Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: Hypertension. 2018 Dec;72(6):1244–1254. doi: 10.1161/HYPERTENSIONAHA.118.11171

Epigenetic Mechanisms and Hypertension

Mingyu Liang 1
PMCID: PMC6314488  NIHMSID: NIHMS1506713  PMID: 30571238

Epigenetic mechanisms contribute to the regulation of physiology and disease by changing gene expression, and epigenetic changes occur as a result of the interplay between DNA and environmental factors (Figure 1). Unlike permanent changes in DNA sequences that change amino acid sequences or gene expression, epigenetic regulation can respond dynamically to environmental and lifestyle factors. On the other hand, this regulation may have more stable, longer-lasting effects than do transient changes in gene expression or cellular regulation independent of changes in gene expression (Figure 1).

Figure 1. Epigenetic mechanisms’ role in disease mechanisms.

Figure 1.

A disease can arise from the expression of mutated proteins, stable changes in gene expression levels, or other abnormalities, including transient changes in gene expression, post-transcriptional or post-translational changes in proteins and changes in other chemical or physical factors. Stable changes in gene expression can occur because of variations in DNA regulatory sequences or effects of environment and lifestyle factors. Epigenetic mechanisms underlie stable changes in DNA function that variations in the sequence of the DNA do not determine directly. Such changes can respond to environment and lifestyle factors dynamically and can result from DNA-environment interactions.

The fundamental importance of epigenetic mechanisms in biology is illustrated best by striking epigenetic phenomena, such as the presence of roughly 200 cell types in the human body that are distinct molecularly and phenotypically, although nearly all of them share identical DNA sequences. Another striking illustration of epigenetic mechanisms’ biological importance is the inactivation of one of the X chromosomes in female cells.

Increasingly strong evidence supports epigenetic mechanisms’ significant role in the development of hypertension. Nevertheless, many fundamental questions remain to be addressed. In this review, I present a tiered approach to classifying evidence of epigenetic regulation and provide a brief overview of the biology of epigenetic mediators and the methods used to study them. I then summarize reports of changes in epigenetic mediators that occur in association with changes in blood pressure (BP) or hypertension. Next, I highlight emerging evidence of epigenetic mediators’ functional role in the development of hypertension. Finally, I outline key questions in the field and propose resources, technologies, and the concepts of a tree-like paradigm and systems molecular medicine that could help advance epigenetic research on hypertension.

I. A Tiered View of the Evidence for Epigenetic Regulation

Researchers in the epigenetics field still grapple with what qualifies as an epigenetic mechanism.1, 2, 3 Most investigators now agree to use the term epigenetics to refer to stable changes in DNA function that are not determined directly by the sequence of the DNA. However, what “stable” means is not always clear. Some would say it indicates that changes must be heritable across multiple generations of the organism, while many others would say the changes have to be heritable only through cell division. However, it can be challenging to test whether a change in DNA function is heritable, even through cell division alone, especially those changes that occur in mammalian species in vivo.

Rather than engaging in another debate about the definition of epigenetics, I propose a practical, tiered approach to examining evidence for epigenetic regulation in this article (Box). Tier 1 includes any evidence that suggests the involvement of any mediators that could underlie stable changes in DNA function, such as DNA methylation, histone modifications, non-coding RNA that regulates transcription, and chromatin conformation.

Box. A three-tier view of the evidence for epigenetic regulation.

Refer to the text for additional details.

Tier 1: Stable changes of genome function not determined directly by changes in DNA sequence

Regulatory mediators:

DNA methylation (mechanisms for reaching Tier 2 are known)

Histone modification

Regulatory RNA

Chromatin conformation

Tier 2: Tier 1 and heritable through mitosis

Examples:

Some long-term effects of early-life experiences

Some maternal effects

Tier 3: Tier 1 and heritable through germ cell reprogramming

Examples:

Transgenerational effects

Some paternal effects

Evidence would be considered as Tier 2 if heritability through cell division is proven. The mechanism underlying the inheritance of DNA methylation marks through cell division is known.4 During DNA replication, DNA methyltransferase 1 (DNMT1), together with other proteins, recognize methylated cytosines in the template strand of DNA and catalyze cytosine methylation in the DNA strand newly synthesized. Therefore, DNA methylation can be considered heritable through cell division unless proven otherwise. Mechanisms that mediate the inheritance of most histone marks, non-coding RNA-determined transcriptional activities, or chromatin conformation through cell division, have not been established.5

Physiological studies sometimes provide indirect or suggestive evidence for the involvement of epigenetic mechanisms across cell divisions. For example, the maternal environment and early-life experiences are known to influence physiology or diseases in adulthood (Box),6 and it has been shown that some of these effects are associated with changes in the epigenetic mediators listed under Tier 17,8. However, whether epigenetic mechanisms contribute to these effects functionally largely remains unknown.

Tier 3 refers to epigenetic changes that are heritable through germ cell reprogramming (Box). Germ cells in species such as humans, mice, and rats, undergo two rounds of epigenetic reprogramming during early embryonic development: one after fertilization and the other during the formation of primordial germ cells.9, 10 This reprogramming, which was believed once to involve the complete erasure of epigenetic marks, could leave behind some marks.11,12,13 The residual marks, perhaps working with trans regulatory factors, such as RNA,14,15 could serve as the molecular basis of epigenetic memory following germ cell reprogramming.

A special type of epigenetic inheritance is transgenerational inheritance.9 An F0 generation’s environment during pregnancy can influence primordial germ cells in the embryo that become germ cells in F1 and individual organisms in F2. Therefore, typically, an epigenetic trait would have to be inherited from F0 to at least F3 to be considered inherited trans-generationally rather than a direct effect of the initial environmental exposure. However, fewer generations are required if the initial environmental exposure is limited to the paternal side or occurs before the formation of primordial germ cells on the maternal side.16

The Basic Biology of Epigenetic Mediators

II.1. DNA methylation

DNA methylation occurs primarily at cytosine in cytosine-guanine dinucleotides (CpG), and involves the addition of a methyl group to carbon number 5 in cytosine to form 5-methylcytosine (5mC).17, 18 Some parts of the genome are methylated heavily and remain in the transcriptionally silent state of heterochromatin generally. In other parts of the genome, the methylation levels can change dynamically in response to developmental or environmental cues. The level of methylation, particularly that of CpG islands (clusters of CpG sites) located in promoter regions, may be correlated inversely with the level of gene transcription activity. However, the relation between methylation and expression levels can be complex. 19,20,21 In addition, DNA methylation also can occur at cytosine in CpH (H=A, C or T).22

DNMT catalyzes the formation of 5mC.4, 23 As described above, DNMT1 is responsible primarily for maintaining DNA methylation patterns during DNA replication and cell division. DNMT3, which includes DNMT3A, 3B, and 3L, mediates de novo formation of 5mC. DNMT3A and 3B are catalytic and can methylate overlapping, but not identical, genomic loci. DNMT2 might be involved in tRNA methylation. Ten-eleven translocases (TET), which include TET1, 2, and 3, catalyze DNA de-methylation.24 TETs convert 5mC to 5-hydroxymethylcytosine (5hmC), which can be converted further to 5-formylcytosine or 5-carboxylcytosine and replaced with unmodified C. These three TET isoforms act on overlapping genomic loci.

II.2. Histone modifications

DNA wraps around eight histone proteins, two each of H2A, H2B, H3, and H4, to form nucleosomes. The N-terminal tails of H3 and H4 are subject to post-translational modifications that influence chromatin structure and gene expression.25,26 Common types of modification include acetylation at select lysine (K) residues, methylation at K or arginine residues, and phosphorylation at serine or threonine residues. Ubiquitylation and sumoylation also can occur.

Histone acetyltransferases (HATs) catalyze histone acetylation using acetyl CoA as the source of the acetyl group. Histone deacetylases (HDACs) catalyze the hydrolytic removal of acetyl groups, histone methyltransferases (HMTs) catalyze the transfer of one to three methyl groups from S-adenosyl-L-methionine to histones, and histone demethylases (HDMTs) remove methyl groups from histones. Each of these enzyme families includes multiple sub-families or members that target different amino acid residues or require different cofactors. Pharmacological inhibitors are available for several of these enzymes.27,28

II.3. Non-coding RNA

The role of non-coding RNAs, such as microRNAs, in post-transcriptional regulation of mRNA stability and translation has been well-established. However, a molecule’s participation in posttranscriptional and translational regulation is not a sufficient basis for considering the molecule a bona fide epigenetic regulator. While several microRNAs have been reported to influence gene transcription activities and potentially could be involved in epigenetic regulation or inheritance, evidence that supports microRNAs’ contribution to stable changes in chromatin structure and genome function is limited in mammalian species. Long non-coding RNAs (lncRNAs) also participate in post-transcriptional regulation. However, some lncRNAs are well-established regulators of gene transcription activities, and part of this regulation might occur through epigenetic mechanisms.29, 30 For example, lncRNA Xist plays a key role in X chromosome inactivation.

Accordingly, this review will cover studies of lncRNAs in hypertension, but recognize that the mechanism underlying the physiological role of lncRNAs is not always clear and could involve both epigenetic and non-epigenetic actions. The reader is referred to other reviews for microRNAs’ role in hypertension31,32.

II.4. Chromatin conformation

Chromatin conformation influences gene transcription by changing the spatial organization and accessibility of DNA regulatory elements.33, 34 DNA segments, such as enhancers and promoters, interact more frequently within than across topologically associating domains (TAD). A TAD may contain multiple DNA loops or sub-TADs ranging from thousands to millions of bp in size. CCCTC-binding factors (CTCF) that form homodimers and interact with other proteins, such as cohesin, maintain the DNA loops at the anchoring point. An important consequence of DNA looping is that enhancers or super-enhancers located thousands of bp from a promoter can be brought within its spatial proximity and regulate the transcriptional activity there. Further, some chromatin conformations bring DNA segments on different chromosomes together spatially.

Epigenetic regulatory mediators can influence each other. Changes in DNA methylation can induce changes in histone modifications, and vice versa. These changes, together with the assembly or disassembly of molecular complexes they induce, often change gene transcriptional activities by changing chromatin conformation and making regulatory DNA elements more or less accessible or active.

Methods Used to Study Epigenetics

Some of the major methods that can be used to detect and quantify epigenetic mediators and features, or study their functional roles are summarized in Table 1. For a more detailed description of several of these techniques, refer to reviews and methods published elsewhere.3550 The method chosen for a study depends on several factors, including whether the study’s focus is genome-wide or on specific loci and the availability of expertise and resources.

Table 1.

Major Methods Used to Study Epigenetic Regulation.

Goal of study Epigenetic
mark
Major Methods Notes
Describing epigenetic mediators DNA methylation Whole genome bisulfite sequencing Bisulfite treatment converts C, but not methylated C, to U. Single base resolution.
RRBS and variants Digestion-based enrichment, followed by bisulfite deep sequencing
MeDIP and similar methods Affinity enrichment, followed by deep sequencing or array-based analysis
Cloning-based bisulfite sequencing To analyze specific genomic loci
Methylation-specific PCR To analyze specific genomic loci
Histone modification ChIP-seq Identifying DNA segments bound by a histone with specific modifications
Regulatory RNA RNA-seq or real-time PCR Some lncRNAs do not have a poly(A) tail.
Chromatin conformation 3C, 4C, Hi-C and variants Mapping genomic proximity contacts globally or from a specific viewpoint
Chromatin accessibility DNase-seq, ATAC-seq Identifying open chromatin
Testing functional role DNA methylation Pharmacological inhibitors Inhibitors used commonly require DNA replication.
Silencing or targeting DNMTs and TETs
Targeted (de)methylation at specific genomic loci Frequently adapting DNA recognition agents developed for genome editing
Histone modification Pharmacological inhibitors Some histone modifying enzymes also can modify non-histone proteins.
Silencing or targeting histone modifying enzymes
Regulatory RNA Modified anti-sense or genetic targeting Some lncRNAs are located in the nucleus.
Examining transgenerational inheritance Multi-generation breeding The number of generations required could be reduced by using paternal or controlled maternal exposure.

RRBS, reduced representation bisulfite sequencing; MeDIP, methylated DNA immunoprecipitation; ChIP-seq, chromatin immunoprecipitation sequencing; 3C, chromatin conformation capture; ATAC, assay for transposase-accessible chromatin.

A key consideration in epigenetic analysis is cell type, because while some epigenetic features are shared across cell types, other features vary. In that respect, epigenetic analysis is similar to the analysis of gene expression and unlike genomic sequencing, although epigenetic analysis often involves DNA analysis. In some cases, epigenetic features detected in one cell or tissue type might serve as biomarkers or surrogates for epigenetic features of another, less accessible cell or tissue type. However, it is important to use caution in such extrapolations, especially in studies of a disease’s cellular mechanisms.

Epigenome-wide association studies (EWAS) can be performed to examine the association of epigenetic marks, especially DNA methylation, with a phenotype.51 The sample size EWAS requires depends on the degree of epigenetic variation and its effect size, but often, it is large and similar to genome-wide association studies (GWAS). In part because of the requirement for large sample sizes, EWAS have often used DNA from accessible peripheral blood. When interpreting results from such studies, it is important to distinguish biomarkers from mechanistic mediators and consider the tissue specificity of some epigenetic marks and whether blood cells are pathophysiologically relevant to the disease of interest.

It is important as well to consider the mechanisms of action and specificity when pharmacological inhibitors of enzymes that mediate changes of DNA methylation and histone modification are used (Table 1). DNA methylation inhibitors used commonly, such as 5-aza-2’-deoxycytidine (decitabine), are cytosine analogs that will prevent methylation after they are incorporated into DNA strands newly synthesized during DNA replication and cell division.52 Therefore, these inhibitors do not prevent de novo DNA methylation that occurs in non-dividing cells effectively. Most of the HDAC inhibitors used commonly inhibit multiple HDACs.28 Moreover, the substrates of HDACs are not limited to histones, and inhibition of HDACs could alter the acetylation of non-histone proteins in addition to histones.

Association of Epigenetic Mediators with BP or Hypertension

Because of space limitations, this review focuses only on epigenetic mechanisms in the context of the pathogenesis of systemic hypertension, and does not address epigenetic mechanisms in pulmonary arterial hypertension or hypertensive end organ damage.

This section summarizes studies that have tested associations between changes in epigenetic mediators and BP or hypertension. Such evidence is available from both human and animal studies. Human studies can test the clinical relevance of epigenetic mediators directly, while animal studies may provide additional mechanistic insights by analyzing BP-relevant tissues that are less accessible in humans.

It is important to recognize that correlation does not prove causality. Studies that have demonstrated a functional role of epigenetic mediators in the regulation of BP or the development of hypertension will be reviewed in a later section in this article. Nonetheless, evidence of association is valuable, especially for epigenetic research of hypertension in its current stage of development, as it can provide biomarkers for diagnostic purposes and a basis for developing mechanistic studies.

IV.1. DNA methylation

IV.1a. Human studies

BP variations and the development of hypertension have been associated with changes in DNA methylation in several human studies. Richard et al., analyzed cross-sectional associations of systolic and diastolic BP with blood-derived genome-wide DNA methylation measured in 17,010 individuals of European, African American, and Hispanic ancestry,53 and identified 13 replicated CpG sites that were associated significantly with BP. These replicated methylation sites explained 1.4% and 2.0% of the interindividual variations in systolic and diastolic BP, respectively, in addition to what known BP genetic variants can explain. In a GWAS of BP in up to 320,251 individuals of East Asian, European, and South Asian ancestry, the sentinel single nucleotide polymorphisms (SNPs) identified are enriched for association with DNA methylation at multiple CpG sites nearby.54 A study of hypertensive and normotensive subjects reported possible differential methylation of some CpG sites in leukocytes.55

Prenatal exposure to air pollutants was associated with lower or higher DNA methylation in long interspersed nuclear elements (LINE1) in newborn blood spots and higher systolic BP in 11-year-old children, depending on the type of pollutant and timing of the exposure.56 The magnitude of associations varied according to genotype for 11 SNPs within DNMT1, DNMT3B, TET2, and thymine DNA glycosylase genes. Bellavia et al. exposed 15 healthy adult participants to concentrated ambient particles for 130 minutes,57 and found that decreased Alu and TLR4 methylation in the blood was associated with higher diastolic BP postexposure and decreased TLR4 methylation was associated with higher systolic BP postexposure.

A study of 1,160 DNA trios from two established birth cohorts (the Cambridge Baby Growth and Wellbeing Studies) and 1367 Hyperglycemia and Adverse Pregnancy Outcome Study participants identified significant associations between polymorphic variants in several fetal imprinted genes and maternal mean arterial BP.58

IV.1b. Animal model studies

Changes in DNA methylation have been associated with hypertension in several animal models used to study the effect of maternal diet on offspring health. Restricting the supply in the periconceptional diet of mature female sheep of methionine, vitamin B12, and folate, all of which are relevant to the methyl cycle metabolism, resulted in adult offspring with elevated BP, most obviously in males. The methylation status of 4% of 1,400 CpG islands examined in the fetal liver was altered, again, particularly in males.59 In Sprague-Dawley rats, feeding the parents a vitamin-D depleted diet resulted in increased BP in the offspring, which was accompanied by hypermethylation of the promoter region of the Pannexin-1 (Panx1) gene in the kidney.60

Embryo transfer experiments have shown that feeding Dahl salt-sensitive SS rats the grain-based 5L2F diet during gestation and lactation attenuated salt-induced hypertension in the offspring substantially compared to the casein-based AIN-76A diet. 61 Several hundred genomic loci exhibited differential methylation in the renal outer medulla of SS rats fed the two diets.62

Liu et al. reported the first base resolution maps of 5mC and 5hmC in a model of hypertension.20 They found that 11% and 1% of CpG sites in the renal outer medulla of SS rats that fell within CpG islands contained significant 5mC and 5hmC, respectively. Feeding SS rats and congenic, less salt-sensitive SS.13BN26 rats a high-salt diet for just seven days changed the 5mC levels of several hundred CpG islands and the 5hmC levels of several dozen islands in a rat strain-specific manner. Many of the islands methylated differentially were intragenic. The study by Liu et al. also was one of the first to report 5hmC maps in any in vivo disease model.

DNA methylation may regulate several genes relevant to BP regulation. For example, the expression pattern of 11β-hydroxysteroid dehydrogenase type 2 (HSD11B2), which is crucial to prevent abnormal stimulation of mineralocorticoid receptors by glucocorticoids in the distal nephron, is correlated inversely with the degree of methylation at its promoter region.63 Other examples of BP-relevant genes that may be regulated by DNA methylation include angiotensinogen64 and αENaC,65 as well as glucocorticoid receptor66 and angiotensin type 1 receptor67, 68 as reviewed previously.16

IV.1c. Studies related to preeclampsia

Preeclampsia is associated with changes in methylation in humans and animal models. A CpG island in the promoter region of serine protease inhibitor A3 is hypomethylated in placentas from patients with preeclampsia and intrauterine growth restriction, which is associated with differential expression of the gene.69

DNA methylation of the promoter of the thromboxane synthase gene TBXAS1 was lower in omental arteries of preeclamptic women, which is associated with up-regulation of thromboxane synthase expression. Treatment with 5-aza-2’-deoxycytidine increased the expression of thromboxane synthase in a neutrophil-like cell line.70

Methylation levels at IGF2 in umbilical cord blood from infants born to women with preeclampsia were significantly lower than those from a normal pregnancy.71 Preeclamptic placenta exhibited DNMT3A dysregulation and a low promoter methylation and high protein expression of insulin-like growth factor-binding protein 5 (IGFBP5). DNMT3A knockdown caused promoter hypomethylation and up-regulation of IGFBP5 and inhibited trophoblast cell migration and invasion, the latter of which IGFBP5 downregulation reversed.72

Children of mothers with preeclampsia and those conceived by assisted reproductive technologies (ART) display vascular dysfunctions. ART mice and the progeny of male ART mice also exhibit vascular dysfunction and higher arterial BP. Methylation at the promoter of the eNOS gene in the aorta increased, and eNOS expression in the carotid artery and NO metabolites in the plasma decreased in ART mice. Butyrate, which is a deacetylase inhibitor, can reverse these changes, suggesting that a cross-talk between DNA methylation and protein acetylation is involved.73

CpG methylation at some sites in the promoter region of estrogen receptor-α and large-conductance Ca2+-activated potassium channel β1 subunit (BKβ1) in sheep uterine arteries was regulated differentially during pregnancy or chronic hypoxia during gestation,74, 75 and treatment with 5-aza-2’-deoxycytidine reversed the effects of hypoxia.76 Pregnancy also increases TET1 expression in uterine arteries in sheep, and treating isolated uterine arteries with the TET inhibitor monomethyl fumarate reversed several effects of pregnancy.77

IV.2. Histone modifications

Several treatments or genetic perturbations that change BP have been shown to change histone modifications. In mice, deletion of Af17, a putative transcription factor, leads to increased dimethylation of histone H3K79, reduced ENaC function, higher urine volume and sodium excretion, and lower BP.78 Stimulation of β2-adrenergic receptor in mice decreases the expression of WNK4, a regulator of sodium reabsorption, which contributes to the development of salt-sensitive hypertension in mice and rats.79 Stimulation of β2-adrenergic receptor in mouse cultured distal convoluted tubule cells results in cyclic AMP-dependent inhibition of histone deacetylase-8 (HDAC8) activity, increased histone acetylation, and the glucocorticoid receptor binding to a negative glucocorticoid-responsive element in the promoter region of WNK4.79

Treating spontaneously hypertensive rats (SHR) with pentaerythritol tetranitrate during pregnancy and lactation reduced the female offspring’s BP, and several potentially anti-hypertensive genes are upregulated in the aorta of eight-month-old female offspring, which is associated with enhanced H3K27 acetylation and H3K4 trimethylation.80 In mice, the expression of hGRK4γ(142V), a variant of G-protein-coupled receptor kinase type 4, results in hypertension. The hGRK4γ(142V) phosphorylates histone deacetylase type 1, promotes its nuclear export to the cytoplasm, and increases expression of angiotensin II type I receptor.81 db/db mice, which exhibit increased BP in addition to diabetes, express more RAGE and PAI-1 in glomeruli than do control db/+ mice. This is associated with increased RNA polymerase II recruitment and permissive histone marks and decreased repressive histone marks at these genes, as well as altered expression of relevant histone modification enzymes.82

Histone acetylation and acetyltransferases have been reported to be important in determining renin cell identity.83, 84 The histone acetyltransferase activity of p300 contributes to its effect of increasing the promoter activity of guanylyl cyclase/natriuretic peptide receptor-A gene (Npr1).85 All-trans retinoic acid increases H3 and H4 acetylation, enhances their recruitment to Ets-1 and Sp1 binding sites within the Npr1 promoter, and increases promoter activity.86 Transcriptional factor Yin Yang 1 associates with histone deacetylase 2 and increases the promoter activity of human B-type natriuretic peptide gene, while treatment with the histone deacetylase inhibitor trichostatin A decreases hBNP reporter activity.87 Other examples of BP-relevant genes that histone modifications may regulate include ENaC88, 89 and Angiotensin converting enzyme 1,90 as reviewed previously.16

IV.3. lncRNA

Several dozen lncRNAs are expressed differentially in the renal outer medulla in SS and SS.13BN26 rats fed a high-salt diet.91 Two of these are located within the congenic region of SS.13BN26. Several hundred lncRNAs are expressed differentially in the kidney between SS and Dahl salt-resistant rats or SHR92 or in the aorta between SHR and Wistar-Kyoto rats.93

Several lncRNAs participate in the regulation of processes relevant to BP regulation.94 For example, treating rats’ vascular smooth muscle cells with angiotensin II results in the differential expression of several lncRNAs.95 LncRNA hexokinase 2 pseudogene 1 (HK2P1) is dysregulated in the decidua of severe preeclampsia patients, and HK2P1 promotes human endometrial stromal cells’ proliferation and differentiation and appears to regulate hexokinase 2 expression via competition for the shared miR-6887-3p.96

IV.4. Chromatin conformation

Stodola et al. used a 4C-seq approach to map the chromatin conformation in the physical proximity of the proximal promoter of the renin gene Ren in rat cardiac endothelial cells.97 Many of the contact regions are located on rat chromosome 13, which harbors Ren, including a Ren enhancer 6 kbp from the proximal promoter identified previously (Figure 2). Other contact regions are distributed across all chromosomes except chromosome X. These contact regions are enriched significantly for genes of which the expression is correlated with Ren and for quantitative trait loci associated with BP, cardiovascular, and renal phenotypes.

Figure 2. Several DNA regions spanning chromosome 13 are in physical proximity of the renin proximal promoter.

Figure 2.

The contact map shown derives from a 4C-seq analysis of rat cardiac microvascular endothelial cells. Top: Blue lines indicate contacts between Ren proximal promoter and distant interacting regions on chromosome 13. Bottom: Contacts with the Ren proximal promoter within 1 Mb of the viewpoint. Location of the Ren proximal promoter viewpoint is indicated with a dashed red line. Red fragments labeled “Exact Interaction” have significant contact with the Ren proximal promoter in multiple samples analyzed. Reproduced from Stodola et al.97

A genetic fine mapping analysis prioritizes rs9349379, a common and intronic SNP, as the putative causal variant in a locus that has been associated with hypertension and four other vascular diseases.98 Genome editing in cultured cells demonstrates that rs9349379 regulates the expression of the endothelin 1 (EDN1) gene located 600 kb upstream of the genetic locus. Chromatin conformation mapping using 4C-seq did not identify substantial contacts between rs9349379 and EDN1 promoter. However, the EDN1 promoter contacts multiple sites extending up to 500 kb in both the 3’ and 5’ directions, which overlap with rs9349379’s contact region. The overlap region appears to contain a super-enhancer.98

Functional Role of Epigenetic Mechanisms in Hypertension

Compared to the numerous reports correlating changes in epigenetic mediators with BP or hypertension, clear evidence of epigenetic changes’ functional role in the regulation of BP or the development of hypertension remains scarce. Some of this evidence is summarized in Table 2 and discussed below.

Table 2.

Evidence of Epigenetic Mediators’ Functional Role in Hypertension.

Target Species/model Approach Effect on BP Reference
Cg08035323 methylation site Humans Bidirectional Mendelian randomization Influencing BP 53
DNA de novo (de)methylation in the kidney SS Rats Intra-renal injection of GapmeRs targeting Dnmt3a and Tet3 Attenuating salt-induced hypertension 62
DNA methylation CBS+/− mice 5-aza-2'-deoxycytidine treatment Attenuating hypertension 99
DNA methylation Rats, long-term intermittent hypoxia Decitabine treatment Attenuating hypertension 100
Histone acetylation* SHR Valproic acid treatment Attenuating hypertension 102
Histone acetylation* DJ-1/park7−/− mice Valproic acid treatment Attenuating increase in systolic BP 107
Histone methylation Mice LSD1 heterozygous knockout Increasing BP 108
lncRNA Rffl-lnc1* Rats Gene targeting and knockin A 19bp indel increases BP 110
lncRNA AK098656* Rats Transgenic overexpression Resulting in hypertension 111
*

Whether epigenetic changes are involved remains to be tested.

BP, blood pressure; SS, Dahl salt-sensitive rats; Dnmt, DNA methyltransferase; Tet, ten-eleven translocase; CBS, Cystathionine-β-synthase; SHR, spontaneously hypertensive rats; LSD1, lysine-specific demethylase-1.

V.1. DNA methylation

In Richard et al.’s study, Mendelian randomization analysis suggested the presence of causal regulatory relations among select methylation sites, BP, and gene expression.53 Methylation at cg08035323 (TAF1B-YWHAQ) appeared to influence BP, while BP influenced methylation at cg00533891 (ZMIZ1), cg00574958 (CPT1A), and cg02711608 (SLC1A5). In addition, the expression of TSPAN2 was a putative mediator of the association between methylation at cg23999170 and BP.

The renal outer medulla consists primarily of non-dividing cells that have differentiated terminally. Changes in DNA methylation in the renal outer medulla of rat models occurred after only a few days on a high-salt diet20, 62 and likely were the consequence of de novo DNA methylation or de-methylation. Liu et al. targeted Dnmt3a and Tet3 by administering anti-Dnmt3a and anti-Tet3 GapmeR’s directly into the renal interstitium in SS rats. The treatment attenuated salt-induced hypertension and prevented the significant differential expression of 76% of the genes that would respond to the high-salt diet otherwise. The study was one of the first to demonstrate a functional role of de novo DNA methylation or de-methylation in the development of diseases primarily involving non-dividing cells.62

Cystathionine-β-synthase heterozygous knockout mice exhibit hyperhomocysteinemia, high BP, global hypermethylation, and up-regulation of Dnmt1 and Dnmt3a, and treatment with 5-aza-2’-deoxycytidine normalizes BP in these mice.99 Sprague-Dawley rats exposed to long-term (30-day) intermittent hypoxia develop hypertension, higher DNA methylation of genes encoding anti-oxidant enzymes, and other abnormalities. These abnormalities do not normalize after recovery, but they can be prevented or normalized by treatment with decitabine either during intermittent hypoxia or the recovery period.100

Treating mice with a compound that attenuates aging-related increases in BP enhances DNA demethylase activity, decreases methylation of Klotho gene in the kidneys, and abolishes aging-associated downregulation of secreted Klotho levels in both kidneys and serum.101

V.2. Histone modification

Treatment with HDAC inhibitor valproic acid attenuates hypertension in SHR102 and a rat model of DOCA-salt hypertension.103 HDAC inhibitors trichostatin A, butyric acid, and MS-275 decrease eNOS protein levels in endothelial cells.104 However, it is unclear whether changes in histone acetylation mediate these effects. HDAC can influence eNOS activity and mineralocorticoid receptors by changing the acetylation of eNOS and mineralocorticoid receptor proteins.103,105 However, in several non-endothelial cell types, treatment with trichostatin A and sodium butyrate induces eNOS expression, which is accompanied by increased acetylation of histone H3 associated with the eNOS 5’-flanking region. Combined treatment with trichostatin A and 5-aza-2’-deoxycytidine synergistically induces eNOS expression in non-endothelial cells.106

DJ-1/park7(−/−) mice exhibit lower eNOS expression, which is associated with increased HDAC1 recruitment and decreased H3 acetylation at the eNOS promoter, as well as higher systolic BP, and treatment with HDAC inhibitor valproic acid attenuated the increase of systolic BP in these mice.107 Mice with heterozygous knockout of lysine-specific demethylase-1 (LSD1, Kdm1a), which induces demethylation of H3K4 or H3K9, have higher BP than do wild-type mice when they are fed a 4% NaCl diet, but not when fed a 0.08% NaCl diet.108 African-Americans and Hispanics, but not Caucasians, who carry the minor allele of one or two LSD1 SNPs, display greater changes in systolic BP in response to changes from a low to liberal salt diet.109

V.3. lncRNA

Several lncRNAs have been reported to influence BP. Targeted disruption of the rat Rffl-lnc1 locus, which is homologous to a human QT-interval locus, causes aberrant, short QT-intervals and elevated BP, while a CRISPR/Cas9 based targeted knock-in rescues the aberrant QT-interval and BP phenotypes in the strain that has a deletion polymorphism.110 Transgenic overexpression of lncRNA AK098656 in rats results in spontaneous development of hypertension.111 However, it is unclear whether these lncRNAs influence BP by regulating stable changes in DNA function.

Key Questions, Concepts, and Technologies to Advance the Field

While increasing evidence supports epigenetic mediators’ role in hypertension, how great the role is remains to be assessed and likely will vary between patients and models. Transgenerational epigenetic inheritance and its role in hypertension remain fascinating, but largely untested, possibilities. In conditions in which fetal programming and early life experiences affect adult hypertension, the specific epigenetic mechanisms involved and the way they contribute to hypertension need to be elucidated further. A large majority of the BP-associated SNPs identified and replicated by GWAS are non-coding SNPs, nearly all of which overlap with potential enhancers, or are in linkage disequilibrium with SNPs that overlap them.112 It is important to understand whether and the way in which epigenetic mechanisms might mediate these SNPs’ effect on gene expression and BP.

Changing the activity of one DNMT or TET or the level of just one histone modification potentially can change many genes’ expression. However, if a patient’s development of hypertension involves a large number of molecular changes, it will be necessary to identify and understand all of those molecular changes to understand truly the way hypertension develops in that patient and the best way to treat it. The tree-like paradigm depicted in Figure 3 can be used as a framework to understand the way numerous molecular changes converge to form regulatory networks that underlie a complex trait or disease.16, 113, 114 An increasing number of investigators are working in the emerging discipline of systems molecular medicine (Figure 4) with the goal to construct the tree of regulatory mechanisms underlying complex physiological traits and diseases.115, 116

Figure 3. A tree-like paradigm for understanding the mechanisms underlying complex traits and diseases.

Figure 3.

The large number of molecular changes, including epigenetic changes, involved in a complex trait, such as BP, are like leaves on a tree that will merge gradually into small and then large branches. Cellular signaling and metabolic pathways are the small branches, and physiological pathways are the large branches. In the case of BP, the pathways ultimately will influence cardiac output or peripheral vascular resistance, the final determinants of BP. The arrows in the graph illustrate the concept of convergence that is fundamental to this tree-like paradigm. Divergence, cross-talks and feedback loops are not depicted, but could contribute importantly to the regulatory network.

Figure 4. The emerging discipline of systems molecular medicine.

Figure 4.

To construct the regulatory network underlying a complex physiological trait or disease, comprehensive molecular profiles, including epigenetic profiles, with sufficient spatial and temporal resolution need to be obtained from patients and clinically relevant models. The molecular compendia will have to be analyzed with a systems view, with respect to both the relations among molecules and in the context of organ systems function. Informative perturbations will need to be applied, followed by reiterations of molecular and systems analyses, to reveal and validate the underlying regulatory network. The discipline of systems molecular medicine is emerging from the intersection of several concepts and disciplines including systems biology, physiology, genomics, translational research, and precision medicine.

In applying the concept of systems molecular medicine to epigenetic research on hypertension, it would be highly valuable to obtain epigenetic profiles from human tissues that are physiologically important in BP regulation and hypertension.117, 118, 119 The epigenetic and expression profiles available from studies such as the Encyclopedia of DNA Elements (ENCODE) Project and the Genotype-Tissue Expression (GTEx) Project are helpful, but the anatomical resolution of the tissues used and their relevance to BP regulation are limited. Data on genome-wide epigenetic profiles in tissues, such as resistance arterioles and nephron segments, from healthy humans and patients in various stages of hypertension development would be a powerful resource to advance epigenetic research on hypertension.112, 120, 121

Using oligonucleotides or conditional knockout to target specific DNA or histone modifying enzymes in a specific tissue or at a specific stage of disease development, coupled with genome-wide analysis of epigenetic mediators, will continue to be a powerful approach to study epigenetic mediators’ functional role in vivo.62 Development or further improvement of experimental techniques to induce methylation and de-methylation at specific genomic loci, alter specific histone modifications, and form or break specific chromatin loops, would be of great value in the study of specific epigenetic features’ functional role. Further, these techniques would have to be adapted for use in whole organisms to study traits such as BP.122, 123

Acknowledgement

The author thanks Ms. Kristin Nord for her editorial assistance.

Sources of funding

The U.S. National Institutes of Health (HL121233, HL125409, GM066730), American Heart Association (15SFRN23910002), and the Advancing a Healthier Wisconsin Endowment supported the work in the author’s laboratory related to the subject of this article.

Footnotes

Disclosures

None.

References

  • 1.Bird A. Perceptions of epigenetics. Nature. 2007; 24;447(7143): 396–8. [DOI] [PubMed] [Google Scholar]
  • 2.Berger SL, Kouzarides T, Shiekhattar R, Shilatifard A. An operational definition of epigenetics. Genes Dev. 2009; 23(7): 781–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ledford H. Language: Disputed definitions. Nature. 2008; 455(7216): 1023–8. [DOI] [PubMed] [Google Scholar]
  • 4.Smith ZD, Meissner A. DNA methylation: Roles in mammalian development. Nat Rev Genet. 2013; 14(3): 204–20. [DOI] [PubMed] [Google Scholar]
  • 5.Reinberg D, Vales LD. Chromatin domains rich in inheritance. Science. 2018; 361(6397): 33–34. [DOI] [PubMed] [Google Scholar]
  • 6.Zandi-Nejad K, Luyckx VA, Brenner BM. Adult hypertension and kidney disease: The role of fetal programming. Hypertension. 2006; 47(3): 502–8. [DOI] [PubMed] [Google Scholar]
  • 7.McGowan PO, Matthews SG. Prenatal Stress, Glucocorticoids, and Developmental Programming of the Stress Response. Endocrinology. 2018; 159(1): 69–82. [DOI] [PubMed] [Google Scholar]
  • 8.Bommarito PA, Martin E, Fry RC. Effects of prenatal exposure to endocrine disruptors and toxic metals on the fetal epigenome. Epigenomics. 2017; 9(3): 333–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Heard E, Martienssen RA. Transgenerational epigenetic inheritance: Myths and mechanisms. Cell. 2014; 157(1): 95–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science. 2001; 293(5532): 1089–93. [DOI] [PubMed] [Google Scholar]
  • 11.Guibert S, Forné T, Weber M. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res. 2012; 22(4): 633–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hackett JA, Sengupta R, Zylicz JJ, Murakami K, Lee C, Down TA, Surani MA. Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine. Science. 2013; 339(6118): 448–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Smith ZD, Chan MM, Mikkelsen TS, Gu H, Gnirke A, Regev A, Meissner A. A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature. 2012; 484(7394): 339–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Carone BR, Fauquier L, Habib N, Shea JM, Hart CE, Li R, Bock C, Li C, Gu H, Zamore PD, Meissner A, Weng Z, Hofmann HA, Friedman N, Rando OJ. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell. 2010; 143(7): 1084–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rassoulzadegan M, Grandjean V, Gounon P, Vincent S, Gillot I, and Cuzin F. RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse. Nature 2006; 441: 469–474. [DOI] [PubMed] [Google Scholar]
  • 16.Liang M, Cowley AW Jr, Mattson DL, Kotchen TA, Liu Y. Epigenomics of hypertension. Semin Nephrol. 2013; 33(4): 392–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bird A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002; 16(1): 6–21. [DOI] [PubMed] [Google Scholar]
  • 18.Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev. 2011; 25(10): 1010–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wagner JR, Busche S, Ge B, Kwan T, Pastinen T, Blanchette M. The relationship between DNA methylation, genetic and expression inter-individual variation in untransformed human fibroblasts. Genome Biol. 2014; 15(2): R37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Liu Y, Liu P, Yang C, Cowley AW Jr., Liang M. Base-resolution maps of 5-methylcytosine and 5-hydroxymethylcytosine in Dahl S Rats: Effect of salt and genomic sequence. Hypertension 2014. April; 63(4): 827–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bahar Halpern K, Vana T, Walker MD. Paradoxical role of DNA methylation in activation of FoxA2 gene expression during endoderm development. J Biol Chem. 2014; 289(34): 23882–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Schultz MD, He Y, Whitaker JW, Hariharan M, Mukamel EA, Leung D, Rajagopal N, Nery JR, Urich MA, Chen H, Lin S, Lin Y, Jung I, Schmitt AD, Selvaraj S, Ren B, Sejnowski TJ, Wang W, Ecker JR. Human body epigenome maps reveal noncanonical DNA methylation variation. Nature. 2015; 523(7559): 212–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lyko F. The DNA methyltransferase family: A versatile toolkit for epigenetic regulation. Nat Rev Genet. 2018; 19(2): 81–92. [DOI] [PubMed] [Google Scholar]
  • 24.Rasmussen KD, Helin K. Role of TET enzymes in DNA methylation, development, and cancer. Genes Dev. 2016; 30(7): 733–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011; 21(3): 381–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lawrence M, Daujat S, Schneider R. Lateral Thinking: How histone modifications regulate gene expression. Trends Genet. 2016; 32(1): 42–56. [DOI] [PubMed] [Google Scholar]
  • 27.Falkenberg KJ, Johnstone RW. Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov. 2014; 13(9): 673–91. [DOI] [PubMed] [Google Scholar]
  • 28.West AC, Johnstone RW. New and emerging HDAC inhibitors for cancer treatment. J Clin Invest. 2014; 124(1): 30–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: Insights into functions. Nat Rev Genet. 2009; 10(3): 155–9. [DOI] [PubMed] [Google Scholar]
  • 30.Zaratiegui M, Martienssen RA. SnapShot: Small RNA-mediated epigenetic modifications. Cell. 2012; 151(2): 456–456.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Marques FZ, Charchar FJ. microRNAs in Essential Hypertension and Blood Pressure Regulation. Adv Exp Med Biol. 2015; 888: 215–35. [DOI] [PubMed] [Google Scholar]
  • 32.Wise IA, Charchar FJ. Epigenetic Modifications in Essential Hypertension. Int J Mol Sci. 2016; 17(4): 451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dekker J, Marti-Renom MA, Mirny LA. Exploring the three-dimensional organization of genomes: interpreting chromatin interaction data. Nat Rev Genet. 2013; 14(6): 390–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bonev B, Cavalli G. Organization and function of the 3D genome. Nat Rev Genet. 2016; 17(12): 772. [DOI] [PubMed] [Google Scholar]
  • 35.Bock C, Tomazou EM, Brinkman AB, Müller F, Simmer F, Gu H, Jäger N, Gnirke A, Stunnenberg HG, Meissner A. Quantitative comparison of genome-wide DNA methylation mapping technologies. Nat Biotechnol. 2010; 28(10): 1106–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Laird PW. Principles and challenges of genome-wide DNA methylation analysis. Nat Rev Genet. 2010; 11(3): 191–203. [DOI] [PubMed] [Google Scholar]
  • 37.Ozsolak F, Milos PM. RNA sequencing: Advances, challenges and opportunities. Nat Rev Genet. 2011; 12(2): 87–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Park PJ. ChIP-seq: Advantages and challenges of a maturing technology. Nat Rev Genet. 2009; 10(10): 669–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Gu H, Smith ZD, Bock C, Boyle P, Gnirke A, Meissner A. Preparation of reduced representation bisulfite sequencing libraries for genome-scale DNA methylation profiling. Nat Protoc. 2011; 6(4): 468–81. [DOI] [PubMed] [Google Scholar]
  • 40.Branco MR, Ficz G, Reik W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat Rev Genet. 2011; 13(1): 7–13. [DOI] [PubMed] [Google Scholar]
  • 41.Song CX, Yi C, He C. Mapping recently identified nucleotide variants in the genome and transcriptome. Nat Biotechnol. 2012; 30(11): 1107–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Booth MJ et al. Quantitative sequencing of 5-methylcytosine and 5-hydroxymethylcytosine at single-base resolution. Science 2012; 336: 934–937. [DOI] [PubMed] [Google Scholar]
  • 43.Yu M. et al. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome. Cell 2012; 149: 1368–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Zentner GE, Henikoff S. Surveying the epigenomic landscape, one base at a time. Genome Biol. 2012; 13(10): 250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, Huarte M, Zuk O, Carey BW, Cassady JP, Cabili MN, Jaenisch R, Mikkelsen TS, Jacks T, Hacohen N, Bernstein BE, Kellis M, Regev A, Rinn JL, Lander ES. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009; 458(7235): 223–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO, Landthaler M, Lin C, Socci ND, Hermida L, Fulci V, Chiaretti S, Foà R, Schliwka J, Fuchs U, Novosel A, Müller RU, Schermer B, Bissels U, Inman J, Phan Q, Chien M, Weir DB, Choksi R, De Vita G, Frezzetti D, Trompeter HI, Hornung V, Teng G, Hartmann G, Palkovits M, Di Lauro R, Wernet P, Macino G, Rogler CE, Nagle JW, Ju J, Papavasiliou FN, Benzing T, Lichter P, Tam W, Brownstein MJ, Bosio A, Borkhardt A, Russo JJ, Sander C, Zavolan M, Tuschl T. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 2007; 129(7): 1401–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pauli A, Valen E, Lin MF, Garber M, Vastenhouw NL, Levin JZ, Fan L, Sandelin A, Rinn JL, Regev A, Schier AF. Systematic identification of long noncoding RNAs expressed during zebrafish embryogenesis. Genome Res. 2012; 22(3): 577–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Li F, Papworth M, Minczuk M, Rohde C, Zhang Y, Ragozin S, Jeltsch A. Chimeric DNA methyltransferases target DNA methylation to specific DNA sequences and repress expression of target genes. Nucleic Acids Res. 2007; 35(1): 100–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nomura W, Barbas CF 3rd. In vivo site-specific DNA methylation with a designed sequence-enabled DNA methylase. J Am Chem Soc. 2007; 129(28): 8676–7. [DOI] [PubMed] [Google Scholar]
  • 50.Xu GL, Bestor TH. Cytosine methylation targeted to pre-determined sequences. Nat Genet. 1997; 17(4): 376–8. [DOI] [PubMed] [Google Scholar]
  • 51.Rakyan VK, Down TA, Balding DJ, Beck S. Epigenome-wide association studies for common human diseases. Nat Rev Genet. 2011; 12(8): 529–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Stresemann C, Lyko F. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer. 2008; 123(1): 8–13. [DOI] [PubMed] [Google Scholar]
  • 53.Richard MA, Huan T, Ligthart S, Gondalia R, Jhun MA, Brody JA, Irvin MR, Marioni R, Shen J, Tsai PC, Montasser ME, Jia Y, Syme C, Salfati EL, Boerwinkle E, Guan W, Mosley TH jr, Bressler J, Morrison AC, Liu C, Mendelson MM, Uitterlinden AG, van Meurs JB, BIOS Consortium, Franco OH, Zhang G, Li Y, Stesart JD, Bis JC, Psaty BM, Chen YI, Kardia SLR, Zhao W, Turner ST, Absher D, Aslibekyan S, Starr JM, McRae AF, Hou L, Just AC, Schwartz JD, Vokonas PS, Menni C, Spector TD, Shuldiner A, Damcott CM, Totter JI, Palmas W, Liu Y, Paus T, Horvath S, O’Connell JR, Guo X, Pausova Z, Assimes TL, Sotodehnia N, Smith JA, Arnett DK, Deary IJ, Baccarelli AA, Bell JT, Whitsel E, Dehghan A, Levy D, Fornage M. DNA Methylation Analysis Identifies Loci for Blood Pressure Regulation. Am J Hum Genet. 2017; 101(6): 888–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kato N, Loh M, Takeuchi F, Verweij N, Wang X, Zhang W, Kelly TN, Saleheen D, Lehne B, Leach IM, Drong AW, Abbott J, Wahl S, Tan ST, Scott WR, Campanella G, Chadeau-Hyam M, Afzal U, Ahluwalia TS, Bonder MJ, Chen P, Dehghan A, Edwards TL, Esko T, Go MJ, Harris SE, Hartiala J, Kasela S, Kasturiratne A, Khor CC, Kleber ME, Li H, Yu Mok Z, Nakatochi M, Sapari NS, Saxena R, Stewart AFR, Stolk L, Tabara Y, Teh AL, Wu Y, Wu JY, Zhang Y, Aits I, Da Silva Couto Alves A, Das S, Dorajoo R, Hopewell JC, Kim YK, Koivula RW, Luan J, Lyytikäinen LP, Nguyen QN, Pereira MA, Postmus I, Raitakari OT, Bryan MS, Scott RA, Sorice R, Tragante V, Traglia M, White J, Yamamoto K, Zhang Y, Adair LS, Ahmed A, Akiyama K, Asif R, Aung T, Barroso I, Bjonnes A, Braun TR, Cai H, Chang LC, Chen CH, Cheng CY, Chong YS, Collins R, Courtney R, Davies G, Delgado G, Do LD, Doevendans PA, Gansevoort RT, Gao YT, Grammer TB, Grarup N, Grewal J, Gu D, Wander GS, Hartikainen AL, Hazen SL, He J, Heng CK, Hixson JE, Hofman A, Hsu C, Huang W, Husemoen LLN, Hwang JY, Ichihara S, Igase M, Isono M, Justesen JM, Katsuya T, Kibriya MG, Kim YJ, Kishimoto M, Koh WP, Kohara K, Kumari M, Kwek K, Lee NR, Lee J, Liao J, Lieb W, Liewald DCM, Matsubara T, Matsushita Y, Meitinger T, Mihailov E, Milani L, Mills R, Mononen N, Müller-Nurasyid M, Nabika T, Nakashima E, Ng HK, Nikus K, Nutile T, Ohkubo T, Ohnaka K, Parish S, Paternoster L, Peng H, Peters A, Pham ST, Pinidiyapathirage MJ, Rahman M, Rakugi H, Rolandsson O, Ann Rozario M, Ruggiero D, Sala CF, Sarju R, Shimokawa K, Snieder H, Sparsø T, Spiering W, Starr JM, Stott DJ, Stram DO, Sugiyama T, Szymczak S, Tang WHW, Tong L, Trompet S, Turjanmaa V, Ueshima H, Uitterlinden AG, Umemura S, Vaarasmaki M, van Dam RM, van Gilst WH, van Veldhuisen DJ, Viikari JS, Waldenberger M, Wang Y, Wang A, Wilson R, Wong TY, Xiang YB, Yamaguchi S, Ye X, Young RD, Young TL, Yuan JM, Zhou X, Asselbergs FW, Ciullo M, Clarke R, Deloukas P, Franke A, Franks PW, Franks S, Friedlander Y, Gross MD, Guo Z, Hansen T, Jarvelin MR, Jørgensen T, Jukema JW, Kähönen M, Kajio H, Kivimaki M, Lee JY, Lehtimäki T, Linneberg A, Miki T, Pedersen O, Samani NJ, Sørensen TIA, Takayanagi R, Toniolo D; BIOS-consortium; CARDIo GRAMplusCD; LifeLines Cohort Study; InterAct Consortium, Ahsan H, Allayee H, Chen YT, Danesh J, Deary IJ, Franco OH, Franke L, Heijman BT, Holbrook JD, Isaacs A, Kim BJ, Lin X, Liu J, März W, Metspalu A, Mohlke KL, Sanghera DK, Shu XO, van Meurs JBJ, Vithana E, Wickremasinghe AR, Wijmenga C, Wolffenbuttel BHW, Yokota M, Zheng W, Zhu D, Vineis P, Kyrtopoulos SA, Kleinjans JCS, McCarthy MI, Soong R, Gieger C, Scott J, Teo YY, He J, Elliott P, Tai ES, van der Harst P, Kooner JS, Chambers JC. Trans-ancestry genome-wide association study identifies 12 genetic loci influencing blood pressure and implicates a role for DNA methylation. Nat Genet. 2015; 47(11): 1282–1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang X, Falkner B, Zhu H, Shi H, Su S, Xu X, Sharma AK, Dong Y, Treiber F, Gutin B, Harshfield G, Snieder H. A genome-wide methylation study on essential hypertension in young African American males. PLoS One. 2013; 8(1): e53938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Breton CV, Yao J, Millstein J, Gao L, Siegmund KD, Mack W, Whitfield-Maxwell L, Lurmann F, Hodis H, Avol E, Gililand FD. Prenatal Air Pollution Exposures, DNA Methyl Transferase Genotypes, and Associations with Newborn LINE1 and Alu Methylation and Childhood Blood Pressure and Carotid Intima-Media Thickness in the Children’s Health Study. Environ Health Perspect. 2016; 124(12): 1905–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bellavia A, Urch B, Speck M, Brook RD, Scott JA, Albetti B, Behbod B, North M, Valeri L, Bertazzi PA, Silverman F, Gold D, Baccarelli AA. DNA hypomethylationm anbient particulate matter, and increased blood pressure: findings from controlled human exposure experiments. J Am Heart Assoc. 2013; 2(3): e000212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Petry CJ, Sanz Marcos N, Pimentel G, Hayes MG, Nodzenski M, Scholtens DM, Hughes IA, Acerini CL, Ong KK, Lowe WL Jr, Dunger DB. Associations Between Fetal Imprinted Genes and Maternal Blood Pressure in Pregnancy. Hypertension. 2016; 68(6): 1459–1466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sinclair KD, Allegrucci C, Singh R, Gardner DS, Sebastian S, Bispham J, Thurston A, Huntley JF, Rees WD. Maloney CA, Lea RG, Craigon J, McEvoy TG, Young LE. DNA methylation, insulin resistance, and blood pressure in offspring determined by maternal periconceptional B vitamin and methionine status. Proc Natl Acad Sci U S A. 2007; 104(49): 19351–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Meems LM, Mahmud H, Buikema H, Tost J, Michel S, Takens J, Verkaik-Schakel RN, Vreeswijk-Baudoin I, Mateo-Leach IV, van der Harst P, Plosch T, de Boer RA. Parental vitamin D deficiency during pregnancy is associated with increased blood pressure in offspring via Panx1 hypermethylation. Am J Physiol Heart Circ Physiol. 2016; 311(6): H1459–H1469. [DOI] [PubMed] [Google Scholar]
  • 61.Geurts AM, Mattson DL, Liu P, Cabacungan E, Skelton MM, Kurth TM, Yang C, Endres BT, Klotz J, Liang M, Cowley AW Jr. Maternal diet during gestation and lactation modifies the severity of salt-induced hypertension and renal injury in dahl salt-sensitive rats. Hypertension. 2015; 65(2): 447–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Liu P, Liu Y, Liu H, Pan X, Li Y, Usa K, Mishra MK, Nie J, Liang M. Role of DNA de novo (de)Methylation in the Kidney in Salt-Induced Hypertension. Hypertension (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Alikhani-Koopaei R, Fouladkou F, Frey FJ, Frey BM. Epigenetic regulation of 11 beta-hydroxysteroid dehydrogenase type 2 expression. J Clin Invest. 2004; 114(8): 1146–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wang F, Demura M, Cheng Y, Zhu A, Karashima S, Yoneda T, Demura Y, Maeda Y, Namiki M, Ono K, Nakamura Y, Sasano H, Akagi T, Yamagishi M, Saijoh K, Takeda Y. Dynamic CCAAT/enhancer binding protein-associated changes of DNA methylation in the angiotensinogen gene. Hypertension. 2014; 63(2): 281–8. [DOI] [PubMed] [Google Scholar]
  • 65.Yu Z, Kong Q, Kone BC. Aldosterone reprograms promoter methylation to regulate αENAC transcription in the collecting duct. Am J Physiol Renal Physiol. 2013; 305(7): F1006–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wyrwoll CS, Mark PJ, Waddell BJ. Developmental programming of renal glucocorticoid sensitivity and the renin-angiotensin system. Hypertension. 2007; 50(3): 579–84. [DOI] [PubMed] [Google Scholar]
  • 67.Rivière G, Lienhard D, Andrieu T, Vieau D, Frey BM, Frey FJ. Epigenetic regulation of somatic angiotensin-converting enzyme by DNA methylation and histone acetylation. Epigenetics. 2011; 6(4): 478–89. [DOI] [PubMed] [Google Scholar]
  • 68.Bogdarina I, Welham S, King PJ, Burns SP, Clark AJ. Epigenetic modification of the renin-angiotensin system in the fetal programming of hypertension. Circ Res. 2007; 100: 520–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Chelbi ST, Mondon F, Jammes H, Buffat C, Mignot TM, Tost J, Busato F, Gut I, Rebourcet R, Laissue P, Tsatsaris V, Goffinet F, Rigourd V, Carbonne B, Ferre E, Vaiman D. Expressional and epigenetic alterations of placental serine protease inhibitors: SERPINA3 is a potential marker of preeclampsia. Hypertension. 2007; 49(1): 76–83. [DOI] [PubMed] [Google Scholar]
  • 70.Mousa AA, Strauss JF 3rd, Walsh SW. Reduced methylation of the thromboxane synthase gene is correlated with its increased vascular expression in preeclampsia. Hypertension. 2012; 59(6): 1249–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.He J, Zhang A, Fang M, Fang R, Ge J, Jiang Y, Zhang H, Han C, Ye X, Yu D, Huang H, Liu Y, Dong M. Methylation levels at IGF2 and GNAS DMRs in infants born to preeclamptic pregnancies. BMC Genomics. 2013; 14:472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jia Y, Li T, Huang X, Xu X, Zhou X, Jia L, Zhu J, Xie D, Wang K, Zhou Q, Jin L, Zhang J, Duan T. Dysregulated DNA Methyltransferase 3A Upregulates IGFBP5 to Suppress Trophoblast Cell Migration and Invasion in Preeclampsia. Hypertension. 2017; 69(2): 356–366. [DOI] [PubMed] [Google Scholar]
  • 73.Rexhaj E, Paoloni-Giacobino A, Rimoldi SF, Ruster DG, Anderegg M, Somm E, Bouillet E, Allemann Y, Sartori C, Scherrer U. Mice generated by in vitro fertilization exhibit vascular dysfunction and shortened life span. J Clin Invest. 2013; 123(12): 5052–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Dasgupta C, Chen M, Zhang H, Yang S, Zhang L. Chronic hypoxia during gestation causes epigenetic repression of the estrogen receptor –α gene in ovine uterine arteries via heightened promoter methylation. Hypertension. 2012; 60(3): 697–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chen M, Dasgupta C, Xiong F, Zhang L. Epigenetic upregulation of large-conductance Ca2+-activated K+ channel expression in uterine vascular adaption to pregnancy. Hypertension. 2014; 64(3): 610–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chen M, Xiao D, Hu XQ, Dasgupta C, Yang S, Zhang L. Hypoxia Represses ER-α Expression and Inhibits Estrogen-Induced Regulation of Ca2+-Activated K+ Channel Activity and Myogenic Tone in Ovine Uterine Arteries: Causal Role of DNA Methylation. Hypertension. 2015; 66(1):44–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hu XQ, Dasgupta C, Chen M, Xiao D, Huang X, Han L, Yang S, Xu Z, Zhang L. Pregnancy Reprograms Large-Conductance Ca2+ -Activated K+ Channel in Uterine Arteries: Roles of Ten-Eleven Translocation Methylcytosine Dioxygenase 1-Medicated Active Demethylation. Hypertension. 2017; 69(6): 1181–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Chen L, Wu H, Pochynyuk OM, Reisenauer MR, Zhang z, Huang L, Zaika OL, Mamenko M, Zhang W, Zhou Q, Liu M, Xia Y, Zhang W. Af17 deficiency increases sodium excretion and decreases blood pressure. J Am Soc Nephrol. 2011; 22(6): 1076–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Mu S, Shimosawa T, Ogura S, Wang H, Uetake Y, Kawakami-Mori F, Marumo T, Yatomi Y, Geller DS, Tanaka H, Fujita T. Epigenetic modulation of the renal β-adrenergic-WNK4 pathway in salt-sensitive hypertension. Nat Med. 2011; 17(5): 573–80. [DOI] [PubMed] [Google Scholar]
  • 80.Wu Z, Siuda D, Xia N, Reifenberg G, Daiber A, Munzel T, Forstermann U, Li H. Maternal treatment of spontaneously hypertensive rats with pentaerythritol tetranitrate reduces blood pressure in female offspring. Hypertension. 2015; 65(1): 232–7. [DOI] [PubMed] [Google Scholar]
  • 81.Wang Z, Zeng C, Villar VA, Chen SY, Konkalmatt P, Wang X, Asico LD, Jones JE, Yang Y, Sanada H, Felder RA, Eisner GM, Weir MR, Armando I, Jose PA. Human GRK4γ142V Variant Promotes Angiotensin II Type I Receptor-Mediated Hypertension via Renal Histone Deacetylase Type 1 Inhibition. Hypertension. 2016; 67(2): 325–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Reddy MA, Sumanth P, Lanting L, Yuan H, Wang M, Mar D, Alpers CE, Bomsztyk K, Natarajan R. Losartan reverses permissive epigenetic changes in renal glomeruli of diabetic db/db mice. Kidney Int. 2014; 85(2): 362–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Pentz ES, Lopez ML, Cordaillat M, Gomez RA. Identity of the renin cell is mediated by cAMP and chromatin remodeling: an in vitro model for studying cell recruitment and plasticity. Am J Physiol Heart Circ Physiol. 2008; 294(2): H699–707. [DOI] [PubMed] [Google Scholar]
  • 84.Pentz ES, Cordaillat M, Carretero OA, Tucker AE, Sequeira Lopez ML, Gomez RA. Histone acetyl transferases CBP and p300 are necessary for maintenance of renin cell identity and transformation of smooth muscle cells to the renin phenotype. Am J Physiol Heart Circ Physiol. 2012; 302(12): H2545–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kumar P, Pandey KN. Cooperative activation of Npr1 gene transcription and expression by interaction of Ets-1 and p300. Hypertension. 2009; 54(1): 172–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kumar P, Garg R, Bolden G, Pandey KN. Interactive roles of Ets-1, Sp1, and acetylated histones in the retinoic acid-dependent activation of guanylyl cyclase/atrial natriuretic peptide receptor-A gene transcription. J Biol Chem. 2010; 285(48): 37521–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Glenn DJ, Wang F, Chen S, Nishimoto M, Gardner DG. Endothelin-stimulated human B-type natriuretic peptide gene expression is mediated by Yin Yang 1 in association with histone deacetylase 2. Hypertension. 2009; 53(3): 549–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang D, Yu ZY, Cruz P, Kong Q, Li S, Kone BC. Epigenetics and the control of epithelial sodium channel expression in collecting duct. Kidney Int. 2009; 75(3): 260–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zhang W, Xia X, Reisenauer MR, Rieg T, Lang F, Kuhl D, Vallon V, Kone BC. Aldosterone-induced Sgk1 relieves Dot1a-Af9-mediated transcriptional repression of epithelial Na+ channel alpha. J Clin Invest. 2007; 117(3): 773–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lee HA, Cho HM, Lee DY, Kim KC, Han HS, Kim IK. Tissue-specific upregulation of angiotensin-converting enzyme 1 in spontaneously hypertensive rats through histone code modifications. Hypertension. 2012; 59(3): 621–6. [DOI] [PubMed] [Google Scholar]
  • 91.Wang F, Li L, Xu H, Liu Y, Yang C, Cowley AW Jr, Wang N, Liu P, Liang M. Characteristics of Long Non-coding RNAs in the Brown Norway Rat and Alterations in the Dahl Salt-Sensitive Rat. Sci Rep. 2014; 4: 7146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gopalakrishnan K, Kumarasamy S, Mell B, Joe B. Genome-wide identification of long noncoding RNAs in rat models of cardiovascular and renal disease. Hypertension. 2015; 65(1): 200–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yao QP, Xie ZW, Wang KX, Zhang P, Han Y, Qi YX, Jiang ZL. Profiles of long noncoding RNAs in hypertensive rats: long noncoding RNA XR007793 regulates cyclic strain-induces proliferation and migration of vascular smooth muscle cells. J Hypertens. 2017; 35(6): 1195–1203. [DOI] [PubMed] [Google Scholar]
  • 94.Yu B, Wang S. Angio-LncRs: LncRNAs that regulate angiogenesis and vascular disease. Theranostics. 2018; 8(13): 3654–3675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Leung A, trac C, Jin W, Lanting L, Akbany A, Saetrom P, Schones DE, Natarajan R. Novel long noncoding RNAs are regulated by angiotensin II in vascular smooth muscle cells. Circ Res. 2013; 113(3): 266–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lv H, Tong J, Yang J, Lv S, Li WP, Zhang C, Chen ZJ. Dysregulated Pseudogene HK2P1 May Contribute to Preeclampsia as a Competing Endogenous RNA for Hexokinase 2 by Impairing Decidualization. Hypertension. 2018; 71(4): 648–658. [DOI] [PubMed] [Google Scholar]
  • 97.Stodola TJ, Liu P, Liu Y, Vallejos AK, Geurts AM, Greene AS, Liang M. Genome-wide map of proximity linkage to renin proximal promoter in rat. Physiol Genomics. 2018; 50(5): 323–331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gupta RM, Hadaya J, Trehan A, Zekavat SM, Roselli C, Klarin D, Emdin CA, Hilvering CRE, Bianchi V, Mueller C, Khera AV, Ryan RJH, Engreitz JM, Issner R, Shoresh N, Epstein CB, de Laat W, Brown JD, Schnabel RB, Bernstein BE, Kathiresan S. A Genetic Variant Associated with Five Vascular Diseases Is a Distal Regulator of Endothelin-1 Gene Expression. Cell. 2017; 170(3): 522–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Pushpakumar S, Kundu S, Narayanan N, Sen U. DNA hypermethgylation in hyperhomocysteinemia contributes to abnormal extracellular matrix metabolism in the kidney. FASEB J. 2015; 29(11): 4713–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nanduri J, Peng YJ, Wang N, Khan SA, Semenza GL, Kumar GK, Prabhakar NR. Epigenetic regulation of redox state mediates persistent cardiorespiratory abnormalities after long-term intermittent hypoxia. J Physiol. 2017; 595(1): 63–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Chen K, Sun Z. Activation of DNA demethylases attenuates aging-associated arterial stiffening and hypertension. Aging Cell. 2018; e12762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Cardinale JP, Sriramula S, Pariaut R, Guggilam A, Mariappan N, Elks CM, Francis J. HDAC inhibition attenuates inflammatory, hypertrophic and hypertensive responses in spontaneously hypertensive rats. Hypertension. 2010; 56(3): 437–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lee HA, Lee DY, Cho HM, Kim SY, Iwasaki Y, Kim IK. Histone deacetylase inhibition attenuates transcriptional activity of mineralocorticoid receptor through its acetylation and prevents development of hypertension. Circ Res. 2013; 112(7): 1004–12. [DOI] [PubMed] [Google Scholar]
  • 104.Rossig L, Li H, Fisslthaler B, Urbich C, Fleming I, Forstermann U, Zeiher AM, Dimmeler S. Inhibitors of histone deacetylation downregulate the expression of endothelial nitric oxide synthase and compromise endothelial cell function in vasorelazation and angiogenesis. Circ Res. 2002; 91(9): 837–44. [DOI] [PubMed] [Google Scholar]
  • 105.Jung SB, Kim CS, Naqvi A, Yamamori T, Mattagajasingh I, Hoffman TA, Cole MP, Kumar A, Dericco JS, Jeon BH, Irani K. Histone deacetylase 3 antagonizes aspirin-stimulated endothelial nitric oxide production by reversing aspirin-induced lysine acetylation of endothelial nitric oxide synthase. Circ Res. 2010; 107(7): 877–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Gan Y, Shen YH, Wang J, Wang X, Utama B, Wang J, Wang XL. Role of histone deacetylation in cell-specific expression of endothelial nitric-oxide synthase. J Biol Chem. 2005; 280(16): 16467–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Won KJ, Jung SH, Jung SH, Lee KP, Lee HM, Lee DY, Park ES, Kim J, Kim B. DJ-1/park7 modulates vasorelaxation and blood pressure via epigenetic modification of endothelial nitric oxide synthase. Cardiovasc Res. 2014; 101(3): 473–81. [DOI] [PubMed] [Google Scholar]
  • 108.Pojoga LH, Williams JS, Yao TM, Kimar A, Raffetto JD, do Nascimento GR, Reslan OM, Adler GK, Williams GH, Shi Y, Khalil RA. Histone demethylase LSD1 deficiency during high-salt diet is associated with enhanced vascular contraction, altered NO-cGMP relaxation pathway, and hypertension. Am J Physiol Heart Circ Physiol. 2011; 301(5): H1862–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Williams JS, Chamarthi B, Goodarzi MO, Pojoga LH, Sun B, Garza AE, Raby BA, Adler GK, Hopkins PN, Brown NJ, Jeunemaitre X, Ferri C, Fang R, Leonor T, Cui J, Guo X, Taylor KD, Ida Chen YD, Xiang A, Raffel LJ, Buchanan TA, Rotter JI, Williams GH, Shi Y. Lysine-specific demethylase 1: an epigenetic regulator of salt-sensitive hypertension. Am J Hypertens. 2012; 25(7): 812–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Cheng X, Waghulde H, Mell B, Morgan EE, Pruett-Miller SM, Joe B. Positional cloning of quantitative trait nucleotides for blood pressure and cardiac QT-interval by targeted CRISPR/Cas9 editing of a novel long non-coding RNA. PLoS Genet. 2017; 13(8): e1006961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Jin L, Lin X, Yang L, Fan X, Wang W, Li S, Li J, Liu X, Bao M, Cui X, Yang J, Cui Q, Geng B, Cai J. AK098656, a Novel Vascular Smooth Muscle Cell-Dominant Long Noncoding RNA, Promotes Hypertension. Hypertension. 2018; 71(2): 262–272. [DOI] [PubMed] [Google Scholar]
  • 112.Liu Y, Usa K, Wang F, Liu P, Geurts AM, Li J, Williams AM, Regner KR, Kong Y, Liu H, Nie J, Liang M. MicroRNA-214-3p in the Kidney Contributes to the Development of Hypertension. J Am Soc Nephrol; published ahead of print July 26, 2018, doi: 10.1681/ASN.2018020117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Williams AM, Liu Y, Regner KR, Jotterand F, Liu P, Liang M. Artificial Intelligence, Physiological Genomics, and Precision Medicine. Physiol Genomics. 2018; 50(4): 237–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Liu Y, Singh RJ, Usa K, Netzel BC, and Liang M. Renal Medullary 11β-Hydroxysteroid Dehydrogenase Type 1 in Dahl salt-sensitive hypertension. Physiol Genomics 2008; 36: 52–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Liang M. Integrative pathway knowledge bases as a tool for systems molecular medicine. Physiol Genomics. 2007; 30(3): 209–12. [DOI] [PubMed] [Google Scholar]
  • 116.Liang M. MicroRNA: a new entrance to the broad paradigm of systems molecular medicine. Physiol Genomics. 2009; 38(2): 113–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kotchen TA, Cowley AW Jr, Liang M. Ushering Hypertension into a New Era of Precision Medicine. JAMA 2016; 315(4): 343–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Touyz RM, Montezano AC, Rios F, Widlansky ME, Liang M. Redox Stress Defines the Small Artery Vasculopathy of Hypertension: How Do We Bridge the Bench-to-Bedside Gap? Circ Res. 2017; 120(11): 1721–1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Mattson DL, Liang M. Hypertension: From GWAS to functional genomics-based precision medicine. Nat Rev Nephrol. 2017; 13(4): 195–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Baker MA, Davis SJ, Liu P, Pan X, Williams AM, Iczkowski KA, Gallagher ST, Bishop K, Regner KR, Liu Y, Liang M. Tissue-Specific MicroRNA Expression Patterns in Four Types of Kidney Disease. J Am Soc Nephrol. 2017; 28(10): 2985–2992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Widlansky ME, Jensen DM, Wang J, Liu Y, Geurts AM, Kriegel AJ, Liu P, Ying R, Zhang G, Casati M, Chu C, Malik M, Branum A, Tanner MJ, Tyagi S, Usa K, Liang M. miR-29 contributes to normal endothelial function and can restore it in cardiometabolic disorders. EMBO Mol Med 2018; 10(3): e8046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Liu XS, Wu H, Ji X, Stelzer Y, Wu X, Czauderna S, Shu J, Dadon D, Young RA, Jaenisch R. Editing DNA Methylation in the Mammalian Genome. Cell. 2016; 167(1): 233–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lei Y, Zhang X, Su J, Jeong M, Gundry MC, Huang YH, Zhou Y, Li W, Goodell MA. Targeted DNA methylation in vivo using an engineered dCas9-MQ1 fusion protein. Nat Commun. 2017; 8:16026. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES