Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Mar 20.
Published in final edited form as: Curr Hypertens Rep. 2013 Dec;15(6):676–686. doi: 10.1007/s11906-013-0388-6

Progress and Future Aspects in Genetics of Human Hypertension

Qi Zhao 1, Tanika N Kelly 1, Changwei Li 1, Jiang He 1
PMCID: PMC4368442  NIHMSID: NIHMS528143  PMID: 24072558

Abstract

Hypertension has become a major global health burden due to its high prevalence and associated increase in risk of cardiovascular disease and premature death. It is well established that hypertension is determined by both genetic and environmental factors and their complex interactions. Recent large-scale meta-analyses of genome-wide association studies (GWAS) have successfully identified a total of 38 loci which achieved genome-wide significance (P < 5×10−8) for their association with blood pressure (BP). Although the heritability of BP explained by these loci is very limited, GWAS meta-analyses have elicited renewed optimism in hypertension genomics research, highlighting novel pathways influencing BP and elucidating genetic mechanisms underlying BP regulation. This review summarizes evolving progress in the rapidly moving field of hypertension genetics and highlights several promising approaches for dissecting the remaining heritability of BP. It also discusses the future translation of genetic findings to hypertension treatment and prevention.

Keywords: Blood pressure, Genetic association studies, Genetic linkage, Genome-wide association study, Hypertension, Rare variants, Sequencing, Risk prediction

Introduction

Elevated blood pressure (BP) is a major global health challenge due to its high prevalence and associated increased risk of cardiovascular disease (CVD) and premature death [14]. An estimated 978 million adults, or 28% of the world’s adult population, had uncontrolled hypertension in 2008 [2]. More alarming, conservative estimates indicate that the global burden of hypertension will increase to more than 1.5 billion by 2025 [4]. As the most important modifiable risk factor for CVD and all-cause mortality, elevated BP was responsible for approximately 7.6 million deaths globally, or 13.5% of all deaths, in 2001 [1, 3].

BP is influenced by both genomic and environmental factors, as well as their interactions. Although BP was established early on as an inheritable trait, with many monogenic forms of BP dysregulation clearly described, our understanding of the genomic architecture of the complex BP phenotype was initially slow to progress [5]. Early genome-wide linkage analyses, candidate gene studies, and genome-wide association studies (GWAS) were relatively unsuccessful in identifying reproducible loci related to BP [612]. However, increased methodological stringency and the recent formation of large BP consortia have enabled important breakthroughs in hypertension genomic research. Through GWAS meta-analyses, numerous loci have now been robustly associated with BP in populations of European and Asian ancestries [5, 1316]. Although much of the heritability of BP still remains unexplained, there is renewed optimism as we turn our attention towards next-generation approaches for the discovery of novel genomic determinants of this complex trait.

Genetics of Hypertension in the Pre-GWAS Era

Monogenic forms of hypertension

Some of the earliest advancements in human BP genomics research involved the identification of the genes responsible for severe inherited forms of hypertension and hypotension. Although many physiological processes are responsible for the regulation of BP, the vast majority of genes identified for monogenic BP disorders play key roles in renal-sodium handling [1723]. Many such genes have been shown to exert their effects by directly or indirectly influencing sodium and water reabsorption in the nephron’s distal tubule, leading to changes in plasma volume, cardiac output, and BP [24]. A number of reports have provided systematic reviews of a variety of types of monogenetic forms of hypertension and their related causal mutations [2529].

Identification of genes responsible for monogenic hypertension and hypotension disorders has provided valuable insights into the genomic mechanisms and biological pathways underlying BP regulation. Furthermore, such research has also provided important clues to investigators of the complex essential hypertension phenotype. For example, in comparison to the rare variants in genes responsible for monogenic BP disorders, investigators have postulated that common genetic variation in these genes may have more modest effects, contributing to the inter-individual variation in the complex BP phenotype [30]. As such, these genes have been the target of myriad candidate gene studies of BP and hypertension [31], and are considered very promising targets for follow-up when present at GWAS-identified loci [13].

Heritability of essential hypertension

Blood pressure has long been established as an inheritable trait, suggesting a significant contribution of genetic factors to this complex phenotype [3234]. The heritability of BP has been shown to range from about 30–60% in pedigree data to as high as 70% in twin studies [3544]. Longitudinal data from the Framingham Heart Study showed that 57% and 56% of inter-individual variability in systolic (SBP) and diastolic BP (DBP), respectively, was due to genetic factors [34]. Data from Nigerian families suggest heritabilities of 34% to 45% and 29% to 43% for SBP and DBP, respectively [43, 44]. Similarly, in the Chinese population, Gu et al estimated significant heritabilities of 31% and 32% for SBP and DBP, respectively [36].

Linkage and Candidate studies

Given the widespread success of genome-wide linkage analyses in the identification of genes for Mendelian disorders, investigators were initially optimistic about using this approach to localize genomic regions harboring susceptibility loci for the complex BP phenotype. Numerous genome-wide linkage scans of SBP, DBP, or hypertension were subsequently conducted, but with somewhat disappointing results. For example, among approximately 34 quantitative trait loci (QTLs) for SBP, DBP, and hypertension phenotypes which achieved a LOD score of 3.0 or higher [34, 4553], only one locus has been replicated in independent samples. Hsueh and colleagues linked 2q31–2q34 to DBP among Old Order Amish families [45], while Morrison and colleagues linked 2q34 to hypertension among African-American families [46]. The failure of linkage analyses highlights the complexity of the genomic mechanisms underlying BP regulation. In addition, it has spurred a general shift away from this approach in favor of more powerful association methods.

To date, over 1,500 genes have been related to BP in human populations, with the vast majority derived from candidate gene association studies [54]. Based on a priori knowledge of biologic function, candidate gene studies offer a powerful approach for detecting genetic variants which influence common complex traits like BP. Despite their popularity, early candidate gene studies of BP were hampered by inconsistent findings, which to some extent may have reflected methodological limitations, including small sample sizes, poor phenotype measurement, inappropriate correction for multiple testing, and lack of verification in independent samples. Some investigators, however, have continued to support the use of candidate gene studies, noting that biologically relevant loci may be missed by GWAS which use very stringent alpha-thresholds for determining statistical significance [55]. Some of the more recent candidate gene studies have successfully identified genetic associations that are reproducible in independent samples [5557]. Such successes are likely the results of the employment of large sample sizes and appropriate correction for multiple testing. Furthermore, more recent candidate gene studies have taken advantage of advances in high-throughput genotyping technology to identify gene variants related to BP utilizing gene-centric arrays which interrogate large numbers of variants in a multitude of genes and biological pathways [57, 58]. Using the HumanCVD BeadChip, which genotypes approximately 50,000 single nucleotide polymorphisms (SNPs) from 2,000 genes demonstrated to associate with CVD-related traits, Johnson and colleagues identified BP-related SNPs in the LSP1/TNNT3, MTHFR-NPPB, AGT, ATP2B1, NPR3, HFE, NOS3 and SOX6 genes among a discovery-stage sample of 25,118 participants and replication study of 59,349 participants [58]. In summary, these findings demonstrate that despite their tarnished reputation, candidate gene studies may still play a role in our quest to discover variants related to BP.

Progress in the GWAS Era

By interrogating a dense panel of SNPs covering the entire genome, GWAS represent an agnostic and powerful approach for the discovery of susceptibility loci for common complex traits. As such, there was initial enthusiasm at the prospect of using GWAS to identify novel BP-related variants. However, in contrast to GWAS for other CVD-related phenotypes [6, 59, 60], early GWAS failed to identify any associations with BP at a level of genome-wide significance (P < 5×10−8) [6, 811]. For example, in the Wellcome Trust Case Control Consortium (WTCCC), investigators used a 500K Affymetrix SNP chip to compare approximately 2,000 cases for each of 7 common diseases, including hypertension, to 3,000 shared controls. In this study, a total of 24 independent association signals were identified for 6 diseases with the exception of hypertension. There were no signals that achieved even a suggestive association of P<5×10−7 with hypertension [6]. While a couple of the more recent GWAS have identified BP loci that meet conventional significance thresholds with evidence of replication [61, 62], the failure of early GWAS created an impetus for the formation of consortia with the purpose of conducting GWAS meta-analyses in very large samples capable of detecting the modest effects of BP loci [5, 1316].

In June 2009, two consortia, CHARGE and Global Blood Pressure Genetics (Global BPgen), reported findings of their large-scale GWAS meta-analyses. With discovery-stage sample sizes of 29,136 and 34,133 participants in CHARGE and Global BPgen, respectively, they together identified 13 independent loci associated with BP at a level of genome-wide significance (P < 5×10−8) [13, 14]. These findings represented an important advance in BP genomics research, providing some of the first robust evidence of genetic association for the BP phenotype. Since the 2009 publications, four additional large BP GWAS meta-analyses have been conducted in European and East Asian populations. These include two from the International Consortium of BP (ICBP), which is the largest GWAS meta-analysis of BP to date, with a discovery-stage sample of approximately 70,000 participants [5, 16]; one from the HYPERGENES Project, with a smaller sample size of 1,865 hypertension cases and 1,750 controls [63]; and one from the Asian Genetic Epidemiology Network (AGEN), with GWAS data from nearly 20,000 East Asian participants and follow-up genotyping in an additional 30,000 [15]. In total, these studies have identified 38 loci robustly associated with BP traits (Table 1).

Table 1.

Genetic variants which achieved P < 5×10−8 in previous GWAS meta-analyses, according to their one mega-base position.

Chr Region Lead SNP Position Nearest Gene Functional Relevance Associated Phenotype(s) Identifying Consortium
1p36 rs880315 10719453 CASZ11 Intron DBP AGEN
rs17367504 11785365 MTHFR1 Intron SBP, DBP, HTN, MAP Global BPgen, AGEN, ICBP2,3

1p13 rs17030613 112971190 CAPZA11 Intron DBP AGEN
rs2932538 113018066 MOV10 Near Promoter SBP, DBP ICBP2

2q24 rs16849225 164615066 FIGN Intergenic SBP AGEN
rs13002573 164623454 FIGN Intergenic PP ICBP3
rs1446468 164671732 FIGN Intergenic SBP, DBP, MAP ICBP3

3p24 rs13082711 27512913 SLC4A7 Intergenic DBP. MAP ICBP2,3

3p22 rs3774372 41852418 ULK41 Missense DBP ICBP2
rs9815354 41887655 ULK41 Intron DBP CHARGE, AGEN

3p21 rs319690 47902488 MAP41 Intron SBP, DBP, MAP ICBP3

3q26 rs419076 170583580 MECOM1 Intron SBP, DBP, MAP ICBP2,3
rs1343040 170668987 MECOM1 Intron MAP ICBP3

4q12 rs871606 54494002 CHIC2 Intergenic PP ICBP3

4q21 rs13149993 81377569 FGF5 Intergenic MAP ICBP3
rs1458038 81383747 FGF5 Intergenic SBP, DBP, MAP ICBP2,3
rs16998073 81403365 FGF5 Intergenic SBP, DBP, HTN, MAP Global BPgen, AGEN, ICBP3

4q24 rs13107325 103407732 SLC39A81 Missense SBP, DBP, MAP ICBP2,3

4q25 rs6825911 111601087 ENPEP Intergenic DBP AGEN

4q32 rs13139571 156864963 GUCY1A31 Intron DBP ICBP2,3

5p13 rs1173756 32825609 NPR31 3′ UTR PP ICBP3
rs1173766 32840285 NPR3 Intergenic SBP AGEN
rs1173771 32850785 NPR3 Intergenic SBP, DBP, HTN.
MAP, PP
ICBP2,3

5q33 rs9313772 157737035 EBF1 Intergenic MAP ICBP3
rs11953630 157777980 EBF1 Intergenic MAP, SBP, DBP ICBP2,3

6p22 rs1799945 26199158 HFE1 Missense MAP, SBP, DBP, HTN ICBP2,3
rs198846 26215442 HIST1H1T Near 3′UTR MAP ICBP3

6p21 rs805303 31724345 BAG61 Intron SBP, DBP, HTN ICBP2

7q22 rs17477177 106199094 PIK3CG Intergenic PP, SBP ICBP3

7q36 rs3918226 150321109 NOS3 Near Promoter HTN HYPERGENES

8q24 rs2071518 120504993 NOV1 3′ UTR PP ICBP3

10p12 rs4373814 18459978 CACNB21 Intron SBP, DBP ICBP2
rs1813353 18747454 CACNB21 Intron MAP, SBP, DBP, HTN ICBP2,3
rs11014166 18748804 CACNB21 Intron MAP, DBP CHARGE, AGEN, ICBP3
rs12258967 18767965 CACNB21 Intron MAP ICBP3

10q21 rs4590817 63137559 C10orf1071 Intron MAP, SBP, DBP, HTN ICBP2,3
rs1530440 63194597 C10orf1071 Intron MAP, DBP Global BPgen, AGEN, ICBP3

10q23 rs9663362 95885167 PLCE11 Intron PP ICBP3
rs932764 95885930 PLCE11 Intron SBP, HTN ICBP2,3

10q24 rs1004467 104584497 CYP17A11 Intron PP, MAP, SBP CHARGE, ICBP3
rs3824755 104585839 CYP17A11 Intron PP ICBP3
rs11191548 104836168 NT5C2 Intergenic PP, MAP, SBP, DBP, HTN Global BPgen, AGEN, ICBP2,3
rs11191593 104929205 NT5C21 Intron MAP ICBP3

10q25 rs2782980 115771517 ADRB1 Intergenic MAP ICBP3

11p15 rs7129220 10307114 ADM Intergenic SBP ICBP2,3

11p15 rs381815 16858844 PLEKHA71 Intron MAP, SBP, DBP CHARGE, AGEN, ICBP2,3

11q22 rs633185 100098748 ARHGAP421 Intron MAP, SBP, DBP, HTN ICBP2,3
rs604723 100115756 ARHGAP421 Intron MAP ICBP3

11q24 rs11222084 129778440 ADAMTS-8 Intergenic PP ICBP3

12q21 rs4842666 88465680 POC1B Intergenic SBP CHARGE
rs11105328 88466521 POC1B Intergenic SBP CHARGE
rs2681472 88533090 ATP2B11 Intron PP, MAP, SBP, DBP, HTN CHARGE, AGEN, ICBP3
rs2681492 88537220 ATP2B11 Intron PP, MAP, SBP, DBP CHARGE, ICBP3
rs11105354 88550654 ATP2B11 Intron SBP, HTN CHARGE
rs12579302 88574634 ATP2B1 Near Promoter SBP, HTN CHARGE
rs17249754 88584717 ATP2B1 Intergenic PP, MAP, SBP, DBP, HTN CHARGE, AGEN, ICBP3
rs11105364 88593407 ATP2B1 Intergenic SBP, HTN CHARGE
rs11105368 88598572 ATP2B1 Intergenic SBP, HTN CHARGE
rs11105378 88614872 ATP2B1 Intergenic SBP, HTN CHARGE
rs12230074 88614998 ATP2B1 Intergenic SBP, HTN CHARGE

12q24 rs3184504 110368991 SH2B31 Missense MAP, SBP, DBP CHARGE, ICBP2,3
rs4766578 110388754 ATXN1 Intron DBP CHARGE
rs10774625 110394602 ATXN1 Intron DBP CHARGE
rs653178 110492139 ATXN21 Intron MAP, DBP CHARGE, Global Bpgen, ICBP3
rs671 110726149 ALDH21 Missense SBP, DBP AGEN
rs11066132 110952589 NAA251 Intron SBP, DBP AGEN
rs2074356 111129784 HECTD41 Intron SBP, DBP AGEN
rs11066280 111302166 HECTD41 Intron SBP, DBP AGEN

12q24 rs2384550 113837114 TBX3 Intergenic DBP CHARGE, AGEN
rs10850411 113872179 TBX3 Intergenic DBP ICBP2,3
rs35444 114036820 TBX3 Intergenic DBP AGEN

15q24 rs1378942 72864420 CSK1 Intron MAP, SBP, DBP, HTN Global BPgen, AGEN, ICBP2,3
rs6495122 72912698 ULK31 Intron MAP, DBP CHARGE, ICBP3

15q26 rs2521501 89238392 FES1 Intron MAP, SBP, DBP ICBP2,3

17q21 rs12946454 40563647 ACBD4 Near Promoter SBP Global Bpgen, AGEN

17q21 rs8069437 42261948 WNT3 Intergenic PP ICBP3
rs17608766 42368270 GOSR21 Intron PP, SBP ICBP2,3

17q21 rs12940887 44757806 ZNF6521 Intron SBP, DBP ICBP2,3
rs16948048 44795465 ZNF652 Near Promoter DBP Global Bpgen, AGEN

18p11 rs8096897 13428905 c18orf11 Intron SBP CHARGE

20p12 rs1327235 10917030 JAG1 Intergenic MAP, SBP, DBP ICBP2,3

20q13 rs6026748 57179210 ZNF831 Intergenic MAP ICBP3
rs6015450 57184512 ZNF831 Intergenic MAP, SBP, DBP, HTN ICBP2,3

AGEN=Asian Genetic Epidemiology Network; CHARGE=Cohorts for Hearts and Aging Research in Genomic Epidemiology; Chr=Chromosome; DBP=Diastolic blood pressure; Global Bpgen=Global Blood Pressure Genetics; HTN=Hypertension; ICBP=International Consortium of Blood Pressure; MAP=Mean arterial pressure; PP=Pulse pressure; SBP=Systolic blood pressure.

1

Corresponding variant is located within this gene;

2

ICBP publication by Ehret and colleagues [5];

3

ICBP publication by Wain and colleagues [16].

Although inference of causal genes and variants based on GWAS signals alone is difficult due to regional linkage disequilibrium (LD) structure, findings from these large GWAS meta-analyses have provided robust association evidence for some biological candidate genes previously suspected to influence BP. For example, meta-analysis of CHARGE and Global BPgen findings revealed an association of SBP with intronic marker rs1004467 (P=1.28×10−13) of the CYP17A1 gene, which is responsible for a monogenic form of hypertension [14, 64]. Similarly, in the GWAS meta-analysis by Global BPgen, Newton-Cheh and colleagues identified a strong signal for SBP at 1p36. The most significant SNP at that locus was rs17367504 (P=7×10−24), an intronic variant of the MTHFR gene, which has been implicated in BP due to its role in regulating homocysteine, a biomarker linked to endothelial dysfunction and hypertension [65]. Several other relevant biological candidates are also present at this locus, including NPPA and NPPB, which encode natriuretic peptides, renin-angiotensinogen-aldosterone system (RAAS) component AGTRAP, and ion channel CLCN6 [13].

While GWAS meta-analyses results have provided association evidence for some genes with known biologic relevance, the majority of loci identified had not been previously implicated in studies of BP regulation in human populations. For example, the ATP2B1 gene at the 12q21 locus achieved genome-wide significance for SBP, DBP, and mean arterial pressure (MAP) in GWAS meta-analyses conducted by CHARGE, Global BPgen, and ICBP [5, 13, 14, 16], but has never been linked with BP regulation. The post hoc investigation into the potential biologic plausibility of ATP2B1 revealed a previous experiment demonstrating increased mRNA expression in the spontaneously hypertensive rat [66]. While some genes at implicated loci, like that of ATP2B1, have demonstrated plausibility for association with BP based on our current knowledge, other loci discovered by GWAS meta-analyses have provided completely novel insights into BP regulation. For example, the SH2B3 locus achieved genome-wide significance for SBP, DBP, and MAP in GWAS meta-analyses by CHARGE, Global BPgen, and ICBP [5, 13, 14, 16]. SH2B3 had been shown previously to exert an effect on cytokine sensitivity in studies of knockout mice and was associated with autoimmune conditions in human populations [14]. Based on these studies, Levy and colleagues speculated that immune response pathways may influence BP by mechanisms not previously appreciated [14].

As the largest consortium of GWAS conducted in the East Asian population, the AGEN Hypertension meta-analysis replicated 7 of the 13 loci that had been identified previously by the CHARGE and Global BPgen consortia, including 4 at a level of genome-wide significance [15]. Of particular importance, the AGEN meta-analysis identified 5 novel loci which achieved P < 5×10−8 for association with BP phenotypes [15]. These findings indicate that the physiologic effects of many common polymorphisms may be generalizable across populations with diverse genetic backgrounds. On the other hand, the success of AGEN also suggests that genomic mechanisms may be discovered in unique populations due to differences in allele frequencies or factors that interact with genes to influence BP. Thus, the investigation of genomic factors influencing BP in populations with differing genetic backgrounds should continue to be pursued. Findings from these studies will be essential to enhancing our understanding of the molecular mechanisms underlying BP regulation.

Promising Approaches for Dissecting the Missing Heritability of Hypertension

To date, most identified genetic variants have displayed modest effect sizes. It was estimated that the currently identified common variants explain only about 0.9% of the variability of BP, leaving a large proportion of heritability unexplained [5]. As we look towards the future, new approaches are being sought to help explain the “missing heritability” of BP. On the horizon are global GWAS meta-analyses, research of gene-gene and gene-environment interactions (including epigenetic studies), and, as we move beyond GWAS, next-generation sequencing studies. While setbacks are likely to occur as we continue to move forward, there is optimism that such work will make headway in our quest to better understand the genomic architecture of BP.

Global GWAS meta-analyses

In the ICBP GWAS meta-analysis, Ehret and colleagues estimated that up to 2.2% of inter-individual variation in BP would eventually be explained by approximately 116 common genetic variants theorized to associate with BP (compared to 0.9% of variation explained by 29 currently identified SNPs) [5]. However, they showed that very large sample sizes would be required to detect these remaining SNPs [5]. Mega-consortia are now being formed that include genetically diverse samples from around the world. By substantially increasing sample sizes, these studies will have power to detect additional BP loci [67, 68]. Furthermore, such research will present an outstanding opportunity to refine genomic signals in the search for causal variants by leveraging LD structure across populations [67, 68]. In undertaking these studies, investigators will likely encounter new challenges, such as how to appropriately account for the genetic heterogeneity that exists between ethnically diverse samples while maximizing study power [67, 68]. However, novel insights into other phenotypes, such as serum proteins, have already been identified by global GWAS meta-analysis approaches [69]. It is likely that BP will soon follow suit.

Gene-gene and gene-environment interactions

Given the commonly accepted belief that complex traits like BP are influenced by the interaction of genetic and environmental factors, it has been suggested that research of such interactions could help explain some of the missing heritability of these traits [30, 70, 71]. Still, there is a paucity of data from GWAS examining how genes interact with each other and with environmental factors to influence BP. Since current methods for detecting interactions have been shown to lack power, investigators may be hesitant to undertake such analyses, especially in light of the early difficulties of BP GWAS in identifying simple single-marker associations [72]. However, before moving completely beyond GWAS, it may be worthwhile to leverage data from existing large consortia to explore the interactions between genes and environmental factors on BP.

Epigenetics

Epigenetics is the study of heritable alterations in phenotypes and gene expression that occur without changes in the DNA sequence [73]. The epigenetic control of gene expression is critical for many cell functions, such as tissue specificity, germline specificity, imprinting, and X-chromosome inactivation [74]. Epigenetic processes include nucleic acid methylation, histone modification, nucleosome positioning, transcription control with DNA-binding proteins and noncoding RNAs, and translation control with microRNAs and RNA-binding proteins. Epigenetic mechanisms have been involved in the pathogenesis of CVD, including hypertension, [73, 75] and suggested as a potential mechanism for explaining a part of missing heritability of these complex diseases [74]. Studies have already shown a loss of global genomic methylation content among hypertension patients, as well as hypermethylation of the HSD11B2 gene [76, 77]. To reveal epigenetic biomarkers implicated in hypertension etiology, progression, and prevention, the National Heart, Lung, and Blood Institute convened a working group of multidisciplinary experts to identify urgently needed studies and resources and the future direction of epigenetic research of hypertension [74]. A better understanding of epigenetic changes in response to environmental and genetic stresses is needed to clarify the factors that act together to determine an individual’s BP.

Rare variants and sequencing studies

Cohen and colleagues achieved early success identifying rare variants with large influence on lipid phenotypes by sequencing extremes of the population distribution, prompting investigators to turn their attention towards clarifying the role of rare genetic variants in the complex BP phenotype [7880]. There is already some suggestion that rare variants could help explain the missing heritability of BP. For example, Ji and colleagues reported that carriers of rare functional mutations in three renal salt-handling genes (SLC12A3, SLC12A1, and KCNJ1) had significantly reduced BP compared to non-carriers [81]. Similarly, Rao et al resequenced a locus of the CHGA gene and discovered a Gly364Ser amino-acid substitution that decreased DBP by approximately 5 mmHg [82]. While these previous studies have sequenced a limited number of genes, the advent of next-generation sequencing technology has made it plausible to deeply sequence large stretches of DNA, whole exomes, or even the entire genome in large population-based studies [83]. As such, the National Heart, Lung, and Blood Institute sponsored an initiative to identify low-frequency and rare variants which may contribute to heart, lung, and blood disorders by conducting whole-exome sequencing in ongoing population-based studies [84]. With much of the sequencing completed and catalogued in the database of Genotypes and Phenotypes (dbGaP), results from the BP working group are eagerly anticipated [85].

Prospects for Translation of Genetic Findings

Development of novel drugs for hypertension treatment

Recent large-scale genetic studies have implicated novel biological pathways in BP regulation, providing potential targets for the treatment of hypertension and the prevention of CVD. However, the translation of genetic findings from GWAS into the clinic remains limited and a topic of intense debate. It takes a considerable length of time to move from a gene target identified by association study to an approved marketed drug, and most GWAS results have become available only in the past few years. In addition, the effect sizes of GWAS-identified BP variants are relatively small (ranging from 0.2–1.0 mm Hg per risk allele) [5], and the merit of their utilization in clinical practice is not clear. Nevertheless, the case for statins in the treatment of high low-density lipoprotein (LDL)-cholesterol provides optimism for the potential use of GWAS-identified BP genes as pharmaceutical targets for antihypertensive drug development [86]. Although statins were developed in the last century, a recent GWAS identified that the gene (HMGCR) encoding the statins’ target protein, 3-hydroxy-3-methylglutaryl coenzyme A reductase, was associated with plasma LDL-cholesterol levels (P for rs12654264 = 1 × 10−20) [87]. Despite an effect size of only 2.7 mg/dl per allele of the rs12654264 variant [87], the statin drug can lower LDL-cholesterol by 40–60% [88].

Although the development of novel drugs based on GWAS findings will take some time, Sanseau and collaborators have suggested a potential shortcut for using emerging genomics research to assuage human disease. In an analysis conducted using data from the National Human Genome Research Institute’s repository of GWAS data and Informa Healthcare’s Pharmaprojects database of drug development projects [89], they found that out of 155 genes that could be mapped to GWAS traits and were also targeted by available drugs, 92 genes were associated with drugs that had indications for diseases that differed from their mapped GWAS traits. These findings suggest that GWAS data may help us identify novel uses for existing drugs, leading to immediate translational opportunities for GWAS findings.

Hypertension risk prediction

Improving risk prediction is a key objective in genomic studies of human diseases and is an important component of “personalized medicine,” including risk stratification, targeted prevention, and therapeutic interventions. However, GWAS-identified variants that have been robustly associated with BP and hypertension have relatively small effect sizes. In addition, most GWAS have used cross-sectional data, and the predictive values of variants identified by such studies need validation in prospective cohorts. Fortunately, investigators have begun to implement large-scale longitudinal cohorts to confirm the associations between GWAS-identified BP variants and both hypertension incidence and BP change over time. For example, Fava and colleagues recently validated a genetic risk score (GRS) with aggregate genetic information from 29 GWAS-BP SNPs. These variants were cumulatively and independently associated with hypertension incidence and BP changes over approximately 23 years’ follow-up among more than 17,000 Swedes [90]. However, their analyses did not show an improvement in the prediction of incident hypertension beyond traditional risk factors. Indeed, the magnitude of association of the GRS with hypertension incidence is substantially lower than that of obesity and prehypertension status, but comparable to that of either positive family history of hypertension or the presence of diabetes. These results suggest that it is still too early to consider GWAS findings in the prediction of hypertension. In the future, however, knowledge of additional BP-related genomic variants and their complex interactions with both genetic and environmental factors could substantially improve the GRS and lead to its translation to the clinical setting.

Antihypertensive pharmacogenomics

Another promising area of genomic research is its application in the prediction of individual response and side effects to antihypertensive therapies. Although this is still far away from clinical application, the past decade has seen substantial growth in the literature surrounding hypertension pharmacogenomics. Most of the studies have been focused on candidate genes, primarily direct protein targets of a drug or involved in the physiological or pharmacological signaling pathways relevant to a drug’s action. For example, genetic variants of several genes from the RAAS (ACE, AGT, AGTR1, AGTR2, and REN) have been widely investigated for their associations with BP response to angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers [91]. Variants of β1-adrenergic receptor (ADRB1) from the sympathetic nervous system and its associated regulatory protein (GRK4) have shown significant interaction effects with β-blocker on BP lowering [9294]. In addition, renal sodium absorption-regulating genes ADD1, WNK1, and NEDD4L have influenced BP response to diuretics in an interactive manner [95]. With the advent of GWAS, Turner et al published the first GWAS of antihypertensive pharmacogenomics [96], in which theyhey identified and validated a region on chromosome 12 that was associated with DBP response to hydrochlorothiazide. This region includes LYZ, YEATS, and FRS2 genes that had not been previously implicated in hypertension or response to diuretics. The study highlights the potential power of the GWAS approach in antihypertensive pharmacogenomics. Other groups are conducting ongoing pharmacogenomics studies that will also utilize GWAS [97].

Although there have been significant advances in hypertension pharmacogenomics research, most of the studies were not sufficiently powered, with relatively small sample size and lack of replication samples. Thus, collaboration among investigators to allow large-scale joint analyses and replication will be essential in advancing this field. Ethnic differences have also been noted in response to the BP-lowering effects of antihypertensive medications, as seen with β-blockers and diuretics [98]. This not only supports the role of genetic factors in determining individuals’ response to antihypertensive medications, but it also highlights the necessity and importance of utilizing multiple ethnicities to identify genetic variants responsible for varied BP response to treatment.

Conclusions

Although the genomic mechanisms underlying BP regulation have yet to be fully elucidated, there have been important advances in the field. Initially slow to progress, genetic association studies seem to have finally delivered on their promise to identify common polymorphisms associated with this trait. While it is true that much of the heritability of BP remains unexplained, the variants robustly identified by previous GWAS meta-analyses already show non-negligible associations with BP and its comorbid conditions. Furthermore, with the formation of global GWAS meta-analysis consortia, the emergence of epigenetics, and the advent of next-generation sequencing technology, the future for BP genomics research is bright. Investigators are optimistic that the coming years will offer a clearer picture of the genomic architecture of BP. Eventually, such insights could be used to identify individuals at high risk for hypertension who may benefit most from primary prevention efforts, and could provide new biological targets for developing more effective hypertension treatment methods. In addition, advances in pharmacogenomics of antihypertensive drugs may be used to develop novel personalized treatments for hypertension. Advancements in all of these areas will have important public health and clinical implications that will help to curb the growing cardiovascular disease epidemic at national and global levels.

Acknowledgments

This work was supported by research grants (R01HL087263 and R01HL090682) from the National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.

Footnotes

Conflict of Interest

Qi Zhao, Tanika N. Kelly, Changwei Li, and Jiang He declare that they have no conflict of interest.

Compliance with Ethics Guidelines

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Contributor Information

Qi Zhao, Email: qizhao@tulane.edu.

Tanika N. Kelly, Email: tkelly@tulane.edu.

Changwei Li, Email: cli8@tulane.edu.

References

Recently published papers of particular interest have been highlighted as:

• Of importance

•• Of major importance

  • 1.Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, Adair-Rohani H, et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2224–60. doi: 10.1016/S0140-6736(12)61766-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Danaei G, Finucane MM, Lin JK, Singh GM, Paciorek CJ, Cowan MJ, et al. National, regional, and global trends in systolic blood pressure since 1980: systematic analysis of health examination surveys and epidemiological studies with 786 country-years and 5.4 million participants. Lancet. 2011;377:568–77. doi: 10.1016/S0140-6736(10)62036-3. [DOI] [PubMed] [Google Scholar]
  • 3.Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJ. Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data. Lancet. 2006;367:1747–57. doi: 10.1016/S0140-6736(06)68770-9. [DOI] [PubMed] [Google Scholar]
  • 4.Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217–23. doi: 10.1016/S0140-6736(05)17741-1. [DOI] [PubMed] [Google Scholar]
  • 5••.Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, et al. Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 2011;478:103–9. doi: 10.1038/nature10405. To date, this is the largest meta-analysis of genome-wide association studies (GWAS) of the phenotypes of blood pressure by the International Consortium of Blood Pressure (ICBP). It included a discovery GWAS among 69,395 individuals and a combined sample of about 200,000 Europeans. It identified 16 novel loci associated with blood pressure. A genetic risk score calculated by GWAS significant variants was associated with hypertension, left ventricular wall thickness, stroke, and coronary artery disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature. 2007;447:661–78. doi: 10.1038/nature05911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Saxena R, Voight BF, Lyssenko V, Burtt NP, de Bakker PI, Chen H, et al. Genome-wide association analysis identifies loci for type 2 diabetes and triglyceride levels. Science. 2007;316:1331–6. doi: 10.1126/science.1142358. [DOI] [PubMed] [Google Scholar]
  • 8.Levy D, Larson MG, Benjamin EJ, Newton-Cheh C, Wang TJ, Hwang SJ, et al. 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]
  • 9.Kato N, Miyata T, Tabara Y, Katsuya T, Yanai K, Hanada H, et al. High-density association study and nomination of susceptibility genes for hypertension in the Japanese National Project. Hum Mol Genet. 2008;17:617–27. doi: 10.1093/hmg/ddm335. [DOI] [PubMed] [Google Scholar]
  • 10.Sabatti C, Service SK, Hartikainen AL, Pouta A, Ripatti S, Brodsky J, 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]
  • 11.Wang Y, O’Connell JR, McArdle PF, Wade JB, Dorff SE, Shah SJ, et al. From the Cover: Whole-genome association study identifies STK39 as a hypertension susceptibility gene. Proc Natl Acad Sci U S A. 2009;106:226–31. doi: 10.1073/pnas.0808358106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Harrap SB. Where are all the blood-pressure genes? Lancet. 2003;361:2149–51. doi: 10.1016/S0140-6736(03)13694-X. [DOI] [PubMed] [Google Scholar]
  • 13.Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, Coin L, et al. Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet. 2009;41:666–76. doi: 10.1038/ng.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, Dehghan A, et al. Genome-wide association study of blood pressure and hypertension. Nat Genet. 2009;41:677–87. doi: 10.1038/ng.384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15••.Kato N, Takeuchi F, Tabara Y, Kelly TN, Go MJ, Sim X, et al. Meta-analysis of genome-wide association studies identifies common variants associated with blood pressure variation in east Asians. Nat Genet. 2011;43:531–8. doi: 10.1038/ng.834. This is the first, largest meta-analysis of genome-wide association studies of blood pressure among East Asians. This study identified five novel genetic variants associated with blood pressure in East Asians and also provided evidence of East Asian-specific blood pressure association at ALDH2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16••.Wain LV, Verwoert GC, O’Reilly PF, Shi G, Johnson T, Johnson AD, et al. Genome-wide association study identifies six new loci influencing pulse pressure and mean arterial pressure. Nat Genet. 2011;43:1005–11. doi: 10.1038/ng.922. This study is the second meta-analysis conducted by the International Consortium of Blood Pressure (ICBP). This study idenified four new genomic loci associated with pulse pressure and one locus associated with both pulse pressure and mean arterial pressure. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Abriel H, Loffing J, Rebhun JF, Pratt JH, Schild L, Horisberger JD, et al. Defective regulation of the epithelial Na+ channel by Nedd4 in Liddle’s syndrome. J Clin Invest. 1999;103:667–73. doi: 10.1172/JCI5713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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;13:183–8. doi: 10.1038/ng0696-183. [DOI] [PubMed] [Google Scholar]
  • 19.Simon DB, Bindra RS, Mansfield TA, Nelson-Williams C, Mendonca E, Stone R, et al. Mutations in the chloride channel gene, CLCNKB, cause Bartter’s syndrome type III. Nat Genet. 1997;17:171–8. doi: 10.1038/ng1097-171. [DOI] [PubMed] [Google Scholar]
  • 20.Birkenhager R, Otto E, Schurmann MJ, Vollmer M, Ruf EM, Maier-Lutz I, et al. Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet. 2001;29:310–4. doi: 10.1038/ng752. [DOI] [PubMed] [Google Scholar]
  • 21.Geller DS, Rodriguez-Soriano J, Vallo Boado A, Schifter S, Bayer M, Chang SS, et al. Mutations in the mineralocorticoid receptor gene cause autosomal dominant pseudohypoaldosteronism type I. Nat Genet. 1998;19:279–81. doi: 10.1038/966. [DOI] [PubMed] [Google Scholar]
  • 22.Sartorato P, Lapeyraque AL, Armanini D, Kuhnle U, Khaldi Y, Salomon R, et al. Different inactivating mutations of the mineralocorticoid receptor in fourteen families affected by type I pseudohypoaldosteronism. J Clin Endocrinol Metab. 2003;88:2508–17. doi: 10.1210/jc.2002-021932. [DOI] [PubMed] [Google Scholar]
  • 23.Viemann M, Peter M, Lopez-Siguero JP, Simic-Schleicher G, Sippell WG. Evidence for genetic heterogeneity of pseudohypoaldosteronism type 1: identification of a novel mutation in the human mineralocorticoid receptor in one sporadic case and no mutations in two autosomal dominant kindreds. J Clin Endocrinol Metab. 2001;86:2056–9. doi: 10.1210/jcem.86.5.7449. [DOI] [PubMed] [Google Scholar]
  • 24.Lifton RP. Molecular genetics of human blood pressure variation. Science. 1996;272:676–80. doi: 10.1126/science.272.5262.676. [DOI] [PubMed] [Google Scholar]
  • 25.Simonetti GD, Mohaupt MG, Bianchetti MG. Monogenic forms of hypertension. Eur J Pediatr. 2012;171:1433–9. doi: 10.1007/s00431-011-1440-7. [DOI] [PubMed] [Google Scholar]
  • 26.Toka HR, Luft FC. Monogenic forms of human hypertension. Semin Nephrol. 2002;22:81–8. doi: 10.1053/snep.2002.30206. [DOI] [PubMed] [Google Scholar]
  • 27.Luft FC. Mendelian forms of human hypertension and mechanisms of disease. Clin Med Res. 2003;1:291–300. doi: 10.3121/cmr.1.4.291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Garovic VD, Hilliard AA, Turner ST. Monogenic forms of low-renin hypertension. Nat Clin Pract Nephrol. 2006;2:624–30. doi: 10.1038/ncpneph0309. [DOI] [PubMed] [Google Scholar]
  • 29.Martinez-Aguayo A, Fardella C. Genetics of hypertensive syndrome. Horm Res. 2009;71:253–9. doi: 10.1159/000208798. [DOI] [PubMed] [Google Scholar]
  • 30.Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, et al. Finding the missing heritability of complex diseases. Nature. 2009;461:747–53. doi: 10.1038/nature08494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Marteau JB, Zaiou M, Siest G, Visvikis-Siest S. Genetic determinants of blood pressure regulation. J Hypertens. 2005;23:2127–43. doi: 10.1097/01.hjh.0000186024.12364.2e. [DOI] [PubMed] [Google Scholar]
  • 32.Cheng LS, Livshits G, Carmelli D, Wahrendorf J, Brunner D. Segregation analysis reveals a major gene effect controlling systolic blood pressure and BMI in an Israeli population. Hum Biol. 1998;70:59–75. [PubMed] [Google Scholar]
  • 33.Perusse L, Moll PP, Sing CF. Evidence that a single gene with gender- and age-dependent effects influences systolic blood pressure determination in a population-based sample. Am J Hum Genet. 1991;49:94–105. [PMC free article] [PubMed] [Google Scholar]
  • 34.Levy D, DeStefano AL, Larson MG, O’Donnell CJ, Lifton RP, Gavras H, et al. 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;36:477–83. doi: 10.1161/01.hyp.36.4.477. [DOI] [PubMed] [Google Scholar]
  • 35.van Rijn MJ, Schut AF, Aulchenko YS, Deinum J, Sayed-Tabatabaei FA, Yazdanpanah M, et al. Heritability of blood pressure traits and the genetic contribution to blood pressure variance explained by four blood-pressure-related genes. J Hypertens. 2007;25:565–70. doi: 10.1097/HJH.0b013e32801449fb. [DOI] [PubMed] [Google Scholar]
  • 36.Gu D, Rice T, Wang S, Yang W, Gu C, Chen CS, et al. Heritability of blood pressure responses to dietary sodium and potassium intake in a Chinese population. Hypertension. 2007;50:116–22. doi: 10.1161/HYPERTENSIONAHA.107.088310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hottenga JJ, Whitfield JB, de Geus EJ, Boomsma DI, Martin NG. Heritability and stability of resting blood pressure in Australian twins. Twin Res Hum Genet. 2006;9:205–9. doi: 10.1375/183242706776382455. [DOI] [PubMed] [Google Scholar]
  • 38.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;45:80–5. doi: 10.1161/01.HYP.0000149952.84391.54. [DOI] [PubMed] [Google Scholar]
  • 39.Hottenga JJ, Boomsma DI, Kupper N, Posthuma D, Snieder H, Willemsen G, et al. Heritability and stability of resting blood pressure. Twin Res Hum Genet. 2005;8:499–508. doi: 10.1375/183242705774310123. [DOI] [PubMed] [Google Scholar]
  • 40.Bochud M, Bovet P, Elston RC, Paccaud F, Falconnet C, Maillard M, et al. High heritability of ambulatory blood pressure in families of East African descent. Hypertension. 2005;45:445–50. doi: 10.1161/01.HYP.0000156538.59873.86. [DOI] [PubMed] [Google Scholar]
  • 41.Fava C, Burri P, Almgren P, Groop L, Hulthen UL, Melander O. Heritability of ambulatory and office blood pressure phenotypes in Swedish families. J Hypertens. 2004;22:1717–21. doi: 10.1097/00004872-200409000-00015. [DOI] [PubMed] [Google Scholar]
  • 42.Fagard R, Brguljan J, Staessen J, Thijs L, Derom C, Thomis M, et al. Heritability of conventional and ambulatory blood pressures. A study in twins. Hypertension. 1995;26:919–24. doi: 10.1161/01.hyp.26.6.919. [DOI] [PubMed] [Google Scholar]
  • 43.Rotimi CN, Cooper RS, Cao G, Ogunbiyi O, Ladipo M, Owoaje E, et al. Maximum-likelihood generalized heritability estimate for blood pressure in Nigerian families. Hypertension. 1999;33:874–8. doi: 10.1161/01.hyp.33.3.874. [DOI] [PubMed] [Google Scholar]
  • 44.Adeyemo AA, Omotade OO, Rotimi CN, Luke AH, Tayo BO, Cooper RS. Heritability of blood pressure in Nigerian families. J Hypertens. 2002;20:859–63. doi: 10.1097/00004872-200205000-00019. [DOI] [PubMed] [Google Scholar]
  • 45.Hsueh WC, Mitchell BD, Schneider JL, Wagner MJ, Bell CJ, Nanthakumar E, et al. QTL influencing blood pressure maps to the region of PPH1 on chromosome 2q31–34 in Old Order Amish. Circulation. 2000;101:2810–6. doi: 10.1161/01.cir.101.24.2810. [DOI] [PubMed] [Google Scholar]
  • 46.Morrison AC, Cooper R, Hunt S, Lewis CE, Luke A, Mosley TH, et al. Genome scan for hypertension in nonobese African Americans: the National Heart, Lung, and Blood Institute Family Blood Pressure Program. Am J Hypertens. 2004;17:834–8. doi: 10.1016/j.amjhyper.2004.04.009. [DOI] [PubMed] [Google Scholar]
  • 47.Koivukoski L, Fisher SA, Kanninen T, Lewis CM, von Wowern F, Hunt S, et al. Meta-analysis of genome-wide scans for hypertension and blood pressure in Caucasians shows evidence of susceptibility regions on chromosomes 2 and 3. Hum Mol Genet. 2004;13:2325–32. doi: 10.1093/hmg/ddh237. [DOI] [PubMed] [Google Scholar]
  • 48.Rice T, Cooper RS, Wu X, Bouchard C, Rankinen T, Rao DC, et al. Meta-analysis of genome-wide scans for blood pressure in African American and Nigerian samples. The National Heart, Lung, and Blood Institute GeneLink Project. Am J Hypertens. 2006;19:270–4. doi: 10.1016/j.amjhyper.2005.09.006. [DOI] [PubMed] [Google Scholar]
  • 49.Mocci E, Concas MP, Fanciulli M, Pirastu N, Adamo M, Cabras V, et al. Microsatellites and SNPs linkage analysis in a Sardinian genetic isolate confirms several essential hypertension loci previously identified in different populations. BMC Med Genet. 2009;10:81. doi: 10.1186/1471-2350-10-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Simino J, Shi G, Kume R, Schwander K, Province MA, Gu CC, et al. Five blood pressure loci identified by an updated genome-wide linkage scan: meta-analysis of the Family Blood Pressure Program. Am J Hypertens. 2011;24:347–54. doi: 10.1038/ajh.2010.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Harrap SB, Wong ZY, Stebbing M, Lamantia A, Bahlo M. Blood pressure QTLs identified by genome-wide linkage analysis and dependence on associated phenotypes. Physiol Genomics. 2002;8:99–105. doi: 10.1152/physiolgenomics.00069.2001. [DOI] [PubMed] [Google Scholar]
  • 52.Hoffmann K, Planitz C, Ruschendorf F, Muller-Myhsok B, Stassen HH, Lucke B, et al. A novel locus for arterial hypertension on chromosome 1p36 maps to a metabolic syndrome trait cluster in the Sorbs, a Slavic population isolate in Germany. J Hypertens. 2009;27:983–90. doi: 10.1097/HJH.0b013e328328123d. [DOI] [PubMed] [Google Scholar]
  • 53.Puppala S, Coletta DK, Schneider J, Hu SL, Farook VS, Dyer TD, et al. Genome-wide linkage screen for systolic blood pressure in the Veterans Administration Genetic Epidemiology Study (VAGES) of Mexican-Americans and confirmation of a major susceptibility locus on chromosome 6q14.1. Hum Hered. 2011;71:1–10. doi: 10.1159/000323143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yu W, Gwinn M, Clyne M, Yesupriya A, Khoury MJ. A navigator for human genome epidemiology. Nat Genet. 2008;40:124–5. doi: 10.1038/ng0208-124. [DOI] [PubMed] [Google Scholar]
  • 55•.Johnson AD, Newton-Cheh C, Chasman DI, Ehret GB, Johnson T, Rose L, et al. Association of hypertension drug target genes with blood pressure and hypertension in 86,588 individuals. Hypertension. 2011;57:903–10. doi: 10.1161/HYPERTENSIONAHA.110.158667. This large-scale study indicates that candidate genes, with clinical and physiological relevance by virtue of their role as antihypertensive drug targets, harbor true blood pressure related variants. The variants on the genes ADRB1 and AGT even reached the genome-wide significance. Thus, the author suggested that revisiting genome-wide association studies from the perspective of biological and clinical knowledge might be useful for discovery and validation of new genetic associations. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Takeuchi F, Yamamoto K, Katsuya T, Sugiyama T, Nabika T, Ohnaka K, et al. Reevaluation of the association of seven candidate genes with blood pressure and hypertension: a replication study and meta-analysis with a larger sample size. Hypertens Res. 2012;35:825–31. doi: 10.1038/hr.2012.43. [DOI] [PubMed] [Google Scholar]
  • 57•.Ganesh SK, Tragante V, Guo W, Guo Y, Lanktree MB, Smith EN, et al. Loci influencing blood pressure identified using a cardiovascular gene-centric array. Hum Mol Genet. 2013;22:1663–78. doi: 10.1093/hmg/dds555. This is a large association study using a cardiovasuclar gene-centric array, examing ~ 50,000 single-nucleotide polymorphisms from ~2,100 candidate genes for cardiovascular phenotypes in 127, 505 individuals of European ancestry. It identified two novel loci associated with blood pressure. This study highlights the potential of candidiate gene studies in identifying genetic factors for complex disease by the use of strigent methodologies. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Johnson T, Gaunt TR, Newhouse SJ, Padmanabhan S, Tomaszewski M, Kumari M, et al. Blood pressure loci identified with a gene-centric array. Am J Hum Genet. 2011;89:688–700. doi: 10.1016/j.ajhg.2011.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Samani NJ, Erdmann J, Hall AS, Hengstenberg C, Mangino M, Mayer B, et al. Genomewide association analysis of coronary artery disease. N Engl J Med. 2007;357:443–53. doi: 10.1056/NEJMoa072366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science. 2007;316:889–94. doi: 10.1126/science.1141634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hiura Y, Tabara Y, Kokubo Y, Okamura T, Miki T, Tomoike H, et al. A genome-wide association study of hypertension-related phenotypes in a Japanese population. Circ J. 2010;74:2353–9. doi: 10.1253/circj.cj-10-0353. [DOI] [PubMed] [Google Scholar]
  • 62.Padmanabhan S, Melander O, Johnson T, Di Blasio AM, Lee WK, Gentilini D, et al. Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet. 2010;6:e1001177. doi: 10.1371/journal.pgen.1001177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Salvi E, Kutalik Z, Glorioso N, Benaglio P, Frau F, Kuznetsova T, et al. 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. 2012;59:248–55. doi: 10.1161/HYPERTENSIONAHA.111.181990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.White PC. Disorders of aldosterone biosynthesis and action. N Engl J Med. 1994;331:250–8. doi: 10.1056/NEJM199407283310408. [DOI] [PubMed] [Google Scholar]
  • 65.Qian X, Lu Z, Tan M, Liu H, Lu D. A meta-analysis of association between C677T polymorphism in the methylenetetrahydrofolate reductase gene and hypertension. Eur J Hum Genet. 2007;15:1239–45. doi: 10.1038/sj.ejhg.5201914. [DOI] [PubMed] [Google Scholar]
  • 66.Monteith GR, Kable EP, Kuo TH, Roufogalis BD. Elevated plasma membrane and sarcoplasmic reticulum Ca2+ pump mRNA levels in cultured aortic smooth muscle cells from spontaneously hypertensive rats. Biochem Biophys Res Commun. 1997;230:344–6. doi: 10.1006/bbrc.1996.5956. [DOI] [PubMed] [Google Scholar]
  • 67.Wang X, Chua HX, Chen P, Ong RT, Sim X, Zhang W, et al. Comparing methods for performing trans-ethnic meta-analysis of genome-wide association studies. Hum Mol Genet. 2013;22:2303–11. doi: 10.1093/hmg/ddt064. [DOI] [PubMed] [Google Scholar]
  • 68.Morris AP. Transethnic meta-analysis of genomewide association studies. Genet Epidemiol. 2011;35:809–22. doi: 10.1002/gepi.20630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Franceschini N, van Rooij FJ, Prins BP, Feitosa MF, Karakas M, Eckfeldt JH, et al. Discovery and fine mapping of serum protein loci through transethnic meta-analysis. Am J Hum Genet. 2012;91:744–53. doi: 10.1016/j.ajhg.2012.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Murcray CE, Lewinger JP, Gauderman WJ. Gene-environment interaction in genome-wide association studies. Am J Epidemiol. 2009;169:219–26. doi: 10.1093/aje/kwn353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bloom JS, Ehrenreich IM, Loo WT, Lite TL, Kruglyak L. Finding the sources of missing heritability in a yeast cross. Nature. 2013;494:234–7. doi: 10.1038/nature11867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Murcray CE, Lewinger JP, Conti DV, Thomas DC, Gauderman WJ. Sample size requirements to detect gene-environment interactions in genome-wide association studies. Genet Epidemiol. 2011;35:201–10. doi: 10.1002/gepi.20569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Baccarelli A, Rienstra M, Benjamin EJ. Cardiovascular epigenetics: basic concepts and results from animal and human studies. Circ Cardiovasc Genet. 2010;3:567–73. doi: 10.1161/CIRCGENETICS.110.958744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cowley AW, Jr, Nadeau JH, Baccarelli A, Berecek K, Fornage M, Gibbons GH, et al. Report of the National Heart, Lung, and Blood Institute Working Group on epigenetics and hypertension. Hypertension. 2012;59:899–905. doi: 10.1161/HYPERTENSIONAHA.111.190116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Millis RM. Epigenetics and hypertension. Curr Hypertens Rep. 2011;13:21–8. doi: 10.1007/s11906-010-0173-8. [DOI] [PubMed] [Google Scholar]
  • 76.Friso S, Pizzolo F, Choi SW, Guarini P, Castagna A, Ravagnani V, et al. Epigenetic control of 11 beta-hydroxysteroid dehydrogenase 2 gene promoter is related to human hypertension. Atherosclerosis. 2008;199:323–7. doi: 10.1016/j.atherosclerosis.2007.11.029. [DOI] [PubMed] [Google Scholar]
  • 77.Smolarek I, Wyszko E, Barciszewska AM, Nowak S, Gawronska I, Jablecka A, et al. Global DNA methylation changes in blood of patients with essential hypertension. Med Sci Monit. 2010;16:CR149–55. [PubMed] [Google Scholar]
  • 78.Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK, Hobbs HH. Low LDL cholesterol in individuals of African descent resulting from frequent nonsense mutations in PCSK9. Nat Genet. 2005;37:161–5. doi: 10.1038/ng1509. [DOI] [PubMed] [Google Scholar]
  • 79.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;305:869–72. doi: 10.1126/science.1099870. [DOI] [PubMed] [Google Scholar]
  • 80.Cohen JC, Pertsemlidis A, Fahmi S, Esmail S, Vega GL, Grundy SM, et al. Multiple rare variants in NPC1L1 associated with reduced sterol absorption and plasma low-density lipoprotein levels. Proc Natl Acad Sci U S A. 2006;103:1810–5. doi: 10.1073/pnas.0508483103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ji W, Foo JN, O’Roak BJ, Zhao H, Larson MG, Simon DB, et al. Rare independent mutations in renal salt handling genes contribute to blood pressure variation. Nat Genet. 2008;40:592–9. doi: 10.1038/ng.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rao F, Wen G, Gayen JR, Das M, Vaingankar SM, Rana BK, et al. Catecholamine release-inhibitory peptide catestatin (chromogranin A(352–372)): naturally occurring amino acid variant Gly364Ser causes profound changes in human autonomic activity and alters risk for hypertension. Circulation. 2007;115:2271–81. doi: 10.1161/CIRCULATIONAHA.106.628859. [DOI] [PubMed] [Google Scholar]
  • 83.Metzker ML. Sequencing technologies - the next generation. Nat Rev Genet. 2010;11:31–46. doi: 10.1038/nrg2626. [DOI] [PubMed] [Google Scholar]
  • 84.Tennessen JA, Bigham AW, O’Connor TD, Fu W, Kenny EE, Gravel S, et al. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science. 2012;337:64–9. doi: 10.1126/science.1219240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mailman MD, Feolo M, Jin Y, Kimura M, Tryka K, Bagoutdinov R, et al. The NCBI dbGaP database of genotypes and phenotypes. Nat Genet. 2007;39:1181–6. doi: 10.1038/ng1007-1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Delles C, Padmanabhan S. Genetics and hypertension: is it time to change my practice? Can J Cardiol. 2012;28:296–304. doi: 10.1016/j.cjca.2012.02.004. [DOI] [PubMed] [Google Scholar]
  • 87.Kathiresan S, Melander O, Guiducci C, Surti A, Burtt NP, Rieder MJ, et al. Six new loci associated with blood low-density lipoprotein cholesterol, high-density lipoprotein cholesterol or triglycerides in humans. Nat Genet. 2008;40:189–97. doi: 10.1038/ng.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Law MR, Wald NJ, Rudnicka AR. Quantifying effect of statins on low density lipoprotein cholesterol, ischaemic heart disease, and stroke: systematic review and meta-analysis. BMJ. 2003;326:1423. doi: 10.1136/bmj.326.7404.1423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sanseau P, Agarwal P, Barnes MR, Pastinen T, Richards JB, Cardon LR, et al. Use of genome-wide association studies for drug repositioning. Nat Biotechnol. 2012;30:317–20. doi: 10.1038/nbt.2151. [DOI] [PubMed] [Google Scholar]
  • 90••.Fava C, Sjogren M, Montagnana M, Danese E, Almgren P, Engstrom G, et al. Prediction of blood pressure changes over time and incidence of hypertension by a genetic risk score in Swedes. Hypertension. 2013;61:319–26. doi: 10.1161/HYPERTENSIONAHA.112.202655. This study longitudinally confirmed the role of the genetic variants identified by genome-wide association studies in blood pressure changes and hypertension incidence over time. This type of study is very necessary to validate blood pressure-related genetic variants identified by cross-sectional studies and to assess the hypertension prediction value of these variants. [DOI] [PubMed] [Google Scholar]
  • 91.Konoshita T. Do genetic variants of the Renin-Angiotensin system predict blood pressure response to Renin-Angiotensin system-blocking drugs?: a systematic review of pharmacogenomics in the Renin-Angiotensin system. Curr Hypertens Rep. 2011;13:356–61. doi: 10.1007/s11906-011-0212-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Liu J, Liu ZQ, Yu BN, Xu FH, Mo W, Zhou G, et al. beta1-Adrenergic receptor polymorphisms influence the response to metoprolol monotherapy in patients with essential hypertension. Clin Pharmacol Ther. 2006;80:23–32. doi: 10.1016/j.clpt.2006.03.004. [DOI] [PubMed] [Google Scholar]
  • 93.Johnson JA, Zineh I, Puckett BJ, McGorray SP, Yarandi HN, Pauly DF. Beta 1-adrenergic receptor polymorphisms and antihypertensive response to metoprolol. Clin Pharmacol Ther. 2003;74:44–52. doi: 10.1016/S0009-9236(03)00068-7. [DOI] [PubMed] [Google Scholar]
  • 94.Vandell AG, Lobmeyer MT, Gawronski BE, Langaee TY, Gong Y, Gums JG, et al. G protein receptor kinase 4 polymorphisms: beta-blocker pharmacogenetics and treatment-related outcomes in hypertension. Hypertension. 2012;60:957–64. doi: 10.1161/HYPERTENSIONAHA.112.198721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Manunta P, Lavery G, Lanzani C, Braund PS, Simonini M, Bodycote C, et al. Physiological interaction between alpha-adducin and WNK1-NEDD4L pathways on sodium-related blood pressure regulation. Hypertension. 2008;52:366–72. doi: 10.1161/HYPERTENSIONAHA.108.113977. [DOI] [PubMed] [Google Scholar]
  • 96.Turner ST, Bailey KR, Fridley BL, Chapman AB, Schwartz GL, Chai HS, et al. Genomic association analysis suggests chromosome 12 locus influencing antihypertensive response to thiazide diuretic. Hypertension. 2008;52:359–65. doi: 10.1161/HYPERTENSIONAHA.107.104273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Johnson JA, Boerwinkle E, Zineh I, Chapman AB, Bailey K, Cooper-DeHoff RM, et al. Pharmacogenomics of antihypertensive drugs: rationale and design of the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR) study. Am Heart J. 2009;157:442–9. doi: 10.1016/j.ahj.2008.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Johnson JA. Ethnic differences in cardiovascular drug response: potential contribution of pharmacogenetics. Circulation. 2008;118:1383–93. doi: 10.1161/CIRCULATIONAHA.107.704023. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES