Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Aug 1.
Published in final edited form as: Cardiovasc Ther. 2010 Mar 29;28(4):e20–e32. doi: 10.1111/j.1755-5922.2010.00137.x

Natural Antioxidants and Hypertension: Promise and Challenges

Tinoy J Kizhakekuttu 1, Michael E Widlansky 1
PMCID: PMC2905473  NIHMSID: NIHMS209319  PMID: 20370791

Abstract

Hypertension reigns as a leading cause of cardiovascular morbidity and mortality worldwide. Excessive reactive oxygen species (ROS) has emerged as a central common pathway by which disparate influences may induce and exacerbate hypertension. Potential sources of excessive ROS in hypertension include NADPH oxidase, mitochondria, xanthine oxidase, endothelium-derived NO synthase (eNOS), cyclooxygenase 1 and 2, cytochrome P450 epoxygenase and transition metals. While a significant body of epidemiological and clinical data suggests that antioxidant rich diets reduce blood pressure and cardiovascular risk, randomized trials and population studies using natural antioxidants have yielded disappointing results. The reasons behind this lack of efficacy are not completely clear, but likely include a combination of 1) ineffective dosing regimens 2) the potential pro-oxidant capacity of some of these agents 3) selection of subjects less likely to benefit from antioxidant therapy (too healthy or too sick), 4) inefficiency of non-specific quenching of prevalent ROS versus prevention of excessive ROS production. Commonly used antioxidants include Vitamins A, C and E, L-arginine, flavanoids, and mitochondria targeted agents, Coenzyme Q10, acetyl-L-carnitine and alpha-lipoic acid. Various reasons, including incomplete knowledge of the mechanisms of action of these agents, lack of target specificity, and potential inter-individual differences in therapeutic efficacy preclude us from recommending any specific natural antioxidant for antihypertensive therapy at this time. This review focuses on recent literature regarding above mentioned issues evaluating naturally occurring antioxidants with respect to their impact on hypertension.

Keywords: Hypertension, Antioxidants, Oxidative Stress

Introduction

Hypertension (HTN) is the most important cardiovascular risk factor worldwide, contributing to half of prevalent coronary heart disease and approximately two thirds of prevalent cerebrovascular disease burdens.1 While a multitude of genetic and environmental factors contribute to this complex disease, excessive reactive oxygen species have emerged as a central common pathway by which disparate influences may induce and exacerbate hypertension.2 Further, a significant body of epidemiological3 and clinical trial data4, 5 suggest that diets known to contain significant concentrations of naturally occurring antioxidants appear to reduce blood pressure and may reduce cardiovascular risk.

In light of these data, there is significant interest in identifying key, naturally-occurring antioxidants to both prevent and treat hypertension. This reviewl focuses on the recent literature evaluating naturally occurring antioxidants with respect to their impact on hypertension.

Role of Oxidative Stress in the Pathogenesis of Hypertension

Reactive oxygen species (ROS) are generated by multiple cellular sources, including NADPH oxidase, mitochondria, xanthine oxidase, uncoupled endothelium-derived NO synthase, cycloxygenase, and lipoxygenase (Table 1).6, 7 The dominant initial ROS species produced by these sources is superoxide (O2). Superoxide is short-lived molecule that can subsequently undergo enzymatic dismutation to hydrogen peroxide. Superoxide can oxidize proteins and lipids, or react with endothelium-derived nitric oxide (NO) to create the reactive nitrogen species peroxynitrite. Peroxynitrite and other reactive nitrogen species can subsequently oxidize proteins, lipids, and critical enzymatic cofactors that may further increase oxidative stress.8, 9 Hydrogen peroxide produced by enzymatic dismutation of O2 can be further convert to highly reactive hydroxyl radical (via Fenton chemistry) that can cause DNA damage.10 The balance between superoxide production and consumption likely keeps the concentration of O2 in the picomolar range and hydrogen peroxide in the nanomolar range.11 These homeostatic levels of reactive oxygen species appear to be important in normal cellular signaling1214 and normal reactions to stressors.15, 16

Table 1.

Potential Sources of Excessive Reactive Oxygen Species in Hypertension

• NADPH Oxidase
• Mitochondria
• Xanthine Oxidase
• Endothelium-Derived NO Synthase (eNOS)
• Cycloxygenase 1 and 2
• Cytochrome P450 epoxygenase
• Transition Metals (e.g. Iron)

While multiple diverse factors likely contribute to the development of hypertension, the pathogenesis of this disease appears related, at least in part, to the development of a state of excessive oxidative stress. Local excessive superoxide production in the kidneys, CNS, and vasculature, along with inflammatory activation, are central findings in hypertension models.17,18 Animal studies demonstrate the development of hypertension with associated increases in oxidative stress and impaired vasodilation in rats exposed to a high-salt and oxidant containing diet.19 Further, infusion of superoxide dismutase lowers oxidative stress and blood pressure in these animal models.2022 Divergent animal models of hypertension, including spontaneous hypertension,23 salt-sensitive hypertension,24,25 renovascular hypertension,26,27 and obesity-related hypertension28 are all associated with excessive oxidative stress. These data suggest, regardless of etiology, excessive ROS is a common factor in the pathogenesis and morbidity of hypertension.

Roles and Interactions of Sources of Oxidative Stress in Hypertension

As delineated in Table 1, multiple diverse sources of reactive oxygen species generation are relevant to the pathogenesis of hypertension. The prominent role of excessive superoxide produced by NADPH oxidase under angiotensin II stimulation in the development of hypertension has recently been extensively reviewed.17 Interestingly, data emerging over the past several years indicate that hypertension-related excessive ROS levels are most likely secondary to regulatory interactions between the major sources of ROS themselves involving ROS in the signaling process.

Mitochondria produce excessive ROS in the setting of spontaneous hypertension29,30 as well as in hypertensive states characterized by salt-sensitivity and elevated endothelial-1 levels.31 Mitochondria may produce superoxide through multiple mechanisms, including the electron transport chain complexes (I, II, and III), monoamine oxidase A and B, and Krebs cycle enzymes.32 Interestingly, extensive regulatory crosstalk between NADPH oxidase and mitochondria appears to modulate superoxide production from both sources.33,34 For example, over-expression of thioredoxin-2, an important mitochondrial-based antioxidant thiol, reduces angiotensin II-induced hypertension35 and lowers basal blood pressure in transgenic mice over-expressing thioredoxin.36

NADPH oxidase and mitochondria also interact with endothelium-derived NO synthase (eNOS) through ROS production, modulating overall NO bioavailability and superoxide production from eNOS. eNOS has been localized to the outer mitochondrial membrane in endothelial cells.37 Beyond quenching NO through direct reaction to create peroxynitrite, superoxide from NADPH oxidase or mitochondria can oxidize tetrahydrobiopterin (BH4), a necessary eNOS cofactor, leading to eNOS uncoupling and superoxide production from eNOS. 38 Further, uncoupling of eNOS leads to reduced NO bioavailability and excessive mitochondrial ROS production. 39

Xanthine oxidase, which generates superoxide by converting hypoxanthine to xanthine, is upregulated by NADPH oxidase under oscillatory shear conditions.40 Xanthine oxidase may also contribute to excessive ROS production in salt-sensitive hypertension, although its relative contribution compared to mitochondria and NADPH oxidase remains to be fully elucidated.41 Prior work also demonstrates potential roles for lipoxygenase, cycloxygenase, cytochrome P450 epoxygenase, and transitional metals in overall cellular superoxide production, but further study is necessary to better delineate the roles of these sources of ROS in hypertension.

While the causal intrinsic and extrinsic factors governing the development of hypertension are very likely multi-factorial, genetic polymorphisms associated with sources of oxidative stress in hypertension may modulate an individual’s potential for elevated ROS and the development of hypertension under genetic and environmental influences.4250 Overall, while there may be a hierarchy of the relative contributions of each ROS source in hypertension, the measured combined local concentrations of superoxide and peroxynitrite most likely reflect a combination of genetic susceptibility, coordinated ROS generation from multiple sources, local environmental influences on sources of ROS production, and overall intrinsic anti-oxidant defense mechanisms.51

Selecting Natural Antioxidants to Treat Hypertension

Randomized trials employing non-pharmacological dietary interventions emphasizing fruits, vegetables, whole grains, and nuts have shown impressive blood pressure lowering results in both hypertensive and normotensive subjects.5254 Similar interventions demonstrated to reduce cardiovascular morbidity and mortality continue to maintain interest in the potential of isolating specific compounds enriched in these diets that may be responsible for the overall dietary benefits.55

The dietary components in these studies are high in compounds known to have antioxidant properties leading many to ascribe the benefits of these diets to their increased content of natural antioxidants. However, prior randomized trials and population studies in healthy populations and patients at high risk for cardiovascular events that have employed combinations of some of these natural antioxidants as dietary supplements have, for the most part, shown disappointing results,5660 6163 The reasons behind these disappointing results are not completely clear, but likely include a combination of 1) ineffective dosing and dosing regimens 2) the potential pro-oxidant capacity and other potentially deleterious effects of these some of these compounds under certain conditions,64, 65 3) selection of subjects less likely to benefit from antioxidant therapy (too healthy or too sick). Populations at intermediate cardiovascular risk may be better suitable to see effects of antioxidants in shorter term studies.66 4) inefficiency of non-specific quenching of prevalent ROS versus prevention of excessive ROS production.67, 68

When considering antioxidant therapy for hypertension, lessons from prior disappointing attempts to reduce blood pressure and cardiovascular risk with antioxidant therapy should be considered. The profile of an ideal agent is outlined in Table 2. The importance of patient selection is being increasingly recognized in light of emerging data suggesting that antioxidant supplementation in healthy subjects may blunt the protective benefits of aerobic exercise training, suggesting ROS generation can be beneficial under certain circumstances.69

Table 2.

Optimal Profile of Natural Antioxidant Agent for Anti-Hypertensive Therapy

• Good oral bioavailability
• Patient-friendly dosing regimen (once or twice daily dosing)
• Concentrates locally in relevant tissues (kidney, brain, and/or vasculature)
• Limited potential for pro-oxidative role and secondary cell signaling that may limit
  effectiveness
• Inhibits the production of ROS rather quenching ROS post-production.
• Good safety profile with limited side effects
• Efficacious for hypertension originating from disparate etiologies
• No adverse interactions with the metabolism of potential concomitant anti-hypertensive
  pharmacological therapy
• Has pleiotropic effects that go beyond blood pressure lowering and translate into
  prevention of/ reversal of/ slower progression of end organ damage

Anti-Hypertensive Profile of Common Natural Antioxidants

Antioxidant Vitamins

Vitamin A Precursors and Derivatives

Vitamin A precursors and derivatives are retinoids that consist of a beta-ionone ring attached to an isoprenoid carbon chain. Foods high in vitamin A include liver, sweet potato, carrot, pumpkin, and broccoli leaf. Initial interest in vitamin A-related compounds focused primarily on beta-carotene, given initial promising epidemiological data with respect to its cardioprotective effects and some correlation with higher plasma levels to lower blood pressure in men.70 However, concerns about beta-carotene’s pro-oxidative potential came to light with a report suggesting adverse mitochondrial effects of beta-carotene cleavage products.71 Further, adverse mortality data with respect to beta-carotene has limited interest in this compound as an effective antihypertensive agent.72

Recently, interest in vitamin A derivatives has turned to lycopene, itself a potent antioxidant,73 found concentrated in tomatoes. One small study has shown a reduction in blood pressure with a tomato-extract based intervention (containing a combination of potential anti-oxidant compounds including lycopene) in patients with stage I hypertension, 74 although second study showed no effect in pre-hypertensive patients. 75

Ascorbic Acid (Vitamin C)

L-ascorbic acid is a six-carbon lactone and, for humans, is an essential nutrient. In Western diets, commonly consumed foods that contain high levels of ascorbic acid include broccoli, lemons, limes, oranges, and strawberries. Toxicity potential of this compound is low, although an increased risk of oxalate renal calculi may exist at higher doses (exceeding 2 grams/day) 76

The initial purported mechanisms for the potential benefits of ascorbate supplementation were centered on quenching of single-electron free radicals. Subsequent research has demonstrated that the plasma concentrations of ascorbate required for this mechanism to be physiologically relevant are not attainable by oral supplementation.77 However, vitamin C can concentrate in local tissues to levels an order of magnitude higher than that of plasma. At this leve, ascobate may to effectively compete for superoxide and reduce thiols.78, 79 Recent data also suggest potential suppressive effects of ascorbate on NADPH oxidase activity.80, 81 Ascorbate appears to have limited pro-oxidant ability.82

Ascorbate’s anti-hypertensive efficacy has been evaluated in multiple small studies. Many,8386 but not all,87 show modest reductions in blood pressure in both normotensive and hypertensive populations. These data also suggest that supplementation has limited effect on systemic antioxidant markers85 and little additional blood pressure benefits are seen beyond the 500 mg daily dose. Large scale randomized trial data specific to ascorbate supplementation and its effects on hypertension are currently lacking. Data from Heart Protection Study (HPS) suggest no significant mortality from supplementation with 250mg/day of ascorbate supplementation.57 However, the relatively low dose of ascorbate, use of combination therapy, and high-risk patient population studied in HPS leave unanswered the key questions of appropriate dosing and target.

α-Tocopherol (Vitamin E)

Vitamin E is a generic term for a group of compounds classified as tocopherols and tocotrienols.88 While there are four isomers in each class of Vitamin E compounds, the overwhelming majority of the active form is α-tocopherol.89 Dietary sources high in vitamin E include avocados, asparagus, vegetable oils, nuts, and leafy green vegetables.

Vitamin E is a potent antioxidant that inhibits LDL and membrane phospholipid oxidation.90 Interestingly, inflammatory cells and neurons have binding proteins for α-tocopherol, the actions of which may include inhibition of NADPH oxidase, lipoxygenase, and cyclo-oxygenase, actions which may lower oxidative stress.91 However, studies demonstrating vitamin E’s pro-oxidant capacity under certain cellular conditions suggest that local condition may influence the vitamin E’s redox activity.65

Initial excitement for vitamin E supplementation was based on the reduction of cardiovascular events seen in the CHAOS study.60 However, follow-up studies have been largely disappointing.9294 While one small study that used vitamin E in combination with zinc, vitamin C, and beta-carotene showed a modest, significant reduction in blood pressure over 8 weeks of therapy,95 other small studies96, 97 show either no effect or a pressor effect from vitamin E supplementation. Further, the more definitive HOPE trial, failed to show blood pressure or mortality benefit for patients at high risk for cardiovascular disease.92

L-Arginine

L-arginine is an amino acid and the main substrate for the production of NO from eNOS in a reaction that is dependent on tetrahydrobiopterin.98 Potential dietary sources include milk products, beef, wheat germ, nuts, and soybeans. Reduced levels of tetrahydrobiopterin leads to uncoupling of reduced NADPH oxidation and NO synthesis, with oxygen as terminal electron acceptor instead of L-arginine, resulting in the generation of superoxide by eNOS. 99101 Low cellular levels of L-arginine have been demonstrated in human hypertension, 102, 103 While L-arginine deficiency itself does not appears to lead to uncoupling of eNOS,104 low levels of L-arginine may lead to reduced levels of bioavailable NO which could contribute to hypertension. Thus, L-arginine supplementation could theoretically reduce blood pressure by allowing for restoration of normal NO bioavailability, perhaps overcoming overall L-arginine deficiency as well as more successfully competing fo the eNOS active site with circulating asymmetric dimet hylarginine, a circulating competitor of L-arginine that may be increased in the setting of hypertension.105

This concept is supported by studies demonstrating the anti-hypertensive effect of L-arginine supplementation in salt-sensitive rats,106 healthy human subjects,107 hypertensive diabetics,108 patients with chronic kidney disease,109 and diabetic patients in combination with N-acetylcysteine, a precursor of glutathione.108 L-arginine’s anti-hypertensive response may be mediated in part by its suppressive effects on angiotensin II and endothelin-1, and its potentiating effects on insulin.110

However, recent concerns about potential deleterious increases in homocysteine in the setting of L-arginine supplementation have been raised.111 The majority of L-arginine is processed into creatine, which leads increased homocysteine levels.112 Homocysteine can increase oxidative stress.113 A recent study confirms this mechanism is relevant to L-arginine metabolism in humans,114 suggesting a potential mechanism for neutralizing the eNOS-related anti-oxidant effects of L-arginine.

Flavonoids

Flavonoids are polyphenolic compounds commonly found in concentrated amounts in multiple fruits, vegetables, and beverages, including apples, berries, grapes, onions, pomegranate, red wine, tea, cocoa, and dark chocolate. The exact structure and composition of the flavonoid compounds varies between food sources, and flavonoid content can be altered based on the manner of food preparation.115 Interest in flavonoids as antioxidant therapy for cardiovascular disease originates from epidemiological data suggesting improved cardiovascular outcomes in individuals with high intake of food and beverages with high flavonoid content115, 116 as well as cellular work suggesting a strong anti-oxidant effect of these compounds.117120

However, the limited oral bioavailability of flavonoids suggests cell signaling mechanisms, rather than free radical quenching activity, is more likely to be root of sustained cardiovascular benefits from flavonoids.120 This concept is consistent with studies demonstrating that flavonoids can inhibit NADPH oxidase through ACE inhibition,121, 122 increase eNOS-specific NO production through the estrogen receptor,123 and alter COX-2 expression.124 Studies investigating the anti-hypertensive effects of flavonoids are inconclusive. While multiple small studies of short duration dark chocolate therapy have demonstrated blood pressure lowering effects in hypertensives,125128 studies in normotensive and pre-hypertensive individuals have demonstrated no benefit.75, 129 Further, tea intake may, at least temporarily, increase blood pressure in certain populations.130, 131 The specific flavonoids and combination of flavonoids that exert the largest beneficial effects remain unknown.

Mitochondria-Related Anti-Oxidants

Coenzyme Q10 (CoQ)

CoQ (2, 3 dimethoxy-5 meth-6-decaprenyl benzoquinone) is derived from mevalonic acid and phenylalanine, and can be supplemented by oral intake. This compound is a key component of the electron transport chain, accepting electrons from Complexes I and II and the glyceraldehyde-3-phosphate shuttle. CoQ levels have been shown to be lower in older adults known to have a greater prevalence of hypertension.132 The mechanism of action of CoQ is not likely to be secondary to a superoxide scavenger effect given CoQ’s hydrophobic properties. CoQ may reduce mitochondrial superoxide production by increasing the efficiency of electron transfer from Complexes I and II down the mitochondrial electron transport chain.133 Coenzyme Q may also exert an antioxidant effect by reducing lipid peroxidation at the level of the plasma membrane.134

Early data from non-controlled studies in human hypertension demonstrate reductions in blood pressure with CoQ supplementation.135, 136 Further, small randomized studies using a CoQ dose of 100–120mg daily have demonstrated significant reductions in blood pressure with minimal side effects in patient with Stage II hypertension.137140 Interestingly, a new, mitochondrial-targeted formulation of CoQ has demonstrated anti-hypertensive efficacy in a rat model.141

Acetyl-L-Carnitine (ALCAR) and α-Lipoic Acid (LA)

LA is a dithiol compound synthesized from octanoic acid in mitochondria. The in vivo and in vitro effects of LA have been thoroughly reviewed elsewhere.142, 143 LA has moderate oral bioavailability.144. While LA is a potent in vitro antioxidant, the limited plasma concentrations achievable with supplementation and rapid clearance of LA suggest free radical scavenger and anti-oxidant recycling activity are unlikely to be the primary in vivo activity of LA. Participation in mitochondrial-associated metabolic pathways, in cell signaling that may improve coupling of eNOS, and anti-inflammatory actions are among the potential beneficial effects of LA supplementation.142, 145 Work in a diabetic rat and multiple different hypertensive rat models has shown the potential for LA supplementation to reduce blood pressure.146149

ALCAR (acetylated L-carnitine) is a key compound in the transport of fatty acids into mitochondria for beta-oxidation. The antioxidant mechanism of ALCAR supplementation appears to be secondary to reductions in mitochondrial ROS production in synergy with concomitant LA therapy.150 The exact intra-mitochondrial mechanism ALCAR’s effects are not clear, and prior work in older rats demonstrates ALCAR potential to be pro-oxidative when used alone.151 Further data suggest ALCAR may be of particular benefit in diabetics with hypertension secondary to their low carnitine levels152 and elevated circulating free fatty acid levels.153, 154

Human data with respect to the anti-hypertensive effects of these compounds is limited to two small studies which have shown some promising results. Consistent with animal data, combined ALCAR and LA therapy reduced systolic blood pressure in coronary artery disease patients with hypertension and/or metabolic syndrome at the time of enrollment.155 Also consistent with prior cell culture and animal work, 32 type 2 diabetic subjects supplemented with 2 grams/day of acetyl-L-carnitine showed significantly lowered blood pressure and improved insulin sensitivity.156

Other Potential Natural Antioxidant Agents

Garlic,157 glutamate,158 N-acetylcysteine,159 sour milk,160, 161 and vitamin D162, 163 all have shown anti-hypertensive effects through anti-oxidant mechanisms that may involve inhibition of sources of excessive ROS. Further work remains to be done to establish the mechanisms and efficacy of these interventions.

Conclusions and Future Directions

A summary of our findings with respect to the above interventions is contained in Table 3. Critical evaluation of the these data reveal several issues and limitations related to our current knowledge of natural antioxidant compounds and their potential anti-hypertensive efficacy that obviate our ability to recommend any individual agent at this time (Table 4). First, the majority of these agents have been discovered to have potential mechanisms of action that were initially unanticipated, including the potential for deleterious, pro-oxidative effects. A greater understanding of the mechanisms of action of the above agents may allow providers to better target therapies to appropriate populations. Second, while interventions such as tomato extract and dark chocolate may hold promise, the identity of the compounds or mix of compounds responsible for the antihypertensive effects of these interventions remain unknown and need to be identified before lycopene or individual flavonoid compounds can be recommended as supplements for anti-hypertensive therapy. Third, small, single center trials often enrolling less than 100 subjects comprise the majority of studies found related to novel antioxidant therapy for hypertension, leaving open concerns with respect to publication bias. In addition, the vast majority of these studies made no systemic measurements of total antioxidant capacity, making to determine whether changes in anti-oxidant capacity accompanied the observed reductions in blood pressure. With the exception of Vitamins A and E (which cannot be recommended at this time), data from larger randomized clinical trials aimed at blood pressure lowering and optimally also measuring cardiovascular endpoints and antioxidant effects would help more clearly distinguish which of the above agents, if any, may be reasonable to recommend to as anti-hypertensive agents as well as help determine if antioxidant actions may be responsible for any ameliorative effects. Thus, despite some interesting findings, the recommendations of the American Heart Association of a dietary strategy rich in fruits and vegetables appears to continue to be the best strategy for non-pharmacological therapy in hypertension.164 Further work is clearly necessary in order to more clearly identify which natural antioxidants have efficacy and their mechanisms of action.

Table 3.

Profiles of Commonly Used Natural Antioxidants for Hypertension

Supplement Antioxidant
Capacity?
Pro-oxidant
Capacity
Lowers BP in
small clinical
trials
Lowers BP in
large randomized
controlled
studies
Vitamin A
  Beta-carotone + +
   Lycopene (tomato extract) + ? + ?
Vitamin C + + ?
Vitamin E + + +/−
L-Arginine + + + ?
Flavonoid
Containing
food/beverages
+ ? +/−(positive for
dark chocolate,
negative for tea)
?
Acetyl-L-Carnitine ? +/− + ?
α-Lipoic Acid + + (in combination
with ALCAR)
?

Table 4.

Current Limitations Precluding Recommendation of Specific Natural Antioxidant Supplements for Hypertension

• Incomplete knowledge of the mechanism(s) of action of these agents
• Lack of target specificity of these agents
• Lack of large, randomized control trials to determine true anti-hypertensive efficacy for
   many of these agents
• Lack of data on “hard” outcome (e.g. death, cardiovascular events)
• Potential differences in therapeutic efficacy in different hypertensive populations

How did you gather, select and analyze the info you considered in your review?

Literature search was conducted by both authors independently using Medline (1966- present) and Cochrane database of systematic reviews. References from the extracted papers, reviews and meta analyses were also consulted to complete the database. About 150 high quality studies specifically focusing on redox physiology and pathophysiology in hypertension, as well as anti-oxidant therapy for hypertension were selected by the authors and analyzed for this review.

What is the take home message?

Reactive oxygen species play a central role in the pathogenesis of hypertension and its vascular complications. While there are several promising anti-oxidants being tested, current data do not support the use of individual compounds or combinations of supplemental antioxidants for the treatment of hypertension at this time. Further work is necessary to identify which natural antioxidants are efficacious in hypertension. Pending future discoveries in this area, the American Heart Association’s recommendation of a diet rich in fruits and vegetables remains to be the best “natural antioxidant” strategy for non-pharmacological therapy in hypertension.

Acknowledgements

Dr. Widlansky is supported by 1K23HL089326. Dr. Kizhakekuttu is supported by a Ruth L. Kirschstein NIH T32 training grant (HL007792-15).

Footnotes

Conflicts of Interest: None

Reference List

  • 1.Whitworth JA. 2003 World Health Organization (WHO)/International Society of Hypertension (ISH) statement on management of hypertension. J Hypertens. 2003 November;21(11):1983–1992. doi: 10.1097/00004872-200311000-00002. [DOI] [PubMed] [Google Scholar]
  • 2.Griendling KK, FitzGerald GA. Oxidative stress and cardiovascular injury: Part II: animal and human studies. Circulation. 2003 October 28;108(17):2034–2040. doi: 10.1161/01.CIR.0000093661.90582.c4. [DOI] [PubMed] [Google Scholar]
  • 3.Dauchet L, Amouyel P, Hercberg S, Dallongeville J. Fruit and vegetable consumption and risk of coronary heart disease: a meta-analysis of cohort studies. J Nutr. 2006 October;136(10):2588–2593. doi: 10.1093/jn/136.10.2588. [DOI] [PubMed] [Google Scholar]
  • 4.Appel LJ, Moore TJ, Obarzanek E, et al. A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group. N Engl J Med. 1997;336(No. 16):1117–1124. doi: 10.1056/NEJM199704173361601. [DOI] [PubMed] [Google Scholar]
  • 5.Esposito K, Marfella R, Ciotola M, et al. Effect of a mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA. 2004 September 22;292(12):1440–1446. doi: 10.1001/jama.292.12.1440. [DOI] [PubMed] [Google Scholar]
  • 6.Fridovich I. Superoxide anion radical (O2−.), superoxide dismutases, and related matters. J Biol Chem. 1997 July 25;272(30):18515–18517. doi: 10.1074/jbc.272.30.18515. [DOI] [PubMed] [Google Scholar]
  • 7.Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res. 2000 November 10;87(10):840–844. doi: 10.1161/01.res.87.10.840. [DOI] [PubMed] [Google Scholar]
  • 8.Laursen JB, Somers M, Kurz S, et al. Endothelial regulation of vasomotion in apoE-deficient mice : Implications for interactions between peroxynitrite and tetrahydrobiopterin. Circulation. 2001 March 6;103(9):1282–1288. doi: 10.1161/01.cir.103.9.1282. [DOI] [PubMed] [Google Scholar]
  • 9.Munzel T, Daiber A, Ullrich V, Mulsch A. Vascular consequences of endothelial nitric oxide synthase uncoupling for the activity and expression of the soluble guanylyl cyclase and the cGMP-dependent protein kinase. Arterioscler Thromb Vasc Biol. 2005 August;25(8):1551–1557. doi: 10.1161/01.ATV.0000168896.64927.bb. [DOI] [PubMed] [Google Scholar]
  • 10.Dizdaroglu M, Gajewski E, Reddy P, Margolis SA. Structure of a hydroxyl radical induced DNA-protein cross-link involving thymine and tyrosine in nucleohistone. Biochemistry. 1989 April 18;28(8):3625–3628. doi: 10.1021/bi00434a071. [DOI] [PubMed] [Google Scholar]
  • 11.Fridovich I. Superoxide dismutases. Annu Rev Biochem. 1975;44:147–159. doi: 10.1146/annurev.bi.44.070175.001051. [DOI] [PubMed] [Google Scholar]
  • 12.Thomas SR, Chen K, Keaney JF., Jr Hydrogen peroxide activates endothelial nitric-oxide synthase through coordinated phosphorylation and dephosphorylation via a phosphoinositide 3-kinase-dependent signaling pathway. J Biol Chem. 2002 February 22;277(8):6017–6024. doi: 10.1074/jbc.M109107200. [DOI] [PubMed] [Google Scholar]
  • 13.Zembowicz A, Hatchett RJ, Jakubowski AM, Gryglewski RJ. Involvement of nitric oxide in the endothelium-dependent relaxation induced by hydrogen peroxide in rabbit aorta. Br J Pharmacol. 1993;110:151–158. doi: 10.1111/j.1476-5381.1993.tb13785.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Drummond GR, Cai H, Davis ME, Ramasamy S, Harrison DG. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression by hydrogen peroxide. Circ Res. 2000 February 18;86(3):347–354. doi: 10.1161/01.res.86.3.347. [DOI] [PubMed] [Google Scholar]
  • 15.Stocker R, Keaney JF., Jr The role of oxidative modifications in atherosclerosis. Physiol Rev. 2004;84:1381–1478. doi: 10.1152/physrev.00047.2003. [DOI] [PubMed] [Google Scholar]
  • 16.Chen K, Thomas SR, Keaney JF., Jr Beyond LDL oxidation: ROS in vascular signal transduction. Free Radic Biol Med. 2003 July 15;35(2):117–132. doi: 10.1016/s0891-5849(03)00239-9. [DOI] [PubMed] [Google Scholar]
  • 17.Harrison DG, Gongora MC. Oxidative stress and hypertension. Med Clin North Am. 2009 May;93(3):621–635. doi: 10.1016/j.mcna.2009.02.015. [DOI] [PubMed] [Google Scholar]
  • 18.Hoch NE, Guzik TJ, Chen W, et al. Regulation of T-cell function by endogenously produced angiotensin II. Am J Physiol Regul Integr Comp Physiol. 2009 February;296(2):R208–R216. doi: 10.1152/ajpregu.90521.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Miyagawa K, Ohashi M, Yamashita S, et al. Increased oxidative stress impairs endothelial modulation of contractions in arteries from spontaneously hypertensive rats. J Hypertens. 2007 February;25(2):415–421. doi: 10.1097/HJH.0b013e3280115b96. [DOI] [PubMed] [Google Scholar]
  • 20.Nakazono K, Watanabe N, Matsuno K, Sasaki J, Sato T, Inoue M. Does superoxide underlie the pathogenesis of hypertension? Proc Natl Acad Sci U S A. 1991;88:10045–10048. doi: 10.1073/pnas.88.22.10045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Laursen JB, Rajagopalan S, Galis Z, Tarpey M, Freemam BA, Harrison DG. Role of superoxide in angiotensin II-induced but not catecholamine-induced hypertension. Circulation. 1997;95:588–593. doi: 10.1161/01.cir.95.3.588. [DOI] [PubMed] [Google Scholar]
  • 22.Schnackenberg CG, Welch WJ, Wilcox CS. Normalization of blood pressure and renal vascular resistance in SHR with a membrane-permeable superoxide dismutase mimetic: role of nitric oxide. Hypertension. 1998 July;32(1):59–64. doi: 10.1161/01.hyp.32.1.59. [DOI] [PubMed] [Google Scholar]
  • 23.Friedman J, Peleg E, Kagan T, Shnizer S, Rosenthal T. Oxidative stress in hypertensive,diabetic, and diabetic hypertensive rats. Am J Hypertens. 2003 December;16(12):1049–1052. doi: 10.1016/j.amjhyper.2003.07.013. [DOI] [PubMed] [Google Scholar]
  • 24.Trolliet MR, Rudd MA, Loscalzo J. Oxidative stress and renal dysfunction in salt-sensitive hypertension. Kidney Blood Press Res. 2001;24(2):116–123. doi: 10.1159/000054217. [DOI] [PubMed] [Google Scholar]
  • 25.Koga Y, Hirooka Y, Araki S, Nozoe M, Kishi T, Sunagawa K. High salt intake enhances blood pressure increase during development of hypertension via oxidative stress in rostral ventrolateral medulla of spontaneously hypertensive rats. Hypertens Res. 2008 November;31(11):2075–2083. doi: 10.1291/hypres.31.2075. [DOI] [PubMed] [Google Scholar]
  • 26.Lerman LO, Nath KA, Rodriguez-Porcel M, et al. Increased oxidative stress in experimental renovascular hypertension. Hypertension. 2001 February;37(2 Part 2):541–546. doi: 10.1161/01.hyp.37.2.541. [DOI] [PubMed] [Google Scholar]
  • 27.Oliveira-Sales EB, Nishi EE, Carillo BA, et al. Oxidative stress in the sympathetic premotor neurons contributes to sympathetic activation in renovascular hypertension. Am J Hypertens. 2009 May;22(5):484–492. doi: 10.1038/ajh.2009.17. [DOI] [PubMed] [Google Scholar]
  • 28.Dobrian AD, Davies MJ, Schriver SD, Lauterio TJ, Prewitt RL. Oxidative stress in a rat model of obesity-induced hypertension. Hypertension. 2001 February;37(2 Part 2):554–560. doi: 10.1161/01.hyp.37.2.554. [DOI] [PubMed] [Google Scholar]
  • 29.Lopez-Campistrous A, Hao L, Xiang W, et al. Mitochondrial dysfunction in the hypertensive rat brain: respiratory complexes exhibit assembly defects in hypertension. Hypertension. 2008 February;51(2):412–419. doi: 10.1161/HYPERTENSIONAHA.107.102285. [DOI] [PubMed] [Google Scholar]
  • 30.Ito H, Torii M, Suzuki T. Decreased superoxide dismutase activity and increased superoxide anion production in cardiac hypertrophy of spontaneously hypertensive rats. Clin Exp Hypertens. 1995 July;17(5):803–816. doi: 10.3109/10641969509033636. [DOI] [PubMed] [Google Scholar]
  • 31.Callera GE, Tostes RC, Yogi A, Montezano AC, Touyz RM. Endothelin-1-induced oxidative stress in DOCA-salt hypertension involves NADPH-oxidase-independent mechanisms. Clin Sci (Lond) 2006 February;110(2):243–253. doi: 10.1042/CS20050307. [DOI] [PubMed] [Google Scholar]
  • 32.Addabbo F, Montagnani M, Goligorsky MS. Mitochondria and reactive oxygen species. Hypertension. 2009 June;53(6):885–892. doi: 10.1161/HYPERTENSIONAHA.109.130054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chandel NS, McClintock DS, Feliciano CE, et al. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem. 2000 August 18;275(33):25130–25138. doi: 10.1074/jbc.M001914200. [DOI] [PubMed] [Google Scholar]
  • 34.Doughan AK, Harrison DG, Dikalov SI. Molecular mechanisms of angiotensin II-mediated mitochondrial dysfunction: linking mitochondrial oxidative damage and vascular endothelial dysfunction. Circ Res. 2008 February 29;102(4):488–496. doi: 10.1161/CIRCRESAHA.107.162800. [DOI] [PubMed] [Google Scholar]
  • 35.Widder JD, Fraccarollo D, Galuppo P, et al. Attenuation of angiotensin II-induced vascular dysfunction and hypertension by overexpression of Thioredoxin 2. Hypertension. 2009 August;54(2):338–344. doi: 10.1161/HYPERTENSIONAHA.108.127928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zhang H, Luo Y, Zhang W, et al. Endothelial-specific expression of mitochondrial thioredoxin improves endothelial cell function and reduces atherosclerotic lesions. Am J Pathol. 2007 March;170(3):1108–1120. doi: 10.2353/ajpath.2007.060960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gao S, Chen J, Brodsky SV, et al. Docking of endothelial nitric oxide synthase (eNOS) to the mitochondrial outer membrane: a pentabasic amino acid sequence in the autoinhibitory domain of eNOS targets a proteinase K-cleavable peptide on the cytoplasmic face of mitochondria. J Biol Chem. 2004 April 16;279(16):15968–15974. doi: 10.1074/jbc.M308504200. [DOI] [PubMed] [Google Scholar]
  • 38.Landmesser U, Dikalov S, Price SR, et al. Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J Clin Invest. 2003 April;111(8):1201–1209. doi: 10.1172/JCI14172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Addabbo F, Ratliff B, Park HC, et al. The Krebs cycle and mitochondrial mass are early victims of endothelial dysfunction: proteomic approach. Am J Pathol. 2009 January;174(1):34–43. doi: 10.2353/ajpath.2009.080650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.McNally JS, Davis ME, Giddens DP, et al. Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide production in response to oscillatory shear stress. Am J Physiol Heart Circ Physiol. 2003 December;285(6):H2290–H2297. doi: 10.1152/ajpheart.00515.2003. [DOI] [PubMed] [Google Scholar]
  • 41.Viel EC, Benkirane K, Javeshghani D, Touyz RM, Schiffrin EL. Xanthine oxidase and mitochondria contribute to vascular superoxide anion generation in DOCA-salt hypertensive rats. Am J Physiol Heart Circ Physiol. 2008 July;295(1):H281–H288. doi: 10.1152/ajpheart.00304.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sandrim VC, de Syllos RW, Lisboa HR, Tres GS, Tanus-Santos JE. Endothelial nitric oxide synthase haplotypes affect the susceptibility to hypertension in patients with type 2 diabetes mellitus. Atherosclerosis. 2006 November;189(1):241–246. doi: 10.1016/j.atherosclerosis.2005.12.011. [DOI] [PubMed] [Google Scholar]
  • 43.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 October;20(10):772–779. doi: 10.1038/sj.jhh.1002057. [DOI] [PubMed] [Google Scholar]
  • 44.Sandrim VC, de Syllos RW, Lisboa HR, Tres GS, Tanus-Santos JE. Influence of eNOS haplotypes on the plasma nitric oxide products concentrations in hypertensive and type 2 diabetes mellitus patients. Nitric Oxide. 2007 May;16(3):348–355. doi: 10.1016/j.niox.2006.12.007. [DOI] [PubMed] [Google Scholar]
  • 45.Dengel DR, Brown MD, Ferrell RE, Reynolds TH, Supiano MA. A preliminary study on T-786C endothelial nitric oxide synthase gene and renal hemodynamic and blood pressure responses to dietary sodium. Physiol Res. 2007;56(4):393–401. doi: 10.33549/physiolres.931002. [DOI] [PubMed] [Google Scholar]
  • 46.San JG, Moreno MU, Olivan S, et al. Functional effect of the p22phox −930A/G polymorphism on p22phox expression and NADPH oxidase activity in hypertension. Hypertension. 2004 August;44(2):163–169. doi: 10.1161/01.HYP.0000134790.02026.e4. [DOI] [PubMed] [Google Scholar]
  • 47.Sawada T, Kishimoto T, Osaki Y, et al. Relation of the Glu298Asp polymorphism of the nitric oxide synthase gene to hypertension and serum cholesterol in Japanese workers. Prev Med. 2008 August;47(2):167–171. doi: 10.1016/j.ypmed.2008.04.006. [DOI] [PubMed] [Google Scholar]
  • 48.Augeri AL, Tsongalis GJ, Van Heest JL, Maresh CM, Thompson PD, Pescatello LS. The endothelial nitric oxide synthase −786 T>C polymorphism and the exercise-induced blood pressure and nitric oxide responses among men with elevated blood pressure. Atherosclerosis. 2009 June;204(2):e28–e34. doi: 10.1016/j.atherosclerosis.2008.12.015. [DOI] [PubMed] [Google Scholar]
  • 49.Ji Q, Ikegami H, Fujisawa T, et al. A common polymorphism of uncoupling protein 2 gene is associated with hypertension. J Hypertens. 2004 January;22(1):97–102. doi: 10.1097/00004872-200401000-00018. [DOI] [PubMed] [Google Scholar]
  • 50.Chaves FJ, Corella D, Blesa S, et al. Xanthine oxidoreductase polymorphisms: influence in blood pressure and oxidative stress levels. Pharmacogenet Genomics. 2007 August;17(8):589–596. doi: 10.1097/01.fpc.0000239970.23723.38. [DOI] [PubMed] [Google Scholar]
  • 51.Wolin MS. Reactive oxygen species and the control of vascular function. Am J Physiol Heart Circ Physiol. 2009 March;296(3):H539–H549. doi: 10.1152/ajpheart.01167.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Moore TJ, Vollmer WM, Appel LJ, et al. Effect of dietary patterns on ambulatory blood pressure: results from the Dietary Approaches to Stop Hypertension (DASH) Trial. DASH Collaborative Research Group. Hypertension. 1999 September;34(3):472–477. doi: 10.1161/01.hyp.34.3.472. [DOI] [PubMed] [Google Scholar]
  • 53.Conlin PR, Chow D, Miller ER, III, et al. The effect of dietary patterns on blood pressure control in hypertensive patients: results from the Dietary Approaches to Stop Hypertension (DASH) trial. Am J Hypertens. 2000 September;13(9):949–955. doi: 10.1016/s0895-7061(99)00284-8. [DOI] [PubMed] [Google Scholar]
  • 54.John JH, Ziebland S, Yudkin P, Roe LS, Neil HA. Effects of fruit and vegetable consumption on plasma antioxidant concentrations and blood pressure: a randomised controlled trial. Lancet. 2002 June 8;359(9322):1969–1974. doi: 10.1016/s0140-6736(02)98858-6. [DOI] [PubMed] [Google Scholar]
  • 55.Parikh A, Lipsitz SR, Natarajan S. Association between a DASH-like diet and mortality in adults with hypertension: findings from a population-based follow-up study. Am J Hypertens. 2009 April;22(4):409–416. doi: 10.1038/ajh.2009.10. [DOI] [PubMed] [Google Scholar]
  • 56.The effect of vitamin E beta carotene on the incidence of lung cancer and other cancers in male smokers. The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. N Engl J Med. 1994 April 14;330(15):1029–1035. doi: 10.1056/NEJM199404143301501. [DOI] [PubMed] [Google Scholar]
  • 57.MRC/BHF Heart Protection Study of antioxidant vitamin supplementation in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet. 2002 July 6;360(9326):23–33. doi: 10.1016/S0140-6736(02)09328-5. [DOI] [PubMed] [Google Scholar]
  • 58.Sesso HD, Buring JE, Christen WG, et al. Vitamins E and C in the prevention of cardiovascular disease in men: the Physicians' Health Study II randomized controlled trial. JAMA. 2008 November 12;300(18):2123–2133. doi: 10.1001/jama.2008.600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Stampfer MJ, Hennekens CH, Manson JE, Colditz GA, Rosner B, Willett WC. Vitamin E consumption and the risk of coronary disease in women. N Engl J Med. 1993;328:1444–1449. doi: 10.1056/NEJM199305203282003. [DOI] [PubMed] [Google Scholar]
  • 60.Stephens NG, Parsons A, Schofield PM, Kelly F, Cheeseman K, Mitchinson MJ. Randomised controlled trial of vitamin E in patients with coronary disease: Cambridge Heart Antioxidant Study (CHAOS) Lancet. 1996;347:781–786. doi: 10.1016/s0140-6736(96)90866-1. [DOI] [PubMed] [Google Scholar]
  • 61.Rimm EB, Stampfer MJ, Ascherio A, Giovannucci E, Colditz GA, Willett WC. Vitamin E consumption and the risk of coronary heart disease in men. N Engl J Med. 1993;328:1450–1456. doi: 10.1056/NEJM199305203282004. [DOI] [PubMed] [Google Scholar]
  • 62.Lee IM, Cook NR, Gaziano JM, et al. Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women's Health Study: a randomized controlled trial. JAMA. 2005 July 6;294(1):56–65. doi: 10.1001/jama.294.1.56. [DOI] [PubMed] [Google Scholar]
  • 63.Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst Rev. 2008;(2):CD007176. doi: 10.1002/14651858.CD007176. [DOI] [PubMed] [Google Scholar]
  • 64.Bowry VW, Stocker R. Tocopherol-mediated peroxidation. the prooxidant effect of vitamin E on the radical-initiated oxidation of human low-density lipoprotein. J Am Chem Soc. 1993;115:6029–6044. [Google Scholar]
  • 65.Weinberg RB, VanderWerken BS, Anderson RA, Stegner JE, Thomas MJ. Pro-oxidant effect of vitamin E in cigarette smokers consuming a high polyunsaturated fat diet. Arterioscler Thromb Vasc Biol. 2001 June;21(6):1029–1033. doi: 10.1161/01.atv.21.6.1029. [DOI] [PubMed] [Google Scholar]
  • 66.Salonen JT, Nyyssonen K, Salonen R, et al. Antioxidant Supplementation in Atherosclerosis Prevention (ASAP) study: a randomized trial of the effect of vitamins E and C on 3-year progression of carotid atherosclerosis. J Intern Med. 2000 November;248(5):377–386. doi: 10.1046/j.1365-2796.2000.00752.x. [DOI] [PubMed] [Google Scholar]
  • 67.Gotoh N, Niki E. Rates of interactions of superoxide with vitamin E, vitamin C, and related compounds as measured by chemiluminescence. Biochim Biophys Acta. 1992;1115:201–207. doi: 10.1016/0304-4165(92)90054-x. [DOI] [PubMed] [Google Scholar]
  • 68.Münzel T, Keaney JF., Jr Are ACE-inhibitors a "magic bullet" against oxidative stress? Circulation. 2001;104(13):1571–1574. doi: 10.1161/hc3801.095585. [DOI] [PubMed] [Google Scholar]
  • 69.Ristow M, Zarse K, Oberbach A, et al. Antioxidants prevent health-promoting effects of physical exercise in humans. Proc Natl Acad Sci U S A. 2009 May 26;106(21):8665–8670. doi: 10.1073/pnas.0903485106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Stamler J, Liu K, Ruth KJ, Pryer J, Greenland P. Eight-year blood pressure change in middle-aged men: relationship to multiple nutrients. Hypertension. 2002 May;39(5):1000–1006. doi: 10.1161/01.hyp.0000016178.80811.d9. [DOI] [PubMed] [Google Scholar]
  • 71.Siems W, Sommerburg O, Schild L, Augustin W, Langhans CD, Wiswedel I. Beta-carotene cleavage products induce oxidative stress in vitro by impairing mitochondrial respiration. FASEB J. 2002 August;16(10):1289–1291. doi: 10.1096/fj.01-0765fje. [DOI] [PubMed] [Google Scholar]
  • 72.Hennekens CH, Buring JE, Manson JE, et al. Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease. N Engl J Med. 1996;334:1145–1149. doi: 10.1056/NEJM199605023341801. [DOI] [PubMed] [Google Scholar]
  • 73.Upritchard JE, Sutherland WH, Mann JI. Effect of supplementation with tomato juice, vitamin E, and vitamin C on LDL oxidation and products of inflammatory activity in type 2 diabetes. Diabetes Care. 2000 June;23(6):733–738. doi: 10.2337/diacare.23.6.733. [DOI] [PubMed] [Google Scholar]
  • 74.Engelhard YN, Gazer B, Paran E. Natural antioxidants from tomato extract reduce blood pressure in patients with grade-1 hypertension: a double-blind, placebo-controlled pilot study. Am Heart J. 2006 January;151(1):100. doi: 10.1016/j.ahj.2005.05.008. [DOI] [PubMed] [Google Scholar]
  • 75.Ried K, Frank OR, Stocks NP. Dark chocolate or tomato extract for prehypertension: a randomised controlled trial. BMC Complement Altern Med. 2009;9:22. doi: 10.1186/1472-6882-9-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Urivetzky M, Kessaris D, Smith AD. Ascorbic acid overdosing: a risk factor for calcium oxalate nephrolithiasis. J Urol. 1992 May;147(5):1215–1218. doi: 10.1016/s0022-5347(17)37521-3. [DOI] [PubMed] [Google Scholar]
  • 77.Jackson TS, Xu A, Vita JA, Keaney JF., Jr Ascorbate prevents the interaction of superoxide and nitric oxide only at very high physiological concentrations. Circ Res. 1998;83:916–922. doi: 10.1161/01.res.83.9.916. [DOI] [PubMed] [Google Scholar]
  • 78.Levine M, Conry-Cantilena C, Wang Y, et al. Vitamin C pharmacokinetics in healthy volunteers: Evidence for a recommended dietary allowance. Proc Natl Acad Sci USA. 1996;93:3704–3709. doi: 10.1073/pnas.93.8.3704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Stamler JS, Singel DJ, Loscalzo J. Biochemistry of nitric oxide and its redox-activated forms. Science. 1992;258:1898–1902. doi: 10.1126/science.1281928. [DOI] [PubMed] [Google Scholar]
  • 80.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 September;38(3 Pt 2):606–611. doi: 10.1161/hy09t1.094005. [DOI] [PubMed] [Google Scholar]
  • 81.Ulker S, McKeown PP, Bayraktutan U. Vitamins reverse endothelial dysfunction through regulation of eNOS and NAD(P)H oxidase activities. Hypertension. 2003 March;41(3):534–539. doi: 10.1161/01.HYP.0000057421.28533.37. [DOI] [PubMed] [Google Scholar]
  • 82.Muhlhofer A, Mrosek S, Schlegel B, et al. High-dose intravenous vitamin C is not associated with an increase of pro-oxidative biomarkers. Eur J Clin Nutr. 2004 August;58(8):1151–1158. doi: 10.1038/sj.ejcn.1601943. [DOI] [PubMed] [Google Scholar]
  • 83.Ghosh SK, Ekpo EB, Shah IU, Girling AJ, Jenkins C, Sinclair AJ. A double-blind, placebo-controlled parallel trial of vitamin C treatment in elderly patients with hypertension. Gerontology. 1994;40(5):268–272. doi: 10.1159/000213595. [DOI] [PubMed] [Google Scholar]
  • 84.Fotherby MD, Williams JC, Forster LA, Craner P, Ferns GA. Effect of vitamin C on ambulatory blood pressure and plasma lipids in older persons. Journal of Hypertension. 2000;18:411–415. doi: 10.1097/00004872-200018040-00009. [DOI] [PubMed] [Google Scholar]
  • 85.Duffy SJ, Gokce N, Holbrook M, et al. Treatment of hypertension with ascorbic acid. Lancet. 1999;354:2048–2049. doi: 10.1016/s0140-6736(99)04410-4. [DOI] [PubMed] [Google Scholar]
  • 86.Mullan BA, Young IS, Fee H, McCance DR. Ascorbic Acid reduces blood pressure and arterial stiffness in type 2 diabetes. Hypertension. 2002 December;40(6):804–809. doi: 10.1161/01.hyp.0000039961.13718.00. [DOI] [PubMed] [Google Scholar]
  • 87.Darko D, Dornhorst A, Kelly FJ, Ritter JM, Chowienczyk PJ. Lack of effect of oral vitamin C on blood pressure, oxidative stress and endothelial function in Type II diabetes. Clin Sci (Lond) 2002 October;103(4):339–344. doi: 10.1042/cs1030339. [DOI] [PubMed] [Google Scholar]
  • 88.Brigelius-Flohe R, Traber MG. Vitamin E: function and metabolism. FASEB J. 1999 July;13(10):1145–1155. [PubMed] [Google Scholar]
  • 89.McDermott JH. Antioxidant nutrients: current dietary recommendations and research update. J Am Pharm Assoc (Wash ) 2000 November;40(6):785–799. doi: 10.1016/s1086-5802(16)31126-3. [DOI] [PubMed] [Google Scholar]
  • 90.Upston JM, Witting PK, Brown AJ, Stocker R, Keaney JF., Jr Effect of vitamin E on aortic lipid oxidation and intimal proliferation after arterial injury in cholesterol-fed rabbits. Free Radic Biol Med. 2001 November 15;31(10):1245–1253. doi: 10.1016/s0891-5849(01)00721-3. [DOI] [PubMed] [Google Scholar]
  • 91.Azzi A, Ricciarelli R, Zingg JM. Non-antioxidant molecular functions of alpha-tocopherol (vitamin E) FEBS Lett. 2002 May 22;519(1–3):8–10. doi: 10.1016/s0014-5793(02)02706-0. [DOI] [PubMed] [Google Scholar]
  • 92.Yusuf S, Dagenais G, Pogue J, Bosch J, Sleight P. Vitamin E supplementation and cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med. 2000;342:154–160. doi: 10.1056/NEJM200001203420302. [DOI] [PubMed] [Google Scholar]
  • 93.Miller ER, III, 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 January 4;142(1):37–46. doi: 10.7326/0003-4819-142-1-200501040-00110. [DOI] [PubMed] [Google Scholar]
  • 94.Lonn E, Bosch J, Yusuf S, et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: a randomized controlled trial. JAMA. 2005 March 16;293(11):1338–1347. doi: 10.1001/jama.293.11.1338. [DOI] [PubMed] [Google Scholar]
  • 95.Galley HF, Thornton J, Howdle PD, Walker BE, Webster NR. Combination oral antioxidant supplementation reduces blood pressure. Clin Sci. 1997 April;92(4):361–365. doi: 10.1042/cs0920361. [DOI] [PubMed] [Google Scholar]
  • 96.Palumbo G, Avanzini F, Alli C, et al. Effects of vitamin E on clinic and ambulatory blood pressure in treated hypertensive patients. Collaborative Group of the Primary Prevention Project (PPP)--Hypertension study. Am J Hypertens. 2000 May;13(5 Pt 1):564–567. doi: 10.1016/s0895-7061(00)00244-2. [DOI] [PubMed] [Google Scholar]
  • 97.Ward NC, Wu JH, Clarke MW, et al. The effect of vitamin E on blood pressure in individuals with type 2 diabetes: a randomized, double-blind, placebo-controlled trial. J Hypertens. 2007 January;25(1):227–234. doi: 10.1097/01.hjh.0000254373.96111.43. [DOI] [PubMed] [Google Scholar]
  • 98.Tiefenbacher CP. Tetrahydrobiopterin: a critical cofactor for eNOS and a strategy in the treatment of endothelial dysfunction? Am J Physiol Heart Circ Physiol. 2001 June;280(6):H2484–H2488. doi: 10.1152/ajpheart.2001.280.6.H2484. [DOI] [PubMed] [Google Scholar]
  • 99.Stroes E, Hijmering M, van ZM, Wever R, Rabelink TJ, van Faassen EE. Origin of superoxide production by endothelial nitric oxide synthase. FEBS Lett. 1998 November 6;438(3):161–164. doi: 10.1016/s0014-5793(98)01292-7. [DOI] [PubMed] [Google Scholar]
  • 100.Govers R, Rabelink TJ. Cellular regulation of endothelial nitric oxide synthase. Am J Physiol Renal Physiol. 2001 February;280(2):F193–F206. doi: 10.1152/ajprenal.2001.280.2.F193. [DOI] [PubMed] [Google Scholar]
  • 101.Katusic ZS. Vascular endothelial dysfunction: does tetrahydrobiopterin play a role? Am J Physiol Heart Circ Physiol. 2001 September;281(3):H981–H986. doi: 10.1152/ajpheart.2001.281.3.H981. [DOI] [PubMed] [Google Scholar]
  • 102.Schlaich MP, Parnell MM, Ahlers BA, et al. Impaired L-arginine transport and endothelial function in hypertensive and genetically predisposed normotensive subjects. Circulation. 2004 December 14;110(24):3680–3686. doi: 10.1161/01.CIR.0000149748.79945.52. [DOI] [PubMed] [Google Scholar]
  • 103.Wang D, Strandgaard S, Iversen J, Wilcox CS. Asymmetric dimethylarginine, oxidative stress, and vascular nitric oxide synthase in essential hypertension. Am J Physiol Regul Integr Comp Physiol. 2009 February;296(2):R195–R200. doi: 10.1152/ajpregu.90506.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Bevers LM, Braam B, Post JA, et al. Tetrahydrobiopterin, but not L-arginine, decreases NO synthase uncoupling in cells expressing high levels of endothelial NO synthase. Hypertension. 2006 January;47(1):87–94. doi: 10.1161/01.HYP.0000196735.85398.0e. [DOI] [PubMed] [Google Scholar]
  • 105.Matsuoka H, Itoh S, Kimoto M, et al. Asymmetrical dimethylarginine, an endogenous nitric oxide synthase inhibitor, in experimental hypertension. Hypertension. 1997 January;29(1 Pt 2):242–247. doi: 10.1161/01.hyp.29.1.242. [DOI] [PubMed] [Google Scholar]
  • 106.Chen PY, Sanders PW. L-arginine abrogates salt-sensitive hypertension in Dahl/Rapp Rats. J Clin Invest. 1991;88:1559–1567. doi: 10.1172/JCI115467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.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 May;13(5 Pt 1):547–551. doi: 10.1016/s0895-7061(99)00233-2. [DOI] [PubMed] [Google Scholar]
  • 108.Martina V, Masha A, Gigliardi VR, 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 May;31(5):940–944. doi: 10.2337/dc07-2251. [DOI] [PubMed] [Google Scholar]
  • 109.Kelly BS, Alexander JW, Dreyer D, et al. Oral arginine improves blood pressure in renal transplant and hemodialysis patients. JPEN J Parenter Enteral Nutr. 2001 July;25(4):194–202. doi: 10.1177/0148607101025004194. [DOI] [PubMed] [Google Scholar]
  • 110.Gokce N, et al. L-arginine amd hypertension. J Nutr. 2004 October;134(10 Suppl) doi: 10.1093/jn/134.10.2807S. 2807S–28011S. [DOI] [PubMed] [Google Scholar]
  • 111.Loscalzo J. Adverse effects of supplemental L-arginine in atherosclerosis: consequences of methylation stress in a complex catabolism? Arterioscler Thromb Vasc Biol. 2003 January 1;23(1):3–5. doi: 10.1161/01.atv.0000040860.71626.9d. [DOI] [PubMed] [Google Scholar]
  • 112.Persky AM, Brazeau GA. Clinical pharmacology of the dietary supplement creatine monohydrate. Pharmacol Rev. 2001 June;53(2):161–176. [PubMed] [Google Scholar]
  • 113.Tyagi N, Sedoris KC, Steed M, Ovechkin AV, Moshal KS, Tyagi SC. Mechanisms of homocysteine-induced oxidative stress. Am J Physiol Heart Circ Physiol. 2005 December;289(6):H2649–H2656. doi: 10.1152/ajpheart.00548.2005. [DOI] [PubMed] [Google Scholar]
  • 114.Jahangir E, Vita JA, Handy D, et al. The effect of l-arginine and creatine on vascular function and homocysteine metabolism. Vasc Med. 2009 August;14(3):239–248. doi: 10.1177/1358863X08100834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Peters U, Poole C, Arab L. Does tea affect cardiovascular disease? a meta-analysis. Am J Epidemiol. 2001 September 15;154(6):495–503. doi: 10.1093/aje/154.6.495. [DOI] [PubMed] [Google Scholar]
  • 116.Bazzano LA, He J, Ogden LG, et al. Fruit and vegetable intake and risk of cardiovascular disease in US adults: the first National Health and Nutrition Examination Survey Epidemiologic Follow-up Study. Am J Clin Nutr. 2002 July;76(1):93–99. doi: 10.1093/ajcn/76.1.93. [DOI] [PubMed] [Google Scholar]
  • 117.Lotito SB, Fraga CG. (+)−Catechin prevents human plasma oxidation. Free Radic Biol Med. 1998 February;24(3):435–441. doi: 10.1016/s0891-5849(97)00276-1. [DOI] [PubMed] [Google Scholar]
  • 118.Stein JH, Keevil JG, Wiebe DA, Aeschlimann S, Folts JD. Purple grape juice improves endothelial function and reduces the susceptibility of LDL cholesterol to oxidation in patients with coronary artery disease. Circulation. 1999 September 7;100(10):1050–1055. doi: 10.1161/01.cir.100.10.1050. [DOI] [PubMed] [Google Scholar]
  • 119.Aviram M, Fuhrman B. Wine flavonoids protect against LDL oxidation and atherosclerosis. Ann N Y Acad Sci. 2002 May;957:146–161. doi: 10.1111/j.1749-6632.2002.tb02913.x. [DOI] [PubMed] [Google Scholar]
  • 120.Lotito SB, Frei B. Consumption of flavonoid-rich foods and increased plasma antioxidant capacity in humans: cause, consequence, or epiphenomenon? Free Radic Biol Med. 2006 December 15;41(12):1727–1746. doi: 10.1016/j.freeradbiomed.2006.04.033. [DOI] [PubMed] [Google Scholar]
  • 121.Aviram M, Dornfeld L. Pomegranate juice consumption inhibits serum angiotensin converting enzyme activity and reduces systolic blood pressure. Atherosclerosis. 2001 September;158(1):195–198. doi: 10.1016/s0021-9150(01)00412-9. [DOI] [PubMed] [Google Scholar]
  • 122.Aviram M, Rosenblat M, Gaitini D, et al. Pomegranate juice consumption for 3 years by patients with carotid artery stenosis reduces common carotid intima-media thickness, blood pressure and LDL oxidation. Clin Nutr. 2004 June;23(3):423–433. doi: 10.1016/j.clnu.2003.10.002. [DOI] [PubMed] [Google Scholar]
  • 123.Anter E, Thomas SR, Schulz E, Shapira OM, Vita JA, Keaney JF., Jr Activation of eNOS by the p38 MAP kinase in response to black tea polyphenols. J Biol Chem. 2004 August 27;45:46637–46643. doi: 10.1074/jbc.M405547200. [DOI] [PubMed] [Google Scholar]
  • 124.Diebolt M, Bucher B, Andriantsitohaina R. Wine polyphenols decrease blood pressure, improve NO vasodilatation, and induce gene expression. Hypertension. 2001 August;38(2):159–165. doi: 10.1161/01.hyp.38.2.159. [DOI] [PubMed] [Google Scholar]
  • 125.Taubert D, Berkels R, Roesen R, Klaus W. Chocolate and blood pressure in elderly individuals with isolated systolic hypertension. JAMA. 2003 August 27;290(8):1029–1030. doi: 10.1001/jama.290.8.1029. [DOI] [PubMed] [Google Scholar]
  • 126.Grassi D, Lippi C, Necozione S, Desideri G, Ferri C. Short-term administration of dark chocolate is followed by a significant increase in insulin sensitivity and a decrease in blood pressure in healthy persons. Am J Clin Nutr. 2005 March;81(3):611–614. doi: 10.1093/ajcn/81.3.611. [DOI] [PubMed] [Google Scholar]
  • 127.Taubert D, Roesen R, Lehmann C, Jung N, Schomig E. Effects of low habitual cocoa intake on blood pressure and bioactive nitric oxide: a randomized controlled trial. JAMA. 2007 July 4;298(1):49–60. doi: 10.1001/jama.298.1.49. [DOI] [PubMed] [Google Scholar]
  • 128.Grassi D, Desideri G, Necozione S, et al. Blood pressure is reduced and insulin sensitivity increased in glucose-intolerant, hypertensive subjects after 15 days of consuming high-polyphenol dark chocolate. J Nutr. 2008 September;138(9):1671–1676. doi: 10.1093/jn/138.9.1671. [DOI] [PubMed] [Google Scholar]
  • 129.Zilkens RR, Burke V, Hodgson JM, Barden A, Beilin LJ, Puddey IB. Red wine and beer elevate blood pressure in normotensive men. Hypertension. 2005 May;45(5):874–879. doi: 10.1161/01.HYP.0000164639.83623.76. [DOI] [PubMed] [Google Scholar]
  • 130.Hodgson JM, Puddey IB, Burke V, Beilin LJ, Jordan N. Effects on blood pressure of drinking green and black tea. J Hypertens. 1999 April;17(4):457–463. doi: 10.1097/00004872-199917040-00002. [DOI] [PubMed] [Google Scholar]
  • 131.Taubert D, Roesen R, Schomig E. Effect of cocoa and tea intake on blood pressure: a meta-analysis. Arch Intern Med. 2007 April 9;167(7):626–634. doi: 10.1001/archinte.167.7.626. [DOI] [PubMed] [Google Scholar]
  • 132.Overvad K, Diamant B, Holm L, Holmer G, Mortensen SA, Stender S. Coenzyme Q10 in health and disease. Eur J Clin Nutr. 1999 October;53(10):764–770. doi: 10.1038/sj.ejcn.1600880. [DOI] [PubMed] [Google Scholar]
  • 133.McCarty MF. Coenzyme Q versus hypertension: does CoQ decrease endothelial superoxide generation? Med Hypotheses. 1999 October;53(4):300–304. doi: 10.1054/mehy.1997.0761. [DOI] [PubMed] [Google Scholar]
  • 134.Houston MC. Nutraceuticals, vitamins, antioxidants, and minerals in the prevention and treatment of hypertension. Prog Cardiovasc Dis. 2005 May;47(6):396–449. doi: 10.1016/j.pcad.2005.01.004. [DOI] [PubMed] [Google Scholar]
  • 135.Langsjoen P, Langsjoen P, Willis R, Folkers K. Treatment of essential hypertension with coenzyme Q10. Mol Aspects Med. 1994;15 Suppl:S265–S272. doi: 10.1016/0098-2997(94)90037-x. [DOI] [PubMed] [Google Scholar]
  • 136.Digiesi V, Cantini F, Oradei A, et al. Coenzyme Q10 in essential hypertension. Mol Aspects Med. 1994;15 Suppl:s257–s263. doi: 10.1016/0098-2997(94)90036-1. [DOI] [PubMed] [Google Scholar]
  • 137.Yamagami T, Takagi M, Akagami H, Kubo H, Toyama S, Okamoto T. Effect of Coenzyme Q10 on essential hypertension: a double blind control study. Elsvier; 1986. pp. 337–343. [Google Scholar]
  • 138.Digiesi V, Cantini F, Brodbeck D. Effect of conexyme Q10 on essential arterial hypertension. Curr Ther Res. 1990;47(5):841–845. [Google Scholar]
  • 139.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 March;13(3):203–208. doi: 10.1038/sj.jhh.1000778. [DOI] [PubMed] [Google Scholar]
  • 140.Burke BE, Neuenschwander R, Olson RD. Randomized, double-blind, placebo-controlled trial of coenzyme Q10 in isolated systolic hypertension. South Med J. 2001 November;94(11):1112–1117. doi: 10.1097/00007611-200111000-00015. [DOI] [PubMed] [Google Scholar]
  • 141.Graham D, Huynh NN, Hamilton CA, et al. Mitochondria-targeted antioxidant MitoQ10 improves endothelial function and attenuates cardiac hypertrophy. Hypertension. 2009 August;54(2):322–328. doi: 10.1161/HYPERTENSIONAHA.109.130351. [DOI] [PubMed] [Google Scholar]
  • 142.Smith AR, Shenvi SV, Widlansky M, Suh JH, Hagen TM. Lipoic acid as a potential therapy for chronic diseases associated with oxidative stress. Curr Med Chem. 2004 May;11(9):1135–1146. doi: 10.2174/0929867043365387. [DOI] [PubMed] [Google Scholar]
  • 143.Shay KP, Moreau RF, Smith EJ, Smith AR, Hagen TM. Alpha-lipoic acid as a dietary supplement: Molecular mechanisms and therapeutic potential. Biochim Biophys Acta. 2009 October;1790(10):1149–1160. doi: 10.1016/j.bbagen.2009.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Teichert J, Kern J, Tritschler HJ, Ulrich H, Preiss R. Investigations on the pharmacokinetics of alpha-lipoic acid in healthy volunteers. Int J Clin Pharmacol Ther. 1998 December;36(12):625–628. [PubMed] [Google Scholar]
  • 145.Zhang WJ, Frei B. Alpha-lipoic acid inhibits TNF-alpha-induced NF-kappaB activation and adhesion molecule expression in human aortic endothelial cells. FASEB J. 2001 November;15(13):2423–2432. doi: 10.1096/fj.01-0260com. [DOI] [PubMed] [Google Scholar]
  • 146.Vasdev S, Ford CA, Parai S, Longerich L, Gadag V. Dietary alpha-lipoic acid supplementation lowers blood pressure in spontaneously hypertensive rats. J Hypertens. 2000 May;18(5):567–573. doi: 10.1097/00004872-200018050-00009. [DOI] [PubMed] [Google Scholar]
  • 147.Kocak G, Aktan F, Canbolat O, 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 December;13(6):308–318. [PubMed] [Google Scholar]
  • 148.Vasdev S, Ford CA, Parai S, Longerich L, Gadag V. Dietary lipoic acid supplementation prevents fructose-induced hypertension in rats. Nutr Metab Cardiovasc Dis. 2000 December;10(6):339–346. [PubMed] [Google Scholar]
  • 149.Takaoka M, Kobayashi Y, Yuba M, Ohkita M, Matsumura Y. Effects of alpha-lipoic acid on deoxycorticosterone acetate-salt-induced hypertension in rats. Eur J Pharmacol. 2001 July 20;424(2):121–129. doi: 10.1016/s0014-2999(01)01120-7. [DOI] [PubMed] [Google Scholar]
  • 150.Hagen TM, Moreau R, Suh JH, Visioli F. Mitochondrial decay in the aging rat heart: evidence for improvement by dietary supplementation with acetyl-L-carnitine and/or lipoic acid. Ann N Y Acad Sci. 2002 April;959:491–507. doi: 10.1111/j.1749-6632.2002.tb02119.x. [DOI] [PubMed] [Google Scholar]
  • 151.Hagen TM, Ingersoll RT, Wehr CM, et al. Acetyl-L-carnitine fed to old rats partially restores mitochondrial function and ambulatory activity. Proc Natl Acad Sci USA. 1998;95(16):9562–9566. doi: 10.1073/pnas.95.16.9562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Tamamogullari N, Silig Y, Icagasioglu S, Atalay A. Carnitine deficiency in diabetes mellitus complications. J Diabetes Complications. 1999 September;13(5–6):251–253. doi: 10.1016/s1056-8727(99)00052-5. [DOI] [PubMed] [Google Scholar]
  • 153.Grekin RJ, Dumont CJ, Vollmer AP, Watts SW, Webb RC. Mechanisms in the pressor effects of hepatic portal venous fatty acid infusion. Am J Physiol. 1997 July;273(1 Pt 2):R324–R330. doi: 10.1152/ajpregu.1997.273.1.R324. [DOI] [PubMed] [Google Scholar]
  • 154.Umpierrez GE, Smiley D, Robalino G, et al. Intravenous intralipid-induced blood pressure elevation and endothelial dysfunction in obese African-Americans with type 2 diabetes. J Clin Endocrinol Metab. 2009 February;94(2):609–614. doi: 10.1210/jc.2008-1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.McMackin CJ, Widlansky ME, Hamburg NM, et al. Effect of combined treatment with alpha-Lipoic acid and acetyl-L-carnitine on vascular function and blood pressure in patients with coronary artery disease. J Clin Hypertens. 2007;9(4):249–255. doi: 10.1111/j.1524-6175.2007.06052.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Ruggenenti P, Cattaneo D, Loriga G, et al. Ameliorating hypertension and insulin resistance in subjects at increased cardiovascular risk: effects of acetyl-L-carnitine therapy. Hypertension. 2009 September;54(3):567–574. doi: 10.1161/HYPERTENSIONAHA.109.132522. [DOI] [PubMed] [Google Scholar]
  • 157.Dhawan V, Jain S. Garlic supplementation prevents oxidative DNA damage in essential hypertension. Mol Cell Biochem. 2005 July;275(1–2):85–94. doi: 10.1007/s11010-005-0824-2. [DOI] [PubMed] [Google Scholar]
  • 158.Stamler J, Brown IJ, Daviglus ML, et al. Glutamic acid, the main dietary amino acid, and blood pressure: the INTERMAP Study (International Collaborative Study of Macronutrients, Micronutrients and Blood Pressure) Circulation. 2009 July 21;120(3):221–228. doi: 10.1161/CIRCULATIONAHA.108.839241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.El MA, Ismael MA, Lu H, Fantus IG, de CJ, Couture R. Comparative effects of N-acetyl-L-cysteine and ramipril on arterial hypertension, insulin resistance, and oxidative stress in chronically glucose-fed rats. Can J Physiol Pharmacol. 2008 November;86(11):752–760. doi: 10.1139/Y08-090. [DOI] [PubMed] [Google Scholar]
  • 160.Nakamura Y, Yamamoto N, Sakai K, Okubo A, Yamazaki S, Takano T. Purification and characterization of angiotensin I-converting enzyme inhibitors from sour milk. J Dairy Sci. 1995 April;78(4):777–783. doi: 10.3168/jds.S0022-0302(95)76689-9. [DOI] [PubMed] [Google Scholar]
  • 161.Hata Y, Yamamoto M, Ohni M, Nakajima K, Nakamura Y, Takano T. A placebo-controlled study of the effect of sour milk on blood pressure in hypertensive subjects. Am J Clin Nutr. 1996 November;64(5):767–771. doi: 10.1093/ajcn/64.5.767. [DOI] [PubMed] [Google Scholar]
  • 162.Kawashima H. Altered vitamin D metabolism in the kidney of the spontaneously hypertensive rat. Biochem J. 1986 August 1;237(3):893–897. doi: 10.1042/bj2370893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Kahonen M, Nappi S, Jolma P, et al. Vascular influences of calcium supplementation and vitamin D-induced hypercalcemia in NaCl-hypertensive rats. J Cardiovasc Pharmacol. 2003 September;42(3):319–328. doi: 10.1097/00005344-200309000-00002. [DOI] [PubMed] [Google Scholar]
  • 164.Lichtenstein AH, Appel LJ, Brands M, et al. Diet and lifestyle recommendations revision 2006: a scientific statement from the American Heart Association Nutrition Committee. Circulation. 2006 July 4;114(1):82–96. doi: 10.1161/CIRCULATIONAHA.106.176158. [DOI] [PubMed] [Google Scholar]

RESOURCES