Hostname: page-component-8448b6f56d-gtxcr Total loading time: 0 Render date: 2024-04-23T12:55:49.942Z Has data issue: false hasContentIssue false

The role of inorganic nitrate and nitrite in CVD

Published online by Cambridge University Press:  01 June 2017

Jacklyn Jackson
Affiliation:
School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, University Drive, Callaghan, NSW, Australia
Amanda J. Patterson
Affiliation:
Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, University Drive, Callaghan, NSW, Australia
Lesley MacDonald-Wicks
Affiliation:
Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, University Drive, Callaghan, NSW, Australia
Mark McEvoy*
Affiliation:
Centre for Clinical Epidemiology and Biostatistics, Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW, 2308, Australia
*
*Corresponding author: Mark McEvoy, email Mark.Mcevoy@newcastle.edu.au
Rights & Permissions [Opens in a new window]

Abstract

CVD is the leading cause of death worldwide, a consequence of mostly poor lifestyle and dietary behaviours. Although whole fruit and vegetable consumption has been consistently shown to reduce CVD risk, the exact protective constituents of these foods are yet to be clearly identified. A recent and biologically plausible hypothesis supporting the cardioprotective effects of vegetables has been linked to their inorganic nitrate content. Approximately 60–80 % inorganic nitrate exposure in the human diet is contributed by vegetable consumption. Although inorganic nitrate is a relatively stable molecule, under specific conditions it can be metabolised in the body to produce NO via the newly discovered nitrate–nitrite–NO pathway. NO is a major signalling molecule in the human body, and has a key role in maintaining vascular tone, smooth muscle cell proliferation, platelet activity and inflammation. Currently, there is accumulating evidence demonstrating that inorganic nitrate can lead to lower blood pressure and improved vascular compliance in humans. The aim of this review is to present an informative, balanced and critical review of the current evidence investigating the role of inorganic nitrate and nitrite in the development, prevention and/or treatment of CVD. Although there is evidence supporting short-term inorganic nitrate intakes for reduced blood pressure, there is a severe lack of research examining the role of long-term nitrate intakes in the treatment and/or prevention of hard CVD outcomes, such as myocardial infarction and cardiovascular mortality. Epidemiological evidence is needed in this field to justify continued research efforts.

Type
Review Article
Copyright
© The Authors 2017 

Introduction

Despite major medical research advancements over the past 50 years, CVD remains the leading cause of death worldwide and is responsible for 39 % of non-communicable disease deaths in populations aged under 70 years( 1 ). The leading non-communicable disease risk factor is hypertension, which is responsible for 13 % of global deaths each year and is a major risk factor for coronary artery disease (CAD), IHD and stroke( 1 ).

The pathogenesis of CVD is influenced by a variety of risk factors that can be broadly categorised as either modifiable or non-modifiable( Reference Buttar, Li and Ravi 2 ). Non-modifiable risk factors cannot be controlled through intervention and include advancing age, sex (men at greater risk than premenopausal women; postmenopausal women at greater risk than men), ethnicity and family history of CVD( Reference Buttar, Li and Ravi 2 ). Modifiable risk factors, on the other hand, have the ability to be manipulated through intervention in order to control, treat or modify the risk factor( Reference Buttar, Li and Ravi 2 ). Established modifiable risk factors for CVD include hypertension, tobacco use, raised blood glucose, physical inactivity, unhealthy diet, raised blood cholesterol/lipids and overweight and obesity( Reference Buttar, Li and Ravi 2 ).

Implementation of various lifestyle strategies which target specific modifiable risk factors can reduce the risk of CVD by up to 80 %( 1 , Reference Buttar, Li and Ravi 2 ), thus indicating that CVD is a chronic and mostly lifestyle-induced disease, to which the majority of current mortality is the consequence of previous exposures to behavioural risk factors such as inappropriate nutrition, insufficient physical activity and tobacco exposure( Reference Buttar, Li and Ravi 2 Reference Clair, Rigotti and Porneala 5 ). In addition, excess weight and central obesity, increased blood pressure, dyslipidaemia, diabetes and low cardiorespiratory fitness are among the factors contributing principally to CVD risk( Reference Buttar, Li and Ravi 2 , Reference Isomaa, Almgren and Tuomi 6 ).

Given the scope and prevalence of CVD within our current food and lifestyle environment, it is clear that preventative measures are the most appropriate to deal with this global health issue in order to reduce the costs to both the community (through improved quality of life) and governments through a reduction in hospitalisations, medication use and rehabilitation( Reference Buttar, Li and Ravi 2 ). Although behavioural factors such as smoking cessation and increased physical activity appear relatively straightforward targets for public health preventative interventions, the definition of a perceived ‘healthy’ diet has changed over time, leading to a general sense of public confusion and uncertainty surrounding the topic( Reference Carpentier and Komsa-Penkova 7 , Reference Harnack, Block and Lane 8 ).

Currently, the most compelling dietary evidence for CVD prevention is linked to whole-diet approaches such as the Mediterranean and Dietary Approaches to Stop Hypertension (DASH) diets( Reference Carpentier and Komsa-Penkova 7 , Reference Sofi, Cesari and Abbate 9 ). Although the cardioprotective effects of these diets may be credited to a whole-diet/whole-food effect, some individual nutritive components of these foods have also been extensively investigated.

The investigation of single nutritive components demonstrates that the evidence is less clear; this is especially noticeable for fruit and vegetable constituents. While whole fruit and vegetable consumption has been consistently shown to reduce CVD risk, as evidenced by various prospective studies showing a direct inverse association between fruit and vegetable intakes and the development of CVD events such as myocardial infarction (MI) and stroke( Reference Dauchet, Amouyel and Hercberg 10 Reference Bazzano, He and Ogden 13 ), the various constituents of fruits and vegetables such as vitamin C, polyphenols, fibre and antioxidants are yet to clearly demonstrate a beneficial link or a physiological pathway for their individual effect( Reference Wang, Ouyang and Liu 14 Reference Vivekananthan, Penn and Sapp 18 ).

A recent and biologically plausible hypothesis for the cardioprotective and blood pressure-lowering effect of vegetables has been linked to their inorganic nitrate (NO3 )/nitrite (NO2 ) content( Reference Hord 19 ). Support for this hypothesis has been implied in studies indicating that nitrate-rich green leafy vegetables and vitamin C-rich fruits and vegetables contribute most to the apparent cardiovascular protective effect of total fruit and vegetable intake( Reference Bhupathiraju, Wedick and Pan 20 , Reference Joshipura, Ascherio and Manson 21 ). Additionally, cardioprotective diets including the DASH, Mediterranean and traditional Japanese diets have been shown to naturally contain high quantities of inorganic nitrate (147–1222 mg/d) relative to a typical Western-style diet (about 75 mg/d)( Reference Hord, Tang and Bryan 22 Reference L’Hirondel and L’Hirondel 24 ).

Within the human body, inorganic nitrate/nitrite (NOx) can be metabolised to produce NO (Fig. 1)( Reference Lundberg and Weitzberg 25 , Reference McKnight, Smith and Drummond 26 ). NO is a highly valuable signalling molecule and has been demonstrated to mediate favourable effects on blood pressure control, platelet function, vascular health and exercise performance( Reference Kapil, Weitzberg and Lundberg 27 Reference Bailey, Winyard and Vanhatalo 30 ). In addition, the utility of inorganic NOx as an NO donor may be of particular relevance given that one serving of nitrate-rich vegetables (such as beetroot) has been estimated to produce more NO under specific conditions than can be endogenously formed by the classical l-arginine–nitric oxide synthase (NOS) pathway each day( Reference Hord 19 , Reference Kelm 31 , Reference Hotchkiss 32 ) (Fig. 1 ( Reference Webb, Patel and Loukogeorgakis 33 )).

Fig. 1 The fate of dietary nitrate. Nitrate is systematically absorbed becoming concentrated in the salivary glands and part of the salivary circulation. Salivary nitrate is reduced to nitrite by oral bacteria. In the stomach nitrite may produce NO. Nitrite transported in arterial circulation can be reduced to NO in low oxygen concentrations which can lead to vasodilation and reductions in blood pressure (Webb A, Patel N, Loukogeorhakis S, et al. Acute blood pressure lowering,vasoprotective, and antiplatelet properties of dietary nitrate via bioconversion to nitrite. Hypertension, vol. 51, pp. 784–790, from http://hyper.ahajournals.org/content/51/3/784.short ( Reference Webb, Patel and Loukogeorgakis 33 )).

Currently, the true effect that dietary/inorganic NOx may have on CVD risk factors and outcomes is poorly understood, but it is a highly worthwhile line of investigation given that an increased daily consumption of nitrate intake represents a potential low-cost and simple treatment option for reducing CVD burden.

Production of nitric oxide in the body

Endogenous production via the l-arginine–nitric oxide synthase pathway

The notion that NOx could be produced endogenously in the body was first considered in the early 1980s, upon finding that NOx excretion was exceeding quantities of ingestion in animal and human models( Reference Li and Förstermann 34 , Reference Green, De Luzuriaga and Wagner 35 ). Later it was demonstrated that l-arginine was the substrate for synthesising nitrogen oxides endogenously via the action of NOS enzymes( Reference Palmer, Ashton and Moncada 36 ).

In healthy individuals the l-arginine–NOS pathway can produce sufficient quantities of NO to maintain health (approximately 1·7 mmol/d)( Reference Kelm 31 , Reference Hotchkiss 32 ). However, conditions such as diabetes mellitus, ageing, hypercholesterolaemia and tobacco exposure have been found to make an impact on the bioactivity of endogenously produced NO via one or more of the following functions( Reference Guzik, Mussa and Gastaldi 37 Reference Ichiki, Ikeda and Haramaki 42 ):

  1. (1) Increased degradation of NO( Reference Taddei, Virdis and Ghiadoni 38 , Reference Ichiki, Ikeda and Haramaki 42 , Reference Bryan and Loscalzo 43 );

  2. (2) Altered phosphorylation and activation of NOS( Reference Taddei, Virdis and Ghiadoni 38 , Reference Bryan and Loscalzo 43 );

  3. (3) Increased production of NOS inhibitors (for example, asymmetric dimethylarginine; ADMA), leading to disruption of NOS activation( Reference Taddei, Virdis and Ghiadoni 38 , Reference Feron, Dessy and Moniotte 39 , Reference Böger, Bode-Böger and Szuba 41 Reference Bryan and Loscalzo 43 );

  4. (4) Deficiency of the NOS substrate, l-arginine( Reference Li and Förstermann 34 , Reference Taddei, Virdis and Ghiadoni 38 , Reference Böger, Bode-Böger and Szuba 41 );

  5. (5) Reduced availability of one or more cofactors essential for NOS function( Reference Li and Förstermann 34 , Reference Taddei, Virdis and Ghiadoni 38 ).

While appropriate medical management, consumption of a healthy diet and moderate exercise can somewhat reverse these effects, it has been postulated that supplementing parts of the NOS pathway may enhance NOS activity and NO production( Reference Taddei, Virdis and Ghiadoni 38 , Reference Böger, Bode-Böger and Szuba 41 , Reference Bryan and Loscalzo 43 ). This has been of particular importance given that increased ADMA levels inhibit NOS function and have been cited as the strongest risk predictor of cardiovascular events, and all-cause and cardiovascular mortality in individuals with CAD( Reference Sibal, Agarwal and Home 44 ). Although it remains unclear whether a change in ADMA can alter CVD risk, interventions such as l-arginine supplementation have been shown to improve endothelial-mediated vasodilation in individuals with elevated ADMA levels( Reference Böger, Bode-Böger and Szuba 41 , Reference Sibal, Agarwal and Home 44 ).

As a result, the effect of l-arginine supplementation has been investigated and short-term supplementation has been shown to improve endothelial function and relieve symptoms in patients with CHD( Reference Creager, Gallagher and Girerd 45 ). Long-term (6 months) supplementation, however, demonstrated no beneficial effect( Reference Bednarz, Jaxa-Chamiec and Maciejewski 46 ). In fact the long-term l-arginine supplementation led to increased rates of death and less cardiovascular improvement compared with the placebo due to the development of arginine toxicity and hyperkalaemia (abnormally high serum K)( Reference Schulman, Becker and Kass 47 , 48 ). In addition, the utility of supplementing arginine is questionable given that arginine is classified as a ‘semi-essential’ or ‘conditionally essential’ amino acid, depending on the developmental stage or health status of the individual( Reference Nakaki and Hishikawa 49 ). However, it is generally accepted that healthy adults should not need to supplement with arginine as their bodies produce physiologically sufficient amounts( 48 ). Arginine is also highly abundant in the diet, as rich dietary sources include meat, dairy products, vegetables, legumes and whole grains( 48 , Reference Nakaki and Hishikawa 49 ).

The ‘arginine paradox’ appears to address this notion, as it refers to the phenomenon that exogenous arginine causes NO-mediated biological effects, despite the fact that NOS are theoretically saturated in the substrate l-arginine( Reference Nakaki and Hishikawa 49 ). A recently published cross-sectional study including 2771 men and women investigated whether regular dietary intakes of l-arginine were associated with serum NOx, as an indicator of systemic NO production( Reference Mirmiran, Bahadoran and Ghasemi 50 ). This study found that increased dietary l-arginine intakes were strongly associated with serum NOx, which was independent of the overall dietary patterns of the study participants and other dietary factors, including intakes of high-nitrate-containing foods (probably due to collection of fasting blood samples)( Reference Mirmiran, Bahadoran and Ghasemi 50 ). Therefore, although there may be some utility in consuming adequate amounts of arginine, which is readily achieved by consumption of a healthy balanced diet, there appears to be no great benefit for the general population to be using arginine supplements. However, dietary intervention to also consume nitrate-rich foods holds much promise for supplementing the NOS pathway via the alternative nitrate–nitrite–NO pathway.

The nitrate–nitrite–nitric oxide pathway

Up until the early 1990s, plasma NOx were considered to be biologically inactive endproducts of NO production in the human body. However, it is now clear that under specific conditions nitrate and nitrite anions can be recycled in vivo back to NO( Reference McKnight, Smith and Drummond 26 , Reference Kapil, Weitzberg and Lundberg 27 , Reference Weitzberg and Lundberg 51 , Reference Zweier, Samouilov and Kuppusamy 52 ).

With a bioavailability of 100 %, ingested inorganic nitrate is swiftly absorbed in the proximal small intestine leading to significantly raised plasma nitrate concentrations for a period of up to 5–6 h post-nitrate ingestion( Reference Kapil, Weitzberg and Lundberg 27 , Reference Webb, Patel and Loukogeorgakis 33 , Reference Hobbs, George and Lovegrove 53 Reference Lundberg and Govoni 55 ). About 75 % of this nitrate is excreted at the kidneys; however, the other 25 % of plasma nitrate is actively extracted by the salivary glands, leading to salivary nitrate concentrations which are ten to twenty times higher than plasma nitrate concentrations( Reference Kapil, Weitzberg and Lundberg 27 , Reference Bryan and Loscalzo 43 , Reference Lundberg and Govoni 55 Reference Bondonno, Liu and Croft 57 ). Salivary nitrate accumulation must occur in order for nitrate to be reduced to nitrite, as anaerobic bacteria in the oral cavity use nitrate as an alternative electron acceptor to oxygen during respiration( Reference Kapil, Weitzberg and Lundberg 27 , Reference Lundberg and Govoni 55 , Reference Pannala, Mani and Spencer 56 , Reference Tannenbaum, Weisman and Fett 58 ). When this nitrite-rich saliva is swallowed it is reduced in the acidic stomach to produce nitrogen oxides including NO( Reference McKnight, Smith and Drummond 26 , Reference Kapil, Weitzberg and Lundberg 27 , Reference Zweier, Samouilov and Kuppusamy 52 , Reference Benjamin, O’Driscoll and Dougall 59 ). Today, this process is widely known as the nitrate–nitrite–NO pathway, and is thought to be one of the body’s major sources of NO generation, especially in situations when NO bioavailability via the conventional l-arginine–NOS pathway is compromised. In addition it has been suggested that the nitrate–nitrite–NO pathway may play a significant role in maintaining levels of bioactive NO and may be critical for maintaining cardiovascular homeostasis in the body( Reference Kapil, Weitzberg and Lundberg 27 , Reference Hobbs, George and Lovegrove 53 , Reference Coggan, Leibowitz and Spearie 60 ).

Noteworthy factors other than inorganic nitrate and nitrite consumption which have been shown to facilitate the nitrate–nitrite–NO pathway include:

  1. (1) Entero-salivary nitrate cycling. Approximately 25 % of plasma nitrate is actively taken up by the salivary glands leading to significant nitrate accumulation in the saliva. Within the oral cavity, anaerobic bacteria reduce nitrate to nitrite via the action of nitrate-reductive enzymes. Nitrite-rich saliva must be swallowed to produce NO in the acidic stomach. The importance of this salivary nitrate cycling has been demonstrated in studies where subjects spat after a dietary load of inorganic nitrate, preventing the opportunity for nitrate to accumulate in the saliva and be reduced to nitrite, therefore preventing NO production and any beneficial effects( Reference Lundberg and Weitzberg 25 , Reference Webb, Patel and Loukogeorgakis 33 , Reference Lundberg, Weitzberg and Gladwin 61 ).

  2. (2) Presence of anaerobic bacteria. Mammalian bacteria can utilise nitrate as an alternative electron acceptor to oxygen during respiration, and is a vital component of the nitrate–nitrite–NO pathway as human cells lack the required nitrate reductase enzymes( Reference Lundberg, Weitzberg and Gladwin 61 ). The importance of these bacteria has been further established in studies of germ-free rats, in which gastric NO formation was negligible post-dietary nitrate load( Reference Sobko, Reinders and Norin 62 ). Additionally, human studies have demonstrated that the use of commercial antibacterial mouthwash in human subjects abolished any blood pressure-lowering effects of a dietary nitrate load, indicating that the mouthwash killed off the commensal facultative bacteria in the mouth, thus preventing the production of nitrite and NO leading to a loss of beneficial health effects( Reference Bondonno, Liu and Croft 63 Reference Duncan, Dougall and Johnston 65 ).

  3. (3) Hypoxic conditions. The rate in which nitrate is reduced to nitrite is thirty times greater during conditions of low oxygen tension, as the oral bacteria use salivary nitrate as an alternative electron acceptor to oxygen during respiration( Reference Duncan, Dougall and Johnston 65 ). Xanthine oxidoreductase has also been shown to catalyse the reduction of nitrite to NO in hypoxic conditions( Reference Zhang, Naughton and Winyard 66 Reference Webb, Bond and McLean 68 ). This could also account for the increased production and utility of NO seen in exercising skeletal muscle or during myocardial ischaemia( Reference Zweier, Samouilov and Kuppusamy 52 , Reference Lundberg, Weitzberg and Gladwin 61 , Reference Duranski, Greer and Dejam 69 ). It is also important to note that plasma nitrite can be reduced to NO along the physiological oxygen gradient of the circulatory system( Reference Gladwin, Raat and Shiva 70 ). Specifically, deoxygenated Hb in the peripheral circulation can act as a nitrite reductase for NO production, as it has been revealed that as Hb deoxygenation increases, more NO is produced( Reference Cosby, Partovi and Crawford 71 Reference Doyle, Pickering and DeWeert 73 ). This provides an explanation for how various human studies have observed vasodilation after a NOx load, in healthy subjects at rest( Reference Webb, Patel and Loukogeorgakis 33 , Reference Ashworth, Mitchell and Blackwell 74 ).

  4. (4) Acidic conditions. Nitrite in the acidic stomach has been shown to spontaneously decompose to NO, a reaction that appears to increase in conditions of reduced pH (increased acidity)( Reference McKnight, Smith and Drummond 26 ). The importance of an acidic stomach for this reaction has been demonstrated in a study showing that NO production via nitrite protonation was inhibited in individuals using proton pump inhibitors (medications which reduce the acidity of gastric juices)( Reference Lundberg, Weitzberg and Lundberg 75 ).

  5. (5) Presence of reducing agents including vitamin C and polyphenols. Both vitamin C and polyphenols are abundant in a vegetable-rich diet, and their presence in the diet has been shown to favour the formation of NO via the nitrate–nitrite–NO pathway and prolong the half-life of NO in the stomach( Reference Mowat, Carswell and Wirz 76 , Reference Gago, Lundberg and Barbosa 77 ).

Sources of dietary inorganic nitrate and nitrite

N is vital to life on Earth and can undergo many chemical and biological changes in order to be amalgamated into living and non-living material. An essential form of environmental N includes inorganic nitrate, as an adequate nitrate supply in the soil is essential for plant growth( Reference Bryan and Loscalzo 43 , Reference Crawford 78 ).

The two major determining factors of the nitrate content of vegetables and fruit include their species and the amount of available nitrate in the soil( Reference Bryan and Loscalzo 43 ). Some species of vegetables such as green leafy vegetables (mean nitrate about 975–3624 mg/kg) and beetroot (mean nitrate about 1992 mg/kg) are naturally high in nitrate; however, environmental factors can lead to great variation among samples( Reference Hord, Tang and Bryan 22 ). These factors include seasonal differences and disruption to normal plant growth, leading to nitrate accumulation in the plant leaves, stems and stalks, due to changes in the photosynthetic conversion of plant nitrate to amino acids( Reference Crawford 78 Reference Kaiser and Brendle-Behnisch 80 ). Therefore, established factors shown to effect the normal growth of plants include drought conditions, high temperatures, shady and cloudy conditions, deficiency of soil nutrients, and excessive soil N( Reference Bryan and Loscalzo 43 ). Additionally, farming practices leading to damaged produce, early harvest, storage and transport conditions, processing and cooking practices will also result in significant variation in vegetable and fruit nitrate content( Reference Bryan and Loscalzo 43 ).

European-based studies have demonstrated that organically grown vegetables have a lower nitrate content than conventionally grown crops, despite the fact that organic fertilisers may cause high nitrate levels in vegetables, depending on the types and amount of organic fertilisers applied( Reference Muramoto 81 ). A California-based study by Muramoto( Reference Muramoto 81 ) reiterated this notion, as it found that spinach grown and harvested during the same season and under the same farming practices had a wide range of nitrate contents. This range appeared greatest in organic spinach, in which the maximum nitrate content measured was 3000 mg/kg, which was five times higher than the minimum (600 mg/kg)( Reference Muramoto 81 ). However, this study also demonstrated that conventionally grown spinach contained on average 30 % more nitrate than spinach grown organically, a result most probably explained due to the wide use of N-containing fertilisers in conventional farming( Reference Muramoto 81 ).

Muramoto( Reference Muramoto 81 ) also found a statistically significant seasonal difference in the nitrate content of iceberg lettuce, as winter samples were found to have on average 52 % more nitrate than summer samples( Reference Muramoto 81 ). This finding is consistent with Ekart et al. ( Reference Ekart, Gorenjal and Madorran 82 ), which found lettuce harvested during summer had a statistically significant lower nitrate content than lettuce harvested during winter (summer harvest: 1209 mg/kg; winter harvest: 2164 mg/kg). In addition, Ekart et al. ( Reference Ekart, Gorenjal and Madorran 82 ) found that washing leafy greens reduced the nitrate content of foods on average by 19 %. Other processing, such as boiling, blanching and sautéing, were found to significantly reduce the nitrate content of spinach by 53, 36 and 30 %, respectively( Reference Ekart, Gorenjal and Madorran 82 ), a finding which could be partly explained due to the water-soluble nature of inorganic nitrate( Reference Omar, Artime and Webb 83 ).

Due to the high variability of nitrate within plant species, accurate and reliable nitrate intake measured from fruit and vegetable consumption is difficult to predict. Despite this, combined vegetable and fruit intake is the major source of exogenous inorganic nitrate exposure and is predicted to constitute 30–90 % of total nitrate intake( Reference Du, Zhang and Lin 84 ). Other sources of nitrate intake include drinking water and meat products; however, their nitrate content is highly regulated to comply with strict government limits( 85 Reference Shuval and Gruener 89 ).

Nitrate occurs naturally in the water supply; however, in most developed countries water nitrate is generally present in concentrations much lower than allowed in the water guidelines (≤50 mg/l)( 85 , 86 , Reference Ward, DeKok and Levallois 88 ). Therefore, nitrate from the water supply is unlikely to contribute significantly to total nitrate intake in comparison with food sources.

Nitrate and nitrite salts (for example, potassium nitrite/sodium nitrate) have been used as food additives in cured meats for many years due to their effectiveness in ensuring microbial safety and their ability to enhance the flavour and appearance of the product( Reference Bryan and Loscalzo 43 ). The maximum levels of nitrate and nitrite allowed as food additives have been defined (Table 1)( 85 , Reference Jukes 90 92 ).

Table 1 Permissions for nitrate and nitrite in food productsFootnote *

* Nitrate salt: potassium nitrate and sodium nitrate. Nitrite salt: potassium nitrite and sodium nitrite.

It has been estimated that approximately 60–80 % of dietary nitrates are derived from vegetables (mainly green leafy and root vegetables), indicating that vegetable intake tends to contribute the greatest quantities of dietary nitrate (Table 2)( Reference Hord, Tang and Bryan 22 , Reference Weitzberg and Lundberg 93 ). This has been further implied by dietary patterns such as the DASH diet, Mediterranean, vegetarian and traditional Japanese diets which tend to include high quantities of vegetables (five or more serves per d) and provide approximately 147–1222 mg nitrate per d( Reference Hord, Tang and Bryan 22 Reference L’Hirondel and L’Hirondel 24 ). This is a relatively high nitrate intake compared with the typical Western-style diet which tends to be low in vegetables (one to three serves per d) and provides about 60–75 mg nitrate per d( Reference L’Hirondel and L’Hirondel 24 ). In addition, processed and cured meats are frequently cited as the major dietary source of nitrite (Table 3)( Reference Hord, Tang and Bryan 22 , Reference Lundberg and Weitzberg 25 , Reference Du, Zhang and Lin 84 , Reference Machha and Schechter 94 ), followed by various fruits and vegetables (Tables 2, 4 and 5) that have been physically damaged or poorly stored, as enzymes present in the plant tissues and/or contaminating bacteria facilitate the reduction of nitrate to nitrite( Reference Bryan and Loscalzo 43 , 85 ).

Table 2 Vegetable sources of nitrate and nitrite with estimated nitrate and/or nitrite contentsFootnote * (Mean values and ranges)

NA, data not available; ND, not detected.

* Data are combined nitrate and nitrite estimates from various published papers, government documents and reviews.

Table 3 Meat-based sources of nitrate and nitrite with estimated nitrate and/or nitrite contentsFootnote * (Mean values and ranges)

ND, not detected; NA, data not available.

* Data are combined nitrate and nitrite estimates from various published papers, government documents and reviews.

Table 4 Fruit sources of nitrate and nitrite with estimated nitrate and/or nitrite contentsFootnote * (Mean values and ranges)

NA, data not available; ND, not detected.

* Data are combined nitrate and nitrite estimates from various published papers, government documents and reviews.

Table 5 Nitrate- and nitrite-containing herbs with estimated nitrate and/or nitrite contentsFootnote * (Mean values and ranges)

ND, not detected.

* Data are combined nitrate and nitrite estimates from various published papers, government documents and reviews.

Nitric oxide in the cardiovascular system

Within the cardiovascular system, basal endothelial NO has a critical role in maintaining cardiovascular health as it controls vascular tone, smooth muscle cell proliferation and growth, platelet activity and aggregation, leucocyte trafficking, expression of adhesion molecules and inflammation( Reference Li and Förstermann 34 , Reference Machha and Schechter 94 Reference De Caterina, Libby and Peng 99 ). However, when the bioavailability of NO is compromised, the beneficial effects of NO are lost and endothelial dysfunction predominates due to the imbalance created between the release of vasoconstrictors and vasodilators (such as NO)( Reference Hobbs, George and Lovegrove 53 , Reference Versari, Daghini and Virdis 100 , Reference Vanhoutte 101 ). This idea has been supported in a study conducted by Kleinbongard et al. ( Reference Kleinbongard, Dejam and Lauer 102 ) which found that plasma nitrite levels are a reliable indicator of endothelial dysfunction and correlate with cardiovascular risk factors in humans. Additionally, endothelial dysfunction has been strongly linked with atherosclerosis development and a number of cardiovascular disorders such as hypertension, CAD, congestive heart failure and peripheral artery disease in multiple longitudinal studies( Reference Hobbs, George and Lovegrove 53 , Reference Vanhoutte 101 , Reference Landmesser and Drexler 103 Reference Gokce, Keaney and Hunter 107 ).

While in the past most of the evidence suggesting a relationship between endothelial dysfunction and clinical events from atherosclerosis development was considered ‘circumstantial’, more recently conducted cross-sectional studies have indicated that severe endothelial dysfunction of the arteries can trigger events of unstable angina and MI( Reference Vita and Keaney 108 , Reference Schächinger, Britten and Zeiher 109 ). Al Suwaidi et al. ( Reference Al Suwaidi, Hamasaki and Higano 104 ) studied 157 patients with mild CAD for 2·3 years, and found an increased incidence of cardiovascular events in patients with impaired endothelium-dependent vasodilation (NO production of endothelium) of the coronary arteries. In another study by Katz et al. ( Reference Katz, Hryniewicz and Hriljac 110 ), 259 subjects with chronic heart failure were assessed prospectively, to which endothelial dysfunction in chronic heart failure was found to significantly increase risk of mortality, thus supporting the notion that coronary endothelial dysfunction plays a role in the pathogenesis of coronary atherosclerosis, risk of cardiac events and death( Reference Al Suwaidi, Hamasaki and Higano 104 , Reference Katz, Hryniewicz and Hriljac 110 ).

Many factors are known to predispose to endothelial dysfunction, due to reductions in NO concentrations and bioavailability in humans( Reference Li and Förstermann 34 , Reference Lidder and Webb 111 , Reference Lundberg, Gladwin and Weitzberg 112 ). These factors are consistent with the modifiable and non-modifiable risk factors for CVD, including hypertension, hypercholesterolaemia, diabetes, tobacco use, physical inactivity, consumption of unhealthy diets and increased age and sex (NO bioavailability is reduced in postmenopausal women, a period in which CVD risk is drastically increased in women)( Reference Li and Förstermann 34 , Reference Lundberg, Gladwin and Weitzberg 112 Reference Celermajer, Sorensen and Spiegelhalter 120 ). Interestingly, improved endothelial function is a common feature of experimental intervention studies, which have shown reductions in cardiovascular risk and improvements in endothelial-dependent vasodilation in the coronary and peripheral circulation( Reference Vita and Keaney 108 ). Such interventions commonly include the use of lipid- and blood pressure-lowering medications, smoking cessation and increased physical activity( Reference Vita and Keaney 108 , Reference Tsuchiya, Asada and Kasahara 117 , Reference Celermajer, Sorensen and Georgakopoulos 121 Reference Hornig, Maier and Drexler 124 ). However, the notion that inorganic nitrate and nitrite either consumed from dietary sources such as green leafy vegetables or supplements is relatively new, and their therapeutic potential as an NO donor via the nitrate-nitrite–NO pathway remains unclear( Reference Lundberg, Gladwin and Weitzberg 112 , Reference DeVan, Brooks and Evans 125 ).

Cardiovascular protective actions of nitric oxide

NO is non-polar and can diffuse freely across cell plasma membranes and is a key signalling molecule capable of many important functions, acting primarily by stimulating intra-cellular receptors within the target cell( Reference Wilson and Hunt 126 ).

Within the vasculature of the cardiovascular system, the primary role for NO’s action is for the regulation of vascular function and blood pressure, a notion which has been clearly demonstrated in animal models in which synthesis of NO was blocked leading to persistently elevated blood pressure( Reference Lundberg, Gladwin and Weitzberg 112 , Reference Channon, Qian and George 127 ). In addition, this interaction has been demonstrated in some recently conducted short-term dietary nitrate trials in human subjects, which showed that peak blood pressure-lowering effects were achieved in synchronisation with peak plasma concentrations of NO (NOx) after a dietary nitrate load( Reference Hobbs, Kaffa and George 28 , Reference Webb, Patel and Loukogeorgakis 33 , Reference Hobbs, Goulding and Nguyen 128 ).

The cellular pathway in which NO exerts this vasodilatory action is well established. NO rapidly diffuses across vascular smooth muscle cell membranes. Within the smooth muscle cells, NO binds to and activates guanylyl cyclase to produce cyclic GMP( Reference Wilson and Hunt 126 ). Once produced, cyclic GMP can have a number of effects in the cells, but many of these effects are mediated through the activation of protein kinase G. Activation of protein kinase G via cyclic GMP leads to the activation of myosin phosphatase which in turn leads to smooth muscle cell relaxation and vasodilation( Reference Wilson and Hunt 126 , Reference Channon, Qian and George 127 ).

In addition to regulating vascular tone, NO can facilitate many other important functions preventing the development of atherosclerosis, which include antiplatelet effects, anti-proliferative effects, anti-inflammatory, and antioxidant effects( Reference Channon, Qian and George 127 , Reference Simon, Stamler and Jaraki 129 , Reference Clapp, Hingorani and Kharbanda 130 ). Although the cellular pathways for these actions are yet to be clearly defined, it is clear that NO is capable of binding to or reacting with a variety of chemical modalities within the cellular environment, including metal-containing proteins, membrane receptors, ion channels, enzymes, transcription factors and oxygen species( Reference Channon, Qian and George 127 , Reference Vallance and Webb 131 ).

Other nitric oxides and possible mechanisms in the cardiovascular system

While NO is the most widely cited bioactive metabolite underpinning the cardiovascular therapeutic benefits of dietary inorganic nitrates and nitrites, it has been suggested that other nitric oxides also play a role( Reference Lundberg and Weitzberg 25 , Reference Weitzberg and Lundberg 93 ). This may be expected, given that dietary constituents in the stomach may react with each other in order to form a variety of bioactive compounds( Reference Lundberg and Weitzberg 25 ). Examples of such compounds include nitrated fatty acids, nitrosothiols and ethyl nitrite( Reference Lundberg and Weitzberg 25 ).

While the biological significance of these compounds is yet to be made clear, the following actions have been suggested:

  1. (1) Ethyl nitrite. Rat models have shown that ethanol from alcoholic drinks can interact with salivary-derived nitrite in the acidic stomach, leading to the production of ethyl nitrite( Reference Lundberg and Weitzberg 25 , Reference Gago, Nyström and Cavaleiro 132 ). Ethyl nitrate is a potent smooth muscle relaxant and may have a vasodilatory role in the cardiovascular system( Reference Gago, Nyström and Cavaleiro 132 ).

  2. (2) Nitrosothiols. In the stomach, nitrite has been shown to induce S-nitrosation within the gastric compartment. S-nitrosothiols are thought to represent a circulating endogenous reservoir of NO acting as an NO donor( Reference Lundberg and Weitzberg 25 ).

  3. (3) Nitrated fatty acids (nitroalkenes). Nitric oxides can react with unsaturated fatty acids to produce nitroalkenes. Analysis of synthetic nitroalkenes derivatives of oleic, linoleic and arachidonic acids reveals that these species possess unique chemical reactions which may support multiple cell signalling events such as vasodilation and reduced inflammation( Reference Lundberg and Weitzberg 25 ). Such events may be mediated through their NO donor capabilities.

Currently the systemic capabilities of these bioactive N compounds remain uncertain; however, it highlights a possible whole-diet effect for exerting a beneficial effect on NO and other relevant cardiovascular signalling molecules. This notion is highlighted by Lundberg and Weitzberg( Reference Lundberg and Weitzberg 25 , Reference Weitzberg and Lundberg 93 ), indicating that various dietary constituents of the Mediterranean diet may interact in the stomach to produce these potentially therapeutic compounds, and may provide an additional explanation for the cardiovascular health benefits/protection seen with this dietary pattern.

Inorganic v. organic nitrate and nitrite

Organic nitrates such as glyceryl trinitrate and isosorbide mononitrate represent the first class of NO donors to reach the clinical setting and have been used extensively in the treatment of various cardiovascular conditions including angina, CAD and heart failure( Reference Omar, Artime and Webb 83 ).

Unlike inorganic nitrates which are relatively simple molecules and naturally occurring in fruits and vegetables, organic nitrates are synthetic compounds produced by a reaction between nitric acid and an alcohol group( Reference Omar, Artime and Webb 83 ). Organic nitrates are complex, non-polar hydrocarbon chains attached to a nitro-oxy-radical (–ONO2), which is responsible for its biological effects (Fig. 2)( Reference Omar, Artime and Webb 83 ).

Fig. 2 Chemical structure of inorganic nitrate/nitrite compared with organic mono-, di-, tri- and tetra-nitrates/nitrites. 5-ISMN, isosorbide-5-mononitrate; ISDN, isosorbide dinitrate; GTN, glyceryl trinitrate; ETN, erythritol tetranitrate; PETriN, pentaerythrityl trinitrate; PETN, pentaerythritol tetranitrate. Reprinted from Omar et al. ( Reference Omar, Artime and Webb 83 ), with permission from Elsevier.

Once organic nitrates are introduced to the blood system, levels rise quickly leading to the rapid onset of their action( Reference Omar, Artime and Webb 83 ). At low doses (≤1·25 mg/kg body weight) organic nitrate has been demonstrated to dilate large conductance veins and large arteries, while at high doses (2·5–5 mg/kg body weight) organic nitrates can also induce dilation of the arterioles of the microcirculation( Reference Omar, Artime and Webb 83 ). These vasodilatory effects of organic nitrates have been shown to reduce cardiac work and lower myocardial oxygen requirements, which may alleviate or even prevent cases of MI( Reference Klemenska and Beresewicz 133 ). In addition, it has been suggested that organic nitrates have anti-aggregatory properties in patients with stable and unstable angina( Reference Klemenska and Beresewicz 133 ).

Today in clinical practice short-acting organic nitrates, most notably in the form of glyceryl trinitrate, are administered during the symptomatic treatment of MI and angina( Reference Omar, Artime and Webb 83 , Reference Klemenska and Beresewicz 133 ). Glyceryl tri-nitrates are generally administered in the form of either a mouth spray or intravenous infusion, to which onset of action is rapid (2–3 min)( Reference Klemenska and Beresewicz 133 ). Although short-term treatment with organic nitrates has some positive impact on endothelial function, acute side effects of their use include hypotension, dizziness, nausea and headache( Reference Omar, Artime and Webb 83 ). Also, despite the high potency of organic nitrates and their long history as being used to treat various CVD, nitrate tolerance is a huge limitation and an undesirable side effect of their use( Reference Omar, Artime and Webb 83 , Reference Klemenska and Beresewicz 133 ).

Nitrate tolerance is a complex phenomenon and is poorly understood; however, it is clearly a result of chronic organic nitrate use to which nitrovasodilator responsiveness is lost( Reference Omar, Artime and Webb 83 ). Nitrate tolerance has been reported to occur within 1–3 d of continuous glyceryl trinitrate treatment in patients with MI, stable angina and chronic congestive heart failure( Reference Klemenska and Beresewicz 133 ). Further, chronic organic nitrate use has also been linked to endothelial dysfunction, increased production of free radicals and the development of vascular tolerance to other endothelium-dependent vasodilators( Reference Omar, Artime and Webb 83 ). Although this phenomenon is poorly understood, recent animal and human studies indicate that increased vascular production of the superoxide anion (O2 ) underlies the mechanism for tolerance( Reference Klemenska and Beresewicz 133 ). This oxidative stress hypothesis of nitrate tolerance is supported by numerous reports demonstrating that the tolerance is prevented by co-administration of antioxidants (for example, vitamin C, vitamin E and folic acid) and interventions which inhibit reactive oxygen species formation (lipid- and blood pressure-lowering medications)( Reference Klemenska and Beresewicz 133 Reference Fontaine, Otto and Fontaine 136 ).

It is interesting to note that the phenomenon of tolerance is not exhibited with the consumption of inorganic nitrates/nitrites; however, despite showing promise in preventing or treating certain cardiovascular conditions, such as hypertension, they have received little attention by the medical community( Reference Kapil, Weitzberg and Lundberg 27 ).

Inorganic nitrate and nitrite: from dietary contaminant to potential therapeutic nutrient

Throughout history, cases of accidental toxic exposure to nitrate and nitrite have been documented; however, the health risk of excessive inorganic nitrate and nitrite consumption appears specific to population subgroups( Reference Hord, Tang and Bryan 22 ). One of these subgroups includes infants aged less than 6 months, to which excessive nitrite exposure has been linked to cases of methaemoglobinaemia (blue baby syndrome)( Reference Greer and Shannon 137 ). As a result, strict regulatory limits have been established to govern the NOx content of the drinking water supply and their use as an additive to processed and cured meats in order to limit exposure to the population( 85 , 86 ).

Methaemoglobinaemia can occur when nitrite oxidises ferrous Fe (Fe2+) in Hb to the ferric state (Fe3+), resulting in methaemoglobin. Methaemoglobin is incapable of binding molecular oxygen, and impairs oxygen delivery to the tissues, causing hypoxia and cyanosis( Reference Greer and Shannon 137 ). While most cases of methomeoglobinaemia have been attributed to the consumption of well water (prone to high nitrate accumulation) used for the preparation of infant formula, there have been reported cases of nitrate poisoning in infants from the ingestion of plant nitrates( 86 , Reference Greer and Shannon 137 ). While Martinez et al. ( Reference Martinez, Sanchez-Valverde and Gil 138 ) found that the use of certain high-nitrate vegetables (herbs and green leafy vegetables) in infant homemade vegetable purée increased methaemoglobinaemia in infants (herbs: OR 5·2, 95% CI 1·1, 24·6; and green leafy vegetables: OR 2·0, 95% CI 0·4, 8·7), the most important factor increasing methaemoglobinaemia was the time lapse between vegetable purée preparation and consumption (OR 17·4, 95% CI 3·5, 86·3 if purée was prepared 24–48 h before; and OR 24·9, 95% CI 3·3, 187·6 if prepared >48 h before)( Reference Martinez, Sanchez-Valverde and Gil 138 ).

To date, human nitrate and nitrite exposure studies have failed to prove a direct link with methaemoglobinaemia, suggesting that NOx exposure alone may not be responsible for methaemoglobinaemia development( Reference Milkowski, Garg and Coughlin 139 , Reference Avery 140 ).

Another population subgroup that is thought to be at health risk due to excessive NOx exposure is high consumers of cured and processed meats( Reference Hord, Tang and Bryan 22 , Reference Bouvard, Loomis and Guyton 141 ). It has been theorised that nitrates and nitrites from processed meats generate N-nitroso compounds which can be carcinogenic( Reference Bingham 142 ).

In October 2015 the International Agency for Research on Cancer (IARC) summarised more than 800 studies conducted globally, and determined that 50 g of processed meat per d increased the risk of colorectal cancer by 18%, and therefore concluded that processed meats are carcinogenic( Reference Bouvard, Loomis and Guyton 141 ). In animal studies N-nitrosamines and related N-nitrosamides have been shown to be carcinogenic in a variety of molecular structures( Reference Gilchrist, Winyard and Benjamin 143 , Reference Magee and Barnes 144 ). However, such direct evidence demonstrating nitrate and nitrite as human carcinogens is severely lacking. This has been reflected in the conclusions of the FAO expert committee who found no consistent increased risk of cancer with increasing consumption of nitrate, as available epidemiological studies did not provide evidence that nitrate is carcinogenic to humans( 145 ).

Currently, researchers are interested in understanding whether the health risks associated with inorganic nitrates/nitrites outweigh the recently discovered health benefits; however, there is a growing consensus that any weak and inconclusive data on inorganic NOx and cancer associations are far outweighed by the potential health benefits of restoring NO homeostasis( Reference Hord, Tang and Bryan 22 , Reference Du, Zhang and Lin 84 , Reference Milkowski, Garg and Coughlin 139 , Reference Gilchrist, Winyard and Benjamin 143 ). In particular this has been demonstrated in various animal and human experimental studies, in which inorganic NOx has been shown to improve outcomes such as blood pressure, endothelial function, platelet function, ischaemia–reperfusion injury, exercise performance and host defence( Reference Gilchrist, Winyard and Benjamin 143 , Reference Kapil, Milsom and Okorie 146 Reference Dykhuizen, Frazer and Duncan 151 ).

Evidence of cardiovascular benefit from animal studies

Intakes of dietary inorganic nitrate have been shown to be strongly cardioprotective in animal studies. Carlström et al. ( Reference Carlström, Persson and Larsson 152 ) indicated this in a four-arm dietary intervention trial in rats. The rats were placed on either a normal-salt diet (control), a high-salt diet, a high-salt diet supplemented with a nutritional (low) dose of nitrate, and a high-salt diet supplemented with a pharmacological (high) dose of nitrate for 8–11 weeks( Reference Carlström, Persson and Larsson 152 ). As expected, results demonstrated that chronic consumption of a high-salt diet develops hypertension; however, when combined with a low nitrate dose, blood pressure was non-statistically significantly lower( Reference Carlström, Persson and Larsson 152 ). On the other hand, the higher nitrate dose lowered blood pressure by a significant 24 mmHg compared with the plain high-salt diet, a magnitude of blood pressure reduction considerably magnified compared with blood pressure reductions observed in another study of healthy normotensive rats using the same nitrate dose( Reference Carlström, Persson and Larsson 152 , Reference Petersson, Carlström and Schreiber 153 ). Similar results were reported by Kanematsu et al. ( Reference Kanematsu, Yamaguchi and Ohnishi 154 ), finding that in hypertensive rats, antihypertensive effects were only apparent with the highest dose of nitrate, yet there was a strong tissue-protective effect seen with lower doses equivalent to modest dietary intakes. Ferguson et al. ( Reference Ferguson, Hirai and Copp 155 ) demonstrated clinically significant reductions in mean arterial pressure with beetroot juice supplementation in exercising rats (control: 137 (sem 3); beetroot juice: 127 (sem 4) mmHg; P<0·05), indicating that clinically significant blood pressure reductions may be achievable in doses attained from dietary sources( Reference Ferguson, Hirai and Copp 155 ).

In addition to significant blood pressure control, Carlström et al. ( Reference Carlström, Persson and Larsson 152 ) found that dietary nitrate supplementation can partly prevent the development of cardiac hypertrophy and high nitrate doses significantly reduced the fibrotic changes which were observed in the high-salt group, two factors which are major predictors of heart failure( Reference Carlström, Persson and Larsson 152 ). Two other studies found that mice ingesting inorganic nitrate led to a significantly reduced infarct size during myocardial ischaemia, an important finding given that reduced infarct size is associated with lower heart failure risk post-MI and mortality( Reference Bryan, Calvert and Elrod 156 Reference Minicucci, Azevedo and Polegato 158 ).

When Baker et al. ( Reference Baker, Su and Fu 149 ) treated rats with an intravenous bolus of sodium nitrite across various doses (0·04, 0·4, 1·0, 4·0, 7·0 and 10·0 mg/kg), before initialising a blockage of the coronary artery, there was a clear dose-dependent effect of nitrite on infarct size. However, it was intriguing to note that protection was only found in doses up to 4·0 mg/kg, an effect which was absent at higher doses( Reference Baker, Su and Fu 149 ). Rats administered with 4·0 mg/kg nitrite exhibited a significant 32% reduction in infarct size compared with controls( Reference Baker, Su and Fu 149 ). Nitrite was also found most effective when administered before and/or during the ischaemic event, but not at the onset of reperfusion( Reference Baker, Su and Fu 149 ). Further, equivalent doses of sodium nitrate had no effect on infarct size, indicating that administration timing and doses are key considerations for nitrite protection from MI( Reference Baker, Su and Fu 149 ).

Thrombosis is largely a result of platelet adhesion, activation and aggregation, and is a common pathology underlying IHD and ischaemic stroke( Reference Nieswandt, Pleines and Bender 159 , Reference Raskob, Angchaisuksiri and Blanco 160 ). NO plays a key role in preventing thrombosis development( Reference Park, Piknova and Huang 161 ). Park et al. ( Reference Park, Piknova and Huang 161 ) demonstrates this notion upon discovering an inverse correlation between NOx levels and platelet activity/aggregation in mice. In addition, Apostoli et al. ( Reference Apostoli, Solomon and Smallwood 162 ) examined the effect of inorganic nitrite on platelet aggregation in endothelial NOS-deficient mice. This study found that inorganic nitrite exerts an antiplatelet effect during endothelial NOS deficiency and suggested that dietary nitrate may reduce platelet hyperactivity during endothelial dysfunction( Reference Apostoli, Solomon and Smallwood 162 ).

Pulmonary hypertension can lead to the remodelling of the artery wall, causing abnormalities of elastic fibres, intimal fibrosis and medial hypertrophy( Reference Moraes, Colucci and Givertz 163 ). This can result in vascular stiffness and is a condition linked to the development of chronic heart failure( Reference Moraes, Colucci and Givertz 163 ). Sodium nitrite interventions in lamb and mouse models have shown reductions in pulmonary hypertension specifically during hypoxic conditions( Reference Hunter, Dejam and Blood 164 , Reference Zuckerbraun, Shiva and Ifedigbo 165 ). However, Casey et al. ( Reference Casey, Badejo and Dhaliwal 166 ) found that intravenous injections of sodium nitrite during normoxic conditions could lead to reductions in pulmonary and systemic arterial pressure and increased cardiac outputs in adult male rats. This suggests that sodium nitrite may have a role in reducing the workload of the heart during pulmonary hypertension, thus protecting the heart and vascular system from associated damage and dysfunction( Reference Casey, Badejo and Dhaliwal 166 ).

Hendgen-Cotta et al. ( Reference Hendgen-Cotta, Luedike and Totzeck 167 ) pre-treated mice with nitrate before inducing chronic limb ischaemia, and nitrate supplementation was found to enhance revascularisation and increased mobilisation of circulating angiogenic cells (CAC), which are important for the recovery and maintenance of healthy endothelial function( Reference Hendgen-Cotta, Luedike and Totzeck 167 ). Heiss et al. ( Reference Heiss, Meyer and Totzeck 168 ), on the other hand, injected inorganic nitrite into healthy mice, and found that nitrite significantly increased CAC at 1 h compared with controls. It is interesting to note, however, that when this test was repeated in endothelial NOS-deficient mice, no CAC mobilisation was observed, indicating that NOS may be required to take part in nitrate-mediated CAC mobilisation( Reference Heiss, Meyer and Totzeck 168 ).

In a study conducted by Sindler et al. ( Reference Sindler, Fleenor and Calvert 169 ) the effect of nitrite in aged, but healthy, mice was investigated and high dietary nitrite doses were found to reverse age-related vascular dysfunction, arterial stiffness and reduce levels of oxidative stress. This is in line with Carlström et al. ( Reference Carlström, Persson and Larsson 152 ) who found that key plasma and urinary oxidative stress markers (malondialdehyde, type VI isoprostane (iPF-VI) and 8-oxo-2’-deoxyguanosine (8-OHdG)) were significantly reduced (despite co-consumption of a high-salt diet) with both low- (0·1 mmol nitrate/d) and high- (1·0 mmol nitrate/d) dose dietary nitrate supplementation, which may be useful in preventing NO degradation and endothelial dysfunction( Reference Carlström, Persson and Larsson 152 , Reference Cai and Harrison 170 ). This is an interesting finding, given that oxidative stress is directly linked with an inflammatory response which is thought to have a central role in the development of atherosclerosis( Reference Weitzberg and Lundberg 93 ).

Stokes et al. ( Reference Stokes, Dugas and Tang 171 ) found that mice fed cholesterol-enriched diets for 3 weeks tend to develop clear signs of vascular disease pathology, including elevated leucocyte adhesion and endothelial dysfunction, an effect which was prevented with nitrite supplementation in the drinking water. In another study by Carlström et al. ( Reference Carlström, Larsen and Nyström 172 ) it was demonstrated that several features of the metabolic syndrome (including visceral fat and circulating TAG, which are strong risk factors for CVD) can be reversed by dietary nitrate supplementation, in amounts which correspond to those derived from endothelial NOS under normal healthy conditions or a vegetable-rich diet( Reference Carlström, Larsen and Nyström 172 ).

Evidence of cardiovascular benefit from human studies

In 2003, Cosby et al. ( Reference Cosby, Partovi and Crawford 71 ) conducted one of the first studies demonstrating a relationship between inorganic nitrite supplementation and blood pressure reductions in healthy human subjects. This study chose to use sodium nitrite (NaNO2 ) infusions providing approximately 75 mg NaNO2 over two 15-min periods, a dose which was found to significantly reduce mean blood pressure by 7 mmHg (P<0·01)( Reference Cosby, Partovi and Crawford 71 ). Similar findings were later established using sodium nitrate (NaNO3 ) in a study conducted by Larsen et al. ( Reference Larsen, Ekblom and Sahlin 173 ). In this study healthy subjects consumed NaNO3 (8·5 mg/kg per d for 3 d) as a dietary supplement, and although systolic blood pressure was not changed during this time compared with placebo (sodium chloride), diastolic blood pressure was significantly reduced on average by 3·7 mmHg (P<0·02) and mean arterial pressure was lowered by 3·2 mmHg (P<0·03)( Reference Larsen, Ekblom and Sahlin 173 ). Soon after, Webb et al. ( Reference Webb, Patel and Loukogeorgakis 33 ) investigated this topic further using beetroot juice (containing approximately 1400 mg inorganic nitrate). Results from Webb et al. ( Reference Webb, Patel and Loukogeorgakis 33 ) showed a peak reduction in systolic blood pressure of 10·4 (sem 3) mmHg (P<0·01), a reduction in diastolic blood pressure of 8·1 (sem 2·1) mmHg (P<0·01) and mean arterial pressure reduction of 8·0 (sem 2·1) mmHg (P<0·01), thus indicating that significant blood pressure reductions are possible with the acute consumption of dietary inorganic nitrate in healthy subjects. This is a notion which has been further supported by a recently conducted systematic review and meta-analysis which found that inorganic nitrate and beetroot juice consumption was associated with greater changes in systolic blood pressure (–4·4 (95% CI –5·9, –2·8) mmHg; P<0·001) than diastolic blood pressure (–1·1 (95% CI –2·2, 0·1) mmHg; P=0·06)( Reference Siervo, Lara and Ogbonmwan 174 ). However, it is important to note that these findings have not been consistent across the literature, as a few recently conducted randomised controlled trials have found that inorganic nitrate consumption from either beetroot juice or from a high-nitrate diet (rich in green leafy vegetables) for 1–2 weeks had little/no effect on the blood pressure of study subjects( Reference Bondonno, Liu and Croft 57 , Reference Bondonno, Liu and Croft 175 , Reference Gilchrist, Winyard and Aizawa 176 ). The exact cause of this variation across studies remains unclear, yet could be due to methodological differences including the study population (for example, healthy subjects v. hypertensive subjects) or the conditions in which NOx was consumed (for example, food v. supplement, dosing or altered environmental conditions such as exercise stress). Nevertheless, this question remains unclear and will require further investigation, in order to better understand the usefulness of dietary/inorganic NOx within the general population.

While the acute effects of dietary inorganic nitrate on blood pressure have been extensively investigated, very few studies have investigated long-term effects. Sobko et al. ( Reference Sobko, Marcus and Govoni 23 ) investigated the effects of a traditional Japanese diet on blood pressure which provided approximately 1140 mg of nitrate per d for a 10 d period. The traditional Japanese diet led to a lower diastolic blood pressure than seen in the non-Japanese diet group (71·3 (sd 7·9) v. 75·8 (sd 7·8) mmHg; P=0·0066), indicating that dietary inorganic nitrate consumption for longer-periods of time may have some blood pressure-lowering effects in healthy individuals; however, a 10 d intervention can hardly be classified as a long-term intervention( Reference Sobko, Marcus and Govoni 23 ). In another 4-week intervention, Kapil et al. ( Reference Kapil, Khambata and Robertson 29 ) assigned hypertensive patients to receive a daily dose of either 250 ml of beetroot juice or placebo (nitrate-depleted beetroot juice). Notably, Kapil et al. ( Reference Kapil, Khambata and Robertson 29 ) found that daily dietary nitrate supplementation significantly reduced mean clinic blood pressure (7·7/2·4 mmHg (range 3·6–11·8/0·0–4·9 mmHg); P<0·001, P=0·05), mean 24 h ambulatory blood pressure (7·7/5·2 mmHg (range 4·1–11·2/2·7–7·7 mmHg); P<0·001 for both) and mean home blood pressure (8·1/3·8 mmHg (range 3·8–12·4/0·7–6·9 mmHg); P<0·001, P<0·01)( Reference Kapil, Khambata and Robertson 29 ).

Currently, the longest intervention study conducted in this area is a 10-week intervention trial from DeVan et al. ( Reference DeVan, Brooks and Evans 125 ). In this study, healthy 50- to 79-year-old subjects were recruited to consume either 0, 80 or 160 mg of sodium nitrite per d for a 10-week period( Reference DeVan, Brooks and Evans 125 ). Results indicated no significant changes in blood pressure at week 10 compared with baseline blood pressure values; however, a significant time × treatment effect for carotid diameter in the nitrite groups was detected, as well as improved endothelial function of the brachial artery, suggesting improved vascular function with chronic inorganic nitrite supplementation despite a lack of an effect seen with blood pressure( Reference DeVan, Brooks and Evans 125 ). However, it is worth noting that the only prospective cohort study on this topic conducted by Golzarand et al. ( Reference Golzarand, Bahadoran and Mirmiran 177 ) found that higher dietary intakes of nitrate-containing vegetables (about 427·6 g/d) in normotensive individuals may have a protective effect against the development of hypertension (highest tertile of nitrate-containing vegetables, OR 0·63 (95% CI 0·41–0·98); P=0·05).

Endothelial dysfunction is one of the key early events involved in the development of atherosclerosis( Reference Raitakari and Celermajer 178 ). Flow-mediated dilatation is commonly used as a measure of endothelial function as reduced flow-mediated dilatation is an indicator of endothelial dysfunction (caused by reduced NO bioavailability) and has been associated with increased severity and duration of blood pressure elevations( Reference Hadi, Carr and Suwaidi 179 ). More recently, dietary inorganic nitrate interventions have been shown to significantly improve flow-mediated dilatation in healthy and hypertensive human subjects consuming spinach, beetroot juice or sodium nitrate capsules( Reference Kapil, Khambata and Robertson 29 , Reference Heiss, Meyer and Totzeck 168 , Reference Bondonno, Yang and Croft 180 , Reference Rodriguez-Mateos, Hezel and Aydin 181 ). Joris & Mensink( Reference Joris and Mensink 182 ) tested the effects of beetroot juice (containing approximately 500 mg nitrate) with a dietary load of fat (56·6 g fat) in overweight and obese subjects (BMI 30·1 (sd 1·9) kg/m2). While the control drink group saw impaired flow-mediated dilatation with dietary fat intake, the consumption of beetroot juice appeared to attenuate this impairment (beetroot juice: –0·37 (sd 2·92) % v. control: –1·56 (sd 2·9) %; P=0·03)( Reference Joris and Mensink 182 ). Additionally, flow-mediated dilatation has been shown to be reduced by approximately 40% after vascular ischaemia; however, Ingram et al. ( Reference Ingram, Fraser and Bleasdale 183 ) demonstrated that sodium nitrite pre-conditioning (providing a nitrite dose before ischaemic event) will prevent ischaemic reperfusion injury by preventing reductions in flow-mediated dilatation and endothelial dysfunction. Similar findings have been reported by Kapil et al. ( Reference Kapil, Khambata and Robertson 29 ) and Webb et al. ( Reference Webb, Patel and Loukogeorgakis 33 ) with beetroot juice pre-conditioning, indicating that higher plasma NOx concentrations achieved by inorganic NOx consumption may have a role for improving cardiovascular outcomes after vascular ischaemic events( Reference Kapil, Khambata and Robertson 29 , Reference Webb, Patel and Loukogeorgakis 33 ).

In addition to flow-mediated dilatation, CAC have been identified as an important indicator of vascular endothelial function, as they have a critical role in vascular repair( Reference Heiss, Jahn and Taylor 184 ). The number of CAC have also been shown to predict the occurrence of CVD and death( Reference Heiss, Meyer and Totzeck 168 ). Therefore it is of interest to note that Heiss et al. ( Reference Heiss, Meyer and Totzeck 168 ) have indicated an important role for dietary nitrate for increasing CAC, showing that a single dose of sodium nitrate (12·7 mg/kg body weight) can double the number of CAC 1–2 h post-nitrate ingestion.

Pulse wave velocity and augmentation index are accepted measurements of arterial stiffness and atherosclerosis, to which higher readings are associated with increased CVD risk( 185 , Reference Chirinos, Zambrano and Chakko 186 ). The role of dietary inorganic nitrate in preventing arterial stiffness has been established, as Kapil et al. ( Reference Kapil, Khambata and Robertson 29 ) found that a 4-week beetroot juice intervention reduced pulse wave velocity and augmentation index in hypertensive subjects. Zamani et al. ( Reference Zamani, Rawat and Shiva-Kumar 187 ) also saw a significantly reduced augmentation index with beetroot juice consumption in patients with symptomatic heart failure (beetroot juice: 132·2 (sd 16·7) %; placebo: 141·2 (sd 21·9) %; mean change –9·1 (sd 15·4) %; P=0·03). Rammos et al. ( Reference Rammos, Hendgen-Cotta and Sobierajski 188 ) investigated the effect of a 4-week sodium nitrate supplementation trial in elderly volunteers with mild hypertension, and found that vascular stiffness was significantly improved in the nitrate-supplemented volunteers. This is a very significant finding given that vascular stiffness tends to naturally increase with age( Reference Liu, Bondonno and Croft 189 ).

In an randomized controlled trial conducted by Jones et al. ( Reference Jones, Pellaton and Velmurugan 190 ), participants prone to MI and undergoing primary percutaneous coronary intervention (non-surgical intervention to treat stenosis) were administered with either a high-dose bolus injection of NaNO2 (1·8 µmol) or NaCl placebo. The nitrite group experienced a significantly (P=0·05) improved myocardial salvage index (established indicator of cardioprotective benefit) relative to placebo( Reference Jones, Pellaton and Velmurugan 190 ). In addition, a subset of participants who exhibited a blocked blood vessel experienced a 19% reduction in infarct size with nitrite treatment compared with placebo( Reference Jones, Pellaton and Velmurugan 190 ). A 1-year follow-up of study participants also found that the nitrite group experienced a significant reduction in major adverse cardiac events (NaNO2 : 2·6% v. NaCl: 15·8%; P=0·04)( Reference Jones, Pellaton and Velmurugan 190 ).

Conclusion

CVD remains the major killer from any disease across the developed world. Currently the available evidence indicates a role for dietary nitrate for improving CVD risk factors, a highly valuable finding given that dietary nitrate from beetroot and green leafy vegetables could represent a relatively simple and cost-effective treatment/preventative strategy for reducing CVD and its sequelae. However, at present it remains unclear whether incidence of CVD morbidity or mortality can be reduced with long-term dietary intakes of inorganic nitrate, as such evidence investigating this question directly has not yet been published. At present, there is an overwhelming need for epidemiological research to be conducted to identify the potential long-term effects of sustained inorganic nitrate and nitrite consumption on the development of CVD and its consequences.

Acknowledgements

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

We acknowledge the contribution of all authors to the writing of the present review and J. J. for conceiving the article. All authors approved the final manuscript.

There are no conflicts of interest.

References

1. World Health Organization (2011) Global status report on noncommunicable diseases 2010. http://www.who.int/nmh/publications/ncd_report_full_en.pdf (accessed December 2015).Google Scholar
2. Buttar, HS, Li, T & Ravi, N (2005) Prevention of cardiovascular diseases: role of exercise, dietary interventions, obesity and smoking cessation. Exp Clin Cardiol 10, 229249.Google ScholarPubMed
3. Cordain, L, Eaton, SB, Sebastian, A, et al. (2005) Origins and evolution of the Western diet: health implications for the 21st century. Am J Clin Nutr 81, 341354.Google Scholar
4. Barengo, NC, Hu, G, Lakka, TA, et al. (2004) Low physical activity as a predictor for total and cardiovascular disease mortality in middle-aged men and women in Finland. Eur Heart J 25, 22042211.Google Scholar
5. Clair, C, Rigotti, NA, Porneala, B, et al. (2013) Association of smoking cessation and weight change with cardiovascular disease among adults with and without diabetes. JAMA 309, 10141021.CrossRefGoogle ScholarPubMed
6. Isomaa, B, Almgren, P, Tuomi, T, et al. (2001) Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care 24, 683689.Google Scholar
7. Carpentier, Y & Komsa-Penkova, R (2011) Clinical Nutrition University. The place of nutrition in the prevention of cardiovascular diseases (CVDs). E Spen Eur E J Clin Nutr Metab 6, e272e282.Google Scholar
8. Harnack, L, Block, G & Lane, S (1997) Influence of selected environmental and personal factors on dietary behavior for chronic disease prevention: a review of the literature. J Nutr Educ 29, 306312.Google Scholar
9. Sofi, F, Cesari, F, Abbate, R, et al. (2008) Adherence to Mediterranean diet and health status: meta-analysis. BMJ 337, a1344.Google Scholar
10. 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
11. Verlangieri, A, Kapeghian, J, El-Dean, S, et al. (1985) Fruit and vegetable consumption and cardiovascular mortality. Med Hypotheses 16, 715.CrossRefGoogle ScholarPubMed
12. He, FJ, Nowson, CA & MacGregor, GA (2006) Fruit and vegetable consumption and stroke: meta-analysis of cohort studies. Lancet 367, 320326.Google Scholar
13. 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
14. Wang, X, Ouyang, Y, Liu, J, et al. (2014) Fruit and vegetable consumption and mortality from all causes, cardiovascular disease, and cancer: systematic review and dose–response meta-analysis of prospective cohort studies. BMJ 349, g4490.CrossRefGoogle ScholarPubMed
15. Ness, AR, Powles, JW & Khaw, K-T (1996) Vitamin C and cardiovascular disease: a systematic review. J Cardiovasc Risk 3, 513521.Google Scholar
16. Chong, MF-F, Macdonald, R & Lovegrove, JA (2010) Fruit polyphenols and CVD risk: a review of human intervention studies. Br J Nutr 104, S28S39.Google Scholar
17. Threapleton, DE, Greenwood, DC, Evans, CE, et al. (2013) Dietary fibre intake and risk of cardiovascular disease: systematic review and meta-analysis. BMJ 347, f6879.Google Scholar
18. Vivekananthan, DP, Penn, MS, Sapp, SK, et al. (2003) Use of antioxidant vitamins for the prevention of cardiovascular disease: meta-analysis of randomised trials. Lancet 361, 20172023.Google Scholar
19. Hord, NG (2011) Dietary nitrates, nitrites, and cardiovascular disease. Curr Atheroscler Rep 13, 484492.Google Scholar
20. Bhupathiraju, SN, Wedick, NM, Pan, A, et al. (2012) Quantity and variety in fruit and vegetable intake and risk of coronary heart disease. Am J Clin Nutr 98, 15141523.Google Scholar
21. Joshipura, KJ, Ascherio, A, Manson, JE, et al. (1999) Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA 282, 12331239.Google Scholar
22. Hord, NG, Tang, Y & Bryan, NS (2009) Food sources of nitrates and nitrites: the physiologic context for potential health benefits. Am J Clin Nutr 90, 110.Google Scholar
23. Sobko, T, Marcus, C, Govoni, M, et al. (2010) Dietary nitrate in Japanese traditional foods lowers diastolic blood pressure in healthy volunteers. Nitric Oxide 22, 136140.Google Scholar
24. L’Hirondel, J & L’Hirondel, J-L (2002) Nitrate and Man: Toxic, Harmless or Beneficial? Wallingford: CABI.Google Scholar
25. Lundberg, J & Weitzberg, E (2010) Chapter 16: Nitric oxide formation from inorganic nitrate and nitrite. In Nitric Oxide, 2nd ed., pp. 539553 [LJ Ignarro, editor]. Burlington, MA: Academic Press.Google Scholar
26. McKnight, G, Smith, L, Drummond, R, et al. (1997) Chemical synthesis of nitric oxide in the stomach from dietary nitrate in humans. Gut 40, 211214.CrossRefGoogle ScholarPubMed
27. Kapil, V, Weitzberg, E, Lundberg, J, et al. (2014) Clinical evidence demonstrating the utility of inorganic nitrate in cardiovascular health. Nitric Oxide 38, 4557.Google Scholar
28. Hobbs, DA, Kaffa, N, George, TW, et al. (2012) Blood pressure-lowering effects of beetroot juice and novel beetroot-enriched bread products in normotensive male subjects. Br J Nutr 108, 20662074.Google Scholar
29. Kapil, V, Khambata, RS, Robertson, A, et al. (2015) Dietary nitrate provides sustained blood pressure lowering in hypertensive patients: a randomized, phase 2, double-blind, placebo-controlled study. Hypertension 65, 320327.Google Scholar
30. Bailey, SJ, Winyard, P, Vanhatalo, A, et al. (2009) Dietary nitrate supplementation reduces the O2 cost of low-intensity exercise and enhances tolerance to high-intensity exercise in humans. J Appl Physiol 107, 11441155.Google Scholar
31. Kelm, M (1999) Nitric oxide metabolism and breakdown. Biochim Biophys Acta 1411, 273289.Google Scholar
32. Hotchkiss, J (1988) Nitrate, nitrite balance, and de novo synthesis of nitrate. Am J Clin Nutr 47, 161162.Google Scholar
33. 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
34. Li, H & Förstermann, U (2000) Nitric oxide in the pathogenesis of vascular disease. J Pathol 190, 244254.Google Scholar
35. Green, LC, De Luzuriaga, KR, Wagner, DA, et al. (1981) Nitrate biosynthesis in man. Proc Natl Acad Sci U S A 78, 77647768.Google Scholar
36. Palmer, R, Ashton, D & Moncada, S (1988) Vascular endothelial cells synthesize nitric oxide from l-arginine. Nature 333, 664666.Google Scholar
37. Guzik, TJ, Mussa, S, Gastaldi, D, et al. (2002) Mechanisms of increased vascular superoxide production in human diabetes mellitus: role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation 105, 16561662.Google Scholar
38. Taddei, S, Virdis, A, Ghiadoni, L, et al. (2001) Age-related reduction of NO availability and oxidative stress in humans. Hypertension 38, 274279.Google Scholar
39. Feron, O, Dessy, C, Moniotte, S, et al. (1999) Hypercholesterolemia decreases nitric oxide production by promoting the interaction of caveolin and endothelial nitric oxide synthase. J Clin Invest 103, 897905.Google Scholar
40. Kharitonov, SA, Robbins, RA, Yates, D, et al. (1995) Acute and chronic effects of cigarette smoking on exhaled nitric oxide. Am J Respir Crit Care Med 152, 609612.CrossRefGoogle ScholarPubMed
41. Böger, RH, Bode-Böger, SM, Szuba, A, et al. (1998) Asymmetric dimethylarginine (ADMA): a novel risk factor for endothelial dysfunction its role in hypercholesterolemia. Circulation 98, 18421847.Google Scholar
42. Ichiki, K, Ikeda, H, Haramaki, N, et al. (1996) Long-term smoking impairs platelet-derived nitric oxide release. Circulation 94, 31093114.Google Scholar
43. Bryan, NS & Loscalzo, J (2011) Nitrite and Nitrate in Human Health and Diesease. New York: Humana Press.Google Scholar
44. Sibal, L, Agarwal, SC, Home, PD, et al. (2010) The role of asymmetric dimethylarginine (ADMA) in endothelial dysfunction and cardiovascular disease. Curr Cardiol Rev 6, 8290.Google Scholar
45. Creager, MA, Gallagher, SJ, Girerd, XJ, et al. (1992) l-Arginine improves endothelium-dependent vasodilation in hypercholesterolemic humans. J Clin Invest 90, 12481253.Google Scholar
46. Bednarz, B, Jaxa-Chamiec, T, Maciejewski, P, et al. (2005) Efficacy and safety of oral l-arginine in acute myocardial infarction. Results of the multicenter, randomized, double-blind, placebo-controlled ARAMI pilot trial. Kardiol Pol 62, 421427.Google Scholar
47. Schulman, SP, Becker, LC, Kass, DA, et al. (2006) l-Arginine therapy in acute myocardial infarction: the Vascular Interaction With Age in Myocardial Infarction (VINTAGE MI) randomized clinical trial. JAMA 295, 5864.Google Scholar
48. The Natural Standard Research Collaboration (2013) Drugs and supplements: arginine. Mayo Foundation for Medical Education and Research. http://www.mayoclinic.org/drugs-supplements/arginine/safety/hrb-20058733 (accessed June 2016).Google Scholar
49. Nakaki, T & Hishikawa, K (2002) The arginine paradox. Nihon Yakurigaku Zasshi 199, 714.CrossRefGoogle Scholar
50. Mirmiran, P, Bahadoran, Z, Ghasemi, A, et al. (2016) The association of dietary l-arginine intake and serum nitric oxide metabolites in adults: a population-based study. Nutrients 8, E311.Google Scholar
51. Weitzberg, E & Lundberg, J (1998) Nonenzymatic nitric oxide production in humans. Nitric Oxide 2, 17.Google Scholar
52. Zweier, JL, Samouilov, A & Kuppusamy, P (1999) Non-enzymatic nitric oxide synthesis in biological systems. Biochim Biophys Acta 1411, 250262.Google Scholar
53. 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
54. van Velzen, AG, Sips, AJ, Schothorst, RC, et al. (2008) The oral bioavailability of nitrate from nitrate-rich vegetables in humans. Toxicol Lett 181, 177181.Google Scholar
55. 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
56. Pannala, AS, Mani, AR, Spencer, JP, et al. (2003) The effect of dietary nitrate on salivary, plasma, and urinary nitrate metabolism in humans. Free Radic Biol Med 34, 576584.Google Scholar
57. Bondonno, CP, Liu, AH, Croft, KD, et al. (2015) Absence of an effect of high nitrate intake from beetroot juice on blood pressure in treated hypertensive individuals: a randomized controlled trial. Am J Clin Nutr 102, 368375.Google Scholar
58. Tannenbaum, S, Weisman, M & Fett, D (1976) The effect of nitrate intake on nitrite formation in human saliva. Food Cosmet Toxoicol 14, 549552.CrossRefGoogle ScholarPubMed
59. Benjamin, N, O’Driscoll, F, Dougall, H, et al. (1994) Stomach NO synthesis. Nature 368, 502.Google Scholar
60. Coggan, AR, Leibowitz, JL, Spearie, CA, et al. (2015) Acute dietary nitrate intake improves muscle contractile function in patients with heart failure a double-blind, placebo-controlled, randomized trial. Circ Heart Fail 8, 914920.Google Scholar
61. Lundberg, JO, Weitzberg, E & Gladwin, MT (2008) The nitrate–nitrite–nitric oxide pathway in physiology and therapeutics. Nat Rev Drug Discov 7, 156167.Google Scholar
62. Sobko, T, Reinders, C, Norin, E, et al. (2004) Gastrointestinal nitric oxide generation in germ-free and conventional rats. Am J Physiol Gastrointest Liver Physiol 287, G993G997.Google Scholar
63. Bondonno, CP, Liu, AH, Croft, KD, et al. (2015) Antibacterial mouthwash blunts oral nitrate reduction and increases blood pressure in treated hypertensive men and women. Am J Hypertens 28, 572575.Google Scholar
64. Govoni, M, Jansson, , Weitzberg, E, et al. (2008) The increase in plasma nitrite after a dietary nitrate load is markedly attenuated by an antibacterial mouthwash. Nitric Oxide 19, 333337.Google Scholar
65. 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
66. Zhang, Z, Naughton, D, Winyard, PG, et al. (1998) Generation of nitric oxide by a nitrite reductase activity of xanthine oxidase: a potential pathway for nitric oxide formation in the absence of nitric oxide synthase activity. Biochem Biophys Res Commun 249, 767772.Google Scholar
67. Millar, TM, Stevens, CR, Benjamin, N, et al. (1998) Xanthine oxidoreductase catalyses the reduction of nitrates and nitrite to nitric oxide under hypoxic conditions. FEBS Lett 427, 225228.Google Scholar
68. Webb, A, Bond, R, McLean, P, et al. (2004) Reduction of nitrite to nitric oxide during ischemia protects against myocardial ischemia–reperfusion damage. Proc Natl Acad Sci U S A 101, 1368313688.Google Scholar
69. Duranski, MR, Greer, JJ, Dejam, A, et al. (2005) Cytoprotective effects of nitrite during in vivo ischemia–reperfusion of the heart and liver. J Clin Invest 115, 12321240.Google Scholar
70. Gladwin, MT, Raat, NJ, Shiva, S, et al. (2006) Nitrite as a vascular endocrine nitric oxide reservoir that contributes to hypoxic signaling, cytoprotection, and vasodilation. Am J Physiol Heart Circ Physiol 60, H2026H2035.Google Scholar
71. 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
72. Brooks, J (1937) The action of nitrite on haemoglobin in the absence of oxygen. Proc R Soc Lond B Biol Sci 123, 368382.Google Scholar
73. Doyle, MP, Pickering, RA, DeWeert, TM, et al. (1981) Kinetics and mechanism of the oxidation of human deoxyhemoglobin by nitrites. J Biol Chem 256, 1239312398.Google Scholar
74. 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
75. Lundberg, J, Weitzberg, E, Lundberg, J, et al. (1994) Intragastric nitric oxide production in humans: measurements in expelled air. Gut 35, 15431546.Google Scholar
76. Mowat, C, Carswell, A, Wirz, A, et al. (1999) Omeprazole and dietary nitrate independently affect levels of vitamin C and nitrite in gastric juice. Gastroenterology 116, 813822.Google Scholar
77. 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
78. Crawford, NM (1995) Nitrate: nutrient and signal for plant growth. Plant Cell 7, 859868.Google Scholar
79. Ysart, G, Clifford, R & Harrison, N (1999) Monitoring for nitrate in UK-grown lettuce and spinach. Food Addit Contam 16, 301306.CrossRefGoogle ScholarPubMed
80. Kaiser, WM & Brendle-Behnisch, E (1991) Rapid modulation of spinach leaf nitrate reductase activity by photosynthesis. I. Modulation in vivo by CO2 availability. Plant Physiol 96, 363367.Google Scholar
81. Muramoto, J (1999) Comparison of Nitrate Content in Leafy Vegetables from Organic and Conventional Farms in California. University of California, Santa Cruz, CA: Center for Agroecology and Sustainable Food Systems.Google Scholar
82. Ekart, K, Gorenjal, AH, Madorran, E, et al. (2013) Study on the influence of food processing on nitrate levels in vegetables. EFSA J 10, 514E.Google Scholar
83. Omar, SA, Artime, E & Webb, AJ (2012) A comparison of organic and inorganic nitrates/nitrites. Nitric Oxide 26, 229240.Google Scholar
84. Du, S-T, Zhang, Y-S & Lin, X-Y (2007) Accumulation of nitrate in vegetables and its possible implications to human health. Agric Sci China 6, 12461255.Google Scholar
85. Food Standards Australia New Zealand (2015) Survey of nitrates and nitrites in food and beverages in Australia. http://www.foodstandards.gov.au/consumer/additives/nitrate/Pages/default.aspx (accessed November 2015).Google Scholar
86. National Health and Medical Research Council & National Resource Management Ministerial Council (2011) Australian Drinking Water Guidelines Paper 6: National Water Quality Management Strategy. Canberra: National Health and Medical Research Council, National Resource Management Ministerial Council, Commonwealth of Australia.Google Scholar
87. United States Food and Drug Administration (2006) Food Additive Status List. Rockville, MD: United States Food and Drug Administration.Google Scholar
88. Ward, MH, DeKok, TM, Levallois, P, et al. (2005) Workgroup report: drinking-water nitrate and health – recent findings and research needs. Environ Health Perspect 113, 16071614.Google Scholar
89. Shuval, HI & Gruener, N (1972) Epidemiological and toxicological aspects of nitrates and nitrites in the environment. Am J Public Health 62, 10451052.Google Scholar
90. Jukes, D (2013) Food Additives in the European Union. Reading: The Department of Food Science and Technology, University of Reading. http://www.reading.ac.uk/foodlaw/additive.htm (accessed April 2017).Google Scholar
91. Food Standards Agency (2015) Food additives legislation guidance to compliance. http://www.food.gov.uk/sites/default/files/multimedia/pdfs/guidance/food-additives-legislation-guidance-to-compliance.pdf (accessed December 2016).Google Scholar
92. United States Department of Health and Human Services (2014) Food additives permitted for direct addition to food for human consumption. www.fda.gov/Food/ingredientsPackagingLabeling/FoodAdditivesUngredients.ucm091048.htm (accessed June 2016).Google Scholar
93. Weitzberg, E & Lundberg, JO (2013) Novel aspects of dietary nitrate and human health. Annu Rev Nutr 33, 129159.CrossRefGoogle ScholarPubMed
94. Machha, A & Schechter, AN (2011) Dietary nitrite and nitrate: a review of potential mechanisms of cardiovascular benefits. Eur J Nutr 50, 293303.Google Scholar
95. Kelm, M & Schrader, J (1990) Control of coronary vascular tone by nitric oxide. Circ Res 66, 15611575.Google Scholar
96. Cornwell, TL, Arnold, E, Boerth, NJ, et al. (1994) Inhibition of smooth muscle cell growth by nitric oxide and activation of cAMP-dependent protein kinase by cGMP. Am J Physiol Cell Physiol 267, C1405C1413.Google Scholar
97. Radomski, M, Palmer, R & Moncada, S (1987) Endogenous nitric oxide inhibits human platelet adhesion to vascular endothelium. Lancet 330, 10571058.Google Scholar
98. Kubes, P, Suzuki, M & Granger, D (1991) Nitric oxide: an endogenous modulator of leukocyte adhesion. Proc Natl Acad Sci U S A 88, 46514655.Google Scholar
99. De Caterina, R, Libby, P, Peng, H-B, et al. (1995) Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J Clin Invest 96, 6068.Google Scholar
100. Versari, D, Daghini, E, Virdis, A, et al. (2009) Endothelium‐dependent contractions and endothelial dysfunction in human hypertension. Br J Pharmacol 157, 527536.Google Scholar
101. Vanhoutte, P (1997) Endothelial dysfunction and atherosclerosis. Eur Heart J 18, Suppl. E, 1929.Google Scholar
102. Kleinbongard, P, Dejam, A, Lauer, T, et al. (2006) Plasma nitrite concentrations reflect the degree of endothelial dysfunction in humans. Free Radic Biol Med 40, 295302.Google Scholar
103. Landmesser, U & Drexler, H (2007) Endothelial function and hypertension. Curr Opin Cardiol 22, 316320.Google Scholar
104. Al Suwaidi, J, Hamasaki, S & Higano, ST (2000) Long-term follow-up of patients with mild coronary artery disease and endothelial dysfunction. Circulation 101, 948954.Google Scholar
105. Neunteufl, T, Katzenschlager, R, Hassan, A, et al. (1997) Systemic endothelial dysfunction is related to the extent and severity of coronary artery disease. Atherosclerosis 129, 111118.Google Scholar
106. Drexler, H, Hayoz, D, Münzel, T, et al. (1992) Endothelial function in chronic congestive heart failure. Am J Cardiol 69, 15961601.Google Scholar
107. Gokce, N, Keaney, JF, Hunter, LM, et al. (2003) Predictive value of noninvasively determined endothelial dysfunction for long-term cardiovascular events inpatients with peripheral vascular disease. J Am Coll Cardiol 41, 17691775.Google Scholar
108. Vita, JA & Keaney, JF (2002) Endothelial function a barometer for cardiovascular risk? Circulation 106, 640642.Google Scholar
109. Schächinger, V, Britten, MB & Zeiher, AM (2000) Prognostic impact of coronary vasodilator dysfunction on adverse long-term outcome of coronary heart disease. Circulation 101, 18991906.Google Scholar
110. Katz, SD, Hryniewicz, K, Hriljac, I, et al. (2005) Vascular endothelial dysfunction and mortality risk in patients with chronic heart failure. Circulation 111, 310314.Google Scholar
111. Lidder, S & Webb, AJ (2013) Vascular effects of dietary nitrate (as found in green leafy vegetables and beetroot) via the nitrate–nitrite–nitric oxide pathway. Br J Clin Pharmacol 75, 677696.Google Scholar
112. Lundberg, JO, Gladwin, MT & Weitzberg, E (2015) Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discov 14, 623641.Google Scholar
113. Jeerooburkhan, N, Jones, LC, Bujac, S, et al. (2001) Genetic and environmental determinants of plasma nitrogen oxides and risk of ischemic heart disease. Hypertension 38, 10541061.Google Scholar
114. Panza, JA, Quyyumi, AA, Brush, JE Jr, et al. (1990) Abnormal endothelium-dependent vascular relaxation in patients with essential hypertension. N Engl J Med 323, 2227.Google Scholar
115. Casino, PR, Kilcoyne, CM, Quyyumi, AA, et al. (1993) The role of nitric oxide in endothelium-dependent vasodilation of hypercholesterolemic patients. Circulation 88, 25412547.Google Scholar
116. Henry, RM, Ferreira, I, Kostense, PJ, et al. (2004) Type 2 diabetes is associated with impaired endothelium-dependent, flow-mediated dilation, but impaired glucose metabolism is not: The Hoorn Study. Atherosclerosis 174, 4956.Google Scholar
117. Tsuchiya, M, Asada, A, Kasahara, E, et al. (2002) Smoking a single cigarette rapidly reduces combined concentrations of nitrate and nitrite and concentrations of antioxidants in plasma. Circulation 105, 11551157.Google Scholar
118. Green, DJ, Maiorana, A, O’Driscoll, G, et al. (2004) Effect of exercise training on endothelium‐derived nitric oxide function in humans. J Physiol 561, 125.Google Scholar
119. Lopez-Garcia, E, Schulze, MB, Fung, TT, et al. (2004) Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr 80, 10291035.Google Scholar
120. Celermajer, DS, Sorensen, KE, Spiegelhalter, DJ, et al. (1994) Aging is associated with endothelial dysfunction in healthy men years before the age-related decline in women. J Am Coll Cardiol 24, 471476.Google Scholar
121. Celermajer, D, Sorensen, K, Georgakopoulos, D, et al. (1993) Cigarette smoking is associated with dose-related and potentially reversible impairment of endothelium-dependent dilation in healthy young adults. Circulation 88, 21492155.Google Scholar
122. Fuentes, F, Lopez-Miranda, J, Sanchez, E, et al. (2001) Mediterranean and low-fat diets improve endothelial function in hypercholesterolemic men. Ann Intern Med 134, 11151119.Google Scholar
123. O’Driscoll, G, Green, D & Taylor, RR (1997) Simvastatin, an HMG-coenzyme A reductase inhibitor, improves endothelial function within 1 month. Circulation 95, 11261131.Google Scholar
124. Hornig, B, Maier, V & Drexler, H (1996) Physical training improves endothelial function in patients with chronic heart failure. Circulation 93, 210214.Google Scholar
125. DeVan, A, Brooks, F, Evans, T, et al. (2014) Safety and efficacy of sodium nitrite supplementation for improving vascular endothelial dysfunction in middle-aged and older healthy adults. FASEB J 28, Suppl., 698.4.Google Scholar
126. Wilson, J & Hunt, T (2014) Molecular Biology of the Cell: The Problems Book. New York: Garland Science.Google Scholar
127. Channon, KM, Qian, H & George, SE (2000) Nitric oxide synthase in atherosclerosis and vascular injury: insights from experimental gene therapy. Arterioscler Thromb Vasc Biol 20, 18731881.Google Scholar
128. 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
129. Simon, DI, Stamler, JS, Jaraki, O, et al. (1993) Antiplatelet properties of protein S-nitrosothiols derived from nitric oxide and endothelium-derived relaxing factor. Arterioscler Thromb Vasc Biol 13, 791799.Google Scholar
130. Clapp, BR, Hingorani, AD, Kharbanda, RK, et al. (2004) Inflammation-induced endothelial dysfunction involves reduced nitric oxide bioavailability and increased oxidant stress. Cardiovasc Res 64, 172178.Google Scholar
131. Vallance, PJ & Webb, DJ (2003) Vascular Endothelium in Human Physiology and Pathophysiology, vol. 7. Boca Raton, FL: CRC Press.Google Scholar
132. Gago, B, Nyström, T, Cavaleiro, C, et al. (2008) The potent vasodilator ethyl nitrite is formed upon reaction of nitrite and ethanol under gastric conditions. Free Radic Biol Med 45, 404412.Google Scholar
133. Klemenska, E & Beresewicz, A (2009) Bioactivation of organic nitrates and the mechanism of nitrate tolerance. Cardiol J 16, 1119.Google Scholar
134. Bassenge, E, Fink, N, Skatchkov, M, et al. (1998) Dietary supplement with vitamin C prevents nitrate tolerance. J Clin Invest 102, 6771.Google Scholar
135. Gori, T, Burstein, JM, Ahmed, S, et al. (2001) Folic acid prevents nitroglycerin-induced nitric oxide synthase dysfunction and nitrate tolerance. Circulation 104, 11191123.Google Scholar
136. Fontaine, D, Otto, A, Fontaine, J, et al. (2003) Prevention of nitrate tolerance by long-term treatment with statins. Cardiovasc Drugs Ther 17, 123128.Google Scholar
137. Greer, FR & Shannon, M (2005) Infant methemoglobinemia: the role of dietary nitrate in food and water. Pediatrics 116, 784786.Google Scholar
138. Martinez, A, Sanchez-Valverde, F, Gil, F, et al. (2013) Methemoglobinemia induced by vegetable intake in infants in northern Spain. J Peadiatr Gastroenterol Nutr 56, 573577.Google Scholar
139. Milkowski, A, Garg, HK, Coughlin, JR, et al. (2010) Nutritional epidemiology in the context of nitric oxide biology: a risk–benefit evaluation for dietary nitrite and nitrate. Nitric Oxide 22, 110119.Google Scholar
140. Avery, AA (1999) Infantile methemoglobinemia: reexamining the role of drinking water nitrates. Environ Health Perspect 107, 583586.Google Scholar
141. Bouvard, V, Loomis, D, Guyton, KZ, et al. (2015) Carcinogenicity of consumption of red and processed meat. Lancet Oncol 16, 15991600.Google Scholar
142. Bingham, SA (1999) High-meat diets and cancer risk. Proc Nutr Soc 58, 243248.Google Scholar
143. Gilchrist, M, Winyard, PG & Benjamin, N (2010) Dietary nitrate – good or bad? Nitric Oxide 22, 104109.Google Scholar
144. Magee, PN & Barnes, J (1956) The production of malignant primary hepatic tumours in the rat by feeding dimethylnitrosamine. Br J Cancer 10, 114122.Google Scholar
145. Assembly of Life Sciences (1981) The Health Effects of Nitrate, Nitrite, and N-Nitroso Compounds: Part 1 of a 2-Part Study. Washington, DC: National Academies Press.Google Scholar
146. Kapil, V, Milsom, AB, Okorie, M, et al. (2010) Inorganic nitrate supplementation lowers blood pressure in humans. Hypertension 56, 274281.Google Scholar
147. Asgary, S, Afshani, M, Sahebkar, A, et al. (2016) Improvement of hypertension, endothelial function and systemic inflammation following short-term supplementation with red beet (Beta vulgaris L.) juice: a randomized crossover pilot study. J Hum Hypertens 30, 627632.Google Scholar
148. Richardson, G, Hicks, S, O’Byrne, S, et al. (2002) The ingestion of inorganic nitrate increases gastric S-nitrosothiol levels and inhibits platelet function in humans. Nitric Oxide 7, 2429.Google Scholar
149. Baker, JE, Su, J, Fu, X, et al. (2007) Nitrite confers protection against myocardial infarction: role of xanthine oxidoreductase, NADPH oxidase and K ATP channels. J Mol Cell Cardiol 43, 437444.Google Scholar
150. Lansley, KE, Winyard, PG, Bailey, SJ, et al. (2011) Acute dietary nitrate supplementation improves cycling time trial performance. Med Sci Sports Exerc 43, 11251131.Google Scholar
151. Dykhuizen, R, Frazer, R, Duncan, C, et al. (1996) Antimicrobial effect of acidified nitrite on gut pathogens: importance of dietary nitrate in host defense. Antimicrob Agents Chemother 40, 14221425.Google Scholar
152. Carlström, M, Persson, AEG, Larsson, E, et al. (2011) Dietary nitrate attenuates oxidative stress, prevents cardiac and renal injuries, and reduces blood pressure in salt-induced hypertension. Cardiovasc Res 89, 574585.Google Scholar
153. Petersson, J, Carlström, 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
154. Kanematsu, Y, Yamaguchi, K, Ohnishi, H, et al. (2008) Dietary doses of nitrite restore circulating nitric oxide level and improve renal injury in l-NAME-induced hypertensive rats. Am J Physiol Renal Physiol 295, F1457F1462.Google Scholar
155. Ferguson, SK, Hirai, DM, Copp, SW, et al. (2013) Impact of dietary nitrate supplementation via beetroot juice on exercising muscle vascular control in rats. J Physiol 591, 547557.Google Scholar
156. Bryan, NS, Calvert, JW, Elrod, JW, et al. (2007) Dietary nitrite supplementation protects against myocardial ischemia–reperfusion injury. Proc Natl Acad Sci U S A 104, 1914419149.Google Scholar
157. Shiva, S, Sack, MN, Greer, JJ, et al. (2007) Nitrite augments tolerance to ischemia/reperfusion injury via the modulation of mitochondrial electron transfer. J Exp Med 204, 20892102.Google Scholar
158. Minicucci, MF, Azevedo, PS, Polegato, BF, et al. (2011) Heart failure after myocardial infarction: clinical implications and treatment. Clin Cardiol 34, 410414.Google Scholar
159. Nieswandt, B, Pleines, I & Bender, M (2011) Platelet adhesion and activation mechanisms in arterial thrombosis and ischaemic stroke. J Thromb Haemost 9, 92104.Google Scholar
160. Raskob, GE, Angchaisuksiri, P, Blanco, AN, et al. (2014) Thrombosis: a major contributor to global disease burden. Semi Throm Hemost 40, 724735.Google Scholar
161. Park, JW, Piknova, B, Huang, PL, et al. (2013) Effect of blood nitrite and nitrate levels on murine platelet function. PLOS ONE 8, e55699.Google Scholar
162. Apostoli, G, Solomon, A, Smallwood, M, et al. (2014) Role of inorganic nitrate and nitrite in driving nitric oxide–cGMP‐mediated inhibition of platelet aggregation in vitro and in vivo . J Thromb Haemost 12, 18801889.Google Scholar
163. Moraes, DL, Colucci, WS & Givertz, MM (2000) Secondary pulmonary hypertension in chronic heart failure. Circulation 102, 17181723.Google Scholar
164. Hunter, CJ, Dejam, A, Blood, AB, et al. (2004) Inhaled nebulized nitrite is a hypoxia-sensitive NO-dependent selective pulmonary vasodilator. Nat Med 10, 11221127.Google Scholar
165. Zuckerbraun, BS, Shiva, S, Ifedigbo, E, et al. (2010) Nitrite potently inhibits hypoxic and inflammatory pulmonary arterial hypertension and smooth muscle proliferation via xanthine oxidoreductase–dependent nitric oxide generation. Circulation 121, 98109.Google Scholar
166. Casey, DB, Badejo, AM, Dhaliwal, JS, et al. (2009) Pulmonary vasodilator responses to sodium nitrite are mediated by an allopurinol-sensitive mechanism in the rat. Am J Physiol Heart Circ Physiol 296, H524H533.Google Scholar
167. Hendgen-Cotta, UB, Luedike, P, Totzeck, M, et al. (2012) Dietary nitrate supplementation improves revascularization in chronic ischemia. Circulation 126, 19831992.Google Scholar
168. Heiss, C, Meyer, C, Totzeck, M, et al. (2012) Dietary inorganic nitrate mobilizes circulating angiogenic cells. Free Radic Biol Med 52, 17671772.Google Scholar
169. Sindler, AL, Fleenor, BS, Calvert, JW, et al. (2011) Nitrite supplementation reverses vascular endothelial dysfunction and large elastic artery stiffness with aging. Aging Cell 10, 429437.Google Scholar
170. Cai, H & Harrison, DG (2000) Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res 87, 840844.Google Scholar
171. Stokes, KY, Dugas, TR, Tang, Y, et al. (2009) Dietary nitrite prevents hypercholesterolemic microvascular inflammation and reverses endothelial dysfunction. Am J Physiol Heart Circ Physiol 296, H1281H1288.Google Scholar
172. Carlström, M, Larsen, FJ, Nyström, T, et al. (2010) Dietary inorganic nitrate reverses features of metabolic syndrome in endothelial nitric oxide synthase-deficient mice. Proc Natl Acad Sci U S A 107, 1771617720.Google Scholar
173. 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.Google Scholar
174. Siervo, M, Lara, J, Ogbonmwan, I, et al. (2013) Inorganic nitrate and beetroot juice supplementation reduces blood pressure in adults: a systematic review and meta-analysis. J Nutr 143, 818826.Google Scholar
175. Bondonno, CP, Liu, AH, Croft, KD, et al. (2014) Short-term effects of nitrate-rich green leafy vegetables on blood pressure and arterial stiffness in individuals with high-normal blood pressure. Free Radic Biol Med 77, 353362.Google Scholar
176. 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
177. Golzarand, M, Bahadoran, Z, Mirmiran, P, et al. (2016) Consumption of nitrate-containing vegetables is inversely associated with hypertension in adults: a prospective investigation from the Tehran Lipid and Glucose Study. J Nephrol 29, 377384.Google Scholar
178. Raitakari, OT & Celermajer, DS (2000) Testing for endothelial dysfunction. Ann Med 32, 293304.Google Scholar
179. Hadi, HA, Carr, CS & Suwaidi, J (2005) Endothelial dysfunction: cardiovascular risk factors, therapy, and outcome. Vasc Health Risk Manag 1, 183198.Google Scholar
180. 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
181. 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
182. 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
183. Ingram, TE, Fraser, AG, Bleasdale, RA, et al. (2013) Low-dose sodium nitrite attenuates myocardial ischemia and vascular ischemia–reperfusion injury in human models. J Am Coll Cardiol 61, 25342541.Google Scholar
184. Heiss, C, Jahn, S, Taylor, M, et al. (2010) Improvement of endothelial function with dietary flavanols is associated with mobilization of circulating angiogenic cells in patients with coronary artery disease. J Am Coll Cardiol 56, 218224.Google Scholar
185. The Reference Values for Arterial Stiffness’ Collaboration (2010) Determinants of pulse wave velocity in healthy people and in the presence of cardiovascular risk factors: ‘establishing normal and reference values’. Eur Heart J 31, 23382350.Google Scholar
186. Chirinos, JA, Zambrano, JP, Chakko, S, et al. (2005) Aortic pressure augmentation predicts adverse cardiovascular events in patients with established coronary artery disease. Hypertension 45, 980985.Google Scholar
187. Zamani, P, Rawat, D, Shiva-Kumar, P, et al. (2014) The effect of inorganic nitrate on exercise capacity in heart failure with preserved ejection fraction. Circulation 114, 371380.Google Scholar
188. Rammos, C, Hendgen-Cotta, UB, Sobierajski, J, et al. (2014) Dietary nitrate reverses vascular dysfunction in older adults with moderately increased cardiovascular risk. J Am Coll Cardiol 63, 15841585.Google Scholar
189. Liu, AH, Bondonno, CP, Croft, KD, et al. (2013) Effects of a nitrate-rich meal on arterial stiffness and blood pressure in healthy volunteers. Nitric Oxide 35, 123130.Google Scholar
190. Jones, DA, Pellaton, C, Velmurugan, S, et al. (2014) Randomized phase 2 trial of intra-coronary nitrite during acute myocardial infarction. Circ Res 114, 437447.Google Scholar
191. Alexander, J, Benford, D, Cockburn, A, et al. (2008) Nitrate in vegetables: Scientific Opinion of the Panel on Contaminants in the Food chain. EFSA J 689, 179.Google Scholar
192. Siciliano, J, Krulick, S, Heisler, EG, et al. (1975) Nitrate and nitrite content of some fresh and processed market vegetables. J Agric Food Chem 23, 461464.Google Scholar
193. Jackson, WA, Steel, JS & Boswell, VR (1967) Nitrates in edible vegetables and vegetable products. Proc Amer Soc Hort Sci 90, 349352.Google Scholar
194. Corré, WJ (1979) Nitrate and Nitrite in Vegetables. Wageningen, the Netherlands: Wageningen University and Research Centre.Google Scholar
195. Richardson, W (1907) The occurrence of nitrates in vegetable foods, in cured meats and elsewhere. J Am Chem Soc 29, 17571767.Google Scholar
196. Wilson, J (1949) Nitrate in foods and its relation to health. Agron J 41, 2022.Google Scholar
197. Lee, DH (1970) Nitrates, nitrites, and methemoglobinemia. Environ Res 3, 484511.Google Scholar
198. White, JW (1975) Relative significance of dietary sources of nitrate and nitrite. J Agric Food Chem 23, 886891.Google Scholar
199. Santamaria, P, Elia, A, Serio, F, et al. (1999) A survey of nitrate and oxalate content in fresh vegetables. J Sci Food Agric 79, 18821888.Google Scholar
200. Öztekin, N, Nutku, MS & Erim, FB (2002) Simultaneous determination of nitrite and nitrate in meat products and vegetables by capillary electrophoresis. Food Chem 76, 103106.Google Scholar
201. Tamme, T, Reinik, M, Roasto, M, et al. (2006) Nitrates and nitrites in vegetables and vegetable-based products and their intakes by the Estonian population. Food Addit Contam 23, 355361.Google Scholar
202. Hsu, J, Arcot, J & Lee, NA (2009) Nitrate and nitrite quantification from cured meat and vegetables and their estimated dietary intake in Australians. Food Chem 115, 334339.Google Scholar
203. Thomson, B, Nokes, C & Cressey, P (2007) Intake and risk assessment of nitrate and nitrite from New Zealand foods and drinking water. Food Addit Contam 24, 113121.Google Scholar
204. Walker, R (1990) Nitrates, nitrites and N‐nitrosocompounds: a review of the occurrence in food and diet and the toxicological implications. Food Addit Contam 7, 717768.Google Scholar
205. Wang, Z, Wei, Y & Li, S (2000) Nitrate accumulation and its regulation by nutrient management in vegetables. In Balanceable Fertilization and High Quality Vegetables Continual Production. Beijing: China Agricultural University.Google Scholar
206. Petersen, A & Stoltze, S (1999) Nitrate and nitrite in vegetables on the Danish market: content and intake. Food Addit Contam 16, 291299.Google Scholar
207. Ysart, G, Miller, P, Barrett, G, et al. (1999) Dietary exposures to nitrate in the UK. Food Addit Contam 16, 521532.Google Scholar
208. Sušin, J, Kmecl, V & Gregorčič, A (2006) A survey of nitrate and nitrite content of fruit and vegetables grown in Slovenia during 1996–2002. Food Addit Contam 23, 385390.Google Scholar
209. Pickston, L, Smith, J & Todd, M (1980) Nitrate and nitrite levels in fruit and vegetables in New Zealand and the effect of storage and pressure cooking on these levels. Food Technol New Zeal 15, 1117.Google Scholar
210. Zhong, W, Hu, C & Wang, M (2002) Nitrate and nitrite in vegetables from north China: content and intake. Food Addit Contam 19, 11251129.Google Scholar
211. Panalaks, T, Iyengar, J & Sen, N (1973) Nitrate, nitrite, and dimethylnitrosamine in cured meat products. J Ass Offic Anal Chem 56, 621625.Google Scholar
212. Sen, NP & Baddoo, PA (1997) Trends in the levels of residual nitrite in Canadian cured meat products over the past 25 years. J Agric Food Chem 45, 47144718.Google Scholar
213. Sen, NP, Baddoo, PA & Seaman, SW (1994) Rapid and sensitive determination of nitrite in foods and biological materials by flow injection or high-performance liquid chromatography with chemiluminescence detection. J Chromatogr A 673, 7784.Google Scholar
214. Panalaks, T, Iyengar, JR, Donaldson, BA, et al. (1974) Further survey of cured meat products for volatile N-nitrosamines. J Assoc Off Anal Chem 57, 806812.Google Scholar
215. Meah, M, Harrison, N & Davies, A (1994) Nitrate and nitrite in foods and the diet. Food Addit Contam 11, 519532.Google Scholar
216. Buege, D, Lee, M & Cassens, R (1978) Residual Nitrite Levels in Meat Products Manufactured by Wisconsin Meat Processors. Madison, WI: University of Wisconsin.Google Scholar
217. Reinik, M, Tamme, T, Roasto, M, et al. (2005) Nitrites, nitrates and N-nitrosoamines in Estonian cured meat products: intake by Estonian children and adolescents. Food Addit Contam 22, 10981105.Google Scholar
218. Cassens, RG (1997) Composition and safety of cured meats in the USA. Food Chem 59, 561566.Google Scholar
219. Kerr, R, Marsh, C, Schroeder, W, et al. (1926) The use of sodium nitrite in the curing of meat. J Agric Res 33, 541551.Google Scholar
220. Coppola, ED, Wickroski, AF & Hanna, JG (1976) Nitrite in meat products determined by fluorescence quenching of p-aminobenzoate ion. J Assoc Off Anal Chem 59, 783786.Google Scholar
221. Siu, DC & Henshall, A (1998) Ion chromatographic determination of nitrate and nitrite in meat products. J Chromatogr A 804, 157160.Google Scholar
222. Greenberg, R (1977) Nitrosopyrrolidine in United States cured meat products. In Proceedings of the International Symposium on Nitrite in Meat Products, 1976, Zeist, the Netherlands, pp. 203–210 [BJ Tinbergen and B Krol, editors]. Wageningen, the Netherlands: Centre for Agricultural Publishing and Documentation.Google Scholar
223. Sen, N, Donaldson, B & Charbonneau, C (1975) Formation of nitrosodimethylamine from the interaction of certain pesticides and nitrite. In N-Nitroso Compounds in the Environment, IARC Scientific Publications , no. 9, pp. 7579 [P Bogovski and EA Walker, editors]. Lyon, France: IARC.Google Scholar
224. Sen, N, Donaldson, B, Seaman, S, et al. (1977) Recent nitrosamine analyses in cooked bacon. J Can Inst Food Sci Technol 10, A13A15.Google Scholar
Figure 0

Fig. 1 The fate of dietary nitrate. Nitrate is systematically absorbed becoming concentrated in the salivary glands and part of the salivary circulation. Salivary nitrate is reduced to nitrite by oral bacteria. In the stomach nitrite may produce NO. Nitrite transported in arterial circulation can be reduced to NO in low oxygen concentrations which can lead to vasodilation and reductions in blood pressure (Webb A, Patel N, Loukogeorhakis S, et al. Acute blood pressure lowering,vasoprotective, and antiplatelet properties of dietary nitrate via bioconversion to nitrite. Hypertension, vol. 51, pp. 784–790, from http://hyper.ahajournals.org/content/51/3/784.short(33)).

Figure 1

Table 1 Permissions for nitrate and nitrite in food products*

Figure 2

Table 2 Vegetable sources of nitrate and nitrite with estimated nitrate and/or nitrite contents* (Mean values and ranges)

Figure 3

Table 3 Meat-based sources of nitrate and nitrite with estimated nitrate and/or nitrite contents* (Mean values and ranges)

Figure 4

Table 4 Fruit sources of nitrate and nitrite with estimated nitrate and/or nitrite contents* (Mean values and ranges)

Figure 5

Table 5 Nitrate- and nitrite-containing herbs with estimated nitrate and/or nitrite contents* (Mean values and ranges)

Figure 6

Fig. 2 Chemical structure of inorganic nitrate/nitrite compared with organic mono-, di-, tri- and tetra-nitrates/nitrites. 5-ISMN, isosorbide-5-mononitrate; ISDN, isosorbide dinitrate; GTN, glyceryl trinitrate; ETN, erythritol tetranitrate; PETriN, pentaerythrityl trinitrate; PETN, pentaerythritol tetranitrate. Reprinted from Omar et al.(83), with permission from Elsevier.