Hostname: page-component-8448b6f56d-gtxcr Total loading time: 0 Render date: 2024-04-24T08:51:19.497Z Has data issue: false hasContentIssue false

Does dietary nitrate say NO to cardiovascular ageing? Current evidence and implications for research

Published online by Cambridge University Press:  10 May 2018

Mario Siervo*
Affiliation:
Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Leech Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
Filippo Scialò
Affiliation:
Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, University of Newcastle, Newcastle upon Tyne NE4 5PL, UK
Oliver M. Shannon
Affiliation:
Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Leech Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
Blossom C.M. Stephan
Affiliation:
Institute of Health and Society and Newcastle University Institute of Ageing, Newcastle University, Biomedical Research Building, Campus of Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
Ammar W. Ashor
Affiliation:
Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Leech Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK Department of Pharmacology, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
*
*Corresponding author: Dr Mario Siervo, email mario.siervo@ncl.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

CVD are characterised by a multi-factorial pathogenesis. Key pathogenetic steps in the development of CVD are the occurrence of endothelial dysfunction and formation of atherosclerotic lesions. Reduced nitric oxide (NO) bioavailability is a primary event in the initiation of the atherosclerotic cascade. NO is a free radical with multiple physiological functions including the regulation of vascular resistance, coagulation, immunity and oxidative metabolism. The synthesis of NO proceeds via two distinct pathways identified as enzymatic and non-enzymatic. The former involves the conversion of arginine into NO by the NO synthases, whilst the latter comprises a two-step reducing process converting inorganic nitrate $({\rm NO}_3^ - )$ into nitrite and subsequently NO.

Inorganic ${\rm NO}_3^ - $ is present in water and food, particularly beetroot and green leafy vegetables. Several investigations have therefore used the non-enzymatic NO pathway as a target for nutritional supplementation (${\rm NO}_3^ - $ salts) or dietary interventions (high-${\rm NO}_3^ - $ foods) to increase NO bioavailability and impact on cardiovascular outcomes. Some studies have reported positive effects of dietary ${\rm NO}_3^ - $ on systolic blood pressure and endothelial function in patients with hypertension and chronic heart failure. Nevertheless, results have been inconsistent and the size of the effect appears to be declining in older individuals. Additionally, there is a paucity of studies for disorders such as diabetes, CHD and chronic kidney failure. Thus, whilst dietary ${\rm NO}_3^ - $ supplementation could represent an effective and viable strategy for the primary and secondary prevention of age-related cardiovascular and metabolic diseases, more large-scale, robust studies are awaited to confirm or refute this notion.

Type
Conference on ‘What governs what we eat?’
Copyright
Copyright © The Authors 2018 

CVD are the leading cause of death worldwide and a major cause of morbidity and disability(Reference Townsend, Wickramasinghe and Bhatnagar1). In the UK, cardiovascular mortality accounts for 19 and 28 % of premature deaths among women and men, respectively(Reference Bhatnagar, Wickramasinghe and Williams2). CVD are characterised by a multifactorial pathogenesis including genetic, diet and lifestyle factors(Reference Frayn and Stanner3). The clinical outcomes of CVD, such as heart failure, atrial fibrillation and cerebrovascular disease, are largely attributed to a reduction in the blood supply to associated organs and tissues. This occurs secondary to thickening of the walls of the blood vessels and formation of obstructive atherosclerotic plaques(Reference Gimbrone and Garcia-Cardena4). Endothelial dysfunction (ED) appears to be a critical step in the initiation of the atherosclerotic process(Reference Gimbrone and Garcia-Cardena4).

The endothelium

The endothelium is a monolayer of cells separating the vascular lumen from the rest of the blood vessel. It is now recognised that the endothelium has vital paracrine, endocrine and autocrine functions(Reference Sena, Pereira and Seica5). Therefore, in addition to helping maintain blood flow, the main function of the endothelium is to serve as an endocrine organ(Reference Michiels6). The endothelium generates several extracellular messengers that mediate multiple functions including preserving haemostatic balance(Reference Rajendran, Rengarajan and Thangavel7). In addition to insulating the thrombogenic sub-endothelial layers, the endothelium secretes molecules that inhibit the inappropriate formation of thrombus including nitric oxide (NO), prostacyclin I2, tissue plasminogen activator and protein C/protein S(Reference Rajendran, Rengarajan and Thangavel7). However, in cases of vessel damage and exposure to certain pro-inflammatory substances, the balance is shifted towards a procoagulant/prothrombotic state(Reference Sena, Pereira and Seica5). This stimulates the endothelium to secrete agents that help with platelet aggregation and clot formation including platelet activating factor, von Willebrand factor and thromboxane A2(Reference Sena, Pereira and Seica5, Reference Sumpio, Riley and Dardik8).

Normal vascular endothelium has anti-proliferative and anti-apoptotic properties that are mediated through the activity of NO, prostacyclin I2 and C-type natriuretic peptide. Moreover, endothelial cells secrete factors that promote proliferation of smooth muscle cells and the formation of new blood vessels, e.g. vascular endothelial growth factor, angiopoietins and adropins(Reference Mensah9). Further, NO secreted by the normal endothelium prevents inflammatory response in the vascular wall secondary to local injury. Dysfunction in the endothelium is characterised by disturbed vasodilator and anticoagulant function, increased adhesiveness of the vessel wall for platelets and leucocytes (inflammation), reduced fibrinolytic activity and breakdown of barrier function causing leakage and oedema formation(Reference Vanhoutte, Shimokawa and Tang10).

Nitric oxide and endothelial function

NO is a free radical gas molecule that is involved in the regulation of multiple physiological processes such as blood pressure (BP), glucose metabolism, inflammation and coagulation(Reference Siervo, Capuano and Colantuoni11). Reduced availability of NO contributes to pathological conditions including hypertension, diabetes, chronic heart failure or kidney failure(Reference Yetik-Anacak and Catravas12). NO is regarded as one of the most important molecules secreted by the endothelium. It is a highly diffusible molecule with a very short half-life (<1 s)(Reference Channon13). The production of NO is catalysed by the nitric oxide synthase enzyme (NOS). There are three isoforms of this enzyme including: endothelial (eNOS), neuronal and inducible(Reference Lei, Vodovotz and Tzeng14). The eNOS is a homodimeric enzyme expressed constitutively in the endothelial cells that facilitate the conversion of the amino acid l-arginine into l-citrulline and NO(Reference Michiels6). This process requires molecular oxygen and reduced NADPH as co-substrates, and the following cofactors: FAD, FMN, tetrahydrobiopterin, haeme and Ca–calmodulin(Reference Forstermann and Sessa15) (see Fig. 1).

Fig. 1. Exogenous and endogenous sources of nitric oxide (NO). NO is produced by a family of enzymes known as NO synthases (NOS) which utilise the substrate l-arginine. The ${\rm NO}_3^ - {\rm -} {\rm NO}_2^ - $–NO pathway has been proposed as an alternative pathway for NO generation. ADMA, asymmetric dimethylarginine; BH4, tetrahydrobiopterin; cGMP, cyclic GMP; ONOO, peroxynitrite; sGC, soluble guanylate cyclase.

The triggers for NO synthesis and release are either mechanical stretching of the vessel wall or release of receptor-mediated agonists such as bradykinin, acetylcholine or histamine(Reference Khazaei, Moien-Afshari and Laher16). These signals lead to an increase in intracellular calcium concentration. Intracellular Ca2+ binds to calmodulin to form Ca2+–calmodulin complex that mobilises eNOS from its binding to caveolin, thereby allowing the activated eNOS to catalyse the synthesis of NO from l-arginine(Reference Khazaei, Moien-Afshari and Laher16). Because of the gaseous nature of NO, it diffuses from where it is synthesised in the endothelium to the vascular smooth muscle where it activates soluble guanylate cyclase leading to increasing intracellular cyclic guanosine monophosphate. The cyclic guanosine monophosphate causes smooth muscle relaxation and, eventually, arterial dilation(Reference Forstermann17).

In addition to arterial dilation, NO has many other vital protective functions in blood vessels including decreasing: (1) smooth muscle proliferation; (2) platelet aggregation; (3) endothelin production; (4) monocytes and platelets adhesion; (5) expression of adhesion molecules; and (6) oxidation of LDL(Reference Vanhoutte, Shimokawa and Tang10). Because of the vital role of NO, researchers have suggested that reduced NO availability is the major cause of ED. This deficiency activates atherogenic processes in the vessel wall, which include vasoconstriction, monocyte activation and adherence to vascular endothelium, proliferation of smooth muscle cells, thrombosis and impaired coagulation and, eventually, atherosclerosis(Reference Versari, Daghini and Virdis18).

Many factors modulate NO synthesis and degradation, and therefore, affect endothelial function (EF). Asymmetric dimethyl l-arginine is a product of protein metabolism formed secondarily to methylation of l-arginine(Reference Giles19). Asymmetric dimethyl l-arginine decreases the synthesis of NO by reducing the expression and/or activity of eNOS. Asymmetric dimethyl l-arginine is increased in many pathological conditions such as hypercholesterolaemia, atherosclerosis, hypertension, chronic heart failure, diabetes mellitus and chronic renal failure(Reference North and Sinclair20). Further, uncoupling of eNOS as a result of the oxidation of tetrahydrobiopterin or depletion of l-arginine and the accumulation of endogenous methylarginines may lead to reduced formation of NO, i.e. the eNOS enzyme is converted from NO-producing enzyme to O2-producing enzyme(Reference Schmidt and Alp21). Overproduction of reactive oxygen species (ROS) is the major cause of reduced NO availability in CVD. NO reacts with superoxide anion with high affinity forming the harmful free radical peroxynitrite (ONOO)(Reference Sena, Pereira and Seica5). Lipid peroxyl radicals and oxidised LDL react with endothelial NO before it reaches the vascular smooth muscle cells and, therefore, inhibit NO from dilating blood vessels(Reference Sena, Pereira and Seica5).

The ability of the endothelium to maintain the integrity of the vessel wall can be affected by both the biochemical and pathophysiological states of the rest of the body. For example, chronic smoking deteriorates EF by decreasing NO production and enhancing its degradation via the generation of oxygen-free radicals(Reference Toda and Toda22). Further, hypercholesterolaemia and high homocysteinaemia may reduce the availability of NO secondary to oxidative stress(Reference Sibal, Agarwal and Home23, Reference Dayal and Lentz24).

Endothelial dysfunction and hypertension

ED has been demonstrated both in the resistance and conduit arteries of several animal models of hypertension(Reference Tang and Vanhoutte25). In human subjects, reduced forearm blood flow responses to endothelium-dependent vasodilator agonists, such as acetylcholine and bradykinin(Reference Linder, Kiowski and Buhler26, Reference Panza, Quyyumi and Brush27), and increased vasoconstrictor responses to locally administered NOS inhibitors(Reference Panza, Casino and Kilcoyne28) have been observed in hypertensive patients. The cause of ED associated with hypertension is speculated to be a reduction in NO bioavailability (increased degradation by oxidative stress, reduced production by eNOS inactivation) and abundance of vasoconstrictor agents in the circulation such as angiotensin II and prostaglandins(Reference Tang and Vanhoutte25).

Vascular ageing

The ageing process is characterised by a progressive decline of cellular integrity and function resulting from the structural modification of macromolecules including formation of oxidised lipid species, advanced glycated products, nitrosylated proteins and DNA mutations(Reference Dobrovic and Kristensen29, Reference Vijg and Suh30). The accumulation of modified molecules and their incorporation into cellular components are responsible for the structural and functional deterioration of tissues and organs with time(Reference Ashor, Siervo, Mathers and Mocchegiani31).

Whilst the complexity of the biological mechanisms contributing to the ageing process is still poorly understood, a comprehensive summary of some of these mechanisms has been proposed recently by Lopez-Otin et al. (Reference Lopez-Otin, Blasco and Partridge32). The authors proposed the following set of hallmarks of ageing: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication(Reference Lopez-Otin, Blasco and Partridge32). Many factors contribute to the age-related molecular damage, but it seems likely that much damage is due to three common stressors including oxidative stress/redox changes, inflammation and metabolic stress(Reference Mathers33) (see Fig. 2).

Fig. 2. Factors associated with vascular ageing and atherosclerosis. The triad of oxidative stress, inflammation and endothelial cell senescence contribute to reduced nitric oxide (NO) availability, endothelial dysfunction and the subsequent atherosclerosis.

Ageing and CVD risk

Age-specific mortality rates from heart disease and stroke increase exponentially with age and account for more than 40 % of all deaths worldwide among individuals aged 65–74 years and almost 60 % at age 85 years and older(Reference Ungvari, Kaley and de Cabo34). In the UK, although death rates from CVD have been declining over the past four decades, IHD is ranked as the number one for years of life lost due to premature mortality(Reference Bhatnagar, Wickramasinghe and Williams2). Importantly, key cardiovascular risk factors including lifestyle factors such as smoking, poor diet and lack of physical activity are the major causes of morbidity measured by disability-adjusted life years(Reference Murray, Richards and Newton35).

Ageing is associated with complex structural and functional changes in all tissues including the vascular system, and these changes increase CVD risk independent of other risk factors such as hypertension, diabetes or hypercholesterolaemia(Reference Jani and Rajkumar36). These functional changes include widespread ED, dilation of the central arteries and increased arterial stiffness(Reference Mitchell, Parise and Benjamin37, Reference Taddei, Virdis and Ghiadoni38). Development of strategies to attenuate ageing of the vascular system could make a substantial contribution to lowering CVD risk and improving the quality of life of older people(Reference El Assar, Angulo and Vallejo39).

Factors that impair endothelial function with ageing

Oxidative stress

ROS encompass a large family of oxidant molecules such as superoxide (O2), hydrogen peroxide (H2O2), hydroxyl radical (OH.) and ONOO. The accumulation of ROS and the resulting oxidative modification of cellular macromolecules (lipids, proteins and nucleic acids/DNA) have been suggested to contribute to ageing in all organisms(Reference Puca, Carrizzo and Villa40). Indeed, increased production of free radicals, secondary to mitochondrial dysfunction, causes oxidative damage to cells including vascular cells(Reference Fusco, Colloca and Lo Monaco41). ROS formation can also lead to a propagation of the activity where the effect of a single reactive molecule can be amplified due to a series of chain reactions causing further damage and the loss of cell homeostasis(Reference Zorov, Juhaszova and Sollott42). Beside their damaging effect, ROS are also important secondary messengers in physiological process regulating enzymatic activity, gene expression and have a key role in response to pathogens infections(Reference Bedard and Krause43). For this reason, ROS production is tightly regulated by key antioxidant enzymes such as superoxide dismutase, glutathione peroxidase and catalase that when not jeopardised are able to keep the balance between the production and elimination of these oxidant species(Reference Birben, Sahiner and Sackesen44).

The major sources of ROS in CVD are represented by NADPH(Reference Drummond and Sobey45, Reference Turgeon, Haddad and Dussault46), mitochondrial respiration(Reference Kornfeld, Hwang and Disatnik47), xanthine oxidase(Reference Panth, Paudel and Parajuli48), lipoxygenase and uncoupled NOS(Reference Kuroda, Ago and Matsushima49). The putative mechanism by which the dysregulated enzymatic functions are linked to CVD are thought to be connected to the excessive O2 generation that may act as a NO scavenger causing both a reduction of NO bioavailability in the vascular tissue and the production of the highly reactive ONOO, which in turn can negatively modulate protein functions through nitrosylation of tyrosine residues. Mitochondria also represent an important source of ROS (mtROS) that have been associated with CVD pathogenesis(Reference Dromparis and Michelakis50), and the role of nitrate ${\rm \;} ({\rm NO}_3^ - )\; $ and nitrite ${\rm \;} ({\rm NO}_2^ - )\; $ in the regulation of mitochondrial function and ROS generation is becoming an area of interest in the context of CVD prevention(Reference Shiva and Gladwin51).

Inflammation

Chronic inflammation is a driver of ageing and contributes to the pathology of many age-related diseases including atherosclerosis(Reference Chung, Cesari and Anton52). Observational and experimental studies have demonstrated the importance of inflammation as a determinant of an unhealthy ageing phenotype. For example, the Whitehall II study reported that a high level of IL-6 almost halved the odds of successful ageing after 10 years (OR 0·53) and increased the risk of cardiovascular events and non-cardiovascular mortality(Reference Akbaraly, Hamer and Ferrie53). Growing evidence suggests important cross-talk between oxidative stress, inflammatory processes and the onset of ED prior to atherosclerosis(Reference Ungvari, Kaley and de Cabo34). ROS induce pro-inflammatory changes in the vascular endothelium, described as endothelial activation, which involves secretion of autocrine/paracrine factors, leucocyte-endothelial interaction and the up-regulation of expression of cellular adhesion molecules(Reference Herrera, Mingorance and Rodriguez-Rodriguez54). Oxidative stress activates redox-sensitive transcription factors including the activator protein and NF-κB, increasing the expression of cytokines (TNF-α, IL-1 and IL-6), adhesion molecules (intercellular adhesion molecule and vascular cell adhesion molecule) and pro-inflammatory enzymes (inducible NOS and cyclooxygenase-2)(Reference El Assar, Angulo and Vallejo39).

Ageing is associated with higher circulating concentrations of cytokines, especially TNF-α, IL-1β and IL-6, which mediate the acute phase protein C-reactive protein(Reference El Assar, Angulo and Vallejo39). These factors contribute significantly to the pro-inflammatory microenvironment and facilitate the development of vascular dysfunction(Reference El Assar, Angulo and Vallejo39). Among middle-aged and older adults, the Framingham heart study showed that brachial flow-mediated dilation is inversely related to C-reactive protein, IL-6 and intercellular adhesion molecule inflammatory markers(Reference Vita, Keaney and Larson55). Further, inhibition of NF-κB signalling improved EF significantly in middle-aged and older adults(Reference Pierce, Lesniewski and Lawson56).

Senescence

Cellular senescence is characterised by telomere shortening and permanent loss of mitotic capability, which are associated with morphological and functional changes and impaired cellular homeostasis(Reference Erusalimsky57). Risk factors for atherosclerosis including oxidative stress, inflammation, smoking, diabetes and hypertension have all been associated with accelerated telomere shortening(Reference Fyhrquist and Saijonmaa58). Telomere length in endothelial cells is inversely proportional to patient age(Reference Aviv, Khan and Skurnick59), and this shortening is exacerbated in older aged patients with coronary artery disease(Reference Ogami, Ikura and Ohsawa60). Cross-sectional studies demonstrate that those with increased arterial stiffness, an indicator of vascular ageing, have shorter telomeres(Reference Nawrot, Staessen and Holvoet61). Hypertensive patients have shorter telomeres than their normotensive peers and hypertensives with shorter telomeres are more likely to develop atherosclerosis over 5 years follow-up(Reference El Assar, Angulo and Vallejo39). The afore-mentioned observations suggest that telomere length might be a potential candidate marker for cardiovascular ageing. Vascular endothelial cell senescence in vivo has also been observed(Reference Minamino, Miyauchi and Yoshida62). The development of more senescent endothelial cells has been linked to a shift from an anti-atherosclerotic phenotype (characterised by decreased levels of NO, eNOS activity and shear stress-induced NO production) to a pro-atherosclerotic phenotype (indicated by increased ROS, thromboxane A2 and endothelin-1). These observations implicate endothelial cell senescence in the initiation and progression of atherosclerosis(Reference Minamino and Komuro63). NO increases telomerase activity and promotes mobilisation of endothelial progenitors cells, which have the potential to delay endothelial cell ageing by replacing damaged endothelial cells to maintain physical and functional integrity of the endothelium(Reference Farsetti, Grasselli and Bacchetti64).

Ageing-associated nitric oxide insufficiency

Vascular NO insufficiency in older people is mediated in part by decreased NO production by eNOS(Reference Cau, Carneiro and Tostes65). There is evidence that eNOS activity is reduced with age because of post-translational modification such as acylation, nitrosylation, glycation or phosphorylation(Reference Cau, Carneiro and Tostes65). Additionally, this reduction in eNOS activity might be secondary to the deficiency of cofactors required in the process of NO production (e.g. tetrahydrobiopterin)(Reference Seals, Kaplon and Gioscia-Ryan66). The age-associated increase in arginase activity may compete with eNOS for the critical substrate required in NO production, l-arginine(Reference Seals, Kaplon and Gioscia-Ryan66). Further, excessive O2 production with ageing may contribute to NO insufficiency. The interaction of O2 with NO produces the highly reactive ONOO−(Reference Sindler, Devan and Fleenor67). Due to its ability to restore reduced NO bioavailability, inorganic ${\rm NO}_3^ - $ represents a potential therapeutic strategy to treat age-associated vascular dysfunction(Reference Sindler, Devan and Fleenor67).

The nitrate–nitrite−nitric oxide pathway

Epidemiological studies have consistently shown a protective effect of higher intake of fruit and vegetables and reduced risk of CVD(Reference Zhan, Liu and Cai68). Whilst the exact mechanism/s through which a fruit- and vegetable-rich diet reduces CVD risk remains to be fully elucidated, an increase in NO bioavailability is likely to be important. Eighty-five per cent of the dietary ${\rm NO}_3^ - $ is derived from vegetables and the remaining is mostly from drinking-water. Dietary intake of ${\rm NO}_2^ - $ principally comes from cured meat, to which ${\rm NO}_2^ - $ salts are added to prevent the development of botulinum toxin and to maintain product taste and colour(Reference Hord, Tang and Bryan69). Vegetables can be categorised according to their ${\rm NO}_3^ - $ contents into three categories: (1) high ${\rm NO}_3^ - $ contents: e.g. rocket, spinach, lettuce and beetroot (>1000 mg/kg); (2) medium ${\rm NO}_3^ - $ contents: e.g. turnip, cabbage, green beans, cucumber and carrot (100–1000 mg/kg); and (3) low ${\rm NO}_3^ - $ contents: e.g. onion and tomato (<100 mg/kg)(Reference Lidder and Webb70). The concentration of ${\rm NO}_3^ - $ in drinking-water varies according to the geographical location and regional rules regarding safe levels of ${\rm NO}_3^ - $ in tap or bottled water(Reference Bryan and Ivy71).

It is thought that the beneficial effect of NO disappears in a few seconds as this gasotransmitter is oxidised to ${\rm NO}_2^ - $ and then to ${\rm NO}_3^ - $. ${\rm NO}_3^ - $ is then excreted in urine as a cumulative by-product of NO metabolism and dietary ${\rm NO}_3^ - \; $ intake(Reference Kelm72). Interestingly, in the past two decades, scientists discovered an alternative pathway for NO source other than the classical l-arginine–eNOS–NO pathway(Reference Zweier, Wang and Samouilov73). The other source of NO was found to be ${\rm NO}_2^ - $, which can be converted back to NO by the action of several enzymes and molecules including deoxyhaemoglobin, deoxymyoglobin, xanthine oxidoreductase, protons, polyphenols and ascorbic acid(Reference Lundberg, Weitzberg and Gladwin74). Of note, this pathway is more active and efficient in cases of hypoxia in which the level of both oxygen and NO are low(Reference Zweier, Samouilov and Kuppusamy75).

Dietary ${\rm NO}_3^ - $ is well absorbed in the upper gastrointestinal tract with approximately 100 % bioavailability and plasma concentration of ${\rm NO}_3^ - $ peaking after 1 h(Reference Lidder and Webb70). About 25 % of the circulating pool of ${\rm NO}_3^ - $ is actively taken up from the blood via an anion exchange channel called sialin and secreted by the salivary glands into the saliva(Reference Bailey, Feelisch and Horowitz76). The salivary ${\rm NO}_3^ - $ is reduced to ${\rm NO}_2^ - $ by facultative anaerobic bacteria in the oral cavity, particularly those residing on the dorsal surface of the tongue(Reference Lundberg, Weitzberg and Gladwin74). This ${\rm NO}_2^ - $ and other inorganic ${\rm NO}_3^ - $ travel to the stomach where they are converted to NO with the help of ascorbic acid. In this strong acidic environment of the stomach, ${\rm NO}_2^ - $ is protonated to form nitrous acid (HNO2)(Reference Bailey, Feelisch and Horowitz76). Nitrous acid can spontaneously give rise to the generation of NO through the following sequence of reactions: 2HNO2 → H2O + N2O3 and N2O3 ↔ NO + NO2(Reference Butler and Feelisch77). The liberated NO has been found to be protective for the gastric mucosa, i.e. enhances blood supply(Reference Lundberg, Weitzberg and Gladwin74). Moreover, the remaining NO, ${\rm NO}_2^ - $ and ${\rm NO}_3^ - $ diffuse to the general circulation and contribute to NO pool(Reference Lundberg, Feelisch and Bjorne78).

In the circulation, ${\rm NO}_2^ - $ may function as a source of NO that is activated in hypoxia and acidic conditions to increase blood flow and regulate BP(Reference Kevil, Kolluru and Pattillo79, Reference Zweier, Li and Samouilov80). There are many mechanisms involved in the bioconversion of ${\rm NO}_2^ - $ to NO in the blood(Reference Kim-Shapiro and Gladwin81). The most common is the reaction of deoxyhaemoglobin (HbFe2+) with ${\rm NO}_2^ - $ in acidic environment, which will liberate NO (${\rm NO}_2^ - $ + HbFe2+ + H+ → NO + HbFe3+ + OH)(Reference Kim-Shapiro and Gladwin81). In addition to HbFe2+, there are many enzymes and proteins that enhance the conversion of ${\rm NO}_2^ - $ to NO such as myoglobin, cytochrome C oxidase, eNOS and xanthine oxidoreductases(Reference Kim-Shapiro and Gladwin81).

Therapeutic effects of inorganic nitrate in patients with CVD

${\rm NO}_3^ - $ has been used in the treatment of CVD including angina and digital ischaemia since medieval times(Reference Butler and Feelisch77, Reference Machha and Schechter82). In the past 30 years, since the discovery of the ${\rm NO}_3^ - {\rm -} {\rm NO}_2^ - $−NO pathway and its contribution to the overall NO pool(Reference Kapil, Weitzberg and Lundberg83), there has been a renewal in using ${\rm NO}_3^ - $ and ${\rm NO}_2^ - $ in experiments and in clinical trials focused on the prevention of CVD. Larsen et al. (Reference Larsen, Ekblom and Sahlin84) in a pioneer study demonstrated the beneficial effect of inorganic ${\rm NO}_3^ - $ in BP reduction; the investigators administered 0·1 mmol sodium ${\rm NO}_3^ - $/kg body weight daily to healthy participants (which corresponds to an intake of 100–300 g of ${\rm NO}_3^ - $-rich vegetables daily) and found after 3 d of ${\rm NO}_3^ - $ supplementation, a 4 mmHg reduction in diastolic BP(Reference Larsen, Ekblom and Sahlin84). Administration of the same dose of ${\rm NO}_3^ - $ to a larger group of individuals produced significant reductions in both systolic and diastolic BP(Reference Larsen, Weitzberg and Lundberg85). After the publication of these seminal studies, there has been a growing interest in the protective effects of dietary ${\rm NO}_3^ - $ on cardio-metabolic outcomes. However, the majority of the studies have been conducted in healthy populations and the evidence on the effects of dietary ${\rm NO}_3^ - $ supplementation in patients with CVD is still limited. A summary of the dietary ${\rm NO}_3^ - $ and ${\rm NO}_2^ - $ interventions conducted in patients with CVD is provided in Table 1.

Table 1. Summary of nutritional interventions testing the effects of dietary nitrate supplementations in patients with CVD

R, randomised; CO, cross-over; P, placebo; BJ, beetroot juice; BP, blood pressure; SBP, systolic blood pressure; ABPM, ambulatory blood pressure monitoring; PAR, parallel; DB, double blind; FMD, flow-mediated dilation; DBP, diastolic blood pressure; PWV, pulse wave velocity; CHF, chronic heart failure; PAD, peripheral arterial disease; CKD, chronic kidney disease.

Electronic search conducted on PubMed on 24 January 2018 using the following algorithm: ‘dietary nitrate’ OR beetroot OR beet root OR ‘inorganic nitrate’. Number of articles retrieved by primary search: 1649. One author screened all articles to include studies that investigated effects of dietary nitrate supplementation in patients with CVD.

* Placebo was not defined.

Four weeks of ${\rm NO}_3^ - $ supplementation (9 mg/kg) to older individuals at higher CVD risk significantly lowered systolic BP by 8 mmHg in comparison with placebo(Reference Rammos, Hendgen-Cotta and Sobierajski86). Supplementing beetroot juice (providing a ${\rm NO}_3^ - $ dose of 300–400 mg) to older overweight, but otherwise healthy, participants for 3 weeks lowered daily home-measured systolic BP by 7 mmHg(Reference Jajja, Sutyarjoko and Lara87). However, BP values were found to have returned to pre-intervention values, 1 week after stopping the beetroot supplementation. Kapil et al. (Reference Kapil, Khambata and Robertson88) conducted the largest and longest trial in stage 1 hypertensive patients and found that dietary ${\rm NO}_3^ - $ improved both systolic and diastolic BP (measured by 24 h monitoring, home monitoring and clinic resting) and EF (measured by flow-mediated dilation and arterial stiffness). In contrast, studies in treated hypertensive patients did not show significant improvement of BP with beetroot administration(Reference Bondonno, Liu and Croft89). Moreover, an individual participant meta-analysis (eighty-five participants) showed that beetroot supplementation lowered 24 h ambulatory BP significantly in younger participants only (<65 years)(Reference Siervo, Lara and Jajja90). Two meta-analyses have demonstrated a significant reduction of systolic BP (−4·4 mm Hg)(Reference Siervo, Lara and Ogbonmwan91) and a significant improvement of EF(Reference Lara, Ashor and Oggioni92) with inorganic ${\rm NO}_3^ - $ or beetroot consumption.

The discovery of the contribution of dietary ${\rm NO}_3^ - $ to NO bioavailability has provided a rationale for the use of ${\rm NO}_3^ - $ to reverse ED secondary to NO insufficiency in cardiovascular and metabolic diseases(Reference Machha and Schechter82). Inorganic ${\rm NO}_2^ - $ supplementation reversed ED significantly in a murine model of hypercholesterolaemia(Reference Stokes, Dugas and Tang93). In human subjects, dietary ${\rm NO}_3^ - $ supplementation has been found to improve flow-mediated dilation and arterial stiffness in hypercholesterolaemic patients(Reference Velmurugan, Gan and Rathod94) and reduce TAG concentrations in patients at higher CVD risk(Reference Zand, Lanza and Garg95).

Data from animal studies have also shown promising results regarding the effect of dietary ${\rm NO}_3^ - $ on biomarkers of metabolic diseases. Supplementation of eNOS-deficient mice suffering from metabolic syndrome with inorganic ${\rm NO}_3^ - $ for 10 weeks reduced visceral fat and circulating TAG concentration and reversed the pre-diabetic phenotype(Reference Carlstrom, Larsen and Nystrom96). Further, supplementing diabetic rats with sodium ${\rm NO}_3^ - $ for 2 months produced significant improvements in glucose homeostasis, lipid profile and oxidative stress markers(Reference Khalifi, Rahimipour and Jeddi97). However, ${\rm NO}_3^ - $ supplementation in human subjects showed no evidence of improvement in glucose and insulin homeostasis in diabetics(Reference Cermak, Hansen and Kouw98Reference Shepherd, Gilchrist and Winyard100) or non-diabetic participants(Reference Larsen, Schiffer and Ekblom101). Potassium ${\rm NO}_3^ - $ supplementation did not improve glucose tolerance in young and older obese individuals but reduced oxidative stress during hyperglycaemia in older individuals(Reference Ashor, Chowdhury and Oggioni102).

Inorganic ${\rm NO}_2^ - $ reversed ageing-related arterial stiffness in older mice. In one study, the plasma ${\rm NO}_2^ - $ concentration in older mice was found to be restored to youthful concentrations with inorganic ${\rm NO}_2^ - $ supplementation(Reference Sindler, Fleenor and Calvert103). In healthy human subjects, Bahra et al. (Reference Bahra, Kapil and Pearl104) observed a significant reduction in arterial stiffness 3 h after ${\rm NO}_3^ - $ ingestion. Further, one study found that daily consumption of ${\rm NO}_3^ - $ (900 mg) for 4 weeks reduced pulse wave velocity in older people at increased CVD risk(Reference Rammos, Hendgen-Cotta and Sobierajski86). However, in another study, arterial compliance increased with no change in pulse wave velocity after 220 mg ${\rm NO}_3^ - $ supplementation in twenty-eight healthy participants(Reference Liu, Bondonno and Croft105).

Inorganic ${\rm NO}_3^ - $ administration inhibits platelet aggregation, and therefore, may reduce thrombotic events in both human subjects and experimental animals(Reference Park, Piknova and Huang106, Reference Richardson, Hicks and O'Byrne107). Two studies have found a positive effect of dietary ${\rm NO}_3^ - $ supplementation on platelet aggregation in hypercholesterolaemic patients(Reference Velmurugan, Gan and Rathod94) and platelet-derived extracellular vesicles in coronary artery disease patients on clopidogrel therapy(Reference Burnley-Hall, Abdul and Androshchuk108). The restoration of blood to a tissue after a period of ischaemia is sometimes associated with severe tissue injury due to a high release of free radicals. Animal studies have demonstrated that the prior administration of inorganic ${\rm NO}_3^ - $ reduces the infarct size in a model of ischaemic-reperfusion injury(Reference Lundberg, Carlstrom and Larsen109). Moreover, low-dose sodium ${\rm NO}_2^ - $ attenuated myocardial ischaemia and vascular reperfusion injury in a human experimental study(Reference Ingram, Fraser and Bleasdale110). However, Schwarz et al. (Reference Schwarz, Singh and Parasuraman111) showed that supplementation with sodium ${\rm NO}_3^ - $ marginally improved exercise performance in patients with chronic angina on prescribed medications. Conversely, positive effects of dietary ${\rm NO}_3^ - $ supplementation were found on exercise tolerance and onset of claudication intermittens in eight patients with peripheral arterial disease(Reference Kenjale, Ham and Stabler112). Dietary ${\rm NO}_3^ - $ supplementation appears to have positive effects on exercise performance and oxygen consumption in patients with chronic heart failure(Reference Coggan, Broadstreet and Mahmood113, Reference Zamani, Tan and Soto-Calderon114), whereas the effects on BP, at rest and during exercise, and cardiac haemodynamics are less replicable(Reference Kapil, Khambata and Robertson88Reference Siervo, Lara and Ogbonmwan91, Reference Gilchrist, Winyard and Aizawa99, Reference Oggioni, Jakovljevic and Klonizakis115Reference Ashor, Lara and Siervo117).

Directions for future research

There is currently limited evidence to support the protective effects of inorganic ${\rm NO}_3^ - $ supplementation on cardiovascular and metabolic outcomes in patients at higher CVD risk. Several studies have been conducted in patients with hypertension and chronic heart failure, but the results have been contrasting, whereas for other cardiovascular disorders such as diabetes, CHD or chronic kidney failure, there is simply a paucity of studies. In addition, the evidence is further weakened by the pilot nature of these studies both in terms of short duration (longest trial is 8 weeks)(Reference Kapil, Weitzberg and Lundberg83) of the interventions and small sample size (largest population is seventy patients)(Reference Schwarz, Singh and Parasuraman111). Future research efforts should be therefore directed at the conduction of more robust, confirmatory trials to provide strong and unbiased evidence on the effects of dietary ${\rm NO}_3^ - $ on cardiovascular outcomes. In consideration of the larger number of studies and overall supportive effects of dietary ${\rm NO}_3^ - $ on BP, priority might be given to the design of trials testing the effects of dietary ${\rm NO}_3^ - $ in larger populations of hypertensive patients with and without anti-hypertensive medications to evaluate whether dietary ${\rm NO}_3^ - $ provides additive effects to background pharmacological treatments of BP. These studies may also take into consideration the recruitment of patients with more severe hypertension (stage 2 or 3) and evaluate whether ethnicity could be a modifying factor of the BP response to dietary ${\rm NO}_3^ - $ supplementation.

Conclusions

NO influences several physiological functions involved in the pathogenesis of CVD such as ROS generation, inflammation and platelet aggregation. Increasing inorganic ${\rm NO}_3^ - $ intake, via supplementation of ${\rm NO}_3^ - $ salts or increased high-${\rm NO}_3^ - $ food consumption (i.e. beetroot, green leafy vegetables) could represent a viable and effective strategy for the prevention of age-related chronic cardiovascular and metabolic diseases. The evidence from randomised clinical trials has so far suggested positive effects of systolic BP and EF but the size of the effect appears to be declining in older patients at higher cardiovascular risk. Therefore, until larger and more robust trials are conducted in patients at higher CVD risk, dietary ${\rm NO}_3^ - $ supplementation cannot be recommended as a nutritional or clinical strategy for the primary and secondary prevention of CVD.

Acknowledgements

The authors are very grateful to the reviewer of this manuscript for their helpful comments and swift feedback.

Financial Support

None.

Conflicts of Interest

None.

Authorship

M. S. is the guarantor of this work and takes responsibility for the integrity of the data. M. S., F. S. and A. W. A. wrote the manuscript; M. S. conducted the systematic search and completed the data extraction; O. M. S. and B. C. M contributed to the discussion and reviewed/edited the manuscript.

References

1.Townsend, N, Wickramasinghe, K, Bhatnagar, P et al. (2012) Coronary Heart Disease Statistics 2012 Edition. London: British Heart Foundation.Google Scholar
2.Bhatnagar, P, Wickramasinghe, K, Williams, J et al. (2015) The epidemiology of cardiovascular disease in the UK 2014. Heart (British Cardiac Society) 101, 11821189.Google ScholarPubMed
3.Frayn, KN, Stanner, S (editors) (2005) Cardiovascular Disease: Diet, Nutrition and Emerging Risk Factors: The Report of a British Nutrition Foundation Task Force. Oxford: Blackwell Pub.CrossRefGoogle Scholar
4.Gimbrone, MA Jr & Garcia-Cardena, G (2016) Endothelial cell dysfunction and the pathobiology of atherosclerosis. Circ Res 118, 620636.CrossRefGoogle ScholarPubMed
5.Sena, CM, Pereira, AM & Seica, R (2013) Endothelial dysfunction – a major mediator of diabetic vascular disease. Biochim Biophys Acta 1832, 22162231.CrossRefGoogle Scholar
6.Michiels, C (2003) Endothelial cell functions. J Cell Physiol 196, 430443.CrossRefGoogle ScholarPubMed
7.Rajendran, P, Rengarajan, T, Thangavel, J et al. (2013) The vascular endothelium and human diseases. Int J Biol Sci 9, 10571069.Google Scholar
8.Sumpio, BE, Riley, JT & Dardik, A (2002) Cells in focus: endothelial cell. Int J Biochem Cell Biol 34, 15081512.CrossRefGoogle ScholarPubMed
9.Mensah, GA (2007) Healthy endothelium: the scientific basis for cardiovascular health promotion and chronic disease prevention. Vascul Pharmacol 46, 310314.Google Scholar
10.Vanhoutte, PM, Shimokawa, H, Tang, EH et al. (2009) Endothelial dysfunction and vascular disease. Acta Physiol (Oxf) 196, 193222.CrossRefGoogle ScholarPubMed
11.Siervo, M, Capuano, L & Colantuoni, A (2010) [Physiology and in vivo measurements of nitric oxide in man]. Clin Ter 161, 173183.Google ScholarPubMed
12.Yetik-Anacak, G & Catravas, JD (2006) Nitric oxide and the endothelium: history and impact on cardiovascular disease. Vascul Pharmacol 45, 268276.CrossRefGoogle ScholarPubMed
13.Channon, KM (2006) The endothelium and the pathogenesis of atherosclerosis. Medicine 34, 173177.CrossRefGoogle Scholar
14.Lei, J, Vodovotz, Y, Tzeng, E et al. (2013) Nitric oxide, a protective molecule in the cardiovascular system. Nitric Oxide 35C, 175185.CrossRefGoogle Scholar
15.Forstermann, U & Sessa, WC (2012) Nitric oxide synthases: regulation and function. Eur Heart J 33, 829837, 837a–837d.CrossRefGoogle ScholarPubMed
16.Khazaei, M, Moien-Afshari, F & Laher, I (2008) Vascular endothelial function in health and diseases. Pathophysiology 15, 4967.CrossRefGoogle ScholarPubMed
17.Forstermann, U (2010) Nitric oxide and oxidative stress in vascular disease. Pflugers Arch 459, 923939.Google Scholar
18.Versari, D, Daghini, E, Virdis, A et al. (2009) Endothelial dysfunction as a target for prevention of cardiovascular disease. Diab Care 32, Suppl. 2, S314S321.CrossRefGoogle ScholarPubMed
19.Giles, TD (2006) Aspects of nitric oxide in health and disease: a focus on hypertension and cardiovascular disease. J Clin Hypertens (Greenwich) 8, 216.Google ScholarPubMed
20.North, BJ & Sinclair, DA (2012) The intersection between aging and cardiovascular disease. Circ Res 110, 10971108.CrossRefGoogle ScholarPubMed
21.Schmidt, TS & Alp, NJ (2007) Mechanisms for the role of tetrahydrobiopterin in endothelial function and vascular disease. Clin Sci (Lond) 113, 4763.CrossRefGoogle ScholarPubMed
22.Toda, N & Toda, H (2010) Nitric oxide-mediated blood flow regulation as affected by smoking and nicotine. Eur J Pharmacol 649, 113.CrossRefGoogle ScholarPubMed
23.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.CrossRefGoogle ScholarPubMed
24.Dayal, S & Lentz, SR (2005) ADMA and hyperhomocysteinemia. Vasc Med 10, Suppl. 1, S27S33.Google Scholar
25.Tang, EH & Vanhoutte, PM (2010) Endothelial dysfunction: a strategic target in the treatment of hypertension? Pflugers Arch 459, 9951004.CrossRefGoogle ScholarPubMed
26.Linder, L, Kiowski, W, Buhler, FR et al. (1990) Indirect evidence for release of endothelium-derived relaxing factor in human forearm circulation in vivo. Blunted response in essential hypertension. Circulation 81, 17621767.CrossRefGoogle ScholarPubMed
27.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.CrossRefGoogle ScholarPubMed
28.Panza, JA, Casino, PR, Kilcoyne, CM et al. (1993) Role of endothelium-derived nitric oxide in the abnormal endothelium-dependent vascular relaxation of patients with essential hypertension. Circulation 87, 14681474.CrossRefGoogle ScholarPubMed
29.Dobrovic, A & Kristensen, LS (2009) DNA methylation, epimutations and cancer predisposition. Int J Biochem Cell Biol 41, 3439.CrossRefGoogle ScholarPubMed
30.Vijg, J & Suh, Y (2013) Genome instability and aging. Annu Rev Physiol 75, 645668.CrossRefGoogle ScholarPubMed
31.Ashor, AW, Siervo, M & Mathers, JC (2016) Chapter 43 – vitamin C, antioxidant status, and cardiovascular aging A2 – Malavolta, Marco. In Molecular Basis of Nutrition and Aging, pp. 609619 [Mocchegiani, E, editor]. San Diego: Academic Press.CrossRefGoogle Scholar
32.Lopez-Otin, C, Blasco, MA, Partridge, L et al. (2013) The hallmarks of aging. Cell 153, 11941217.CrossRefGoogle ScholarPubMed
33.Mathers, JC (2015) Impact of nutrition on the ageing process. Br J Nutr 113, Suppl, S18S22.CrossRefGoogle ScholarPubMed
34.Ungvari, Z, Kaley, G, de Cabo, R et al. (2010) Mechanisms of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci 65, 10281041.CrossRefGoogle ScholarPubMed
35.Murray, CJ, Richards, MA, Newton, JN et al. (2013) UK health performance: findings of the global burden of disease study 2010. Lancet 381, 9971020.CrossRefGoogle ScholarPubMed
36.Jani, B & Rajkumar, C (2006) Ageing and vascular ageing. Postgrad Med J 82, 357362.CrossRefGoogle ScholarPubMed
37.Mitchell, GF, Parise, H, Benjamin, EJ et al. (2004) Changes in arterial stiffness and wave reflection with advancing age in healthy men and women: the Framingham heart study. Hypertension 43, 12391245.CrossRefGoogle ScholarPubMed
38.Taddei, S, Virdis, A, Ghiadoni, L et al. (2001) Age-related reduction of NO availability and oxidative stress in humans. Hypertension 38, 274279.CrossRefGoogle ScholarPubMed
39.El Assar, M, Angulo, J, Vallejo, S et al. (2012) Mechanisms involved in the aging-induced vascular dysfunction. Front Physiol 3, 132.CrossRefGoogle ScholarPubMed
40.Puca, AA, Carrizzo, A, Villa, F et al. (2013) Vascular ageing: the role of oxidative stress. Int J Biochem Cell Biol 45, 556559.Google Scholar
41.Fusco, D, Colloca, G, Lo Monaco, MR et al. (2007) Effects of antioxidant supplementation on the aging process. Clin Interv Aging 2, 377387.Google ScholarPubMed
42.Zorov, DB, Juhaszova, M & Sollott, SJ (2014) Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 94, 909950.CrossRefGoogle ScholarPubMed
43.Bedard, K & Krause, KH (2007) The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87, 245313.CrossRefGoogle ScholarPubMed
44.Birben, E, Sahiner, UM, Sackesen, C et al. (2012) Oxidative stress and antioxidant defense. World Allergy Organ J 5, 919.CrossRefGoogle ScholarPubMed
45.Drummond, GR & Sobey, CG (2014) Endothelial NADPH oxidases: which NOX to target in vascular disease? Trends Endocrinol Metab 25, 452463.CrossRefGoogle ScholarPubMed
46.Turgeon, J, Haddad, P, Dussault, S et al. (2012) Protection against vascular aging in Nox2-deficient mice: impact on endothelial progenitor cells and reparative neovascularization. Atherosclerosis 223, 122129.CrossRefGoogle ScholarPubMed
47.Kornfeld, OS, Hwang, S, Disatnik, MH et al. (2015) Mitochondrial reactive oxygen species at the heart of the matter: new therapeutic approaches for cardiovascular diseases. Circ Res 116, 17831799.CrossRefGoogle ScholarPubMed
48.Panth, N, Paudel, KR & Parajuli, K (2016) Reactive oxygen species: a key hallmark of cardiovascular disease. Adv Med 2016, 12.CrossRefGoogle ScholarPubMed
49.Kuroda, J, Ago, T, Matsushima, S et al. (2010) NADPH oxidase 4 (Nox4) is a major source of oxidative stress in the failing heart. Proc Natl Acad Sci USA 107, 1556515570.CrossRefGoogle Scholar
50.Dromparis, P & Michelakis, ED (2013) Mitochondria in vascular health and disease. Annu Rev Physiol 75, 95126.CrossRefGoogle ScholarPubMed
51.Shiva, S & Gladwin, MT (2009) Nitrite mediates cytoprotection after ischemia/reperfusion by modulating mitochondrial function. Basic Res Cardiol 104, 113119.CrossRefGoogle ScholarPubMed
52.Chung, HY, Cesari, M, Anton, S et al. (2009) Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res Rev 8, 1830.CrossRefGoogle ScholarPubMed
53.Akbaraly, TN, Hamer, M, Ferrie, JE et al. (2013) Chronic inflammation as a determinant of future aging phenotypes. CMAJ 185, E763E770.CrossRefGoogle ScholarPubMed
54.Herrera, MD, Mingorance, C, Rodriguez-Rodriguez, R et al. (2010) Endothelial dysfunction and aging: an update. Ageing Res Rev 9, 142152.CrossRefGoogle ScholarPubMed
55.Vita, JA, Keaney, JF Jr, Larson, MG et al. (2004) Brachial artery vasodilator function and systemic inflammation in the Framingham offspring study. Circulation 110, 36043609.CrossRefGoogle ScholarPubMed
56.Pierce, GL, Lesniewski, LA, Lawson, BR et al. (2009) Nuclear factor-KB activation contributes to vascular endothelial dysfunction via oxidative stress in overweight/obese middle-aged and older humans. Circulation 119, 12841292.CrossRefGoogle Scholar
57.Erusalimsky, JD (2009) Vascular endothelial senescence: from mechanisms to pathophysiology. J Appl Physiol (1985) 106, 326332.CrossRefGoogle ScholarPubMed
58.Fyhrquist, F & Saijonmaa, O (2012) Telomere length and cardiovascular aging. Ann Med 44, Suppl. 1, S138S142.CrossRefGoogle ScholarPubMed
59.Aviv, H, Khan, MY, Skurnick, J et al. (2001) Age dependent aneuploidy and telomere length of the human vascular endothelium. Atherosclerosis 159, 281287.CrossRefGoogle ScholarPubMed
60.Ogami, M, Ikura, Y, Ohsawa, M et al. (2004) Telomere shortening in human coronary artery diseases. Arterioscler Thromb Vasc Biol 24, 546550.Google Scholar
61.Nawrot, TS, Staessen, JA, Holvoet, P et al. (2010) Telomere length and its associations with oxidized-LDL, carotid artery distensibility and smoking. Front Biosci (Elite Ed) 2, 11641168.Google ScholarPubMed
62.Minamino, T, Miyauchi, H, Yoshida, T et al. (2002) Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation 105, 15411544.Google Scholar
63.Minamino, T & Komuro, I (2007) Vascular cell senescence: contribution to atherosclerosis. Circ Res 100, 1526.CrossRefGoogle ScholarPubMed
64.Farsetti, A, Grasselli, A, Bacchetti, S et al. (2009) The telomerase tale in vascular aging: regulation by estrogens and nitric oxide signaling. J Appl Physiol (1985) 106, 333337.CrossRefGoogle ScholarPubMed
65.Cau, SB, Carneiro, FS & Tostes, RC (2012) Differential modulation of nitric oxide synthases in aging: therapeutic opportunities. Front Physiol 3, 218.CrossRefGoogle ScholarPubMed
66.Seals, DR, Kaplon, RE, Gioscia-Ryan, RA et al. (2014) You're only as old as your arteries: translational strategies for preserving vascular endothelial function with aging. Physiology (Bethesda, Md) 29, 250264.Google ScholarPubMed
67.Sindler, AL, Devan, AE, Fleenor, BS et al. (2014) Inorganic nitrite supplementation for healthy arterial aging. J Appl Physiol (1985) 116, 463477.CrossRefGoogle ScholarPubMed
68.Zhan, J, Liu, YJ, Cai, LB et al. (2017) Fruit and vegetable consumption and risk of cardiovascular disease: a meta-analysis of prospective cohort studies. Crit Rev Food Sci Nutr 57, 16501663.CrossRefGoogle ScholarPubMed
69.Hord, NG, Tang, Y & Bryan, NS (2009) Food sources of nitrates and nitrites: the physiologic context for potential health benefits1–3. Am J Clin Nutr 90, 110.CrossRefGoogle Scholar
70.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.CrossRefGoogle ScholarPubMed
71.Bryan, NS & Ivy, JL (2015) Inorganic nitrite and nitrate: evidence to support consideration as dietary nutrients. Nutr Res (New York, NY) 35, 643654.CrossRefGoogle ScholarPubMed
72.Kelm, M (1999) Nitric oxide metabolism and breakdown. Biochim Biophys Acta (BBA) – Bioenerg 1411, 273289.CrossRefGoogle ScholarPubMed
73.Zweier, JL, Wang, P, Samouilov, A et al. (1995) Enzyme-independent formation of nitric oxide in biological tissues. Nat Med 1, 804809.Google Scholar
74.Lundberg, JO, Weitzberg, E & Gladwin, MT (2008) The nitrate–nitrite–nitric oxide pathway in physiology and therapeutics. Nat Rev Drug Discov 7, 156.CrossRefGoogle ScholarPubMed
75.Zweier, JL, Samouilov, A & Kuppusamy, P (1999) Non-enzymatic nitric oxide synthesis in biological systems. Biochim Biophys Acta 1411, 250262.Google Scholar
76.Bailey, JC, Feelisch, M, Horowitz, JD et al. (2014) Pharmacology and therapeutic role of inorganic nitrite and nitrate in vasodilatation. Pharmacol Ther 144, 303320.CrossRefGoogle ScholarPubMed
77.Butler, AR & Feelisch, M (2008) Therapeutic uses of inorganic nitrite and nitrate: from the past to the future. Circulation 117, 21512159.CrossRefGoogle ScholarPubMed
78.Lundberg, JO, Feelisch, M, Bjorne, H et al. (2006) Cardioprotective effects of vegetables: is nitrate the answer? Nitric Oxide 15, 359362.CrossRefGoogle ScholarPubMed
79.Kevil, CG, Kolluru, GK, Pattillo, CB et al. (2011) Inorganic nitrite therapy: historical perspective and future directions. Free Radic Biol Med 51, 576593.CrossRefGoogle ScholarPubMed
80.Zweier, JL, Li, H, Samouilov, A et al. (2010) Mechanisms of nitrite reduction to nitric oxide in the heart and vessel wall. Nitric Oxide 22, 8390.Google Scholar
81.Kim-Shapiro, DB & Gladwin, MT (2014) Mechanisms of nitrite bioactivation. Nitric Oxide 38, 5868.CrossRefGoogle ScholarPubMed
82.Machha, A & Schechter, AN (2012) Inorganic nitrate: a major player in the cardiovascular health benefits of vegetables? Nutr Rev 70, 367372.CrossRefGoogle Scholar
83.Kapil, V, Weitzberg, E, Lundberg, JO et al. (2014) Clinical evidence demonstrating the utility of inorganic nitrate in cardiovascular health. Nitric Oxide 38, 4557.Google Scholar
84.Larsen, FJ, Ekblom, B, Sahlin, K et al. (2006) Effects of dietary nitrate on blood pressure in healthy volunteers. N Engl J Med 355, 27922793.CrossRefGoogle ScholarPubMed
85.Larsen, FJ, Weitzberg, E, Lundberg, JO et al. (2007) Effects of dietary nitrate on oxygen cost during exercise. Acta Physiol (Oxf) 191, 5966.CrossRefGoogle ScholarPubMed
86.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
87.Jajja, A, Sutyarjoko, A, Lara, J et al. (2014) Beetroot supplementation lowers daily systolic blood pressure in older, overweight subjects. Nutr Res (New York, NY) 34, 868875.CrossRefGoogle ScholarPubMed
88.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.CrossRefGoogle ScholarPubMed
89.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.CrossRefGoogle ScholarPubMed
90.Siervo, M, Lara, J, Jajja, A et al. (2015) Ageing modifies the effects of beetroot juice supplementation on 24-h blood pressure variability: an individual participant meta-analysis. Nitric Oxide 47, 97105.CrossRefGoogle Scholar
91.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.CrossRefGoogle ScholarPubMed
92.Lara, J, Ashor, AW, Oggioni, C et al. (2016) Effects of inorganic nitrate and beetroot supplementation on endothelial function: a systematic review and meta-analysis. Eur J Nutr 55, 451459.Google Scholar
93.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.CrossRefGoogle ScholarPubMed
94.Velmurugan, S, Gan, JM, Rathod, KS et al. (2016) Dietary nitrate improves vascular function in patients with hypercholesterolemia: a randomized, double-blind, placebo-controlled study. Am J Clin Nutr 103, 2538.CrossRefGoogle ScholarPubMed
95.Zand, J, Lanza, F, Garg, HK et al. (2011) All-natural nitrite and nitrate containing dietary supplement promotes nitric oxide production and reduces triglycerides in humans. Nutr Res (New York, NY) 31, 262269.CrossRefGoogle ScholarPubMed
96.Carlstrom, M, Larsen, FJ, Nystrom, T et al. (2010) Dietary inorganic nitrate reverses features of metabolic syndrome in endothelial nitric oxide synthase-deficient mice. Proc Natl Acad Sci USA 107, 1771617720.CrossRefGoogle ScholarPubMed
97.Khalifi, S, Rahimipour, A, Jeddi, S et al. (2015) Dietary nitrate improves glucose tolerance and lipid profile in an animal model of hyperglycemia. Nitric Oxide 44, 2430.CrossRefGoogle Scholar
98.Cermak, NM, Hansen, D, Kouw, IW et al. (2015) A single dose of sodium nitrate does not improve oral glucose tolerance in patients with type 2 diabetes mellitus. Nutr Res (New York, NY) 35, 674680.CrossRefGoogle Scholar
99.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.CrossRefGoogle ScholarPubMed
100.Shepherd, AI, Gilchrist, M, Winyard, PG et al. (2015) Effects of dietary nitrate supplementation on the oxygen cost of exercise and walking performance in individuals with type 2 diabetes: a randomized, double-blind, placebo-controlled crossover trial. Free Radic Biol Med 86, 200208.CrossRefGoogle ScholarPubMed
101.Larsen, FJ, Schiffer, TA, Ekblom, B et al. (2014) Dietary nitrate reduces resting metabolic rate: a randomized, crossover study in humans. Am J Clin Nutr 99, 843850.Google Scholar
102.Ashor, AW, Chowdhury, S, Oggioni, C et al. (2016) Inorganic nitrate supplementation in young and old obese adults does not affect acute glucose and insulin responses but lowers oxidative stress. J Nutr 146, 22242232.Google Scholar
103.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.CrossRefGoogle ScholarPubMed
104.Bahra, M, Kapil, V, Pearl, V et al. (2012) Inorganic nitrate ingestion improves vascular compliance but does not alter flow-mediated dilatation in healthy volunteers. Nitric Oxide 26, 197202.CrossRefGoogle Scholar
105.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.CrossRefGoogle ScholarPubMed
106.Park, JW, Piknova, B, Huang, PL et al. (2013) Effect of blood nitrite and nitrate levels on murine platelet function. PLoS ONE 8, e55699.CrossRefGoogle ScholarPubMed
107.Richardson, G, Hicks, SL, 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.CrossRefGoogle ScholarPubMed
108.Burnley-Hall, N, Abdul, F, Androshchuk, V et al. (2018) Dietary nitrate supplementation reduces circulating platelet-derived extracellular vesicles in coronary artery disease patients on clopidogrel therapy: a randomised, double-blind, placebo-controlled study. Thromb Haemost 118, 112122.Google ScholarPubMed
109.Lundberg, JO, Carlstrom, M, Larsen, FJ et al. (2011) Roles of dietary inorganic nitrate in cardiovascular health and disease. Cardiovasc Res 89, 525532.CrossRefGoogle ScholarPubMed
110.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
111.Schwarz, K, Singh, S, Parasuraman, SK et al. (2017) Inorganic nitrate in angina study: a randomized double-blind placebo-controlled trial. J Am Heart Assoc 6, e006478.CrossRefGoogle ScholarPubMed
112.Kenjale, AA, Ham, KL, Stabler, T et al. (2011) Dietary nitrate supplementation enhances exercise performance in peripheral arterial disease. J Appl Physiol (1985) 110, 15821591.CrossRefGoogle ScholarPubMed
113.Coggan, AR, Broadstreet, SR, Mahmood, K et al. (2017) Dietary nitrate increases VO2peak and performance but does not alter ventilation or efficiency in patients with heart failure with reduced ejection fraction. J Card Fail 24, 6573.CrossRefGoogle ScholarPubMed
114.Zamani, P, Tan, V, Soto-Calderon, H et al. (2017) Pharmacokinetics and pharmacodynamics of inorganic nitrate in heart failure with preserved ejection fraction. Circ Res 120, 11511161.CrossRefGoogle ScholarPubMed
115.Oggioni, C, Jakovljevic, DG, Klonizakis, M et al. (2018) Dietary nitrate does not modify blood pressure and cardiac output at rest and during exercise in older adults: a randomised cross-over study. Int J Food Sci Nutr 69, 7483.CrossRefGoogle Scholar
116.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.CrossRefGoogle ScholarPubMed
117.Ashor, AW, Lara, J & Siervo, M (2017) Medium-term effects of dietary nitrate supplementation on systolic and diastolic blood pressure in adults: a systematic review and meta-analysis. J Hypertens 35, 13531359.CrossRefGoogle ScholarPubMed
118.Ghosh, SM, Kapil, V, Fuentes-Calvo, I et al. (2013) Enhanced vasodilator activity of nitrite in hypertension: critical role for erythrocytic xanthine oxidoreductase and translational potential. Hypertension 61, 10911102.CrossRefGoogle ScholarPubMed
119.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.CrossRefGoogle ScholarPubMed
120.Kerley, CP, Dolan, E & Cormican, L (2017) Nitrate-rich beetroot juice selectively lowers ambulatory pressures and LDL cholesterol in uncontrolled but not controlled hypertension: a pilot study. Ir J Med Sci 186, 895902.Google Scholar
121.Shaltout, HA, Eggebeen, J, Marsh, AP et al. (2017) Effects of supervised exercise and dietary nitrate in older adults with controlled hypertension and/or heart failure with preserved ejection fraction. Nitric Oxide: Biol Chem 69, 7890.CrossRefGoogle ScholarPubMed
122.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.CrossRefGoogle ScholarPubMed
123.Eggebeen, J, Kim-Shapiro, DB, Haykowsky, M et al. (2016). One week of daily dosing with beetroot juice improves submaximal endurance and blood pressure in older patients with heart failure and preserved ejection fraction. JACC Heart Fail 4, 428437.Google Scholar
124.Chirinos, JA, Londono-Hoyos, F, Zamani, P et al. (2017) Effects of organic and inorganic nitrate on aortic and carotid haemodynamics in heart failure with preserved ejection fraction. Eur J Heart Fail 19, 15071515.Google Scholar
125.Kemmner, S, Lorenz, G, Wobst, J et al. (2017) Dietary nitrate load lowers blood pressure and renal resistive index in patients with chronic kidney disease: a pilot study. Nitric Oxide 64, 715.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Exogenous and endogenous sources of nitric oxide (NO). NO is produced by a family of enzymes known as NO synthases (NOS) which utilise the substrate l-arginine. The ${\rm NO}_3^ - {\rm -} {\rm NO}_2^ - $–NO pathway has been proposed as an alternative pathway for NO generation. ADMA, asymmetric dimethylarginine; BH4, tetrahydrobiopterin; cGMP, cyclic GMP; ONOO, peroxynitrite; sGC, soluble guanylate cyclase.

Figure 1

Fig. 2. Factors associated with vascular ageing and atherosclerosis. The triad of oxidative stress, inflammation and endothelial cell senescence contribute to reduced nitric oxide (NO) availability, endothelial dysfunction and the subsequent atherosclerosis.

Figure 2

Table 1. Summary of nutritional interventions testing the effects of dietary nitrate supplementations in patients with CVD