Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Aug 6.
Published in final edited form as: Pharmacogenomics J. 2019 Feb 6;19(4):315–336. doi: 10.1038/s41397-019-0082-4

Genetic Polymorphisms Associated with Reactive Oxygen Species and Blood Pressure Regulation

Santiago Cuevas 1, Van Anthony M Villar 2, Pedro A Jose 2
PMCID: PMC6650341  NIHMSID: NIHMS1517349  PMID: 30723314

Abstract

Hypertension is the most prevalent cause of cardiovascular disease and kidney failure but only about 50% of patients achieve adequate blood pressure control, in part, due to inter-individual genetic variations in the response to antihypertensive medication. Significant strides have been made toward the understanding of the role of reactive oxygen species (ROS) in the regulation of the cardiovascular system. However, the role of ROS in human hypertension is still unclear. Polymorphisms of some genes involved in the regulation of ROS production are associated with hypertension, suggesting their potential influence on blood pressure control and response to antihypertensive medication. This review provides an update on the genes associated with the regulation of ROS production in hypertension and discusses the controversies on the use of antioxidants in the treatment of hypertension, including the antioxidant effects of antihypertensive drugs.

Keywords: Hypertension, Oxidative Stress, Reactive Oxygen Species, Pharmacogenomics, antioxidant treatment

Introduction

Hypertension is the most prevalent cause of cardiovascular disease and kidney failure1, but the prevention and treatment of hypertension are still a challenge24. According to the 2017 High Blood Pressure Clinical Practice Guidelines, in adults (≥20 years of age), a “doctor’s office” reading of 120-129 mm Hg for systolic blood pressure (SBP) even with less than 80 mm Hg for diastolic blood pressure (DBP), is considered as elevated BP2,3. An SBP of 130-139 mm Hg or a DBP of 80-89 is now considered as Stage 1 hypertension, while the previous definition of hypertension ≥140 SBP or ≥90 DBP is now considered as Stage 2 hypertension. In the general population, only about 50% of treated patients achieve adequate blood pressure control2,4. The poor efficacy of hypertension treatment and the inter-individual variations in the response to antihypertensive medications have many causes, including non-compliance, but genetic variations could be important contributory factors4,5.

Reactive oxygen species (ROS) are inevitable by-products of aerobic existence6. Disturbance in the normal redox state of cells, either due to the overproduction of ROS or low production of antioxidants, can lead to oxidative stress and specific types of oxygen radicals, such as superoxide anion, H2O2 and hydroxyl radical, may damage all components of the cell, including proteins, lipids, and DNA7,8.

The deleterious effects of ROS and their role in the pathogenesis of hypertension have been extensively demonstrated in experimental models810, but the benefits of antioxidant drug treatment in human hypertension are not clear8,1012. This may be related to the fact that ROS are not always harmful; ROS are able to regulate the activity of cellular signaling pathways such as Ca2+ signaling13,14, and are involved in the regulation of several cells functions such as phenotypic modulation, migration and adhesion, vascular tone, apoptosis and sodium reabsorption between others1318. The oxidative environment in the cell influences gene transcription, post-transcription, translation, and post-translation of proteins. Modifications of the oxidative status may eventually regulate the expression and activity of many proteins such as nuclear factor-kappaB, Nrf2, p38 mitogen-activated protein kinase, NH(2)-terminal Jun kinases/stress-activated protein kinases, hexosamines, and others13,1927 evidencing that ROS may be essential for the normal function of cells and biological systems.

Pharmacogenomics aims to individualize therapy based on the individual’s genetic profile. There are numerous endogenous oxidants and antioxidant proteins (Figure 1) in different organs, including the kidney, brain, and cardiovascular system, that keep a normal redox balance in the body. Genetic polymorphisms that affect the expression and activity of some of these pro-oxidant or antioxidant genes are associated with human hypertension (Table 1). These polymorphisms could influence the response to antihypertensive drugs, i.e., pharmacogenomics. This review provides an update on the genes associated with the regulation of ROS production in hypertension and discusses the controversies on the use of antioxidants in the treatment of hypertension, including the antioxidant effects of antihypertensive drugs.

Figure 1. Genes associated with redox balance regulation and Hypertension.

Figure 1.

The presence of the SNPs in pro-oxidant and antioxidant genes could increase the ROS production in the brain, cardiovascular system, heart, and kidney to induce hypertension.

Table 1.

Genes associated with hypertension and oxidative stress in humans and/or animal models of hypertension.

Genes associated with oxidative stress Associated with hypertension in animal models SNPs Associated with hypertension in humans
Polymorphisms of pro-oxidant genes

Activating transcription factor 1 (ATF1) rs11169571 [73]

Aminopeptidase A (APA) Aminopeptidase-A (stop), rs2290105 [84, 85]

Angiotensinogen (AGT) [37,76,77] −6G>A, −20A>C, −152A>G, −217G>A, rs5050 M235T (rs699) [7883]

Angiotensin II type 1 receptor (AGTR1) [86, 87] rs5186 (1166A>C) [84]
Arachidonate 15-lipoxygenase (ALOX1){351] 261R>Q [350]

Cycloxygenase-2 (COX2, PTGS2) −765G>C, rs2143417 [8890]

Cytochrome P450 [9194] CYP2C8*3, CYP 450 2J2(−50G>T), CYP3A5*3 (rs776746) (Japanese), CYP3Ar5*1 (European- and African-Americans), CYP4A11 (rs1126742), CYP17A (rs11191548) [91,96103,200,356358]

DNA-Binding Factor NFkB [104]

Endothelin I rs5335 (70C>G), rs5370 (198G>T) [105,359]

Hemojuvelin (HJV) rs16827043, rs7536827 [106]

Interleukin-6 (IL-6) [107] rs1800795 (−174G>C), rs1800796 (−572C>G) [108111]

Interleukin-17A (IL-17A) [112] rs2275913 (G>A) [113]

Iron regulatory protein (HFE) rs1799945 (63H>D) [114]

Leptin (LEP) [115,116] II/I tetra nucleotide repeat, rs799039(G2548A) [117,118]

Leptin receptor (LEPR) rs1137101 (223Q>R), rs1137100 (109K>R) [117119]

Myeloperoxidase (MPO) rs2333227(−463G>A) [120124]

NADPH oxidase p22phox (CYBA) [25,125,126] rs9932581 (930A>G), rs78935588 (640A>G) rs7195830 (49A>G), −675A>T rs4673 (242C>T), rs8854A, rs9932581G [127139]

NADPH oxidase 1 (NOX1) [140, 141]

NADPH oxidase 2 (NOX2) [142145]

NADPH oxidase 4 (NOX4) [147]

Neutrophil cytosol factor 2 (NCF2) rs12094228, rs16861188, and rs12066019 [148]

Nitric oxide synthase 3 (NOS3) [149] 894G>T (rs1799983) [150,151]

RAC1 rs6967221 [152]

Xanthine dehydrogenase/oxidase (XDH) rs11904439, rs148756340 [240]
Polymorphisms of antioxidant genes

Catalase (CAT) [153] rs769214 (−844G>A), rs1001179 (−262C>T), −20C>T, rs769217 [154,155,157]
[230,279]

Cystathione γ-lyase (CSE) [158160] rs482843 [161]

DJ-1 (PARK7) [162,163] [164]

Dopamine 1 receptor (DRD1) [32,165] (−48A>G, −94G>A rs1799914, rs4867798) [166170]

Dopamine 2 receptor (DRD2) [32,252] rs6276, rs6277, rs1800497 [171176]

Dopamine 3 receptor (DRD3) [178180] rs9880168 [169]

Dopamine 4 receptor (DRD4) −521C>T, DRD4 long allele [182184]

Dopamine 5 receptor (DRD5) [185188] No associations published


Fibroblast growth factor 5 (FGF5) rs16998073 [192,193,200]

Glutathione [197]

Glutathione peroxidase (GPX1, GPX3, GPX4)) [194] rs713041 (718C>T) rs3828599 [195,196]

Glutathione S-transferase Alpha 1 (GSTA1) GSTA1*B allele GSTA1*B allele +GSTM1 null [374]

Glutathione S-transferase Mu 1 (GSTM1) GSTM1 null
GSTM1 + GSTT1 null
[198,199,202]
[374]

Glutathione S-transferase Mu 3 (GSTM3) −63A>C [203]

Glutathione S-transferase Pi 1 (GSTP1) A313 [201]

Glutathione S-transferase Theta 1 (GSTT1) GSTT1 null [198]

Heme oxygenase-1 (HO-1) [204,205] <27 GT repeats rs9607267 [206]

Heme oxygenase-2 (HO-2) [207]

Kidney androgen-regulated protein (KAP) [208210]

Methylenetetrahydrofolate reductase (MTHFR) 677C>T [201,211]

Nuclear factor (erythroid-derived 2)-like 2, also known as NFE2L2 (NRF2) [163]

Paraoxonase 1 (PON1) 192Q>R, −108C>T [212,213]

Paraoxonase 2 (PON2) [146]

Peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-α) [214] 482G>S, 482G>S+1704A>G haplotype [215217]

Sestrin 2 (SESN2) [218]

Superoxide dismutase 1 (Cu-Zn SOD) [220226]

Superoxide dismutase 2 (Mn SOD) [219,227,228] [150]

Superoxide dismutase 3 (EC SOD) [189,231,232] rs13306703 + rs2536512 +/− rs1799895 [190,230]

Thioredoxin (TXN) rs2301241 (−793T>C) [230]

Thioredoxin interacting protein (TXNIP) [234]

Thioredoxin reductase (TXNRD2) [235]

Uncoupling protein 2 (UCP2) [236238] −866 G/A [239]

1. Mechanisms by which ROS regulate blood pressure

The role of oxidative stress in hypertension has been extensively studied and several mechanisms have been described by which ROS regulate blood pressure (Figure 1).

1.1. Endothelial damage:

ROS cause endothelial dysfunction in blood vessels, including renal afferent arterioles and enhance the renal arteriolar vasoconstrictor response to angiotensin II9,10,12,13,22. Some of the benefits of superoxide scavengers in hypertension are caused by enhancement of vasodilation and an increase in renal arterial perfusion23.

1.2. Stiffening of vessels:

Recent studies in humans have shown that aortic stiffening precedes the development of hypertension24. Mice with smooth muscle overexpression of p22phox, a component of NADPH oxidase, develop renal inflammation, fibrosis, and renal dysfunction, prior to the increase in blood pressure, supporting the notion that arterial stiffening induced by oxidative stress and inflammation causes hypertension25.

1.3. Glomerular damage:

Glomerulonephritis without renal insufficiency can be associated with hypertension26. ROS can produce glomerular injury by damaging the podocytes, as has been described in Dahl salt-sensitive hypertensive rats27. The antioxidant tempol reduces glomerular sclerosis and proteinuria in these animals, supporting a role of ROS in the glomerular injury in Dahl salt-sensitive rats28.

1.4. Renin-angiotensin system:

The development and progression of hypertension due to increased production of ROS have been related to renal vasoconstriction caused by an increase in renal afferent nerve activity and myogenic response and secretion of vasoconstrictor hormones, such as angiotensin II, endothelin-1, and thromboxane29. Angiotensin II increases ROS production, inflammation, and renal tubular ion and water transport, and decreases dopamine receptor expression and function, resulting in hypertension4,5,8,1517,2934 . Increasing oxidative stress is one mechanism by which angiotensin II causes renal dysfunction and tissue damage1,9,10,12,13,22,23,3138.

1.5. NaCl retention:

ROS can regulate ion transport1618,23,3564. Superoxide, produced by NADPH oxidase, enhances NaCl transport in the renal proximal tubule16,17,39,44,45,59, thick ascending limb of Henle41,46,48, and collecting duct18,47,48. The voltage-gated proton channel participates in the increased production of superoxide in the renal outer medulla of Dahl salt-sensitive rats46. It should be born in mind, however, that ROS can inhibit Na+/K+-ATPase and NHE3 activity in the renal proximal tubule48,5053.

1.6. Inflammation:

ROS activate pro-inflammatory transcription factors, such as NFκB and activator protein-1 and increase the expression of pro-inflammatory proteins65. ROS cause the activation, adhesion, and infiltration of inflammatory cells in tissues and organs, including the adipose tissue8,10,66. Immune cells, such as macrophages and granulocytes, release ROS to destroy engulfed bacterial or fungal pathogens and this could trigger oxidative stress67,68. Vascular stretch is associated with hypertension that could be related to an increase in ROS production and inflammation8,10,25,69,70.

1.7. Sympathetic nervous system:

Renal ROS induce sympathetic activation in renovascular hypertension71; chronic antioxidant treatment reduces blood pressure in hypertension characterized by sympathoexcitation and renal oxidative stress71. Oxidative stress in the brain, specifically in the cardiovascular regulating center, causes hypertension72.

2. Genes associated with oxidative stress and blood pressure regulation.

Table 1 lists the genes involved in redox balance that have been associated with hypertension. Table 1 also lists the single nucleotide polymorphisms (SNPs) that are associated with human hypertension, as well as the genes associated with oxidative stress and hypertension in animal models73252.

2.1. Effects of antihypertensive drugs on oxidative stress

Table 2 lists the antioxidant drugs shown to reduce blood pressure in humans and animal models of hypertension253328. These antioxidants have different mechanisms of action and various combinations may have synergistic effects on the regulation of blood pressure. For example, the antihypertensive effect of the combination of zinc sulfate, ascorbic acid, α-tocopherol, and β-carotene may be due to an increase in the bioavailability of NO324. By contrast, antioxidant drugs such as vitamin E, under certain conditions, can also increase the blood pressure in mice325. Vitamin E at doses greater than 150 IU daily increases the risk of all-cause mortality in humans326. The combination of vitamin C and polyphenols has also been reported to increase blood pressure variability229, and the antioxidant properties observed in vitro may not be observed in vivo328. The effect of chemicals on ROS production and blood pressure is complex and not easily predictable.

Table 2.

Antioxidant drugs that decrease blood pressure in humans with hypertension and animal models of hypertension.

Antioxidant Species Mechanism of action References
N-acetyl cysteine human, mouse, rat Direct antioxidant, precursor of cysteine reduced glutathione, breaks disulphides [243247]

Apocynin mouse, rat Prevents NADPH oxidase assembly [248252]

Allicin/Aliin/S-allylcysteine (garlic) human Reduces 8-hydroxy-2’deoxyguanosine, malondialdehyde, angiotensin II-generated ROS [253256]

L-arginine human, mouse, rat Substrate for NO production [257259]

Bardoxolone human, mouse, rat Nrf2 inducer [260,261]

L-carnitine human, mouse, rat Key compound in the transport of long-chain fatty acids into mitochondria for β-oxidation [262,264266]

Catechins
Black tea (theaflavin-polyphenol) rat Stimulates NO and H2S production, Decreases endothelin-1 and angiotensin II [267]
Green tea (polyphenols) human Antioxidant, inhibits catechol-O-methyl transferase, NO release [267269]

Coenzyme Q10 human, mouse, rat Reduces mitochondrial superoxide production by increasing the efficiency of electron transfer from Complexes I and II down the mitochondrial electron transport chain [270273]

Curcurmin human, mouse, rat A herbal supplement used as a food additive with antioxidant properties at low concentrations, induces HO-1 [274277]

Hemin mouse, rat HO-1 inducer (can induce mitochondrial dysfunction) [278280]

Hesperidin human, rat Free radical scavenger and enhancer of antioxidant pathways via ERK/Nrf2, inhibits RAS [281283]

α-Lipoic acid human, mouse, rat Free radical scavenger and activator of anti-oxidant recycling [284286]

Melatonin human, rat Free radical scavenger and up-regulator of antioxidant enzymes. [287290]

Quercetin human, mouse, rat Free radical scavenger [291294, 419, 420]

Resveratrol human, mouse, rat Activator of sirtuins and PGC-1α, involved in stress response, and Nrf2 [295, 296, 421]

Tempol human, mouse, rat Redox-cycling nitroxide and SOD mimetic [9,16,28,29,34,297,422]

Troxerutin mouse, rat Flavonoid (hydroxyethylrutoside) with antioxidant properties [298,299]

Vitamin C human, mouse, rat Free radical scavenger [11,300306]

Vitamin D human, mouse, rat Inhibits iron-dependent liposomal lipid peroxidation [10,307319]

Vitamin E human, mouse, rat Free radical scavenger, impairs ROS signaling 11,303,320323]

2.2. Pharmacogenomics of antioxidant drugs

Increased ROS production is involved in the pathogenesis of many1,5,817,22,23,25,2844,48,57,60,6264,68,71,72,86,89,92,104,138,140154,162,163,173,189,197,210,214,218,219,221233,236244,246252,262,263.270273,278280,295,305.329331, but not all cases232234 of hypertension. Deletion of the gene that encodes thioredoxin reductase 2 increases ROS production but blood pressure is actually decreased235. Nevertheless, genetic polymorphisms in pro-oxidant or antioxidant genes may affect the redox balance in the kidney, cardiovascular system, and brain (Figure 1), among others. Therefore, genetic polymorphisms may be involved in the inter-individual variability of the effects of antihypertensive medications. Many genes involved in ROS production and their polymorphisms associated with hypertension have been identified (Table 1).

2.2.1. Polymorphisms in Pro-oxidant Genes

Angiotensinogen (AGT) is converted to angiotensin I by renin and angiotensin 1 to angiotensin II by angiotensin converting enzyme (ACE); angiotensin II induces oxidative stress by stimulation of NADPH oxidase activity15,22,23,2931,36,37,42,47,48,56,57. Polymorphisms in AGT are associated with hypertension in humans7883; a haplotype of human AGT gene containing −217A or −6G increases blood pressure in transgenic mice76,77.

The NADPH oxidase (NOX) family has seven members which are classified into three groups: group 1 is comprised of NOX1, NOX2, NOX3, NOX4; group 2 has NOX5 as the only member, and group 3 is comprised of DOUX1 and DOUX248,332. Increased NOX activity is implicated in many disease states, including hypertension and renal disease5,10,15,23,25,2932,36,47,48,56,57,125147,332344.

p22phox (CYBA, cytochrome B-245 alpha chain) is a membrane-associated protein that plays a crucial role in the activation of NOX1, NOX2, NOX448, and NOX5333. Mutations of CYBA lead to autosomal recessive forms of chronic granulomatous disease334. Germline deletion of CYBA in mice335 or silencing of CYBA in Sprague-Dawley rats does not affect basal blood pressure but ameliorates angiotensin II-induced hypertension125,126. However, smooth muscle-specific overexpression of p22phox in mice increases blood pressure that is normalized in the offspring of dams crossed with Rag1−/− mice25. Polymorphisms in the CYBA promoter in the spontaneously hypertensive rat (SHR) increase the gene expression of CYBA336. Several polymorphisms of CYBA that could affect the production of ROS have also been reported in humans,334,. Some other CYBA gene variants are associated with decreased NOX2-dependent ROS generation but their association with blood pressure has not been studied128. Other CYBA gene variants are associated with increased ROS production and hypertension in several ethnic groups129,130,131,134136,337. However, although CYBA 242C>T is associated with endothelial dysfunction, it is not associated with hypertension in an Asian-Indian population338. A meta-analysis found no association of CYBA 242C>T with hypertension134. CYBA 242C>T may be protective of coronary artery disease in an Asian population132 but increases the risk of diabetes mellitus133. In an Asian-Indian population, the haplotypes rs8854A/rs9932581G/rs4873C and rs8854G/rs9932581G/rs4873C are positively associated with increased blood pressure and oxidative stress while the haplotype rs8854G/rs9932581A/rs4873T is inversely correlated with blood pressure and oxidative stress339.

NOX5 gene, which is present in humans but not rodents, is expressed to a greater degree than the other isoforms in renal proximal tubule cells from hypertensive humans340. Certain NOX5 SNPs have been reported to be associated with decreased (NOX5 77M>K) activity and ROS production341. However, mice with podocyte-specific human NOX5 expression develop renal disease and high blood pressure342. Genes that interact with NOXs have polymorphisms that may also be associated with increased ROS production and hypertension. For example, a polymorphism in the 3’UTR (rs11169571 [T>C] of the activating transcription factor 1 [ATF]) may be involved in essential hypertension by induction of NOX1 and increase in ROS production343.

The minor T allele of rs6967221 in RAC1, one of the cytosolic components of NOX1, NOX2, and NOX3, is associated with a decreased systolic blood pressure response to high sodium intake152.

Endothelin-1 (ET-1) is a potent vasoconstrictor which can increase ROS production by stimulation of NADPH oxidase activity344. A polymorphism of type A endothelin-1 receptor (rs5335, 70C>G) is associated with increased night-time blood pressure105. Polymorphism at rs9349379 in PHACTR1, a distal regulator of EDN1, is associated with a lower risk of hypertension345.

Myeloperoxidase (MPO) produces hypochlorous acid (HOCl) and chloride anion (Cl−) (or equivalent) from H2O2 during the neutrophil’s respiratory burst. MPO released during chronic inflammation produces tissue damage and high MPO levels may exacerbate diseases associated with atherosclerosis. However, MPO-deficient mice unexpectedly have increased atherosclerosis, relative to their wild-type littermates346, indicating that the role of MPO in cardiovascular disease is still unclear or that this murine model may not reflect human disease. The −463G>A polymorphism located in the promoter region of the MPO gene has been associated with hypertensive nephrosclerosis in patients on dialysis120 and hypertension with or without carotid atherosclerosis in Chinese121,122. However, this polymorphism has been associated with a decreased risk of hypertension in Russian females123.

Xanthine dehydrogenase (XDH), aka xanthine oxidoreductase (XOR) and xanthine oxidase (XO) are interconvertible single gene products. XDH is the primary form but is converted to XO irreversibly by proteolysis or reversibly by oxidation of Cys residues. XO catalyzes hypoxanthine or xanthine to form hydrogen peroxide and uric acid while XDH produces NADH347. In the blood, XDH exists mainly as XO242. XOD is extensively expressed in body organs, such as the liver, muscle, brain, and kidney348. XDH-mediated increase in ROS has been described in salt-sensitive hypertension and glucocorticoid induced hypertension242. In a Spanish cohort, −337G>A and 565+64T>C and their haplotypes were found to be associated with higher systolic and diastolic blood pressures and malondialdehyde242. The variation in uric acid production, as related to polymorphisms of XDH, increases the risk of hypertension240.

Lipoxygenases catalyze the dioxygenation of polyunsaturated fatty acids to their corresponding hydroperoxy derivatives. Arachidonate 15-lipoxygenase (ALOX15) gene rs2664593 has been reported to be associated with air pollution and increased left ventricular mass349. A nonsynonymous polymorphism in ALOX12, 261R>Q, has been reported to be associated with essential hypertension and urinary levels of 12-hydroxyeicosatetraenoic acid (12(S)-HETE)350. Mice lacking macrophage 12/15 lipoxygenase are resistant to L-NAME and DOCA-salt hypertension351.

Cyclooxygenase-2 (COX2, PTGS2) can produce ROS, which can increase cyclooxygenase expression and activity92. −765GC+CC genotypes of PTGS2 are inconsistently associated with chronic obstructive pulmonary disease that could be related to increased ROS production90,91. PTGS2 SNPs have been associated with increased high blood pressure in humans352. Germline deletion of Cox-2 in mice increases blood pressure353.

The mitochondrion, which is one of the most important sources of ROS, has been extensively associated with oxidative stress and hypertension8,12,13,145,228. ROS-induced hypertension could involve the mitochondria in the brain72 and in the kidney354,355,356. Cytochrome P450 genes are important sources of ROS in the mitochondria, endoplasmic reticulum, and plasma membrane. P450 proteins are a family of hemoproteins that catalyze the oxygenation of a wide variety of compounds and, in general, is the terminal oxidase enzyme in the electron transfer chain in the mitochondria95. The efficiency of electron transfer depends on many conditions. For example, SNPs in the gene encoding Cytochrome P450 affect the regulation of ROS production and the redox balance357,358. SNPs in the cytochrome P450 gene family have also been associated with high blood pressure in several different populations 96,98,360,361 but protective in a North Americans 97. CYP-epoxygenase decreases renal sodium transport, in part, by inhibition of ENaC activity in the cortical collecting duct362. CYP17A1 (rs11191548) is associated with increased left ventricular mass in patients with hypertension and preserved left ventricular ejection fraction363.

2.2.2. Polymorphisms of antioxidant genes

Oxidative stress can occur not only from an increase in pro-oxidant activity but also from impaired antioxidant activity. SNPs of genes that decrease antioxidant gene function/expression could induce oxidative stress and increase blood pressure. Our group and others have provided evidence for the importance of the antioxidant properties of dopamine receptors in the kidney in the regulation of renal sodium transport and blood pressure, as well as dopamine receptor-mediated non-renal mechanisms in the regulation of blood pressure5,16,32,3840,62,64,165,170,173,174,178188,364367. Germline deletion of the DRD2 results in oxidative stress dependent-hypertension252. DJ-1 (Park7) and paraoxonase 2 (PON2)146,163 are involved in the antioxidant properties of the D2R in the kidney. Polymorphisms associated with deficiency of DRD2 expression are associated with essential hypertension in different populations171,172,174. PON1 SNPs (e.g., −108C>T, 192Q>R) are risk factors for endothelial dysfunction and hypertension212,213. Genetic depletion of DJ-1, a mitochondrial antioxidant368, results in renal oxidative stress and high blood pressure in mice163. Dysfunction of mitochondrial proteins that decrease ROS production (e.g., SOD2, UCP2 [vide infra]) may be involved in the target-organ damage associated with hypertension369.

Polymorphisms in uncoupling protein 2 (UCP2), a mitochondrial gene with antioxidant properties, are associated with an increased risk for diabetic kidney disease370. In addition, a common human polymorphism of the UCP2 gene, −866 G>A, has been associated in hypertension239.

Glutathione (GSH) is another antioxidant enzyme that plays a role in blood pressure regulation371. Glutathione S-transferases (GSTs) catalyze the conjugation of the reduced form of GSH. The GST superfamily constitutes up to 10% of the cytosolic protein in some mammalian organs372. Low blood level of GST-π concentration is predictive of the time of the onset of stroke373. The GSTA1*B allele is considered as a genetic risk factor for hypertension in Japanese374. The association between the GSTT1 null and hypertension was reported in Italian women but not men198 and GSTM1 null genotype with hypertension in Korean men and women199. The GST P1b-1b genotype causes prolonged exposure to ROS and increased risk of pre-eclampsia375; GSTP1 313A>G with preeclampsia in Maya-Mestizo women201. However, a meta-analysis showed no association of GSTM1 and GSTT1 polymorphisms and the risk of hypertension376.

Glutathione peroxidases (GPXs) are important in the reduction of lipid hydroperoxides and H2O2 to water; GPX4 rs713041 (718T>C) may be a predictor of cerebral stroke in hypertensive Russians196. GPX3 s3828599 (T>C) is associated with hypertension in Han Chinese195.

Heme oxygenase catalyzes the degradation of heme, resulting in the formation of iron, carbon monoxide, and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase188. HO-1 short repeats (<25) are associated with lower risk of cardiovascular disease; HO-1 short repeats are associated with increased HO-1 activity206.

Extracellular superoxide dismutase (EC-SOD, aka SOD3, Cu-Zn SOD) protects the tissues from oxidative stress by converting the toxic superoxide anion into less toxic hydrogen peroxide (H2O2)810,12,13,222,224,231233,377. The T-A or T-A-C haplotype, rs13306703 and rs2536512 with or without 17998895 in SOD3 gene increases the risk for essential hypertension in a Japanese population190. By contrast, 172G>A (rs2536512) polymorphism, by itself, is associated with a decreased risk for hypertension in Spaniards230 but is not associated with hypertension in other populations233. Germline global deletion of SOD3 in mice causes oxidative stress and hypertension189. However, an earlier and later study by others did not find SOD3 knockout mice to be hypertensive but found them to have increased hypertensinogenic response to NO inhibition or angiotensin II infusion231,232.

Catalase (CAT) catalyzes the conversion of H2O2 to water and oxygen. SNPs in the CAT gene promoter region, CAT-844 AA and CAT-262 CT or TT, have been associated with essential hypertension among Chinese157, smoking Russians154, Greeks155, but not African-Americans and Caucasians156. However, CAT haplotype [−844G,−89A,−20T] relative to the CAT haplotype [−844A,−89T,−20C] was predictive of a decrease in diastolic blood pressure after bariatric surgery in a French population378. In individuals with low-level lead exposure, CAT rs769217, C>T, is associated with increased blood markers of oxidative stress and hypertension379. By contrast, CAT rs1049982, −20 C>T, is associated with lower blood pressure230. In individuals with a family history of hypertension, 20-35% of the variation of plasma hydrogen peroxide may be due to genetic factors380, 381.

The transcriptional coactivator peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 α) is an important regulator of energy control382 and is a master regulator of manganese SOD2 and UCP-2, both of which are mitochondrial proteins with antioxidant properties383. Polymorphisms in PGC-1α gene have been associated with hypertension in several studies, in males with Gly482Ser+A1704G haplotype, but the 482SS is protective of hypertension in Caucasian males in two studies and females in one study215217.

These aforementioned studies show that SNPs of genes involved in redox balance are involved in blood pressure regulation.

3. Treatment of oxidative stress in humans with hypertension

Despite the numerous studies demonstrating a role of oxidative stress in cardiovascular diseases and the beneficial effects of antioxidants in the treatment of hypertension in animal models (Table 2), it has been difficult to demonstrate a role of oxidative stress in the pathogenesis and treatment of hypertension in humans305,384. Indeed, oxidative stress may be the consequence and not the cause of hypertension in humans385.

Several antioxidant drugs, such as vitamin C, vitamin D, vitamin E, and bardoxolone alone or in combination302307,310312,314317,319,320,324,384,386 with other antioxidants have been shown to prevent the deleterious effects of oxidative stress or hypoxia in different cardiovascular and renal diseases, including hypertension but some with undesirable side effects308,309,318,325,326,327,387,388. A meta-analysis in 135,967 participants in 19 clinical trials showed that high doses of vitamin E increased mortality387. The authors of a more recent meta-analysis concluded that supplements with vitamin E decreased cardiovascular mortality risk and folic acid decreased the risk for cardiovascular disease, while β-carotene, eicosapentanoic acid, magnesium, selenium, vitamins D and K, and zinc did not show significant risk reduction of cause-specific death or cardiovascular disease388. Thiosulfate, a hydrogen sulfide donor, which can decrease oxidative stress, has been reported to improve vascular endothelial function in hypertensive humans389,390. Bardoxolone, a Nrf2 agonist that increases the expression of several antioxidants, was initially shown to improve renal function in humans with advanced chronic kidney disease and type 2 diabetes391,. However, it was withdrawn from further clinical trials because of serious adverse events, including heart failure and cardiovascular events, and mortality392,393.

Antioxidant treatment with ascorbic acid was initially shown to lower blood pressure in a limited number of patients with hypertension394,395. However, larger studies have not found a clear beneficial effect of antioxidant vitamins on the development or control of blood pressure396,397. The combination of ascorbic acid and polyphenols actually resulted in a higher blood pressure variation327. Therefore, there is insufficient evidence to support the use of dietary supplements in the primary prevention of cardiovascular diseases388. However, the period of treatment and doses398 could be crucial in the beneficial or deleterious effects of antioxidant therapy. What is evident from these published data is that the effect of the ROS on the cardiovascular system is more complex than expected and innovative approaches must be formulated to resolve these discrepant results.

3.1. Antioxidant effect of antihypertensive drugs

The ability of some angiotensin II type 1 receptor blockers305,399 and ACE inhibitors305,400 to reduce ROS production and oxidative stress is well known. The classical renin-angiotensin system increases ROS production22,23,31,3437,56,57,64 and thus, the beneficial effects of some of the antihypertensive drugs may be due to their ability to inhibit NADPH activity. The sulfhydrylated ACE inhibitors (e.g., captopril, epicaptopril, and S-zofenopril but not enalaprilat, perindoprilat, or quinaprilat401,402) contain a thiol radical that per se has antioxidant properties and may prefer to scavenge general radicals rather than superoxide radicals400,401. Although, the antioxidant effect of sulfhydrylated ACE inhibitors has been ascribed to the thiol group, the vasodilatory effect of S-zofenopril may be due to hydrogen sulfide403. The antioxidant effects of other antihypertensive drugs, such as β adrenoceptor blockers305,404,405 and calcium channel blockers305,406 have been reported, as well. Hypertension and oxidative stress associated with chronic ethanol intake can be prevented by the β-adrenoceptor blocker, nebivolol407. Therefore, part of the beneficial effects of some antihypertensive drugs may due to their ability to decrease ROS production. However, a novel angiotensin II type 1 receptor blocker has been reported to induce oxidative stress in a hepatocellular cell line HepG2408.

These disparate effects of anti-hypertensive drugs on ROS production and blood pressure regulation may be related to the fact that, as aforementioned, ROS have beneficial effects on cell function10,1321. Anti-oxidants at high concentrations may have pro-oxidant effects409 and the excessive antioxidation could have deleterious consequences. For example, a small but continuous production of ROS expression during physical exercise enhances antioxidant defenses and induces the expression of antioxidant enzymes; vitamin C supplementation decreases the endurance capacity in humans and rats410 and diminishes some of the increased skeletal muscle adaptations following acute exercise411. While physiological doses of anti-oxidants may be beneficial, excessive antioxidation could have deleterious consequences because the “remodeling” of skeletal muscles with exercise is dependent on reactive oxygen and nitrogen signaling412. The duration of the antioxidant effects may also be transient. For example, the biomarkers of oxidative DNA damage were attenuated by daily consumption of blueberries for 4 weeks in pre- and stage 1-hypertensive postmenopausal women, however, these effects were not found after 8 weeks413.

Increased production of mitochondrial ROS plays a role in the pathogenesis of diabetic nephropathy414 and hypertension355. However, ROS produced by NOX4 can induce endothelial angiogenesis and protect against chronic cardiac overload415. Moreover, diabetic complications are associated with a decrease in mitochondrial ROS production but may help in the preservation of renal glomerular function during hyperglycemia 416,417. Therefore, “normal” physiological levels of mitochondrial superoxide are important for healthy mitochondrial function418.

The amount of ROS formed, type of ROS formed, i.e., superoxide versus H2O2, source, duration, and their subcellular locations may be determinants on the consequences of ROS production on cell function. It is universally accepted that a redox imbalance induced by an excessive and uncontrolled ROS production could have deleterious consequences on blood pressure regulation. However, the excessive intake or expression of antioxidants could also have delirious consequences on the cardiovascular system.

4. Conclusion

Several genetic polymorphisms that affect pro-oxidant and antioxidant systems, directly or indirectly, are associated with hypertension. Antioxidants can reduce the blood pressure in humans and animal models of hypertension. Antihypertensive drugs can also have anti-oxidant effects. However, an indiscriminate decrease in ROS production can have deleterious consequences. ROS are involved in the regulation of essential cellular processes. Thus, the long-term administration of drugs with antioxidant properties may impair vital cellular function, resulting in undesirable side effects. Studies are needed to elucidate the role of pharmacogenomics in redox balance in the treatment of hypertension.

Acknowledgments

Funding:

These studies were supported in part by grants from the NIH, 5P01HL074940-11, 7R01DK039308-31, 5P01 HL068686-15, 5R01HL023081-35, and 2R01HL092196-10.

Footnotes

Conflict(s) of Interest/Disclosure(s); None

References:

  • 1.Oparil S, Acelajado MC, Bakris GL, Berlowitz DR, Cífková R, Dominiczak AF et al. Hypertension. Nat Rev Dis Primers. 2018; 4 :18014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Muntner P, Carey RM, Gidding S, Jones DW, Taler SJ, Wright JT Jr. Whelton PK. Potential U.S. Population Impact of the 2017 ACC/AHA High Blood Pressure Guideline. J Am Coll Cardiol. 2018; 71: 109–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Greenland P, Peterson E. The New 2017 ACC/AHA Guidelines “Up the Pressure” on Diagnosis and Treatment of Hypertension. JAMA. 2017; 318: 2083–2084. [DOI] [PubMed] [Google Scholar]
  • 4.Cooper-DeHoff RM, Johnson JA. Hypertension pharmacogenomics: in search of personalized treatment approaches. Nat Rev Nephrol 2016; 12: 110–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Armando I, Villar VA, Jose PA. Genomics and pharmacogenomics of salt-sensitive hypertension. Curr Hypertens Rev 2015; 11: 49–56. [PubMed] [Google Scholar]
  • 6.Holterman CE, Thibodeau JF, Kennedy CR. NADPH oxidase 5 and renal disease. Curr Opin Nephrol Hypertens 2015; 24, 81–87. [DOI] [PubMed] [Google Scholar]
  • 7.Weidinger A, Kozlov AV. Biological activities of reactive oxygen and nitrogen species: Oxidative stress versus signal transduction. Biomolecules 2015; 15: 472–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Loperena R, Harrison DG. Oxidative stress and hypertensive diseases. Med Clin North Am 2017; 101: 169–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wilcox CS. Asymmetric dimethylarginine and reactive oxygen species: unwelcome twin visitors to the cardiovascular and kidney disease tables. Hypertension 2012; 59: 375–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sorriento D, De Luca N, Trimarco B, et al. The Antioxidant Therapy: New Insights in the Treatment of Hypertension. Front Physiol. 2018; 9: 258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Roberts JM, Myatt L, Spong CY et al. Eunice Kennedy Shriver National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Vitamins C and E to prevent complications of pregnancy-associated hypertension. N Engl J Med 2010; 362: 1282–1291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Montezano AC, Dulak-Lis M, Tsiropoulou S et al. Oxidative stress and human hypertension: vascular mechanisms, biomarkers, and novel therapies. Can J Cardiol 2015; 31: 631–641. [DOI] [PubMed] [Google Scholar]
  • 13.Brown DI, Griendling KK. Regulation of signal transduction by reactive oxygen species in the cardiovascular system. Circ Res 2015; 116: 531–549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ma R, Chaudhari S, Li W. Canonical transient receptor potential 6 channel: A new target of reactive oxygen species in renal physiology and pathology. Antioxid Redox Signal 2016, 25, 732–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ratliff BB, Abdulmahdi W, Pawar R, Wolin MS. Oxidant mechanisms in renal injury and disease. Antioxid Redox Signal 2016; 25: 119–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Banday AA, Lokhandwala MF. Transcriptional regulation of renal dopamine D1 receptor function during oxidative stress. Hypertension 2015; 65: 1064–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yan Y, Shapiro AP, Haller S et al. Involvement of reactive oxygen species in a feed-forward mechanism of Na/K-ATPase-mediated signaling transduction. J Biol Chem 2013; 288: 34249–34258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lu X, Wang F, Liu M et al. Activation of ENaC in collecting duct cells by prorenin and its receptor PRR: involvement of Nox4-derived hydrogen peroxide. Am J Physiol Renal Physiol 2016; 310: F1243–1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Silva-Islas CA, Maldonado PD. Canonical and non-canonical mechanisms of Nrf2 activation. Pharmacol Res. 2018; 134: 92–99. [DOI] [PubMed] [Google Scholar]
  • 20.Chung HS, Wang SB, Venkatraman V, Murray CI, Van Eyk JE. Cysteine oxidative posttranslational modifications: emerging regulation in the cardiovascular system. Circ Res 2013; 112: 382–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes 2003; 52: 1–8. [DOI] [PubMed] [Google Scholar]
  • 22.Wang D, Chen Y, Chabrashvili T et al. Role of oxidative stress in endothelial dysfunction and enhanced responses to angiotensin II of afferent arterioles from rabbits infused with angiotensin II. J Am Soc Nephrol 2003; 14: 2783–2789. [DOI] [PubMed] [Google Scholar]
  • 23.Kopkan L, Castillo A, Navar LG et al. Enhanced superoxide generation modulates renal function in ANG II-induced hypertensive rats. Am J Physiol Renal Physiol 2006; 290: F80–F86. [DOI] [PubMed] [Google Scholar]
  • 24.Kaess BM, Rong J, Larson MG et al. Aortic stiffness, blood pressure progression, and incident hypertension. JAMA 2012; 308: 875–881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wu J, Saleh MA, Kirabo A et al. Immune activation caused by vascular oxidation promotes fibrosis and hypertension. J Clin Invest 2016; 126; 50–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zucchelli P, Zuccalà A, Mancini E. Hypertension in primary glomerulonephritis without renal insufficiency. Nephrol Dial Transplant 1989; 4: 605–610. [PubMed] [Google Scholar]
  • 27.Nagase M, Shibata S, Yoshida S et al. Podocyte injury underlies the glomerulopathy of Dahl salt-hypertensive rats and is reversed by aldosterone blocker. Hypertension 2006; 47: 1084–1093. [DOI] [PubMed] [Google Scholar]
  • 28.Meng S, Cason GW, Gannon AW et al. Oxidative stress in Dahl salt-sensitive hypertension. Hypertension 2003; 41: 1346–1352. [DOI] [PubMed] [Google Scholar]
  • 29.Araujo M, Wilcox CS. Oxidative stress in hypertension: role of the kidney. Antioxid Redox Signal 2014; 20: 74–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lerman LO, Nath KA, Rodriguez-Porcel M et al. Increased oxidative stress in experimental renovascular hypertension. Hypertension 2001; 37: 541–546. [DOI] [PubMed] [Google Scholar]
  • 31.Lin Y, Luo R et al. Intrarenal renin-angiotensin system mediates fatty acid-induced ER stress in the kidney. Am J Physiol Renal Physiol 2016; 310: F351–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cuevas S, Villar VA, Jose PA, Armando I. Renal dopamine receptors, oxidative stress, and hypertension. Int J Mol Sci 2013; 14: 17553–17572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rukavina Mikusic NL, Kravetz MC, Kouyoumdzian NM et al. Signaling pathways involved in renal oxidative injury: role of the vasoactive peptides and the renal dopaminergic system. J Signal Transduct 2014; 2014: 731350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Stec DE, Juncos LA, Granger JP. Renal intramedullary infusion of tempol normalizes the blood pressure response to intrarenal blockade of heme oxygenase-1 in angiotensin II-dependent hypertension. J Am Soc Hypertens 2016; 10:346–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Silva GB, Garvin JL. Angiotensin II-dependent hypertension increases Na transport- related oxygen consumption by the thick ascending limb. Hypertension 2008; 52: 1091–1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Luo H, Wang X, Chen C et al. Oxidative stress causes imbalance of renal renin angiotensin system (RAS) components and hypertension in obese Zucker rats. J Am Heart Assoc 2015; 4: e001559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ramkumar N, Kohan DE. Proximal tubule angiotensinogen modulation of arterial pressure. Curr Opin Nephrol Hypertens 2013; 22: 32–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Labandeira-Garcia JL, Rodriguez-Pallares J, Villar-Cheda B et al. Angiotensin system and dopaminergic degeneration in the substantia nigra. Aging Dis 2011; 2: 257–274. [PMC free article] [PubMed] [Google Scholar]
  • 39.Escano CS, Armando I, Wang X et al. Renal dopaminergic defect in C57Bl/6J mice. Am J Physiol Regul Integr Comp Physiol 2009; 297: R1660–1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chugh G, Lokhandwala MF, Asghar M. Altered functioning of both renal dopamine D1 and angiotensin II type 1 receptors causes hypertension in old rats. Hypertension 2012; 59: 1029–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ortiz PA, Garvin JL. Superoxide stimulates NaCl absorption by the thick ascending limb. Am J Physiol Renal Physiol 2002; 283: F957–962. [DOI] [PubMed] [Google Scholar]
  • 42.Persson P, Hansell P, Palm F. NADPH oxidase inhibition reduces tubular sodium transport and improves kidney oxygenation in diabetes. Am J Physiol Regul Integr Comp Physiol 2012; 302: R1443–1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Cowley AW Jr, Abe M, Mori T et al. Reactive oxygen species as important determinants of medullary flow, sodium excretion, and hypertension. Am J Physiol Renal Physiol 2015; 308: F179–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Simão S, Gomes P, Pinho MJ, Soares-da-Silva P. H2O2 stimulates Cl- /HCO3- exchanger activity through oxidation of thiol groups in immortalized SHR renal proximal tubular epithelial cells. J Cell Biochem 2011; 112: 3660–3665. [DOI] [PubMed] [Google Scholar]
  • 45.Shah PT, Martin R, Yan Y, Shapiro JI, Liu J. Carbonylation modification regulates Na/K-ATPase signaling and salt sensitivity: A review and a hypothesis. Front Physiol 2016; 7: 256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.O’Connor PM, Guha A, Stilphen CA, Sun J, Jin C. Proton channels and renal hypertensive injury: a key piece of the Dahl salt-sensitive rat puzzle? Am J Physiol Regul Integr Comp Physiol 2016; 310: R679–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sun P, Yue P, Wang WH. Angiotensin II stimulates epithelial sodium channels in the cortical collecting duct of the rat kidney. Am J Physiol Renal Physiol 2012; 302: F679–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Sedeek M, Nasrallah R, Touyz RM, Hébert RL. NADPH oxidases, reactive oxygen species, and the kidney: friend and foe. J Am Soc Nephrol. 2013; 24:1512–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cabral PD, Garvin JL. Luminal flow regulates NO and O2(−) along the nephron. Am J Physiol Renal Physiol 2011; 300: F1047–1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Yan Y, Shapiro AP, Haller S et al. Involvement of reactive oxygen species in a feed-forward mechanism of Na/K-ATPase-mediated signaling transduction. J Biol Chem 2013; 288: 34249–34258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Andreoli SP1, McAteer JA, Seifert SA, Kempson SA. Oxidant-induced alterations in glucose and phosphate transport in LLC-PK1 cells: mechanisms of injury. Am J Physiol 1993; 265: F377–3784. [DOI] [PubMed] [Google Scholar]
  • 52.Kurella EG, Tyulina OV, Boldyrev AA. Oxidative resistance of Na/K-ATPase. Cell Mol Neurobiol 1999; 19:133–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Panico C, Luo Z, Damiano S, Artigiano F, Gill P, Welch WJ. Renal proximal tubular reabsorption is reduced in adult spontaneously hypertensive rats: roles of superoxide and Na+/H+ exchanger 3. Hypertension 2009; 54:1291–1297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Richards SL, Wilkins KA, Swarbreck SM et al. The hydroxyl radical in plants: from seed to seed. J Exp Bot 2015; 66: 37–46. [DOI] [PubMed] [Google Scholar]
  • 55.Nikinmaa M, Bogdanova A, Lecklin T. Oxygen dependency of the adrenergic Na/H exchange in rainbow trout erythrocytes is diminished by a hydroxyl radical scavenger. Acta Physiol Scand 2003; 178: 149–154. [DOI] [PubMed] [Google Scholar]
  • 56.Massey KJ, Hong NJ, Garvin JL. Angiotensin II stimulates superoxide production in the thick ascending limb by activating NOX4. Am J Physiol Cell Physiol 2012; 303: C781–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kopkan L, Cervenka L. Renal interactions of renin-angiotensin system, nitric oxide and superoxide anion: implications in the pathophysiology of salt-sensitivity and hypertension. Physiol Res 2009; 58: S55–67. [DOI] [PubMed] [Google Scholar]
  • 58.Crajoinas RO, Lessa LM, Carraro-Lacroix LR et al. Posttranslational mechanisms associated with reduced NHE3 activity in adult vs. young prehypertensive SHR. Am J Physiol Renal Physiol. 2010; 299: F872–881. [DOI] [PubMed] [Google Scholar]
  • 59.Boer PA, Morelli JM, Figueiredo JF, Gontijo JA. Early altered renal sodium handling determined by lithium clearance in spontaneously hypertensive rats (SHR): role of renal nerves. Life Sci 2005; 76: 1805–1815. [DOI] [PubMed] [Google Scholar]
  • 60.Pinto V, Pinho MJ, Hopfer U, Jose PA, Soares-da-Silva P. Oxidative stress and the genomic regulation of aldosterone-stimulated NHE1 activity in SHR renal proximal tubular cells. Mol Cell Biochem 2008; 310: 191–201. [DOI] [PubMed] [Google Scholar]
  • 61.Huang WH, Wang Y, Askari A. (Na+ + K+)-ATPase: inactivation and degradation induced by oxygen radicals. Int J Biochem 1992; 24: 621–626. [DOI] [PubMed] [Google Scholar]
  • 62.Banday AA, Lokhandwala MF. Transcription factor Nrf2 protects renal dopamine D1 receptor function during oxidative stress. Hypertension 2013; 62: 512–7. [DOI] [PubMed] [Google Scholar]
  • 63.Zou L, Linck V, Zhai YJ, Galarza-Paez L, Li L, Yue Q, Al-Khalili O, Bao HF, Ma HP, Thai TL, Jiao J, Eaton DC.. Knockout of mitochondrial voltage-dependent anion channel type 3 increases reactive oxygen species (ROS) levels and alters renal sodium transport. J Biol Chem. 2018; 293: 1666–1675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chugh G, Lokhandwala MF, Asghar M. Oxidative stress alters renal D1 and AT1 receptor functions and increases blood pressure in old rats. Am J Physiol Renal Physiol 2011; 300: F133–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sen CK, Packer L. Antioxidant and redox regulation of gene transcription. FASEB J 1996; 10:709–720. [DOI] [PubMed] [Google Scholar]
  • 66.Furukawa S, Fujita T, Shimabukuro M et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest 2004; 114: 1752–1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Thayer TC, Delano M, Liu C et al. Superoxide production by macrophages and T cells is critical for the induction of autoreactivity and type 1 diabetes. Diabetes 2011; 60: 2144–2151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cachofeiro V, Goicochea M, de Vinuesa SG et al. Oxidative stress and inflammation, a link between chronic kidney disease and cardiovascular disease. Kidney Int Suppl 2008:S4–9. [DOI] [PubMed] [Google Scholar]
  • 69.Manning AM, Bell FP, Rosenbloom CL, et al. NF-kappa B is activated during acute inflammation in vivo in association with elevated endothelial cell adhesion molecule gene expression and leukocyte recruitment. J Inflamm 1995; 45: 283–296. [PubMed] [Google Scholar]
  • 70.Theuer J, Dechend R, Muller DN et al. Angiotensin II induced inflammation in the kidney and in the heart of double transgenic rats. BMC Cardiovasc Disord 2002; 2: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nishi EE, Oliveira-Sales EB, Bergamaschi CT, Oliveira TG, Boim MA, Campos RR. Chronic antioxidant treatment improves arterial renovascular hypertension and oxidative stress markers in the kidney in Wistar rats. Am J Hypertens 2010; 23: 473–480. [DOI] [PubMed] [Google Scholar]
  • 72.Hurr C, Young CN. Neural control of non-vasomotor organs in hypertension. Curr Hypertens Rep. 2016; 18: 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Yang S, Gao Y, Liu G et al. The human ATF1 rs11169571 polymorphism increases essential hypertension risk through modifying miRNA binding. FEBS Lett 2015; 589: 2087–2093. [DOI] [PubMed] [Google Scholar]
  • 74.Zhang L, Miyaki K, Araki J, Song Y, Kimura T, Omae K, Muramatsu M. Interaction of angiotensin I-converting enzyme insertion-deletion polymorphism and daily salt intake influences hypertension in Japanese men. Hypertens Res. 2006; 29: 751–758. [DOI] [PubMed] [Google Scholar]
  • 75.Todoroki M, Minami J, Ishimitsu T, Ohrui M, Matsuoka H. Relation between the angiotensin-converting enzyme insertion/deletion polymorphism and blood pressure in Japanese male subjects. J Hum Hypertens. 2003; 17: 713–718. [DOI] [PubMed] [Google Scholar]
  • 76.Jain S, Vinukonda G, Fiering SN, Kumar A. A haplotype of human angiotensinogen gene containing −217A increases blood pressure in transgenic mice compared with −217G. Am J Physiol Regul Integr Comp Physiol 2008; 295: R1849–1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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; 285:41172–41186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kayima J, Liang J, Natanzon Y, Nankabirwa J, Ssinabulya I, Nakibuuka J, et al. Association of genetic variation with blood pressure traits among East Africans. Clin Genet. 2017; 92: 487–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kumar A, Li Y, Patil S, Jain S. A haplotype of the angiotensinogen gene is associated with hypertension in african americans. Clin Exp Pharmacol Physiol 2005; 32: 495–502. [DOI] [PubMed] [Google Scholar]
  • 80.Zeng R, Wang QP, Fang MX, Zhuang J, Fan RX. Association of A-20C polymorphism in the angiotensinogen gene with essential hypertension: a meta-analysis. Genet Mol Res 2015; 14: 12984–12992. [DOI] [PubMed] [Google Scholar]
  • 81.Norat T, Bowman R, Luben R, Welch A, Khaw KT, Wareham N et al. Blood pressure and interactions between the angiotensin polymorphism AGT M235T and sodium intake: a cross-sectional population study. Am J Clin Nutr 2008; 88:392–397. [DOI] [PubMed] [Google Scholar]
  • 82.Purkait P, Halder K, Thakur S, et al. Association of angiotensinogen gene SNPs and haplotypes with risk of hypertension in eastern Indian population. Clin Hypertens. 2017; 23 :12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sethi AA, Nordestgaard BG, Tybjaerg-Hansen A. Angiotensinogen gene polymorphism, plasma angiotensinogen, and risk of hypertension and ischemic heart disease: a meta-analysis. Arterioscler Thromb Vasc Biol. 2003. ;23 :1269–1275. [DOI] [PubMed] [Google Scholar]
  • 84.Forman JP, Fisher ND, Pollak MR, Cox DG, Tonna S, Curhan GC. Renin-angiotensin system polymorphisms and risk of hypertension: influence of environmental factors. J Clin Hypertens 2008; 10: 459–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sano M, Kuroi N, Nakayama T, Sato N, Izumi Y, Soma M. Association study: the aminopeptidase a gene and essential hypertension. Int J Biomed Sci. 2005;1 : 16–22. [PMC free article] [PubMed] [Google Scholar]
  • 86.Te Riet L, van Esch JH, Roks AJ, van den Meiracker AH, Danser AH. Hypertension: renin-angiotensin-aldosterone system alterations. Circ Res 2015: 116, 960–975. [DOI] [PubMed] [Google Scholar]
  • 87.Van Haaster MC, McDonough AA, Gurley SB. Blood pressure regulation by the angiotensin type 1 receptor in the proximal tubule. Curr Opin Nephrol Hypertens. 2018; 27: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Cipollone F, Toniato E, Martinotti S, Fazia M, Iezzi A, Cuccurullo C et al. A polymorphism in the cyclooxygenase 2 gene as an inherited protective factor against myocardial infarction and stroke. JAMA 2004; 291: 2221–2228. [DOI] [PubMed] [Google Scholar]
  • 89.Martínez-Revelles S, Avendaño MS, García-Redondo AB et al. Reciprocal relationship between reactive oxygen species and cyclooxygenase-2 and vascular dysfunction in hypertension. Antioxid Redox Signal 2013; 18: 51–65. [DOI] [PubMed] [Google Scholar]
  • 90.Iwai N, Tago N, Yasui N, Kokubo Y, Inamoto N, Tomoike H, Shioji K. Genetic analysis of 22 candidate genes for hypertension in the Japanese population. J Hypertens. 2004; 22: 1119–1126. [DOI] [PubMed] [Google Scholar]
  • 91.Fava C, Ricci M, Melander O, Minuz P. Hypertension, cardiovascular risk and polymorphisms in genes controlling the cytochrome P450 pathway of arachidonic acid: A sex-specific relation? Prostaglandins Other Lipid Mediat. 2012; 98: 75–85. [DOI] [PubMed] [Google Scholar]
  • 92.Jennings BL, Montanez DE, May ME Jr et al. Cytochrome P450 1B1 contributes to increased blood pressure and cardiovascular and renal dysfunction in spontaneously hypertensive rats. Cardiovasc Drugs Ther 2014; 28: 145–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Savas Ü, Wei S, Hsu MH, Falck JR, Guengerich FP, Capdevila JH, Johnson EF. 20-Hydroxyeicosatetraenoic Acid (HETE)-dependent Hypertension in Human Cytochrome P450 (CYP) 4A11 Transgenic Mice: NORMALIZATION OF BLOOD PRESSURE BY SODIUM RESTRICTION, HYDROCHLOROTHIAZIDE, OR BLOCKADE OF THE TYPE 1 ANGIOTENSIN II RECEPTOR. J Biol Chem. 2016; 291: 16904–16919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Jennings BL, Moore JA, Pingili AK, Estes AM, Fang XR, Kanu A, Gonzalez FJ, Malik KU. Disruption of the cytochrome P-450 1B1 gene exacerbates renal dysfunction and damage associated with angiotensin II-induced hypertension in female mice. Am J Physiol Renal Physiol. 2015; 308: F981–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Dennerlein S, Rehling P. Human mitochondrial COX1 assembly into cytochrome c oxidase at a glance. J Cell Sci 2015; 128; 833–837. [DOI] [PubMed] [Google Scholar]
  • 96.Polonikov AV, Ivanov VP, Solodilova MA, et al. A common polymorphism G-50T in cytochrome P450 2J2 gene is associated with increased risk of essential hypertension in a Russian population. Dis Markers 2008: 24: 119–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.King LM, Gainer JV, David GL et al. Single nucleotide polymorphisms in the CYP2J2 and CYP2C8 genes and the risk of hypertension. Pharmacogenet Genomics 2005: 15: 7–13. [DOI] [PubMed] [Google Scholar]
  • 98.Zhang L, Miyaki K, Wang W, Muramatsu M. CYP3A5 polymorphism and sensitivity of blood pressure to dietary salt in Japanese men. J Hum Hypertens 2010; 24: 345–350. [DOI] [PubMed] [Google Scholar]
  • 99.Tzveova R, Naydenova G, Yaneva T, et al. Gender-Specific Effect of CYP2C8*3 on the Risk of Essential Hypertension in Bulgarian Patients. Biochem Genet. 2015; 53: 319–333. [DOI] [PubMed] [Google Scholar]
  • 100.Bochud M, Eap CB, Elston RC, et al. Association of CYP3A5 genotypes with blood pressure and renal function in African families. J Hypertens. 2006; 24: 923–929. [DOI] [PubMed] [Google Scholar]
  • 101.Plenty NL, Faulkner JL, Cotton J, et al. Arachidonic acid metabolites of CYP4A and CYP4F are altered in women with preeclampsia. Prostaglandins Other Lipid Mediat. 2018; 136: 15–22. [DOI] [PubMed] [Google Scholar]
  • 102.Fu Z, Nakayama T, Sato N, Izumi Y, et al. haplotype of the CYP4A11 gene associated with essential hypertension in Japanese men. J Hypertens. 2008; 26: 453–461. [DOI] [PubMed] [Google Scholar]
  • 103.Zhang H, Jin L, Mu T, et al. Associations of CYP4A11 gene-gene and gene-smoking interactions with essential hypertension in the male eastern Chinese Han population. Clin Exp Hypertens. 2017; 39: 448–453. [DOI] [PubMed] [Google Scholar]
  • 104.Luo H, Wang X, Wang J et al. Chronic NF-κB blockade improves renal angiotensin II type 1 receptor functions and reduces blood pressure in Zucker diabetic rats. Cardiovasc Diabetol 2015; 14: 76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rahman T, Baker M, Hall DH, Avery PJ, Keavney B. Common genetic variation in the type A endothelin-1 receptor is associated with ambulatory blood pressure: a family study. J Hum Hypertens. 2008; 22: 282–288. [DOI] [PubMed] [Google Scholar]
  • 106.Nikkari ST, Visto AL, Määttä KM, Kunnas TA. Minor variant of rs 16827043 in the iron regulator hemojuvelin gene (HJV) contributes to hypertension: The TAMRISK study. Medicine (Baltimore). 2017; 96: e6052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Manhiani MM, Seth DM, Banes-Berceli AK et al. The role of IL-6 in the physiologic versus hypertensive blood pressure actions of angiotensin II. Physiol Rep 2015; 3: e12595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bayoumy NM, Al-Sharaidh AS, Babay ZH, Abdulgader AM. The role of interleukin-6 promoter polymorphism −174G/C in Saudi women with hypertensive disorders of pregnancy. Saudi Med J 2013; 34: 689–694. [PubMed] [Google Scholar]
  • 109.Losito A, Kalidas K, Santoni S, Jeffery S. Association of interleukin-6 −174G/C promoter polymorphism with hypertension and left ventricular hypertrophy in dialysis patients. Kidney Int 2003; 64: 616–622. [DOI] [PubMed] [Google Scholar]
  • 110.Riikola A1, Sipilä K, Kähönen M, Jula A, Nieminen MS, Moilanen L, Kesäniemi YA, Lehtimäki T, Hulkkonen J. Interleukin-6 promoter polymorphism and cardiovascular risk factors: the Health 2000 Survey. Atherosclerosis. 2009; 207: 466–470. [DOI] [PubMed] [Google Scholar]
  • 111.Ma H, Sun G, Wang W, Zhou Y, Liu D, Tong Y, Lu Z. Association Between Interleukin-6 −572 C>G and −174 G>C Polymorphisms and Hypertension: A Meta-analysis of Case-control Studies. Medicine (Baltimore). 2016; 95 :e2416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Saleh MA, Norlander AE, Madhur MS. Inhibition of Interleukin 17-A but not Interleukin-17F Signaling Lowers Blood Pressure and Reduces End-organ Inflammation in Angiotensin II-induced Hypertension. JACC Basic Transl Sci. 2016; 1: 606–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Shuang L, Li Z, Chen F, et al. Association between interleukin-17 gene polymorphisms and risk of coronary artery disease. Int J Clin Exp Pathol. 2015; 8: 11653–11658. [PMC free article] [PubMed] [Google Scholar]
  • 114.Lu X, Wang L, Lin X, Huang J, Charles Gu C et al. Genome-wide association study in Chinese identifies novel loci for blood pressure and hypertension. Hum Mol Genet. 2015; 24: 865–874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Huby AC, Otvos L Jr, Belin de Chantemèle EJ. Leptin induces hypertension and endothelial dysfunction via aldosterone-dependent mechanisms in obese female mice. Hypertension 2016; 67: 1020–1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.do Carmo JM, da Silva AA, Wang Z, Fang T, Aberdein N, de Lara Rodriguez CE, Hall JE. Obesity-Induced Hypertension: Brain Signaling Pathways. Curr Hypertens Rep. 2016; 18: 58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Li YX, Zhang Q, Shang XM, Li YQ, Liu XK, Liu CQ et al. Association of two well-defined polymorphisms in leptin and leptin receptor genes with hypertension and circulating leptin: a meta-analysis. Arch Med Res 2015; 46: 38–46. [DOI] [PubMed] [Google Scholar]
  • 118.Farias DR, Franco-Sena AB, Rebelo F, Salles GF, Struchiner CJ, Martins MC et al. Polymorphisms of Leptin (G2548A) and Leptin Receptor (Q223R and K109R) Genes and Blood Pressure During Pregnancy and the Postpartum Period: A Cohort. Am J Hypertens. 2017; 30: 130–140. [DOI] [PubMed] [Google Scholar]
  • 119.Rosmond R, Chagnon YC, Holm G, Chagnon M, Pérusse L, Lindell K, Carlsson B, Bouchard C, Björntorp P. Hypertension in obesity and the leptin receptor gene locus. J Clin Endocrinol Metab 2000; 85: 3126–3131 [DOI] [PubMed] [Google Scholar]
  • 120.Doi K, Noiri E, Maeda R, Nakao A, Kobayashi S, Tokunaga K et al. Functional polymorphism of the myeloperoxidase gene in hypertensive nephrosclerosis dialysis patients. Hypertens Res 2007; 30: 1193–1198. [DOI] [PubMed] [Google Scholar]
  • 121.Liu YC, Chung CJ, Shiue HS, Cheng YY, Huang SR, Su CT et al. Genetic polymorphisms of myeloperoxidase and their effect on hypertension. Blood Press 2013; 22: 282–289. [DOI] [PubMed] [Google Scholar]
  • 122.Fang J, Ma L, Zhang S, Fang Y, Su P, Ma H. Association of myeloperoxidase gene variation with carotid atherosclerosis in patients with essential hypertension. Mol Med Rep 2013; 7: 313–317. [DOI] [PubMed] [Google Scholar]
  • 123.Bushueva O, Solodilova M, Ivanov V, Polonikov A. Gender-specific protective effect of the −463G>A polymorphism of myeloperoxidase gene against the risk of essential hypertension in Russians. J Am Soc Hypertens 2015; 9: 902–906. [DOI] [PubMed] [Google Scholar]
  • 124.Hasanpour Z, Javanmard SH, Gharaaty M, Sadeghi M. Association between serum myeloperoxidase levels and coronary artery disease in patients without diabetes, hypertension, obesity, and hyperlipidemia. Adv Biomed Res 2016; 5: 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Nouri P, Gill P, Li M, Wilcox CS, Welch WJ. p22phox in the macula densa regulates single nephron GFR during angiotensin II infusion in rats. Am J Physiol Heart Circ Physiol 2007; 292: H1685–1689. [DOI] [PubMed] [Google Scholar]
  • 126.Modlinger P, Chabrashvili T, Gill PS, Mendonca M, Harrison DG, Griendling KK et al. RNA silencing in vivo reveals role of p22phox in rat angiotensin slow pressor response. Hypertension 2006; 47: 238–244. [DOI] [PubMed] [Google Scholar]
  • 127.Kumar R, Kohli S, Ali Z. CYBA (p22phox) variants associate with blood pressure and oxidative stress markers in hypertension: a replication study in populations of diverse altitudes. Hypertens Res 2015; 38: 498–506. [DOI] [PubMed] [Google Scholar]
  • 128.Bedard K, Attar H, Bonnefont J, Jaquet V, Borel C, Plastre O et al. Three common polymorphisms in the CYBA gene form a haplotype associated with decreased ROS generation. Hum Mutat 2009: 30: 1123–1133. [DOI] [PubMed] [Google Scholar]
  • 129.Petrovič D Association of the −262C/T polymorphism in the catalase gene promoter and the C242T polymorphism of the NADPH oxidase P22phox gene with essential arterial hypertension in patients with diabetes mellitus type 2. Clin Exp Hypertens 2014; 36: 36–39. [DOI] [PubMed] [Google Scholar]
  • 130.Schreiber R, Bellinazzi VR, Sposito AC, Mill JG, Krieger JE, Pereira AC et al. Influence of the C242T polymorphism of the p22-phox gene (CYBA) on the interaction between urinary sodium excretion and blood pressure in an urban Brazilian population. PLoS One 2013. 8: e81054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Castejon AM, Bracero J, Hoffmann IS, Alfieri AB, Cubeddu LX. NAD(P)H oxidase p22phox gene C242T polymorphism, nitric oxide production, salt sensitivity and cardiovascular risk factors in Hispanics. J Hum Hypertens 2006; 20: 772–779. [DOI] [PubMed] [Google Scholar]
  • 132.Di Castelnuovo A, Soccio M, Iacoviello L, Evangelista V, Consoli A, Vanuzzo D et al. The C242T polymorphism of the p22phox component of NAD(P)H oxidase and vascular risk. Two case-control studies and a meta-analysis. Thromb Haemost 2008; 99: 594–601. [DOI] [PubMed] [Google Scholar]
  • 133.Sun Q, Yin Y, Zhu Z, Yan Z. Association of the C242T polymorphism in the NAD(P)H oxidase p22 phox gene with type 2 diabetes mellitus risk: a meta-analysis. Curr Med Res Opin 2014; 30: 415–422. [DOI] [PubMed] [Google Scholar]
  • 134.Qin YW, Peng J, Liang BY, Su L, Chen Q, Xie JJ et al. The A930G polymorphism of p22phox (CYBA) gene but not C242T variation is associated with hypertension: a meta-analysis. PLoS One 2013; 8: e82465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Nowak T, Niemiec P, Górczyńska-Kosiorz S, Balcerzyk A, Iwanicki T, Krauze J et al. The CYBA Gene (α) 49A>G polymorphism (rs7195830) is associated with hypertension in patients with coronary artery disease. Biomed Res Int 2016; 2016: 1539671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Moreno MU1, San José G, Fortuño A, Beloqui O, Redón J, Chaves FJ et al. A novel CYBA variant, the −675A/T polymorphism, is associated with essential hypertension. J Hypertens 2007; 25: 1620–1626. [DOI] [PubMed] [Google Scholar]
  • 137.Niemiec P, Zak I, Emich-Widera E et al. The C242T polymorphism of the gene encoding cytochrome b-245 alpha is not associated with paediatric ischaemic stroke: family-based and case-control study. Neurol Neurochir Pol 2010; 44: 453–458. [DOI] [PubMed] [Google Scholar]
  • 138.Feairheller DL, Brown MD, Park JY, Brinkley TE, Basu S, Hagberg JM et al. Exercise training, NADPH oxidase p22phox gene polymorphisms, and hypertension. Med Sci Sports Exerc 2009; 41: 1421–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Schreiber R, Ferreira-Sae MC, Ronchi JA, Pio-Magalhães JA, Cipolli JA, Matos-Souza JR et al. The C242T polymorphism of the p22-phox gene (CYBA) is associated with higher left ventricular mass in Brazilian hypertensive patients. BMC Med Genet 2011; 12: 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gavazzi G, Banfi B, Deffert C, Fiette L, Schappi M, Herrmann F et al. Decreased blood pressure in NOX1-deficient mice. FEBS Lett 2006; 580: 497–504. [DOI] [PubMed] [Google Scholar]
  • 141.Sirokmány G, Donkó Á, Geiszt M. Nox/Duox Family of NADPH Oxidases: Lessons from Knockout Mouse Models. Trends Pharmacol Sci. 2016. ;37: 318–327. [DOI] [PubMed] [Google Scholar]
  • 142.Fujii A, Nakano D, Katsuragi M, Ohkita M, Takaoka M, Ohno Y et al. Role of gp91phox-containing NADPH oxidase in the deoxycorticosterone acetate-salt-induced hypertension. Eur J Pharmacol 2006; 552: 131–134. [DOI] [PubMed] [Google Scholar]
  • 143.Haque MZ, Majid DS. High salt intake delayed angiotensin II-induced hypertension in mice with a genetic variant of NADPH oxidase. Am J Hypertens 2011; 24:114–118. [DOI] [PubMed] [Google Scholar]
  • 144.Murdoch CE, Alom-Ruiz SP, Wang M, Zhang M, Walker S, Yu B et al. Role of endothelial Nox2 NADPH oxidase in angiotensin II-induced hypertension and vasomotor dysfunction. Basic Res Cardiol 2011; 106: 527–538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zhang A, Jia Z, Wang N, Tidwell TJ, Yang T. Relative contributions of mitochondria and NADPH oxidase to deoxycorticosterone acetate-salt hypertension in mice. Kidney Int 2011; 80: 51–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yang Y, Zhang Y, Cuevas S, Villar VA, Escano C, D Asico L et al. Paraoxonase 2 decreases renal reactive oxygen species production, lowers blood pressure, and mediates dopamine D2 receptor-induced inhibition of NADPH oxidase. Free Radic Biol Med 2012; 53: 437–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ray R, Murdoch CE, Wang M, Santos CX, Zhang M, Alom-Ruiz S et al. Endothelial Nox4 NADPH oxidase enhances vasodilatation and reduces blood pressure in vivo. Arterioscler Thromb Vasc Biol 2011; 31: 1368–1376. [DOI] [PubMed] [Google Scholar]
  • 148.Li H, Han X, Hu Z, Huang J, Chen J, Hixson JE, Rao DC, He J, Gu D, Chen S. Associations of NADPH oxidase-related genes with blood pressure changes and incident hypertension: The GenSalt Study. J Hum Hypertens. 2018; 32: 287–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Lai EY, Wellstein A, Welch WJ, Wilcox CS. Superoxide modulates myogenic contractions of mouse afferent arterioles. Hypertension. 2011; 58: 650–656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Hsueh YM, Lin P, Chen HW, Shiue HS, Chung CJ, Tsai CT, Huang YK, Chiou HY, Chen CJ. Genetic polymorphisms of oxidative and antioxidant enzymes and arsenic-related hypertension. J Toxicol Environ Health A. 2005; 68: 1471–1484. [DOI] [PubMed] [Google Scholar]
  • 151.Conen D, Glynn RJ, Buring JE, Ridker PM, Zee RY. Association of renin-angiotensin and endothelial nitric oxide synthase gene polymorphisms with blood pressure progression and incident hypertension: prospective cohort study. J Hypertens 2008; 26: 1780–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Han X, Hu Z, Chen J, Huang J, Huang C, Liu F, Gu C, Yang X, Hixson JE, Lu X, Wang L, Liu DP, He J, Chen S, Gu D. Associations Between Genetic Variants of NADPH Oxidase-Related Genes and Blood Pressure Responses to Dietary Sodium Intervention: The GenSalt Study. Am J Hypertens 2017; 30: 427–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Godin N, Liu F, Lau GJ, Brezniceanu et al. Catalase overexpression prevents hypertension and tubular apoptosis in angiotensinogen transgenic mice. Kidney Int 2010; 77: 1086–1097. [DOI] [PubMed] [Google Scholar]
  • 154.Bushueva OY, Ivanov VP, Ryzhaeva VN, Ponomarenko IV, Churnosov MI, Polonikov AV. [Association of the −844G>A polymorphism in the catalase gene with the increased risk of essential hypertension in smokers]. Ter Arkh. 2016; 88: 50–54 [DOI] [PubMed] [Google Scholar]
  • 155.Kouremenos N, Zacharopoulou IV, Triantafyllidi H, Zacharopoulos GV, Mornos C, Filippatos G, et al. Genes and genetic variations involved in the development of hypertension: focusing on a Greek patient cohort. Hellenic J Cardiol. 2014; 55: 9–16. [PubMed] [Google Scholar]
  • 156.Zhou XF, Cui J, DeStefano AL et al. Polymorphisms in the promoter region of catalase gene and essential hypertension. Dis Markers 2005; 21: 3–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Jiang Z, Akey JM, Shi J et al. A polymorphism in the promoter region of catalase is associated with blood pressure levels. Hum Genet 2001; 109: 95–98. [DOI] [PubMed] [Google Scholar]
  • 158.Yang G, Wu L, Jiang B, Yang W, Qi J, Cao K et al. H2S as a physiologic vasorelaxant: hypertension in mice with deletion of cystathionine gamma-lyase. Science. 2008; 322: 587–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Ishii I, Akahoshi N, Yamada H, Nakano S, Izumi T, Suematsu M. Cystathionine gamma-Lyase-deficient mice require dietary cysteine to protect against acute lethal myopathy and oxidative injury. J Biol Chem 2010; 285: 26358–26368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Gupta V, Kapopara PR, Khan AA, Arige V, Subramanian L, Sonawane PJ, Sasi BK, Mahapatra NR. Functional promoter polymorphisms direct the expression of cystathionine gamma-lyase gene in mouse models of essential hypertension. J Mol Cell Cardiol. 2017; 102: 61–73. [DOI] [PubMed] [Google Scholar]
  • 161.Mrozikiewicz PM, Bogacz A, Omielańczyk M, et al. The importance of rs1021737 and rs482843 polymorphisms of cystathionine gamma-lyase in the etiology of preeclampsia in the Caucasian population. Ginekol Pol. 2015; 86: 119–125. [DOI] [PubMed] [Google Scholar]
  • 162.Cuevas S, Zhang Y, Yang Y et al. Role of renal DJ-1 in the pathogenesis of hypertension associated with increased reactive oxygen species production. Hypertension 2012; 59: 446–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Cuevas S, Yang Y, Konkalmatt P et al. Role of nuclear factor erythroid 2-related factor 2 in the oxidative stress-dependent hypertension associated with the depletion of DJ-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Yamane T, Murao S, Kozuka M, Shimizu M, Suzuki J, Kubo C et al. Serum DJ-1 level is positively associated with improvements in some aspects of metabolic syndrome in Japanese women through lifestyle intervention. Nutr Res 2014; 34: 851–855. [DOI] [PubMed] [Google Scholar]
  • 165.Albrecht FE, Drago J, Felder RA et al. Role of the D1A dopamine receptor in the pathogenesis of genetic hypertension. J Clin Invest 1996; 97: 2283–2288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Sato M, Soma M, Nakayama T, Kanmatsuse K. Dopamine D1 receptor gene polymorphism is associated with essential hypertension. Hypertension 2000; 36: 183–186. [DOI] [PubMed] [Google Scholar]
  • 167.Orun O, Nacar C, Cabadak H et al. Investigation of the association between dopamine D1 receptor gene polymorphisms and essential hypertension in a group of Turkish subjects. Clin Exp Hypertens 2011; 33: 418–421. [DOI] [PubMed] [Google Scholar]
  • 168.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] [PMC free article] [PubMed] [Google Scholar]
  • 169.Yang H, Zhong L, Bai S, Dong Y, Wang Y, Li Q et al. Association of Dopamine D1 and D3 receptor gene polymorphisms with essential hypertension in 3 ethnic groups in China. Med Sci Monit Basic Res 2017; 23: 234–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Staessen JA, Kuznetsova T, Zhang H, Maillard M, Bochud M, Hasenkamp 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. [DOI] [PubMed] [Google Scholar]
  • 171.Rosmond R, Rankinen T, Chagnon M, Pérusse L, Chagnon YC, Bouchard C, Björntorp P. Polymorphism in exon 6 of the dopamine D(2) receptor gene (DRD2) is associated with elevated blood pressure and personality disorders in men. J Hum Hypertens. 2001; 15: 553–558 . [DOI] [PubMed] [Google Scholar]
  • 172.Jiang X, Konkalmatt P, Yang Y et al. Single-nucleotide polymorphisms of the dopamine D2 receptor increase inflammation and fibrosis in human renal proximal tubule cells. Hypertension 2014; 63: e74–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.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; 4:196–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Fang YJ, Thomas GN, Xu ZL, Fang JQ, Critchley JA, Tomlinson B. An affected pedigree member analysis of linkage between the dopamine D2 receptor gene TaqI polymorphism and obesity and hypertension. Int J Cardiol 2005; 102: 111–116. [DOI] [PubMed] [Google Scholar]
  • 175.Duan J, Wainwright MS, Comeron JM, Saitou N, Sanders AR, Gelernter J et al. Synonymous mutations in the human dopamine receptor D2 (DRD2) affect mRNA stability and synthesis of the receptor. Hum Mol Genet 2003; 12: 205–216. [DOI] [PubMed] [Google Scholar]
  • 176.Thompson J, Thomas N, Singleton A, Piggott M, Lloyd S, Perry EK et al. D2 dopamine receptor gene (DRD2) Taq1 A polymorphism: reduced dopamine D2 receptor binding in the human striatum associated with the A1 allele. Pharmacogenetics 1997; 7: 479–484. [DOI] [PubMed] [Google Scholar]
  • 177.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: 5741–5749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Johnson TL, Tulis DA, Keeler BE, Virag JA, Lust RM, Clemens S. The dopamine D3 receptor knockout mouse mimics aging-related changes in autonomic function and cardiac fibrosis. PLoS One 2013; 8: e74116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Asico LD, Ladines C, Fuchs S, Accili D, Carey RM, Semeraro C et al. Disruption of the dopamine D3 receptor gene produces renin-dependent hypertension. J Clin Invest 1998; 102: 493–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wang X, Escano CS, Asico L, et al. Upregulation of renal D5 dopamine receptor ameliorates the hypertension in D3 dopamine receptor-deficient mice. Hypertension. 2013: 62: 295–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Shimada S, Hirabayashi M, Ishige K, Kosuge Y, Kihara T, Ito Y. Activation of dopamine D4 receptors is protective against hypoxia/reoxygenation-induced cell death in HT22 cells. J Pharmacol Sci 2010; 114:217–224. [DOI] [PubMed] [Google Scholar]
  • 182.Korobochka R, Gritsenko I, Gonen R, Ebstein RP, Ohel G. Association between a functional dopamine D4 receptor promoter region polymorphism (-C521T) and pre-eclampsia: a family-based study. Mol Hum Reprod. 2006; 12: 85–88. [DOI] [PubMed] [Google Scholar]
  • 183.Sen S, Nesse R, Sheng L, Stoltenberg SF, Gleiberman L, Burmeister M, Weder AB. Association between a dopamine-4 receptor polymorphism and blood pressure. Am J Hypertens. 2005; 18: 1206–1210. [DOI] [PubMed] [Google Scholar]
  • 184.Norman SM, Sullivan KM, Liu F, et al. Blood Pressure and Heart Rate Changes During Clozapine Treatment. Psychiatr Q. 2017; 88: 545–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Asico L, Zhang X, Jiang J, et al. Lack of renal dopamine D5 receptors promotes hypertension. J Am Soc Nephrol 2011; 22: 82–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Hollon TR, Bek MJ, Lachowicz JE, et al. Mice lacking D5 dopamine receptors have increased sympathetic tone and are hypertensive. J Neurosci. 2002; 22: 10801–10810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Yang Z, Asico LD, Yu P, el al. D5 dopamine receptor regulation of reactive oxygen species production, NADPH oxidase, and blood pressure. Am J Physiol Regul Integr Comp Physiol. 2006; 290: R96–R104. [DOI] [PubMed] [Google Scholar]
  • 188.Lu Q, Yang Y, Villar VA, et al. D5 dopamine receptor decreases NADPH oxidase, reactive oxygen species and blood pressure via heme oxygenase-1. Hypertens Res. 2013; 36: 684–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Kawakami T, Puri N, Sodhi K, Bellner L, Takahashi T, Morita K et al. Reciprocal effects of oxidative stress on heme oxygenase expression and activity contributes to reno-vascular abnormalities in EC-SOD knockout mice. Int J Hypertens 2012; 2012: 740203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Naganuma T, Nakayama T, Sato N et al. A haplotype-based case-control study examining human extracellular superoxide dismutase gene and essential hypertension. Hypertens Res 2008; 31: 1533–1540. [DOI] [PubMed] [Google Scholar]
  • 191.Park SY, Lee HJ, Ji SM, Kim ME, Jigden B, Lim JE et al. ANTXR2 is a potential causative gene in the genome-wide association study of the blood pressure locus 4q21. Hypertens Res 2014; 37: 811–817. [DOI] [PubMed] [Google Scholar]
  • 192.Rhee MY, Yang SJ, Oh SW et al. Novel genetic variations associated with salt sensitivity in the Korean population. Hypertens Res 2011; 34: 606–611. [DOI] [PubMed] [Google Scholar]
  • 193.Ren Y, Jiao X, Zhang L. Expression level of fibroblast growth factor 5 (FGF5) in the peripheral blood of primary hypertension and its clinical significance. Saudi J Biol Sci. 2018; 25: 469–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Ardanaz N, Yang XP, Cifuentes ME, Haurani MJ, Jackson KW, Liao TD et al. Lack of glutathione peroxidase 1 accelerates cardiac-specific hypertrophy and dysfunction in angiotensin II hypertension. Hypertension. 2010; 55: 116–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Hao Y, Wu BG, Shi J, Chen YL, Sun ZQ, Zheng LQ et al. Association of tag SNPs of GPx-3 with essential hypertension in rural Han Chinese in Fuxin, Liaoning, China. Chin Med J (Engl) 2011; 124: 2113–2116. [PubMed] [Google Scholar]
  • 196.Polonikov AV, Vialykh EK, Churnosov MI, Illig T, Freidin MB, Vasil’eva OV et al. The C718T polymorphism in the 3’-untranslated region of glutathione peroxidase-4 gene is a predictor of cerebral stroke in patients with essential hypertension. Hypertens Res 2012; 35: 507–512. [DOI] [PubMed] [Google Scholar]
  • 197.Vaziri ND, Wang XQ, Oveisi F, Rad B. Induction of oxidative stress by glutathione depletion causes severe hypertension in normal rats. Hypertension. 2000; 36: 142–146. [DOI] [PubMed] [Google Scholar]
  • 198.Polimanti R, Piacentini S, Lazzarin N, Re MA, Manfellotto D, Fuciarelli M. Glutathione S-transferase variants as risk factor for essential hypertension in Italian patients. Mol Cell Biochem 2011; 357: 227–233. [DOI] [PubMed] [Google Scholar]
  • 199.Han JH, Lee HJ, Choi HJ, Yun KE, Kang MH. Lymphocyte DNA damage and plasma antioxidant status in Korean subclinical hypertensive patients by glutathione S-transferase polymorphism. Nutr Res Pract 2017; 11: 214–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Xi B, Shen Y, Reilly KH, Wang X, Mi J. Recapitulation of four hypertension susceptibility genes (CSK, CYP17A1, MTHFR, and FGF5) in East Asians. Metabolism 2013; 62: 196–203. [DOI] [PubMed] [Google Scholar]
  • 201.Canto P, Canto-Cetina T, Juárez-Velázquez R, Rosas-Vargas H, Rangel-Villalobos H, Canizales-Quinteros S, Velázquez-Wong AC et al. Methylenetetrahydrofolate reductase C677T and glutathione S-transferase P1 A313G are associated with a reduced risk of preeclampsia in Maya-Mestizo women. Hypertens Res 2008: 31, 1015–1019. [DOI] [PubMed] [Google Scholar]
  • 202.Cruz-Gonzalez I, Corral E, Sanchez-Ledesma M, Sanchez-Rodriguez A, Martin-Luengo C, Gonzalez-Sarmiento R. An association between resistant hypertension and the null GSTM1 genotype. J Hum Hypertens 2009; 23 :556–558. [DOI] [PubMed] [Google Scholar]
  • 203.Liu J1, Li M, Qi ML,et al. [Association of −63A/C polymorphism of glutathione S-transferase M3 gene with essential hypertension in Chinese population]. [Article in Chinese] Zhonghua Yi Xue Yi Chuan Xue Za Zhi 2007; 24: 582–585. [PubMed] [Google Scholar]
  • 204.Ndisang JF, Zhao W, Wang R. Selective regulation of blood pressure by heme oxygenase-1 in hypertension. Hypertension 2002; 40, 315–321. [DOI] [PubMed] [Google Scholar]
  • 205.Wenzel P, Rossmann H, Müller C, et al. Heme oxygenase-1 suppresses a pro-inflammatory phenotype in monocytes and determines endothelial function and arterial hypertension in mice and humans. Eur Heart J. 2015; 36: 3437–3446. [DOI] [PubMed] [Google Scholar]
  • 206.Daenen KE, Martens P, Bammens B. Association of HO-1 (GT)n Promoter Polymorphism and Cardiovascular Disease: A Reanalysis of the Literature. Can J Cardiol. 2016; 32: 160–168. [DOI] [PubMed] [Google Scholar]
  • 207.Stout JM, Gousset MU, Drummond HA, Gray W 3rd, Pruett BE, Stec DE. Sex-specific effects of heme oxygenase-2 deficiency on renovascular hypertension. J Am Soc Hypertens 2013; 7: 328–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Tornavaca O, Pascual G, Barreiro ML, Grande MT, Carretero A, Riera M et al. Kidney androgen-regulated protein transgenic mice show hypertension and renal alterations mediated by oxidative stress. Circulation 2009; 119: 1908–1917. [DOI] [PubMed] [Google Scholar]
  • 209.de Quixano BB, Villena JA, Aranda M, et al. Kidney Androgen-Regulated Protein (KAP) Transgenic Mice Are Protected Against High-Fat Diet Induced Metabolic Syndrome. Sci Rep. 2017; 7: 16102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Grande MT, Pascual G, Riolobos AS, Clemente-Lorenzo M, Bardaji B, Barreiro L et al. Increased oxidative stress, the renin-angiotensin system, and sympathetic overactivation induce hypertension in kidney androgen-regulated protein transgenic mice. Free Radic Biol Med 2011; 51:1831–1841. [DOI] [PubMed] [Google Scholar]
  • 211.Cheng J, Tao F, Liu Y, Venners SA, Hsu YH, Jiang et al. Associations of methylenetetrahydrofolate reductase C677T genotype with blood pressure levels in Chinese population with essential hypertension. Clin Exp Hypertens. 2018; 40: 207–212. [DOI] [PubMed] [Google Scholar]
  • 212.Dell’Omo G, Penno G, Pucci L, Lucchesi D, Del Prato S, Pedrinelli R. Q192R Paraoxonase (PON)1 polymorphism, insulin sensitivity, and endothelial function in essential hypertensive men. Clin Med Insights Cardiol 2014; 8: 57–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Turgut Cosan D, Colak E, Saydam F, Yazıcı HU, Degirmenci I, Birdane A, et al. Association of paraoxonase 1 (PON1) gene polymorphisms and concentration with essential hypertension. Clin Exp Hypertens. 2016; 38: 602–607. [DOI] [PubMed] [Google Scholar]
  • 214.Zhao Q, Zhang J, Wang H. PGC-1α overexpression suppresses blood pressure elevation in DOCA-salt hypertensive mice. Biosci Rep 2015; 21: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Oberkofler H, Hölzl B, Esterbauer H, Xie M, Iglseder B, Krempler F, Paulweber B et al. Peroxisome proliferator-activated receptor-gamma coactivator-1 gene locus: associations with hypertension in middle-aged men. Hypertension 2003; 41: 368–372. [DOI] [PubMed] [Google Scholar]
  • 216.Cheurfa N, Reis AF, Dubois-Laforgue D, Bellanné-Chantelot C, Timsit J, Velho G. The Gly482Ser polymorphism in the peroxisome proliferator-activated receptor-gamma coactivator-1 gene is associated with hypertension in type 2 diabetic men. Diabetologia 2004; 47: 1980–1983. [DOI] [PubMed] [Google Scholar]
  • 217.Andersen G, Wegner L, Jensen DP, Glümer C, Tarnow L, Drivsholm T et al. PGC-1alpha Gly482Ser polymorphism associates with hypertension among Danish whites. Hypertension 2005; 45: 565–570. [DOI] [PubMed] [Google Scholar]
  • 218.Yang Y, Cuevas S, Yang S, Villar VA, Escano C, Asico L et al. Sestrin2 decreases renal oxidative stress, lowers blood pressure, and mediates dopamine D2 receptor-induced inhibition of reactive oxygen species production. Hypertension 2014; 64: 825–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Jin K, Vaziri ND. Salt-sensitive hypertension in mitochondrial superoxide dismutase deficiency is associated with intra-renal oxidative stress and inflammation. Clin Exp Nephrol 2014; 18: 445–452. [DOI] [PubMed] [Google Scholar]
  • 220.Kandinov B, Drory VE, Tordjman K, Korczyn AD. Blood pressure measurements in a transgenic SOD1-G93A mouse model of amyotrophic lateral sclerosis. Amyotroph Lateral Scler 2012; 13: 509–513. [DOI] [PubMed] [Google Scholar]
  • 221.Carlström M, Lai EY, Ma Z, Steege A, Patzak A, Eriksson UJ et al. Superoxide dismutase 1 limits renal microvascular remodeling and attenuates arteriole and blood pressure responses to angiotensin II via modulation of nitric oxide bioavailability. Hypertension 2010; 56: 907–913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Wakisaka Y, Chu Y, Miller JD, Rosenberg GA, Heistad DD. Critical role for copper/zinc-superoxide dismutase in preventing spontaneous intracerebral hemorrhage during acute and chronic hypertension in mice. Stroke 2010; 41: 790–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Zhang XM1, Ellis EF. Superoxide dismutase decreases mortality, blood pressure, and cerebral blood flow responses induced by acute hypertension in rats. Stroke. 1991; 22: 489–494. [DOI] [PubMed] [Google Scholar]
  • 224.Collister JP, Taylor-Smith H, Drebes D, Nahey D, Tian J, Zimmerman MC. Angiotensin II-Induced Hypertension Is Attenuated by Overexpressing Copper/Zinc Superoxide Dismutase in the Brain Organum Vasculosum of the Lamina Terminalis. Oxid Med Cell Longev. 2016; 2016: 3959087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Lopes RA, Neves KB, Tostes RC, Montezano AC, Touyz RM. Downregulation of Nuclear Factor Erythroid 2-Related Factor and Associated Antioxidant Genes Contributes to Redox-Sensitive Vascular Dysfunction in Hypertension. Hypertension 2015; 66: 1240–1250. [DOI] [PubMed] [Google Scholar]
  • 226.Hatcher J, Gu H, Cheng ZJ. SOD1 Overexpression Preserves Baroreflex Control of Heart Rate with an Increase of Aortic Depressor Nerve Function. Oxid Med Cell Longev 2016; 2016: 3686829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W et al. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ Res 2010; 107: 106–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Kang PT, Chen CL, Ohanyan V, Luther DJ, Meszaros JG, Chilian WM et al. Overexpressing superoxide dismutase 2 induces a supernormal cardiac function by enhancing redox-dependent mitochondrial function and metabolic dilation. J Mol Cell Cardiol 2015; 88: 14–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Hodgson JM, Croft KD, Woodman RJ, Puddey IB, Bondonno CP, Wu JH, et al. Effects of vitamin E, vitamin C and polyphenols on the rate of blood pressure variation: results of two randomised controlled trials. Br J Nutr. 2014; 112 :1551–1561. [DOI] [PubMed] [Google Scholar]
  • 230.Mansego ML, Solar Gde M, Alonso MP, Martínez F, Sáez GT, Escudero JC et al. Polymorphisms of antioxidant enzymes, blood pressure and risk of hypertension. J Hypertens 2011; 29: 492–500. [DOI] [PubMed] [Google Scholar]
  • 231.Jonsson LM, Rees DD, Edlund T, Marklund SL. Nitric oxide and blood pressure in mice lacking extracellular-superoxide dismutase. Free Radic Res 2002; 36: 755–758. [DOI] [PubMed] [Google Scholar]
  • 232.Lob HE, Vinh A, Li L, Blinder Y, Offermanns S, Harrison DG. Role of vascular extracellular superoxide dismutase in hypertension. Hypertension 2011; 58: 232–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Dong X, Li D, Liu H, Zhao Y. SOD3 and eNOS genotypes are associated with SOD activity and NOx. Exp Ther Med 2014; 8: 328–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Yoshioka J, Imahashi K, Gabel SA, Chutkow WA, Burds AA, Gannon J et al. Targeted deletion of thioredoxin-interacting protein regulates cardiac dysfunction in response to pressure overload. Circ Res 2007; 101: 1328–1338. [DOI] [PubMed] [Google Scholar]
  • 235.Kiermayer C, Northrup E, Schrewe A, Walch A, de Angelis MH, Schoensiegel F et al. Heart-specific knockout of the mitochondrial thioredoxin reductase (Txnrd2) induces metabolic and contractile dysfunction in the aging myocardium. J Am Heart Assoc 2015; 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Rubattu S, Bianchi F, Busceti CL, Cotugno M, Stanzione R, Marchitti S et al. Differential modulation of AMPK/PPARα/UCP2 axis in relation to hypertension and aging in the brain, kidneys and heart of two closely related spontaneously hypertensive rat strains. Oncotarget 2015; 6: 18800–18818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Liu L, Liu J, Tian XY, Wong WT, Lau CW, Xu A et al. Uncoupling protein-2 mediates DPP-4 inhibitor-induced restoration of endothelial function in hypertension through reducing oxidative stress. Antioxid Redox Signal 2014; 21: 1571–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Di Castro S, Scarpino S, Marchitti S, Bianchi F, Stanzione R, Cotugno M et al. Differential modulation of uncoupling protein 2 in kidneys of stroke-prone spontaneously hypertensive rats under high-salt/low-potassium diet. Hypertension 2013; 61: 534–541. [DOI] [PubMed] [Google Scholar]
  • 239.Ji Q, Ikegami H, Fujisawa T, Kawabata Y, Ono M, Nishino M et al. A common polymorphism of uncoupling protein 2 gene is associated with hypertension. J Hypertens 2004; 22: 97–102. [DOI] [PubMed] [Google Scholar]
  • 240.Scheepers LE, Wei FF, Stolarz-Skrzypek K, Malyutina S, Tikhonoff V, Thijs L, et al. Xanthine oxidase gene variants and their association with blood pressure and incident hypertension: a population study. J Hypertens. 2016; 34: 2147–2154. [DOI] [PubMed] [Google Scholar]
  • 241.Khambata RS, Ghosh SM, Ahluwalia A. “Repurposing” of xanthine oxidoreductase as a nitrite reductase: A new paradigm for therapeutic targeting in hypertension. Antioxid Redox Signal 2015; 23: 340–353. [DOI] [PubMed] [Google Scholar]
  • 242.Chaves FJ, Corella D, Blesa S, Mansego ML, Marín P, Portoles O et al. Xanthine oxidoreductase polymorphisms: influence in blood pressure and oxidative stress levels. Pharmacogenet Genomics 2007; 17: 589–596. [DOI] [PubMed] [Google Scholar]
  • 243.Pechánová O, Zicha J, Kojsová S, Dobesová Z, Jendeková L, Kunes J. Effect of chronic N-acetylcysteine treatment on the development of spontaneous hypertension. Clin Sci (Lond) 2006; 110: 235–242. [DOI] [PubMed] [Google Scholar]
  • 244.Martina V, Masha A, Gigliardi VR, Brocato L, Manzato E, Berchio A et al. Long-term N-acetylcysteine and L-arginine administration reduces endothelial activation and systolic blood pressure in hypertensive patients with type 2 diabetes. Diabetes Care 2008; 31: 940–94. [DOI] [PubMed] [Google Scholar]
  • 245.Renke M, Tylicki L, Rutkowski P, Larczynski W, Neuwelt A, Aleksandrowicz E et al. The effect of N-acetylcysteine on blood pressure and markers of cardiovascular risk in non-diabetic patients with chronic kidney disease: a placebo-controlled, randomized, cross-over study. Med Sci Monit 2010; 16: PI13–118. [PubMed] [Google Scholar]
  • 246.Wang LP, Fan SJ, Li SM, Wang XJ, Gao JL, Yang XH. Oxidative stress promotes myocardial fibrosis by upregulating KCa3.1 channel expression in AGT-REN double transgenic hypertensive mice. Pflugers Arch 2017; 469: 1061–1071. [DOI] [PubMed] [Google Scholar]
  • 247.Aldini G, Altomare A, Baron G, Vistoli G, Carini M, Borsani L, Sergio F. N-Acetylcysteine as an antioxidant and disulphide breaking agent: the reasons why. Free Radic Res. 2018. May 9:1–12. doi: 10.1080/10715762.2018.1468564 [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 248.Zhang Y, Cuevas S, Asico LD, Escano C, Yang Y, Pascua AM et al. Deficient dopamine D2 receptor function causes renal inflammation independently of high blood pressure. PLoS One 2012; 7: e38745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Potje SR, Troiano JA, Graton ME, Ximenes VF, Nakamune AC, Antoniali C. Hypotensive and vasorelaxant effect of Diapocynin in normotensive rats. Free Radic Biol Med 2017; 106: 148–157. [DOI] [PubMed] [Google Scholar]
  • 250.Bhatia K, Elmarakby AA, El-Remessy AB, Sullivan JC. Oxidative stress contributes to sex differences in angiotensin II-mediated hypertension in spontaneously hypertensive rats. Am J Physiol Regul Integr Comp Physiol. 2012; 302 :R274–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Li Z, Wang Y, Man RY, Vanhoutte PM. Upregulation of heme oxygenase-1 potentiates EDH-type relaxations in the mesenteric artery of the spontaneously hypertensive rat. Am J Physiol Heart Circ Physiol 2013; 305: H1471–1483. [DOI] [PubMed] [Google Scholar]
  • 252.Armando I, Wang X, Villar VA, Jones JE, Asico LD, Escano C et al. Reactive oxygen species-dependent hypertension in dopamine D2 receptor-deficient mice. Hypertension 2007; 49: 672–678. [DOI] [PubMed] [Google Scholar]
  • 253.Wang HP, Yang J, Qin LQ, Yang XJ. Effect of garlic on blood pressure: a meta-analysis. J Clin Hypertens (Greenwich). 2015; 17: 223–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Ried K Garlic Lowers Blood Pressure in Hypertensive Individuals, Regulates Serum Cholesterol, and Stimulates Immunity: An Updated Meta-analysis and Review. J Nutr. 2016; 146: 389S–396S. [DOI] [PubMed] [Google Scholar]
  • 255.Dhawan V, Jain S. Garlic supplementation prevents oxidative DNA damage in essential hypertension. Mol Cell Biochem. 2005; 275: 85–94. [DOI] [PubMed] [Google Scholar]
  • 256.Castro C, Lorenzo AG, González A, Cruzado M. Garlic components inhibit angiotensin II-induced cell-cycle progression and migration: Involvement of cell-cycle inhibitor p27(Kip1) and mitogen-activated protein kinase. Mol Nutr Food Res. 2010; 54: 781–787 [DOI] [PubMed] [Google Scholar]
  • 257.Siani A, Pagano E, Iacone R, Iacoviello L, Scopacasa F, Strazzullo P. Blood pressure and metabolic changes during dietary L-arginine supplementation in humans. Am J Hypertens 2000; 13: 547–551. [DOI] [PubMed] [Google Scholar]
  • 258.Rajapakse NW, Karim F, Evans RG, Kaye DM, Head GA. Augmented endothelial-specific L-Arginine transport blunts the contribution of the sympathetic nervous system to obesity induced hypertension in mice. PLoS One 2015; 10: e0131424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Vanhoutte PM, Zhao Y, Xu A, Leung SW Thirty Years of Saying NO: Sources, Fate, Actions, and Misfortunes of the Endothelium-Derived Vasodilator Mediator. Circ Res. 2016; 119: 375–396. [DOI] [PubMed] [Google Scholar]
  • 260.Zoja C, Corna D, Nava V, Locatelli M, Abbate M, Gaspari F et al. Analogs of bardoxolone methyl worsen diabetic nephropathy in rats with additional adverse effects. Am J Physiol Renal Physiol 2013; 304: F808–819. [DOI] [PubMed] [Google Scholar]
  • 261.de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H et al. BEACON Trial Investigators. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013; 369: 2492–2503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Blanca AJ, Ruiz-Armenta MV, Zambrano S, Miguel-Carrasco JL, Arias JL, Arévalo M et al. Inflammatory and fibrotic processes are involved in the cardiotoxic effect of sunitinib: Protective role of l-carnitine. Toxicol Lett 2016; 241: 9–18. [DOI] [PubMed] [Google Scholar]
  • 263.Graham D, Huynh NN, Hamilton CA, Beattie E, Smith RA, Cochemé HM et al. Mitochondria-targeted antioxidant MitoQ10 improves endothelial function and attenuates cardiac hypertrophy. Hypertension 2009; 54: 322–328. [DOI] [PubMed] [Google Scholar]
  • 264.Ruggenenti P, Cattaneo D, Loriga G, Ledda F, Motterlini N, Gherardi G, Orisio S, Remuzzi G. Ameliorating hypertension and insulin resistance in subjects at increased cardiovascular risk: effects of acetyl-L-carnitine therapy. Hypertension 2009; 54: 567–574. [DOI] [PubMed] [Google Scholar]
  • 265.Takahashi R, Asai T, Murakami H, Murakami R, Tsuzuki M, Numaguchi Y, Matsui H, Murohara T, Okumura K. Pressure overload-induced cardiomyopathy in heterozygous carrier mice of carnitine transporter gene mutation. Hypertension 2007; 50: 497–450. [DOI] [PubMed] [Google Scholar]
  • 266.Mingorance C, Duluc L, Chalopin M, Simard G, Ducluzeau PH, Herrera MD et al. Propionyl-L-carnitine corrects metabolic and cardiovascular alterations in diet-induced obese mice and improves liver respiratory chain activity. PLoS One 2012; 7: e34268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.San Cheang W, Yuen Ngai C, Yen Tam Y,et al. Black tea protects against hypertension-associated endothelial dysfunction through alleviation of endoplasmic reticulum stress. Sci Rep. 2015; 5: 10340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Serban C, Sahebkar A, Ursoniu S, Andrica F, Banach M. Effect of sour tea (Hibiscus sabdariffa L.) on arterial hypertension: a systematic review and meta-analysis of randomized controlled trials. J Hypertens. 2015; 33: 1119–1127. [DOI] [PubMed] [Google Scholar]
  • 269.Sugita M, Kapoor MP, Nishimura A, Okubo T. Influence of green tea catechins on oxidative stress metabolites at rest and during exercise in healthy humans. Nutrition. 2016; 32: 321–331. [DOI] [PubMed] [Google Scholar]
  • 270.Burke BE, Neuenschwander R, Olson RD. Randomized, double-blind, placebo-controlled trial of coenzyme Q10 in isolated systolic hypertension. South Med J 2001; 94: 1112–1117. [DOI] [PubMed] [Google Scholar]
  • 271.Singh RB, Niaz MA, Rastogi SS, Shukla PK, Thakur AS. Effect of hydrosoluble coenzyme Q10 on blood pressures and insulin resistance in hypertensive patients with coronary artery disease. J Hum Hypertens 1999; 13: 203–208. [DOI] [PubMed] [Google Scholar]
  • 272.Luna-Sánchez M, Hidalgo-Gutiérrez A, Hildebrandt TM, Chaves-Serrano J, Barriocanal-Casado E, Santos-Fandila Á et al. CoQ deficiency causes disruption of mitochondrial sulfide oxidation, a new pathomechanism associated with this syndrome. EMBO Mol Med 2017; 9: 78–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Ankola DD, Viswanad B, Bhardwaj V, Ramarao P, Kumar MN. Development of potent oral nanoparticulate formulation of coenzyme Q10 for treatment of hypertension: can the simple nutritional supplements be used as first line therapeutic agents for prophylaxis/therapy? Eur J Pharm Biopharm 2007; 67: 361–369. [DOI] [PubMed] [Google Scholar]
  • 274.Nakmareong S, Kukongviriyapan U, Pakdeechote P, Kukongviriyapan V, Kongyingyoes B, Donpunha W, Prachaney P, Phisalaphong C. Tetrahydrocurcumin alleviates hypertension, aortic stiffening and oxidative stress in rats with nitric oxide deficiency. Hypertens Res 2012; 35: 418–425. [DOI] [PubMed] [Google Scholar]
  • 275.Kukongviriyapan U, Pannangpetch P, Kukongviriyapan V, Donpunha W, Sompamit K, Surawattanawan P. Curcumin protects against cadmium-induced vascular dysfunction, hypertension and tissue cadmium accumulation in mice. Nutrients 2014; 21: 1194–1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Cicero AFG, Fogacci F, Colletti A. Food and plant bioactives for reducing cardiometabolic disease risk: an evidence based approach. Food Funct. 2017. June; 8: 2076–2088. [DOI] [PubMed] [Google Scholar]
  • 277.Wang S, Ye Q, Tu J, Zhang M, Ji B. Curcumin protects against hypertension aggravated retinal ischemia/reperfusion in a rat stroke model. Clin Exp Hypertens. 2017; 39: 711–717. [DOI] [PubMed] [Google Scholar]
  • 278.El-Bassossy HM, Hassan N, Zakaria MN. Heme oxygenase-1 alleviates vascular complications associated with metabolic syndrome: Effect on endothelial dependent relaxation and NO production. Chem Biol Interact 2014; 223: 109–115. [DOI] [PubMed] [Google Scholar]
  • 279.Sirivarasai J, Kaojarern S, Chanprasertyothin S, Panpunuan P, Petchpoung K, et al. Environmental lead exposure, catalase gene, and markers of antioxidant and oxidative stress relation to hypertension: an analysis based on the EGAT study. Biomed Res Int. 2015; 2015: 856319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Lu Q, Yang Y, Villar VA, Asico L, Jones JE, Yu P et al. D5 dopamine receptor decreases NADPH oxidase, reactive oxygen species and blood pressure via heme oxygenase-1. Hypertens Res 2013; 36: 684–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Parhiz H, Roohbakhsh A, Soltani F, Rezaee R, Iranshahi M. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: an updated review of their molecular mechanisms and experimental models. Phytother Res 2015; 29: 323–331. [DOI] [PubMed] [Google Scholar]
  • 282.Wunpathe C, Potue P, Maneesai P, et al. Hesperidin Suppresses Renin-Angiotensin System Mediated NOX2 Over-Expression and Sympathoexcitation in 2K-1C Hypertensive Rats. Am J Chin Med. 2018: 13: 1–17. [DOI] [PubMed] [Google Scholar]
  • 283.Salden BN, Troost FJ, de Groot E, et al. Randomized clinical trial on the efficacy of hesperidin 2S on validated cardiovascular biomarkers in healthy overweight individuals. Am J Clin Nutr. 2016; 104: 1523–1153. [DOI] [PubMed] [Google Scholar]
  • 284.Koçak G, Aktan F, Canbolat O, Ozoğul C, Elbeğ S, Yildizoglu-Ari N et al. Alpha-lipoic acid treatment ameliorates metabolic parameters, blood pressure, vascular reactivity and morphology of vessels already damaged by streptozotocin-diabetes. Diabetes Nutr Metab 2000; 13: 308–318. [PubMed] [Google Scholar]
  • 285.Mohammadi V, Dehghani S, Askari G. Does alpha-lipoic acid supplement regulate blood pressure? A systematic review of randomized, double-blind placebo-controlled clinical trials. Int J Prev Med 2017; 8: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.de Queiroz TM, Xia H, Filipeanu CM, Braga VA, Lazartigues E. α-Lipoic acid reduces neurogenic hypertension by blunting oxidative stress-mediated increase in ADAM17. Am J Physiol Heart Circ Physiol 2015; 309: H926–934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Grossman E, Laudon M, Zisapel N. Effect of melatonin on nocturnal blood pressure: meta-analysis of randomized controlled trials. Vasc Health Risk Manag 2011; 7: 577–584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Rahman MM, Kwon HS, Kim MJ, Go HK, Oak MH, Kim DH. Melatonin supplementation plus exercise behavior ameliorate insulin resistance, hypertension and fatigue in a rat model of type 2 diabetes mellitus. Biomed Pharmacother 2017; 92: 606–614. [DOI] [PubMed] [Google Scholar]
  • 289.Klimentova J, Cebova M, Barta A, Matuskova Z, Vrankova S, Rehakova R et al. Effect of melatonin on blood pressure and nitric oxide generation in rats with metabolic syndrome. Physiol Res 2016; 65: S373–380. [DOI] [PubMed] [Google Scholar]
  • 290.Qiao YF, Guo WJ, Li L, Shao S, Qiao X, Shao JJ et la. Melatonin attenuates hypertension-induced renal injury partially through inhibiting oxidative stress in rats. Mol Med Rep 2016; 13: 21–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Plotnikov MB, Aliev OI, Sidekhmenova AV, Shamanaev AY, Anishchenko AM, Nosarev AV et al. Modes of hypotensive action of dihydroquercetin in arterial hypertension. Bull Exp Biol Med 2017; 162: 353–356. [DOI] [PubMed] [Google Scholar]
  • 292.Gormaz JG, Quintremil S, Rodrigo R. Cardiovascular Disease: A target for the pharmacological effects of quercetin. Curr Top Med Chem 2015; 15: 1735–1742. [DOI] [PubMed] [Google Scholar]
  • 293.Egert S, Bosy-Westphal A, Seiberl J, Kurbitz C, Settler U, Plachta-Danielzik S et al. Quercetin reduces systolic blood pressure and plasma oxidised lowdensity lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: a double-blinded, placebo-controlled cross-over study. Br J Nutr 2009; 102: 1065–1074. [DOI] [PubMed] [Google Scholar]
  • 294.Ko WC, Shih CM, Chen MC, Lai YH, Chen JH, Chen CM et al. Suppressive effects of 3-O-methylquercetin on ovalbumin-induced airway hyperresponsiveness. Planta Med 2004; 70: 1123–1127. [DOI] [PubMed] [Google Scholar]
  • 295.Dolinsky VW, Chakrabarti S, Pereira TJ, Oka T, Levasseur J, Beker D et al. Resveratrol prevents hypertension and cardiac hypertrophy in hypertensive rats and mice. Biochim Biophys Acta 2013; 1832: 1723–1733. [DOI] [PubMed] [Google Scholar]
  • 296.Tain YL, Lin YJ, Sheen JM, et al. Resveratrol prevents the combined maternal plus postweaning high-fat-diets-induced hypertension in male offspring. J Nutr Biochem. 2017; 48: 120–127. [DOI] [PubMed] [Google Scholar]
  • 297.Diao Z, Asico LD, Villar VAM, et al. Increased renal oxidative stress in salt-sensitive human GRK4γ486V transgenic mice. Free Radic Biol Med. 2017; 106: 80–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Geetha R, Yogalakshmi B, Sreeja S, Bhavani K, Anuradha CV. Troxerutin suppresses lipid abnormalities in the heart of high-fat-high-fructose diet-fed mice. Mol Cell Biochem 2014; 387: 123–134. [DOI] [PubMed] [Google Scholar]
  • 299.Yu Y, Zheng G. Troxerutin protects against diabetic cardiomyopathy through NF-κB/AKT/IRS1 in a rat model of type 2 diabetes. Mol Med Rep 2017; 15: 3473–3478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Jaja SI, Ogungbemi SI, Kehinde MO. Electrocardiographic and blood pressure changes in apparently healthy male subjects following oral, chronic, low-dose vitamin C supplementation and/or change in posture. Nig Q J Hosp Med 2008; 18: 96–100. [DOI] [PubMed] [Google Scholar]
  • 301.Chen X, Touyz RM, Park JB, Schiffrin EL. Antioxidant effects of vitamins C and E are associated with altered activation of vascular NADPH oxidase and superoxide dismutase in stroke-prone SHR. Hypertension 2001; 38: 606–611. [DOI] [PubMed] [Google Scholar]
  • 302.Bruno RM, Daghini E, Ghiadoni L, Sudano I, Rugani I, Varanini M, Passino C, Emdin M, Taddei S. Effect of acute administration of vitamin C on muscle sympathetic activity, cardiac sympathovagal balance, and baroreflex sensitivity in hypertensive patients. Am J Clin Nutr 2012; 96: 302–308. [DOI] [PubMed] [Google Scholar]
  • 303.Rodrigo R, Prat H, Passalacqua W, Araya J, Bächler JP. Decrease in oxidative stress through supplementation of vitamins C and E is associated with a reduction in blood pressure in patients with essential hypertension. Clin Sci (Lond) 2008; 114: 625–634. [DOI] [PubMed] [Google Scholar]
  • 304.Donpunha W, Kukongviriyapan U, Sompamit K, Pakdeechote P, Kukongviriyapan V, Pannangpetch P. Protective effect of ascorbic acid on cadmium-induced hypertension and vascular dysfunction in mice. Biometals 2011; 24: 105–115. [DOI] [PubMed] [Google Scholar]
  • 305.Ahmad KA, Yuan Yuan D, Nawaz W, Ze H, Zhuo CX, Talal B et al. Antioxidant therapy for management of oxidative stress induced hypertension. Free Radic Res 2017; 51:428–438. [DOI] [PubMed] [Google Scholar]
  • 306.Juraschek SP, Guallar E, Appel LJ, Miller ER 3rd. Effects of vitamin C supplementation on blood pressure: a meta-analysis of randomized controlled trials. Am J Clin Nutr 2012; 95: 1079–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Witham MD, Nadir MA, Struthers AD. Effect of vitamin D on blood pressure: a systematic review and meta-analysis. J Hypertens 2009; 27: 1948–1954. [DOI] [PubMed] [Google Scholar]
  • 308.Mirhosseini NZ, Knaus SJ, Bohaychuk K, Singh J, Vatanparast HA, Weber LP. Both high and low plasma levels of 25-hydroxy vitamin D increase blood pressure in a normal rat model. Br J Nutr 2016. 116: 1889–1900. [DOI] [PubMed] [Google Scholar]
  • 309.Pilz S, Gaksch M, Kienreich K, Grübler M, Verheyen N, Fahrleitner-Pammer A et al. Effects of vitamin D on blood pressure and cardiovascular risk factors: a randomized controlled trial. Hypertension 2015. 65: 1195–1201. [DOI] [PubMed] [Google Scholar]
  • 310.Ni W, Watts SW, Ng M, Chen S, Glenn DJ, Gardner DG. Elimination of vitamin D receptor in vascular endothelial cells alters vascular function. Hypertension 2014; 64: 1290–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 311.Pelham CJ, Drews EM, Agrawal DK. Vitamin D controls resistance artery function through regulation of perivascular adipose tissue hypoxia and inflammation. J Mol Cell Cardiol 2016; 98: 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 312.Franczyk A, Stolarz-Skrzypek K, Wesołowska A, Czarnecka D. Vitamin D and vitamin D receptor activators in treatment of hypertension and cardiovascular disease. Cardiovasc Hematol Disord Drug Targets 2014; 14: 34–44. [DOI] [PubMed] [Google Scholar]
  • 313.Faulkner JL, Amaral LM, Cornelius DC, Cunningham MW, Ibrahim T, Heep A, Campbell N et al. Vitamin D supplementation reduces some AT1-AA-induced downstream targets implicated in preeclampsia including hypertension. Am J Physiol Regul Integr Comp Physiol 2017; 312: R125–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Beveridge LA, Struthers AD, Khan F, Jorde R, Scragg R, Macdonald HM et al. D-PRESSURE Collaboration. Effect of Vitamin D Supplementation on Blood Pressure: A Systematic Review and Meta-analysis Incorporating Individual Patient Data. JAMA Intern Med 2015; 175: 745–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 315.Trehan N, Afonso L, Levine DL, Levy PD. Vitamin D Deficiency, Supplementation, and Cardiovascular Health. Crit Pathw Cardiol 2017; 16: 109–118. [DOI] [PubMed] [Google Scholar]
  • 316.Al Mheid I, Quyyumi AA. Vitamin D and Cardiovascular Disease: Controversy Unresolved. J Am Coll Cardiol 2017; 70: 89–100. [DOI] [PubMed] [Google Scholar]
  • 317.Legarth C, Grimm D, Wehland M, Bauer J, Krüger M. The Impact of Vitamin D in the Treatment of Essential Hypertension. Int J Mol Sci. 2018; 19: E455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Mirhosseini NZ, Knaus SJ, Bohaychuk K, et al. Both high and low plasma levels of 25-hydroxy vitamin D increase blood pressure in a normal rat model. Br J Nutr. 2016; 116: 1889–1900. [DOI] [PubMed] [Google Scholar]
  • 319.Oh J, Riek AE, Zhang RM, et al. Deletion of JNK2 prevents vitamin-D-deficiency-induced hypertension and atherosclerosis in mice. J Steroid Biochem Mol Biol. 2018; 177: 179–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320.Boshtam M, Rafiei M, Sadeghi K, Sarraf-Zadegan N. Vitamin E can reduce blood pressure in mild hypertensives. Int J Vitam Nutr Res 2002; 72: 309–314. [DOI] [PubMed] [Google Scholar]
  • 321.Barbagallo M, Dominguez LJ, Tagliamonte MR, Resnick LM, Paolisso G. Effects of vitamin E and glutathione on glucose metabolism: role of magnesium. Hypertension 1999; 34: 1002–6. [DOI] [PubMed] [Google Scholar]
  • 322.Wong SK, Chin KY, Suhaimi FH, Ahmad F, Ima-Nirwana S. Vitamin E as a Potential Interventional Treatment for Metabolic Syndrome: Evidence from Animal and Human Studies. Front Pharmacol 2017; 8: 444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323.Azzi A Many tocopherols, one vitamin E. Mol Aspects Med. 2018; 61: 92–103. [DOI] [PubMed] [Google Scholar]
  • 324.Galley HF, Thornton J, Howdle PD, Walker BE, Webster NR. Combination oral antioxidant supplementation reduces blood pressure. Clin Sci (Lond) 1997; 92: 361–365. [DOI] [PubMed] [Google Scholar]
  • 325.Wigley RD, Vlieg M. Alpha tocopherol and blood pressure in PN/n mice. Aust J Exp Biol Med Sci 1978; 56: 631–637. [DOI] [PubMed] [Google Scholar]
  • 326.Saremi A, Arora R. Vitamin E and cardiovascular disease. Am J Ther. 2010; 17 :e56–65. [DOI] [PubMed] [Google Scholar]
  • 327.Hodgson JM, Croft KD, Woodman RJ, Puddey IB, Bondonno CP, Wu JH et al. Effects of vitamin E, vitamin C and polyphenols on the rate of blood pressure variation: results of two randomised controlled trials. Br J Nutr 2014; 112: 1551–1561. [DOI] [PubMed] [Google Scholar]
  • 328.Konety BR, Leman E, Vietmeier B, Arlotti J, Dhir R, Getzenberg RH. In vitro and in vivo effects of vitamin D (calcitriol) administration on the normal neonatal and prepubertal prostate. J Urol 2000; 164: 1812–1818. [PubMed] [Google Scholar]
  • 329.Montezano AC, Touyz RM. Reactive oxygen species, vascular Noxs, and hypertension: focus on translational and clinical research. Antioxid Redox Signal 2014; 20: 164–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330.Ando K, Fujita M. Reactive oxygen species and the central nervous system in salt-sensitive hypertension: possible relationship with obesity-induced hypertension. Clin Exp Pharmacol Physiol 2012; 39: 111–116. [DOI] [PubMed] [Google Scholar]
  • 331.Giordano FJ. Oxygen, oxidative stress, hypoxia, and heart failure. J Clin Invest 2005; 115: 500–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol 2004; 4: 181–189. [DOI] [PubMed] [Google Scholar]
  • 333.Montezano AC, Tsiropoulou S, Dulak-Lis M, Harvey A, Camargo Lde L, Touyz RM. Redox signaling, Nox5 and vascular remodeling in hypertension. Curr Opin Nephrol Hypertens 2015; 24, 425–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Stasia MJ. CYBA encoding p22(phox), the cytochrome b558 alpha polypeptide: gene structure, expression, role and physiopathology. Gene 2016; 586: 27–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335.Wang H, Albadawi H, Siddiquee Z, Stone JM, Panchenko MP, Watkins MT et al. Altered vascular activation due to deficiency of the NADPH oxidase component p22phox. Cardiovasc Pathol 2014; 23: 35–42. [DOI] [PubMed] [Google Scholar]
  • 336.Zalba G, San José G, Beaumont FJ, Fortuño MA, Fortuño A, Díez J et al. Polymorphisms and promoter overactivity of the p22(phox) gene in vascular smooth muscle cells from spontaneously hypertensive rats. Circ Res 2001; 88: 217–222. [DOI] [PubMed] [Google Scholar]
  • 337.Schreiber R, Ferreira-Sae MC, Tucunduva AC, Mill JG, Costa FO, Krieger JE et al. CYBA C242T polymorphism is associated with obesity and diabetes mellitus in Brazilian hypertensive patients. Diabet Med 2012; 29: e55–61. [DOI] [PubMed] [Google Scholar]
  • 338.Rafiq A, Aslam K, Malik R, Afroze D. C242T polymorphism of the NADPH oxidase p22PHOX gene and its association with endothelial dysfunction in asymptomatic individuals with essential systemic hypertension. Mol Med Rep 2014; 9: 1857–1862. [DOI] [PubMed] [Google Scholar]
  • 339.Kumar R, Kohli S, Ali Z, Duhan K, Ram R, Gupta M et al. CYBA (p22phox) variants associate with blood pressure and oxidative stress markers in hypertension: a replication study in populations of diverse altitudes. Hypertens Res 2015; 38: 498–506. [DOI] [PubMed] [Google Scholar]
  • 340.Yu P, Han W, Villar VA, Yang Y, Lu Q, Lee H et al. Unique role of NADPH oxidase 5 in oxidative stress in human renal proximal tubule cells. Redox Biol 2014; 2: 570–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Wang Y, Chen F, Le B, Stepp DW, Fulton DJ. Impact of Nox5 polymorphisms on basal and stimulus-dependent ROS generation. PLoS One 2014; 9: e100102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 342.Holterman CE, Thibodeau JF, Towaij C, Gutsol A, Montezano AC, Parks RJ, et al. Nephropathy and elevated BP in mice with podocyte-specific NADPH oxidase 5 expression. J Am Soc Nephrol 2014; 25: 784–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Yang S, Gao Y, Liu G, Li J, Shi K, Du B et al. The human ATF1 rs11169571 polymorphism increases essential hypertension risk through modifying miRNA binding. FEBS Lett 2015; 589: 2087–2093. [DOI] [PubMed] [Google Scholar]
  • 344.Heimlich JB, Speed JS, O’Connor PM, Pollock JS, Townes TM, Meiler SE, et al. E Endothelin-1 contributes to the progression of renal injury in sickle cell disease via reactive oxygen species. Br J Pharmacol 2016; 173: 386–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345.Gupta RM, Hadaya J, Trehan A, Zekavat SM, Roselli C, Klarin D, et al. A Genetic Variant Associated with Five Vascular Diseases Is a Distal Regulator of Endothelin-1 Gene Expression. Cell 2017; 170: 522–533 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 346.Brennan ML, Anderson MM, Shih DM, Qu XD, Wang X, Mehta AC et al. : Increased atherosclerosis in myeloperoxidase-deficient mice. J Clin Invest 2001: 107; 419–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347.Nishino T, Okamoto K, Eger BT, Pai EF, Nishino T. Mammalian xanthine oxidoreductase - mechanism of transition from xanthine dehydrogenase to xanthine oxidase. FEBS J. 2008; 275: 3278–3289. [DOI] [PubMed] [Google Scholar]
  • 348.Wright RM, Vaitaitis GM, Wilson CM, Repine TB, Terada LS, Repine JE. cDNA cloning, characterization, and tissue-specific expression of human xanthine dehydrogenase/xanthine oxidase. Proc Natl Acad Sci USA 1993; 90: 10690–10694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Van Hee VC, Adar SD, Szpiro AA et al. Common genetic variation, residential proximity to traffic exposure, and left ventricular mass: the multi-ethnic study of atherosclerosis. Environ Health Perspect 2010; 118: 962–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 350.Quintana LF, Guzmán B, Collado S, Clària J, Poch E. A coding polymorphism in the 12-lipoxygenase gene is associated to essential hypertension and urinary 12(S)-HETE. Kidney Int 2006; 69: 526–30. [DOI] [PubMed] [Google Scholar]
  • 351.Kriska T, Cepura C, Magier D, Siangjong L, Gauthier KM, Campbell WB. Mice lacking macrophage 12/15-lipoxygenase are resistant to experimental hypertension. Am J Physiol Heart Circ Physiol. 2012; 302: H2428–2438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Huan T, Esko T, Peters MJ et al. A meta-analysis of gene expression signatures of blood pressure and hypertension. PLoS Genet 2015; 11: e1005035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Staehr M, Hansen PB, Madsen K, Vanhoutte PM, Nüsing RM, Jensen BL. Deletion of cyclooxygenase-2 in the mouse increases arterial blood pressure with no impairment in renal NO production in response to chronic high salt intake. Am J Physiol Regul Integr Comp Physiol. 2013; 304: R899–8907. [DOI] [PubMed] [Google Scholar]
  • 354.Eirin A, Lerman A, Lerman LO. Mitochondria: a pathogenic paradigm in hypertensive renal disease. Hypertension 2015; 65: 264–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Lee H, Abe Y, Lee I, Shrivastav S, Crusan AP, Hüttemann M et al. Increased mitochondrial activity in renal proximal tubule cells from young spontaneously hypertensive rats. Kidney Int 2014; 85: 561–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 356.Dikalova AE, Bikineyeva AT, Budzyn K, Nazarewicz RR, McCann L, Lewis W et al. Therapeutic targeting of mitochondrial superoxide in hypertension. Circ Res 2010. ; 107: 106–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 357.Bajpai P, Srinivasan S, Ghosh J, Nagy LD, Wei S, Guengerich FP, Avadhani NG. Targeting of splice variants of human cytochrome P450 2C8 (CYP2C8) to mitochondria and their role in arachidonic acid metabolism and respiratory dysfunction. J Biol Chem. 2014; 289: 29614–29630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 358.Yan HC, Liu JH, Li J, He BX, Yang L, Qiu J et al. Association between the CYP4A11 T8590C variant and essential hypertension: new data from Han Chinese and a meta-analysis. PLoS One 2013; 8: e80072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Gregoski MJ, Buxbaum SG, Kapuku G, Dong Y, Zhu H, Davis M, Gonto K, Treiber FA. Interactive influences of ethnicity, endothelin-1 gene, and everyday discrimination upon nocturnal ambulatory blood pressure. Ann Behav Med. 2013; 45: 377–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Hermann M, Hellermann JP, Quitzau K. CYP4A11 polymorphism correlates with coronary endothelial dysfunction in patients with coronary artery disease--the ENCORE Trials. Atherosclerosis 2009; 207: 476–479. [DOI] [PubMed] [Google Scholar]
  • 361.Polonikov AV, Ivanov VP, Solodilova MA. CYP2E1 gene promoter polymorphism −1293G>C increases the risk of essential hypertension in men with alcohol abuse. Bull Exp Biol Med 2013; 155: 734–737. [DOI] [PubMed] [Google Scholar]
  • 362.Capdevila J, Wang W. Role of cytochrome P450 epoxygenase in regulating renal membrane transport and hypertension. Curr Opin Nephrol Hypertens 2013; 22: 163–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Huber M, Lezius S, Reibis R et al. A single nucleotide polymorphism near the CYP17A1 gene is associated with left ventricular mass in hypertensive patients under pharmacotherapy. Int J Mol Sci 2015; 16: 17456–17468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 364.Yao B, Harris RC, Zhang MZ. Intrarenal dopamine attenuates deoxycorticosterone acetate/high salt-induced blood pressure elevation in part through activation of a medullary cyclooxygenase 2 pathway. Hypertension 2009; 54: 1077–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Zhang MZ, Harris RC. Antihypertensive mechanisms of intra-renal dopamine. Curr Opin Nephrol Hypertens 2015; 24: 117–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Zhang MZ, Yao B, Wang S, Fan X, Wu G, Yang H, et al. Intrarenal dopamine deficiency leads to hypertension and decreased longevity in mice. J Clin Invest. 2011; 121: 2845–2854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Zhang MZ, Wang Y, Yao B, Gewin L, Wei S, Capdevila JH, Harris RC.Role of epoxyeicosatrienoic acids (EETs) in mediation of dopamine’s effects in the kidney. Am J Physiol Renal Physiol. 2013; 305: F1680–1686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 368.Andres-Mateos E, Perier C, Zhang L, Blanchard-Fillion B, Greco TM, Thomas B et la. DJ-1 gene deletion reveals that DJ-1 is an atypical peroxiredoxin-like peroxidase. Proc Natl Acad Sci USA 2007; 104: 14807–14812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369.Rubattu S, Pagliaro B, Pierelli G, Santolamazza C, Castro SD, Mennuni S, Volpe M et al. Pathogenesis of target organ damage in hypertension: role of mitochondrial oxidative stress. Int J Mol Sci 2014; 16: 823–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 370.de Souza BM, Michels M, Sortica DA, Bouças AP, Rheinheimer J, Buffon MP et al. Polymorphisms of the UCP2 gene are associated with glomerular filtration rate in type 2 diabetic patients and with decreased UCP2 gene expression in human kidney. PLoS One 2015; 10: e0132938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 371.Rybka J, Kupczyk D, Kędziora-Kornatowska K, Motyl J, Czuczejko J, Szewczyk-Golec K et al. Glutathione-related antioxidant defense system in elderly patients treated for hypertension. Cardiovasc Toxicol 2011; 11: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372.Boyer TD. The glutathione S-transferases: an update. Hepatology 1989; 9: 486–496. [DOI] [PubMed] [Google Scholar]
  • 373.Palmer CNTurck N, Robin X, Walter N, Fouda C, Hainard A, Sztajzel R, Wagner G, Hochstrasser DF, Montaner J, Burkhard PR, Sanchez JC. Blood glutathione S-transferase-π as a time indicator of stroke onset. PLoS One. 2012; 7: e43830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 374.Oniki K, Hori M, Takata K, Yokoyama T, Mihara S, Marubayashi T et al. Association between glutathione S-transferase A1, M1 and T1 polymorphisms and hypertension. Pharmacogenet Genomics 2008; 18: 275–277. [DOI] [PubMed] [Google Scholar]
  • 375.Zusterzeel PL, Visser W, Peters WH, Merkus HW, Nelen WL, Steegers EA. Polymorphism in the glutathione S transferase P1 gene and risk for preeclampsia. Obstet Gynecol 2000; 96: 50–54. [DOI] [PubMed] [Google Scholar]
  • 376.Ge B, Song Y, Zhang Y, Liu X, Wen Y, Guo X et al. Glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) null polymorphisms and the risk of hypertension: a meta-analysis. PLoS One 2015; 10: e0118897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 377.Wang Y, Branicky R, Noë A, Hekimi S. Superoxide dismutases: Dual roles in controlling ROS damage and regulating ROS signaling. J Cell Biol. 2018; 217: 1915–1928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 378.Alili R, Nivet-Antoine V, Saldmann A, Golmard JL, Cottart CH, Laguillier C, Giral P, Beaudeux JL, Bouillot JL, Poitou C, Clément K, Hébert-Schuster M. Human catalase gene promoter haplotype and cardiometabolic improvement after bariatric surgery. Gene. 2018; 656: 17–21. [DOI] [PubMed] [Google Scholar]
  • 379.Kasperczyk S, Kasperczyk J, Ostałowska A et al. The role of the antioxidant enzymes in erythrocytes in the development of arterial hypertension among humans exposed to lead. Biol Trace Elem Res 2009; 130: 95–106. [DOI] [PubMed] [Google Scholar]
  • 380.Lacy F, O’Connor DT, Schmid-Schönbein GW. Plasma hydrogen peroxide production in hypertensives and normotensive subjects at genetic risk of hypertension. J Hypertens 1998; 16: 291–303. [DOI] [PubMed] [Google Scholar]
  • 381.Lacy F, Kailasam MT, O’Connor DT, Schmid-Schönbein GW, Parmer RJ et al. Plasma hydrogen peroxide production in human essential hypertension: role of heredity, gender, and ethnicity. Hypertension 2000; 36: 878–884. [DOI] [PubMed] [Google Scholar]
  • 382.Puigserver P1, Spiegelman BM. Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator. Endocr Rev 2003; 24: 78–90. [DOI] [PubMed] [Google Scholar]
  • 383.Chen SD, Yang DI, Lin TK, Shaw FZ, Liou CW, Chuang YC. Roles of oxidative stress, apoptosis, PGC-1α and mitochondrial biogenesis in cerebral ischemia. Int J Mol Sci 2011; 12: 7199–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Togliatto G, Lombardo G, Brizzi MF. The Future Challenge of Reactive Oxygen Species (ROS) in Hypertension: From Bench to Bed Side. Int J Mol Sci. 2017; 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 385.Mihalj M, Tadzic R, Vcev A, Rucevic S, Drenjancevic I. Blood pressure reduction is associated with the changes in oxidative stress and endothelial activation in hypertension, regardless of antihypertensive therapy. Kidney Blood Press Res 2016; 41: 721–735. [DOI] [PubMed] [Google Scholar]
  • 386.Chin MP, Bakris GL, Block GA, et al. Bardoxolone Methyl Improves Kidney Function in Patients with Chronic Kidney Disease Stage 4 and Type 2 Diabetes: Post-Hoc Analyses from Bardoxolone Methyl Evaluation in Patients with Chronic Kidney Disease and Type 2 Diabetes Study. Am J Nephrol. 2018; 47: 40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 387.Miller ER, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementation may increase all-cause mortality. Ann Intern Med 2005; 142: 37–46. [DOI] [PubMed] [Google Scholar]
  • 388.Schwingshackl L, Boeing H, Stelmach-Mardas M, Gottschald M, Dietrich S, et al. Dietary Supplements and Risk of Cause-Specific Death, Cardiovascular Disease, and Cancer: A Systematic Review and Meta-Analysis of Primary Prevention Trials. Adv Nutr 2017; 8: 27–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389.Xiao L, Dong JH, Teng X, Jin S, Xue HM, Liu SY, et al. Hydrogen sulfide improves endothelial dysfunction in hypertension by activating peroxisome proliferator-activated receptor delta/endothelial nitric oxide synthase signaling. J Hypertens. 2018; 36: 651–665. [DOI] [PubMed] [Google Scholar]
  • 390.Van Goor H, van den Born JC, Hillebrands JL, Joles JA. Hydrogen sulfide in hypertension. Curr Opin Nephrol Hypertens 2016; 25: 107–113. [DOI] [PubMed] [Google Scholar]
  • 391.de Zeeuw D, Akizawa T, Audhya P et al. BEACON Trial Investigators. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 2013; 369: 2492–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392.Rossing P Diabetic nephropathy: Could problems with bardoxolone methyl have been predicted? Nat Rev Nephrol 2013; 9: 128–30. [DOI] [PubMed] [Google Scholar]
  • 393.Himmelfarb J, Tuttle KR. Bardoxolone methyl in type 2 diabetes and advanced chronic kidney disease. N Engl J Med 2014; 370: 1768–1769. [DOI] [PubMed] [Google Scholar]
  • 394.Duffy SJ, Gokce N, Holbrook M, Huang A, Frei B, Keaney JF Jr et al. Treatment of hypertension with ascorbic acid. Lancet 1999; 354: 2048–2049. [DOI] [PubMed] [Google Scholar]
  • 395.Mullan BA, Young IS, Fee H, McCance DR. Ascorbic acid reduces blood pressure and arterial stiffness in type 2 diabetes. Hypertension 2002; 40: 804–809. [DOI] [PubMed] [Google Scholar]
  • 396.Czernichow S, Bertrais S, Blacher J Galan P, Briançon S, Favier A et al. Effect of supplementation with antioxidants upon long-term risk of hypertension in the SU.VI.MAX study: association with plasma antioxidant levels. J Hypertens 2005; 23: 2013–2018. [DOI] [PubMed] [Google Scholar]
  • 397.Kim MK, Sasaki S, Sasazuki S, Okubo S, Hayashi M, Tsugane S. Lack of long-term effect of vitamin C supplementation on blood pressure. Hypertension 2002; 40: 797–803. [DOI] [PubMed] [Google Scholar]
  • 398.Borghi C, Cicero AF. Nutraceuticals with a clinically detectable blood pressure-lowering effect: a review of available randomized clinical trials and their meta-analyses. Br J Clin Pharmacol. 2017; 83: 163–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 399.Kamper M, Tsimpoukidi O, Chatzigeorgiou A, Lymberi M, Kamper EF. The antioxidant effect of angiotensin II receptor blocker, losartan, in streptozotocin-induced diabetic rats. Transl Res 2010; 156, 26–36. [DOI] [PubMed] [Google Scholar]
  • 400.Molteni A, Ward WF, Ts’ao CH, Taylor J et al. Cytostatic properties of some angiotensin I converting enzyme inhibitors and of angiotensin II type I receptor antagonists. Curr Pharm 2003; 9: 751–761. [DOI] [PubMed] [Google Scholar]
  • 401.Chopra M1, McMurray J, Stewart J, Dargie HJ, Smith WE. Free radical scavenging: a potentially beneficial action of thiol-containing angiotensin converting enzyme inhibitors. Biochem Soc Trans. 1990; 18, 1184–1185. [DOI] [PubMed] [Google Scholar]
  • 402.Dalbeth N, Edwards J, Fairchild S, Callan M, Hall FC. The non-thiol angiotensin-converting enzyme inhibitor quinapril suppresses inflammatory arthritis. Rheumatology (Oxford). 2005; 44: 24–31. [DOI] [PubMed] [Google Scholar]
  • 403.Bucci M1, Vellecco V, Cantalupo A, Brancaleone V, Zhou Z, Evangelista S, Calderone V, et al. Hydrogen sulfide accounts for the peripheral vascular effects of zofenopril independently of ACE inhibition. Cardiovasc Res. 2014; 102: 138–147. [DOI] [PubMed] [Google Scholar]
  • 404.Moser M, Frishman W. Results of therapy with carvedilol, a beta-blocker vasodilator with antioxidant properties, in hypertensive patients. Am J Hypertens 1998; 1 Pt 2: 15S–22S. [DOI] [PubMed] [Google Scholar]
  • 405.Gomes A, Costa D, Lima JL, Fernandes E. Antioxidant activity of beta-blockers: an effect mediated by scavenging reactive oxygen and nitrogen species? Bioorg Med Chem 2006; 14: 4568–4577. [DOI] [PubMed] [Google Scholar]
  • 406.Tomino Y Renoprotective effects of the L-/T-type calcium channel blocker benidipine in patients with hypertension. Curr Hypertens Rev. 2013; 9: 108–114. [DOI] [PubMed] [Google Scholar]
  • 407.do Vale GT, Simplicio JA, Gonzaga NA, Yokota R, Ribeiro AA, Casarini DE, et al. Nebivolol prevents vascular oxidative stress and hypertension in rats chronically treated with ethanol. Atherosclerosis. 2018; 274: 67–76. [DOI] [PubMed] [Google Scholar]
  • 408.Ahmadian E, Khosroushahi AY, Eftekhari A, Farajnia S, Babaei H, Eghbal MA. Novel angiotensin receptor blocker, azilsartan induces oxidative stress and NFkB-mediated apoptosis in hepatocellular carcinoma cell line HepG2. Biomed Pharmacother 2018; 99: 939–946. [DOI] [PubMed] [Google Scholar]
  • 409.Yang CS, Ho CT, Zhang J, Wan X, Zhang K, Lim J. Antioxidants: Differing Meanings in Food Science and Health Science. J Agric Food Chem. 2018; 66: 3063–3068. [DOI] [PubMed] [Google Scholar]
  • 410.Gomez-Cabrera MC, Domenech E, Romagnoli M, Arduini A, Borras C, Pallardo FV et al. Oral administration of vitamin C decreases muscle mitochondrial biogenesis and hampers training-induced adaptations in endurance performance. Am J Clin Nutr 2008; 87: 142–149. [DOI] [PubMed] [Google Scholar]
  • 411.Morrison D, Hughes J, Della Gatta PA, Mason S, Lamon S, Russell AP, et al. Vitamin C and E supplementation prevents some of the cellular adaptations to endurance-training in humans. Free Radic Biol Med. 2015; 89: 852–862. [DOI] [PubMed] [Google Scholar]
  • 412.Merry TL, Ristow M. . Do antioxidant supplements interfere with skeletal muscle adaptation to exercise training? J Physiol. 2016; 594: 5135–5147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 413.Johnson SA, Feresin RG, Navaei N, Figueroa A, Elam ML, Akhavan NS, et al. Effects of daily blueberry consumption on circulating biomarkers of oxidative stress, inflammation, and antioxidant defense in postmenopausal women with pre- and stage 1-hypertension: a randomized controlled trial. Food Funct 2017; 8: 372–380. [DOI] [PubMed] [Google Scholar]
  • 414.Guo K, Lu J, Huang Y, Wu M, Zhang L, Yu H et al. Protective role of PGC-1α in diabetic nephropathy is associated with the inhibition of ROS through mitochondrial dynamic remodeling. PLoS One 2015; 10: e0125176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 415.Zhang M, Brewer AC, Schröder K et al. NADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesis. Proc Natl Acad Sci U S A 2010; 107: 18121–18126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 416.Coughlan MT, Sharma K. Challenging the dogma of mitochondrial reactive oxygen species overproduction in diabetic kidney disease. Kidney Int 2016; 90: 272–279. [DOI] [PubMed] [Google Scholar]
  • 417.Towler DA. Mitochondrial ROS deficiency and diabetic complications: AMP[K]-lifying the adaptation to hyperglycemia. J Clin Invest. 2013; 123: 4573–4576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.Craige SM, Chen K, Pei Y, Li C, Huang X, Chen C et al. NADPH oxidase 4 promotes endothelial angiogenesis through endothelial nitric oxide synthase activation. Circulation 2011; 124: 731–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 419.Brüll V, Burak C, Stoffel-Wagner B, Wolffram S, Nickenig G, Müller C, et al. Effects of a quercetin-rich onion skin extract on 24 h ambulatory blood pressure and endothelial function in overweight-to-obese patients with (pre-)hypertension: a randomised double-blinded placebo-controlled cross-over trial. Br J Nutr. 2015; 114: 1263–1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 420.Patel RV, Mistry BM, Shinde SK, Syed R, Singh V, Shin HS. Therapeutic potential of quercetin as a cardiovascular agent. Eur J Med Chem. 2018; 155: 889–904. [DOI] [PubMed] [Google Scholar]
  • 421.Tain YL, Lee WC, Wu KLH, Leu S, Chan JYH. Resveratrol Prevents the Development of Hypertension Programmed by Maternal Plus Post-Weaning High-Fructose Consumption Through Modulation of Oxidative Stress, Nutrient-Sensing Signals, and Gut Microbiota. Mol Nutr Food Res. 2018. April 30:e1800066. doi: 10.1002/mnfr.201800066 [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 422.Banday AA, Lokhandwala MF. Oxidative stress impairs cGMP-dependent protein kinase activation and vasodilator-stimulated phosphoprotein serine-phosphorylation. Clin Exp Hypertens. 2018; 9: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES