Skip to main content

Advertisement

Log in

Primary Pediatric Hypertension: Current Understanding and Emerging Concepts

  • Therapeutic Trials (M Weir, Section Editor)
  • Published:
Current Hypertension Reports Aims and scope Submit manuscript

Abstract

The rising prevalence of primary pediatric hypertension and its tracking into adult hypertension point to the importance of determining its pathogenesis to gain insights into its current and emerging management. Considering that the intricate control of BP is governed by a myriad of anatomical, molecular biological, biochemical, and physiological systems, multiple genes are likely to influence an individual’s BP and susceptibility to develop hypertension. The long-term regulation of BP rests on renal and non-renal mechanisms. One renal mechanism relates to sodium transport. The impaired renal sodium handling in primary hypertension and salt sensitivity may be caused by aberrant counter-regulatory natriuretic and anti-natriuretic pathways. The sympathetic nervous and renin-angiotensin-aldosterone systems are examples of antinatriuretic pathways. An important counter-regulatory natriuretic pathway is afforded by the renal autocrine/paracrine dopamine system, aberrations of which are involved in the pathogenesis of hypertension, including that associated with obesity. We present updates on the complex interactions of these two systems with dietary salt intake in relation to obesity, insulin resistance, inflammation, and oxidative stress. We review how insults during pregnancy such as maternal and paternal malnutrition, glucocorticoid exposure, infection, placental insufficiency, and treatments during the neonatal period have long-lasting effects in the regulation of renal function and BP. Moreover, these effects have sex differences. There is a need for early diagnosis, frequent monitoring, and timely management due to increasing evidence of premature target organ damage. Large controlled studies are needed to evaluate the long-term consequences of the treatment of elevated BP during childhood, especially to establish the validity of the current definition and treatment of pediatric 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 •• Of major importance

  1. Yoon SS, Carroll MD, Fryar CD. Hypertension prevalence and control among adults: United States, 2011-2014. NCHS Data Brief. 2015;220:1–8.

    Google Scholar 

  2. Falkner B. Recent clinical and translational advances in pediatric hypertension. Hypertension. 2015;65(5):926–31. doi:10.1161/HYPERTENSIONAHA.114.03586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bucher BS, Ferrarini A, Weber N, Bullo M, Bianchetti MG, Simonetti GD. Primary hypertension in childhood. Curr Hypertens Rep. 2013;15(5):444–52. doi:10.1007/s11906-013-0378.

    Article  CAS  PubMed  Google Scholar 

  4. Roulet C, Bovet P, Brauchli T, Simeoni U, Xi B, Santschi V, et al. Secular trends in blood pressure in children: a systematic review. J Clin Hypertens (Greenwich). 2016; doi:10.1111/jch.12955.

  5. Moyer VA. U.S. Preventive Services Task Force. Screening for primary hypertension in children and adolescents: U.S. Preventive Services Task Force recommendation statement. Ann Intern Med. 2013;159(9):613–9.

    Article  PubMed  Google Scholar 

  6. • Lee CG. The emerging epidemic of hypertension in Asian children and adolescents. Curr Hypertens Rep. 2014;16:495. doi:10.1007/s11906-014-0495-z. This study shows that increased prevalence of pediatric hypertension is worldwide and associated with the obesity epidemic.

    Article  PubMed  Google Scholar 

  7. Yan W, Li X, Zhang Y, Niu D, Mu K, Ye Y, et al. Reevaluate secular trends of body size measurements and prevalence of hypertension among Chinese children and adolescents in past two decades. J Hypertens. 2016;34(12):2337–43.

    Article  CAS  PubMed  Google Scholar 

  8. Thompson M, Dana T, Bougatsos C, Blazina I, Norris SL. Screening for hypertension in children and adolescents to prevent cardiovascular disease. Pediatrics. 2013;131(3):490–525. doi:10.1542/peds.2012-3523.

    Article  PubMed  Google Scholar 

  9. Rao G. Diagnosis, epidemiology, and management of hypertension in children. Pediatrics. 2016;138(2) doi:10.1542/peds.2015-3616.

  10. Magnussen CG, Smith KJ. Pediatric blood pressure and adult preclinical markers of cardiovascular disease. Clin Med Insights Blood Disord. 2016;4(9):1–8.

    Article  Google Scholar 

  11. • Chu C, Dai Y, Mu J, Yang R, Wang M, Yang J, et al. Associations of risk factors in childhood with arterial stiffness 26 years later: the Hanzhong adolescent hypertension cohort. J Hypertens. 2017;35(Suppl 1):S10–5. doi:10.1097/HJH.0000000000001242. Premature target organ damage (long-term arterial stiffness) with pediatric hypertension evaluated by brachial-ankle pulse wave velocity.

    Article  CAS  PubMed  Google Scholar 

  12. National High Blood Pressure Education Program Working Group on High Blood Pressure in Children and Adolescents. The fourth report on the diagnosis, evaluation, and treatment of high blood pressure in children and adolescents. Pediatrics. 2004;114(2 Suppl 4th Report):555–76.

    Article  Google Scholar 

  13. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. National High Blood Pressure Education Program. Bethesda: National Heart, Lung, and Blood Institute (US); 2004.

    Google Scholar 

  14. Viera AJ, Lin FC, Hinderliter AL, Shimbo D, Person SD, Pletcher MJ, et al. Nighttime blood pressure dipping in young adults and coronary artery calcium 10–15 years later: the coronary artery risk development in young adults study. Hypertension. 2012;59:1157–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Flynn J, Zhang Y, Solar-Yohay S, Shi V. Clinical and demographic characteristics of children with hypertension. Hypertension. 2012;60(4):1047–54. doi:10.1161/HYPERTENSIONAHA.112.197525.

    Article  CAS  PubMed  Google Scholar 

  16. Brady TM, Feld LG. Pediatric approach to hypertension. Semin Nephrol. 2009;29(4):379–88.

    Article  PubMed  Google Scholar 

  17. •• Barker DJ. The fetal origins of hypertension. J Hypertens Suppl. 1996;14:S117–20. Early studies on “thrifty phenotype” introduced by Barker.

    CAS  PubMed  Google Scholar 

  18. • Fraser A, Nelson SM, Macdonald-Wallis C, Sattar N, Lawlor DA. Hypertensive disorders of pregnancy and cardiometabolic health in adolescent offspring. Hypertension. 2013;62:614–20. The Avon longitudinal study exemplifies fetal programming where insults during pregnancy (gestational hypertension) are associated with increased blood pressure in the offspring.

    Article  CAS  PubMed  Google Scholar 

  19. Von Ehr J, von Versen-Höynck F. Implications of maternal conditions and pregnancy course on offspring’s medical problems in adult life. Arch Gynecol Obstet. 2016;294(4):673–9. doi:10.1007/s00404-016-4178-7.

    Article  Google Scholar 

  20. Lopez-Lopez J, Jaramillo PL, Camacho PA, Arbelaez DG, Cohen DD. The link between fetal programming, inflammation, muscular strength, and blood pressure. Mediat Inflamm. 2015;710613 doi:10.1155/2015/710613.

  21. Lewandowski AJ, Davis EF, Yu G, Digby JE, Boardman H, Whitworth P, et al. Elevated blood pressure in preterm-born offspring associates with a distinct antiangiogenic state and microvascular abnormalities in adult life. Hypertension. 2015;65(3):607–14. doi:10.1161/HYPERTENSIONAHA.114.04662.

    Article  CAS  PubMed  Google Scholar 

  22. Alexander BT, Dasinger JH, Intapad S. Fetal programming and cardiovascular pathology. Compr Physiol. 2015;5(2):997–1025. doi:10.1002/cphy.c140036.

    Article  PubMed  PubMed Central  Google Scholar 

  23. • Brenner BM, Lawler EV, Mackenzie HS. The hyperfiltration theory: a paradigm shift in nephrology. Kidney Int. 1996;49(6):1774–7. The Brenner’s hypothesis supports the idea that decreased development of nephrons from prenatal insults leads to compensatory hypertrophy of remaining nephrons, hyperfiltration, and hastened decline of renal function.

    Article  CAS  PubMed  Google Scholar 

  24. • Gurusinghe S, Palvanov A, Bittman ME, Singer P, Frank R, Chorny N, et al. Kidney volume and ambulatory blood pressure in children. J Clin Hypertens (Greenwich). 2016; doi:10.1111/jch.12954. This study showed that kidney volume is positively correlated with SBP in ages 4 – 20 years.

  25. • Shahzad T, Radajewski S, Chao CM, Bellusci S, Ehrhardt H. Pathogenesis of bronchopulmonary dysplasia: when inflammation meets organ development. Mol Cell Pediatr. 2016;3(1):23. doi:10.1186/s40348-016-0051-9. Excessive oxygen exposure in preterm infants can injure vasculogenesis increasing future risk for hypertension.

    Article  PubMed  PubMed Central  Google Scholar 

  26. •• Grigore D, Ojeda NB, Alexander BT. Sex differences in the fetal programming of hypertension. Gend Med. 2008;5(Suppl A):S121–32. doi:10.1016/j.genm.2008.03.012. Testosterone has permissive effect in the development of hypertension in the offspring as evidenced by decrease in nephron number, increase in endothelin production and increase in reactive oxygen species.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Ojeda NB, Intapad S, Alexander BT. Sex differences in the developmental programming of hypertension. Acta Physiol (Oxf). 2014;210(2):307–16. doi:10.1111/apha.12206.

    Article  CAS  Google Scholar 

  28. Thornburg KL, O'Tierney PF, Louey S. Review: The placenta is a programming agent for cardiovascular disease. Placenta. 2010;31(Suppl):S54–9. doi:10.1016/j.placenta.2010.01.002.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Baum M. Role of the kidney in the prenatal and early postnatal programming of hypertension. Am J Physiol Ren Physiol. 2010;298(2):F235–47. doi:10.1152/ajprenal.00288.2009.

    Article  CAS  Google Scholar 

  30. Tain YL, Lee WC, Wu KL, Leu S, Chan JY. Targeting arachidonic acid pathway to prevent programmed hypertension in maternal fructose-fed male adult rat offspring. J Nutr Biochem. 2016;38:86–92.

    Article  CAS  PubMed  Google Scholar 

  31. Gray C, Al-Dujaili EA, Sparrow AJ, Gardiner SM, Craigon J, Welham SJ, et al. Excess maternal salt intake produces sex-specific hypertension in offspring: putative roles for kidney and gastrointestinal sodium handling. PLoS One. 2013;8(8):e72682. doi:10.1371/journal.pone.0072682.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Maruyama K, Kagota S, Van Vliet BN, Wakuda H, Shinozuka K. A maternal high salt diet disturbs cardiac and vascular function of offspring. Life Sci. 2015;136:42–51. doi:10.1016/j.lfs.2015.06.023.

    Article  CAS  PubMed  Google Scholar 

  33. Porter JP, King SH, Honeycutt AD. Prenatal high-salt diet in the Sprague-Dawley rat programs blood pressure and heart rate hyperresponsiveness to stress in adult female offspring. Am J Phys Regul Integr Comp Phys. 2007;293(1):R334–42.

    CAS  Google Scholar 

  34. Contreras RJ, Wong DL, Henderson R, Curtis KS, Smith JC. High dietary NaCl early in development enhances mean arterial pressure of adult rats. Physiol Behav. 2000;71(1–2):173–81.

    Article  CAS  PubMed  Google Scholar 

  35. Bo L, Jiang L, Zhou A, Wu C, Li J, Gao Q, et al. Maternal high-salt diets affected pressor responses and microvasoconstriction via PKC/BK channel signaling pathways in rat offspring. Mol Nutr Food Res. 2015;59(6):1190–9. doi:10.1002/mnfr.201400841.

    Article  CAS  PubMed  Google Scholar 

  36. Piecha G, Koleganova N, Ritz E, Müller A, Fedorova OV, Bagrov AY, et al. High salt intake causes adverse fetal programming—vascular effects beyond blood pressure. Nephrol Dial Transplant. 2012;27(9):3464–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Koleganova N, Piecha G, Ritz E, Becker LE, Müller A, Weckbach M, et al. Both high and low maternal salt intake in pregnancy alter kidney development in the offspring. Am J Physiol Ren Physiol. 2011;301(2):F344–54.

    Article  CAS  Google Scholar 

  38. Tain YL, Lee WC, Leu S, Wu K, Chan J. High salt exacerbates programmed hypertension in maternal fructose-fed male offspring. Nutr Metab Cardiovasc Dis. 2015;25(12):1146–51. doi:10.1016/j.numecd.2015.08.002.

    Article  CAS  PubMed  Google Scholar 

  39. • Li S, Fang Q, Zhou A, Wu L, Shi A, Cao L, et al. Intake of high sucrose during pregnancy altered large-conductance Ca2+-activated K+ channels and vessel tone in offspring's mesenteric arteries. Hypertens Res. 2013;36(2):158–65. Not only high maternal salt intake but also high sucrose diet increased the offspring’s sensitivity to angiotensin II similar to a maternal high salt intake.

    Article  CAS  PubMed  Google Scholar 

  40. Khan IY, Dekou V, Douglas G, Jensen R, Hanson MA, Poston L, et al. A high-fat diet during rat pregnancy or suckling induces cardiovascular dysfunction in adult offspring. Am J Phys Regul Integr Comp Phys. 2005;288(1):R127–33.

    CAS  Google Scholar 

  41. • Masuyama H, Mitsui T, Eguchi T, Tamada S, Hiramatsu Y. The effects of paternal high-fat diet exposure on offspring metabolism with epigenetic changes in the mouse adiponectin and leptin gene promoters. Am J Physiol Endocrinol Metab. 2016;311(1):E236–45. doi:10.1152/ajpendo.00095.2016. High paternal fat diet also plays a role in metabolic syndrome in the offspring.

    Article  PubMed  Google Scholar 

  42. Elahi MM, Cagampang FR, Mukhtar D, Anthony FW, Ohri SK, Hanson MA. Long-term maternal high-fat feeding from weaning through pregnancy and lactation predisposes offspring to hypertension, raised plasma lipids and fatty liver in mice. Br J Nutr. 2009;102(4):514.

    Article  CAS  PubMed  Google Scholar 

  43. Elahi MM, Matata BM. Effects of maternal high-fat diet and statin treatment on bone marrow endothelial progenitor cells and cardiovascular risk factors in female mice offspring fed a similar diet. Nutrition. 2017;35:6–13. doi:10.1016/j.nut.2016.10.01.

    Article  CAS  PubMed  Google Scholar 

  44. • Nguyen LT, Chen H, Pollock CA, Saad S. Sirtuins-mediators of maternal obesity-induced complications in offspring? FASEB J. 2016;30(4):1383–90. doi:10.1096/fj.15-280743. Sirtuins are proposed as one of the mediators of the fetal programming of childhood obesity.

    Article  CAS  PubMed  Google Scholar 

  45. Gregory EF, Goldshore MA, Henderson JL, Weatherford RD, Showell NN. Infant growth following maternal participation in a gestational weight management intervention. Child Obes. 2016;12(3):219–25. doi:10.1089/chi.2015.0238.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Zimanyi MA, Bertram JF, Black MJ. Nephron number and blood pressure in rat offspring with maternal high-protein diet. Pediatr Nephrol. 2002;17(12):1000–4.

    Article  PubMed  Google Scholar 

  47. Lee JH, Lee H, Lee SM, Kang PJ, Kim KC, Hong YM. Changes of blood pressure, abdominal visceral fat tissue and gene expressions in fetal programming induced rat model after amlodipine-losartan combination treatment. Clin Hypertens. 2016;22:12. doi:10.1186/s40885-016-0046-9.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Hoppe CC, Evans RG, Bertram JF, Moritz KM. Effects of dietary protein restriction on nephron number in the mouse. Am J Phys Regul Integr Comp Phys. 2007;292:R1768–74.

    CAS  Google Scholar 

  49. de Belchior AC, Freire DD Jr, da Costa CP, Vassallo DV, Padilha AS, Dos Santos L. Maternal protein restriction compromises myocardial contractility in the young adult rat by changing proteins involved in calcium handling. J Appl Physiol. 2016;120(3):344–50. doi:10.1152/japplphysiol.00246.2015.

    Article  PubMed  CAS  Google Scholar 

  50. • Harrison M, Langley-Evans SC. Intergenerational programming of impaired nephrogenesis and hypertension in rats following maternal protein restriction during pregnancy. Br J Nutr. 2009;101(7):1020–30. doi:10.1017/S0007114508057607. This study demonstrates that maternal protein restriction is associated with 2 generations of offsprings with high blood pressure.

    Article  CAS  PubMed  Google Scholar 

  51. Bertram C, Khan O, Ohri S, Phillips DI, Matthews SG, Hanson MA. Transgenerational effects of prenatal nutrient restriction on cardiovascular and hypothalamic-pituitary-adrenal function. J Physiol. 2008;586(8):2217–29. doi:10.1113/jphysiol.2007.147967.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. McMullen S, Langley-Evans SC. Maternal low-protein diet in rat pregnancy programs blood pressure through sex-specific mechanisms. Am J Phys Regul Integr Comp Phys. 2005;288(1):R85–90.

    CAS  Google Scholar 

  53. Harvey TJ, Murphy RM, Morrison JL, Posterino GS. Maternal nutrient restriction alters Ca2+ handling properties and contractile function of isolated left ventricle bundles in male but not female juvenile rats. PLoS One. 2015;10(9):e0138388. doi:10.1371/journal.pone.0138388.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. de Brito Alves JL, de Oliveira JM, Ferreira DJ, Barros MA, Nogueira VO, Alves DS, et al. Maternal protein restriction induced-hypertension is associated to oxidative disruption at transcriptional and functional levels in the medulla oblongata. Clin Exp Pharmacol Physiol. 2016;43(12):1177–84. doi:10.1111/1440-1681.12667.

    Article  PubMed  CAS  Google Scholar 

  55. Wattez JS, Delahaye F, Barella LF, Dickes-Coopman A, Montel V, Breton C, et al. Short- and long-term effects of maternal perinatal undernutrition are lowered by cross-fostering during lactation in the male rat. J Dev Orig Health Dis. 2014;5(2):109–20. doi:10.1017/S2040174413000548.

    Article  CAS  PubMed  Google Scholar 

  56. • Simonds SE, Pryor JT, Cowley MA. Does leptin cause an increase in blood pressure in animals and humans? Curr Opin Nephrol Hypertens. 2017;26(1):20–5. Chronic administration of leptin does not increase blood pressure.

    Article  CAS  PubMed  Google Scholar 

  57. Thanos PK, Zhuo J, Robison L, Kim R, Ananth M, Choai I, et al. Suboptimal maternal diets alter mu opioid receptor and dopamine type 1 receptor binding but exert no effect on dopamine transporters in the offspring brain. Int J Dev Neurosci. 2016 Sep 22. pii: S0736-5748(16)30211–8. doi:10.1016/j.ijdevneu.2016.09.008.

  58. Amare AT, Schubert KO, Klingler-Hoffmann M, Cohen-Woods S, Baune BT. The genetic overlap between mood disorders and cardiometabolic diseases: a systematic review of genome wide and candidate gene studies. Transl Psychiatry. 2017;7(1):e1007. doi:10.1038/tp.2016.261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang H, Sun ZQ, Liu SS, Yang LN. Association between GRK4 and DRD1 gene polymorphisms and hypertension: a meta-analysis. Clin Interv Aging. 2015;11:17–27. doi:10.2147/CIA.S94510.

    Article  PubMed  PubMed Central  Google Scholar 

  60. • Armando I, Konkalmatt P, Felder RA, Jose PA. The renal dopaminergic system: novel diagnostic and therapeutic approaches in hypertension and kidney disease. Transl Res. 2015;165(4):505–11. This is an updated review on the renal dopaminergic system and how dysfunction of which would cause decreased sodium excretion and increased blood pressure.

    Article  CAS  PubMed  Google Scholar 

  61. Choi MR, Kouyoumdzian NM, Rukavina Mikusic NL, Kravetz MC, Rosón MI, Rodríguez Fermepin M, et al. Renal dopaminergic system: pathophysiological implications and clinical perspectives. World J Nephrol. 2015;4(2):196–212.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Zhang MZ, Harris RC. Antihypertensive mechanisms of intra-renal dopamine. Curr Opin Nephrol Hypertens. 2015;24(2):117–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Asghar M, Tayebati SK, Lokhandwala MF, Hussain T. Potential dopamine-1 receptor stimulation in hypertension management. Curr Hypertens Rep. 2011;13(4):294–302.

    Article  CAS  PubMed  Google Scholar 

  64. Rayner B, Ramesar R. The importance of G protein-coupled receptor kinase 4 (GRK4) in pathogenesis of salt sensitivity, salt sensitive hypertension and response to antihypertensive treatment. Int J Mol Sci. 2015;16(3):5741–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Rayner BL, Spence JD. Hypertension in blacks: insights from Africa. J Hypertens. 2017;35(2):234–9. doi:10.1097/HJH.000000000000117.

    Article  CAS  PubMed  Google Scholar 

  66. Dagan A, Habib S, Gattineni J, Dwarakanath V, Baum M. Prenatal programming of rat thick ascending limb chloride transport by low-protein diet and dexamethasone. Am J Phys Regul Integr Comp Phys. 2009;297(1):R93–9. doi:10.1152/ajpregu.91006.2008.

    CAS  Google Scholar 

  67. Wesseling S, Koeners MP, Joles JA. Salt sensitivity of blood pressure: developmental and sex-related effects. Am J Clin Nutr. 2011;94(6 Suppl):1928S–32S.

    Article  CAS  PubMed  Google Scholar 

  68. • Khorram O, Chuang TD, Pearce WJ. Long-term effects of maternal undernutrition on offspring carotid artery remodeling: role of miR-29c. J Dev Orig Health Dis. 2015 Aug;6(4):342–9. doi: 10.1017/S2040174415001208. Epub 2015 May 26. Maternal undernutrition (decreased protein intake) increases extracellular matrix proteins in the carotid arteries related to suppression of miR-29C.

  69. Sathishkumar K, Balakrishnan MP, Yallampalli C. Enhanced mesenteric arterial responsiveness to angiotensin II is androgen receptor-dependent in prenatally protein-restricted adult female rat offspring. Biol Reprod. 2015;92(2):55.

    Article  PubMed  CAS  Google Scholar 

  70. Cooke CL, Zhao L, Gysler S, Arany E, Regnault TR. Sex-specific effects of low protein diet on in utero programming of renal G-protein coupled receptors. J Dev Orig Health Dis. 2014;5(1):36–44. doi:10.1017/S2040174413000524.

    Article  CAS  PubMed  Google Scholar 

  71. Chisaka T, Mogi M, Nakaoka H, Kan-No H, Tsukuda K, Wang XL, et al. Low-protein diet-induced fetal growth restriction leads to exaggerated proliferative response to vascular injury in postnatal life. Am J Hypertens. 2016;29(1):54–62.

    Article  PubMed  Google Scholar 

  72. de Lima MC, Scabora JE, Lopes A, Mesquita FF, Torres D, Boer PA, et al. Early changes of hypothalamic angiotensin II receptors expression in gestational protein-restricted offspring: effect on water intake, blood pressure and renal sodium handling. J Renin-Angiotensin-Aldosterone Syst. 2013;14(3):271–82. doi:10.1177/1470320312456328.

    Article  PubMed  CAS  Google Scholar 

  73. Casas-Agustench P, Iglesias-Gutiérrez E, Dávalos A. Mother’s nutritional miRNA legacy: nutrition during pregnancy and its possible implications to develop cardiometabolic disease in later life. Pharmacol Res. 2015;100:322–34.

    Article  CAS  PubMed  Google Scholar 

  74. •• Koeners MP, Wesseling S, Ulu A, Sepúlveda RL, Morisseau C, Braam B, et al. Soluble epoxide hydrolase in the generation and maintenance of high blood pressure in spontaneously hypertensive rats. Am J Physiol Endocrinol Metab. 2011;300(4):E691–8. doi:10.1152/ajpendo.00710.2010. Fetal programming may be reversed through inhibition of soluble epoxide hydrolase.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. •• Tain YL, Lee CT, Chan JY, Hsu CN. Maternal melatonin or N-acetylcysteine therapy regulates hydrogen sulfide-generating pathway and renal transcriptome to prevent prenatal NG-Nitro-L-arginine-methyl ester (L-NAME)-induced fetal programming of hypertension in adult male offspring. Am J Obstet Gynecol. 2016;215(5):636.e1–636.e72. Maternal antioxidant treatment with melatonin and N-acetylcysteine may reverse fetal programming of hypertension.

    Article  CAS  Google Scholar 

  76. •• de Bem GF, da Costa CA, de Oliveira PR, Cordeiro VS, Santos IB, de Carvalho LC, et al. Protective effect of Euterpe oleracea Mart (açaí) extract on programmed changes in the adult rat offspring caused by maternal protein restriction during pregnancy. J Pharm Pharmacol. 2014;66(9):1328–38. doi:10.1111/jphp.12258. This study shows that fetal programming may be reversible in experimental conditions by administration of acai extract.

    Article  PubMed  CAS  Google Scholar 

  77. Szeto IM, Aziz A, Das PJ, Taha AY, Okubo N, Reza-Lopez S, et al. High multivitamin intake by Wistar rats during pregnancy results in increased food intake and components of the metabolic syndrome in male offspring. Am J Phys Regul Integr Comp Phys. 2008;295(2):R575–82. doi:10.1152/ajpregu.90354.2008.

    CAS  Google Scholar 

  78. • Pannia E, Cho CE, Kubant R, Sánchez-Hernández D, Huot PS, Chatterjee D, et al. A high multivitamin diet fed to Wistar rat dams during pregnancy increases maternal weight gain later in life and alters homeostatic, hedonic and peripheral regulatory systems of energy balance. Behav Brain Res. 2015;278:1–11. doi:10.1016/j.bbr.2014.09.019. Excessive maternal vitamin intake alters the metabolism of offspring.

    Article  CAS  PubMed  Google Scholar 

  79. Szeto IM, Das PJ, Aziz A, Anderson GH. Multivitamin supplementation of Wistar rats during pregnancy accelerates the development of obesity in offspring fed an obesogenic diet. Int J Obes. 2009;33(3):364–72. doi:10.1038/ijo.2008.281.

    Article  CAS  Google Scholar 

  80. Cho CE, Sánchez-Hernández D, Reza-López SA, Huot PS, Kim YI, Anderson GH. Obesogenic phenotype of offspring of dams fed a high multivitamin diet is prevented by a post-weaning high multivitamin or high folate diet. Int J Obes. 2013;37(9):1177–82. doi:10.1038/ijo.2012.210.

    Article  CAS  Google Scholar 

  81. Dodic M, Abouantoun T, O'Connor A, Wintour EM, Moritz KM. Programming effects of short prenatal exposure to dexamethasone in sheep. Hypertension. 2002;40:729–34.

    Article  CAS  PubMed  Google Scholar 

  82. Dodic M, Samuel C, Moritz KM, Wintour EM, Morgan J, et al. Impaired cardiac functional reserve and left ventricular hypertrophy in adult sheep after prenatal dexamethasone exposure. Circ Res. 2002;89:623–9.

    Article  Google Scholar 

  83. O'Regan D, Kenyon CJ, Seckl JR, Holmes MC. Glucocorticoid exposure in late gestation in the rat permanently programs gender-specific differences in adult cardiovascular and metabolic physiology. Am J Physiol Endocrinol Metab. 2004;287:E863–70.

    Article  PubMed  CAS  Google Scholar 

  84. •• Dickinson H, Walker DW, Wintour EM, Moritz KM. Maternal dexamethasone treatment at midgestation reduces nephron number and alters renal gene expression in the fetal spiny mouse. Am J Phys Regul Integr Comp Phys. 2007;292:R453–61. Prenatal exposure to glucocorticoids reduces nephron numbers increasing risk of hypertension later in life.

    CAS  Google Scholar 

  85. Marshall AC, Shaltout HA, Nautiyal M, Rose JC, Chappell MC, Diz D. Fetal betamethasone exposure attenuates angiotensin-(1-7)-Mas receptor expression in the dorsal medulla of adult sheep. Peptides. 2013 Jun;44:25–31.. doi:10.1016/j.peptides.2013.03.018. Epub 2013 Mar 26.

  86. Marshall AC, Shaltout HA, Pirro NT, Rose JC, Diz DI, Chappell MC. Antenatal betamethasone exposure is associated with lower ANG-(1-7) and increased ACE in the CSF of adult sheep. Am J Phys Regul Integr Comp Phys. 2013;305(7):R679–88. doi:10.1152/ajpregu.00321.2013.

    CAS  Google Scholar 

  87. Cuffe JS, Burgess DJ, O'Sullivan L, Singh RR, Moritz KM. Maternal corticosterone exposure in the mouse programs sex-specific renal adaptations in the renin-angiotensin-aldosterone system in 6-month offspring. Physiol Rep. 2016 Apr;4(8). pii: e12754. doi:10.14814/phy2.12754.

  88. O'Sullivan L, Cuffe JS, Koning A, Singh RR, Paravicini TM, Moritz KM. Excess prenatal corticosterone exposure results in albuminuria, sex-specific hypotension, and altered heart rate responses to restraint stress in aged adult mice. Am J Physiol Ren Physiol. 2015;308(10):F1065–73. doi:10.1152/ajprenal.00676.2014.

    Article  CAS  Google Scholar 

  89. Tain YL, Sheen JM, Chen CC, Yu HR, Tiao MM, Kuo HC, et al. Maternal citrulline supplementation prevents prenatal dexamethasone-induced programmed hypertension. Free Radic Res. 2014;48(5):580–6. doi:10.3109/10715762.2014.895341.

    Article  CAS  PubMed  Google Scholar 

  90. Benediktsson R, Lindsay RS, Noble J, Seckl JR, Edwards CR. Glucocorticoid exposure in utero: new model for adult hypertension. Lancet. 1993;341:339–41.

  91. Dickinson H, Walker DW, Wintour EM, Moritz K. Maternal dexamethasone treatment at midgestation reduces nephron number and alters renal gene expression in the fetal spiny mouse. Am J Phys Regul Integr Comp Phys. 2007;292(1):R453–61.

    CAS  Google Scholar 

  92. Sheen JM, Hsieh CS, Tain YL, Li SW, Yu HR, Chen CC, et al. Programming effects of prenatal glucocorticoid exposure with a postnatal high-fat diet in diabetes mellitus. Int J Mol Sci. 2016;17(4):533. doi:10.3390/ijms17040533.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Rexhepaj R, Boini KM, Huang DY, Amann K, Artunc F, Wang K, et al. Role of maternal glucocorticoid inducible kinase SGK1 in fetal programming of blood pressure in response to prenatal diet. Am J Phys Regul Integr Comp Phys. 2008;294(6):R2008–13. doi:10.1152/ajpregu.00737.2007.

    CAS  Google Scholar 

  94. Woods LL. Maternal glucocorticoids and prenatal programming of hypertension. Am J Phys Regul Integr Comp Phys. 2006;291(4):R1069–75.

    CAS  Google Scholar 

  95. O'Sullivan L, Cuffe JS, Paravicini TM, Campbell S, Dickinson H, Singh RR, et al. Prenatal exposure to dexamethasone in the mouse alters cardiac growth patterns and increases pulse pressure in aged male offspring. PLoS One. 2013;8(7):e69149. doi:10.1371/journal.pone.0069149.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. • Wang X, Wang J, Luo H, Chen C, Pei F, Cai Y, et al. Prenatal lipopolysaccharide exposure causes mesenteric vascular dysfunction through the nitric oxide and cyclic guanosine monophosphate pathway in offspring. Free Radic Biol Med. 2015;86:322–30. doi:10.1016/j.freeradbiomed.2015.05.040. Prenatal LPS exposure increases oxidative stress which causes vascular dysfunction in offspring.

    Article  CAS  PubMed  Google Scholar 

  97. • Wang X, Luo H, Chen C, Chen K, Wang J, Cai Y, et al. Prenatal lipopolysaccharide exposure results in dysfunction of the renal dopamine D1 receptor in offspring. Free Radic Biol Med. 2014;76:242–50. Prenatal LPS exposure causes dysfunction of renal dopaminergic system decreasing sodium excretion.

    Article  CAS  PubMed  Google Scholar 

  98. Hao XQ, Zhang HG, Yuan ZB, Yang DL, Hao LY, Li XH. Prenatal exposure to lipopolysaccharide alters the intrarenal renin-angiotensin system and renal damage in offspring rats. Hypertens Res. 2010;33(1):76–82. doi:10.1038/hr.2009.185.

    Article  CAS  PubMed  Google Scholar 

  99. Gao M, Zhang X, Chen X, Mi C, Tang Y, Zhou J, et al. Prenatal exposure to lipopolysaccharide results in local RAS activation in the adipose tissue of rat offspring. PLoS One. 2014 Oct 31;9(10):e111376. doi:10.1371/journal.pone.0111376.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Wei YL, Li XH, Zhou JZ. Prenatal exposure to lipopolysaccharide results in increases in blood pressure and body weight in rats. Acta Pharmacol Sin. 2007;28(5):651–6.

    Article  CAS  PubMed  Google Scholar 

  101. •• Sanada H, Yoneda M, Yatabe J, Williams SM, Bartlett J, White MJ, et al. Common variants of the G protein-coupled receptor type 4 are associated with human essential hypertension and predict the blood pressure response to angiotensin receptor blockade. Pharmacogenomics J. 2016;16(1):3–9. doi:10.1038/tpj.2015.6. This study reviews the GRK4 variants and their response to antihypertensives (angiotensin receptor blocker).

    Article  CAS  PubMed  Google Scholar 

  102. • Jose PA, Felder RA, Yang Z, Zeng C, Eisner GM. Gastrorenal axis. Hypertension. 2016;67(6):1056–63. An oral sodium load causes gastrin release which then stimulates the renal dopaminergic system to excrete the sodium.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Baharnoori M, Bhardwaj SK, Srivastava LK. Effect of maternal lipopolysaccharide administration on the development of dopaminergic receptors and transporter in the rat offspring. PLoS One. 2013;8(1):e54439. doi:10.1371/journal.pone.0054439.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Luchicchi A, Lecca S, Melis M, De Felice M, Cadeddu F, Frau R, et al. Maternal immune activation disrupts dopamine system in the offspring. Int J Neuropsychopharmacol. 2016 Jul 5;19(7). pii: pyw007. doi:10.1093/ijnp/pyw007. Print 2016 Jul.

  105. Trøseid M, Nestvold TK, Rudi K, Thoresen H, Nielsen EW, Lappegård KT. Plasma lipopolysaccharide is closely associated with glycemic control and abdominal obesity: evidence from bariatric surgery. Diabetes Care. 2013;36(11):3627–32. doi:10.2337/dc13-0451.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Alexander BT. Placental insufficiency leads to development of hypertension in growth-restricted offspring. Hypertension. 41(3):457–62.

  107. Master JS, Zimanyi MA, Yin KV, Moritz KM, Gallo LA, Tran M, et al. Transgenerational left ventricular hypertrophy and hypertension in offspring after uteroplacental insufficiency in male rats. Clin Exp Pharmacol Physiol. 2014;41(11):884–9.

    Article  CAS  PubMed  Google Scholar 

  108. Tare M, Parkington HC, Bubb KJ, Wlodek ME. Uteroplacental insufficiency and lactational environment separately influence arterial stiffness and vascular function in adult male rats. Hypertension. 2012;60(2):378–86. doi:10.1161/HYPERTENSIONAHA.112.190876.

    Article  CAS  PubMed  Google Scholar 

  109. •• Mazzuca MQ, Wlodek ME, Dragomir NM, Parkington HC, Tare M. Uteroplacental insufficiency programs regional vascular dysfunction and alters arterial stiffness in female offspring. J Physiol. 2010;588(Pt 11):1997–2010. doi:10.1113/jphysiol.2010.187849. This study discusses the role of uteroplacental insufficiency in fetal programming of hypertension.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Gallo LA, Tran M, Cullen-McEwen LA, Denton KM, Jefferies AJ, Moritz KM, et al. Transgenerational programming of fetal nephron deficits and sex-specific adult hypertension in rats. Reprod Fertil Dev. 2014;26(7):1032–43. doi:10.1071/RD13133.

    Article  PubMed  Google Scholar 

  111. Dodson RB, Rozance PJ, Fleenor BS, Petrash CC, Shoemaker LG, Hunter KS, et al. Increased arterial stiffness and extracellular matrix reorganization in intrauterine growth-restricted fetal sheep. Pediatr Res. 2013;73(2):147–54. doi:10.1038/pr.2012.156.

    Article  CAS  PubMed  Google Scholar 

  112. Bauer R, Walter B, Bauer K, Klupsch R, Patt S, Zwiener U. Intrauterine growth restriction reduces nephron number and renal excretory function in newborn piglets. Acta Physiol Scand. 2002;176(2):83–90.

    Article  CAS  PubMed  Google Scholar 

  113. Baserga M, Bares AL, Hale MA, Callaway CW, McKnight RA. Lane PH, et al Uteroplacental insufficiency affects kidney VEGF expression in a model of IUGR with compensatory glomerular hypertrophy and hypertension. Early Hum Dev. 2009;85(6):361–7. doi:10.1016/j.earlhumdev.2008.12.015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. • Ojeda NB, Grigore D, Yanes LL, Iliescu R, Robertson EB, Zhang H, et al. Testosterone contributes to marked elevations in mean arterial pressure in adult male intrauterine growth restricted offspring. Am J Phys Regul Integr Comp Phys. 2007;292(2):R758–63. Sex differences is also present in uteroplacental insufficiency modulation of blood pressure.

    CAS  Google Scholar 

  115. US Burden of Disease Collaborators. The state of US health, 1990-2010: burden of diseases, injuries, and risk factors. JAMA. 2013;310:591–608.

    Article  PubMed Central  CAS  Google Scholar 

  116. •• Armando I, Villar VA, Jose PA. Dopamine and renal function and blood pressure regulation. Compr Physiol. 2011;1(3):1075–117. doi:10.1002/cphy.c100032. Comprehensive review of the renal dopaminergic system and GRK4 and role in blood pressure regulation.

    PubMed  Google Scholar 

  117. Glazier AM, Nadeau JH, Aitman TJ. Finding genes that underlie complex traits. Science. 2002;298:2345–9.

    Article  CAS  PubMed  Google Scholar 

  118. Jain S, Tillinger A, Mopidevi B, Pandey VG, Chauhan CK, Fiering SN, et al. Transgenic mice with -6A haplotype of the human angiotensinogen gene have increased blood pressure compared with -6G haplotype. J Biol Chem. 2010;285(52):41172–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Jain S, Prater A, Pandey V, Rana A, Puri N, Kumar A. A haplotype of angiotensin receptor type 1 associated with human hypertension increases blood pressure in transgenic mice. J Biol Chem. 2013;288(52):37048–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Mopidevi B, Kaw MK, Puri N, Ponnala M, Jain S, Rana A, et al. Variable transcriptional regulation of the human aldosterone synthase gene causes salt-dependent high blood pressure in transgenic mice. Circ Cardiovasc Genet. 2015;8(1):30–9. doi:10.1161/CIRCGENETICS.114.000694.

    Article  CAS  PubMed  Google Scholar 

  121. Wang Z, Zeng C, Villar VA, Chen SY, Konkalmatt P, Wang X, et al. 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:10.1161/HYPERTENSIONAHA.115.05962.

    CAS  PubMed  Google Scholar 

  122. Felder RA, Sanada H, Xu J, Yu PY, Wang Z, Watanabe H, et al. G protein-coupled receptor kinase 4 gene variants in human essential hypertension. Proc Natl Acad Sci U S A. 2002;99(6):3872–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Rhee MY, Yang SJ, Oh SW, Park Y, Kim CI, Park HK, et al. Novel genetic variations associated with salt sensitivity in the Korean population. Hypertens Res. 2011;34(5):606–11. doi:10.1038/hr.2010.278.

    Article  CAS  PubMed  Google Scholar 

  124. Carey RM, Schoeffel CD, Gildea JJ, Jones JE, McGrath HE, Gordon LN, et al. Salt sensitivity of blood pressure is associated with polymorphisms in the sodium-bicarbonate cotransporter. Hypertension. 2012;60(5):1359–66. doi:10.1161/HYPERTENSIONAHA.112.196071.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Wang Y, O’Connell JR, McArdle PF, Wade JB, Dorff SE, Shah SJ, et al. Whole-genome association study identifies STK39 as a hypertension susceptibility gene. Proc Natl Acad Sci U S A. 2009;106:226–31.

    Article  CAS  PubMed  Google Scholar 

  126. Diao Z, Asico LD, Villar VA, Zheng X, Cuevas S, Armando I, et al. Increased renal oxidative stress in salt-sensitive human GRK4γ486V transgenic mice. Free Radic Biol Med. 2017;106:80–90. doi:10.1016/j.freeradbiomed.2017.02.021.

    Article  CAS  PubMed  Google Scholar 

  127. Cabrera CP, Ng FL, Warren HR, Barnes MR, Munroe PB, Caulfield MJ. Exploring hypertension genome-wide association studies findings and impact on pathophysiology, pathways, and pharmacogenetics. Wiley Interdiscip Rev Syst Biol Med. 2015;7(2):73–90.

    Article  CAS  PubMed  Google Scholar 

  128. Padmanabhan S, Caulfield M, Dominiczak AF. Genetic and molecular aspects of hypertension. Circ Res. 2015;116(6):937–59. doi:10.1161/CIRCRESAHA.116.303647.

    Article  CAS  PubMed  Google Scholar 

  129. Huan T, Esko T, Peters MJ, Pilling LC, Schramm K, Schurmann C, et al. International consortium for blood pressure GWAS (ICBP). A meta-analysis of gene expression signatures of blood pressure and hypertension. PLoS Genet. 2015;11(3):e1005035. doi:10.1371/journal.pgen.1005035.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Staessen JA, Kuznetsova T, Zhang H, Maillard M, Bochud M, Hasenkaml S, et al. Blood pressure and renal sodium handling in relation to genetic variation in the DRD1 promoter and GRK4. Hypertension. 2008;51:1643–50.

    Article  CAS  PubMed  Google Scholar 

  131. Martinez Cantarin MP, Ertel A, Deloach S, Fortina P, Scott K, Burns TL, et al. Variants in genes involved in functional pathways associated with hypertension in African Americans. Clin Transl Sci. 2010;3(6):279–86. doi:10.1111/j.1752-8062.2010.00242.x.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav. 2014;13:69–86.

    Article  CAS  PubMed  Google Scholar 

  133. Banday AA, Lau YS, Lokhandwala MF. Oxidative stress causes renal dopamine D1 receptor dysfunction and salt-sensitive hypertension in Sprague-Dawley rats. Hypertension. 2008;51:367–75.

    Article  CAS  PubMed  Google Scholar 

  134. Doyle K, Fitzpatrick FA. Redox signaling, alkylation (carbonylation) of conserved cysteines inactivates class I histone deacetylases 1, 2, and 3 and antagonizes their transcriptional repressor function. J Biol Chem. 2010;285:17417–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. • Gildea JJ, Carlson JM, Schoeffel CD, Carey RM, Felder RA. Urinary exosome miRNome analysis and its applications to salt sensitivity of blood pressure. Clin Biochem. 2013;46:1131–4. This study discusses new methods of diagnosing salt sensitivity through increased miR-124 expression in urinary exosomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Walesky C, Edwards G, Borude P, Gunewardena S, O'Neil M, Yoo B, et al. Hepatocyte nuclear factor 4 alpha deletion promotes diethylnitrosamine-induced hepatocellular carcinoma in rodents. Hepatology. 2013;57:2480–90.

  137. Heideman MR, Wilting RH. Yanover, Velds a, de Jong J, Kerkhoven RM, et al. dosage-dependent tumor suppression by histone deacetylases 1 and 2 through regulation of c-Myc collaborating genes and p53 function. Blood. 2013;121:2038–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Stocks T, Van Hemelrijck M, Manjer J, Bjørge T, Ulmer H, Hallmans G, et al. Blood pressure and risk of cancer incidence and mortality in the metabolic syndrome and cancer project. Hypertension. 2012;59:802–10.

    Article  CAS  PubMed  Google Scholar 

  139. D'Elia L, Galletti F, Strazzullo P. Dietary salt intake and risk of gastric cancer. Cancer Treat Res. 2014;159:83–95.

    Article  PubMed  CAS  Google Scholar 

  140. Hasenkamp S, Telgmann R, Staessen JA, Hagedorn C, Dördelmann C, Bek M, et al. Characterization and functional analyses of the human G protein-coupled receptor kinase 4 gene promoter. Hypertension. 2008;52:737–46.

    Article  CAS  PubMed  Google Scholar 

  141. Escano CS, Armando I, Wang X, Asico LD, Pascua A, Yang Y, et al. Renal dopaminergic defect in C57Bl/6J mice. Am J Phys Regul Integr Comp Phys. 2009;297:R1660–9.

    CAS  Google Scholar 

  142. Zhao Y, Vanhoutte PM, Leung SW. α1-Adrenoceptor activation of PKC-ε causes heterologous desensitization of thromboxane receptors in the aorta of spontaneously hypertensive rats. Br J Pharmacol. 2015;172(14):3687–701.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Sanada H, Yatabe J, Midorikawa S, Katoh T, Hashimoto S, Watanabe T, et al. Amelioration of genetic hypertension by suppression of renal G protein-coupled receptor kinase type 4 expression. Hypertension. 2006;47(6):1131–9.

    Article  CAS  PubMed  Google Scholar 

  144. Rassler B. The renin-angiotensin system in the development of salt-sensitive hypertension in animal models and humans. Pharmaceuticals (Basel). 2010;3(4):940–60.

    Article  CAS  Google Scholar 

  145. Gildea JJ, Lahiff DT, Van Sciver RE, Weiss RS, Shah N, McGrath HE, et al. A linear relationship between the ex-vivo sodium mediated expression of two sodium regulatory pathways as a surrogate marker of salt sensitivity of blood pressure in exfoliated human renal proximal tubule cells: the virtual renal biopsy. Clin Chim Acta. 2013;421:236–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Yang J, Villar VA, Armando I, Jose PA, Zeng C. G protein-coupled receptor kinases: crucial regulators of blood pressure. J Am Heart Assoc. 2016 Jul 7;5(7). pii: e003519. doi:10.1161/JAHA.116.003519.

  147. Sun CJ, Zhang WY. G protein-coupled receptor kinase 4 gene variants are not associated with preeclampsia in Northern Han Chinese. Hypertens Res. 2010;33(7):683–7. doi:10.1038/hr.2010.57.

    Article  CAS  PubMed  Google Scholar 

  148. Wolski H, Marek P, Drews K, Barlik M, Kurzawińska G, Oarowski M, et al. DRD1 and DRD4 dopamine receptors in the etiology of preeclampsia. Ginekol Pol. 2015;86(9):672–7.

    Article  PubMed  Google Scholar 

  149. •• Bhatnagar V, O'Connor DT, Brophy VH, Schork NJ, Richard E, Salem RM, et al. AASK Study Investigators. G-protein-coupled receptor kinase 4 polymorphisms and blood pressure response to metoprolol among African Americans: sex-specificity and interactions. Am J Hypertens. 2009;22(3):332–8. doi:10.1038/ajh.2008.341. AASK study discusses the GRK4 polymorphisms among African Americans and response to beta blockers.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. •• Vandell AG, Lobmeyer MT, Gawronski BE, Langaee TY, Gong Y, Gums JG, et al. G protein receptor kinase 4 polymorphisms: β-blocker pharmacogenetics and treatment-related outcomes in hypertension. Hypertension. 2012;60(4):957–64. doi:10.1161/HYPERTENSIONAHA.112.198721. This study discusses the different GRK4 polymorphisms and response to beta blockers and risk for adverse outcomes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Wagner F, Malice MP, Wiegert E, McGrath HE, Gildea J, Mitta S, et al. A comparison of the natriuretic and kaliuretic effects of cicletanine and hydrochlorothiazide in prehypertensive and hypertensive humans. J Hypertens. 2012;30(4):819–27. doi:10.1097/HJH.0b013e32835022a8.

    Article  CAS  PubMed  Google Scholar 

  152. Wang Z, Asico LD, Escano CS, Felder RA, Jose PA. Human G protein-coupled receptor kinase type 4 γ (hGRK4γ) wild-type prevents salt sensitivity while its variant, hGatatRK4γ486V, promotes salt sensitivity in transgenic mice: role of genetic background. Hypertension. 2006;48:e27.

    Article  CAS  Google Scholar 

  153. Sanada H, Yatabe J, Yatabe MS, Yokokawa H, Williams S, Bartlett J, et al. G protein coupled receptor type 4 gene variants and response to antihypertensive medication. Circulation. 2009;120:S1087.

    Google Scholar 

  154. Litwin M, Niemirska A, Sladowska J, Antoniewicz J, Daszkowska J, Wierzbicka A, et al. Left ventricular hypertrophy and arterial wall thickening in children with essential hypertension. Pediatr Nephrol. 2006;21:811–9.

    Article  PubMed  Google Scholar 

  155. Flynn JT. Pediatric hypertension update. Curr Opin Nephrol Hypertens. 2010;19:292–7.

    Article  PubMed  Google Scholar 

  156. • Kupferman JC, Lande MB, Adams HR, Pavlakis SG. Primary hypertension and neurocognitive and executive functioning in school-age children. Pediatr Nephrol. 2013;28:401–8. doi:10.1007/s00467-012-2215-8. Premature target organ damage (brain) of pediatric hypertension.

    Article  PubMed  Google Scholar 

  157. Litwin M, Niemirska A. Intima-media thickness measurements in children with cardiovascular risk factors. Pediatr Nephrol. 2009;24:707–19.

    Article  PubMed  Google Scholar 

  158. • Folkow B. Structure and function of the arteries in hypertension. Am Heart J. 1987;114:938–48. Folkow’s hypothesis that hypertension thickens medial layer of blood vessels which begets hypertension.

    Article  CAS  PubMed  Google Scholar 

  159. Srinivasan SR, Myers L, Berenson GS. Changes in metabolic syndrome variables since childhood in prehypertensive and hypertensive subjects: the Bogalusa heart study. Hypertension. 2006;48:33–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported, in part, by grants from the National Institutes of Health (R37 HL023081, R01DK039308, R01HL092196, P01HL068686, and P01HL074940 [PAJose]) and a minigrant from the National Kidney Foundation of Maryland (V.A. Villar).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andrew C. Tiu.

Ethics declarations

Conflict of Interest

The authors declare no conflicts of interest relevant to this manuscript.

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.

Additional information

This article is part of the Topical Collection on Therapeutic Trials

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tiu, A.C., Bishop, M.D., Asico, L.D. et al. Primary Pediatric Hypertension: Current Understanding and Emerging Concepts. Curr Hypertens Rep 19, 70 (2017). https://doi.org/10.1007/s11906-017-0768-4

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11906-017-0768-4

Keywords

Navigation