Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Sep 1.
Published in final edited form as: Curr Opin Cardiol. 2010 May;25(3):229–237. doi: 10.1097/HCO.0b013e3283383e2c

Blood pressure and human genetic variation in the general population

Pankaj Arora 1,2,3, Christopher Newton-Cheh 1,2,3
PMCID: PMC2947583  NIHMSID: NIHMS203472  PMID: 20224392

Abstract

Purpose of review

Hypertension is a complex trait with multiple environmental and genetic contributors. Linkage studies of rare Mendelian disorders of hypertension and hypotension have produced the most notable progress toward understanding the heritable basis of blood pressure. Association studies to identify common variants have been limited in the past by small sample sizes and most findings lack replication.

Recent Findings

Recently, well-powered, targeted candidate gene and genome-wide association studies have reported reproducible associations between common genetic variants and blood pressure.

Summary

Identification of novel blood pressure genes will lead to an improved understanding of blood pressure regulation and the potential for novel therapies.

Keywords: Blood pressure, genetics, association

Introduction

Elevated blood pressure (BP) or hypertension (HTN) increases the risk for stroke, congestive heart failure, coronary heart disease, and end-stage renal disease [13]. Lifestyle changes and antihypertensive therapy are associated with reductions in morbidity and mortality [46]. However, current population surveys indicate that control rates are generally poor [7]. An improved understanding of the environmental and genetic determinants of BP variation could point to novel avenues to prevent HTN and its complications.

The genetic basis of HTN at the population level has only recently begun to yield to intense investigation. Prior Current Opinion in Cardiology reviews from Binder et al. in 2007 and Hamet et al. in 2007 covered genome-wide linkage approaches in family- and population-based studies and early candidate gene studies. The current review focuses on genetic discoveries in 2008 and 2009.

Population burden of hypertension

More than a quarter of the world’s adult population is hypertensive [8]. In North America roughly 1 in 3 adults have HTN. Most individuals eventually become hypertensive if they live long enough, with a lifetime risk of ~90% in individuals who are normotensive at age 55 [9]. Despite the high prevalence, almost one third of all hypertensive individuals are unaware, half untreated, and three fourths uncontrolled [3].

Hypertension is a dichotomous trait, but BP shows a continuous relationship to outcomes. A meta analysis including one million adults found increasing risk of cardiovascular mortality with increasing BP starting as low as 115/75 mm Hg [1]. The Framingham Heart Study found high-normal BP (SBP 130 to 139 mm Hg, DBP 85 to 89 mm Hg) to be associated with increased cardiovascular risk [10]. These observations are consistent with a graded and continuous relationship of increasing BP with risk of cardiovascular disease.

Observational studies have reported the association of elevated BP with higher salt intake [11], excessive alcohol consumption [12], higher BMI [13] and reduced physical activity [14]. Interventional trials have established that modification of each of these risk factors lowers BP effectively, in some cases in excess of the BP-lowering effect of single-medication therapy [35,15,16].

Genetic factors influence BP and hypertension

Intra-individual BP variation, measurement error, strong environmental contributions, and anti-hypertensive therapy of varying intensity pose challenges to the genetic study of BP. Despite a large number of covarying factors, family studies have shown significant genetic contributions to inter-individual differences in BP [1720]. Long-term BP estimates have been shown to be highly heritable, with 50–60% of long-term systolic or diastolic BP (SBP, DBP) attributable to additive genetic factors [20].

The genetic architecture—a function of the number of variants as well as their frequency and strength of effect—that underlies this heritability has been unknown. Genome-wide linkage studies in rare families with Mendelian forms of HTN and hypotension successfully identified rare amino acid-altering variants that have relatively strong effects on salt-handling genes (Table 1). These have been well reviewed elsewhere [21]. While these rare variants clearly play a major role in BP variation within the families that harbor them, what role they play in overall BP variation has been unclear. Studies in the past two years have added importantly to our understanding of this genetic architecture.

Table 1. Mendelian syndromes of hypertension and hypotension.

Shown below are the genes that are well described to harbor rare mutations that cause Mendelian forms of hypertension and hypotension. Different mutations in some of the same genes (*) can lead to both hyper- or hypo-tension depending on whether they result in gain or loss of protein function.

gene gene name (alias) disease gain(+) or
loss(−) of
function
refs
Hypertension
CYP11B1 cytochrome P450, family 11, subfamily B, polypeptide 1 11 β hydroxylase deficiency [22]
CYP11B2* cytochrome P450, family 11, subfamily B, polypeptide 2
(aldosterone synthase)
glucocorticoid remediable aldosteronism + [23]
CYP17A1 cytochrome P450, family 17, subfamily A, polypeptide 1 combined 17α-hydroxylase/17,20-lyase
deficiency
[24,25]
CYP17A1 cytochrome P450, family 17, subfamily A, polypeptide 1 isolated 17,20-lyase deficiency [26]
HSD11B2 hydroxysteroid (11-beta) dehydrogenase 2 apparent mineralocorticoid excess [27,28]
NR3C2* mineralocorticoid receptor (MCR) mineralocorticoid receptor mutation + [29]
SCNN1B*
SCNN1G*
sodium channel, nonvoltage-gated 1 beta and gamma
subunits
Liddle’s Syndrome + [30
32]
WNK1 WNK lysine deficient protein kinase 1 pseudohypoaldosteronism type II (Gordon
Syndrome)
+ [33]
WNK4 WNK lysine deficient protein kinase 4 pseudohypoaldosteronism type II (Gordon
Syndrome)
−/+ [33
35]
Hypotension
BSND Bartter syndrome, infantile, with sensorineural deafness Bartter’s syndrome Type 4 [36]
CASR calcium-sensing receptor Bartter’s syndrome Type 5 + [37,38]
CLCNKB chloride channel Kb Bartter’s syndrome Type 3 [39]
CYP11B2* cytochrome P450, family 11, subfamily B, polypeptide 2 corticosterone methyloxidase type I and II
deficiency
[40,41]
CYP21A2 cytochrome P450, family 21, subfamily A, polypeptide 2 21-hydroxylase deficiency [42,43]
HSD3B2 hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid
delta-isomerase 2
3 β hydroxysteroid dehydrogenase
deficiency type II
[44]
KCNJ1 potassium inwardly-rectifying channel, subfamily J, member
1 (ROMK)
Bartter’s syndrome Type 2 [45]
NR3C2* mineralocorticoid receptor pseudohypoaldosteronism type I [46]
SCNN1A
SCNN1B*
SCNN1G*
sodium channel, nonvoltage-gated 1 alpha, beta and gamma
subunits
pseudohypoaldosteronism type I [47]
SLC12A1 solute carrier family 12 member 1 (Na-K-2Cl cotransporter,
NKCC2, loop diuretic target)
Bartter’s syndrome Type 1 [48]
SLC12A3 solute carrier family 12 member 3 (Na-Cl cotransporter,
NCCT, thiazide diuretic target)
Gitelman’s syndrome [49]
*

gene associated with both hypertension and hypotension

Do rare variants at the population level influence BP?

Richard Lifton’s group and others have identified many disease-causing mutations in renal salt-handling genes, placing these genes and pathways centrally in BP regulation (Table 1). Whether genetic variation in these genes at the population level contributes to inter-individual differences in BP was an open question. It is possible that rare mutations in a gene lead to a detectable phenotype in a handful of families, but that low prevalence (due to negative selection) makes variation in these genes a negligible contributor to differences in BP in general. A series of reports on a gene responsible for Gitelman’s syndrome, an autosomal recessive syndrome characterized by hyplochloremic metabolic alkalosis, hyopokalemia and hypocalciuria, have expanded our understanding of the genetic architecture of BP.

Homozygous mutations in families

In a linkage study in 12 families, Simon et al. mapped Gitelman’s syndrome to the SLC12A3 gene encoding the thiazide-sensitive Na-Cl cotransporter (NCCT) [49]. They identified homozygous loss-of-function mutations in SLC12A3 that result in salt wasting from the distal convoluted tubule. The authors speculated that heterozygous carriers of mutant alleles (one mutated copy, one normal copy of the gene) might be protected against development of HTN.

Heterozygous mutations in a large family with Gitelman’s syndrome

Cruz et al. examined an Amish kindred with Gitelman’s syndrome, including 60 individuals with zero, 113 with one and 26 with two copies of the mutant allele [50]. Blood pressure was significantly lower in heterozygous children but not in adults, perhaps due to limited power, age effects or improved dietary compensation in adults. These observations in a single family raised the intriguing possibility that heterozygous mutations in renal salt-handling genes at the population level might affect BP, not just in families selected for Gitelman’s syndrome.

Heterozygous mutations in the general population

In 2008, Ji et al. reported a screen in 3,125 individuals (many in families) from the community-based Framingham Heart Study for mutations in 3 candidate genes in which homozygous mutations cause Gitelman’s and Bartter’s syndromes: NCCT (SLC12A3); NKCC2 (SLC12A1); and ROMK (KCNJ1) (Table 1) [51] ••. Using criteria of complete conservation across multi-species comparisons with rare allele frequency (<1 per 1,000) or known Gitelman’s or Bartter’s mutations, they identified a final set of 30 putative functional mutations (15 in SLC12A3, 10 in SLC12A1 and 5 in KCNJ1) found in 49 subjects. 80% of the carriers of at least one mutation had long-term SBP values below the mean of the entire cohort (P=0.001). Carriers had 6.3 mm Hg lower SBP (P=0.0009) and 3.4 mm Hg lower DBP (P=0.0003) compared to non-carriers. The BP reduction among mutation carriers was observed in all age groups, with a ~60% reduction in the risk of developing HTN by age 60 (P<0.0003).

Collectively, these studies chart the identification of a gene influencing BP through linkage in rare families with recessive hypotension syndromes, use of large kindreds to examine within-family effects of heterozygosity and ultimately extension to the population level. Along with the finding that rare mutations in ABCA1 result in marked reduction in high-density lipoprotein levels in the general population [52], these are the first demonstrations that rare mutations of strong effect contribute to variation in common cardiovascular traits. Whether similar unbiased (cf genome-wide linkage) or targeted (cf sequencing of candidate genes) approaches could identify common variants that contribute to BP variation at the population level was unclear until recently. The literature is full of reports of small numbers of variants in modest-sized studies that failed to be replicated and led to a general sense that association studies of common variants were hopeless.

Common variants at the population level

Linkage studies are well suited to detect rare variants with strong effects that are conclusively scored in individual families for conditions that are relatively rare but has been an utter failure for common complex diseases [53]. By contrast, association studies are better suited to detect modest genetic effects of common variants [54]. Association studies were plagued by poor reproducibility, stemming from false positive reports due to inappropriately permissively significance thresholds and the inability to validate suggestive findings in underpowered replication attempts [55]. The creation of a reference sequence of the human genome, generation of large databases of common genetic variation, growth of high-throughput genotyping platforms and development of rigorous methodological approaches set the stage for the discoveries that followed.

First validated common BP variants identified

Atrial and B-type natriuretic peptides (ANP, BNP) have direct vasodilatory and natriuretic effects and antagonize the renin-angiotensin-aldosterone system [5658]. In mouse models, knockout of the NPPA (encoding ANP) or natriuretic peptide receptor A (NPR-A) genes [59,60] result in salt-sensitive HTN and ANP overexpression [61] lowers BP. The known physiologic effects in humans and animal models make the natriuretic peptide system a compelling candidate to influence interindividual differences in BP, but until recently there was no clear evidence that ANP or BNP influence BP regulation in humans.

In 2009, Newton-Cheh et al. reported a candidate gene association study at the locus on chromosome 1 where NPPA and NPPB lie in tandem (Table 2) [62] ••. Thirteen single nucleotide polymorphisms (SNPs) were genotyped in a discovery sample from the Framingham Heart Study (n=1,705) and tested for association with plasma ANP and BNP, with subsequent replication in samples from Sweden and Finland (total n=14,743). Three non-coding SNPs showed consistent association with ANP or BNP at significance levels that were unambiguous (P ranging from 10−10 to 10−70), presumably through shared regulatory effects given the concordant effects on both peptides.

Table 2. Common variants implicated in recent candidate gene and GWAS studies with p<5×10−8.

Fourteen variants at 13 independent loci are shown; the first two variants are at the same locus but are incompletely correlated and may represent different signals of association. P-values for variants observed in more than one study (or a close proxy) are based on meta-analysis of reported studies.

Nearby genes SNP Chr Allele
frequency
Trait Effect
(mm Hg)
p-value Reference
NPPA/NPPB rs5068 1 0.94 SBP 1.10 3×10−9 [62,63]
MTHFR/CLCN6/NPPA/NPPB rs17367504 1 0.86 SBP 0.85 2×10−13 [63]
CYP17A1 rs11191548 10 0.91 SBP 1.16 7×10−24 [63,64]
PLCD3 rs12946454 17 0.28 SBP 0.57 1×10−8 [63]
FGF5 rs16998073 4 0.21 DBP 0.50 1×10−21 [63]
c10orf107 rs1530440 10 0.81 DBP 0.39 1×10−9 [63]
SH2B3 rs3184504 12 0.47 DBP 0.46 3×10−18 [63,64]
CYP1A2/CSK/LMAN1L rs1378942 15 0.36 DBP 0.43 1×10−23 [63,64]
ZNF652 rs16948048 17 0.39 DBP 0.31 5×10−9 [63]
PLEKHA7 rs381815 11 0.26 SBP 0.65 2×10−9 [64]
ATP2B1 rs2681492 12 0.80 SBP 0.85 6×10−17 [64,65]*
ULK4 rs9815354 3 0.17 DBP 0.49 3×10−9 [64]
CACNB2 rs11014166 10 0.66 DBP 0.37 1×10−8 [64]
TBX3/TBX5 rs2384550 12 0.65 DBP 0.35 4×10−8 [64]
*

A correlated SNP was reported in Cho et al just missed genome-wide significant association with SBP (P=1×10−7).

With the addition of further samples from Sweden (final n=29,717), we found that two alleles associated with higher ANP and BNP were also associated with lower SBP (0.9–1.5 mm Hg) and DBP (0.3–0.8 mm Hg) and decreased odds of HTN (0.85–0.90). BP associations had much more modest significance than those of natriuretic peptide concentrations (P ranging from 1×10−6 to 6×10−5) [62]. The identification of cis-acting non-coding common genetic variants that influence plasma concentrations of ANP and BNP as well as BP provided the first in vivo evidence in humans of the relevance of endogenous variation in the natriuretic peptide system in BP regulation in humans.

It is worth pointing out that these weak BP effects could not be detected in such a small discovery sample size (n=1,705) were it not for the use intermediate ANP/BNP traits which showed much stronger association. Unbiased screens of the genome for BP variants, requiring stringent p-value thresholds to distinguish false- from true-positive associations (e.g. P<5×10−8) would require even larger sample sizes to detect such an effect.

Candidate gene studies are like the drunk searching under the lamppost for his keys—one can only look where biologic involvement is already suspected—and thus do not expand our understanding of human biology as much as unbiased surveys that test thousands of genes comprehensively. Genome-wide association studies (GWAS) have only borne fruit for BP when very large sample sizes allowed stringent P-value thresholds to be met.

GWAS in small to moderately large studies do not reach genome-wide significance

The first few published GWA studies published by us and others in 2007, including the Wellcome Trust Case Control Consortium (WTCCC, 2000 hypertensive cases, 3000 population-based controls) [66], the Diabetics Genetics Initiative (DGI, 1,464 cases with type 2 diabetes and 1467 matched controls) [67] and the Framingham Heart Study 100K project (1,327 individuals for SBP and DBP) [68] failed to identify any loci meeting stringent genome-wide significance thresholds. Genome-wide significance has been variably defined but is generally accepted at <5×10−8, which represents Bonferroni correction for the estimated 1 million independent common variant tests in the genome for individuals of European ancestry (0.05/1,000,000) [69]. In 2009, further GWA studies of BP traits and/or HTN in populations of different ancestries and of moderate size have subsequently been reported but none achieved genome-wide significance [65,7074]. A few are highlighted.

In 2009, Wang et al. [72] reported a GWAS in 542 Amish subjects and found a SNP at 2q24.3 with modest association with SBP (P=8×10−5). They genotyped an additional 1,347 individuals and looked up results from published GWA studies for a total sample size of 7,125, but did not reach genome-wide significance (P=2×10−7). The SNP lies within an intron of the serine threonine kinase 39 (STK39) gene which encodes the SPAK protein, known to regulate NCCT and NKCC2 in the kidney [75,76]. No replication of this association has been published.

In 2009, Cho et al. [65]• reported a GWAS from 2 population-based cohorts of 8,842 individuals with replication in 7,861 individuals all of Korean ancestry. SNP rs17249754 near ATP2B1 was associated with SBP with borderline significance (P=1×10−7). This SNP is located near the ATPase calcium ion transporting plasma membrane 1 (ATP2B1) gene, which encodes a protein involved in calcium movement from cytosol to the extracellular compartment [77]. A correlated SNP at ATP2B1 was subsequently reported to be associated with SBP and HTN in a GWAS in European samples (see below).

Adeyemo et al. reported the first GWAS for BP and HTN in an African-American sample n=1,017 individuals, some in families) [74]. The authors selected 17 SNPs for follow-up replication in 980 West Africans. Of these, none were genome-wide significant upon joint analysis with the original GWAS result (all P>3×10−6).

Large GWAS meta-analyses and replication identify 13 novel loci

In 2009, two meta-analyses of genome-wide association studies of BP, Global BPgen [63]•• and CHARGE BP [64] ••, reported in parallel the first genome-wide significant associations between common genetic variants and BP.

Global BPgen consortium identifies 8 BP loci

Newton-Cheh et al. reported findings of the Global BPgen consortium including a stage 1 GWAS meta-analysis in 34,433 individuals from 17 cohorts of European ancestry ascertained through population-based sampling and controls from case-control collections. In total, 2.5 million genotyped and imputed SNPs were tested for association with SBP and DBP under an additive genetic model, adjusting for age, age2, sex, body mass index, with imputation in individuals treated with anti-hypertensives. Twelve independent SNPs were selected from the stage 1 GWAS meta-analysis and directly genotyped in up to 71,225 European-derived and 12,889 Asian ancestry individuals (stage 2a). Twenty SNPs associated with SBP or DBP were looked up in the CHARGE BP results (stage 2b).

Eight loci met genome-wide significance for association with BP (3 SBP, 5 DBP) on meta-analysis of stage 1+2a+2b (P range 10−23 to 10−8, n≤134,258, Table 2) [63]. Effect sizes were quite modest, generally ≤ ≈1 mm Hg systolic or ≈0.5 mm Hg diastolic. All variants were associated with both SBP and DBP, even if ascertained on association with one trait, and all were associated with dichotomous HTN. Of the 8 loci, two harbor genes known to be involved in BP physiology and six do not.

Two loci contain known BP genes

The MTHFR/CLCN6/NPPA/NPPB locus included a SNP rs17367504 associated with BP (Table 2). The SNP is partially correlated to the top SNP from our prior candidate gene study and is associated with plasma ANP concentration making it likely to mediate its effect through regulation of the natriuretic peptide system. However, rs17367504 is also strongly associated with hepatic expression of CLCN6 [78], which encodes a renally-expressed chloride channel, suggesting that a simple SNP→ANP→BP model may not hold. The Global BPgen study did confirm the association of the rs5068 SNP from the candidate gene study with SBP (joint P=3×10−9, Table 2) [62,63]. Whether multiple independent variants at the locus exist, potentially acting through different mechanisms, will require much more work.

One locus contains a gene involved in Mendelian HTN, CYP17A1 (Table 1, Table 2). CYP17A1 encodes a protein with both 17α-hydroxylase activity—mediating a key step in the biosynthesis of glucocorticoids—and 17,20-lyase activity involved in sex steroidogenesis [26,79]. Missense mutations in CYP17A1 cause adrenal hyperplasia with HTN and hypokalemia. There are many other genes in the associated interval at this locus, but the well-annotated Mendelian HTN gene certainly stands out as a strong candidate to underlie the common variant association.

Thus, strong candidate genes at these loci appear to be good places to start to explain these common variant associations. But ultimately, it will be necessary to determine the causal variants and to elucidate their relationships to gene expression for regulatory noncoding variants or to gene function via altered proteins for coding or splice variants. Unfortunately, the genetic code for regulatory machinery is only beginning to be understood and smoking-gun amino-acid altering variants are a minority of common variant associations. Of the remaining 6 loci identified by Global BPgen, the 12q24 locus includes an amino-acid altering variant.

A missense SNP in SH2B3 is associated with multiple traits

The missense SNP rs3184504 in the SH2B3 (SH2B adaptor protein 3) gene codes for an arginine to tryptophan at amino acid 262 (R262W) in the lymphocyte-specific adapter protein (LNK). A relationship of this gene to BP physiology has not previously been recognized. In earlier GWA studies, the minor T allele (262W) of this SNP was found to be associated in humans with increased odds of autoimmune diseases including type 1 diabetes [80], celiac disease [81], myocardial infarction [82,83] as well as higher eosinophil and other blood cell counts [82,83] (Table 3). The mouse knockout of LNK is associated with higher eosinophil, lymphocyte and platelet counts, suggesting by extension that the 262W allele results in loss of function (Table 3). The increased risk of autoimmune disease seems consistent with a potentially augmented immunologic activity. In fact, strong signatures of positive selection on the 262W allele in European-derived individuals suggest a selective advantage in human history, perhaps through fighting infection.

Table 3. SH2B3 262W allele in humans mirrors the mouse knockout.

Shown are the effects in a mouse knockout and in humans for the minor 262W allele.

eosinophils lymphocyte platelets type 1
diabetes
risk
celiac
disease
risk
myocardial
infarction
diastolic
blood
pressure
selection
mouse
knockout
[85]
SH2B3
262W
allele

[82,83]

[82]

[82]

[80]

[81]

[82,83]

[63,64]
positive
[63,83]

The role of SH2B3 in BP regulation or myocardial infarction pathogenesis is less clear but could be due to an effect outside of the hematopoietic lineage. In an endothelial cell model, exposure to TNF-α has been found to upregulate LNK, inhibiting VCAM1 expression and ERK1/2 and increasing eNOS activity via the PI3K/AKT pathway [84]. Given the role of the endothelial cell in atherothrombosis and smooth muscle tone, it seems possible that loss of LNK function from the 262W allele could lead to increased BP and risk of myocardial infarction through altered endothelial function.

CHARGE BP consortium

In a parallel publication, the CHARGE BP consortium reported results using a sample consisting of 29,136 European-derived individuals [64]•• with validation of top results in the Global BPgen GWAS. CHARGE BP identified 8 genome-wide significant loci, including 3 in common with Global BPgen and 5 unique to CHARGE BP (Table 2). The 5 additional loci have not previously been annotated to harbor genes involved in BP regulation, with the exception of CACNB2 which encodes a beta subunit of voltage-gated calcium channels, including the L-type dihydropyridine-sensitive channel.

Conclusions and questions raised

Until recently only Mendelian families yielded robust genetic BP findings. Large sample sizes, comprehensive genetic screens and rigorous p-value thresholds have allowed human genetics to expose additional novel pathways involved in BP regulation through the study of unselected population samples. However, in total the common and rare variants identified to date that influence BP explain less than a percent or two of overall population variation in BP [51,63,64], far less than age or body mass index. It seems unlikely, but requires formal testing, that genetic testing will add clinical utility to predicting future HTN in patients, beyond that available from a scale, a BP cuff and their age.

Why are common BP variant effects so weak? BP regulation requires that organ perfusion be achieved through a very dynamic range of daily activities and environments, from sleeping to running through feast or famine. This has likely led to many redundant systems that allow tight regulation of BP and poor tolerance of genetic variants of strong effect that are unable to rise to high frequency due to negative selection.

Why are so few of the antihypertensive targets represented among the common BP variants identified to date? It is possible that evolution has been particularly intolerant of genetic variation in the targets of antihypertensive therapies. However, it is equally possible that hundreds of genes are involved in BP, and that antihypertensive targets represent a small fraction of the total number of BP genes.

What do the weak common variant effects tell us about the potential of novel targets to have large effects on BP control? Not much. The minor allele of the SNP rs3846663 has been associated with a trivial 2.5 mg/dL higher low-density lipoprotein (LDL) cholesterol in a genome-wide association study [86]. If one did not already know that HMG-CoA reductase is involved in LDL cholesterol synthesis, then finding this SNP in an intron of the HMGCR gene could allow identification of this target for cholesterol-lowering therapy.

Next steps

Even larger GWA studies are likely to identify additional common genetic variants of even weaker effects. The ongoing ICBP-GWAS consortium, including the Global BPgen and CHARGE BP consortia as well as others, is one such example, with >70,000 individuals with genome-wide genotyping. Sequencing will enable fine mapping of association signals and can potentially identify less common, stronger effect alleles (more “Mendelian”) that may be more easily recognized by alteration of amino acids and more tractable for physiologic study because of their greater effect sizes. High-resolution phenotyping of individuals selected on genotype can potentially home in on the mechanisms underlying association of a particular genetic variant, as has recently been applied to the IL2RA gene [87]. Identification of novel genetic influences on BP is just a start, as was identification of the first genes underlying familial hypercholesterolemia in the 1970s, with the promise of an improved understanding of BP regulation and identification of novel therapeutic targets.

Acknowledgements

Dr. Newton-Cheh is supported by NIH (K23-HL080025 and R01-HL098283), a Doris Duke Charitable Foundation Clinical Scientist Development Award, and a Burroughs Wellcome Fund Career Award for Medical Scientists.

Footnotes

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

References

  • 1.Lewington S, Clarke R, Qizilbash N, Peto R, Collins R. 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.Klag MJ, Whelton PK, Randall BL, Neaton JD, Brancati FL, Ford CE, Shulman NB, Stamler J. Blood pressure and end-stage renal disease in men. N Engl J Med. 1996;334:13–18. doi: 10.1056/NEJM199601043340103. [DOI] [PubMed] [Google Scholar]
  • 3.Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL, Jr, Jones DW, Materson BJ, Oparil S, Wright JT, Jr, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure: the JNC 7 report. JAMA. 2003;289:2560–2572. doi: 10.1001/jama.289.19.2560. [DOI] [PubMed] [Google Scholar]
  • 4.Sacks FM, Svetkey LP, Vollmer WM, Appel LJ, Bray GA, Harsha D, Obarzanek E, Conlin PR, Miller ER, 3rd, Simons-Morton DG, et al. Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. DASH-Sodium Collaborative Research Group. N Engl J Med. 2001;344:3–10. doi: 10.1056/NEJM200101043440101. [DOI] [PubMed] [Google Scholar]
  • 5.Whelton SP, Chin A, Xin X, He J. Effect of aerobic exercise on blood pressure: a meta-analysis of randomized, controlled trials. Ann Intern Med. 2002;136:493–503. doi: 10.7326/0003-4819-136-7-200204020-00006. [DOI] [PubMed] [Google Scholar]
  • 6.Neal B, MacMahon S, Chapman N. Effects of ACE inhibitors, calcium antagonists, and other blood-pressure-lowering drugs: results of prospectively designed overviews of randomised trials. Blood Pressure Lowering Treatment Trialists' Collaboration. Lancet. 2000;356:1955–1964. doi: 10.1016/s0140-6736(00)03307-9. [DOI] [PubMed] [Google Scholar]
  • 7.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]
  • 8.Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217–223. doi: 10.1016/S0140-6736(05)17741-1. [DOI] [PubMed] [Google Scholar]
  • 9.Vasan RS, Beiser A, Seshadri S, Larson MG, Kannel WB, D'Agostino RB, Levy D. Residual lifetime risk for developing hypertension in middle-aged women and men: The Framingham Heart Study. JAMA. 2002;287:1003–1010. doi: 10.1001/jama.287.8.1003. [DOI] [PubMed] [Google Scholar]
  • 10.Vasan RS, Larson MG, Leip EP, Evans JC, O'Donnell CJ, Kannel WB, Levy D. Impact of high-normal blood pressure on the risk of cardiovascular disease. N.Engl.J.Med. 2001;345:1291–1297. doi: 10.1056/NEJMoa003417. [DOI] [PubMed] [Google Scholar]
  • 11.Elliott P, Stamler J, Nichols R, Dyer AR, Stamler R, Kesteloot H, Marmot M. Intersalt revisited: further analyses of 24 hour sodium excretion and blood pressure within and across populations. Intersalt Cooperative Research Group. Bmj. 1996;312:1249–1253. doi: 10.1136/bmj.312.7041.1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Arkwright PD, Beilin LJ, Rouse I, Armstrong BK, Vandongen R. Effects of alcohol use and other aspects of lifestyle on blood pressure levels and prevalence of hypertension in a working population. Circulation. 1982;66:60–66. doi: 10.1161/01.cir.66.1.60. [DOI] [PubMed] [Google Scholar]
  • 13.Field AE, Coakley EH, Must A, Spadano JL, Laird N, Dietz WH, Rimm E, Colditz GA. Impact of overweight on the risk of developing common chronic diseases during a 10-year period. Arch Intern Med. 2001;161:1581–1586. doi: 10.1001/archinte.161.13.1581. [DOI] [PubMed] [Google Scholar]
  • 14.Blair SN, Goodyear NN, Gibbons LW, Cooper KH. Physical fitness and incidence of hypertension in healthy normotensive men and women. JAMA. 1984;252:487–490. [PubMed] [Google Scholar]
  • 15.Effects of weight loss and sodium reduction intervention on blood pressure and hypertension incidence in overweight people with high-normal blood pressure. The Trials of Hypertension Prevention, phase II. The Trials of Hypertension Prevention Collaborative Research Group. Arch Intern Med. 1997;157:657–667. [PubMed] [Google Scholar]
  • 16.Xin X, He J, Frontini MG, Ogden LG, Motsamai OI, Whelton PK. Effects of alcohol reduction on blood pressure: a meta-analysis of randomized controlled trials. Hypertension. 2001;38:1112–1117. doi: 10.1161/hy1101.093424. [DOI] [PubMed] [Google Scholar]
  • 17.Snieder H, Harshfield GA, Treiber FA. Heritability of blood pressure and hemodynamics in African- and European-American youth. Hypertension. 2003;41:1196–1201. doi: 10.1161/01.HYP.0000072269.19820.0D. [DOI] [PubMed] [Google Scholar]
  • 18.Hottenga JJ, Boomsma DI, Kupper N, Posthuma D, Snieder H, Willemsen G, de Geus EJ. Heritability and stability of resting blood pressure. Twin Res Hum Genet. 2005;8:499–508. doi: 10.1375/183242705774310123. [DOI] [PubMed] [Google Scholar]
  • 19.Kupper N, Willemsen G, Riese H, Posthuma D, Boomsma DI, de Geus EJ. Heritability of daytime ambulatory blood pressure in an extended twin design. Hypertension. 2005;45:80–85. doi: 10.1161/01.HYP.0000149952.84391.54. [DOI] [PubMed] [Google Scholar]
  • 20.Levy D, DeStefano AL, Larson MG, O'Donnell CJ, Lifton RP, Gavras H, Cupples LA, Myers RH. Evidence for a gene influencing blood pressure on chromosome 17. Genome scan linkage results for longitudinal blood pressure phenotypes in subjects from the framingham heart study. Hypertension. 2000;36:477–483. doi: 10.1161/01.hyp.36.4.477. [DOI] [PubMed] [Google Scholar]
  • 21.Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hypertension. Cell. 2001;104:545–556. doi: 10.1016/s0092-8674(01)00241-0. [DOI] [PubMed] [Google Scholar]
  • 22.White PC, Dupont J, New MI, Leiberman E, Hochberg Z, Rosler A. A mutation in CYP11B1 (Arg-448----His) associated with steroid 11 beta-hydroxylase deficiency in Jews of Moroccan origin. J Clin Invest. 1991;87:1664–1667. doi: 10.1172/JCI115182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lifton RP, Dluhy RG, Powers M, Rich GM, Cook S, Ulick S, Lalouel JM. A chimaeric 11 beta-hydroxylase/aldosterone synthase gene causes glucocorticoid-remediable aldosteronism and human hypertension. Nature. 1992;355:262–265. doi: 10.1038/355262a0. [DOI] [PubMed] [Google Scholar]
  • 24.Kagimoto M, Winter JS, Kagimoto K, Simpson ER, Waterman MR. Structural characterization of normal and mutant human steroid 17 alpha-hydroxylase genes: molecular basis of one example of combined 17 alpha-hydroxylase/17,20 lyase deficiency. Mol Endocrinol. 1988;2:564–570. doi: 10.1210/mend-2-6-564. [DOI] [PubMed] [Google Scholar]
  • 25.Rosa S, Duff C, Meyer M, Lang-Muritano M, Balercia G, Boscaro M, Topaloglu AK, Mioni R, Fallo F, Zuliani L, et al. P450c17 deficiency: clinical and molecular characterization of six patients. J Clin Endocrinol Metab. 2007;92:1000–1007. doi: 10.1210/jc.2006-1486. [DOI] [PubMed] [Google Scholar]
  • 26.Geller DH, Auchus RJ, Mendonca BB, Miller WL. The genetic and functional basis of isolated 17,20-lyase deficiency. Nat Genet. 1997;17:201–205. doi: 10.1038/ng1097-201. [DOI] [PubMed] [Google Scholar]
  • 27.Mune T, Rogerson FM, Nikkila H, Agarwal AK, White PC. Human hypertension caused by mutations in the kidney isozyme of 11 beta-hydroxysteroid dehydrogenase. Nat Genet. 1995;10:394–399. doi: 10.1038/ng0895-394. [DOI] [PubMed] [Google Scholar]
  • 28.Wilson RC, Dave-Sharma S, Wei JQ, Obeyesekere VR, Li K, Ferrari P, Krozowski ZS, Shackleton CH, Bradlow L, Wiens T, et al. A genetic defect resulting in mild low-renin hypertension. Proc Natl Acad Sci U S A. 1998;95:10200–10205. doi: 10.1073/pnas.95.17.10200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Geller DS, Farhi A, Pinkerton N, Fradley M, Moritz M, Spitzer A, Meinke G, Tsai FT, Sigler PB, Lifton RP. Activating mineralocorticoid receptor mutation in hypertension exacerbated by pregnancy. Science. 2000;289:119–123. doi: 10.1126/science.289.5476.119. [DOI] [PubMed] [Google Scholar]
  • 30.Shimkets RA, Warnock DG, Bositis CM, Nelson-Williams C, Hansson JH, Schambelan M, Gill JR, Jr, Ulick S, Milora RV, Findling JW, et al. Liddle's syndrome: heritable human hypertension caused by mutations in the beta subunit of the epithelial sodium channel. Cell. 1994;79:407–414. doi: 10.1016/0092-8674(94)90250-x. [DOI] [PubMed] [Google Scholar]
  • 31.Hansson JH, Nelson-Williams C, Suzuki H, Schild L, Shimkets R, Lu Y, Canessa C, Iwasaki T, Rossier B, Lifton RP. Hypertension caused by a truncated epithelial sodium channel gamma subunit: genetic heterogeneity of Liddle syndrome. Nat Genet. 1995;11:76–82. doi: 10.1038/ng0995-76. [DOI] [PubMed] [Google Scholar]
  • 32.Abriel H, Loffing J, Rebhun JF, Pratt JH, Schild L, Horisberger JD, Rotin D, Staub O. Defective regulation of the epithelial Na+ channel by Nedd4 in Liddle's syndrome. J Clin Invest. 1999;103:667–673. doi: 10.1172/JCI5713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wilson FH, Disse-Nicodeme S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, et al. Human hypertension caused by mutations in WNK kinases. Science. 2001;293:1107–1112. doi: 10.1126/science.1062844. [DOI] [PubMed] [Google Scholar]
  • 34.Wilson FH, Kahle KT, Sabath E, Lalioti MD, Rapson AK, Hoover RS, Hebert SC, Gamba G, Lifton RP. Molecular pathogenesis of inherited hypertension with hyperkalemia: the Na-Cl cotransporter is inhibited by wild-type but not mutant WNK4. Proc Natl Acad Sci U S A. 2003;100:680–684. doi: 10.1073/pnas.242735399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kahle KT, Wilson FH, Leng Q, Lalioti MD, O'Connell AD, Dong K, Rapson AK, MacGregor GG, Giebisch G, Hebert SC, et al. WNK4 regulates the balance between renal NaCl reabsorption and K+ secretion. Nat Genet. 2003;35:372–376. doi: 10.1038/ng1271. [DOI] [PubMed] [Google Scholar]
  • 36.Birkenhager R, Otto E, Schurmann MJ, Vollmer M, Ruf EM, Maier-Lutz I, Beekmann F, Fekete A, Omran H, Feldmann D, et al. Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet. 2001;29:310–314. doi: 10.1038/ng752. [DOI] [PubMed] [Google Scholar]
  • 37.Watanabe S, Fukumoto S, Chang H, Takeuchi Y, Hasegawa Y, Okazaki R, Chikatsu N, Fujita T. Association between activating mutations of calcium-sensing receptor and Bartter's syndrome. Lancet. 2002;360:692–694. doi: 10.1016/S0140-6736(02)09842-2. [DOI] [PubMed] [Google Scholar]
  • 38.Vargas-Poussou R, Huang C, Hulin P, Houillier P, Jeunemaitre X, Paillard M, Planelles G, Dechaux M, Miller RT, Antignac C. Functional characterization of a calcium-sensing receptor mutation in severe autosomal dominant hypocalcemia with a Bartter-like syndrome. J Am Soc Nephrol. 2002;13:2259–2266. doi: 10.1097/01.asn.0000025781.16723.68. [DOI] [PubMed] [Google Scholar]
  • 39.Simon DB, Bindra RS, Mansfield TA, Nelson-Williams C, Mendonca E, Stone R, Schurman S, Nayir A, Alpay H, Bakkaloglu A, et al. Mutations in the chloride channel gene, CLCNKB, cause Bartter's syndrome type III. Nat Genet. 1997;17:171–178. doi: 10.1038/ng1097-171. [DOI] [PubMed] [Google Scholar]
  • 40.Pascoe L, Curnow KM, Slutsker L, Rosler A, White PC. Mutations in the human CYP11B2 (aldosterone synthase) gene causing corticosterone methyloxidase II deficiency. Proc Natl Acad Sci U S A. 1992;89:4996–5000. doi: 10.1073/pnas.89.11.4996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mitsuuchi Y, Kawamoto T, Miyahara K, Ulick S, Morton DH, Naiki Y, Kuribayashi I, Toda K, Hara T, Orii T, et al. Congenitally defective aldosterone biosynthesis in humans: inactivation of the P-450C18 gene (CYP11B2) due to nucleotide deletion in CMO I deficient patients. Biochem Biophys Res Commun. 1993;190:864–869. doi: 10.1006/bbrc.1993.1128. [DOI] [PubMed] [Google Scholar]
  • 42.Bongiovanni AM, Root AW. The adrenogenital syndrome. N Engl J Med. 1963;268:1391–1399. doi: 10.1056/NEJM196306202682505. concl. [DOI] [PubMed] [Google Scholar]
  • 43.Amor M, Parker KL, Globerman H, New MI, White PC. Mutation in the CYP21B gene (Ile-172----Asn) causes steroid 21-hydroxylase deficiency. Proc Natl Acad Sci U S A. 1988;85:1600–1604. doi: 10.1073/pnas.85.5.1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bongiovanni AM. The adrenogenital syndrome with deficiency of 3 beta-hydroxysteroid dehydrogenase. J Clin Invest. 1962;41:2086–2092. doi: 10.1172/JCI104666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Simon DB, Karet FE, Rodriguez-Soriano J, Hamdan JH, DiPietro A, Trachtman H, Sanjad SA, Lifton RP. Genetic heterogeneity of Bartter's syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet. 1996;14:152–156. doi: 10.1038/ng1096-152. [DOI] [PubMed] [Google Scholar]
  • 46.Geller DS, Rodriguez-Soriano J, Vallo Boado A, Schifter S, Bayer M, Chang SS, Lifton RP. Mutations in the mineralocorticoid receptor gene cause autosomal dominant pseudohypoaldosteronism type I. Nat Genet. 1998;19:279–281. doi: 10.1038/966. [DOI] [PubMed] [Google Scholar]
  • 47.Chang SS, Grunder S, Hanukoglu A, Rosler A, Mathew PM, Hanukoglu I, Schild L, Lu Y, Shimkets RA, Nelson-Williams C, et al. Mutations in subunits of the epithelial sodium channel cause salt wasting with hyperkalaemic acidosis, pseudohypoaldosteronism type 1. Nat Genet. 1996;12:248–253. doi: 10.1038/ng0396-248. [DOI] [PubMed] [Google Scholar]
  • 48.Simon DB, Karet FE, Hamdan JM, DiPietro A, Sanjad SA, Lifton RP. Bartter's syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotransporter NKCC2. Nat Genet. 1996;13:183–188. doi: 10.1038/ng0696-183. [DOI] [PubMed] [Google Scholar]
  • 49.Simon DB, Nelson-Williams C, Bia MJ, Ellison D, Karet FE, Molina AM, Vaara I, Iwata F, Cushner HM, Koolen M, et al. Gitelman's variant of Bartter's syndrome, inherited hypokalaemic alkalosis, is caused by mutations in the thiazide-sensitive Na-Cl cotransporter. Nat Genet. 1996;12:24–30. doi: 10.1038/ng0196-24. [DOI] [PubMed] [Google Scholar]
  • 50.Cruz DN, Simon DB, Nelson-Williams C, Farhi A, Finberg K, Burleson L, Gill JR, Lifton RP. Mutations in the Na-Cl cotransporter reduce blood pressure in humans. Hypertension. 2001;37:1458–1464. doi: 10.1161/01.hyp.37.6.1458. [DOI] [PubMed] [Google Scholar]
  • 51. 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.. •• Richard Lifton’s group describes the association between rare heterozygous alleles in three candidate genes previously associated with monogenic forms of hypotension and lower BP in a population-based study. This is the first study to demonstrate that rare variants play a significant role in BP regulation at the population level.
  • 52.Cohen JC, Kiss RS, Pertsemlidis A, Marcel YL, McPherson R, Hobbs HH. Multiple rare alleles contribute to low plasma levels of HDL cholesterol. Science. 2004;305:869–872. doi: 10.1126/science.1099870. [DOI] [PubMed] [Google Scholar]
  • 53.Altmuller J, Palmer LJ, Fischer G, Scherb H, Wjst M. Genomewide scans of complex human diseases: true linkage is hard to find. Am.J.Hum.Genet. 2001;69:936–950. doi: 10.1086/324069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.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]
  • 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.Anderson JV, Struthers AD, Payne NN, Slater JD, Bloom SR. Atrial natriuretic peptide inhibits the aldosterone response to angiotensin II in man. Clin Sci (Lond) 1986;70:507–512. doi: 10.1042/cs0700507. [DOI] [PubMed] [Google Scholar]
  • 57.Harris PJ, Thomas D, Morgan TO. Atrial natriuretic peptide inhibits angiotensin-stimulated proximal tubular sodium and water reabsorption. Nature. 1987;326:697–698. doi: 10.1038/326697a0. [DOI] [PubMed] [Google Scholar]
  • 58.Itoh H, Pratt RE, Dzau VJ. Atrial natriuretic polypeptide inhibits hypertrophy of vascular smooth muscle cells. J Clin Invest. 1990;86:1690–1697. doi: 10.1172/JCI114893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.John SW, Krege JH, Oliver PM, Hagaman JR, Hodgin JB, Pang SC, Flynn TG, Smithies O. Genetic decreases in atrial natriuretic peptide and salt-sensitive hypertension. Science. 1995;267:679–681. doi: 10.1126/science.7839143. [DOI] [PubMed] [Google Scholar]
  • 60.Lopez MJ, Wong SK, Kishimoto I, Dubois S, Mach V, Friesen J, Garbers DL, Beuve A. Salt-resistant hypertension in mice lacking the guanylyl cyclase-A receptor for atrial natriuretic peptide. Nature. 1995;378:65–68. doi: 10.1038/378065a0. [DOI] [PubMed] [Google Scholar]
  • 61.Schillinger KJ, Tsai SY, Taffet GE, Reddy AK, Marian AJ, Entman ML, Oka K, Chan L, O'Malley BW. Regulatable atrial natriuretic peptide gene therapy for hypertension. Proc.Natl.Acad.Sci.U.S.A. 2005;102:13789–13794. doi: 10.1073/pnas.0506807102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Newton-Cheh C, Larson MG, Vasan RS, Levy D, Bloch KD, Surti A, Guiducci C, Kathiresan S, Benjamin EJ, Struck J, et al. 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.. •• The first confirmed common variant associated with blood pressure. The report describes the association of common noncoding alleles near two candidate genes (NPPA and NPPB) with increased circulating plasma natriuretic peptide concentrations and lower BP and hypertension risk.
  • 63. Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, Coin L, Najjar SS, Zhao JH, Heath SC, Eyheramendy S, et al. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet. 2009 doi: 10.1038/ng.361.. •• This study by the Global BPgen consortium reported meta-analysis of GWAS with replication (total n over 100,000) identifying 8 loci associated with SBP and DBP.
  • 64. Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, Dehghan A, Glazer NL, Morrison AC, Johnson AD, Aspelund T, et al. Genome-wide association study of blood pressure and hypertension. Nat Genet. 2009 doi: 10.1038/ng.384.. •• This study by the CHARGE BP consortium reported meta-analysis of GWAS with in silico replication in Global BPgen, identifying 8 loci associated with SBP and DBP.
  • 65.Cho YS, Go MJ, Kim YJ, Heo JY, Oh JH, Ban HJ, Yoon D, Lee MH, Kim DJ, Park M, et al. 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]
  • 66.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]
  • 67.Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, Roix JJ, Kathiresan S, Hirschhorn JN, Daly MJ, et al. 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]
  • 68.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]
  • 69.Pe'er I, Yelensky R, Altshuler D, Daly MJ. Estimation of the multiple testing burden for genomewide association studies of nearly all common variants. Genet Epidemiol. 2008;32:381–385. doi: 10.1002/gepi.20303. [DOI] [PubMed] [Google Scholar]
  • 70.Kato N, Miyata T, Tabara Y, Katsuya T, Yanai K, Hanada H, Kamide K, Nakura J, Kohara K, Takeuchi F, et al. High-density association study and nomination of susceptibility genes for hypertension in the Japanese National Project. Hum.Mol.Genet. 2008;17:617–627. doi: 10.1093/hmg/ddm335. [DOI] [PubMed] [Google Scholar]
  • 71.Sabatti C, Service SK, Hartikainen AL, Pouta A, Ripatti S, Brodsky J, Jones CG, Zaitlen NA, Varilo T, Kaakinen M, et al. Genome-wide association analysis of metabolic traits in a birth cohort from a founder population. Nat Genet. 2009;41:35–46. doi: 10.1038/ng.271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wang Y, O'Connell JR, McArdle PF, Wade JB, Dorff SE, Shah SJ, Shi X, Pan L, Rampersaud E, Shen H, et al. From the Cover: Whole-genome association study identifies STK39 as a hypertension susceptibility gene. Proc Natl Acad Sci U S A. 2009;106:226–231. doi: 10.1073/pnas.0808358106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Org E, Eyheramendy S, Juhanson P, Gieger C, Lichtner P, Klopp N, Veldre G, Doring A, Viigimaa M, Sober S, et al. 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]
  • 74.Adeyemo A, Gerry N, Chen G, Herbert A, Doumatey A, Huang H, Zhou J, Lashley K, Chen Y, Christman M, et al. 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]
  • 75.Richardson C, Rafiqi FH, Karlsson HK, Moleleki N, Vandewalle A, Campbell DG, Morrice NA, Alessi DR. Activation of the thiazide-sensitive Na+-Cl− cotransporter by the WNK-regulated kinases SPAK and OSR1. J Cell Sci. 2008;121:675–684. doi: 10.1242/jcs.025312. [DOI] [PubMed] [Google Scholar]
  • 76.Moriguchi T, Urushiyama S, Hisamoto N, Iemura S, Uchida S, Natsume T, Matsumoto K, Shibuya H. WNK1 regulates phosphorylation of cation-chloride-coupled cotransporters via the STE20-related kinases, SPAK and OSR1. J Biol Chem. 2005;280:42685–42693. doi: 10.1074/jbc.M510042200. [DOI] [PubMed] [Google Scholar]
  • 77.Pande J, Mallhi KK, Sawh A, Szewczyk MM, Simpson F, Grover AK. Aortic smooth muscle and endothelial plasma membrane Ca2+ pump isoforms are inhibited differently by the extracellular inhibitor caloxin 1b1. Am J Physiol Cell Physiol. 2006;290:C1341–C1349. doi: 10.1152/ajpcell.00573.2005. [DOI] [PubMed] [Google Scholar]
  • 78.Schadt EE, Molony C, Chudin E, Hao K, Yang X, Lum PY, Kasarskis A, Zhang B, Wang S, Suver C, et al. Mapping the genetic architecture of gene expression in human liver. PLoS Biol. 2008;6:e107. doi: 10.1371/journal.pbio.0060107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lin D, Harikrishna JA, Moore CC, Jones KL, Miller WL. Missense mutation serine106----proline causes 17 alpha-hydroxylase deficiency. J Biol Chem. 1991;266:15992–15998. [PubMed] [Google Scholar]
  • 80.Todd JA, Walker NM, Cooper JD, Smyth DJ, Downes K, Plagnol V, Bailey R, Nejentsev S, Field SF, Payne F, et al. 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]
  • 81.Hunt KA, Zhernakova A, Turner G, Heap GA, Franke L, Bruinenberg M, Romanos J, Dinesen LC, Ryan AW, Panesar D, et al. 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]
  • 82.Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P, Jonsdottir GM, Thorleifsson G, Helgadottir H, Steinthorsdottir V, Stefansson H, et al. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009 doi: 10.1038/ng.323. [DOI] [PubMed] [Google Scholar]
  • 83.Soranzo N, Spector TD, Mangino M, Kuhnel B, Rendon A, Teumer A, Willenborg C, Wright B, Chen L, Li M, et al. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet. 2009;41:1182–1190. doi: 10.1038/ng.467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Fitau J, Boulday G, Coulon F, Quillard T, Charreau B. The adaptor molecule Lnk negatively regulates tumor necrosis factor-alpha-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]
  • 85.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]
  • 86.Kathiresan S, Willer CJ, Peloso GM, Demissie S, Musunuru K, Schadt EE, Kaplan L, Bennett D, Li Y, Tanaka T, et al. Common variants at 30 loci contribute to polygenic dyslipidemia. Nat Genet. 2009;41:56–65. doi: 10.1038/ng.291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dendrou CA, Plagnol V, Fung E, Yang JH, Downes K, Cooper JD, Nutland S, Coleman G, Himsworth M, Hardy M, et al. Cell-specific protein phenotypes for the autoimmune locus IL2RA using a genotype-selectable human bioresource. Nat Genet. 2009;41:1011–1015. doi: 10.1038/ng.434. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES