table 1.

Comparison of study design, discovery, and replication samples for blood pressure traits in two published genome-wide association meta-analyses of individuals of European ancestry

Discovery Number of studies Mean age across studies (years) Mean BMI across studies (kg/m2) HTN (%) Treated HTN Primary traitsb Strategy for SNP selection for replication Replication
Studiesa Sample size Ancestry In silico De novo genotyping
CHARGE14 29,136 European 6 cohort studies (one family design) 38–72 25–27 17–60% 5–35% SBP, DBP and HTN P < 4 × 10−7 34,433 from Global Bpgen N/A
Global BPgen13 34,433 European 13 population- based (including isolates) and controls only from 4 case–control studies 31–59c 23.5–27.4 15–52% 2–24% SBP, DBP P < 10−5 (2 SNPs with P < 5 × 10−8) 29,136 from CHARGE 71,225 individuals of European ancestry (13 cohorts), 12,889 Indian Asians

BMI, body mass index; DBP, diastolic blood pressure; HTN, hypertension; N/A, not available; SBP, systolic blood pressure; SNP, single-nucleotide polymorphism.

a

CHARGE BP discovery studies included: the Age, Gene/Environment Susceptibility Reykjavik Study (AGES), Cardiovascular Heath Study (CHS), Framingham Heart Study (FHS), Rotterdam Study and Rotterdam Extension Study (RES); Global BPgen discovery studies included: the Baltimore Longitudinal Study of Aging, British 1958 Birth Cohort (B58C-T1DGC and B58C-WTCCC), Cohorte Lausannoise (CoLaus), Diabetes Genetic Initiative (DGI), European Prospective Investigation of Cancer-Norfolk-Genome Wide Association Study (EPIC-Norfolk-GWAS), Fenland Study, Finland-United States Investigation of NIDDM Genetics (FUSION) Study, Invecchiarie in Chianti (InCHIANTI), Kooperative Gesundheitsforschung in der Region Augsburg (KORA), the Myocardial Infarction Genetics Consortium (MIGen), Northern Finland Birth Cohort of 1996 (NFBC1966), SardiNIA, Study of Health in Pomerania (SHIP), the Precocius Coronary Artery Disease (PROCARDIS), Supplementation en Vitamines et Mineraux Antioxydants (SU.VI.MAX), and TwinsUK.

b

Analyses were adjusted for age, age2, body mass index, study site, and population stratification. Method of adjustment for blood pressure medications in quantitative analyses were: CHARGE, addition of 10 mm Hg to SBP and 5 mm Hg to DBP; Global BPgen, addition of 15 mm Hg to SBP and 10 mm Hg to DBP.

c

Individuals >70 years were excluded.

Table 2.

Loci associated with blood pressure CHARGE and Global BPgen meta-analyses

Loci Nearby genes Trait: P valuea SNP Coded allele Coded allele frequency β (s.e.) Total number with replication Reference
1p36 MTHFR SBP: 2 × 10−13 rs17367504 G 0.14 −0.85 (0.11) 82,973 13
CLCN6
NPPA
NPPB
AGTRAP
3p22.1 ULK4 DBP: 3 × 10−9 rs9815354 A 0.17 0.49 (0.08) Up to 63,569 14
4q21 PRDM8 DBP: 1 × 10−21 rs16998073 T 0.21 0.50 (0.05) 101,623 13
FGF5
c4orf22
10q21 c10orf107 DBP: 1 × 10−9 rs1530440 T 0.19 −0.39 (0.06) 87,273 13
TMEM26 SBP: 1 × 10−10 rs1004467 A 0.90 1.05 (0.16) Up to 63,569
RTKN2
RHOBTB1
ARID5B
10q24 CYP17A1 SBP: 7 × 10−24 rs11191548 T 0.91 1.16 (0.12) 132,552 14,13
AS3MT SBP: 1 × 10−10 rs1004467 A 0.90 1.05 (0.16) Up to 63,569
CNNM2
NT5C2
10p12 CACNB2 DBP: 1 × 10−8 rs11014166 A 0.66 0.37 (0.06) Up to 63,569 14
11p15 PLEKHA7 SBP: 2 × 10−9 rs381815 T 0.26 0.65 (0.11) Up to 63,569 14
12q21 ATP2B1 SBP: 4 × 10−11 rs2681492 T 0.80 0.85 (0.13) Up to 63,569 14
DBP:2 × 10−9 rs2681472 A 0.83 0.50 (0.08) Up to 63,569
HTN: 2 × 10−11 rs2681472 A 0.83 0.15 (0.02) Up to 63,569
12q24 SHS2B3 DBP: 3 × 10−18 rs653178 T 0.53 −0.46 (0.05) 79,661 14,13
ATXN2 SBP: 5 × 10−9 rs3184504 T 0.48 0.58 (0.10) Up to 63,569
DBP: 3 × 10−14 0.49 0.48 (0.06) Up to 63,569
12q24.21 TBX3 DBP: 4 × 10−8 rs2384550 A 0.35 −0.35 (0.06) Up to 63,569 14
TBX5
15q24 CYP1A1 DBP: 1 × 10−23 rs1378942 C 0.36 0.43 (0.04) 134,258 14,13
CYP1A2 DBP: 2 × 10−10 rs6495122 A 0.42 0.40 (0.06) Up to 63,569
CSK
LMAN1L
CPLX3
ARID3B
ULK3
17q21 ZNF652 DBP: 5 × 10−9 rs16948048 G 0.39 0.31 (0.05) 82,441 13
PHB
17q21 PLCD3 SBP: 1 × 10−8 rs12946454 T 0.28 0.57 (0.10) 77,690 13
ABCD4
HEXIM1
HEXIM2

DBP, diastolic blood pressure; HTN, hypertension; SBP, systolic blood pressure; SNP, single-nucleotide polymorphisms.

a

Only P value <5 × 10−8 are reported.

The main findings of these two meta-analyses are summarized in Table 2. CHARGE identified eight loci associated with SBP and DBP near or in CYP17A1 (SBP), PLEKHA7 (SBP), ATP2B1 (SBP, DBP, HTN), SH2B3-ATXN2 (SBP, DBP), ULK4 (DBP), TBX3-TBX5 (DBP), CSK-ULK3 (DBP) and CACNB2 (DBP) (Table 2). Global BPgen identified eight loci for BP in regions near or in MTHFR-CLCN6-NPPA-NPPB (SBP), CNNM2-NT5C2-CYP17A1 (SBP), PLCD3 (SBP), FGF5 (DBP), C10orf107 (DBP), SH2B3-ATXN2 (DBP), CSK (DBP), and ZNF652 (DBP). Three loci were identified in both meta-analyses: CYP17A1, SH2B3-ATXN2 and CSK-ULK3.

As expected for common variants, the association effect sizes were small (<1 mm Hg) and explained a small proportion of the phenotypic variation. Studies identified the same regions for SBP and DBP. Although this is not surprising given the high correlation between SBP and DBP, it remains to be determined whether genetic predisposition applies to different types of HTN and those more common among certain demographic groups (e.g., isolated systolic HTN in older adults). Given complex and different regulatory mechanisms at play, alternative phenotypic traits may prove to be informative as strategies for investigation of BP genes, such as BP responses to stressors and responsiveness to dietary and pharmacologic interventions.20

Some of the BP-associated regions identified through GWAS extend over several genes. Because of the strong LD or local correlation between SNPs in Caucasians it is difficult to localize the causal gene(s) for some of these loci. For example, the 10q24 region identified for SBP spans ~430 kb and includes six genes.13,14 The 3p22.1 and 12q24.21 loci similarly apply to regions with multiple genes.14 The study of populations with different evolutionary history and LD structure may provide an opportunity to narrow such regions in efforts to identify the causal gene(s).

Of several identified loci only two regions, 1p36 and 10q24, harbor genes in a pathway known to affect BP (CYP17A1 and NPPA/NPPB, respectively). Mutations in the CYP17A1 gene, which encodes the cytochrome P450 enzyme CYP17A1, cause congenital adrenal hyperplasia and hypokalemic HTN.21 Common variants in the NPPA and NPPB genes associated with natriuretic peptide concentrations have been previously shown to contribute to BP and HTN risk.22 Even though candidate gene analysis have not been proven successful in revealing variants associated with BP, these findings suggest that common variants in these genes may indeed contribute to BP variation in the population at large. In fact, a recent study of WKN1, in which mutations lead to HTN and hyperkalemia of the Gordon syndrome, identified associations of both common and rare genetic variants in the WKN1 gene with BP and potassium excretion. Such results indicate that targeted candidate gene sequencing efforts may provide informative discovery sets for both common and rare variants, in genes associated with known pathways of BP control including monogenic disorder of hypotension and HTN.

The 12q24 region associated with BP contains ~15 genes in addition to SH2B3 and has been associated pleiotropically with multiple phenotypes. rs3184504 of SH2B3 encodes an arginine to tryptophan substitution at amino acid 262 (R262W) in the lymphocyte-specific adapter protein (LNK). In addition to BP, the allele corresponding to the 262W variant has been associated with higher blood cell counts (hemoglobin/hematocrit, eosinophils, and platelets) as well as increased risk of autoimmune diseases (celiac disease and type 1 diabetes) and myocardial infarction.2326 Molecules that contain an SH2 domain in their structure, like LNK, regulate growth factor and cytokine receptor–mediated pathways implicated in hematopoiesis and immunity.27,28 Cells isolated from individuals homozygous for the SH2B3 risk allele display increased production of proinflammatory cytokines such as interleukin-1-β. There is a strong signal of positive selection at the SH2B3 locus in Europeans, which has been postulated as due to improved host defense against infections.24,29 Consistent with the association of the SH2B3 locus with myocardial infarction, LNK also may play a role in platelet activation and stabilization of a developing thrombus.30 The relationship to BP physiology may be less apparent, though LNK has also been reported to regulate bone marrow–derived endothelial progenitor cell proliferation and mobilization31 as well as cytokine-induced signaling nitric oxide synthase activity in human ECs.32

Gender differences in BP are well-documented in different populations3,33,34 and have been largely attributed to sex hormone effects.35 Sex-specific quantitative trait loci have been described for BP traits.36,37 In contrast, BP-associated SNPs identified in recent GWAS by Global BPgen did not show evidence for sex-specific effects.13 Potential interactions with important environmental determinants of BP, such as diet, body mass, physical activity, and behavioral factors,38 have yet to be investigated systematically.

Findings from other GWAS of individuals of European ancestry are summarized in Table 3, as studies of smaller sample sizes, most of which contributed data to the above described meta-analyses. Currently, a joint meta-analysis of Global BPgen and CHARGE is underway, which may uncover additional loci with even smaller effects. In addition, studies using high-density custom arrays for fine mapping of regions identified in GWAS are in progress.39

Table 3.

Additional published GWAS by race/ethnicities

Study Race–ethnicity/country Genotyping platform Discovery samples Replication samples Traits Findings SNP (P value: trait)
WTCCC18 Europeans/Europe Affymetrix GeneChip 500K Mapping Array 2,000 hypertensive cases, 3,000 controls None HTN rs2820037 (P = 8 × 10−7: HTN)
Levy et al.19 European Americans/United States Affymetrix Human Mapping 100K Up to 1,327 individuals None SBP, DBP, long-term averaged SBP and DBP rs10493340 (P = 2 × 10−7: SBP) and rs1963982 (P = 3 × 10−6: DBP)
Adeyemo et al.43 African Americans/United States and West Africans/Africa Affymetrix Genome-Wide Human SNP Array 6.0 509 case and 508 control African Americans 980 nondiabetic West African case–controlsa HTN, SBP, and DBP (among controls only) Rs5743185 (P = 2 × 10−11), rs16877320 (P = 3 × 10−9), rs11160059 (P = 2 × 10−8), rs17365948 (P = 2 × 10−8), rs12279202 (P = 5 × 10−8)
Org et al.69 Europeans/Germany, Estonia, and United Kingdom Affymetrix GeneChip 500K Mapping Array Up to 1,017 individuals ~3,766 individuals from three studies HTN, SBP, and DBP rs11646213 (P = 8.3 × 10−6)b
Yang et al.17 Han Chinese/China Affymetrix Human Mapping 100K 175 cases, 175 age- and sex-matched controls 833 cases and 833 normotensive controls HTN No significant findings for single locus analyses
Wang et al.70 Amish and non-Amish European Americans/United States Affymetrix Human Mapping 100K 542 Amish subjects (119 with type 2 diabetes, 132 with impaired glucose tolerance, and 291 with normal glucose tolerance) 1,347 Amish; ~5,800 individuals from four European-American studies SBP and DBP rs4977950 (P = 9 × 10−8: SBP), SNPs in STK39 (P < 10−7)
Cho et al.42 Korean/Korea Affymetrix Genome-Wide Human SNP array 5.0 8,842 Korean individuals 7,861 Korean individuals SBP and DBP rs17249754c (P = 9 × 10−7 for discovery and 1 × 10−7 for combined meta-analysis: SBP)
Takeuchi et al.41 Japanese/Japan Infinium HumanHap550 BeadArray (Illumina) 1,526 individuals for GWAS 3,526 individuals from a case–control study and >16,000 individuals from two cohort studies used for replication of published findings HTN, SBP, and DBP No significant findings for genome- wide association analysis; replication of seven described loci: CASZ1, MTHFR, ITGA9, FSF5, CYP17A1- CNNM2, ATP2B1, and CSK-ULK3

DBP, diastolic blood pressure; GWAS, genome-wide association studies; HTN, hypertension; SBP, systolic blood pressure; SNP, single-nucleotide polymorphism; WTCCC, Wellcome Trust Case Control Consortium.

a

Replication using α = 0.05.

b

Near the CDH13 gene.

c

ATP2B.

GWAS discovery in individuals of non-European ancestry

Differences in allele frequency and LD patterns as well as in disease prevalence between populations of different ancestry may be helpful in identifying loci and functional variants for complex diseases.40 Few GWAS of complex diseases in non-European populations have been published and only four studies are known to have investigated BP traits (Table 3). A study of Han Chinese (175 hypertensive cases and 175 normotensive controls) failed to identify genome-wide associated loci.17 A GWAS of 1,526 Japanese individuals (403 cases and 452 controls) similarly did not reveal significant loci associated with HTN.41 These studies had limited statistical power due to study size and the restricted focus on HTN. Interestingly, a study of over 16,000 Korean individuals identified a BP-associated SNP (rs17249754, P = 1 × 10−7 in combined meta-analysis) near the ATP2B gene, the region also identified in the CHARGE Consortium.14,42

Despite the high prevalence of HTN in African Americans, only one BP GWAS in individuals of African ancestry has been published to date. Included in the discovery sample were 509 hypertensives and 508 normotensive controls enrolled in Washington DC.43 This study identified several loci for SBP and DBP but these findings did not replicate in independent samples comprising 980 unrelated nondiabetic West Africans enrolled in the Africa America Diabetes (AADM) study.43 Additional GWAS involving larger number of US minority participants are currently underway, including the Women’s Heath Initiative minority sample (WHI-SHARe) and the Candidate Gene Association Resource (CARe).44

Identification of BP genetic variants in African-American and other minority populations poses particular challenges. Foremost are the limited availability of large samples, a reduced coverage of common variants on current genotyping platforms, and a lack of well-validated genotype imputation methods in populations of mixed genetic ancestry. In addition, population stratification may lead to increased false positives but also may obscure true associations. Additional strategies for gene finding in populations of African ancestry include admixture models based on alleles that differ in frequency across populations of different ancestry and are thus suitable to the study of diseases, such as HTN, with disparities among racial/ethnic groups.45,46

Rare variants

Although GWAS have led to the discovery of new genes or genomic regions associated with BP traits, the functional variants underlying these associations remain largely unknown. It is therefore possible that the true functional variants, whether common or rare, have larger effect sizes than the proxy SNPs identified in GWAS. Localization or fine mapping of associated genomic regions will likely contribute to understanding the biological processes involved in BP regulation.

Contributions by low-frequency genetic variants to complex traits47 including BP have been demonstrated.48,49 Mutations in salt-handling genes have been identified in rare families with Mendelian forms of HTN or hypotension. In the community-based Framingham Heart Study, putatively functional rare variants in renal salt–handling genes originally identified in patients with Gitelman’s or Bartter’s syndrome (SLC12A3, SLC12A1, and KCNJ1) were shown to contribute to BP variation at the population level.49 Different mutations within the same gene can lead to low or high BP depending on whether the mutation is associated with gain or loss of function of the encoded protein. Low-frequency variants (minor allele frequency <2–3%) may contribute to large variability of the trait in the aggregate, or may individually have a larger effect.

A recent Wellcome Trust Case Control Consortium (WTCCC) investigation used haplotype analyses to genome-wide association data to identify the contribution of rare genetic variants to several common complex diseases.50 The study identified a novel locus for HTN (ZFATI), not tagged by SNPs, which replicated in an independent sample. However, the replication was not in the same block suggesting a role for multiple rare variants within the gene/region influencing the susceptibility to HTN.

Collectively, the rare and common variants identified to date explain <2% of population variation in BP. Because rare non-synonymous variants in salt-handling genes have had large effects on BP in the population,49 it is possible that a multitude of highly penetrant, low-frequency alleles affecting a variety of pathways involved in BP regulation explain a substantial proportion of the remaining heritability.47 An effective way to identify low-frequency genetic variants responsible for heritable disease–related traits is through DNA sequencing, which can directly identify disease mutations in the genome that are not tagged by GWAS panels of common SNPs. Advances in next-generation DNA technologies and the resultant substantial reductions in the cost of DNA sequencing have enabled direct sequencing of the entire genome (whole-genome sequencing). An alternative approach is whole-genome analysis of all coding sequences (exome sequencing), which focuses variant discovery on coding regions that are more likely to be functional. Such techniques are actively being pursued in novel variant discovery.51 The 1,000 Genomes project, which is sequencing genomes from individuals of 10 different ethnicities (http://1000genomes.org/page.php?page=home), will provide a catalog of human variants to help in the identification of disease-causing variants. Whole-genome sequencing has been successfully applied to discover mutations in patients with rare Mendelian disorders52 and efforts such as the National Heart, Lung, and Blood Institute–funded Exome Sequencing Project, apply exonic whole-genome sequencing to larger numbers of samples in populations and less highly penetrant, more complex phenotypes, such as BP. These studies allow for the identification of variants in a broad range of allele frequency by sequencing participants from the extreme tails of the distribution of traits such as BP, but also pose significant analytic challenges in the evaluation of very rare variants at reasonable statistical power.

Structural variations

Structural variants refer to DNA segments that differ in copy number between individuals and they include insertions–deletions (indels), inversions, duplications, and other copy number variations.53 In addition to their association with rare Mendelian disorders, copy number variations may have an important role in genetic susceptibility to common diseases.54,55 Recent efforts to map and validate structural variants have identified several challenges, including methodological limitations in approaches to capture the spectrum of genetic variations and in the quality of the data.56 The WTCCC has performed a comprehensive analysis of copy number variation in Caucasians for eight common traits including HTN.55 The study replicated three loci where copy number variations were associated with various diseases, but not for the trait HTN. Interestingly, several validated common copy number variations were well tagged by SNPs and therefore could be studied using GWAS data.55 Conrad et al. recently showed that 474 of 1,521 polymorphic trait-associated SNPs identified in GWAS of individuals of European ancestry fell within a recombination hotspot interval that also contained a copy number variation with correlations of 0.5 or higher.54 As mapping of structural variations continues to be refined, the impact of these variants in complex diseases may be more evident.

EPIDEMIOLOGIC ARCHITECTURE OF BP-ASSOCIATED GENETIC VARIANTS

As evidence for new loci for complex diseases accumulates, obvious logical next step is to assess the epidemiologic architecture of these variants in diverse populations. Although the functional variants underlying the risk are mostly unknown, the population impact of such variants is yet to be determined, particularly in public health–relevant environmental and behavioral contexts such as diet, physical activity, psychosocial factors, and age. Integration of individual-level and group-level (social, cultural, and behavioral) contexts into molecular/genetic analyses has been recently proposed.57 Comparability in, and high quality of phenotypic characterization and of exposure data across studies will be essential to such efforts.

The degree to which associations with mature genetic variants can be generalized to populations of diverse ethnic/racial ancestry has not been explored sufficiently. A recent study attempted to validate findings of BP GWAS of European ancestry studies to Japanese subjects.41 This study of 24,000 Japanese individuals from three cohort studies replicated associations with BP or HTN at 7 of 13 investigated loci including MTHFR, FGF5, CYP17A1, ATP2B1, CSK-ULK3, ITGA9, and CASZ1. The SNP in SH2B3 was monomorphic in this study’s samples. Interestingly, two SNPs, rs880315 (CASZ1) and rs16998073 (FGF5), showed significant interpopulation effect heterogeneity, with larger effect in Japanese than Europeans. A study of 8,512 Korean individuals replicated associations of BP with ATP2B1, CSK-ULK3, CYP17A1, and PLEKHA7.58 In African Americans, Adeyemo et al. replicated associations with BP for SNPs in STK39 and CDH13.43

The recently funded Population Architecture using Genomics and Epidemiology (PAGE) Study (www.pagestudy.org) proposes a comprehensive analysis of the association of genetic variants with complex traits. The PAGE consortium consists of four large ongoing NIH-funded population-based studies or consortia: the Women’s Health Initiative (WHI),59 Epidemiologic Architecture for Genes Linked to Environment (EAGLE),60 the Multiethnic Cohort study (MEC),61 and CALICo (Causal Variants Across the Life Course), which includes five cohort studies: the Atherosclerosis Risk in Communities study (ARIC),62 the Coronary Artery Risk in Young Adults (CARDIA),63 the Cardiovascular Health Study (CHS),64 the Study of Latinos (HCHS/SOL), and the Strong Heart Study.65,66 These data from multiple prospective cohort and case–control studies allow analyses of genetic variants within diverse cultural, ancestral, and socioeconomic settings and replication of SNP associations in populations of different ancestry including African Americans, Hispanics, American Indians, and Asians is currently ongoing. Studies such as PAGE may provide insights into genetic susceptibility to BP elevation and HTN across race and ethnic groups and the role of environmental and cultural factors in this regard.

LIMITATIONS AND OPPORTUNITIES

Current genetic discovery efforts have been mostly limited to resting BP measured at a single time point, mostly because large sample sizes with phenotypic measurements are required. The study of BP variability or ambulatory BP monitoring may contribute to deepen current insights into the genetic susceptibility of BP levels, their variability, and temporal changes.67 Similarly, gene discovery efforts based on BP responses to nonpharmacological (diet and physical activity) and pharmacological interventions may help guide prevention efforts and HTN therapy.

Beyond the challenges outlined for the identification of rare and common SNPs and structural variants, and in addition to studies exploring gene–gene and gene–environment interactions, research on the effect of sex chromosomes and mitochondrial variants on BP are needed. Finally, epigenetic effects on BP phenotypes have been understudied to date, despite experimental models indicating that HSD11B2 is repressed by DNA methylation, suggesting that epigenetic mechanisms affect interindividual differences in the expression of BP regulation.68

Acknowledgments

A.P.R. was supported by NIH grant R01 HL071862; N.F. by AHA 0675001N, R01 HL089651, and (U01) RFA-HG-07-014; and G.H. by (U01) RFA-HG-07-014.

Footnotes

Disclosure: The authors declared no conflict of interest.

References

  • 1.Lewington S, Clarke R, Qizilbash N, Peto R, Collins R. Prospective Studies Collaboration. Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet. 2002;360:1903–1913. doi: 10.1016/s0140-6736(02)11911-8. [DOI] [PubMed] [Google Scholar]
  • 2.Hsu CY, McCulloch CE, Darbinian J, Go AS, Iribarren C. Elevated blood pressure and risk of end-stage renal disease in subjects without baseline kidney disease. Arch Intern Med. 2005;165:923–928. doi: 10.1001/archinte.165.8.923. [DOI] [PubMed] [Google Scholar]
  • 3.Burt VL, Whelton P, Roccella EJ, Brown C, Cutler JA, Higgins M, Horan MJ, Labarthe D. Prevalence of hypertension in the US adult population. Results from the Third National Health and Nutrition Examination Survey, 1988–1991. Hypertension. 1995;25:305–313. doi: 10.1161/01.hyp.25.3.305. [DOI] [PubMed] [Google Scholar]
  • 4.Hajjar I, Kotchen TA. Trends in prevalence, awareness, treatment, and control of hypertension in the United States, 1988–2000. JAMA. 2003;290:199–206. doi: 10.1001/jama.290.2.199. [DOI] [PubMed] [Google Scholar]
  • 5.Guyton AC, Coleman TG, Cowley AV, Jr, Scheel KW, Manning RD, Jr, Norman RA., Jr Arterial pressure regulation. Overriding dominance of the kidneys in long-term regulation and in hypertension. Am J Med. 1972;52:584–594. doi: 10.1016/0002-9343(72)90050-2. [DOI] [PubMed] [Google Scholar]
  • 6.Guyton AC. Long-term arterial pressure control: an analysis from animal experiments and computer and graphic models. Am J Physiol. 1990;259:R865–R877. doi: 10.1152/ajpregu.1990.259.5.R865. [DOI] [PubMed] [Google Scholar]
  • 7.Izzo JL., Jr The sympathoadrenal system in the maintenance of elevated arterial pressure. J Cardiovasc Pharmacol. 1984;6 (Suppl 3):S514–S521. [PubMed] [Google Scholar]
  • 8.Hall JE. Historical perspective of the renin-angiotensin system. Mol Biotechnol. 2003;24:27–39. doi: 10.1385/MB:24:1:27. [DOI] [PubMed] [Google Scholar]
  • 9.Hopkins PN, Hunt SC. Genetics of hypertension. Genet Med. 2003;5:413–429. doi: 10.1097/01.gim.0000096375.88710.a6. [DOI] [PubMed] [Google Scholar]
  • 10.Samani NJ. Genome scans for hypertension and blood pressure regulation. Am J Hypertens. 2003;16:167–171. doi: 10.1016/s0895-7061(02)03244-2. [DOI] [PubMed] [Google Scholar]
  • 11.Binder A. A review of the genetics of essential hypertension. Curr Opin Cardiol. 2007;22:176–184. doi: 10.1097/HCO.0b013e3280d357f9. [DOI] [PubMed] [Google Scholar]
  • 12.Kitsios GD, Zintzaras E. Synopsis and data synthesis of genetic association studies in hypertension for the adrenergic receptor family genes: the CUMAGAS-HYPERT database. Am J Hypertens. 2010;23:305–313. doi: 10.1038/ajh.2009.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, Coin L, Najjar SS, Zhao JH, Heath SC, Eyheramendy S, Papadakis K, Voight BF, Scott LJ, Zhang F, Farrall M, Tanaka T, Wallace C, Chambers JC, Khaw KT, Nilsson P, van der Harst P, Polidoro S, Grobbee DE, Onland-Moret NC, Bots ML, Wain LV, Elliott KS, Teumer A, Luan J, Lucas G, Kuusisto J, Burton PR, Hadley D, McArdle WL, Brown M, Dominiczak A, Newhouse SJ, Samani NJ, Webster J, Zeggini E, Beckmann JS, Bergmann S, Lim N, Song K, Vollenweider P, Waeber G, Waterworth DM, Yuan X, Groop L, Orho-Melander M, Allione A, Di Gregorio A, Guarrera S, Panico S, Ricceri F, Romanazzi V, Sacerdote C, Vineis P, Barroso I, Sandhu MS, Luben RN, Crawford GJ, Jousilahti P, Perola M, Boehnke M, Bonnycastle LL, Collins FS, Jackson AU, Mohlke KL, Stringham HM, Valle TT, Willer CJ, Bergman RN, Morken MA, Doring A, Gieger C, Illig T, Meitinger T, Org E, Pfeufer A, Wichmann HE, Kathiresan S, Marrugat J, O’Donnell CJ, Schwartz SM, Siscovick DS, Subirana I, Freimer NB, Hartikainen AL, McCarthy MI, O’Reilly PF, Peltonen L, Pouta A, de Jong PE, Snieder H, van Gilst WH, Clarke R, Goel A, Hamsten A, Peden JF. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet. 2009;41:666–676. doi: 10.1038/ng.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, Dehghan A, Glazer NL, Morrison AC, Johnson AD, Aspelund T, Aulchenko Y, Lumley T, Kottgen A, Vasan RS, Rivadeneira F, Eiriksdottir G, Guo X, Arking DE, Mitchell GF, Mattace-Raso FU, Smith AV, Taylor K, Scharpf RB, Hwang SJ, Sijbrands EJ, Bis J, Harris TB, Ganesh SK, O’Donnell CJ, Hofman A, Rotter JI, Coresh J, Benjamin EJ, Uitterlinden AG, Heiss G, Fox CS, Witteman JC, Boerwinkle E, Wang TJ, Gudnason V, Larson MG, Chakravarti A, Psaty BM, van Duijn CM. Genome-wide association study of blood pressure and hypertension. Nat Genet. 2009;41:677–687. doi: 10.1038/ng.384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bhangale TR, Rieder MJ, Nickerson DA. Estimating coverage and power for genetic association studies using near-complete variation data. Nat Genet. 2008;40:841–843. doi: 10.1038/ng.180. [DOI] [PubMed] [Google Scholar]
  • 16.Frazer KA, Murray SS, Schork NJ, Topol EJ. Human genetic variation and its contribution to complex traits. Nat Rev Genet. 2009;10:241–251. doi: 10.1038/nrg2554. [DOI] [PubMed] [Google Scholar]
  • 17.Yang J, Benyamin B, McEvoy BP, Gordon S, Henders AK, Nyholt DR, Madden PA, Heath AC, Martin NG, Montgomery GW, Goddard ME, Visscher PM. Common SNPs explain a large proportion of the heritability for human height. Nat Genet. 2010;42:565–569. doi: 10.1038/ng.608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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]
  • 19.Levy D, Larson MG, Benjamin EJ, Newton-Cheh C, Wang TJ, Hwang SJ, Vasan RS, Mitchell GF. Framingham Heart Study 100K Project: genome-wide associations for blood pressure and arterial stiffness. BMC Med Genet. 2007;8 (Suppl 1):S3. doi: 10.1186/1471-2350-8-S1-S3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lynch AI, Boerwinkle E, Davis BR, Ford CE, Eckfeldt JH, Leiendecker-Foster C, Arnett DK. Pharmacogenetic association of the NPPA T2238C genetic variant with cardiovascular disease outcomes in patients with hypertension. JAMA. 2008;299:296–307. doi: 10.1001/jama.299.3.296. [DOI] [PubMed] [Google Scholar]
  • 21.Scaroni C, Biason A, Carpenè G, Opocher G, Mantero F. 17-α-Hydroxylase deficiency in three siblings: short- and long-term studies. J Endocrinol Invest. 1991;14:99–108. doi: 10.1007/BF03350278. [DOI] [PubMed] [Google Scholar]
  • 22.Newton-Cheh C, Larson MG, Vasan RS, Levy D, Bloch KD, Surti A, Guiducci C, Kathiresan S, Benjamin EJ, Struck J, Morgenthaler NG, Bergmann A, Blankenberg S, Kee F, Nilsson P, Yin X, Peltonen L, Vartiainen E, Salomaa V, Hirschhorn JN, Melander O, Wang TJ. Association of common variants in NPPA and NPPB with circulating natriuretic peptides and blood pressure. Nat Genet. 2009;41:348–353. doi: 10.1038/ng.328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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, Jönsson 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;41:342–347. doi: 10.1038/ng.323. [DOI] [PubMed] [Google Scholar]
  • 24.Soranzo N, Rendon A, Gieger C, Jones CI, Watkins NA, Menzel S, Döring A, Stephens J, Prokisch H, Erber W, Potter SC, Bray SL, Burns P, Jolley J, Falchi M, Kühnel B, Erdmann J, Schunkert H, Samani NJ, Illig T, Garner SF, Rankin A, Meisinger C, Bradley JR, Thein SL, Goodall AH, Spector TD, Deloukas P, Ouwehand WH. A novel variant on chromosome 7q22.3 associated with mean platelet volume, counts, and function. Blood. 2009;113:3831–3837. doi: 10.1182/blood-2008-10-184234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Todd JA, Walker NM, Cooper JD, Smyth DJ, Downes K, Plagnol V, Bailey R, Nejentsev S, Field SF, Payne F, Lowe CE, Szeszko JS, Hafler JP, Zeitels L, Yang JH, Vella A, Nutland S, Stevens HE, Schuilenburg H, Coleman G, Maisuria M, Meadows W, Smink LJ, Healy B, Burren OS, Lam AA, Ovington NR, Allen J, Adlem E, Leung HT, Wallace C, Howson JM, Guja C, Ionescu-Tîrgoviste C, Simmonds MJ, Heward JM, Gough SC, Dunger DB, Wicker LS, Clayton DG. Genetics of Type 1 Diabetes in Finland; Wellcome Trust Case Control Consortium. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet. 2007;39:857–864. doi: 10.1038/ng2068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hunt KA, Zhernakova A, Turner G, Heap GA, Franke L, Bruinenberg M, Romanos J, Dinesen LC, Ryan AW, Panesar D, Gwilliam R, Takeuchi F, McLaren WM, Holmes GK, Howdle PD, Walters JR, Sanders DS, Playford RJ, Trynka G, Mulder CJ, Mearin ML, Verbeek WH, Trimble V, Stevens FM, O’Morain C, Kennedy NP, Kelleher D, Pennington DJ, Strachan DP, McArdle WL, Mein CA, Wapenaar MC, Deloukas P, McGinnis R, McManus R, Wijmenga C, van Heel DA. Newly identified genetic risk variants for celiac disease related to the immune response. Nat Genet. 2008;40:395–402. doi: 10.1038/ng.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Takaki S, Morita H, Tezuka Y, Takatsu K. Enhanced hematopoiesis by hematopoietic progenitor cells lacking intracellular adaptor protein, Lnk. J Exp Med. 2002;195:151–160. doi: 10.1084/jem.20011170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Velazquez L, Cheng AM, Fleming HE, Furlonger C, Vesely S, Bernstein A, Paige CJ, Pawson T. Cytokine signaling and hematopoietic homeostasis are disrupted in Lnk-deficient mice. J Exp Med. 2002;195:1599–1611. doi: 10.1084/jem.20011883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Zhernakova A, Elbers CC, Ferwerda B, Romanos J, Trynka G, Dubois PC, de Kovel CG, Franke L, Oosting M, Barisani D, Bardella MT, Joosten LA, Saavalainen P, van Heel DA, Catassi C, Netea MG, Wijmenga C. Finnish Celiac Disease Study Group. Evolutionary and functional analysis of celiac risk loci reveals SH2B3 as a protective factor against bacterial infection. Am J Hum Genet. 2010;86:970–977. doi: 10.1016/j.ajhg.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Takizawa H, Nishimura S, Takayama N, Oda A, Nishikii H, Morita Y, Kakinuma S, Yamazaki S, Okamura S, Tamura N, Goto S, Sawaguchi A, Manabe I, Takatsu K, Nakauchi H, Takaki S, Eto K. Lnk regulates integrin alphaIIbbeta3 outside-in signaling in mouse platelets, leading to stabilization of thrombus development in vivo. J Clin Invest. 2010;120:179–190. doi: 10.1172/JCI39503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kwon SM, Suzuki T, Kawamoto A, Ii M, Eguchi M, Akimaru H, Wada M, Matsumoto T, Masuda H, Nakagawa Y, Nishimura H, Kawai K, Takaki S, Asahara T. Pivotal role of lnk adaptor protein in endothelial progenitor cell biology for vascular regeneration. Circ Res. 2009;104:969–977. doi: 10.1161/CIRCRESAHA.108.192856. [DOI] [PubMed] [Google Scholar]
  • 32.Fitau J, Boulday G, Coulon F, Quillard T, Charreau B. The adaptor molecule Lnk negatively regulates tumor necrosis factor-α-dependent VCAM-1 expression in endothelial cells through inhibition of the ERK1 and -2 pathways. J Biol Chem. 2006;281:20148–20159. doi: 10.1074/jbc.M510997200. [DOI] [PubMed] [Google Scholar]
  • 33.Harrap SB, Stebbing M, Hopper JL, Hoang HN, Giles GG. Familial patterns of covariation for cardiovascular risk factors in adults: the Victorian Family Heart Study. Am J Epidemiol. 2000;152:704–715. doi: 10.1093/aje/152.8.704. [DOI] [PubMed] [Google Scholar]
  • 34.Reckelhoff JF. Gender differences in the regulation of blood pressure. Hypertension. 2001;37:1199–1208. doi: 10.1161/01.hyp.37.5.1199. [DOI] [PubMed] [Google Scholar]
  • 35.Dubey RK, Oparil S, Imthurn B, Jackson EK. Sex hormones and hypertension. Cardiovasc Res. 2002;53:688–708. doi: 10.1016/s0008-6363(01)00527-2. [DOI] [PubMed] [Google Scholar]
  • 36.Weiss LA, Pan L, Abney M, Ober C. The sex-specific genetic architecture of quantitative traits in humans. Nat Genet. 2006;38:218–222. doi: 10.1038/ng1726. [DOI] [PubMed] [Google Scholar]
  • 37.Franceschini N, MacCluer JW, Göring HH, Cole SA, Rose KM, Almasy L, Diego V, Laston S, Lee ET, Howard BV, Best LG, Fabsitz RR, Roman MJ, North KE. A quantitative trait loci-specific gene-by-sex interaction on systolic blood pressure among American Indians: the Strong Heart Family Study. Hypertension. 2006;48:266–270. doi: 10.1161/01.HYP.0000231651.91523.7e. [DOI] [PubMed] [Google Scholar]
  • 38.Franceschini N, Rose KM, Storti KL, Rutherford S, Voruganti VS, Laston S, Göring HH, Dyer TD, Umans JG, Lee ET, Best LG, Fabsitz RR, Cole SA, MacCluer JW, North KE. Social- and behavioral-specific genetic effects on blood pressure traits: the Strong Heart Family Study. Circ Cardiovasc Genet. 2009;2:396–401. doi: 10.1161/CIRCGENETICS.109.853630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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:e3583. doi: 10.1371/journal.pone.0003583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rosenberg NA, Huang L, Jewett EM, Szpiech ZA, Jankovic I, Boehnke M. Genome-wide association studies in diverse populations. Nat Rev Genet. 2010;11:356–366. doi: 10.1038/nrg2760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Takeuchi F, Isono M, Katsuya T, Yamamoto K, Yokota M, Sugiyama T, Nabika T, Fujioka A, Ohnaka K, Asano H, Yamori Y, Yamaguchi S, Kobayashi S, Takayanagi R, Ogihara T, Kato N. Blood pressure and hypertension are associated with 7 loci in the Japanese population. Circulation. 2010;121:2302–2309. doi: 10.1161/CIRCULATIONAHA.109.904664. [DOI] [PubMed] [Google Scholar]
  • 42.Cho YS, Go MJ, Kim YJ, Heo JY, Oh JH, Ban HJ, Yoon D, Lee MH, Kim DJ, Park M, Cha SH, Kim JW, Han BG, Min H, Ahn Y, Park MS, Han HR, Jang HY, Cho EY, Lee JE, Cho NH, Shin C, Park T, Park JW, Lee JK, Cardon L, Clarke G, McCarthy MI, Lee JY, Lee JK, Oh B, Kim HL. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat Genet. 2009;41:527–534. doi: 10.1038/ng.357. [DOI] [PubMed] [Google Scholar]
  • 43.Adeyemo A, Gerry N, Chen G, Herbert A, Doumatey A, Huang H, Zhou J, Lashley K, Chen Y, Christman M, Rotimi C. A genome-wide association study of hypertension and blood pressure in African Americans. PLoS Genet. 2009;5:e1000564. doi: 10.1371/journal.pgen.1000564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Musunuru K, Lettre G, Young T, Farlow DN, Pirruccello JP, Ejebe KG, Keating BJ, Yang Q, Chen MH, Lapchyk N, Crenshaw A, Ziaugra L, Rachupka A, Benjamin EJ, Cupples LA, Fornage M, Fox ER, Heckbert SR, Hirschhorn JN, Newton-Cheh C, Nizzari MM, Paltoo DN, Papanicolaou GJ, Patel SR, Psaty BM, Rader DJ, Redline S, Rich SS, Rotter JI, Taylor HA, Jr, Tracy RP, Vasan RS, Wilson JG, Kathiresan S, Fabsitz RR, Boerwinkle E, Gabriel SB. NHLBI Candidate Gene Association Resource. Candidate gene association resource (CARe): design, methods, and proof of concept. Circ Cardiovasc Genet. 2010;3:267–275. doi: 10.1161/CIRCGENETICS.109.882696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Deo RC, Patterson N, Tandon A, McDonald GJ, Haiman CA, Ardlie K, Henderson BE, Henderson SO, Reich D. A high-density admixture scan in 1,670 African Americans with hypertension. PLoS Genet. 2007;3:e196. doi: 10.1371/journal.pgen.0030196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhu X, Luke A, Cooper RS, Quertermous T, Hanis C, Mosley T, Gu CC, Tang H, Rao DC, Risch N, Weder A. Admixture mapping for hypertension loci with genome-scan markers. Nat Genet. 2005;37:177–181. doi: 10.1038/ng1510. [DOI] [PubMed] [Google Scholar]
  • 47.Bodmer W, Bonilla C. Common and rare variants in multifactorial susceptibility to common diseases. Nat Genet. 2008;40:695–701. doi: 10.1038/ng.f.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Newhouse SJ, Wallace C, Dobson R, Mein C, Pembroke J, Farrall M, Clayton D, Brown M, Samani N, Dominiczak A, Connell JM, Webster J, Lathrop GM, Caulfield M, Munroe PB. Haplotypes of the WNK1 gene associate with blood pressure variation in a severely hypertensive population from the British Genetics of Hypertension study. Hum Mol Genet. 2005;14:1805–1814. doi: 10.1093/hmg/ddi187. [DOI] [PubMed] [Google Scholar]
  • 49.Ji W, Foo JN, O’Roak BJ, Zhao H, Larson MG, Simon DB, Newton-Cheh C, State MW, Levy D, Lifton RP. Rare independent mutations in renal salt handling genes contribute to blood pressure variation. Nat Genet. 2008;40:592–599. doi: 10.1038/ng.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Feng T, Zhu X. Genome-wide searching of rare genetic variants in WTCCC data. Hum Genet. 2010;128:269–280. doi: 10.1007/s00439-010-0849-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol. 2008;26:1135–1145. doi: 10.1038/nbt1486. [DOI] [PubMed] [Google Scholar]
  • 52.Ng PC, Kirkness EF. Whole genome sequencing. Methods Mol Biol. 2010;628:215–226. doi: 10.1007/978-1-60327-367-1_12. [DOI] [PubMed] [Google Scholar]
  • 53.Conrad DF, Hurles ME. The population genetics of structural variation. Nat Genet. 2007;39:S30–S36. doi: 10.1038/ng2042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Conrad DF, Pinto D, Redon R, Feuk L, Gokcumen O, Zhang Y, Aerts J, Andrews TD, Barnes C, Campbell P, Fitzgerald T, Hu M, Ihm CH, Kristiansson K, Macarthur DG, Macdonald JR, Onyiah I, Pang AW, Robson S, Stirrups K, Valsesia A, Walter K, Wei J, Tyler-Smith C, Carter NP, Lee C, Scherer SW, Hurles ME. Wellcome Trust Case Control Consortium. Origins and functional impact of copy number variation in the human genome. Nature. 2010;464:704–712. doi: 10.1038/nature08516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Craddock N, Hurles ME, Cardin N, Pearson RD, Plagnol V, Robson S, Vukcevic D, Barnes C, Conrad DF, Giannoulatou E, Holmes C, Marchini JL, Stirrups K, Tobin MD, Wain LV, Yau C, Aerts J, Ahmad T, Andrews TD, Arbury H, Attwood A, Auton A, Ball SG, Balmforth AJ, Barrett JC, Barroso I, Barton A, Bennett AJ, Bhaskar S, Blaszczyk K, Bowes J, Brand OJ, Braund PS, Bredin F, Breen G, Brown MJ, Bruce IN, Bull J, Burren OS, Burton J, Byrnes J, Caesar S, Clee CM, Coffey AJ, Connell JM, Cooper JD, Dominiczak AF, Downes K, Drummond HE, Dudakia D, Dunham A, Ebbs B, Eccles D, Edkins S, Edwards C, Elliot A, Emery P, Evans DM, Evans G, Eyre S, Farmer A, Ferrier IN, Feuk L, Fitzgerald T, Flynn E, Forbes A, Forty L, Franklyn JA, Freathy RM, Gibbs P, Gilbert P, Gokumen O, Gordon-Smith K, Gray E, Green E, Groves CJ, Grozeva D, Gwilliam R, Hall A, Hammond N, Hardy M, Harrison P, Hassanali N, Hebaishi H, Hines S, Hinks A, Hitman GA, Hocking L, Howard E, Howard P, Howson JM, Hughes D, Hunt S, Isaacs JD, Jain M, Jewell DP, Johnson T, Jolley JD, Jones IR, Jones LA, Kirov G, Langford CF, Lango-Allen H, Lathrop GM, Lee J, Lee KL, Lees C, Lewis K, Lindgren CM, Maisuria-Armer M, Maller J, Mansfield J, Martin P, Massey DC, McArdle WL, McGuffin P, McLay KE, Mentzer A, Mimmack ML, Morgan AE, Morris AP, Mowat C, Myers S, Newman W, Nimmo ER, O’Donovan MC, Onipinla A, Onyiah I, Ovington NR, Owen MJ, Palin K, Parnell K, Pernet D, Perry JR, Phillips A, Pinto D, Prescott NJ, Prokopenko I, Quail MA, Rafelt S, Rayner NW, Redon R, Reid DM, Renwick, Ring SM, Robertson N, Russell E, St Clair D, Sambrook JG, Sanderson JD, Schuilenburg H, Scott CE, Scott R, Seal S, Shaw-Hawkins S, Shields BM, Simmonds MJ, Smyth DJ, Somaskantharajah E, Spanova K, Steer S, Stephens J, Stevens HE, Stone MA, Su Z, Symmons DP, Thompson JR, Thomson W, Travers ME, Turnbull C, Valsesia A, Walker M, Walker NM, Wallace C, Warren-Perry M, Watkins NA, Webster J, Weedon MN, Wilson AG, Woodburn M, Wordsworth BP, Young AH, Zeggini E, Carter NP, Frayling TM, Lee C, McVean G, Munroe PB, Palotie A, Sawcer SJ, Scherer SW, Strachan DP, Tyler-Smith C, Brown MA, Burton PR, Caulfield MJ, Compston A, Farrall M, Gough SC, Hall AS, Hattersley AT, Hill AV, Mathew CG, Pembrey M, Satsangi J, Stratton MR, Worthington J, Deloukas P, Duncanson A, Kwiatkowski DP, McCarthy MI, Ouwehand W, Parkes M, Rahman N, Todd JA, Samani NJ, Donnelly P. Wellcome Trust Case Control Consortium. Genome-wide association study of CNVs in 16,000 cases of eight common diseases and 3,000 shared controls. Nature. 2010;464:713–720. doi: 10.1038/nature08979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pang AW, MacDonald JR, Pinto D, Wei J, Rafiq MA, Conrad DF, Park H, Hurles ME, Lee C, Venter JC, Kirkness EF, Levy S, Feuk L, Scherer SW. Towards a comprehensive structural variation map of an individual human genome. Genome Biol. 2010;11:R52. doi: 10.1186/gb-2010-11-5-r52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Coughlin SS. Invited commentary: genetic variants and individual- and societal-level risk factors. Am J Epidemiol. 2010;171:24–26. doi: 10.1093/aje/kwp379. [DOI] [PubMed] [Google Scholar]
  • 58.Hong KW, Jin HS, Lim JE, Kim S, Go MJ, Oh B. Recapitulation of two genomewide association studies on blood pressure and essential hypertension in the Korean population. J Hum Genet. 2010;55:336–341. doi: 10.1038/jhg.2010.31. [DOI] [PubMed] [Google Scholar]
  • 59.Design of the Women’s Health Initiative clinical trial and observational study. The Women’s Health Initiative Study Group. Control Clin Trials. 1998;19:61–109. doi: 10.1016/s0197-2456(97)00078-0. [DOI] [PubMed] [Google Scholar]
  • 60.Centers for Disease Control and Prevention. Daily dietary fat and total food-energy intakes—Third National Health and Nutrition Examination Survey, Phase 1, 1988–91. MMWR Morb Mortal Wkly Rep. 1994;43:116–117. 123–125. [PubMed] [Google Scholar]
  • 61.Kolonel LN, Henderson BE, Hankin JH, Nomura AM, Wilkens LR, Pike MC, Stram DO, Monroe KR, Earle ME, Nagamine FS. A multiethnic cohort in Hawaii and Los Angeles: baseline characteristics. Am J Epidemiol. 2000;151:346–357. doi: 10.1093/oxfordjournals.aje.a010213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.The Atherosclerosis Risk in Communities (ARIC) Study: design and objectives. The ARIC investigators. Am J Epidemiol. 1989;129:687–702. [PubMed] [Google Scholar]
  • 63.Hughes GH, Cutter G, Donahue R, Friedman GD, Hulley S, Hunkeler E, Jacobs DR, Jr, Liu K, Orden S, Pirie P. Recruitment in the Coronary Artery Disease Risk Development in Young Adults (Cardia) Study. Control Clin Trials. 1987;8:68S–73S. doi: 10.1016/0197-2456(87)90008-0. [DOI] [PubMed] [Google Scholar]
  • 64.Fried LP, Borhani NO, Enright P, Furberg CD, Gardin JM, Kronmal RA, Kuller LH, Manolio TA, Mittelmark MB, Newman A. The Cardiovascular Health Study: design and rationale. Ann Epidemiol. 1991;1:263–276. doi: 10.1016/1047-2797(91)90005-w. [DOI] [PubMed] [Google Scholar]
  • 65.Lee ET, Welty TK, Fabsitz R, Cowan LD, Le NA, Oopik AJ, Cucchiara AJ, Savage PJ, Howard BV. The Strong Heart Study. A study of cardiovascular disease in American Indians: design and methods. Am J Epidemiol. 1990;132:1141–1155. doi: 10.1093/oxfordjournals.aje.a115757. [DOI] [PubMed] [Google Scholar]
  • 66.North KE, Howard BV, Welty TK, Best LG, Lee ET, Yeh JL, Fabsitz RR, Roman MJ, MacCluer JW. Genetic and environmental contributions to cardiovascular disease risk in American Indians: the strong heart family study. Am J Epidemiol. 2003;157:303–314. doi: 10.1093/aje/kwf208. [DOI] [PubMed] [Google Scholar]
  • 67.Rothwell PM. Limitations of the usual blood-pressure hypothesis and importance of variability, instability, and episodic hypertension. Lancet. 2010;375:938–948. doi: 10.1016/S0140-6736(10)60309-1. [DOI] [PubMed] [Google Scholar]
  • 68.Alikhani-Koopaei R, Fouladkou F, Frey FJ, Frey BM. Epigenetic regulation of 11 β-hydroxysteroid dehydrogenase type 2 expression. J Clin Invest. 2004;114:1146–1157. doi: 10.1172/JCI21647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Org E, Eyheramendy S, Juhanson P, Gieger C, Lichtner P, Klopp N, Veldre G, Döring A, Viigimaa M, Sõber S, Tomberg K, Eckstein G, Kelgo P, Rebane T, Shaw-Hawkins S, Howard P, Onipinla A, Dobson RJ, Newhouse SJ, Brown M, Dominiczak A, Connell J, Samani N, Farrall M, Caulfield MJ, Munroe PB, Illig T, Wichmann HE, Meitinger T, Laan M. KORA; BRIGHT. Genome-wide scan identifies CDH13 as a novel susceptibility locus contributing to blood pressure determination in two European populations. Hum Mol Genet. 2009;18:2288–2296. doi: 10.1093/hmg/ddp135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang Y, O’Connell JR, McArdle PF, Wade JB, Dorff SE, Shah SJ, Shi X, Pan L, Rampersaud E, Shen H, Kim JD, Subramanya AR, Steinle NI, Parsa A, Ober CC, Welling PA, Chakravarti A, Weder AB, Cooper RS, Mitchell BD, Shuldiner AR, Chang YP. From the cover: whole-genome association study identifies STK39 as a hypertension susceptibility gene. Proc Natl Acad Sci USA. 2009;106:226–231. doi: 10.1073/pnas.0808358106. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES