Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2013 Oct 16;306(1):F1–F11. doi: 10.1152/ajprenal.00334.2013

Genetics of hypertension: discoveries from the bench to human populations

Nora Franceschini 1, Thu H Le 2,
PMCID: PMC3921821  PMID: 24133117

Abstract

Hypertension is a complex trait that is influenced by both heritable and environmental factors. The search for genes accounting for the susceptibility to hypertension has driven parallel efforts in human research and in research using experimental animals in controlled environmental settings. Evidence from rodent models of genetic hypertension and human Mendelian forms of hypertension and hypotension have yielded mechanistic insights into the pathways that are perturbed in blood pressure homeostasis, most of which converge at the level of renal sodium reabsorption. However, the bridging of evidence from these very diverse approaches to identify mechanisms underlying hypertension susceptibility and the translation of these findings to human populations and public health remain a challenge. Furthermore, findings from genome-wide association studies still require functional validation in experimental models. In this review, we highlight results and implications from key studies in experimental and clinical hypertension to date.

Keywords: genes; genetics, hypertension


hypertension is a leading cause of renal and cardiovascular disease morbidity, mortality, and disability worldwide (48, 64, 67). Hypertension affects one in every three US adults (10, 39) with a disproportionate burden in US minorities, particularly African Americans (43). In most cases, the etiology of hypertension is unknown (essential hypertension). Blood pressure is an important predictor of health in populations. The American Heart Association Strategy Impact Goals to improve cardiovascular health to all Americans by 2020 include an ideal blood pressure target (<120/80 mmHg) among seven attributes for cardiovascular health (72), therefore recognizing the importance of blood pressure levels for population health.

Several mechanisms and pathways for blood pressure regulation have been described in studies of humans and experimental animal models (41, 42, 44, 51), and their dysfunction has been shown to lead to hypotensive and hypertensive disease states. However, the mechanisms contributing to blood pressure homeostasis in the general population are complex and are likely modulated by genetic factors and their interactions with environmental and behavioral factors. Studies of genetic determinants of blood pressure in humans and experimental animal models can provide complementary information on disease susceptibility. In this regard, gene-targeting studies in animals can clarify the biology and function of genes affecting blood pressure. Genetic studies in humans provide population-specific information on disease susceptibility for variants affecting blood pressure in the context of a diverse diet, physical activity, and other behavioral factors and can also inform on allelic heterogeneity and ancestry-specific genetic susceptibility (for example, ALDH2 variants in east Asians) (59). The difficult task remains in the bridging of evidence from controlled experimental data to translate the findings to human populations and public health. Conversely, recent genome-wide scans in humans have uncovered previously unsuspected genes for blood pressure that will need functional validation in experimental settings. This review aims to provide an update of the current knowledge of the genetics of hypertension by discussing 1) evidence from animal models, 2) evidence from human Mendelian disorders, and 3) evidence from population studies.

Genetic Models of Hypertension in Rodents: Role of the Kidney

Over 50 years ago, the research community developed rat models of hypertension by which to study essential hypertension in humans. Through selective breeding, several commonly employed rat models of genetic hypertension were generated: the spontaneously hypertensive rats (SHR), the Dahl salt-sensitive rats (DS), and the Milan hypertensive rats. Though these models represent only a rare form of human hypertension with a Mendelian mode of inheritance, studies using these inbred strains have generated key insights. In particular, kidney cross-transplantation studies using hypertensive and normotensive rats derived from one single strain overwhelmingly demonstrated that the genotype of the donor kidney plays a principal role in determining chronic blood pressure levels in the recipient (6, 20, 81, 89). Dahl and colleagues (20) concluded from these studies that “genetic factors operating through the kidney can chronically modify blood pressure”.

As blood pressure is a quantitative trait, much effort has been focused on the identification of genes underlying quantitative trait loci (QTLs) that influence blood pressure regulation. The utilization of traditional crosses and linkage analysis for gene mapping in these hypertensive rat strains led the way for successful identification of numerous blood pressure QTLs on virtually every chromosome in the rat. Over 350 blood pressure QTLs across the rat genome have been reported to date (Rat Genome Database: http://rgd.mcw.edu/). Despite integrative approaches using congenic strategies for fine-mapping, gene expression arrays, and comparative and functional genomics, identification of causative gene(s) underlying a QTL has remained a significant challenge. Factors contributing to this bottleneck include small phenotypic effects of a causative gene and epistatic interactions of genes that influence blood pressure but with opposing effects.

There are, however, a few examples of successful gene identification that deserve particular mention since they provide mechanistic insights into the regulation of blood pressure. The first of these examples was linkage analysis in the Dahl rats that identified genetic polymorphisms in the 11β-hydroxylase gene (Cyp11b1) that cosegregated in a Mendelian manner with the adrenal capacity to synthesize 18-hydroxy-11-deoxycorticosterone (18-OH-DOC) and blood pressure (17). Compared with the Dahl salt-sensitive strain and 11 other strains, the Dahl salt-resistant strain was found to have a different 11β-hydroxylase allele by restriction fragment length polymorphism and 5 amino acid substitutions in the 11β-hydroxylase protein that were associated with a reduced capacity to synthesize 18-OH-DOC (17). Through a series of studies using congenic strategy that resulted in the generation of a 177-kb congenic segment containing nine known genes, the Cyp11b1 gene is the only gene among the nine genes in this locus that has known biological functions related to blood pressure (38). Although earlier binding studies demonstrated that 18-OH-DOC has one-fiftieth binding affinity to mineralocorticoid receptors compared with aldosterone (30, 84), the mechanism by which the 11β-hydroxylase variants affect blood pressure can be explained by observations showing that circulating levels of 18-OH-DOC in the rat are much higher than that of aldosterone (84). Thus, at elevated levels, 18-OH-DOC can compete with aldosterone for binding to the mineralocorticoid receptors (84). While these studies may explain the blood pressure difference between the Dahl salt-sensitive and Dahl salt-resistant rats, other rat strains such as Lewis rats that carry the same 11β-hydroxylase allele as the Dahl salt-sensitive rats (17) are normotensive (18). Thus blood pressure in the normotensive Lewis rats and Dahl salt-resistant rats may be governed by different genes (18). Furthermore, there may be another genetic factor(s) that influences salt sensitivity in the Dahl background. Mutations in the CYB11B1 gene have also been identified in rare Mendelian forms of human hypertension, such as congenital adrenal hyperplasia (99) and glucocorticoid-remediable hyperaldosteronism (6869) described below. Importantly, the concordance across species illustrates the evolutionary conservation of this pathway in blood pressure regulation.

Concordance in humans and rats has also been demonstrated for the role of the α-adducin (ADD1) gene in hypertension. Adducin is a membrane-skeleton protein composed of α- and β-subunits that are encoded by genes on separate chromosomes (12). In renal tubular cells in culture, adducin has been shown to associate with cell-to-cell contact sites in a calcium- and phosphorylation-dependent manner (58). Candidate gene analysis revealed that functional polymorphisms in the α-adducin gene (ADD1) are associated with hypertension in Milan rats and salt-sensitive hypertension in humans (7, 19). In a subsequent whole genome scan for blood pressure QTLs in the Milan hypertensive and normotensive strains, the α-adducin locus reached the threshold of significance for linkage to blood pressure. Functional studies later revealed that polymorphisms in the α-adducin gene may influence blood pressure by regulating the activity of the Na+-K+-ATPase pump that drives renal tubular sodium reabsorption (31).

The SHR has been the most widely studied rat model of hypertension. Although multiple blood pressure QTLs have been identified in this model, it was not until efforts to map intermediate phenotypes of the metabolic syndrome in an SHR-derived strain that successful identification of a gene underlying a blood pressure QTL was achieved. In this regard, linkage studies of adipocyte insulin sensitivity and catecholamines (3), followed by a series of congenic fine-mapping strategies, gene expression analysis, and comparative genomics, resulted in the identification that a loss-of-function mutation in the gene that encodes the CD36 fatty acid transporter could explain these intermediate phenotypes of metabolic syndrome (84). The generation of multiple transgenic lines showed that expression of wild-type CD36 rescued the metabolic syndrome in the SHR (85) and led to the observation that differences in renal expression of CD36 could explain the differences in blood pressure between the transgenic line with lower blood pressure and those with high blood pressures (84, 86). Kidney cross-transplant studies later confirmed that the level of renal CD36 determines the level of blood pressure (84, 86). The mechanism by which renal expression of the CD36 fatty acid transporter influences blood pressure is not understood, and it remains to be determined whether it influences blood pressure in humans.

The identification that genetic variants in the genes encoding 11β-hydroxylase, α-adducin, and CD36 influence blood pressure in the rat models of genetic hypertension illustrates a mechanism that is consistent with the hypothesis set forth by Arthur Guyton and colleagues 40 years ago (42). They argued that the kidney, by regulating sodium excretion, plays a central role in hypertension (42). This notion is further reinforced by findings from studies in Mendelian forms of hypertension and hypotension in humans described below.

Genetics of Mendelian Forms of Hypertension and Hypotension in Humans: Role of the Kidney

Much of our understanding of genetic mechanisms of blood pressure regulation in humans has resulted from investigations of rare Mendelian forms of hypertension and hypotension. In this regard, Lifton and others have identified over 20 genes with mutations with large effects on blood pressure. These genes primarily encode sodium transporters or channels in the kidney, or components belonging to the mineralocorticoid pathway that ultimately influences the activity of the mineralocorticoid receptor which regulates the epithelial sodium channel activity, thereby affecting renal salt reabsorption (the reader is referred to Refs. 22 and 70 for an extensive review of these genes). As mentioned above, mutation in the Cyp11b1 gene resulting in decreased enzymatic capacity to synthesize 18-OH-DOC is thought to afford protection for the Dahl salt-resistant rat from salt-induced hypertension. In humans, loss of regulatory control mutation resulting from chimeric gene duplication due to unequal crossing over between the two closely linked aldosterone synthase and 11β-hydroxylase genes is attributable to the rare syndrome of glucocorticoid-remediable aldosteronism. In this instance, the aldosterone synthase activity is no longer under the control of angiotensin II but is controlled by the regulatory region of 11β-hydroxylase that is regulated by adrenocorticotropic hormone (ACTH) (68, 69). More recently, mutations in the WNK (with no K-lysine) serine-threonine kinases WNK1 and WNK4 were demonstrated to cause pseudohypoaldosteronism type II disorder of hypertension (100), due to alteration in their coordinated actions to regulate the transport of sodium, chloride, and potassium in the distal nephron (57). Taken together, evidence from rat models of genetic hypertension and human monogenic forms of hypertension illustrate that the kidney, by regulating sodium balance, plays a key role in blood pressure regulation. A logical question follows whether this paradigm holds true in human essential hypertension.

Genetics of Hypertension and Blood Pressure in Human Populations

Blood pressure has a substantial heritable component (30–50%) in humans, as illustrated by familial aggregation of hypertension and the highly penetrant genetic variants causing monogenic forms of hypertension and hypotension mentioned above. To address genetic causes of hypertension in the general population, earlier studies included association analyses of candidate genes belonging to biological or physiological pathways known to affect blood pressure. In particular, genes encoding components of the renin-angiotensin system have been analyzed as candidate genes for inherited forms of hypertension in humans. For example, the M235T variant of the human angiotensinogen gene (AGT) has been shown to be linked to hypertension and associated with a modestly elevated plasma angiotensinogen concentration in human hypertensive siblings (53). Although amino acid 235 is in a nonconserved portion of the angiotensinogen protein and variation of this amino acid does not affect protein stability, the M235T variant was later found to be in linkage disequilibrium (LD) with a single nucleotide substitution variant in the promoter of the AGT gene that was associated with increased transcriptional activity of the gene (50). Patients carrying the variant allele were demonstrated to have increased levels of AGT mRNA (50).

To provide direct proof of a causal relationship that mutations in the angiotensinogen gene that increase circulating plasma levels of angiotensinogen could directly increase blood pressure, Kim and Smithies (95) used a targeted gene duplication and deletion approach to generate mouse lines with one to four functional copies of the Agt gene (61). Through this gene titration approach, they demonstrated a positive correlation between plasma angiotensinogen levels and the number of Agt gene copies, as well as a direct linear relationship between blood pressures and number of copies of Agt genes (61). Despite the biological plausibility, the finding in a small cohort of hypertensive siblings and the proof of concept in the mice, the effect of the AGT M235T variant on blood pressure was not duplicated in some human populations (26, 76).

Similarly, inconsistencies have been seen in association studies of the insertion/deletion polymorphism of the angiotensin-converting enzyme (ACE) gene that correlates with circulating plasma ACE levels (21, 40, 54, 90). The Gstm1 gene is also an example of challenges in translating findings from experimental models to human hypertension. Gstm1, a member of the glutathione-S-transferase superfamily of genes, was identified as a strong positional and functional candidate gene for hypertension in the stroke-prone spontaneously hypertensive rat (SHRSP) (52). Decreased expression of the GSTM1 enzyme along with an associated increase in oxidative stress have been demonstrated in the kidneys of SHRSP rats (75). Reduction in GSTM1 levels in vascular smooth muscle cells in vitro causes increased production of reactive oxygen species and cell proliferation (102). Although the common deletion variant of GSMT1 was found to be associated with end-organ damage such as accelerated kidney disease progression in hypertension-associated kidney disease (14) and end-stage renal disease (2) in different human cohorts, it has not been demonstrated to be associated with hypertension in humans (23). Multiple other candidate genes analyses, as well as studies using family-based, genome-wide linkage analysis for hypertension have shown inconsistent results (8, 47, 92).

Recent discovery efforts have used hypothesis-free genome-wide association scans (GWAS) to identify genomic loci for blood pressure variation and hypertension susceptibility. These studies have been successful in identifying common genetic variants (allele frequency >5%) for complex diseases including blood pressure traits (28). Additional complementary methods used for gene discovery in individuals with recent admixture, such as African Americans, are admixture mapping analyses, which are based on alleles that differ in frequency across populations of different ancestry and are thus suitable to the study of diseases, such as hypertension, with disparities among racial/ethnic groups (24, 103). Findings for blood pressure traits using GWAS and admixture mapping analyses in populations studies are described below.

Overview of genome-wide methods to study genetic determinants in humans.

GWAS use dense sets of genetic markers (500,000 to 1 million) to investigate associations of single nucleotide polymorphism (SNP) with complex traits. GWA studies are usually powered for common variants, which are defined as having the minor allele frequency of at least 5% in a population. To increase sample size for variant discovery, data are combined across studies using meta-analyses. Additional SNPs are imputed using reference panels of ancestry-specific populations from the International HapMap Project (34) or the 1,000 Genomes Project (1). For populations with admixture (e.g., Hispanics, African Americans) and/or not represented in the HapMap samples (American Indians), a strategy to improve imputation quality is to obtain study-specific reference haplotypes by using genotype data from a subset of individuals (for example, a sample of individuals chosen to be representative of individuals in the study have dense genotyping, which is then used to estimate haplotypes for imputation in the remaining individuals in the study) (71).

The associations rely on the LD or correlation of typed (or imputed) SNPs with unknown functional variants. Therefore, SNPs identified in GWAS are usually proxies of untyped functional variants, which has implications for fine-mapping of genomic regions (5). Population stratification can lead to spurious associations at markers that have different allele frequency among ancestry subpopulations, such as alleles that are more common among African Americans than European ancestry individuals. In other words, the observed differences in allele frequencies between the cases and controls are due to systematic differences in ancestry rather than association of genes with disease (35). Adjustments for population stratification are usually done using principal components and other methods (87).

To adjust for multiple testing and to decrease false-positive rates (type I error), stringent P values are used (usually 5.0 × 10−8 for GWAS). For variants reaching the P value threshold, further validation is required by replication of the associations in additional individuals for the same trait (15). Consortia and collaborative studies have enabled meta-analyses of multiple GWAS based on large samples and statistical power sufficient to identify novel common variants. The National Human Genome Research Institute (NHGRI) has an ongoing repository of all published SNP-trait associations with P values <1.0 × 10−5 (assessed on the genome catalog website) (45).

Discovery efforts for common variants of blood pressure: GWAS studies.

Most of the large published studies to date have targeted individuals of European ancestry. Two large meta-analyses of GWAS of European ancestry individuals lead by the Cohorts for Heart and Aging Research in Genome Epidemiology (CHARGE) and Global Blood Pressure Genetics (Global BP Gen) consortia revealed 13 new loci for systolic or diastolic blood pressure (CYP17A1, PLEKHA7, ATP2B1, SH2B3-ATXN2, ULK4, TBX3-TBX5,CSK-ULK3, CACNB2,MTHFR-CLCN6-NPPA-NPPB, PLCD3, FGF5, C10orf107, and ZNF652) (65, 77). These studies were followed by publication of results from the International Consortium for Blood Pressure (ICBP), which included CHARGE and Global BP Gen samples, and additional studies for a discovery sample of 69,395 individuals, and combined discovery and replication samples of ∼200,000 individuals of European ancestry (28). ICBP successfully identified 16 additional novel loci for a total of 29 independent variants identified in 28 loci. Nine of these loci were replicated in East Asians and six in individuals of South Asian ancestry, but none of the identified SNPs was associated with blood pressure traits in individuals of African Ancestry (28). However, the identified variants explained only a very small proportion of the blood pressure trait variability (0.9%), suggesting the presence of additional variants to be discovered that may account for the missing heritability of the trait (28). Some of these loci were known or suspected to be involved in blood pressure regulation and/or were related to kidney development and/or function in animal models (Table 1). However, the overlap of genes identified in rat models of hypertension or human Mendelian forms of hypertension or hypotension with those identified in GWAS is small. The underlying reasons for the lack of overlap are unknown but could be speculated to be contributed by differences in phenotypes, studied genetic variants, inaccuracy of blood pressure measurements, and environmental factors in humans. For example, most of the published GWAS were performed in individuals recruited from the population who are unlikely to have severe hypertension or hypotension. In contrast, human monogenic disorders and hypertensive animal models have extreme values of blood pressure distribution. GWAS tests common genetic variants, which usually have small effects on traits and thus require very large sample sizes, whereas experimental models and Mendelian disorders display large effects that can be detected even with small samples. In addition, the lack of significant capture of these genes in human GWAS may also be due to low coverage of genotyping at some loci (and lack of proxies due to LD), bias in blood pressure measurement in studies, and the effect of environmental factors not accounted for in the analyses.

Table 1.

Genes near loci identified in GWAS associated with blood pressure and/or hypertension and their biological relevance in human and animal studies

Gene (Name)*/Reference(s) Mechanism(s) Human Evidence Animal Evidence
CYP17A1 (cytochrome P-450 enzyme) (66, 78 Pathways of steroid synthesis Congenital adrenal hyperplasia (MIM no. 202110)
ADM (adrenomedullin) (28, 97) Vasodilation and angiogenesis promotion Protein found in human pheochromocytoma
NPPA/NPPB (natriuretic peptide A and B) (77) Precursors for atrial- and B-type natriuretic peptides
NPR3 (natriuretic peptide clearance receptor) (28, 59, 97) Natriuretic pathways
GUCY1A3-GUCY1B3 (α− and β−subunits of soluble guanylate cyclase) (28, 97) Both units form the guanylate cyclase enzyme that activates nitric oxide. Vasodilatory mechanism for nitrovasodilator drugs Knockout mice of either gene develop hypertension (36).
EDN3 (endothelin 3) (65, 97) Potent vasoconstrictor
SLC4A7 (electroneutral sodium bicarbonate cotransporter) (28, 97) Kidney sodium-bicarbonate cotransporter
PLCE1 (phospholipase C-epsilon-1 isoform) (28) Podocyte development Nephrotic syndrome type 3 (MIM no. 610725)
HFE (hemochromatosis) (28, 97) Ion transport (iron); diabetes Hemochromatosis (MIM no. 613609)
SLC39A8 (solute carrier family 39, zinc transporter) (28, 97) Ion transport (cadmium)
ENPEP (glutamyl aminopeptidase (aminopeptidase A) (59) Glutamyl aminopeptidase facilitates conversion of angiotensinogen II
ATP2B1 (ATPase, Ca2+ transporting, plasma membrane 1) (16, 59, 65, 96) Calcium homeostasis VSMC target tissue knockout mice show higher blood pressure than wild-type and augmented vasoconstrictive response of femoral artery to phenylephrine stimulation (62).
NOS3 (nitric oxide synthase 3 (endothelial cell) (91) Vasodilatory effects
PIK3CG (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit gamma) (97) Mice lacking PIK3γ have shown resistance to the SBP-lowering effects of β-adrenergic receptor agonist (82).
NOV (nephroblastoma overexpressed) (28, 94) Angiogenesis, proliferation and inhibition of vascular smooth muscle cell growth and migration (29) Mice null for the nephroblastoma-overexpressed protein (CCN3) have reduced neointimal thickening (93).
ADRB1 (adrenoceptor beta 1) (97) Mediates physiological effects of the hormone epinephrine and the neurotransmitter norepinephrine Associated with resting heart rate in humans (88)

VSMC, vascular smooth muscle cells; SBP, systolic blood pressure.

*

Listed genes are from genome-wide association scans (GWAS) studies.

Populations vary significantly in disease risks, allele frequencies, and LD patterns by ancestry. While these factors may pose challenges for the study of complex traits, they can actually be used advantageously for gene discovery. For example, some variants may have a higher allele frequency or a larger effect size in some populations, which would increase the power to identify the associations (59). The Asian Genetic Epidemiology Network BP (AGEN-BP) work group recently published its blood pressure GWAS (59). Using a much smaller discovery sample compared with European ancestry ICBP (19,608 East Asian individuals) and a two-stage replication in up to 30,000 additional East Asians, they identified four novel loci and a variant near the NPR3 locus, previously described in ICBP for European ancestry individuals (28, 59). When the associations at 13 loci in East Asians and individuals of European ancestry are compared, differences in allele frequency and in the magnitude of effect estimates affected the P values and sample size required to reach significance. For example, the CASZ1 SNP rs880315 (at-risk C allele frequency 0.59 in East Asians and 0.35 in Europeans) that did not reach genome-wide significant levels in Europeans but was found to show a significant association with blood pressure in the AGEN-BP meta-analysis (59). The study also identified a novel blood pressure SNP at the TBX3 locus, demonstrating the presence of allelic heterogeneity at this locus (28, 59, 65). Additional blood pressure studies have been recently published for a total of ∼50 common genetic variants associated with blood pressure or hypertension through GWAS or admixture mapping (28, 32, 46, 56, 65, 77, 79, 83, 101, 103). These studies include GWAS using gene-centric arrays (37, 56), and studies of individuals of diverse ancestry including African Americans (32, 33, 103) and Asians (16).

Several of the blood pressure-associated regions identified through GWAS extend over multiple genes due to the strong LD or local correlation between SNPs in individuals of European ancestry. Studying populations with less SNP correlation may thus facilitate finding causal gene(s) in these loci, i.e., fine-mapping genomic regions. In the Genetic Epidemiology NeTwork of Blood Pressure (COGENT-BP) consortium, a large GWAS meta-analyses of blood pressure of 29,378 individuals of African ancestry from 19 discovery cohorts, we were able to fine map four known loci by reducing the length of the genomic region of associations (EBF1, ATP2B1, NT5C2, and ULK4) (33).

Several collaborative efforts to study blood pressure traits in non-European ancestry individuals have been established and are expected to yield novel findings in the coming months. These consortia include the Japanese Millennium Genome Project, the Korea Association Resource consortium, the Asian Genetic Epidemiology Network, the NHLBI Candidate-Gene Association Resource (African Americans), the COGENT-BP (African and African Americans), and the Genomics of Blood Pressure for Hispanics Consortium (GHBP, US and Mexican Hispanics). Loci identified may provide new insights into the biology of blood pressure regulation and may reveal targets for therapy and for population-level risk reduction.

Population-specific and rare/low-frequency variants.

In addition to allelic heterogeneity, both population-specific variants and variants with effects across ancestries (cosmopolitan ones) may contribute to hypertension risk. The AGEN-BP GWAS described an association at the ALDH2 locus for a SNP in LD with rs671, a variant previously associated with hypertension mediated by alcohol consumption in Asians (59). The rs671 variant is common in East Asians (MAF >5%) but not polymorphic in Europeans, and is an example of a population-specific common variant influencing the blood pressure trait. Population specificity becomes even more important for low-frequency and rare variants (11).

The small proportion of the variance in quantitative, blood pressure-related traits explained by common SNPs in GWAS suggests the presence of additional rare (<1%) and low-frequency (1–0.5%) variants influencing these traits (28). Rare variants constitute the vast majority of polymorphic sites in human populations (74), are of recent origin (73), tend to be population specific (11), and may have large phenotypic effects (63). Aggregations of rare variants may account for some GWAS signals (synthetic association) (25, 60). Sequencing of the human exome, which represents 5% of the human genome, is cost effective (4) and has successfully identified rare, highly penetrant and novel alleles in genes underlying monogenic disorders (4, 13, 80) and some complex diseases (49). For example, exome sequencing revealed that partial or total loss-of-function rare variants of MTNR1B contribute to type 2 diabetes risk (9). More recently, in an effort to identify genes underlying the complex cardiovascular disease traits, the NHLBI Exome Sequencing Project (ESP) has selected individuals from the extremes of the blood pressure distribution for whole exome sequencing. Current high-throughput, commercial exome genotyping arrays which have predominantly common variants have become affordable for use in population-based studies and can be supplemented by the addition of population-specific and low-frequency variants identified through sequencing.

More recent population studies using fine-mapping genotyping or sequencing of blood pressure candidate genes have identified common and rare variants accounting for blood pressure distribution. For example, common variants in the NPPA and NPPB genes were associated with natriuretic peptide concentrations and found to contribute to interindividual blood pressure variation and hypertension risk (79). In a novel approach, known and predicted functional mutations in the genes responsible for the rare Mendelian hypotensive disorders due to renal salt wasting, i.e., Gitelman's syndrome [SLC12A3 (NCCT)] and Bartter's syndrome [SLC12A1 (NKCC2) and KCNJ1 (ROMK)] were identified through direct sequencing of 2,492 participants of the Framingham Heart Study (FHS) (55). Heterozygote carriers of these mutations were demonstrated to have clinically significant lower blood pressure and were protected from the development of hypertension compared with noncarriers (55). At least 1 in 64 FHS participants are carriers of a functional mutation of one of the three genes, explaining ∼1% of blood pressure variation in the FHS population (55), larger than the fraction contributed by the more common variants identified in GWAS mentioned above. Future sequencing studies targeting genes with known function in blood pressure regulation in animal models and in monogenic disorders of hypotension and hypertension in humans may prove useful for discovery of both common and rare variants that can explain even a larger fraction of blood pressure variation in the general population.

Developments on the Horizon

Future research using approaches that leverage functional and biological information may further help in understanding the pathophysiology of complex traits such as hypertension. For example, the ENCODE (Encyclopedia of DNA Elements) Project has mapped and characterized a variety of functional elements including promoters, enhancers, repressor or silencer sequences, transcription factor binding sites, methylation sites, DNase I hypersensitive sites, and chromatin modifications in several cell lines including vascular endothelial cells (27). Pathway analysis can identify enriched pathways in a GWAS set and prioritize regions or candidate genes in genetic studies (98). Additional resources such as metabolomics could be integrated to genetic data to investigate downstream biological system pathways. Integration of these resources with genome-wide genotyped variants and whole genome sequencing variants presents both challenges and opportunities to better understand the genetic architecture of complex traits, including hypertension.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

Author contributions: N.F. and T.L. drafted manuscript; N.F. and T.L. edited and revised manuscript; N.F. and T.L. approved final version of manuscript.

ACKNOWLEDGMENTS

This work was supported by the National Institutes of Health Grant DK094907 to T. H. Le and HHSN268201100004C/NHLBI-WH-11-10 to N. Franceschini.

REFERENCES

  • 1.Abecasis GR, Altshuler D, Auton A, Brooks LD, Durbin RM, Gibbs RA, Hurles ME, McVean GA. A map of human genome variation from population-scale sequencing. Nature 467: 1061–1073, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Agrawal S, Tripathi G, Khan F, Sharma R, Baburaj VP. Relationship between GSTs gene polymorphism and susceptibility to end stage renal disease among North Indians. Ren Fail 29: 947–953, 2007 [DOI] [PubMed] [Google Scholar]
  • 3.Aitman TJ, Gotoda T, Evans AL, Imrie H, Heath KE, Trembling PM, Truman H, Wallace CA, Rahman A, Dore C, Flint J, Kren V, Zidek V, Kurtz TW, Pravenec M, Scott J. Quantitative trait loci for cellular defects in glucose and fatty acid metabolism in hypertensive rats. Nat Genet 16: 197–201, 1997 [DOI] [PubMed] [Google Scholar]
  • 4.Bamshad MJ, Ng SB, Bigham AW, Tabor HK, Emond MJ, Nickerson DA, Shendure J. Exome sequencing as a tool for Mendelian disease gene discovery. Nat Rev Genet 12: 745–755, 2011 [DOI] [PubMed] [Google Scholar]
  • 5.Bhangale TR, Rieder MJ, Nickerson DA. Estimating coverage and power for genetic association studies using near-complete variation data. Nat Genet 40: 841–843, 2008 [DOI] [PubMed] [Google Scholar]
  • 6.Bianchi G, Fox U, Di Francesco GF, Bardi U, Radice M. The hypertensive role of the kidney in spontaneously hypertensive rats. Clin Sci Mol Med Suppl 45, Suppl 1: 135s–139s, 1973 [DOI] [PubMed] [Google Scholar]
  • 7.Bianchi G, Tripodi G, Casari G, Salardi S, Barber BR, Garcia R, Leoni P, Torielli L, Cusi D, Ferrandi M, Pinna LA, Barelles FE, Ferrari P. Two point mutations within the adducin genes are involved in blood pressure variation. Proc Natl Acad Sci USA 91: 3999–4003, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Binder A. A review of the genetics of essential hypertension. Curr Opin Cardiol 22: 176–184, 2007 [DOI] [PubMed] [Google Scholar]
  • 9.Bonnefond A, Clement N, Fawcett K, Yengo L, Vaillant E, Guillaume JL, Dechaume A, Payne F, Roussel R, Czernichow S, Hercberg S, Hadjadj S, Balkau B, Marre M, Lantieri O, Langenberg C, Bouatia-Naji N, Charpentier G, Vaxillaire M, Rocheleau G, Wareham NJ, Sladek R, McCarthy MI, Dina C, Barroso I, Jockers R, Froguel P. Rare MTNR1B variants impairing melatonin receptor 1B function contribute to type 2 diabetes. Nat Genet 44: 297–301, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.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 25: 305–313, 1995 [DOI] [PubMed] [Google Scholar]
  • 11.Bustamante CD, Burchard EG, De la Vega FM. Genomics for the world. Nature 475: 163–165, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Casari G, Barlassina C, Cusi D, Zagato L, Muirhead R, Righetti M, Nembri P, Amar K, Gatti M, Macciardi F, Binelli G, Bianchi G. Association of the alpha-adducin locus with essential hypertension. Hypertension 25: 320–326, 1995 [DOI] [PubMed] [Google Scholar]
  • 13.Chambers JC, Zhang W, Lord GM, van der Harst P, Lawlor DA, Sehmi JS, Gale DP, Wass MN, Ahmadi KR, Bakker SJ, Beckmann J, Bilo HJ, Bochud M, Brown MJ, Caulfield MJ, Connell JM, Cook HT, Cotlarciuc I, Davey Smith G, de Silva R, Deng G, Devuyst O, Dikkeschei LD, Dimkovic N, Dockrell M, Dominiczak A, Ebrahim S, Eggermann T, Farrall M, Ferrucci L, Floege J, Forouhi NG, Gansevoort RT, Han X, Hedblad B, Homan van der Heide JJ, Hepkema BG, Hernandez-Fuentes M, Hypponen E, Johnson T, de Jong PE, Kleefstra N, Lagou V, Lapsley M, Li Y, Loos RJ, Luan J, Luttropp K, Marechal C, Melander O, Munroe PB, Nordfors L, Parsa A, Peltonen L, Penninx BW, Perucha E, Pouta A, Prokopenko I, Roderick PJ, Ruokonen A, Samani NJ, Sanna S, Schalling M, Schlessinger D, Schlieper G, Seelen MA, Shuldiner AR, Sjogren M, Smit JH, Snieder H, Soranzo N, Spector TD, Stenvinkel P, Sternberg MJ, Swaminathan R, Tanaka T, Ubink-Veltmaat LJ, Uda M, Vollenweider P, Wallace C, Waterworth D, Zerres K, Waeber G, Wareham NJ, Maxwell PH, McCarthy MI, Jarvelin MR, Mooser V, Abecasis GR, Lightstone L, Scott J, Navis G, Elliott P, Kooner JS. Genetic loci influencing kidney function and chronic kidney disease. Nat Genet 42: 373–375, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chang J, Ma JZ, Zeng Q, Cechova S, Gantz A, Nievergelt C, O'Connor D, Lipkowitz M, Le TH. Loss of GSTM1, a NRF2 target, is associated with accelerated progression of hypertensive kidney disease in the African American Study of Kidney Disease (AASK). Am J Physiol Renal Physiol 304: F348–F355, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chanock SJ, Manolio T, Boehnke M, Boerwinkle E, Hunter DJ, Thomas G, Hirschhorn JN, Abecasis G, Altshuler D, Bailey-Wilson JE, Brooks LD, Cardon LR, Daly M, Donnelly P, Fraumeni JF, Jr, Freimer NB, Gerhard DS, Gunter C, Guttmacher AE, Guyer MS, Harris EL, Hoh J, Hoover R, Kong CA, Merikangas KR, Morton CC, Palmer LJ, Phimister EG, Rice JP, Roberts J, Rotimi C, Tucker MA, Vogan KJ, Wacholder S, Wijsman EM, Winn DM, Collins FS. Replicating genotype-phenotype associations. Nature 447: 655–660, 2007 [DOI] [PubMed] [Google Scholar]
  • 16.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, Oh B, Kim HL. A large-scale genome-wide association study of Asian populations uncovers genetic factors influencing eight quantitative traits. Nat Genet 41: 527–534, 2009 [DOI] [PubMed] [Google Scholar]
  • 17.Cicila GT, Rapp JP, Wang JM, St Lezin E, Ng SC, Kurtz TW. Linkage of 11 beta-hydroxylase mutations with altered steroid biosynthesis and blood pressure in the Dahl rat. Nat Genet 3: 346–353, 1993 [DOI] [PubMed] [Google Scholar]
  • 18.Crespo K, Chauvet C, Blain M, Menard A, Roy J, Deng AY. Normotension in Lewis and Dahl salt-resistant rats is governed by different genes. J Hypertens 29: 460–465, 2011 [DOI] [PubMed] [Google Scholar]
  • 19.Cusi D, Barlassina C, Azzani T, Casari G, Citterio L, Devoto M, Glorioso N, Lanzani C, Manunta P, Righetti M, Rivera R, Stella P, Troffa C, Zagato L, Bianchi G. Polymorphisms of alpha-adducin and salt sensitivity in patients with essential hypertension. Lancet 349: 1353–1357, 1997 [DOI] [PubMed] [Google Scholar]
  • 20.Dahl LK, Heine M. Primary role of renal homografts in setting chronic blood pressure levels in rats. Circ Res 36: 692–696, 1975 [DOI] [PubMed] [Google Scholar]
  • 21.Das M, Pal S, Ghosh A. Angiotensin converting enzyme gene polymorphism (insertion/deletion) and hypertension in adult Asian Indians: a population-based study from Calcutta, India. Hum Biol 80: 303–312, 2008 [DOI] [PubMed] [Google Scholar]
  • 22.Delles C, McBride MW, Graham D, Padmanabhan S, Dominiczak AF. Genetics of hypertension: from experimental animals to humans. Biochim Biophys Acta 1802: 1299–1308, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Delles C, Padmanabhan S, Lee WK, Miller WH, McBride MW, McClure JD, Brain NJ, Wallace C, Marcano AC, Schmieder RE, Brown MJ, Caulfield MJ, Munroe PB, Farrall M, Webster J, Connell JM, Dominiczak AF. Glutathione S-transferase variants and hypertension. J Hypertens 26: 1343–1352, 2008 [DOI] [PubMed] [Google Scholar]
  • 24.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 3: e196, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Dickson SP, Wang K, Krantz I, Hakonarson H, Goldstein DB. Rare variants create synthetic genome-wide associations. PLoS Biol 8: e1000294, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Doria A, Onuma T, Gearin G, Freire MB, Warram JH, Krolewski AS. Angiotensinogen polymorphism M235T, hypertension, and nephropathy in insulin-dependent diabetes. Hypertension 27: 1134–1139, 1996 [DOI] [PubMed] [Google Scholar]
  • 27.Dunham I, Kundaje A, Aldred SF, Collins PJ, Davis CA, Doyle F, Epstein CB, Frietze S, Harrow J, Kaul R, Khatun J, Lajoie BR, Landt SG, Lee BK, Pauli F, Rosenbloom KR, Sabo P, Safi A, Sanyal A, Shoresh N, Simon JM, Song L, Trinklein ND, Altshuler RC, Birney E, Brown JB, Cheng C, Djebali S, Dong X, Ernst J, Furey TS, Gerstein M, Giardine B, Greven M, Hardison RC, Harris RS, Herrero J, Hoffman MM, Iyer S, Kelllis M, Kheradpour P, Lassmann T, Li Q, Lin X, Marinov GK, Merkel A, Mortazavi A, Parker SC, Reddy TE, Rozowsky J, Schlesinger F, Thurman RE, Wang J, Ward LD, Whitfield TW, Wilder SP, Wu W, Xi HS, Yip KY, Zhuang J, Bernstein BE, Green ED, Gunter C, Snyder M, Pazin MJ, Lowdon RF, Dillon LA, Adams LB, Kelly CJ, Zhang J, Wexler JR, Good PJ, Feingold EA, Crawford GE, Dekker J, Elinitski L, Farnham PJ, Giddings MC, Gingeras TR, Guigo R, Hubbard TJ, Kellis M, Kent WJ, Lieb JD, Margulies EH, Myers RM, Starnatoyannopoulos JA, Tennebaum SA, Weng Z, White KP, Wold B, Yu Y, Wrobel J, Risk BA, Gunawardena HP, Kuiper HC, Maier CW, Xie L, Chen X, Mikkelsen TS, Gillespie S, Goren A, Ram O, Zhang X, Wang L, Issner R, Coyne MJ, Durham T, Ku M, Truong T, Eaton ML, Dobin A, Tanzer A, Lagarde J, Lin W, Xue C, Williams BA, Zaleski C, Roder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Batut P, Bell I, Bell K, Chakrabortty S, Chrast J, Curado J, Derrien T, Drenkow J, Dumais E, Dumais J, Duttagupta R, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Howald C, Jha S, Johnson R, Kapranov P, King B, Kingswood C, Li G, Luo OJ, Park E, Preall JB, Presaud K, Ribeca P, Robyr D, Ruan X, Sammeth M, Sandu KS, Schaeffer L, See LH, Shahab A, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Trout D, Walters N, Wang H, Hayashizaki Y, Reymond A, Antonarakis SE, Hannon GJ, Ruan Y, Carninci P, Sloan CA, Learned K, Malladi VS, Wong MC, Barber GP, Cline MS, Dreszer TR, Heitner SG, Karolchik D, Kirkup VM, Meyer LR, Long JC, Maddren M, Raney BJ, Grasfeder LL, Giresi PG, Battenhouse A, Sheffield NC, Showers KA, London D, Bhinge AA, Shestak C, Schaner MR, Kim SK, Zhang ZZ, Mieczkowski PA, Mieczkowska JO, Liu Z, McDaniell RM, Ni Y, Rashid NU, Kim MJ, Adar S, Zhang Z, Wang T, Winter D, Keefe D, Iyer VR, Sandhu KS, Zheng M, Wang P, Gertz J, Vielmetter J, Partridge EC, Varley KE, Gasper C, Bansal A, Pepke S, Jain P, Amrhein H, Bowling KM, Anaya M, Cross MK, Muratet MA, Newberry KM, McCue K, Nesmith AS, Fisher-Aylor KI, Pusey B, DeSalvo G, Parker SL, Balasubramanian S, Davis NS, Meadows SK, Eggleston T, Newberry JS, Levy SE, Absher DM, Wong WH, Blow MJ, Visel A, Pennachio LA, Elnitski L, Petrykowska HM, Abyzov A, Aken B, Barrell D, Barson G, Berry A, Bignell A, Boychenko V, Bussotti G, Davidson C, Despacio-Reyes G, Diekhans M, Ezkurdia I, Frankish A, Gilbert J, Gonzalez JM, Griffiths E, Harte R, Hendrix DA, Hunt T, Jungreis I, Kay M, Khurana E, Leng J, Lin MF, Loveland J, Lu Z, Manthravadi D, Mariotti M, Mudge J, Mukherjee G, Notredame C, Pei B, Rodriguez JM, Saunders G, Sboner A, Searle S, Sisu C, Snow C, Steward C, Tapanari E, Tress ML, van Baren MJ, Washieti S, Wilming L, Zadissa A, Zhengdong Z, Brent M, Haussler D, Valencia A, Raymond A, Addleman N, Alexander RP, Auerbach RK, Bettinger K, Bhardwaj N, Boyle AP, Cao AR, Cayting P, Charos A, Cheng Y, Eastman C, Euskirchen G, Fleming JD, Grubert F, Habegger L, Hariharan M, Harmanci A, Iyenger S, Jin VX, Karczewski KJ, Kasowski M, Lacroute P, Lam H, Larnarre-Vincent N, Lian J, Lindahl-Allen M, Min R, Miotto B, Monahan H, Moqtaderi Z, Mu XJ, O'Geen H, Ouyang Z, Patacsil D, Raha D, Ramirez L, Reed B, Shi M, Slifer T, Witt H, Wu L, Xu X, Yan KK, Yang X, Struhl K, Weissman SM, Tenebaum SA, Penalva LO, Karmakar S, Bhanvadia RR, Choudhury A, Domanus M, Ma L, Moran J, Victorsen A, Auer T, Centarin L, Eichenlaub M, Gruhl F, Heerman S, Hoeckendorf B, Inoue D, Kellner T, Kirchmaier S, Mueller C, Reinhardt R, Schertel L, Schneider S, Sinn R, Wittbrodt B, Wittbrodt J, Jain G, Balasundaram G, Bates DL, Byron R, Canfield TK, Diegel MJ, Dunn D, Ebersol AK, Frum T, Garg K, Gist E, Hansen RS, Boatman L, Haugen E, Humbert R, Johnson AK, Johnson EM, Kutyavin TM, Lee K, Lotakis D, Maurano MT, Neph SJ, Neri FV, Nguyen ED, Qu H, Reynolds AP, Roach V, Rynes E, Sanchez ME, Sandstrom RS, Shafer AO, Stergachis AB, Thomas S, Vernot B, Vierstra J, Vong S, Weaver MA, Yan Y, Zhang M, Akey JA, Bender M, Dorschner MO, Groudine M, MacCoss MJ, Navas P, Stamatoyannopoulos G, Stamatoyannopoulos JA, Beal K, Brazma A, Flicek P, Johnson N, Lukk M, Luscombe NM, Sobral D, Vaquerizas JM, Batzoglou S, Sidow A, Hussami N, Kyriazopoulou-Panagiotopoulou S, Libbrecht MW, Schaub MA, Miller W, Bickel PJ, Banfai B, Boley NP, Huang H, Li JJ, Noble WS, Bilmes JA, Buske OJ, Sahu AO, Kharchenko PV, Park PJ, Baker D, Taylor J, Lochovsky L. An integrated encyclopedia of DNA elements in the human genome. Nature 489: 57–74, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang SJ, Pihur V, Vollenweider P, O'Reilly PF, Amin N, Bragg-Gresham JL, Teumer A, Glazer NL, Launer L, Zhao JH, Aulchenko Y, Heath S, Sober S, Parsa A, Luan J, Arora P, Dehghan A, Zhang F, Lucas G, Hicks AA, Jackson AU, Peden JF, Tanaka T, Wild SH, Rudan I, Igl W, Milaneschi Y, Parker AN, Fava C, Chambers JC, Fox ER, Kumari M, Go MJ, van der Harst P, Kao WH, Sjogren M, Vinay DG, Alexander M, Tabara Y, Shaw-Hawkins S, Whincup PH, Liu Y, Shi G, Kuusisto J, Tayo B, Seielstad M, Sim X, Nguyen KD, Lehtimaki T, Matullo G, Wu Y, Gaunt TR, Onland-Moret NC, Cooper MN, Platou CG, Org E, Hardy R, Dahgam S, Palmen J, Vitart V, Braund PS, Kuznetsova T, Uiterwaal CS, Adeyemo A, Palmas W, Campbell H, Ludwig B, Tomaszewski M, Tzoulaki I, Palmer ND, Aspelund T, Garcia M, Chang YP, O'Connell JR, Steinle NI, Grobbee DE, Arking DE, Kardia SL, Morrison AC, Hernandez D, Najjar S, McArdle WL, Hadley D, Brown MJ, Connell JM, Hingorani AD, Day IN, Lawlor DA, Beilby JP, Lawrence RW, Clarke R, Hopewell JC, Ongen H, Dreisbach AW, Li Y, Young JH, Bis JC, Kahonen M, Viikari J, Adair LS, Lee NR, Chen MH, Olden M, Pattaro C, Bolton JA, Kottgen A, Bergmann S, Mooser V, Chaturvedi N, Frayling TM, Islam M, Jafar TH, Erdmann J, Kulkarni SR, Bornstein SR, Grassler J, Groop L, Voight BF, Kettunen J, Howard P, Taylor A, Guarrera S, Ricceri F, Emilsson V, Plump A, Barroso I, Khaw KT, Weder AB, Hunt SC, Sun YV, Bergman RN, Collins FS, Bonnycastle LL, Scott LJ, Stringham HM, Peltonen L, Perola M, Vartiainen E, Brand SM, Staessen JA, Wang TJ, Burton PR, Soler Artigas M, Dong Y, Snieder H, Wang X, Zhu H, Lohman KK, Rudock ME, Heckbert SR, Smith NL, Wiggins KL, Doumatey A, Shriner D, Veldre G, Viigimaa M, Kinra S, Prabhakaran D, Tripathy V, Langefeld CD, Rosengren A, Thelle DS, Corsi AM, Singleton A, Forrester T, Hilton G, McKenzie CA, Salako T, Iwai N, Kita Y, Ogihara T, Ohkubo T, Okamura T, Ueshima H, Umemura S, Eyheramendy S, Meitinger T, Wichmann HE, Cho YS, Kim HL, Lee JY, Scott J, Sehmi JS, Zhang W, Hedblad B, Nilsson P, Smith GD, Wong A, Narisu N, Stancakova A, Raffel LJ, Yao J, Kathiresan S, O'Donnell CJ, Schwartz SM, Ikram MA, Longstreth WT, Jr, Mosley TH, Seshadri S, Shrine NR, Wain LV, Morken MA, Swift AJ, Laitinen J, Prokopenko I, Zitting P, Cooper JA, Humphries SE, Danesh J, Rasheed A, Goel A, Hamsten A, Watkins H, Bakker SJ, van Gilst WH, Janipalli CS, Mani KR, Yajnik CS, Hofman A, Mattace-Raso FU, Oostra BA, Demirkan A, Isaacs A, Rivadeneira F, Lakatta EG, Orru M, Scuteri A, Ala-Korpela M, Kangas AJ, Lyytikainen LP, Soininen P, Tukiainen T, Wurtz P, Ong RT, Dorr M, Kroemer HK, Volker U, Volzke H, Galan P, Hercberg S, Lathrop M, Zelenika D, Deloukas P, Mangino M, Spector TD, Zhai G, Meschia JF, Nalls MA, Sharma P, Terzic J, Kumar MV, Denniff M, Zukowska-Szczechowska E, Wagenknecht LE, Fowkes FG, Charchar FJ, Schwarz PE, Hayward C, Guo X, Rotimi C, Bots ML, Brand E, Samani NJ, Polasek O, Talmud PJ, Nyberg F, Kuh D, Laan M, Hveem K, Palmer LJ, van der Schouw YT, Casas JP, Mohlke KL, Vineis P, Raitakari O, Ganesh SK, Wong TY, Tai ES, Cooper RS, Laakso M, Rao DC, Harris TB, Morris RW, Dominiczak AF, Kivimaki M, Marmot MG, Miki T, Saleheen D, Chandak GR, Coresh J, Navis G, Salomaa V, Han BG, Zhu X, Kooner JS, Melander O, Ridker PM, Bandinelli S, Gyllensten UB, Wright AF, Wilson JF, Ferrucci L, Farrall M, Tuomilehto J, Pramstaller PP, Elosua R, Soranzo N, Sijbrands EJ, Altshuler D, Loos RJ, Shuldiner AR, Gieger C, Meneton P, Uitterlinden AG, Wareham NJ, Gudnason V, Rotter JI, Rettig R, Uda M, Strachan DP, Witteman JC, Hartikainen AL, Beckmann JS, Boerwinkle E, Vasan RS, Boehnke M, Larson MG, Jarvelin MR, Psaty BM, Abecasis GR, Chakravarti A, Elliott P, van Duijn CM, Newton-Cheh C, Levy D, Caulfield MJ, Johnson T. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature 478: 103–109, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ellis PD, Chen Q, Barker PJ, Metcalfe JC, Kemp PR. Nov gene encodes adhesion factor for vascular smooth muscle cells and is dynamically regulated in response to vascular injury. Arterioscler Thromb Vasc Biol 20: 1912–1919, 2000 [DOI] [PubMed] [Google Scholar]
  • 30.Feldman HI, Appel LJ, Chertow GM, Cifelli D, Cizman B, Daugirdas J, Fink JC, Franklin-Becker ED, Go AS, Hamm LL, He J, Hostetter T, Hsu CY, Jamerson K, Joffe M, Kusek JW, Landis JR, Lash JP, Miller ER, Mohler ER, 3rd, Muntner P, Ojo AO, Rahman M, Townsend RR, Wright JT. The Chronic Renal Insufficiency Cohort (CRIC) Study: Design and Methods. J Am Soc Nephrol 14: S148–S153, 2003 [DOI] [PubMed] [Google Scholar]
  • 31.Ferrandi M, Salardi S, Tripodi G, Barassi P, Rivera R, Manunta P, Goldshleger R, Ferrari P, Bianchi G, Karlish SJ. Evidence for an interaction between adducin and Na+-K+-ATPase: relation to genetic hypertension. Am J Physiol Heart Circ Physiol 277: H1338–H1349, 1999 [DOI] [PubMed] [Google Scholar]
  • 32.Fox ER, Young JH, Li Y, Dreisbach AW, Keating BJ, Musani SK, Liu K, Morrison AC, Ganesh S, Kutlar A, Ramachandran VS, Polak JF, Fabsitz RR, Dries DL, Farlow DN, Redline S, Adeyemo A, Hirschorn JN, Sun YV, Wyatt SB, Penman AD, Palmas W, Rotter JI, Townsend RR, Doumatey AP, Tayo BO, Mosley TH, Jr, Lyon HN, Kang SJ, Rotimi CN, Cooper RS, Franceschini N, Curb JD, Martin LW, Eaton CB, Kardia SL, Taylor HA, Caulfield MJ, Ehret GB, Johnson T, Chakravarti A, Zhu X, Levy D. Association of genetic variation with systolic and diastolic blood pressure among African Americans: the Candidate Gene Association Resource study. Hum Mol Genet 20: 2273–2284, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Franceschini N, Fox E, Zhang Z, Edwards TL, Nalls MA, Sung YJ, Tayo BO, Sun YV, Gottesman O, Adeyemo A, Johnson AD, Young JH, Rice K, Duan Q, Chen F, Li Y, Tang H, Fornage M, Keene KL, Andrews JS, Smith JA, Faul JD, Guangfa Z, Guo W, Liu Y, Murray SS, Musani SK, Srinivasan S, Velez Edwards DR, Wang H, Becker LC, Bovet P, Bochud M, Broeckel U, Burnier M, Carty C, Chasman DI, Ehret G, Chen WM, Chen G, Chen W, Ding J, Dreisbach AW, Evans MK, Guo X, Garcia ME, Jensen R, Keller MF, Lettre G, Lotay V, Martin LW, Moore JH, Morrison AC, Mosley TH, Ogunniyi A, Palmas W, Papanicolaou G, Penman A, Polak JF, Ridker PM, Salako B, Singleton AB, Shriner D, Taylor KD, Vasan R, Wiggins K, Williams SM, Yanek LR, Zhao W, Zonderman AB, Becker DM, Berenson G, Boerwinkle E, Bottinger E, Cushman M, Eaton C, Nyberg F, Heiss G, Hirschhron JN, Howard VJ, Karczewsk KJ, Lanktree MB, Liu K, Loos R, Margolis K, Snyder M, Psaty BM, Schork NJ, Weir DR, Rotimi CN, Sale MM, Harris T, Kardia SL, Hunt SC, Arnett D, Redline S, Cooper RS, Risch NJ, Rao DC, Rotter JI, Chakravarti A, Reiner AP, Levy D, Keating BJ, Zhu X. Genome-wide association analysis of blood-pressure traits in African-ancestry individuals reveals common associated genes in African and non-African populations. Am J Hum Genet 93: 545–554, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Frazer KA, Ballinger DG, Cox DR, Hinds DA, Stuve LL, Gibbs RA, Belmont JW, Boudreau A, Hardenbol P, Leal SM, Pasternak S, Wheeler DA, Willis TD, Yu F, Yang H, Zeng C, Gao Y, Hu H, Hu W, Li C, Lin W, Liu S, Pan H, Tang X, Wang J, Wang W, Yu J, Zhang B, Zhang Q, Zhao H, Zhou J, Gabriel SB, Barry R, Blumenstiel B, Camargo A, Defelice M, Faggart M, Goyette M, Gupta S, Moore J, Nguyen H, Onofrio RC, Parkin M, Roy J, Stahl E, Winchester E, Ziaugra L, Altshuler D, Shen Y, Yao Z, Huang W, Chu X, He Y, Jin L, Liu Y, Sun W, Wang H, Wang Y, Xiong X, Xu L, Waye MM, Tsui SK, Xue H, Wong JT, Galver LM, Fan JB, Gunderson K, Murray SS, Oliphant AR, Chee MS, Montpetit A, Chagnon F, Ferretti V, Leboeuf M, Olivier JF, Phillips MS, Roumy S, Sallee C, Verner A, Hudson TJ, Kwok PY, Cai D, Koboldt DC, Miller RD, Pawlikowska L, Taillon-Miller P, Xiao M, Tsui LC, Mak W, Song YQ, Tam PK, Nakamura Y, Kawaguchi T, Kitamoto T, Morizono T, Nagashima A, Ohnishi Y, Sekine A, Tanaka T, Tsunoda T, Deloukas P, Bird CP, Delgado M, Dermitzakis ET, Gwilliam R, Hunt S, Morrison J, Powell D, Stranger BE, Whittaker P, Bentley DR, Daly MJ, de Bakker PI, Barrett J, Chretien YR, Maller J, McCarroll S, Patterson N, Pe'er I, Price A, Purcell S, Richter DJ, Sabeti P, Saxena R, Schaffner SF, Sham PC, Varilly P, Stein LD, Krishnan L, Smith AV, Tello-Ruiz MK, Thorisson GA, Chakravarti A, Chen PE, Cutler DJ, Kashuk CS, Lin S, Abecasis GR, Guan W, Li Y, Munro HM, Qin ZS, Thomas DJ, McVean G, Auton A, Bottolo L, Cardin N, Eyheramendy S, Freeman C, Marchini J, Myers S, Spencer C, Stephens M, Donnelly P, Cardon LR, Clarke G, Evans DM, Morris AP, Weir BS, Mullikin JC, Sherry ST, Feolo M, Skol A, Zhang H, Matsuda I, Fukushima Y, Macer DR, Suda E, Rotimi CN, Adebamowo CA, Ajayi I, Aniagwu T, Marshall PA, Nkwodimmah C, Royal CD, Leppert MF, Dixon M, Peiffer A, Qiu R, Kent A, Kato K, Niikawa N, Adewole IF, Knoppers BM, Foster MW, Clayton EW, Watkin J, Muzny D, Nazareth L, Sodergren E, Weinstock GM, Yakub I, Birren BW, Wilson RK, Fulton LL, Rogers J, Burton J, Carter NP, Clee CM, Griffiths M, Jones MC, McLay K, Plumb RW, Ross MT, Sims SK, Willey DL, Chen Z, Han H, Kang L, Godbout M, Wallenburg JC, L'Archeveque P, Bellemare G, Saeki K, An D, Fu H, Li Q, Wang Z, Wang R, Holden AL, Brooks LD, McEwen JE, Guyer MS, Wang VO, Peterson JL, Shi M, Spiegel J, Sung LM, Zacharia LF, Collins FS, Kennedy K, Jamieson R, Stewart J. A second generation human haplotype map of over 3.1 million SNPs. Nature 449: 851–861, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Freedman ML, Reich D, Penney KL, McDonald GJ, Mignault AA, Patterson N, Gabriel SB, Topol EJ, Smoller JW, Pato CN, Pato MT, Petryshen TL, Kolonel LN, Lander ES, Sklar P, Henderson B, Hirschhorn JN, Altshuler D. Assessing the impact of population stratification on genetic association studies. Nat Genet 36: 388–393, 2004 [DOI] [PubMed] [Google Scholar]
  • 36.Friebe A, Mergia E, Dangel O, Lange A, Koesling D. Fatal gastrointestinal obstruction and hypertension in mice lacking nitric oxide-sensitive guanylyl cyclase. Proc Natl Acad Sci USA 104: 7699–7704, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ganesh SK, Tragante V, Guo W, Guo Y, Lanktree MB, Smith EN, Johnson T, Castillo BA, Barnard J, Baumert J, Chang YP, Elbers CC, Farrall M, Fischer ME, Franceschini N, Gaunt TR, Gho JM, Gieger C, Gong Y, Isaacs A, Kleber ME, Mateo Leach I, McDonough CW, Meijs MF, Mellander O, Molony CM, Nolte IM, Padmanabhan S, Price TS, Rajagopalan R, Shaffer J, Shah S, Shen H, Soranzo N, van der Most PJ, Van Iperen EP, Van Setten JA, Vonk JM, Zhang L, Beitelshees AL, Berenson GS, Bhatt DL, Boer JM, Boerwinkle E, Burkley B, Burt A, Chakravarti A, Chen W, Cooper-Dehoff RM, Curtis SP, Dreisbach A, Duggan D, Ehret GB, Fabsitz RR, Fornage M, Fox E, Furlong CE, Gansevoort RT, Hofker MH, Hovingh GK, Kirkland SA, Kottke-Marchant K, Kutlar A, Lacroix AZ, Langaee TY, Li YR, Lin H, Liu K, Maiwald S, Malik R, Murugesan G, Newton-Cheh C, O'Connell JR, Onland-Moret NC, Ouwehand WH, Palmas W, Penninx BW, Pepine CJ, Pettinger M, Polak JF, Ramachandran VS, Ranchalis J, Redline S, Ridker PM, Rose LM, Scharnag H, Schork NJ, Shimbo D, Shuldiner AR, Srinivasan SR, Stolk RP, Taylor HA, Thorand B, Trip MD, van Duijn CM, Verschuren WM, Wijmenga C, Winkelmann BR, Wyatt S, Young JH, Boehm BO, Caulfield MJ, Chasman DI, Davidson KW, Doevendans PA, Fitzgerald GA, Gums JG, Hakonarson H, Hillege HL, Illig T, Jarvik GP, Johnson JA, Kastelein JJ, Koenig W, Marz W, Mitchell BD, Murray SS, Oldehinkel AJ, Rader DJ, Reilly MP, Reiner AP, Schadt EE, Silverstein RL, Snieder H, Stanton AV, Uitterlinden AG, van der Harst P, van der Schouw YT, Samani NJ, Johnson AD, Munroe PB, de Bakker PI, Zhu X, Levy D, Keating BJ, Asselbergs FW. Loci influencing blood pressure identified using a cardiovascular gene-centric array. Hum Mol Genet 22: 1663–1678, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Garrett MR, Rapp JP. Defining the blood pressure QTL on chromosome 7 in Dahl rats by a 177-kb congenic segment containing Cyp11b1. Mamm Genome 14: 268–273, 2003 [DOI] [PubMed] [Google Scholar]
  • 39.Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Magid D, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER, Moy CS, Mussolino ME, Nichol G, Paynter NP, Schreiner PJ, Sorlie PD, Stein J, Turan TN, Virani SS, Wong ND, Woo D, Turner MB. Executive summary: heart disease and stroke statistics—2013 update: a report from the American Heart Association. Circulation 127: 143–152, 2013 [DOI] [PubMed] [Google Scholar]
  • 40.Gu XX, Spaepen M, Guo C, Fagard R, Amery A, Lijnen P, Cassiman JJ. Lack of association between the I/D polymorphism of the angiotensin-converting enzyme gene and essential hypertension in a Belgian population. J Hum Hypertens 8: 683–685, 1994 [PubMed] [Google Scholar]
  • 41.Guyton AC. Long-term arterial pressure control: an analysis from animal experiments and computer and graphic models. Am J Physiol Regul Integr Comp Physiol 259: R865–R877, 1990 [DOI] [PubMed] [Google Scholar]
  • 42.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 52: 584–594, 1972 [DOI] [PubMed] [Google Scholar]
  • 43.Hajjar I, Kotchen TA. Trends in prevalence, awareness, treatment, and control of hypertension in the United States, 1988–2000. JAMA 290: 199–206, 2003 [DOI] [PubMed] [Google Scholar]
  • 44.Hall JE. Historical perspective of the renin-angiotensin system. Mol Biotechnol 24: 27–39, 2003 [DOI] [PubMed] [Google Scholar]
  • 45.Hindorff LA, MacArthur J, Morales J, Junkins HA, Hall PN, Klemm AK, Manolio TA. A Catalog of Published Genome-Wide Association Studies. [Online]. www.genome.gov/gwastudies [06/September/2013].
  • 46.Ho JE, Levy D, Rose L, Johnson AD, Ridker PM, Chasman DI. Discovery and replication of novel blood pressure genetic loci in the Women's Genome Health Study. J Hypertens 29: 62–69, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hopkins PN, Hunt SC. Genetics of hypertension. Genet Med 5: 413–429, 2003 [DOI] [PubMed] [Google Scholar]
  • 48.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 165: 923–928, 2005 [DOI] [PubMed] [Google Scholar]
  • 49.Huyghe JR, Jackson AU, Fogarty MP, Buchkovich ML, Stancakova A, Stringham HM, Sim X, Yang L, Fuchsberger C, Cederberg H, Chines PS, Teslovich TM, Romm JM, Ling H, McMullen I, Ingersoll R, Pugh EW, Doheny KF, Neale BM, Daly MJ, Kuusisto J, Scott LJ, Kang HM, Collins FS, Abecasis GR, Watanabe RM, Boehnke M, Laakso M, Mohlke KL. Exome array analysis identifies new loci and low-frequency variants influencing insulin processing and secretion. Nat Genet 45: 197–201, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Inoue I, Nakajima T, Williams CS, Quackenbush J, Puryear R, Powers M, Cheng T, Ludwig EH, Sharma AM, Hata A, Jeunemaitre X, Lalouel JM. A nucleotide substitution in the promoter of human angiotensinogen is associated with essential hypertension and affects basal transcription in vitro. J Clin Invest 99: 1786–1797, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Izzo JL., Jr The sympathoadrenal system in the maintenance of elevated arterial pressure. J Cardiovasc Pharmacol 6, Suppl 3: S514–S521, 1984 [PubMed] [Google Scholar]
  • 52.Jeffs B, Negrin CD, Graham D, Clark JS, Anderson NH, Gauguier D, Dominiczak AF. Applicability of a “speed” congenic strategy to dissect blood pressure quantitative trait loci on rat chromosome 2. Hypertension 35: 179–187, 2000 [DOI] [PubMed] [Google Scholar]
  • 53.Jeunemaitre X, Soubrier F, Kotelevtsev YV, Lifton RP, Williams CS, Charru A, Hunt SC, Hopkins PN, Williams RR, Lalouel JM, Corvol P. Molecular basis of human hypertension: role of angiotensinogen. Cell 71: 169–180, 1992 [DOI] [PubMed] [Google Scholar]
  • 54.Ji LD, Zhang LN, Shen P, Wang P, Zhang YM, Xing WH, Xu J. Association of angiotensinogen gene M235T and angiotensin-converting enzyme gene I/D polymorphisms with essential hypertension in Han Chinese population: a meta-analysis. J Hypertens 28: 419–428, 2010 [DOI] [PubMed] [Google Scholar]
  • 55.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 40: 592–599, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Johnson T, Gaunt TR, Newhouse SJ, Padmanabhan S, Tomaszewski M, Kumari M, Morris RW, Tzoulaki I, O'Brien ET, Poulter NR, Sever P, Shields DC, Thom S, Wannamethee SG, Whincup PH, Brown MJ, Connell JM, Dobson RJ, Howard PJ, Mein CA, Onipinla A, Shaw-Hawkins S, Zhang Y, Davey Smith G, Day IN, Lawlor DA, Goodall AH, Fowkes FG, Abecasis GR, Elliott P, Gateva V, Braund PS, Burton PR, Nelson CP, Tobin MD, van der Harst P, Glorioso N, Neuvrith H, Salvi E, Staessen JA, Stucchi A, Devos N, Jeunemaitre X, Plouin PF, Tichet J, Juhanson P, Org E, Putku M, Sober S, Veldre G, Viigimaa M, Levinsson A, Rosengren A, Thelle DS, Hastie CE, Hedner T, Lee WK, Melander O, Wahlstrand B, Hardy R, Wong A, Cooper JA, Palmen J, Chen L, Stewart AF, Wells GA, Westra HJ, Wolfs MG, Clarke R, Franzosi MG, Goel A, Hamsten A, Lathrop M, Peden JF, Seedorf U, Watkins H, Ouwehand WH, Sambrook J, Stephens J, Casas JP, Drenos F, Holmes MV, Kivimaki M, Shah S, Shah T, Talmud PJ, Whittaker J, Wallace C, Delles C, Laan M, Kuh D, Humphries SE, Nyberg F, Cusi D, Roberts R, Newton-Cheh C, Franke L, Stanton AV, Dominiczak AF, Farrall M, Hingorani AD, Samani NJ, Caulfield MJ, Munroe PB. Blood pressure loci identified with a gene-centric array. Am J Hum Genet 89: 688–700, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kahle KT, Ring AM, Lifton RP. Molecular physiology of the WNK kinases. Annu Rev Physiol 70: 329–355, 2008 [DOI] [PubMed] [Google Scholar]
  • 58.Kaiser HW, O'Keefe E, Bennett V. Adducin: Ca++-dependent association with sites of cell-cell contact. J Cell Biol 109: 557–569, 1989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Kato N, Takeuchi F, Tabara Y, Kelly TN, Go MJ, Sim X, Tay WT, Chen CH, Zhang Y, Yamamoto K, Katsuya T, Yokota M, Kim YJ, Ong RT, Nabika T, Gu D, Chang LC, Kokubo Y, Huang W, Ohnaka K, Yamori Y, Nakashima E, Jaquish CE, Lee JY, Seielstad M, Isono M, Hixson JE, Chen YT, Miki T, Zhou X, Sugiyama T, Jeon JP, Liu JJ, Takayanagi R, Kim SS, Aung T, Sung YJ, Zhang X, Wong TY, Han BG, Kobayashi S, Ogihara T, Zhu D, Iwai N, Wu JY, Teo YY, Tai ES, Cho YS, He J. Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet 43: 531–538, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kent JW., Jr Rare variants, common markers: synthetic association and beyond. Genet Epidemiol 35, Suppl 1: S80–S84, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kim HS, Krege JH, Kluckman KD, Hagaman JR, Hodgin JB, Best CF, Jennette JC, Coffman TM, Maeda N, Smithies O. Genetic control of blood pressure and the angiotensinogen locus. Proc Natl Acad Sci USA 92: 2735–2739, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kobayashi Y, Hirawa N, Tabara Y, Muraoka H, Fujita M, Miyazaki N, Fujiwara A, Ichikawa Y, Yamamoto Y, Ichihara N, Saka S, Wakui H, Yoshida S, Yatsu K, Toya Y, Yasuda G, Kohara K, Kita Y, Takei K, Goshima Y, Ishikawa Y, Ueshima H, Miki T, Umemura S. Mice lacking hypertension candidate gene ATP2B1 in vascular smooth muscle cells show significant blood pressure elevation. Hypertension 59: 854–860, 2012 [DOI] [PubMed] [Google Scholar]
  • 63.Kryukov GV, Pennacchio LA, Sunyaev SR. Most rare missense alleles are deleterious in humans: implications for complex disease and association studies. Am J Hum Genet 80: 727–739, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lawes CM, Velez EdwardsVander Hoorn S, Rodgers A. Global burden of blood-pressure-related disease, 2001. Lancet 371: 1513–1518, 2008 [DOI] [PubMed] [Google Scholar]
  • 65.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 41: 677–687, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.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 41: 677–687, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.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 360: 1903–1913, 2002 [DOI] [PubMed] [Google Scholar]
  • 68.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 355: 262–265, 1992 [DOI] [PubMed] [Google Scholar]
  • 69.Lifton RP, Dluhy RG, Powers M, Rich GM, Gutkin M, Fallo F, Gill JR, Jr, Feld L, Ganguly A, Laidlaw JC, Murnaghan DJ, Kaufman C, Stockigt JR, Ulick S, Lalouel JM. Hereditary hypertension caused by chimaeric gene duplications and ectopic expression of aldosterone synthase. Nat Genet 2: 66–74, 1992 [DOI] [PubMed] [Google Scholar]
  • 70.Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hypertension. Cell 104: 545–556, 2001 [DOI] [PubMed] [Google Scholar]
  • 71.Liu EY, Buyske S, Aragaki AK, Peters U, Boerwinkle E, Carlson C, Carty C, Crawford DC, Haessler J, Hindorff LA, Marchand LL, Manolio TA, Matise T, Wang W, Kooperberg C, North KE, Li Y. Genotype imputation of Metabochip SNPs using a study-specific reference panel of ∼4,000 haplotypes in African Americans from the Women's Health Initiative. Genet Epidemiol 36: 107–117, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lloyd-Jones DM, Hong Y, Labarthe D, Mozaffarian D, Appel LJ, Van Horn L, Greenlund K, Daniels S, Nichol G, Tomaselli GF, Arnett DK, Fonarow GC, Ho PM, Lauer MS, Masoudi FA, Robertson RM, Roger V, Schwamm LH, Sorlie P, Yancy CW, Rosamond WD. Defining and setting national goals for cardiovascular health promotion and disease reduction: the American Heart Association's strategic Impact Goal through 2020 and beyond. Circulation 121: 586–613, 2010 [DOI] [PubMed] [Google Scholar]
  • 73.Lupski JR, Belmont JW, Boerwinkle E, Gibbs RA. Clan genomics and the complex architecture of human disease. Cell 147: 32–43, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Marth GT, Yu F, Indap AR, Garimella K, Gravel S, Leong WF, Tyler-Smith C, Bainbridge M, Blackwell T, Zheng-Bradley X, Chen Y, Challis D, Clarke L, Ball EV, Cibulskis K, Cooper DN, Fulton B, Hartl C, Koboldt D, Muzny D, Smith R, Sougnez C, Stewart C, Ward A, Yu J, Xue Y, Altshuler D, Bustamante CD, Clark AG, Daly M, DePristo M, Flicek P, Gabriel S, Mardis E, Palotie A, Gibbs R. The functional spectrum of low-frequency coding variation. Genome Biol 12: R84, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.McBride MW, Brosnan MJ, Mathers J, McLellan LI, Miller WH, Graham D, Hanlon N, Hamilton CA, Polke JM, Lee WK, Dominiczak AF. Reduction of Gstm1 expression in the stroke-prone spontaneously hypertension rat contributes to increased oxidative stress. Hypertension 45: 786–792, 2005 [DOI] [PubMed] [Google Scholar]
  • 76.Mondry A, Loh M, Liu P, Zhu A, Nagel M. Polymorphisms of the insertion/deletion ACE and M235T AGT genes and hypertension: surprising new findings and meta-analysis of data. Bio Med Central Nephrol 6: 1, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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, Seedorf U, Syvanen AC, Tognoni G, Lakatta EG, Sanna S, Scheet P, Schlessinger D, Scuteri A, Dorr M, Ernst F, Felix SB, Homuth G, Lorbeer R, Reffelmann T, Rettig R, Volker U, Galan P, Gut IG, Hercberg S, Lathrop GM, Zelenika D, Deloukas P, Soranzo N, Williams FM, Zhai G, Salomaa V, Laakso M, Elosua R, Forouhi NG, Volzke H, Uiterwaal CS, van der Schouw YT, Numans ME, Matullo G, Navis G, Berglund G, Bingham SA, Kooner JS, Connell JM, Bandinelli S, Ferrucci L, Watkins H, Spector TD, Tuomilehto J, Altshuler D, Strachan DP, Laan M, Meneton P, Wareham NJ, Uda M, Jarvelin MR, Mooser V, Melander O, Loos RJ, Elliott P, Abecasis GR, Caulfield M, Munroe PB. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet 41: 666–676, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.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 41: 348–353, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ng SB, Turner EH, Robertson PD, Flygare SD, Bigham AW, Lee C, Shaffer T, Wong M, Bhattacharjee A, Eichler EE, Bamshad M, Nickerson DA, Shendure J. Targeted capture and massively parallel sequencing of 12 human exomes. Nature 461: 272–276, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Okamoto K, Aoki K, Nosaka S, Fukushima M. Cardiovascular diseases in the spontaneously hypertensive Rat. Jpn Circ J 28: 943–952, 1964 [DOI] [PubMed] [Google Scholar]
  • 82.Oudit GY, Crackower MA, Eriksson U, Sarao R, Kozieradzki I, Sasaki T, Irie-Sasaki J, Gidrewicz D, Rybin VO, Wada T, Steinberg SF, Backx PH, Penninger JM. Phosphoinositide 3-kinase gamma-deficient mice are protected from isoproterenol-induced heart failure. Circulation 108: 2147–2152, 2003 [DOI] [PubMed] [Google Scholar]
  • 83.Padmanabhan S, Melander O, Johnson T, Di Blasio AM, Lee WK, Gentilini D, Hastie CE, Menni C, Monti MC, Delles C, Laing S, Corso B, Navis G, Kwakernaak AJ, van der Harst P, Bochud M, Maillard M, Burnier M, Hedner T, Kjeldsen S, Wahlstrand B, Sjogren M, Fava C, Montagnana M, Danese E, Torffvit O, Hedblad B, Snieder H, Connell JM, Brown M, Samani NJ, Farrall M, Cesana G, Mancia G, Signorini S, Grassi G, Eyheramendy S, Wichmann HE, Laan M, Strachan DP, Sever P, Shields DC, Stanton A, Vollenweider P, Teumer A, Volzke H, Rettig R, Newton-Cheh C, Arora P, Zhang F, Soranzo N, Spector TD, Lucas G, Kathiresan S, Siscovick DS, Luan J, Loos RJ, Wareham NJ, Penninx BW, Nolte IM, McBride M, Miller WH, Nicklin SA, Baker AH, Graham D, McDonald RA, Pell JP, Sattar N, Welsh P, Munroe P, Caulfield MJ, Zanchetti A, Dominiczak AF. Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet 6: e1001177, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Pravenec M, Kurtz TW. Molecular genetics of experimental hypertension and the metabolic syndrome: from gene pathways to new therapies. Hypertension 49: 941–952, 2007 [DOI] [PubMed] [Google Scholar]
  • 85.Pravenec M, Landa V, Zidek V, Musilova A, Kren V, Kazdova L, Aitman TJ, Glazier AM, Ibrahimi A, Abumrad NA, Qi N, Wang JM, St Lezin EM, Kurtz TW. Transgenic rescue of defective Cd36 ameliorates insulin resistance in spontaneously hypertensive rats. Nat Genet 27: 156–158, 2001 [DOI] [PubMed] [Google Scholar]
  • 86.Pravenec M, Zidek V, Landa V, Viklicky O, Sugimoto K, Qi N, Kurtz TW. Genetic variation in the renal expression of CD36 regulates blood pressure in the sponteneously hypertensive rat. Proceedings of the 21st Scientific Meeting of the International Society of Hypertension Abstract 56, 2006 [Google Scholar]
  • 87.Price AL, Zaitlen NA, Reich D, Patterson N. New approaches to population stratification in genome-wide association studies. Nat Rev Genet 11: 459–463, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ranade K, Jorgenson E, Sheu WH, Pei D, Hsiung CA, Chiang FT, Chen YD, Pratt R, Olshen RA, Curb D, Cox DR, Botstein D, Risch N. A polymorphism in the beta1 adrenergic receptor is associated with resting heart rate. Am J Hum Genet 70: 935–942, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rettig R, Grisk O. The kidney as a determinant of genetic hypertension: evidence from renal transplantation studies. Hypertension 46: 463–468, 2005 [DOI] [PubMed] [Google Scholar]
  • 90.Rigat B, Hubert C, Alhenc-Gelas F, Cambien F, Corvol P, Soubrier F. An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene accounting for half the variance of serum enzyme levels. J Clin Invest 86: 1343–1346, 1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Salvi E, Kutalik Z, Glorioso N, Benaglio P, Frau F, Kuznetsova T, Arima H, Hoggart C, Tichet J, Nikitin YP, Conti C, Seidlerova J, Tikhonoff V, Stolarz-Skrzypek K, Johnson T, Devos N, Zagato L, Guarrera S, Zaninello R, Calabria A, Stancanelli B, Troffa C, Thijs L, Rizzi F, Simonova G, Lupoli S, Argiolas G, Braga D, D'Alessio MC, Ortu MF, Ricceri F, Mercurio M, Descombes P, Marconi M, Chalmers J, Harrap S, Filipovsky J, Bochud M, Iacoviello L, Ellis J, Stanton AV, Laan M, Padmanabhan S, Dominiczak AF, Samani NJ, Melander O, Jeunemaitre X, Manunta P, Shabo A, Vineis P, Cappuccio FP, Caulfield MJ, Matullo G, Rivolta C, Munroe PB, Barlassina C, Staessen JA, Beckmann JS, Cusi D. Genomewide association study using a high-density single nucleotide polymorphism array and case-control design identifies a novel essential hypertension susceptibility locus in the promoter region of endothelial NO synthase. Hypertension 59: 248–255, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Samani NJ. Genome scans for hypertension and blood pressure regulation. Am J Hypertens 16: 167–171, 2003 [DOI] [PubMed] [Google Scholar]
  • 93.Shimoyama T, Hiraoka S, Takemoto M, Koshizaka M, Tokuyama H, Tokuyama T, Watanabe A, Fujimoto M, Kawamura H, Sato S, Tsurutani Y, Saito Y, Perbal B, Koseki H, Yokote K. CCN3 inhibits neointimal hyperplasia through modulation of smooth muscle cell growth and migration. Arterioscler Thromb Vasc Biol 30: 675–682, 2010 [DOI] [PubMed] [Google Scholar]
  • 94.Slavin TP, Feng T, Schnell A, Zhu X, Elston RC. Two-marker association tests yield new disease associations for coronary artery disease and hypertension. Hum Genet 130: 725–733, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Smithies O, Kim HS. Targeted gene duplication and disruption for analyzing quantitative genetic traits in mice. Proc Natl Acad Sci USA 91: 3612–3615, 1994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Tabara Y, Kohara K, Kita Y, Hirawa N, Katsuya T, Ohkubo T, Hiura Y, Tajima A, Morisaki T, Miyata T, Nakayama T, Takashima N, Nakura J, Kawamoto R, Takahashi N, Hata A, Soma M, Imai Y, Kokubo Y, Okamura T, Tomoike H, Iwai N, Ogihara T, Inoue I, Tokunaga K, Johnson T, Caulfield M, Munroe P, Umemura S, Ueshima H, Miki T. Common variants in the ATP2B1 gene are associated with susceptibility to hypertension: the Japanese Millennium Genome Project. Hypertension 56: 973–980, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wain LV, Verwoert GC, O'Reilly PF, Shi G, Johnson T, Johnson AD, Bochud M, Rice KM, Henneman P, Smith AV, Ehret GB, Amin N, Larson MG, Mooser V, Hadley D, Dorr M, Bis JC, Aspelund T, Esko T, Janssens AC, Zhao JH, Heath S, Laan M, Fu J, Pistis G, Luan J, Arora P, Lucas G, Pirastu N, Pichler I, Jackson AU, Webster RJ, Zhang F, Peden JF, Schmidt H, Tanaka T, Campbell H, Igl W, Milaneschi Y, Hottenga JJ, Vitart V, Chasman DI, Trompet S, Bragg-Gresham JL, Alizadeh BZ, Chambers JC, Guo X, Lehtimaki T, Kuhnel B, Lopez LM, Polasek O, Boban M, Nelson CP, Morrison AC, Pihur V, Ganesh SK, Hofman A, Kundu S, Mattace-Raso FU, Rivadeneira F, Sijbrands EJ, Uitterlinden AG, Hwang SJ, Vasan RS, Wang TJ, Bergmann S, Vollenweider P, Waeber G, Laitinen J, Pouta A, Zitting P, McArdle WL, Kroemer HK, Volker U, Volzke H, Glazer NL, Taylor KD, Harris TB, Alavere H, Haller T, Keis A, Tammesoo ML, Aulchenko Y, Barroso I, Khaw KT, Galan P, Hercberg S, Lathrop M, Eyheramendy S, Org E, Sober S, Lu X, Nolte IM, Penninx BW, Corre T, Masciullo C, Sala C, Groop L, Voight BF, Melander O, O'Donnell CJ, Salomaa V, d'Adamo AP, Fabretto A, Faletra F, Ulivi S, Del Greco F, Facheris M, Collins FS, Bergman RN, Beilby JP, Hung J, Musk AW, Mangino M, Shin SY, Soranzo N, Watkins H, Goel A, Hamsten A, Gider P, Loitfelder M, Zeginigg M, Hernandez D, Najjar SS, Navarro P, Wild SH, Corsi AM, Singleton A, de Geus EJ, Willemsen G, Parker AN, Rose LM, Buckley B, Stott D, Orru M, Uda M, van der Klauw MM, Zhang W, Li X, Scott J, Chen YD, Burke GL, Kahonen M, Viikari J, Doring A, Meitinger T, Davies G, Starr JM, Emilsson V, Plump A, Lindeman JH, Hoen PA, Konig IR, Felix JF, Clarke R, Hopewell JC, Ongen H, Breteler M, Debette S, Destefano AL, Fornage M, Mitchell GF, Smith NL, Holm H, Stefansson K, Thorleifsson G, Thorsteinsdottir U, Samani NJ, Preuss M, Rudan I, Hayward C, Deary IJ, Wichmann HE, Raitakari OT, Palmas W, Kooner JS, Stolk RP, Jukema JW, Wright AF, Boomsma DI, Bandinelli S, Gyllensten UB, Wilson JF, Ferrucci L, Schmidt R, Farrall M, Spector TD, Palmer LJ, Tuomilehto J, Pfeufer A, Gasparini P, Siscovick D, Altshuler D, Loos RJ, Toniolo D, Snieder H, Gieger C, Meneton P, Wareham NJ, Oostra BA, Metspalu A, Launer L, Rettig R, Strachan DP, Beckmann JS, Witteman JC, Erdmann J, van Dijk KW, Boerwinkle E, Boehnke M, Ridker PM, Jarvelin MR, Chakravarti A, Abecasis GR, Gudnason V, Newton-Cheh C, Levy D, Munroe PB, Psaty BM, Caulfield MJ, Rao DC, Tobin MD, Elliott P, van Duijn CM. Genome-wide association study identifies six new loci influencing pulse pressure and mean arterial pressure. Nat Genet 43: 1005–1011, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wang K, Li M, Hakonarson H. Analysing biological pathways in genome-wide association studies. Nat Rev Genet 11: 843–854, 2010 [DOI] [PubMed] [Google Scholar]
  • 99.White PC. Inherited forms of mineralocorticoid hypertension. Hypertension 28: 927–936, 1996 [DOI] [PubMed] [Google Scholar]
  • 100.Wilson FH, Disse-Nicodeme S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, Lifton RP. Human hypertension caused by mutations in WNK kinases. Science 293: 1107–1112, 2001 [DOI] [PubMed] [Google Scholar]
  • 101.Yang HC, Liang YJ, Chen JW, Chiang KM, Chung CM, Ho HY, Ting CT, Lin TH, Sheu SH, Tsai WC, Chen JH, Leu HB, Yin WH, Chiu TY, Chern CL, Lin SJ, Tomlinson B, Guo Y, Sham PC, Cherny SS, Lam TH, Thomas GN, Pan WH. Identification of IGF1, SLC4A4, WWOX, and SFMBT1 as hypertension susceptibility genes in Han Chinese with a genome-wide gene-based association study. PLoS One 7: e32907, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Yang Y, Parsons KK, Chi L, Malakauskas SM, Le TH. Glutathione S-transferase-micro1 regulates vascular smooth muscle cell proliferation, migration, and oxidative stress. Hypertension 54: 1360–1368, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zhu X, Young JH, Fox E, Keating BJ, Franceschini N, Kang S, Tayo B, Adeyemo A, Sun YV, Li Y, Morrison A, Newton-Cheh C, Liu K, Ganesh SK, Kutlar A, Vasan RS, Dreisbach A, Wyatt S, Polak J, Palmas W, Musani S, Taylor H, Fabsitz R, Townsend RR, Dries D, Glessner J, Chiang CW, Mosley T, Kardia S, Curb D, Hirschhorn JN, Rotimi C, Reiner A, Eaton C, Rotter JI, Cooper RS, Redline S, Chakravarti A, Levy D. Combined admixture mapping and association analysis identifies a novel blood pressure genetic locus on 5p13: contributions from the CARe consortium. Hum Mol Genet 20: 2285–2295, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES