Skip to main content
Log in

Epigenetics and Hypertension

  • Published:
Current Hypertension Reports Aims and scope Submit manuscript

Abstract

Epigenetics refers to mechanisms for environment–gene interactions (mainly by methylation of DNA and modification of histones) that do not alter the underlying base sequence of the gene. This article reviews evidence for epigenetic contributions to hypertension. For example, DNA methylation at CpG islands and histone acetylation pathways are known to limit nephron development, thereby unmasking hypertension associated with exposure to a high-salt diet. Maternal water deprivation and protein deficiency are shown to increase expression of renin-angiotensin system genes in the offspring. The methylation pattern of a serine protease inhibitor gene in human placentas is shown to be a marker for preeclampsia-associated hypertension. Mental stress induces phenylethanolamine n-methyltransferase, which may act as a DNA methylase and mimic the gene-silencing effects of methyl CpG binding protein-2 on the norepinephrine transporter gene, which, in turn, may exaggerate autonomic responsiveness. A disrupter of telomeric silencing (Dot1) is known to modulate the expression of a connective-tissue growth-factor gene associated with blood vessel remodeling, which could alter vascular compliance and elastance. Dot1a also interacts with the Af9 gene to produce high sodium channel permeability and silences the hydroxysteroid dehydrogenase-11β2 gene, thereby preventing metabolism of cortisol to cortisone and overstimulating aldosterone receptors. These findings indicate targets for environment–gene interactions in various hypertensive states and in essential hypertension.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance

  1. Garcia EA, Newhouse S, Caulfield MJ, Munroe PB: Genes and hypertension. Curr Pharm Des 2003, 9:1679–1689.

    Article  CAS  PubMed  Google Scholar 

  2. Kreutz R, Hübner N, James MR, et al.: Dissection of a quantitative trait locus for genetic hypertension on rat chromosome 10. Proc Natl Acad Sci U S A 1995, 12:8778–8782.

    Article  Google Scholar 

  3. Ehret GB: Genome-wide association studies: contribution of genomics to understanding blood pressure and essential hypertension. Curr Hypertens Rep 2010, 12:17–25.

    Article  PubMed  Google Scholar 

  4. Kunes J, Zicha J: The interaction of genetic and environmental factors in the etiology of hypertension. Physiol Res 2009, 58(Suppl 2):S33–S41.

    PubMed  Google Scholar 

  5. Bodnar LM, Catov JM, Simhan HN, et al.: Maternal vitamin D deficiency increases the risk of preeclampsia. J Clin Endocrinol Metab 2007, 92:3517–3522.

    Article  CAS  PubMed  Google Scholar 

  6. Parra M, Rodrigo R, Barja P, et al.: Screening test for preeclampsia through assessment of uteroplacental blood flow and biochemical markers of oxidative stress and endothelial dysfunction. Am J Obstet Gynecol 2005, 193:1486–1491.

    Article  CAS  PubMed  Google Scholar 

  7. Harskamp RE, Zeeman GG: Preeclampsia: at risk for remote cardiovascular disease. Am J Med Sci 2007, 334:291–295.

    Article  PubMed  Google Scholar 

  8. Koleganova N, Piecha G, Ritz E: Prenatal causes of kidney disease. Blood Purif 2009, 27:48–52.

    Article  CAS  PubMed  Google Scholar 

  9. Jeske YW, So A, Kelemen L, et al.: Examination of chromosome 7p22 candidate genes RBaK, PMS2 and GNA12 in familial hyperaldosteronism type II. Clin Exp Pharmacol Physiol 2008, 35:380–385.

    Article  CAS  PubMed  Google Scholar 

  10. Washietl S, Hofacker IL, Stadler PF: Fast and reliable prediction of noncoding RNAs. Proc Natl Acad Sci U S A 2005, 102:2454–2459.

    Article  CAS  PubMed  Google Scholar 

  11. Chelbi ST, Mondon F, Jammes H, et al.: Expressional and epigenetic alterations of placental serine protease inhibitors: SERPINA3 is a potential marker of preeclampsia. Hypertension 2007, 49:76–83.

    Article  CAS  PubMed  Google Scholar 

  12. Chan GC, Fish JE, Mawji IA, et al.: Epigenetic basis for the transcriptional hyporesponsiveness of the human inducible nitric oxide synthase gene in vascular endothelial cells. J Immunol 2005, 175:3846–3861.

    CAS  PubMed  Google Scholar 

  13. Kang TJ, Yuzawa S, Suga H: Expression of histone H3 tails with combinatorial lysine modifications under the reprogrammed genetic code for the investigation on epigenetic markers. Chem Biol 2008, 15:1166–1174.

    Article  CAS  PubMed  Google Scholar 

  14. Irmak MK, Sizlan A. Essential hypertension seems to result from melatonin-induced epigenetic modifications in area postrema. Med Hypotheses 2006, 66:1000–1007.

    Article  CAS  PubMed  Google Scholar 

  15. Morimoto S, Sasaki S, Itoh H, et al.: Sympathetic activation and contribution of genetic factors in hypertension with neurovascular compression of the rostral ventrolateral medulla. J Hypertens 1999, 17:1577–1582.

    Article  CAS  PubMed  Google Scholar 

  16. • Sharma R, Ottenhof T, Rzeczkowska PA, Niles LP: Epigenetic targets for melatonin: induction of histone H3 hyperacetylation and gene expression in C17.2 neural stem cells. J Pineal Res 2008, 45:277–284. This reference is an in vitro study that provides strong evidence for epigenetic regulation of neural stem cells by histone acetylation, chromatin restructuring, and gene transcription. Epigenetic alteration of such stem cells could play a role in blood pressure control and hypertension by virtue of expressing melatonin receptors at brainstem vasomotor control centers.

    Article  CAS  PubMed  Google Scholar 

  17. Viswanathan M, Laitinen JT, Saavedra JM: Differential expression of melatonin receptors in spontaneously hypertensive rats. Neuroendocrinology 1992, 56:864–870.

    Article  CAS  PubMed  Google Scholar 

  18. • Esler M, Eikelis N, Schlaich M, et al.: Human sympathetic nerve biology: parallel influences of stress and epigenetics in essential hypertension and panic disorder. Ann N Y Acad Sci 2008, 1148:338–348. This reference describes a novel hypothesis for epigenetic regulation resulting from induction of the phenylethanolamine N-methyltransferase (PNMT) gene; co-release of this gene’s product, epinephrine; and silencing of the norepinephrine transporter gene by methylation-related inhibitory transcription factor-2 (MECP2). This sequence of events appears to be common to mental stress-related diseases such as panic disorder and essential hypertension.

    Article  CAS  PubMed  Google Scholar 

  19. • Yu Z, Kong Q, Kone BC: CREB trans-activation of disruptor of telomeric silencing-1 mediates forskolin inhibition of CTGF transcription in mesangial cells. Am J Physiol Renal Physiol 2010, 298:F617–F624. This reference describes an in vitro study in mouse mesangial cells that provides strong evidence for epigenetic regulation of the connective tissue growth factor (CTGF) gene by forskolin, a medicinal plant product. A complex mechanism is reported, which involves forskolin attaching to a cAMP response element binding (CREB), histone disruption of telomeric silencing (Dot), and hypermethylation of histone H3K79 at the CTGF promoter region, which could modify the expression of CTGF and play roles in blood vessel remodeling and renal fibrotic processes involved in hypertension.

    Article  CAS  PubMed  Google Scholar 

  20. Wandji SA, Gadsby JE, Barber JA, Hammond JM: Messenger ribonucleic acids for MAC25 and connective tissue growth factor (CTGF) are inversely regulated during folliculogenesis and early luteogenesis. Endocrinology 2000, 141:2648–2657.

    Article  CAS  PubMed  Google Scholar 

  21. Rodriguez-Iturbe B: Arteriolar remodeling in essential hypertension: Are connective tissue growth factor and transforming growth factor involved? Kidney Int 2006, 69:1104–1105.

    Article  CAS  PubMed  Google Scholar 

  22. Gumz ML, Stow LR, Lynch IJ, et al.: The circadian clock protein Period 1 regulates expression of the renal epithelial sodium channel in mice. J Clin Invest 2009, 119:2423–2434.

    Article  CAS  PubMed  Google Scholar 

  23. Steger DJ, Lefterova MI, Ying L, et al.: DOT1L/KMT4 recruitment and H3K79 methylation are ubiquitously coupled with gene transcription in mammalian cells. Mol Cell Biol 2008, 28:2825–2839.

    Article  CAS  PubMed  Google Scholar 

  24. • Zhang D, Yu ZY, Cruz P, et al.: Epigenetics and the control of epithelial sodium channel expression in collecting duct. Kidney Int 2009, 75:260–267. This reference presents strong evidence for a novel hypothesis and a complex mechanism wherein aldosterone, the sodium-conserving product of the RAS, disrupts interaction of a telomere-silencing molecule (Dot1a) and a fused acute lymphoblastic leukemia–multiple lineage leukemia (ALL, MLL) gene on chromosome 9 (Af9). This mechanism can be induced by glucocorticoid kinase-1 phosphorylation of Af9, which inhibits expression of Dot1a and Af9 and results in histone H3K79 hypomethylation and disinhibition of an epithelial sodium channel promoter. Such a mechanism could contribute to the high sodium-channel permeability described in human populations with predilections for salt-sensitive hypertension.

    Article  CAS  PubMed  Google Scholar 

  25. Lee JY, Prineas RJ, Eaton JW: Heritability of erythrocyte sodium permeability: a possible genetic marker for hypertension. Ann Clin Lab Sci 2009, 39:241–250.

    PubMed  Google Scholar 

  26. Alikhani-Koopaei R, Fouladkou F, Frey FJ, Frey BM: Epigenetic regulation of 11 beta-hydroxysteroid dehydrogenase type 2 expression. J Clin Invest 2004, 114:1146–1157.

    CAS  PubMed  Google Scholar 

  27. • Friso S, Pizzolo F, Choi SW, et al.: Epigenetic control of 11 beta-hydroxysteroid dehydrogenase 2 gene promoter is related to human hypertension. Atherosclerosis 2008, 199:323–327. This reference describes a human study providing strong evidence that epigenetic regulation of a gene that breaks down cortisol to cortisone metabolites contributes to exaggerated responsiveness of the RAS to salt and to salt-sensitive hypertension.

    Article  CAS  PubMed  Google Scholar 

  28. Pippal JB, Fuller PJ: Structure-function relationships in the mineralocorticoid receptor. J Mol Endocrinol 2008, 41:405–413.

    Article  CAS  PubMed  Google Scholar 

  29. Ferrari P, Sansonnens A, Dick B, Frey FJ: In vivo 11beta-HSD-2 activity: variability, salt-sensitivity, and effect of licorice. Hypertension 2001, 38:1330–1336.

    Article  CAS  PubMed  Google Scholar 

  30. Yang HM, Lohmeier TE: Influence of endogenous angiotensin on the renovascular response to norepinephrine. Hypertension 1993, 21:695–703.

    CAS  PubMed  Google Scholar 

  31. Farah V, Elased KM, Morris M: Genetic and dietary interactions: role of angiotensin AT1a receptors in response to a high-fructose diet. Am J Physiol Heart Circ Physiol 2007, 293:H1083–H1089.

    Article  CAS  PubMed  Google Scholar 

  32. Shim CY, Ha JW, Park S, et al.: Exaggerated blood pressure response to exercise is associated with augmented rise of angiotensin II during exercise. J Am Coll Cardiol 2008, 52:287–292.

    Article  CAS  PubMed  Google Scholar 

  33. • Guan J, Mao C, Xu F, et al.: Prenatal dehydration alters renin-angiotensin system associated with angiotensin-increased blood pressure in young offspring. Hypertens Res 2009, 32:1104–1111. This reference describes an experimental animal study providing strong evidence for epigenetic regulation of the RAS by demonstrating upregulation and exaggerated vasopressor responses in response to 3 days of prenatal exposure to limiting maternal water intake. Such exaggerated responses are found in humans with genetic predilections for hypertension.

    Article  CAS  PubMed  Google Scholar 

  34. Bogdarina I, Welham S, King PJ, et al.: Epigenetic modification of the renin-angiotensin system in the fetal programming of hypertension. Circ Res 2007, 100:520–526.

    Article  CAS  PubMed  Google Scholar 

  35. Zhang Z, Rhinehart K, Solis G, et al.: Chronic ANG II infusion increases NO generation by rat descending vasa recta. Am J Physiol Heart Circ Physiol 2005, 288:H29–H36.

    Article  CAS  PubMed  Google Scholar 

  36. Welch WJ, Chabrashvili T, Solis G, et al.: Role of extracellular superoxide dismutase in the mouse angiotensin slow pressor response. Hypertension 2006, 48:934–941.

    Article  CAS  PubMed  Google Scholar 

  37. Guzik TJ, Sadowski J, Guzik B, et al.: Coronary artery superoxide production and nox isoform expression in human coronary artery disease. Arterioscler Thromb Vasc Biol 2006, 26:333–339.

    Article  CAS  PubMed  Google Scholar 

  38. Wang D, Chabrashvili T, Borrego L, et al.: Angiotensin II infusion alters vascular function in mouse resistance vessels: roles of O and endothelium. J Vasc Res 2006, 43:109–119.

    Article  CAS  PubMed  Google Scholar 

  39. Modlinger P, Chabrashvili T, Gill PS, et al.: RNA silencing in vivo reveals role of p22phox in rat angiotensin slow pressor response. Hypertension 2006, 47:238–244.

    Article  CAS  PubMed  Google Scholar 

  40. Marfella CG, Henninger N, LeBlanc SE, et al.: A mutation in the mouse Chd2 chromatin remodeling enzyme results in a complex renal phenotype. Kidney Blood Press Res 2008, 31:421–432.

    Article  CAS  PubMed  Google Scholar 

  41. Bader M: Tissue renin-angiotensin-aldosterone systems: targets for pharmacological therapy. Annu Rev Pharmacol Toxicol 2010, 50:439–465.

    Article  CAS  PubMed  Google Scholar 

  42. Le Clair C, Abbi T, Sandhu H, Tappia PS: Impact of maternal undernutrition on diabetes and cardiovascular disease risk in adult offspring. Can J Physiol Pharmacol 2009, 87:161–179.

    Article  PubMed  CAS  Google Scholar 

  43. • Goyal R, Goyal D, Leitzke A, et al.: Brain renin-angiotensin system: fetal epigenetic programming by maternal protein restriction during pregnancy. Reprod Sci 2010, 17:227–238. This reference describes an experimental animal study providing strong evidence for epigenetic regulation of the brain RAS. A decrement in expression of an angiotensin-converting enzyme gene related to hypomethylation of the CpG islands at the promoter region is demonstrated. Such an effect could contribute to cognitive deficits associated with hypertension in humans.

    Article  PubMed  CAS  Google Scholar 

Download references

Disclosure

No potential conflicts of interest relevant to this article were reported.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Richard M. Millis.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Millis, R.M. Epigenetics and Hypertension. Curr Hypertens Rep 13, 21–28 (2011). https://doi.org/10.1007/s11906-010-0173-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11906-010-0173-8

Keywords

Navigation