Hostname: page-component-76fb5796d-5g6vh Total loading time: 0 Render date: 2024-04-25T16:56:30.877Z Has data issue: false hasContentIssue false

Role of flavonoids and nitrates in cardiovascular health

Published online by Cambridge University Press:  19 January 2017

Julie A. Lovegrove*
Affiliation:
Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Reading RG6 6AP, UK Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Whiteknights, Reading RG6 6AP, UK
Alex Stainer
Affiliation:
Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Whiteknights, Reading RG6 6AP, UK
Ditte A. Hobbs
Affiliation:
Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Reading RG6 6AP, UK Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Whiteknights, Reading RG6 6AP, UK
*
*Corresponding author: Professor J. A. Lovegrove, email j.a.lovegrove@reading.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

CVD remain the leading cause of death globally. Effective dietary strategies for their reduction are of high priority. Increasing evidence suggests that phytochemicals, particularly dietary flavonoids and nitrates, are key modulators of CVD risk reduction through impact on multiple risk factors. The aim of this review is to explore the evidence for the impact of flavonoid- and nitrate-rich foods and supplements on CVD risk, with specific reference to their importance as mediators of vascular health and platelet function. There is accumulating evidence to support benefits of dietary flavonoids on cardiovascular health. Dose-dependent recovery of endothelial function and lowering of blood pressure have been reported for the flavanol (-)-epicatechin, found in cocoa, apples and tea, through production and availability of endothelial nitric oxide (NO). Furthermore, flavonoids, including quercetin and its metabolites, reduce in vitro and ex vivo platelet function via inhibition of phosphorylation-dependent cellular signalling pathways, although further in vivo studies are required to substantiate these mechanistic effects. Hypotensive effects of dietary nitrates have been consistently reported in healthy subjects in acute and chronic settings, although there is less evidence for these effects in patient groups. Proposed mechanisms of actions include endothelial-independent NO availability, which is dependent on the entro-salivary circulation and microbial conversion of dietary nitrate to nitrite in the mouth. In conclusion, flavonoid- and nitrate-rich foods show promising effects on vascular function, yet further randomly controlled studies are required to confirm these findings and to determine effective doses.

Type
Conference on ‘Phytochemicals and health: new perspectives on plant-based nutrition’
Copyright
Copyright © The Authors 2017 

CVD are the leading cause of mortality globally, accounting for about 31 % of deaths each year( 1 ). In the UK, CVD was the second most common cause of death in 2014, responsible for 27 % of all mortalities( Reference Townsend, Bhatnagar and Wilkins 2 ). There are several recognised risk factors for CVD, including raised serum LDL-cholesterol and TAG, low serum HDL-cholesterol, elevated blood pressure (BP), diabetes and obesity many of which can be modified by lifestyle choices, including diet( Reference Lewington, Whitlock and Clarke 3 ). Epidemiological data from the 1970s indicated that CHD rates were higher in countries with low fruit and vegetable consumption( Reference Ness and Powles 4 ). This has been supported by a number of more recent studies that have shown that dietary patterns rich in fruit and vegetables are associated with reduced rates of CHD, stroke and CVD mortality( Reference Joshipura, Hu and Manson 5 Reference Joshipura, Ascherio and Manson 10 ). Some researchers have attempted to identify the types of fruit and vegetables responsible for the reduced risk of CVD. Joshipura et al.( Reference Joshipura, Hu and Manson 5 ) showed that people in the highest quintile of fruit and vegetable intake had a 20 % lower relative risk (RR) for CHD compared with those in the lowest quintile of intake. In addition, each one serving per day increase in fruit and vegetable intake was associated with a 4 % lower risk of CHD. They also found that vitamin C-rich fruits (6 % lower RR per one serving per day increase) and particularly green leafy vegetables (23 % lower RR per one serving per day increase) had the largest protective effects( Reference Joshipura, Hu and Manson 5 ).

Fruit and vegetables are a rich source of phytochemicals such as flavonoids and dietary nitrates, which have been shown to independently exert a number of health effects and could be responsible, at least in part, for the apparent protective effects of fruit and vegetable consumption. The aim of this review is to provide a brief overview of evidence related to the effects of flavonoids and dietary nitrates on cardiovascular health with particular reference to vascular and platelet function.

Dietary flavonoids

The main categories of phytochemicals are polyphenols, which include flavonoids, terpenoids, nitrogen-containing alkaloids and sulphur-containing compounds. Flavonoids are produced by plants as secondary metabolites and have biological roles in plant pigmentation, flavour, growth, reproduction, predator and pathogen resistance( Reference Bravo 11 ). They are present in a variety of foods, including vegetables, fruit, nuts, grains, red wine and chocolate, in concentrations that vary due to a number of factors, including environmental stress, such as UV exposure( Reference Ordidge, Garcia-Macias and Battey 12 ). Flavonoids consist of two benzene rings linked by a three-carbon chain (C6–C3–C6) as shown in Fig. 1. The flavonoid classes differ due to the C ring structural differences, number of phenolic hydroxyl groups and their substitutions, and are commonly divided into seven structural subclasses namely: isoflavones, flavanols or catechins, flavanonols, flavonols, flavanones, flavones, anthocyanins and anthocyanidins( Reference Leonarduzzi, Testa and Sottero 13 ) (Fig. 2). The small structural variations between subclasses are related to considerable differences in biological functions.

Fig. 1. Generic structure of a flavonoid consisting of two benzene rings linked by a 3-carbon chain.

Fig. 2. Structure of the seven classes of flavonoids shown as aglycones.

Absorption and metabolism of flavonoids

Flavonoids are commonly found in the diet as conjugated esters, glycosides or polymers, which have limited bioavailability, requiring intestinal enzyme hydrolysis or colonic microbiota fermentation before absorption into the circulation. Aglycones formed in the intestine by cleavage of flavonoid side chains can enter the epithelial cell by passive diffusion( Reference Day, Canada and Diaz 14 ). However, polar glucosides can be actively transported into epithelial cells via the sodium-GLUT1, where they are hydrolysed by intracellular enzymes to the aglycone( Reference Day, Mellon and Barron 15 ). The importance of the latter absorption route is unclear, but glycosylated flavonoids and aglycones have been shown to inhibit the sodium-GLUT1, potentially reducing dietary glucose absorption( Reference Johnston, Clifford and Morgan 16 ). Before transport to the circulation, the aglycones also undergo further metabolism (phase II) and conjugation, including glucuronidation, methylation or sulphation. Efflux of the metabolites back into the intestine also occurs via transporters, including multidrug resistance protein and P-glycoprotein and the GLUT2( Reference Manzano and Williamson 17 ). Further phase II metabolism occurs in the liver via portal vein transportation and further recycling into the intestinal lumen via the enterohepatic recirculation in bile( Reference Donovan, Crespy and Manach 18 ). Some flavonoids, particularly polyphenol sugar conjugates, pass unabsorbed into the colon and are associated with marked modulation of the colonic gut microbiota( Reference Klinder, Shen and Heppel 19 ) and the production of principally small phenolic acid and aromatic catabolites, which are subsequently absorbed into the circulation( Reference Koutsos, Tuohy and Lovegrove 20 ). These metabolites can be subjected to further metabolism in the liver before they are efficiently excreted in the urine in quantities far higher than those that entered the circulation via the intestine( Reference Jaganath, Mullen and Edwards 21 ). Due to the extensive metabolism and rapid excretion, plasma concentrations do not reflect quantitative absorption and total urinary metabolite excretion can be a more valuable biomarker of intake. Evidence for tissue accumulation of polyphenols and their metabolites is very limited and while this cannot be ruled out, it is believed that frequent ingestion of flavonoid-rich foods is required to maintain constant circulating levels (see review( Reference Del Rio, Rodriguez-Mateos and Spencer 22 )). Detailed studies using stable isotopes have allowed the determination of metabolic pathways of certain polyphenol subclasses. Anthocyanins, found in foods such as berries, are reported to have low bioavailability, but recent data have shown they are extensively metabolised to a diverse range of metabolites, which has highlighted a previous underestimation of anthocyanin absorption and metabolism( Reference de Ferrars, Czank and Zhang 23 ).

Flavonoid intake and CVD risk: epidemiological studies

Epidemiological studies have produced strong evidence for the negative association between high fruit and vegetable consumption and CVD mortality( Reference Dauchet, Amouyel and Dallongeville 24 , Reference Hu and Willett 25 ). However, it is difficult to identify the specific mediator(s) of health due to the numerous potential bioactive compounds present in fruit and vegetables. Observational studies suggest that intakes of flavonoids are associated with a decreased risk of CVD, although the findings are not entirely consistent, because of variation in population studied, dose and specific flavonoid consumed. Data from a large post-menopausal cohort identified a negative association between flavonoid-rich diets and CVD mortality( Reference Mink, Scrafford and Barraj 26 ). Further evidence showed intakes of flavanol- and procyanidin-rich foods were associated with decreased risk of chronic non-communicable diseases particularly CVD( Reference Desch, Kobler and Schmidt 27 , Reference Heiss, Keen and Kelm 28 ). In 2005, Arts and Hollman collated data from fifteen prospective cohort studies; of these thirteen provided evidence for a positive association between dietary flavanols, procyanidins, flavones and flavanones and CVD health, with a reduction of CVD mortality of approximately 65 %( Reference Arts and Hollman 29 ). Systematic reviews( Reference Wang, Nie and Zhou 30 Reference Hollman, Geelen and Kromhout 32 ) that have focused on flavonol intake have reported inconsistent findings, including an inverse association between high flavonol intake and CHD or stroke mortality( Reference Huxley and Neil 31 , Reference Hollman, Geelen and Kromhout 32 ) compared with no association between flavonol intake and CHD risk( Reference Wang, Nie and Zhou 30 ). However, a more recent comprehensive systematic review and meta-analysis of fourteen studies identified that intakes of anthocyanidins, proanthocyanidins, flavones, flavanones and flavan-3-ols were associated with lower CVD; RR of 11, 10, 12, 12 and 13 %, respectively, when comparing the highest and lowest categories of intake; with a 5 % lower RR for CVD for every 10 mg/d increment in flavonol intake( Reference Wang, Ouyang and Liu 33 ).

An inverse association between flavanol intake and CVD mortality was initially identified in the Iowa women health study, which followed 34 489 women, free of CVD at study inclusion( Reference Arts, Jacobs and Harnack 34 ) while, a subsequent follow-up found no association between reduced CVD risk and flavanol intake, instead an association with procyanidin intake( Reference Mink, Scrafford and Barraj 26 ). These seemingly contrasting findings were due to the different ways the data from chocolate and seeded grapes were categorised in the dietary assessment, and emphasise the importance of standardisation of dietary assessment, and the possible benefits of using biomarkers of intake. Evidence from prospective cohort studies generally supports the hypothesis that a greater intake of dietary flavonoids is associated with a lower risk of CVD, although there are inconsistencies in potential benefit. Further supportive evidence from well-performed randomly controlled dietary intervention studies is required to establish a direct relationship between different flavonoid sub-groups and CVD risk.

Chronic and acute effects of flavonoid intake on micro- and macrovascular function

The vascular endothelium plays a key role in the regulation of vascular homeostasis, and alterations in endothelial function contribute to the pathogenesis and clinical expression of CVD( Reference Vita and Keaney 35 ). Many factors impact adversely on the endothelium; these include diabetes mellitus, smoking, physical inactivity, ageing, hypertension, systemic inflammation, dyslipidaemia and insulin resistance, with diet being key in modulating endothelial function( Reference Fung, McCullough and Newby 36 , Reference Meigs, Hu and Rifai 37 ). Prospective cohort studies have supported the association between endothelial function and an increased risk of CVD events and have identified the latter as a valuable holistic surrogate marker of CVD risk( Reference Halcox, Donald and Ellins 38 , Reference Vita 39 ). Endothelial dysfunction has been associated with the development of atherosclerosis and CVD( Reference Drexler and Hornig 40 ) and is most commonly measured in the brachial artery by flow-mediated dilatation (FMD), which uses non-invasive ultrasound before and after increasing shear stress by reactive hyperaemia, with the degree of dilation reflecting arterial nitric oxide (NO) release. Another commonly used technique is laser Doppler imaging with iontophoresis, which measures the endothelial function of the peripheral microcirculation. The degree of endothelial dysfunction occurring in the microcirculation has been shown to be proportional to that occurring in the coronary arteries( Reference Stehouwer 41 ). This technique measures the response of cutaneous blood vessels to transdermal delivery of two contrasting vasoactive agents: acetylcholine (endothelium-dependent vasodilator) and sodium nitroprusside (endothelium-independent vasodilator) by iontophoresis. A reduced local vasodilatory response to acetylcholine is associated with endothelial dysfunction( Reference Ramsay, Ferrell and Greer 42 ).

Consumption of fruit rich in anthocyanins and proanthocyanidins in the form of purple grape juice or grape seed extract (for between 14 and 28 d) significantly increased FMD in volunteers with angiographically documented CHD or above average vascular risk( Reference Chong, Macdonald and Lovegrove 43 ). Furthermore, consumption of pomegranate, containing tannins and anthocyanins, for a period of 90 d to 3 years resulted in improvements in carotid intermedia thickness, a measure of the extent of atherosclerosis in the carotid artery, in those with increased CVD risk( Reference Chong, Macdonald and Lovegrove 43 ). The FLAVURS study investigated the dose-dependent effect (+2, +4 and +6 additional portions/d) of flavonoid-rich and flavonoid-poor fruit and vegetables compared with habitual diet, on microvascular reactivity, determined by laser Doppler imaging with iontophoresis and other CVD risk markers. After two additional portions of flavonoid-rich fruit and vegetables, equivalent to an estimated increase in total dietary flavonoids from 36 (sem 5) to 140 (sem 14) mg/d( Reference Chong, George and Alimbetov 44 ), a significant increase in endothelium-dependent microvascular reactivity was observed in men. In addition, reduced C-reactive protein, E-selectin and vascular cell adhesion molecule and increased plasma NOx was observed with four additional flavonoid-rich portions, compared with the control and low-flavonoid intervention( Reference Macready, George and Chong 45 ). These data support vascular improvements reported in a previous study investigating a similar single dose of flavonoid-rich foods( Reference Dohadwala, Holbrook and Hamburg 46 ). Identification of the specific flavonoid bioactive is not possible in studies that include a variety of foods, yet these data demonstrate that dietary relevant doses of total flavonoids can contribute to vascular health and could be considered as useful strategies for CVD risk factor reduction.

The majority of the population is in a postprandial state for most of the day and it is recognised that acute physiological responses to meals are a major contributor to overall CVD risk. Flavonoid-rich foods have been implicated in modulating postprandial responses. For example, blueberries are a rich source of flavonoids, particularly anthocyanin, flavanol oligomer and chlorogenic acid( Reference Rodriguez-Mateos, Del Pino-Garcia and George 47 ). Acute improvements in vascular function, measured by FMD, were observed in healthy men in a time- and dose-dependent manner (up to a concentration of 766 mg total polyphenols)( Reference Rodriguez-Mateos, Rendeiro and Bergillos-Meca 48 ) with little observed effect of processing( Reference Rodriguez-Mateos, Del Pino-Garcia and George 47 ). These beneficial effects on postprandial vascular reactivity are not confined to blueberries, as a mixed fruit puree containing, 457 mg (-)-epicatechin increased microvascular reactivity and plasma NOx ( Reference George, Waroonphan and Niwat 49 ). Although the fruit puree contained varied flavonoids, the potential vascular benefits of (-)-epicatechin are supported by a meta-analysis of six randomly controlled trials, which found that 70–177 mg (-)-epicatechin, from cocoa or chocolate sources significantly increased postprandial FMD by 3·99 % at 90–149 min post-ingestion( Reference Hooper, Kay and Abdelhamid 50 ). These data indicate that different classes of flavonoids in the form of foods can significantly improve postprandial vascular function and possible CVD risk.

Flavanols, as a subgroup of flavonoids, have been extensively studied and increasing evidence has shown that higher intake of flavanol-rich foods improve arterial function in numerous groups including those at risk for CVD, with established CVD( Reference Heiss, Dejam and Kleinbongard 51 ) and more recently healthy young and ageing individuals( Reference Sansone, Rodriguez-Mateos and Heuel 52 ). The mechanisms of action are not totally understood, but causality between intake and an improvement in arterial function has been demonstrated( Reference Schroeter, Heiss and Balzer 53 ). The important dietary flavonol, (-)-epicatechin, is naturally present is high concentrations in cocoa, apples and tea and a number of systematic reviews and meta-analyses, including a recent study of forty-two randomised controlled human dietary intervention studies on supplemental and flavan-3-ols-rich chocolate and cocoa, reported significant acute and chronic (up to 18 weeks) dose-dependent cardiovascular benefits, including recovery of endothelial function, lowering of BP and some improvements in insulin sensitivity and serum lipids( Reference Hooper, Kay and Abdelhamid 50 , Reference Ried, Sullivan and Fakler 54 , Reference Hooper, Kroon and Rimm 55 ). Furthermore green and black tea (rich in (-)-epicatechin) was also reported to reduce BP and LDL-cholesterol in a systematic review and meta-analysis of a small number of studies, but these findings need confirmation in long-term trials, with low risk of bias( Reference Hartley, Flowers and Holmes 56 ). The extensive studies into the vascular effects of (-)-epicatechin and their impact on other CVD risk markers has prompted some to propose specific dietary recommendation for these flanonoids and a broader recommendation on flavanol-rich fruit and vegetables for CVD risk reduction, although further evidence may be required before specific recommendations are considered.( Reference Schroeter, Heiss and Spencer 57 )

Possible mechanisms of flavonoids and vascular effects

Despite the high antioxidant potential of a number of classes of flavonoids, there is limited evidence to support this mechanism of action due to the low plasma concentrations of flavonoids compared with other endogenous or exogenous antioxidants( Reference Hollman, Cassidy and Comte 58 ). Many of the vascular effects of flavonoids have been associated with molecular signalling cascades and related regulation of cellular function. One of the potential mechanisms of action is the association between flavonoid, particularly (-)-epicatechin, and prolonged, augmented NO synthesis, the primary modulator of vascular dilation( Reference Schroeter, Heiss and Balzer 53 ). NO production from L-arginine is regulated by three NO synthase (NOS) enzymes: endothelial NOS (eNOS), neuronal NOS and inducible NOS with lower production and/or availability of NO as the main effect on endothelial dysfunction. Several in vitro and human studies have reported potent vasorelaxant activity of certain flavonoids related to activation of eNOS( Reference Schroeter, Heiss and Balzer 53 , Reference Almeida Rezende, Pereira and Cortes 59 ). A common polymorphism in the eNOS gene is the Glu298Asp SNP that modifies its coding sequence, replacing a glutamate residue at position 298 with an aspartate residue. This polymorphism has been linked to increased risk of cardiovascular events putatively through reduced NO production by eNOS( Reference Hingorani, Liang and Fatibene 60 , Reference Tian, Zeng and Wang 61 ). Interestingly, in a small acute randomised control study a significant genotype interaction with endothelium-dependent microvascular dilation was observed after consumption of fruit and vegetable puree containing 456 mg (-)-epicatichin. Wild-type, GG, participants (non-risk group) showed an increased endothelial vasodilation at 180 min compared with control, with no effect in T allele carriers. This supports the importance of (-)-epicatichin in eNOS activation and NO availability, with little vascular effect of (-)-epicatichin in those with impaired eNOS function. This nutrient–gene interaction may explain in part, the large variation in individual vascular responses to flavonoid consumption, but requires further confirmatory studies( Reference George, Waroonphan and Niwat 62 ).

Flavonoids have also been reported to modulate xanthine oxidase activity, resulting in decreased oxidative injury and consequential increased NO( Reference Cos, Ying and Calomme 63 ). The vascular effects induced by phenolics may also be mediated by the inhibition of Ca2+ channels and/or the blockage of the protein kinase C-mediated contractile mechanism, as has been observed for caffeic acid phenyl ester and sodium ferulate, respectively( Reference Chen, Ye and Li 64 ). Furthermore, benefits may be mechanistically linked to the actions of circulating phenolic metabolites on inhibition of neutrophil nicotinamide adenine dinucleotide phosphate oxidase activity, which prevents NO degradation and increases its availability( Reference Rodriguez-Mateos, Rendeiro and Bergillos-Meca 48 ). More recently a possible role of flavonoid promotion of endothelium-derived hyperpolarising factor in vasodilation, which induces hyperpolarisation, thus leading to dilation of the vascular smooth muscle cell has been identified( Reference Ndiaye, Chataigneau and Chataigneau 65 ). In summary, there are multiple potential mechanisms by which flavonoids and their metabolites can modulate vascular function( Reference Mladenka, Zatloukalova and Filipsky 66 ) and these may act in an additive or synergistic manner. It is evident that dietary relevant doses of flavonoids are associated with vascular benefit with varied proposed modulating mechanisms that require elucidation in further studies.

Flavonoids and platelet aggregation

Platelets are small nucleated cell fragments that are produced by megakaryocytes in the bone marrow( Reference Italiano, Lecine and Shivdasani 67 , Reference Patel, Hartwig and Italiano 68 ) and play a critical role in haemostasis through formation of aggregates over arterial wall injuries( Reference Ruggeri 69 ). When platelet activation becomes impaired, thrombosis can occur, a pathophysiological condition, which can lead to blockage of coronary arteries or impaired blood supply to the brain, leading to events such as myocardial infarction or stroke( Reference Gibbins 70 ). Many studies have previously shown the ability of flavonoids to inhibit platelet function( Reference Guerrero, Lozano and Castillo 71 Reference Gadi, Bnouham and Aziz 73 ). Quercetin is found in many foods such as apples, onions, tea and wine, and present in significant quantities in many diets( Reference Hertog, Hollman and Katan 74 ). Further understanding of how quercetin modulates platelet function is of relevance to establish a mechanistic link between flavonols and CVD risk.

Hubbard et al.( Reference Hubbard, Wolffram and Lovegrove 75 ) observed a significant inhibition of ex vivo platelet aggregation after ingestion of quercetin-4′-O-β-D-glucoside at a dose of 150 mg and 300 mg, with peak quercetin metabolite concentrations of 4·66 µm and 9·72 µm, respectively. These data were supported by a further small human study, which reported peak plasma quercetin metabolite concentrations of 2·59 µm and significant inhibition of ex vivo collagen-stimulated platelet aggregation 60 and 240 min after consumption of a high-quercetin onion soup rich in quercetin glucosides (68·8 mg total quercetin) compared with a matched low quercetin onion control (4·1 mg total quercetin)( Reference Hubbard, Wolffram and de Vos 76 ). Inhibition of spleen tyrosine kinase and phospholipase Cγ2, two key platelet proteins involved with the collagen-stimulated signalling pathway were also observed, and confirms this as one potential mechanism of action. These data are in agreement with previous in vitro studies displaying the ability of quercetin to inhibit collagen, ADP and thrombin-stimulated platelet aggregation, as well as inhibiting collagen-stimulated mitogen-activated protein kinases and phosphoinositide 3-kinase phosphorylation( Reference Oh, Endale and Park 77 , Reference Hubbard, Stevens and Cicmil 78 ).

Flavonoids undergo significant endogenous metabolism and it is important to determine the bioactivity of metabolites as well as the aglycones by understanding structure–activity relationships and how functional groups affect platelet function. Anti-platelet effects of tamarixetin, quercetin-3-sulphate and quercetin-3-glucuronide, as well as the structurally distinct flavonoids apigenin and catechin, quercetin and its plasma metabolites were determined. Quercetin and apigenin significantly inhibited collagen-stimulated platelet aggregation and 5-hydroxytryptamine secretion with similar potency, and logIC50 values for inhibition of aggregation of −5·17 (sem 0·04) and −5·31 (sem 0·04), respectively( Reference Wright, Moraes and Kemp 79 ). Flavones (such as apigenin) are characterised by a non-hydroxylated C-ring, whereas the C-ring of flavonols (e.g. quercetin) contain a C-3 hydroxyl group (Fig. 1). Catechin was less effective, with an inhibitory potency two orders of magnitude lower than quercetin, suggesting that in vivo, metabolites of quercetin and apigenin may be more relevant in the inhibition of platelet function. Flavan-3-ols such as catechin possess a non-planar, C-3 hydroxylated C ring, which is not substituted with a C-4 carbonyl group (as is found in flavonols). Quercetin-3-sulphate and tamarixetin (a methylated quercetin metabolite) were less potent than quercetin, with a reduction from high to moderate potency upon addition of a C-4′ methyl or C-3′-sulphate group, but at concentrations above 20 µm, all achieved substantial inhibition of platelet aggregation and 5-hydroxytryptamine release. Quercetin-3-glucuronide caused much lower levels of inhibition, providing evidence for reduced potency upon glucuronidation of the C ring. Jasuja et al. have shown quercetin-3-glucuronide to potently inhibit protein disulphide isomerase, an oxidoreductase important in thrombus formation( Reference Jasuja, Passam and Kennedy 80 ). X-ray crystallographic analyses of flavonoid-kinase complexes have shown that flavonoid ring systems and the hydroxyl groups are important features for kinase binding( Reference Lu, Chang and Baratte 81 , Reference Wright, Watson and McGuffin 82 ) supporting the evidence for structure-specific effects on platelet function. Taken together, this evidence shows the importance of understanding the structural differences of flavonoids, and how specific functional groups on polyphenols can lead to enhanced or reduced effects in different stages of haemostasis and thrombosis. This evidence may also facilitate the design of small-molecular inhibitors and inform specific dietary advice.

In summary, flavonoids are generally poorly absorbed and substantially metabolised to aid rapid elimination. Many flavonoid subgroups reach the colon in their native state, and are fermented by the microbiota, which produces small phenolic metabolites with potential bioactivity after absorption. CVD risk reduction from high fruit and vegetable intake may be due, in part, to benefits from flavonoid ingestion. In particular, (-)-epicatechin, a key flavanol, has been causally linked with increased arterial endothelial-dependent dilation measured by FMD, with a putative increase in NO bioavailability. Other potential mechanisms of action include modulation of NADPH oxidase activity and reduction of NO degradation. Furthermore, flavonoids, particularly quercetin and its metabolites, reduce in vitro and ex vivo platelet function, possibly via inhibiting phosphorylation in cell signalling cascades. Further research will be required to determine the biological effects of flavonoid subgroups in vivo, and the minimal effective dose of these compounds before it is possible to make any specific dietary recommendations.

Inorganic nitrate and nitrite

Inorganic nitrate and nitrite were previously considered largely inactive by products of the oxidation of NO endogenously. However, emerging evidence suggest these anions are important storage forms of NO, which can be reduced to bioactive NO under certain conditions. Nitrate is particularly abundant in vegetables such as beetroot and green leafy varieties (spinach, lettuce and rocket) where it is absorbed from the soil and transported to the leaf where it accumulates. Nitrate is important for plant function and is the main growth-limiting factor. In UK diets, estimates from 1997 suggest that the average nitrate intake is approximately 52 mg/d, with vegetables being the main source of nitrate, contributing about 70 % of daily intakes with the remaining nitrate derived from drinking-water( Reference Ysart, Miller and Barrett 83 ) (Fig. 3).

Fig. 3. Diagram of inorganic nitrate metabolism via the nitrate–nitrite–nitric oxide (NO) pathway (adapted from Hobbs et al.( Reference Hobbs, George and Lovegrove 115 )). A proportion of ingested nitrate (NO3 , - - -▸) is converted directly to nitrite (NO2 , →) by facultative anaerobic bacteria, that reside in plaque and on the dorsum of the tongue, during mastication in the mouth (a); the remainder is swallowed and is rapidly absorbed from the upper gastrointestinal tract. Approximately 25 % is removed from the circulation and concentrated in the salivary glands and re-secreted into the mouth, where it is reduced to nitrite. Some of the salivary nitrite enters the acidic environment of the stomach once swallowed (b), where NO is produced non-enzymically from nitrite after formation of nitrous acid (HNO2) and then NO and other nitrogen oxides. The NO generated kills pathogenic bacteria and stimulates mucosal blood flow and mucus generation. The remaining nitrite is absorbed into the circulation; in blood vessels (c) nitrite forms vasodilatory NO after a reaction with deoxygenated Hb (deoxy-Hb). Approximately 60 % of ingested nitrate is excreted in urine within 48 h. Oxy-Hb, oxygenated Hb.

The consumption of inorganic nitrate either from dietary or supplemental sources have been shown to exert a number of important vascular effects such as BP lowering, protection against ischemia-reperfusion injury, inhibiting platelet aggregation, preserving or improving endothelial dysfunction and enhancing exercise performance( Reference Omar, Webb and Lundberg 84 ).

The nitrate–nitrite–nitric oxide pathway

The continuous generation of NO from L-arginine by the enzymatic action of eNOS in the presence of oxygen within endothelial cells is important for maintenance of vascular homeostasis. Indeed reduced production or bioavailability of NO is associated with a number of cardiovascular and metabolic disorders( Reference Cannon 85 ). The nitrate–nitrite–NO pathway is a NOS and oxygen independent pathway for the generation of bioactive NO, and is an important alternative pathway for NO production, particularly during periods of hypoxia( Reference Lundberg, Weitzberg and Cole 86 ). Ingested nitrate, obtained mainly from green leafy vegetables and beetroot, is readily absorbed in the upper part of the gastrointestinal tract where it mixes with NO produced from NOS( Reference Wagner, Schultz and Deen 87 ).

Circulating concentrations of ingested nitrate peak after approximately 1 h( Reference Webb, Patel and Loukogeorgakis 88 ) and remains elevated for up to 5–6 h post-ingestion. The majority of ingested nitrate (65–75 %) is excreted in urine with a very small proportion of nitrate (<1 %) reaching the large bowl, which is excreted in the faeces( Reference Bartholomew and Hill 89 ). The remaining nitrate is reabsorbed by the salivary glands and concentrated up to 20-fold, reaching concentrations of 10 mm in the saliva( Reference Lundberg and Govoni 90 ). Salivary nitrate is converted to nitrite via a two-electron reduction, a reaction that mammalian cells are unable to perform, during anaerobic respiration by nitrate reductases produced by facultative and obligate anaerobic commensal oral bacteria( Reference Lundberg, Weitzberg and Cole 86 , Reference Duncan, Dougall and Johnston 91 ). The importance of oral bacteria in the nitrate–nitrite–NO pathway has been demonstrated in a number of studies( Reference Webb, Patel and Loukogeorgakis 88 , Reference Petersson, Carlstrom and Schreiber 92 , Reference Kapil, Haydar and Pearl 93 ). When the nitrite rich saliva reaches the acidic environment of the stomach some of it reacts to form nitrous acid, which further decomposes to NO and other reactive nitrogen oxides( Reference Benjamin, O'Driscoll and Dougall 94 ). The remaining nitrite (approximately 95 %) is absorbed into the circulation( Reference Hunault, van Velzen and Sips 95 ) where it forms NO via the action of a number of different nitrite reductases, which have selective activity under oxygen/hypoxic/ischaemic conditions. These include Hb( Reference Cosby, Partovi and Crawford 96 ), myoglobin( Reference Shiva, Huang and Grubina 97 ), cytoglobin and neuroglobin( Reference Petersen, Dewilde and Fago 98 ), xanthine oxidoreductase( Reference Webb, Milsom and Rathod 99 ), aldehyde oxidase( Reference Li, Cui and Kundu 100 ), aldehyde dehydrogenase type 2( Reference Badejo, Hodnette and Dhaliwal 101 ), eNOS( Reference Webb, Milsom and Rathod 99 ), cytochrome P450( Reference Li, Liu and Cui 102 ) and the mitochondrial electron transport chain( Reference Nohl, Staniek and Sobhian 103 ). It is likely that the majority of the cardioprotective effects observed from dietary nitrate consumption are via the conversion of nitrite to NO in blood and tissues.

Vascular effects of dietary nitrate and nitrite

Beneficial effects of nitrate consumption on vascular related function were first identified by Larsen et al.( Reference Larsen, Ekblom and Sahlin 104 ), who showed that supplementation of healthy human subjects for 3 d with sodium nitrate reduced BP. Since then, a number of studies have shown that dietary nitrate-rich vegetable sources such as beetroot juice, spinach, rocket and breads also lower BP and vascular function in healthy subjects( Reference Webb, Patel and Loukogeorgakis 88 , Reference Hobbs, Goulding and Nguyen 105 Reference Jonvik, Nyakayiru and Pinckaers 107 ).

Endothelial dysfunction

A hallmark of endothelial dysfunction is the reduced bioavailability of NO, either through reduced eNOS activity or expression, or via increased NO consumption by free radicals and reactive oxygen species( Reference Vanhoutte 108 ) as discussed earlier. It has been shown that consumption of 500 ml beetroot juice containing 23 mm nitrate reversed the deleterious effects of a mild ischaemia-reperfusion injury to the forearms of healthy subjects and preserved the FMD response, whereas the response was reduced by 60 % in the control subjects( Reference Webb, Patel and Loukogeorgakis 88 , Reference Kapil, Haydar and Pearl 93 ). Hobbs et al.( Reference Hobbs, Goulding and Nguyen 105 ) found that consumption of bread enriched with beetroot increased endothelium-independent blood flow in healthy subjects measured by laser Doppler imaging with iontophoresis. In healthy overweight and slightly obese subjects consumption of 140 ml beetroot juice (500 mg nitrate) or control alongside a mixed meal (57 g fat) attenuated postprandial impairment of FMD( Reference Joris and Mensink 109 ). More recently, daily consumption of dietary nitrate in the form of beetroot juice over a 6-week period resulted in a 1·1 % increase in the FMD response compared with a 0·3 % worsening in the control group( Reference Velmurugan, Gan and Rathod 110 ). However, not all studies have found a beneficial effect of dietary nitrate on endothelial function, with no effects of 250 ml beetroot juice (7·5 mm nitrate) on FMD response in patients with type 2 diabetes( Reference Gilchrist, Winyard and Aizawa 111 ). Furthermore, supplemental potassium nitrate consumption (8 mm nitrate) did not affect FMD response in healthy subjects, although a significant reduction (0·3 m/s) in pulse wave velocity and systolic BP (4 mmHg) at 3 h compared with the potassium chloride control was reported. This suggests that although inorganic nitrate did not alter endothelial function, it did appear to increase blood flow in combination with reductions in BP.

Organic and inorganic nitrate/nitrites are both effective in vascular health, yet it has been proposed that inorganic dietary nitrate may be a more appropriate choice for vascular modulation than organic nitrate supplements( Reference Omar, Artime and Webb 112 ). The enterosalivary circulation is key for the effects of inorganic nitrate and prevents a sudden effect, or toxic circulating concentrations of nitrite, in addition to prolonging the vascular effects. In contrast, supplemental organic nitrate, which does not require the enterosalivary circulation for absorption, has rapid pharmacodynamic responses, causing potent acute effects, immediate vasodilation and in chronic use considerably limited by the development of tolerance and endothelial dysfunction. The more subtle and controlled effects of inorganic nitrate may compensate for diminished endothelial function, and also has no reported tolerance. Therefore, with the increasing recognition of the limitations of organic nitrate supplementation, and continuing discovery of beneficial effects of inorganic nitrate/nitrite, dietary inorganic forms may prove to be the optimum strategy for vascular health( Reference Omar, Artime and Webb 112 ).

Endothelial nitric oxide synthase Glu298Asp polymorphism and nitrate interactions

Variation in response to nitrates could be due to genetic polymorphisms. Healthy men retrospectively genotyped for the Glu298Asp polymorphism (7 GG and 7 T carriers), showed a differential postprandial BP response after consumption of beetroot-enriched bread compared with the control bread. A significantly lower diastolic BP in the T carriers was observed with a concomitant tendency for higher plasma NOx concentration. Despite the small study size these data suggests that carriers of the T allele, which limits endogenous NO production from endothelial eNOS( Reference Liu, Wang and Liu 113 ), were more responsive to dietary nitrate. Crosstalk between NOS-dependent pathway and the nitrate–nitrite–NO pathway in control of vascular NO homeostasis could be a possible explanation for these observations( Reference Carlstrom, Liu and Yang 114 ), although future suitably powered studies are needed to confirm these findings. This nutrient–gene interaction is in contrast to that demonstrated for the same eNOS polymorphism and dietary flavonoids (described earlier), and confirms the differential proposed mechanisms by which flavonoids and nitrates impact on NO availability and vascular function.

Blood pressure

Dietary nitrate has been shown to reduced systolic BP and/or diastolic BP, and increase circulating nitrate/nitrite (see review( Reference Hobbs, George and Lovegrove 115 )). These findings are supported by more recent acute and chronic studies conducted in healthy younger populations (Table 1). A recent meta-analysis of four randomised clinical trials in older adults (55–76 years) revealed that consumption of beetroot juice did not have a significant effect on BP. However, consumption of beetroot juice containing 9·6 mm/d for 3 d( Reference Kelly, Fulford and Vanhatalo 116 ), or 4·8–6·4 mm nitrate/l for 3 weeks( Reference Jajja, Sutyarjoko and Lara 117 ) by older adults (60–70 years) significantly lowered resting systolic BP by 5 and 7·3 mmHg respectively, compared with the control. These inconsistent findings highlight the need for further studies to determine effects in older population groups.

Table 1. The acute and chronic effects of dietary or inorganic nitrate on blood pressure in healthy subjects since 2014

M, male; F, female; y, years; n/a, not available; SBP, systolic blood pressure; DBP, diastolic blood pressure; MAP, mean arterial pressure.

* Refers to differences from baseline.

It was concluded from data collated from eight studies conducted in patient groups that dietary nitrate may help to reduce BP in hypertensive subjects, but not in patients with type 2 diabetes, although only one study could be found in the latter population group( Reference Gee and Ahluwalia 118 ). Furthermore, minimal effects were reported in obese insulin resistant individuals( Reference Fuchs, Nyakayiru and Draijer 119 ), and those with chronic obstructive pulmonary disease despite relatively high doses of dietary nitrate (13·5 and 9·6 mm/d, respectively), although the intervention period was limited (2–3 d)( Reference Shepherd, Wilkerson and Dobson 120 , Reference Leong, Basham and Yong 121 ). In contrast consumption of beetroot juice (7·6 mm/d) by fifteen individuals with chronic obstructive pulmonary disease significantly lowered diastolic BP by 8·2 mmHg (P = 0·019)( Reference Berry, Justus and Hauser 122 ). Additional studies are required to confirm these findings.

Platelet aggregation

Dietary and supplemental nitrate have been reported to significantly reduce platelet aggregation in healthy individuals( Reference Webb, Patel and Loukogeorgakis 88 , Reference Velmurugan, Gan and Rathod 110 , Reference Velmurugan, Kapil and Ghosh 123 ). However, a lack of effect was observed in women from one study. A proposed explanation for this sex difference was reduced soluble guanylyl cyclase activity( Reference Velmurugan, Kapil and Ghosh 123 ), a hypothesis supported by studies in mice( Reference Buys, Sips and Vermeersch 124 , Reference Chan, Bubb and Noyce 125 ), although further conformational studies are required.

Metabolic function

The consumption of sodium nitrate by eNOS deficient mice reversed features of the metabolic syndrome including improvements in BP, bodyweight, abdominal fat accumulation, circulating TAG levels and glucose homeostasis( Reference Carlstrom, Larsen and Nystrom 126 ). The improvements in glucose homeostasis by inorganic nitrate have been shown in a number of other mouse studies( Reference Nystrom, Ortsater and Huang 127 Reference Khalifi, Rahimipour and Jeddi 130 ). For example, Khalifi et al.( Reference Khalifi, Rahimipour and Jeddi 130 ) examined the effects of dietary nitrate in glucose tolerance and lipid profile in type 2 diabetic rats, and found that supplementation of drinking-water with 100 mg/l sodium nitrate prevented an increase in systolic BP and serum glucose, improved glucose tolerance and restored dyslipidaemia in an animal model of hyperglycaemia. A possible mechanism for the beneficial effects of nitrate on glucose homeostasis may be the nitrite-mediated induction of GLUT4 translocation( Reference Jiang, Torregrossa and Potts 131 ), which enhances cellular uptake of glucose. More recent data have also shown that dietary nitrate may increase browning of white adipose tissue, which may have antiobesity and antidiabetic effects( Reference Roberts, Ashmore and Kotwica 132 ). However, there are few studies that have investigated the effects of dietary nitrate on glucose homeostasis in human subjects. Gilchrist et al.( Reference Gilchrist, Winyard and Aizawa 111 ) found that consumption of 250 ml beetroot juice (7·5 mm nitrate) for 2 weeks by individuals with type 2 diabetes increased plasma nitrate and nitrite concentrations, but did not improve insulin sensitivity measured by the hyperinsulinaemic isoglycaemic clamp. In support of this Cermak et al.( Reference Cermak, Hansen and Kouw 133 ) found that acute ingestion of sodium nitrate (0·15 mm nitrate/kg bodyweight) did not attenuate the postprandial rise in plasma glucose or insulin following an oral glucose tolerance test in individuals with type 2 diabetes.

In summary, organic nitrate is now considered to have important benefits on vascular health. While these benefits include the lowering of postprandial and longer-term BP in healthy groups, limited data in patient groups prevent the wider translation of these findings. Nitrate-rich foods have some reported benefits on measures of vascular function, with mechanistic links to increasing endothelial-independent NO availability through the reduction of nitrate to nitrite, and NO. The importance of the entero-salivary circulation and reduction of nitrate to nitrite by oral microbiota is essential for the functional effects of dietary nitrate. Evidence for the more controlled and sustained physiological effects of dietary nitrates on vascular health has prompted consideration of their potential advantage over the rapid effects of nitrate supplements. Further research is required to determine the lowest effective dose and specific mechanisms of action, particularly in patients with hypertension and cardiometabolic disease.

Interactions of nitrate–nitrite with flavonoids

Dietary flavonoids and nitrate affect vascular health by different mechanisms. Flavonoids are proposed to modulate endothelial-dependent NO release, and nitrates impact on NO production from nitrite intermediates and it is possible that their combined consumption may result in additive or synergistic vascular responses. Furthermore formation of NO and other reactive nitrogen species in the stomach is enhanced by increasing nitrite concentrations, lower stomach pH and the presence of vitamin C or polyphenols( Reference Carlsson, Wiklund and Engstrand 134 Reference Peri, Pietraforte and Scorza 136 ). Bondonno et al.( Reference Bondonno, Yang and Croft 106 ) investigated the independent and additive effects of consumption of flavonoid-rich apples and nitrate-rich spinach. They found that the combination of nitrate and flavonoids did not result in additive effects on NO status, endothelial function or BP, although independent effects of flavonoid-rich apples and nitrate-rich spinach on these outcomes were reported. More recently, Rodriguez-Mateos et al.( Reference Rodriguez-Mateos, Hezel and Aydin 137 ) investigated interactions between cocoa flavanols and nitrate, and demonstrated additive effects on FMD response when cocoa flavanols and nitrate were consumed at low doses in combination. In addition, cocoa flavonoids enhanced nitrate-related gastric NO formation, supporting previous studies and suggests nutrient–nutrient interactions may modulate vascular function. Thus there is some evidence to suggest that nitrates and flavonoids, when consumed in combination, may exert additive effects on cardiovascular health, but due to the extremely limited data, confirmatory studies are required.

Conclusions

There is an increasing body of evidence to suggest that dietary flavonoids, particularly flavonols and anthocyanidins, improve vascular function and lower BP at doses achievable in diets that are high in foods such as fruit, vegetables, cocoa and teas. The potential mechanisms of actions are not fully understood, although increased NO availability via endothelial-dependent mechanisms have been proposed as a key modulator. Cell-signalling-mediated mechanisms are also important in both platelet and vascular function. Dietary inorganic nitrates are also dietary modulators of vascular health, primarily through the formation of NO via the nitrate–nitrite–NO pathway. Promising effects of inorganic nitrate consumption on BP in healthy, hypertensive and other patient groups have been identified, although many of the current studies are limited in power and design, particularly those in specific patient groups. It is recognised that greater potential benefit may be gained from dietary nitrates compared with organic supplements, with the latter causing an immediate and severe reduction in BP and endothelial dysfunction. Research is required to determine whether dietary nitrates can be used in combination with hypotensive therapy, which may reduce or eliminate the requirement for medication and the associated side-effects. Consumption of diets rich in flavonoids and nitrates may be important in reducing CVD risk and promoting vascular benefit, although results have been inconsistent and more long-term studies are required to determine dose-dependent effects and the specific mechanisms of action.

Financial Support

None.

Conflict of Interest

None.

Authorship

J. A. L., A. S. and D. A. H. are joint authors of this manuscript.

References

1. World Health Organization (2014) Global status report on noncommunicable diseases 2014. Available at: http://apps.who.int/iris/bitstream/10665/148114/1/9789241564854_eng.pdf?ua=1 (accessed 14 June 2016).Google Scholar
2. Townsend, N, Bhatnagar, P, Wilkins, E et al. (2015) Cardiovascular Disease Statistics, 2015. London: British Heart Foundation.Google Scholar
3. Lewington, S, Whitlock, G, Clarke, R et al. (2007) Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths. Lancet 370, 18291839.Google Scholar
4. Ness, AR & Powles, JW (1997) Fruit and vegetables, and cardiovascular disease: a review. Int J Epidemiol 26, 113.CrossRefGoogle ScholarPubMed
5. Joshipura, KJ, Hu, FB, Manson, JE et al. (2001) The effect of fruit and vegetable intake on risk for coronary heart disease. Ann Intern Med 134, 11061114.Google Scholar
6. Crowe, FL, Roddam, AW, Key, TJ et al. (2011) Fruit and vegetable intake and mortality from ischaemic heart disease: results from the European Prospective Investigation into Cancer and Nutrition (EPIC)-Heart study. Eur Heart J 32, 12351243.Google Scholar
7. Nakamura, K, Nagata, C, Oba, S et al. (2008) Fruit and vegetable intake and mortality from cardiovascular disease are inversely associated in Japanese women but not in men. J Nutr 138, 11291134.CrossRefGoogle Scholar
8. Dauchet, L, Amouyel, P, Hercberg, S et al. (2006) Fruit and vegetable consumption and risk of coronary heart disease: a meta-analysis of cohort studies. J Nutr 136, 25882593.Google Scholar
9. Bazzano, LA, He, J, Ogden, LG et al. (2002) 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 76, 9399.Google Scholar
10. Joshipura, KJ, Ascherio, A, Manson, JE et al. (1999) Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA – J Am Med Assoc 282, 12331239.Google Scholar
11. Bravo, L (1998) Polyphenols: chemistry, dietary sources, metabolism, and nutritional significance. Nutr Rev 56, 317333.Google Scholar
12. Ordidge, M, Garcia-Macias, P, Battey, NH et al. (2012) Development of colour and firmness in strawberry crops is UV light sensitive, but colour is not a good predictor of several quality parameters. J Sci Food Agri 92, 15971604.CrossRefGoogle Scholar
13. Leonarduzzi, G, Testa, G, Sottero, B et al. (2010) Design and development of Nanovehicle-based delivery systems for preventive or Therapeutic supplementation with flavonoids. Curr Med Chem 17, 7495.CrossRefGoogle ScholarPubMed
14. Day, AJ, Canada, FJ, Diaz, JC et al. (2000) Dietary flavonoid and isoflavone glycosides are hydrolysed by the lactase site of lactase phlorizin hydrolase. FEBS Lett 468, 166170.CrossRefGoogle ScholarPubMed
15. Day, AJ, Mellon, F, Barron, D et al. (2001) Human metabolism of dietary flavonoids: identification of plasma metabolites of quercetin. Free Rad Res 35, 941952.CrossRefGoogle ScholarPubMed
16. Johnston, KL, Clifford, MN & Morgan, LM (2003) Coffee acutely modifies gastrointestinal hormone secretion and glucose tolerance in humans: glycemic effects of chlorogenic acid and caffeine. Am J Clin Nutr 78, 728733.Google Scholar
17. Manzano, S & Williamson, G (2010) Polyphenols and phenolic acids from strawberry and apple decrease glucose uptake and transport by human intestinal Caco-2 cells. Mol Nutr Food Res 54, 17731780.CrossRefGoogle ScholarPubMed
18. Donovan, JL, Crespy, V, Manach, C et al. (2001) Catechin is metabolized by both the small intestine and liver of rats. J Nutr 131, 17531757.CrossRefGoogle ScholarPubMed
19. Klinder, A, Shen, Q, Heppel, S et al. (2016) Impact of increasing fruit and vegetables and flavonoid intake on the human gut microbiota. Food Funct 7, 17881796.Google Scholar
20. Koutsos, A, Tuohy, KM & Lovegrove, JA (2015) Apples and cardiovascular health -- is the gut microbiota a core consideration? Nutrients 7, 39593998.CrossRefGoogle ScholarPubMed
21. Jaganath, IB, Mullen, W, Edwards, CA et al. (2006) The relative contribution of the small and large intestine to the absorption and metabolism of rutin in man. Free Rad Res 40, 10351046.Google Scholar
22. Del Rio, D, Rodriguez-Mateos, A, Spencer, JPE et al. (2013) Dietary (Poly)phenolics in Human Health: structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid Redox Signal 18, 18181892.Google Scholar
23. de Ferrars, RM, Czank, C, Zhang, Q et al. (2014) The pharmacokinetics of anthocyanins and their metabolites in humans. Br J Pharmacol 171, 32683282.Google Scholar
24. Dauchet, L, Amouyel, P & Dallongeville, J (2005) Fruit and vegetable consumption and risk of stroke - A meta-analysis of cohort studies. Neurology 65, 11931197.Google Scholar
25. Hu, FB & Willett, WC (2002) Optimal diets for prevention of coronary heart disease. JAMA – J Am Med Assoc 288, 25692578.Google Scholar
26. Mink, PJ, Scrafford, CG, Barraj, LM et al. (2007) Flavonoid intake and cardiovascular disease mortality: a prospective study in postmenopausal women. Am J Clin Nutr 85, 895909.Google Scholar
27. Desch, S, Kobler, D, Schmidt, J et al. (2010) Low vs. higher-dose dark chocolate and blood pressure in cardiovascular high-risk patients. Am J Hypertens 23, 694700.Google Scholar
28. Heiss, C, Keen, CL & Kelm, M (2010) Flavanols and cardiovascular disease prevention. Eur Heart J 31, 2583–U2532.Google Scholar
29. Arts, ICW & Hollman, PCH (2005) Polyphenols and disease risk in epidemiologic studies. Am J Clin Nutr 81, 317s325s.Google Scholar
30. Wang, ZM, Nie, ZL, Zhou, B et al. (2012) Flavonols intake and the risk of coronary heart disease: a meta-analysis of cohort studies. Atherosclerosis 222, 270273.Google Scholar
31. Huxley, RR & Neil, HAW (2003) The relation between dietary flavonol intake and coronary heart disease mortality: a meta-analysis of prospective cohort studies. Eur J Clin Nutr 57, 904908.Google Scholar
32. Hollman, PCH, Geelen, A & Kromhout, D (2010) Dietary Flavonol intake may lower stroke risk in men and women. J Nutr 140, 600604.CrossRefGoogle ScholarPubMed
33. Wang, X, Ouyang, YY, Liu, J et al. (2014) Flavonoid intake and risk of CVD: a systematic review and meta-analysis of prospective cohort studies. Br J Nutr 111, 111.Google Scholar
34. Arts, IC, Jacobs, DR Jr, Harnack, LJ et al. (2001) Dietary catechins in relation to coronary heart disease death among postmenopausal women. Epidemiology 12, 668675.CrossRefGoogle ScholarPubMed
35. Vita, JA & Keaney, JF Jr (2002) Endothelial function: a barometer for cardiovascular risk? Circulation 106, 640642.Google Scholar
36. Fung, TT, McCullough, ML, Newby, PK et al. (2005) Diet-quality scores and plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr 82, 163173.CrossRefGoogle Scholar
37. Meigs, JB, Hu, FB, Rifai, N et al. (2004) Biomarkers of endothelial dysfunction and risk of type 2 diabetes mellitus. JAMA – J Am Med Assoc 291, 19781986.CrossRefGoogle ScholarPubMed
38. Halcox, JP, Donald, AE, Ellins, E et al. (2009) Endothelial function predicts progression of carotid intima-media thickness. Circulation 119, 10051012.Google Scholar
39. Vita, JA (2005) Polyphenols and cardiovascular disease: effects on endothelial and platelet function. Am J Clin Nutr 81, 292S297S.CrossRefGoogle ScholarPubMed
40. Drexler, H & Hornig, B (1999) Endothelial dysfunction in human disease. J Mol Cell Cardiol 31, 5160.Google Scholar
41. Stehouwer, CD (1999) Is measurement of endothelial dysfunction clinically useful? Eur J Clin Invest 29, 459461.Google Scholar
42. Ramsay, JE, Ferrell, WR, Greer, IA et al. (2002) Factors critical to iontophoretic assessment of vascular reactivity: implications for clinical studies of endothelial dysfunction. J Cardiovasc Pharmacol 39, 917.Google Scholar
43. Chong, MFF, Macdonald, R & Lovegrove, JA (2010) Fruit polyphenols and CVD risk: a review of human intervention studies. Br J Nutr 104, S28S39.CrossRefGoogle Scholar
44. Chong, MF, George, TW, Alimbetov, D et al. (2013) Impact of the quantity and flavonoid content of fruits and vegetables on markers of intake in adults with an increased risk of cardiovascular disease: the FLAVURS trial. Eur J Nutr 52, 361378.Google Scholar
45. Macready, AL, George, TW, Chong, MF et al. (2014) Flavonoid-rich fruit and vegetables improve microvascular reactivity and inflammatory status in men at risk of cardiovascular disease--FLAVURS: a randomized controlled trial. Am J Clin Nutr 99, 479489.Google Scholar
46. Dohadwala, MM, Holbrook, M, Hamburg, NM et al. (2011) Effects of cranberry juice consumption on vascular function in patients with coronary artery disease. Am J Clin Nutr 93, 934940.Google Scholar
47. Rodriguez-Mateos, A, Del Pino-Garcia, R, George, TW et al. (2014) Impact of processing on the bioavailability and vascular effects of blueberry (poly)phenols. Mol Nutr Food Res 58, 19521961.Google Scholar
48. Rodriguez-Mateos, A, Rendeiro, C, Bergillos-Meca, T et al. (2013) Intake and time dependence of blueberry flavonoid-induced improvements in vascular function: a randomized, controlled, double-blind, crossover intervention study with mechanistic insights into biological activity. Am J Clin Nutr 98, 11791191.Google Scholar
49. George, TW, Waroonphan, S, Niwat, C et al. (2013) Effects of acute consumption of a fruit and vegetable puree-based drink on vasodilation and oxidative status. Br J Nutr 109, 14421452.CrossRefGoogle ScholarPubMed
50. Hooper, L, Kay, C, Abdelhamid, A et al. (2012) Effects of chocolate, cocoa, and flavan-3-ols on cardiovascular health: a systematic review and meta-analysis of randomized trials. Am J Clin Nutr 95, 740751.Google Scholar
51. Heiss, C, Dejam, A, Kleinbongard, P et al. (2003) Vascular effects of cocoa rich in flavan-3-ols. JAMA – J Am Med Assoc 290, 10301031.CrossRefGoogle Scholar
52. Sansone, R, Rodriguez-Mateos, A, Heuel, J et al. (2015) Cocoa flavanol intake improves endothelial function and Framingham Risk Score in healthy men and women: a randomised, controlled, double-masked trial: the Flaviola Health Study. Br J Nutr 114, 12461255.Google Scholar
53. Schroeter, H, Heiss, C, Balzer, J et al. (2006) (-)-Epicatechin mediates beneficial effects of flavanol-rich cocoa on vascular function in humans. Proc Natl Acad Sci USA 103, 10241029.Google Scholar
54. Ried, K, Sullivan, TR, Fakler, P et al. (2012) Effect of cocoa on blood pressure. Cochrane Database Syst Rev CD008893.Google Scholar
55. Hooper, L, Kroon, PA, Rimm, EB et al. (2008) Flavonoids, flavonoid-rich foods, and cardiovascular risk: a meta-analysis of randomized controlled trials. Am J Clin Nutr 88, 3850.Google Scholar
56. Hartley, L, Flowers, N, Holmes, J et al. (2013) Green and black tea for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev, CD009934.Google Scholar
57. Schroeter, H, Heiss, C, Spencer, JP et al. (2010) Recommending flavanols and procyanidins for cardiovascular health: current knowledge and future needs. Mol Aspects Med 31, 546557.Google Scholar
58. Hollman, PC, Cassidy, A, Comte, B et al. (2011) The biological relevance of direct antioxidant effects of polyphenols for cardiovascular health in humans is not established. J Nutr 141, 989S1009S.Google Scholar
59. Almeida Rezende, B, Pereira, AC, Cortes, SF et al. (2016) Vascular effects of flavonoids. Curr Med Chem 23, 87102.Google Scholar
60. Hingorani, AD, Liang, CF, Fatibene, J et al. (1999) A common variant of the endothelial nitric oxide synthase (Glu298-->Asp) is a major risk factor for coronary artery disease in the UK. Circulation 100, 15151520.Google Scholar
61. Tian, GX, Zeng, XT, Wang, XB et al. (2013) Association between the endothelial nitric oxide synthase gene Glu298Asp polymorphism and coronary heart disease: a metaanalysis of 39 casecontrol studies. Mol Med Rep 7, 13101318.Google Scholar
62. George, TW, Waroonphan, S, Niwat, C et al. (2012) The Glu298Asp single nucleotide polymorphism in the endothelial nitric oxide synthase gene differentially affects the vascular response to acute consumption of fruit and vegetable puree based drinks. Mol Nutr Food Res 56, 10141024.Google Scholar
63. Cos, P, Ying, L, Calomme, M et al. (1998) Structure-activity relationship and classification of flavonoids as inhibitors of xanthine oxidase and superoxide scavengers. J Nat Prod 61, 7176.Google Scholar
64. Chen, GP, Ye, Y, Li, L et al. (2009) Endothelium-independent vasorelaxant effect of sodium ferulate on rat thoracic aorta. Life Sci 84, 8188.Google Scholar
65. Ndiaye, M, Chataigneau, T, Chataigneau, M et al. (2004) Red wine polyphenols induce EDHF-mediated relaxations in porcine coronary arteries through the redox-sensitive activation of the PI3-kinase/Akt pathway. Br J Pharmacol 142, 11311136.Google Scholar
66. Mladenka, P, Zatloukalova, L, Filipsky, T et al. (2010) Cardiovascular effects of flavonoids are not caused only by direct antioxidant activity. Free Radic Biol Med 49, 963975.Google Scholar
67. Italiano, JE Jr, Lecine, P, Shivdasani, RA et al. (1999) Blood platelets are assembled principally at the ends of proplatelet processes produced by differentiated megakaryocytes. J Cell Biol 147, 12991312.CrossRefGoogle ScholarPubMed
68. Patel, SR, Hartwig, JH & Italiano, JE Jr (2005) The biogenesis of platelets from megakaryocyte proplatelets. J Clin Invest 115, 33483354.Google Scholar
69. Ruggeri, ZM (2002) Platelets in atherothrombosis. Nat Med 8, 12271234.Google Scholar
70. Gibbins, JM (2004) Platelet adhesion signalling and the regulation of thrombus formation. J Cell Sci 117, 34153425.Google Scholar
71. Guerrero, JA, Lozano, ML, Castillo, J et al. (2005) Flavonoids inhibit platelet function through binding to the thromboxane A2 receptor. J Thromb Haemost 3, 369376.Google Scholar
72. Tzeng, SH, Ko, WC, Ko, FN et al. (1991) Inhibition of platelet aggregation by some flavonoids. Thromb Res 64, 91100.Google Scholar
73. Gadi, D, Bnouham, M, Aziz, M et al. (2012) Flavonoids purified from parsley inhibit human blood platelet aggregation and adhesion to collagen under flow. J Complement Integr Med 9, Article 19.CrossRefGoogle ScholarPubMed
74. Hertog, MGL, Hollman, PCH & Katan, MB (1992) Content of potentially anticarcinogenic flavonoids of 28 vegetables and 9 fruits commonly consumed in the Netherlands. J Agri Food Chem 40, 23792383.Google Scholar
75. Hubbard, GP, Wolffram, S, Lovegrove, JA et al. (2004) Ingestion of quercetin inhibits platelet aggregation and essential components of the collagen-stimulated platelet activation pathway in humans. J Thromb Haemost 2, 21382145.Google Scholar
76. Hubbard, GP, Wolffram, S, de Vos, R et al. (2006) Ingestion of onion soup high in quercetin inhibits platelet aggregation and essential components of the collagen-stimulated platelet activation pathway in man: a pilot study. Br J Nutr 96, 482488.CrossRefGoogle ScholarPubMed
77. Oh, WJ, Endale, M, Park, SC et al. (2012) Dual roles of quercetin in platelets: Phosphoinositide-3-kinase and MAP kinases inhibition, and cAMP-dependent vasodilator-stimulated phosphoprotein stimulation. Evid-Based Compl Alt. [Epublication 17 December 2012]CrossRefGoogle ScholarPubMed
78. Hubbard, GP, Stevens, JM, Cicmil, M et al. (2003) Quercetin inhibits collagen-stimulated platelet activation through inhibition of multiple components of the glycoprotein VI signaling pathway. J Thromb Haemost 1, 10791088.Google Scholar
79. Wright, B, Moraes, LA, Kemp, CF et al. (2010) A structural basis for the inhibition of collagen-stimulated platelet function by quercetin and structurally related flavonoids. Br J Pharmacol 159, 13121325.Google Scholar
80. Jasuja, R, Passam, FH, Kennedy, DR et al. (2012) Protein disulfide isomerase inhibitors constitute a new class of antithrombotic agents. J Clin Invest 122, 21042113.Google Scholar
81. Lu, H, Chang, DJ, Baratte, B et al. (2005) Crystal structure of a human cyclin-dependent kinase 6 complex with a flavonol inhibitor, fisetin. J Med Chem 48, 737743.Google Scholar
82. Wright, B, Watson, KA, McGuffin, LJ et al. (2015) GRID and docking analyses reveal a molecular basis for flavonoid inhibition of Src family kinase activity. J Nutr Biochem 26, 11561165.Google Scholar
83. Ysart, G, Miller, P, Barrett, G et al. (1999) Dietary exposures to nitrate in the UK. Food Addit Contam 16, 521532.Google Scholar
84. Omar, SA, Webb, AJ, Lundberg, JO et al. (2016) Therapeutic effects of inorganic nitrate and nitrite in cardiovascular and metabolic diseases. J Intern Med 279, 315336.Google Scholar
85. Cannon, RO III (1998) Role of nitric oxide in cardiovascular disease: focus on the endothelium. Clin Chem 44, 18091819.Google Scholar
86. Lundberg, JO, Weitzberg, E, Cole, JA et al. (2004) Nitrate, bacteria and human health. Nat Rev Micro 2, 593602.Google Scholar
87. Wagner, DA, Schultz, DS, Deen, WM et al. (1983) Metabolic fate of an oral dose of 15N-labeled nitrate in humans: effect of diet supplementation with ascorbic acid. Cancer Res 43, 19211925.Google Scholar
88. Webb, AJ, Patel, N, Loukogeorgakis, S et al. (2008) Acute blood pressure lowering, vasoprotective, and antiplatelet properties of dietary nitrate via bioconversion to nitrite. Hypertension 51, 784790.Google Scholar
89. Bartholomew, B & Hill, MJ (1984) The pharmacology of dietary nitrate and the origin of urinary nitrate. Food Chem Toxicol 22, 789795.Google Scholar
90. Lundberg, JO & Govoni, M (2004) Inorganic nitrate is a possible source for systemic generation of nitric oxide. Free Radic Biol Med 37, 395400.Google Scholar
91. Duncan, C, Dougall, H, Johnston, P et al. (1995) Chemical generation of nitric oxide in the mouth from the enterosalivary circulation of dietary nitrate. Nat Med 1, 546551.Google Scholar
92. Petersson, J, Carlstrom, M, Schreiber, O et al. (2009) Gastroprotective and blood pressure lowering effects of dietary nitrate are abolished by an antiseptic mouthwash. Free Radic Biol Med 46, 10681075.Google Scholar
93. Kapil, V, Haydar, SM, Pearl, V et al. (2013) Physiological role for nitrate-reducing oral bacteria in blood pressure control. Free Radic Biol Med 55, 93100.Google Scholar
94. Benjamin, N, O'Driscoll, F, Dougall, H et al. (1994) Stomach NO synthesis. Nature 368, 502.Google Scholar
95. Hunault, CC, van Velzen, AG, Sips, AJ et al. (2009) Bioavailability of sodium nitrite from an aqueous solution in healthy adults. Toxicol Lett 190, 4853.Google Scholar
96. Cosby, K, Partovi, KS, Crawford, JH et al. (2003) Nitrite reduction to nitric oxide by deoxyhemoglobin vasodilates the human circulation. Nat Med 9, 14981505.Google Scholar
97. Shiva, S, Huang, Z, Grubina, R et al. (2007) Deoxymyoglobin is a nitrite reductase that generates nitric oxide and regulates mitochondrial respiration. Circ Res 100, 654661.Google Scholar
98. Petersen, MG, Dewilde, S & Fago, A (2008) Reactions of ferrous neuroglobin and cytoglobin with nitrite under anaerobic conditions. J Inorg Biochem 102, 17771782.Google Scholar
99. Webb, AJ, Milsom, AB, Rathod, KS et al. (2008) Mechanisms underlying erythrocyte and endothelial nitrite reduction to nitric oxide in hypoxia: role for xanthine oxidoreductase and endothelial nitric oxide synthase. Circ Res 103, 957964.Google Scholar
100. Li, H, Cui, H, Kundu, TK et al. (2008) Nitric oxide production from nitrite occurs primarily in tissues not in the blood: critical role of xanthine oxidase and aldehyde oxidase. J Biol Chem 283, 1785517863.Google Scholar
101. Badejo, AM Jr, Hodnette, C, Dhaliwal, JS et al. (2010) Mitochondrial aldehyde dehydrogenase mediates vasodilator responses of glyceryl trinitrate and sodium nitrite in the pulmonary vascular bed of the rat. Am J Physiol Heart Circ Physiol 299, H819826.Google Scholar
102. Li, H, Liu, X, Cui, H et al. (2006) Characterization of the mechanism of cytochrome P450 reductase-cytochrome P450-mediated nitric oxide and nitrosothiol generation from organic nitrates. J Biol Chem 281, 1254612554.Google Scholar
103. Nohl, H, Staniek, K, Sobhian, B et al. (2000) Mitochondria recycle nitrite back to the bioregulator nitric monoxide. Acta Biochim Pol 47, 913921.Google Scholar
104. Larsen, FJ, Ekblom, B, Sahlin, K et al. (2006) Effects of dietary nitrate on blood pressure in healthy volunteers. N Engl J Med 355, 27922793.CrossRefGoogle ScholarPubMed
105. Hobbs, DA, Goulding, MG, Nguyen, A et al. (2013) Acute ingestion of beetroot bread increases endothelium-independent vasodilation and lowers diastolic blood pressure in healthy men: a randomized controlled trial. J Nutr 143, 13991405.Google Scholar
106. Bondonno, CP, Yang, X, Croft, KD et al. (2012) Flavonoid-rich apples and nitrate-rich spinach augment nitric oxide status and improve endothelial function in healthy men and women: a randomized controlled trial. Free Radic Biol Med 52, 95102.Google Scholar
107. Jonvik, KL, Nyakayiru, J, Pinckaers, PJ et al. (2016) Nitrate-rich vegetables increase plasma nitrate and nitrite concentrations and lower blood pressure in healthy adults. J Nutr 146, 986993.Google Scholar
108. Vanhoutte, PM (2009) Endothelial dysfunction: the first step toward coronary arteriosclerosis. Circ J 73, 595601.Google Scholar
109. Joris, PJ & Mensink, RP (2013) Beetroot juice improves in overweight and slightly obese men postprandial endothelial function after consumption of a mixed meal. Atherosclerosis 231, 7883.Google Scholar
110. Velmurugan, S, Gan, JM, Rathod, KS et al. (2016) Dietary nitrate improves vascular function in patients with hypercholesterolemia: a randomized, double-blind, placebo-controlled study. Am J Clin Nutr 103, 2538.Google Scholar
111. Gilchrist, M, Winyard, PG, Aizawa, K et al. (2013) Effect of dietary nitrate on blood pressure, endothelial function, and insulin sensitivity in type 2 diabetes. Free Radic Biol Med 60, 8997.Google Scholar
112. Omar, SA, Artime, E & Webb, AJ (2012) A comparison of organic and inorganic nitrates/nitrites. Nitric Oxide 26, 229240.Google Scholar
113. Liu, J, Wang, L, Liu, Y et al. (2015) The association between endothelial nitric oxide synthase gene G894 T polymorphism and hypertension in Han Chinese: a case-control study and an updated meta-analysis. Ann Hum Biol 42, 184194.Google Scholar
114. Carlstrom, M, Liu, M, Yang, T et al. (2015) Cross-talk between nitrate-nitrite-NO and NO synthase pathways in control of vascular NO homeostasis. Antioxid Redox Signal 23, 295306.Google Scholar
115. Hobbs, DA, George, TW & Lovegrove, JA (2013) The effects of dietary nitrate on blood pressure and endothelial function: a review of human intervention studies. Nutr Res Rev 26, 210222.Google Scholar
116. Kelly, J, Fulford, J, Vanhatalo, A et al. (2013) Effects of short-term dietary nitrate supplementation on blood pressure, O2 uptake kinetics, and muscle and cognitive function in older adults. Am J Physiol Regul Integr Comp Physiol 304, R73R83.Google Scholar
117. Jajja, A, Sutyarjoko, A, Lara, J et al. (2014) Beetroot supplementation lowers daily systolic blood pressure in older, overweight subjects. Nutr Res 34, 868875.Google Scholar
118. Gee, LC & Ahluwalia, A (2016) Dietary nitrate lowers blood pressure: epidemiological, pre-clinical experimental and clinical trial evidence. Curr Hypertens Rep 18, 17.CrossRefGoogle ScholarPubMed
119. Fuchs, D, Nyakayiru, J, Draijer, R et al. (2016) Impact of flavonoid-rich black tea and beetroot juice on postprandial peripheral vascular resistance and glucose homeostasis in obese, insulin-resistant men: a randomized controlled trial. Nutr Metab (Lond) 13, 34.Google Scholar
120. Shepherd, AI, Wilkerson, DP, Dobson, L et al. (2015) The effect of dietary nitrate supplementation on the oxygen cost of cycling, walking performance and resting blood pressure in individuals with chronic obstructive pulmonary disease: a double blind placebo controlled, randomised control trial. Nitric Oxide 48, 3137.Google Scholar
121. Leong, P, Basham, JE, Yong, T et al. (2015) A double blind randomized placebo control crossover trial on the effect of dietary nitrate supplementation on exercise tolerance in stable moderate chronic obstructive pulmonary disease. BMC Pulm Med 15, 52.Google Scholar
122. Berry, MJ, Justus, NW, Hauser, JI et al. (2015) Dietary nitrate supplementation improves exercise performance and decreases blood pressure in COPD patients. Nitric Oxide 48, 2230.Google Scholar
123. Velmurugan, S, Kapil, V, Ghosh, SM et al. (2013) Antiplatelet effects of dietary nitrate in healthy volunteers: involvement of cGMP and influence of sex. Free Radic Biol Med 65, 15211532.Google Scholar
124. Buys, ES, Sips, P, Vermeersch, P et al. (2008) Gender-specific hypertension and responsiveness to nitric oxide in sGCalpha1 knockout mice. Cardiovasc Res 79, 179186.Google Scholar
125. Chan, MV, Bubb, KJ, Noyce, A et al. (2012) Distinct endothelial pathways underlie sexual dimorphism in vascular auto-regulation. Br J Pharmacol 167, 805817.Google Scholar
126. Carlstrom, M, Larsen, FJ, Nystrom, T et al. (2010) Dietary inorganic nitrate reverses features of metabolic syndrome in endothelial nitric oxide synthase-deficient mice. Proc Natl Acad Sci USA 107, 1771617720.Google Scholar
127. Nystrom, T, Ortsater, H, Huang, Z et al. (2012) Inorganic nitrite stimulates pancreatic islet blood flow and insulin secretion. Free Radic Biol Med 53, 10171023.Google Scholar
128. Ohtake, K, Nakano, G, Ehara, N et al. (2015) Dietary nitrite supplementation improves insulin resistance in type 2 diabetic KKA(y) mice. Nitric Oxide 44, 3138.Google Scholar
129. Essawy, SS, Abdel-Sater, KA & Elbaz, AA (2014) Comparing the effects of inorganic nitrate and allopurinol in renovascular complications of metabolic syndrome in rats: role of nitric oxide and uric acid. Arch Med Sci 10, 537545.Google Scholar
130. Khalifi, S, Rahimipour, A, Jeddi, S et al. (2015) Dietary nitrate improves glucose tolerance and lipid profile in an animal model of hyperglycemia. Nitric Oxide 44, 2430.CrossRefGoogle Scholar
131. Jiang, H, Torregrossa, AC, Potts, A et al. (2014) Dietary nitrite improves insulin signaling through GLUT4 translocation. Free Radic Biol Med 67, 5157.Google Scholar
132. Roberts, LD, Ashmore, T, Kotwica, AO et al. (2015) Inorganic nitrate promotes the browning of white adipose tissue through the nitrate-nitrite-nitric oxide pathway. Diabetes 64, 471484.Google Scholar
133. Cermak, NM, Hansen, D, Kouw, IW et al. (2015) A single dose of sodium nitrate does not improve oral glucose tolerance in patients with type 2 diabetes mellitus. Nutr Res 35, 674680.Google Scholar
134. Carlsson, S, Wiklund, NP, Engstrand, L et al. (2001) Effects of pH, nitrite, and ascorbic acid on nonenzymatic nitric oxide generation and bacterial growth in urine. Nitric Oxide 5, 580586.Google Scholar
135. Gago, B, Lundberg, JO, Barbosa, RM et al. (2007) Red wine-dependent reduction of nitrite to nitric oxide in the stomach. Free Radic Biol Med 43, 12331242.Google Scholar
136. Peri, L, Pietraforte, D, Scorza, G et al. (2005) Apples increase nitric oxide production by human saliva at the acidic pH of the stomach: a new biological function for polyphenols with a catechol group? Free Radic Biol Med 39, 668681.Google Scholar
137. Rodriguez-Mateos, A, Hezel, M, Aydin, H et al. (2015) Interactions between cocoa flavanols and inorganic nitrate: additive effects on endothelial function at achievable dietary amounts. Free Radic Biol Med 80, 121128.Google Scholar
138. Ashworth, A, Mitchell, K, Blackwell, JR et al. (2015) High-nitrate vegetable diet increases plasma nitrate and nitrite concentrations and reduces blood pressure in healthy women. Public Health Nutr 18, 26692678.Google Scholar
139. Jovanovski, E, Bosco, L, Khan, K et al. (2015) Effect of spinach, a high dietary nitrate source, on Arterial stiffness and related hemodynamic measures: a randomized, controlled trial in healthy adults. Clin Nutr Res 4, 160167.Google Scholar
140. Keen, JT, Levitt, EL, Hodges, GJ et al. (2015) Short-term dietary nitrate supplementation augments cutaneous vasodilatation and reduces mean arterial pressure in healthy humans. Microvasc Res 98, 4853.Google Scholar
Figure 0

Fig. 1. Generic structure of a flavonoid consisting of two benzene rings linked by a 3-carbon chain.

Figure 1

Fig. 2. Structure of the seven classes of flavonoids shown as aglycones.

Figure 2

Fig. 3. Diagram of inorganic nitrate metabolism via the nitrate–nitrite–nitric oxide (NO) pathway (adapted from Hobbs et al.(115)). A proportion of ingested nitrate (NO3, - - -▸) is converted directly to nitrite (NO2, →) by facultative anaerobic bacteria, that reside in plaque and on the dorsum of the tongue, during mastication in the mouth (a); the remainder is swallowed and is rapidly absorbed from the upper gastrointestinal tract. Approximately 25 % is removed from the circulation and concentrated in the salivary glands and re-secreted into the mouth, where it is reduced to nitrite. Some of the salivary nitrite enters the acidic environment of the stomach once swallowed (b), where NO is produced non-enzymically from nitrite after formation of nitrous acid (HNO2) and then NO and other nitrogen oxides. The NO generated kills pathogenic bacteria and stimulates mucosal blood flow and mucus generation. The remaining nitrite is absorbed into the circulation; in blood vessels (c) nitrite forms vasodilatory NO after a reaction with deoxygenated Hb (deoxy-Hb). Approximately 60 % of ingested nitrate is excreted in urine within 48 h. Oxy-Hb, oxygenated Hb.

Figure 3

Table 1. The acute and chronic effects of dietary or inorganic nitrate on blood pressure in healthy subjects since 2014