Skip to main content

From nitrate to NO: potential effects of nitrate-reducing bacteria on systemic health and disease

Abstract

Current research has described improving multisystem disease and organ function through dietary nitrate (DN) supplementation. They have provided some evidence that these floras with nitrate (NO3) reductase are mediators of the underlying mechanism. Symbiotic bacteria with nitrate reductase activity (NRA) are found in the human digestive tract, including the mouth, esophagus and gastrointestinal tract (GT). Nitrate in food can be converted to nitrite under the tongue or in the stomach by these symbiotic bacteria. Then, nitrite is transformed to nitric oxide (NO) by non-enzymatic synthesis. NO is currently recognized as a potent bioactive agent with biological activities, such as vasodilation, regulation of cardiomyocyte function, neurotransmission, suppression of platelet agglutination, and prevention of vascular smooth muscle cell proliferation. NO also can be produced through the conventional l-arginine–NO synthase (l-NOS) pathway, whereas endogenous NO production by l-arginine is inhibited under hypoxia–ischemia or disease conditions. In contrast, exogenous NO3/NO2/NO activity is enhanced and becomes a practical supplemental pathway for NO in the body, playing an essential role in various physiological activities. Moreover, many diseases (such as metabolic or geriatric diseases) are primarily associated with disorders of endogenous NO synthesis, and NO generation from the exogenous NO3/NO2/NO route can partially alleviate the disease progression. The imbalance of NO in the body may be one of the potential mechanisms of disease development. Therefore, the impact of these floras with nitrate reductase on host systemic health through exogenous NO3/NO2/NO pathway production of NO or direct regulation of floras ecological balance is essential (e.g., regulation of body homeostasis, amelioration of diseases, etc.). This review summarizes the bacteria with nitrate reductase in humans, emphasizing the relationship between the metabolic processes of this microflora and host systemic health and disease. The potential effects of nitrate reduction bacteria on human health and disease were also highlighted in disease models from different human systems, including digestive, cardiovascular, endocrine, nervous, respiratory, and urinary systems, providing innovative ideas for future disease diagnosis and treatment based on nitrate reduction bacteria.

Introduction

With the increase in microbiological studies and advances in high-throughput sequencing technology in recent years, several publications on the contribution of microbiota in systemic health and the underlying mechanisms of action have emerged [1,2,3,4,5]. Among them, the bacteria with nitrate reductase are also gaining popularity among researchers [6], which can affect the systemic health and disease of the host by regulating nitrate metabolism [7] (including digestive system [8,9,10,11], cardiovascular system [12,13,14], endocrine system [15,16,17], nervous system [18,19,20], respiratory system [21,22,23] and urinary tract-related diseases [24,25,26]). There are symbiotic bacteria with NRA (such as micropore bacteria, actinomycetes, Escherichia coli, etc.) in the human oral cavity (OC), esophagus and GT, which are closely related to nitrate metabolism [11, 27,28,29,30]. Previous studies have shown that these symbiotic bacteria can reduce nitrate to nitrite, increasing nitrite concentration in plasma and saliva. Nitrite is further reduced to NO and exerts its biological activity after being swallowed into the GT [31]. Recent studies have described that DN supplementation can improve cognitive ability [21, 32, 33], skeletal muscle function [34, 35], cardiovascular function [36, 37] and other physiological functions closely related to human health [7, 38,39,40], and provide some evidence that the bacteria with nitrate reductase are the intermediary of the potential mechanism [41]. One of the mechanisms by which these floras affect human health is their involvement in the production of the signaling molecule NO, which is involved in most physiological activities in the human body (e.g., participation in metabolism and maintenance of cardiovascular homeostasis, dilation of blood vessels, inhibition of atherosclerotic angiopathy, etc.). Therefore, it can be inferred that bacteria with nitrate reductase in the human body are indispensable to human health and disease. The mechanism of its effect on human health deserves further study.

For more than 50 years, DN has been linked to the formation of nitrosamines and the development of cancer [42,43,44,45,46]. As a result, there are strict rules about acceptable levels of nitrate in our diet (e.g., food and drinking water, processed foods and cured meats). It has been observed that the lethal dose of oral nitrate in humans is about 330 mg/kg b.w., and the toxicity of sodium nitrite is about t times that of sodium nitrate. Dietary exposure estimates show that adults consuming 400 g of mixed vegetables does not exceed the daily intake of nitrate, which is within the daily intake range even considering nitrate exposure from other dietary sources. The acceptable daily intake (ADI) of nitrate determined by the former Food Science Council (SCF) was 3.7 mg/kg b.w./day, equivalent to 222 mg of nitrate per day for 60 kg adults and was reconfirmed by the Joint FAO/WHO Expert Committee on Food Additives (JECFA) in 2002. At present, the human intake dose of nitrate and nitrite is within the safe range, far from the toxic dose of nitrate or nitrite [47]. In contrast, in the 1980s–1990s, numerous studies had shown that nitrate could not only biosynthesize in our bodies but also be reduced to NO and other closely related bioactive nitrogen oxides [48,49,50,51,52]. In recent years, it is believed that DN plays a powerful NO-like biological activity in human health [7]. This bioactivity is achieved through a series of reduction reactions. First, nitrite is formed by nitrate-reducing bacteria (NRB) and then transformed into NO and other bioactive nitrogen oxides by non-enzymatic synthesis [53]. With 80% of the body's nitrate coming from dietary leafy green vegetables, studies have shown that naturally occurring nitrate in vegetables is a beneficial active ingredient for systemic health compared to the nitrate added to processed foods and cured meats, and experts generally agree that supplementing nitrate with beetroot juice and other vegetable products may not be harmful [54]. DN is the precursor of signal molecule NO, and DN supplementation can improve NO bioavailability through the NO3/NO2/NO pathway [55]. NO is a mediator with biological activities, such as regulation of cardiomyocyte function [56], modulation of neurotransmission [57, 58], the principle of platelet function [59], anti-inflammation [60, 61], and prevention of vascular smooth muscle cell proliferation [62, 63]. Various studies demonstrated that supplementation of DN can diastole blood vessels [64], lower blood pressure (BP) [65], and improve oxygen consumption efficiency [66, 67]. One of the mechanisms of the effect of DN on human health is the production of NO through the NO3/NO2/NO pathway. Among them, NRB, which plays a vital role in nitrate metabolism, are colonized in the digestive tract (including the oral cavity, esophagus, and GT). Most of the nitrate in the human body is reduced in the OC. Therefore, most reports and studies on the effects of NRB in the human body on general health are biased towards oral floras [68, 69]. Considering the critical role of NO in human health, the body’s overall health may be intertwined with the existence of these bacteria. This review summarizes the relationship between the bacteria with nitrate reductase in human floras, the metabolic process, and the general health of the host. Through several disease models of different systems, the importance of bacteria with nitrate reductase in the human body to whole body health was highlighted to study and discover the methods and strategies to treat related diseases by interfering with the growth and metabolism of these NRB in the future.

Relationship between NRB and their metabolic process and host's general health

NRB and DN

NRB

NRB is a kind of bacteria with NRA. Nitrate is metabolized to nitrite by these symbiotic bacteria and further reduced to NO. This process has become essential to regulate NO homeostasis and signal transduction. There is no nitrate reductase in the human body, but there are symbiotic bacteria with NRA in the human digestive tract (including the OC, esophagus and GT) [11, 29, 31, 37, 69, 70] (Fig. 1). Therefore, NRB play an essential role in the NO3/NO2/NO pathway in intestinal salivary circulation.

Fig. 1
figure 1

Summary of NRB in the human OC, esophagus, and GT

The microbial community of the OC consists of more than 700 prokaryotic taxa and 50–100 billion bacteria (including various NRB) [71,72,73]. NRB was first isolated from experimental rats in 1997 (including Staphylococcus minor, Staphylococcus intermedius, Pasteurella and Streptococcus). In addition, up to 65% of these bacteria are found deep in the posterior tongue [74]. Bacteria with nitrate reductase prefer an anaerobic environment; therefore, they are colonized in the deep fossa of the tongue. The most abundant species include Prevotella melanogaster, Heterotrichia, Haemophilus parainfluenzae, Neisseria flavus, Neisseria fines, and Clostridium nucleatum subsp. nucleatum, Campylobacter, C. labialis, and Prevotella intermedia [75,76,77,78]. Recently, some researchers have shown that the NRB in the human mouth mainly includes thick-walled bacteria (Staphylococcus, Streptococcus, Veronococcus), actinomycetes (Rosella, actinomycetes), Proteus (Neisseria, Haemophilus, campylobacter, Pasteurella), Bacteroides (Proteus) and so on [40]. Regardless of age, the most abundant group of NRB was micropore bacteria, and the level of NRB abundance in the mouth was positively correlated with the amount of DN supplement. The abundance of NRB would increase under a DN load [79]. A comparative analysis of the oral microbiota of vegetarians and omnivores by Hansen and colleagues [80] revealed that a higher proportion of Neisseria and Prevotella was associated with the intake of nitrate-rich vegetables. This suggests that a diet rich in nitrate increases the abundance of NRB and enhances the ability of oral floras to reduce nitrate to nitrite. In addition, the enzymatic activity of NRB varies in a bell-shaped pattern with age (peaking at 30–50 years). It may also vary according to the oral chemical environment (pH, saliva composition, periodontitis), diet type, hygiene practices, and gender [81]. More bacteria with NRA have been discovered in the OC, and the species are more and more abundant, indicating that NRB may play an indispensable role in human health. If NRB is deficient, the NO homeostasis in the body may be broken, and the health of the body will be affected to varying degrees. Tribble et al. [82] have demonstrated through 16S rRNA gene sequencing and analysis that healthy individuals with oral hygiene habits experienced a remarkable reduction in the diversity and abundance of bacteria with nitrate reductase in the mouth after using chlorhexidine mouthwash for a week (twice daily). The decrease in NRB led to a statistically considerable increase in systolic BP, an outcome confirmed in the previous reports [83]. In addition, NRB were also colonized in the oesophagus, which is directly connected to the OC, mainly by Lactobacillus spp., Streptococcus spp., Streptococcus spp., Actinomyces spp., and Prevotella spp. [84]. Some studies have shown that compared with the normal control group, the concentration of NRB in esophageal effusion samples of patients with non-progressive bulimia is significantly higher, especially in the Veronococcus spp., Lactobacillus spp. and Peptococcus spp. [85].

Gastric intestinal floras have been extensively studied in the last decade, especially the relationship between gastrointestinal floras and human health [86]. Bacteria with NRA (e.g., Streptococcus spp., Prevotella spp.) are also present in the GT [86,87,88,89]. Gastrointestinal NRB may have a role like that of the proven oral NRB involved in nitrate metabolism and further acting on the organism’s health. A nitrate-rich diet has recently been shown to increase the abundance of the faucal phylum bacteroides and reduce the quantity of the thick-walled phylum. This phenotype is associated with lower body weight and mass index [28]. Nevertheless, it has also been noted that 16S amplicon sequencing of faces collected from rats fed with high or low nitrate concentrations for 3 weeks found no differences in microbiome [90]. In addition, Rocha et al. [91] found that under antibiotic-induced bacterial symbiosis disorder, inorganic nitrate supplementation for 1 week could prevent the partial loss of rat faucal microflora, but there was no statistical difference. Does DN affect the metabolism of the gastrointestinal microflora? It has long been confirmed that DN enhances the ability of the GT to resist disease and infection, and antibiotic treatment that inhibits oral NRB increases the sensitivity of gastroenteritis. Nitrate in the diet can reverse the imbalance of gastrointestinal microflora caused by antibiotic therapy and enhance the defense of the GT [92, 93]. It is hypothesized that DN intake may modulate gastrointestinal flora metabolism and promote local redox-reduction interactions, thereby exerting beneficial effects on gastrointestinal flora and health status. Dysregulated NO metabolism is associated with ulcerative colitis (UC) [94]. NO can kill bacteria and regulate gastrointestinal mucosal blood flow (MBF) and mucus production, thus protecting the GT. Therefore, one of the mechanisms by which nitrate affects the GT is the role of NO produced by nitrate metabolism, and NRB takes an irreplaceable part in nitrate metabolism. Most DN is reduced to nitrite in the mouth through the enter salivary cycle. However, some nitrate enter the GT directly through the esophagus. There are more kinds of NRB in the OC, and most (about 80%) of the nitrate is reduced to nitrite in the OC through the intestinal saliva circulation, which is eventually catalyzed to generate other NO-related compounds. Most of them are absorbed by the stomach and upper small intestine before reaching the large intestine, and denitrification rarely occurs in the gastrointestinal tract, especially in the lower intestine [95]. Although it has been reported that long-term exposure to nitrate in food and drinking water is associated with an increased risk of colon cancer, its risk is limited to people with low vitamin C intake and high meat intake, suggesting that its risk may be affected by a combination of food and dietary nitrate [27, 96]. The formation of nitroso compounds (NOC) in the gastrointestinal tract is affected by a variety of environmental factors, including various nitrosation reagents, foods, gastric acid, and intestinal microflora [27]. Nitrate and nitrite in the diet come from vegetables and fruits. These foods contain many nitrosation inhibitors that can prevent cancer development [97]. In addition, European Food Safety Authority (EFSA) also pointed out in its 2008 report that epidemiological studies have not shown that nitrate intake from diet or drinking water is associated with an increased risk of cancer [47]. In summary, dietary intake of natural nitrate is almost impossible to denitrify in the lower intestine and is unlikely to produce carcinogenic NOC; the formation of NOC in the gastrointestinal tract is affected by a variety of environmental factors, and eating fruits and vegetables can inhibit the formation of NOC. Therefore, the current research mainly focuses on the effects of nitrate-reducing bacteria in the oral cavity on systemic health. These results further supported the concept that NRB affects human health through the enter salivary circulating NO3/NO2/NO pathway. Recently, studies on the effects of NRB on whole-body health have emerged, and the relationship between NRB and organismal fitness has become a focus of research.

DN

Nitrate is widely present in water, soil, air, and plants. In addition, nitrate in humans is obtained from two primary sources: dietary intake (exogenous nitrate) and endogenous NO oxidation (endogenous nitrate) [98]. The human body takes nitrate from food, which accounts for about 80–85% of the total nitrate intake [99,100,101]. DN is primarily found in vegetables, including celery, radish, beet, etc. In addition, nitrate additives contained in meat are also part of the DN source [102]. Most of the DN is absorbed into the blood after entering the GT, and about 75% of the nitrate is excreted into the body in the form of urine through the kidney. About 25% of the remaining nitrate is enriched into the parotid gland with blood circulation and secreted into the mouth in the form of saliva, of which about 20% of the nitrate is reduced to nitrite under the action of NRB in the mouth [103,104,105]. The remaining nitrate and metabolic nitrite in saliva enter the stomach with swallowing and then enter a new round of the intestinal salivary circulation (80% of nitrite in the human body is produced by the intestinal salivary circulation) (Fig. 2); or NRB can reduce nitrate to nitrite in the acidic environment of the stomach. Then, nitrite is further reduced to biologically active NO and other biologically active nitrogen oxides by acid disproportionation, different hemoglobin (e.g., deoxyhemoglobin, deoxy myoglobin, neurohemoglobin, and cytoglobin), xanthine oxidase, proteins in the mitochondria (cytochrome c oxidase), and nitrite reductase. These nitrogen oxides can be signaled by nitrification, direct NO, and nitridation (Fig. 3) [98, 103, 106]. NO, a metabolite of DN, plays a vital role in protecting the cardiovascular system and gastrointestinal mucosa, regulating cardiomyocyte function and nerve transmission function, and playing a vital role in metabolic diseases. Symbiotic bacteria with nitrate reductase in the human body play an extremely significant role in the NO3/NO2/NO pathway, which reduces nitrate to nitrite. In addition, NO can be produced through the l-NOS path, a process regulated by NOS and its redox state [107]. Among them, NOS includes three subtypes: neuronal NOS (nNOS), inducible NOS (iNOS) and endothelial NOS (eNOS) [41, 108,109,110]. However, in the presence of hypoxia, ischemia, disease or ageing, NOS enzyme activity decreases, and endogenous NO production through the l-NOS pathway is reduced. Currently, the NO3/NO2/NO pathway activity is enhanced to maintain NO homeostasis [111, 112]. NO participates in numerous physiological functions of the human body (such as regulation of cardiomyocyte function, nerve transmission function, platelet function, etc.), so NO homeostasis is closely related to human health (Fig. 4). NO homeostasis is strongly associated with nitrate metabolism. NRB in the OC is essential in the process of nitrate metabolism [113, 114]. In addition, nitrate and nitrite are often used to suppress the development of microorganisms in meat products to prolong their shelf life. Nitrate and nitrite have been considered potential carcinogens for decades and are considered harmful [54, 115]. This stereotype has led to the neglect of the health benefits of nitrate. However, recent studies have failed to prove that nitrate harm humans [116,117,118]. The World Health Organization (WHO) issued guidelines on nitrate and nitrite in drinking water in 2011, arguing that nitrate are not carcinogens based on laboratory animal and epidemiological studies [119]. In addition, many studies have shown that dietary nitrate has many benefits to human health under the action of NRB (especially oral NRB).

Fig. 2
figure 2

Metabolic process of nitrate in human body

Fig. 3
figure 3

Biochemical processes of nitrate metabolism

Fig. 4
figure 4

Bacteria with nitrate reductase ultimately reduce nitrate from dietary sources to NO via the enterosalivary cycle; the body can synthesize NO through the l-NOS pathway, but this pathway is inhibited by inflammatory factors or reactive oxygen species stimulation

In addition, sialin is a membrane protein highly expressed in the parotid gland, a multifunctional anion transporter member of the SLC17 family [120]. Sialin is a salivary gland nitrate transporter with essential physiological effects in modulating NO3/NO2/NO homeostasis in the body (Fig. 5) [31]. Sialin was distributed in serous vesicle cells and lysosomal basal lateral membrane in the salivary gland [121]. The impaired function of the membrane protein sialin may have deleterious effects on human physiological functions [122]. Qin et al. [121] found proof of the physiological correlation of sialin in transporting circulating NO3 into the porcine salivary gland, and inhibition of sialin expression reduced nitrate transport capacity. The discovery that the membrane protein sialin transports circulating nitrate into the parotid gland are critical for future research on the effects of NRB on human disease and health [123, 124].

Fig. 5
figure 5

Process of Sialin transporting nitrate in salivary gland cells

Effect of bacteria with nitrate reductase in the human microflora on systemic health

Many published studies show that DN is beneficial to human health, and NRB and NO play a significant part in the beneficial effects of nitrate. With the increase of age, or in some disease states, the pathway of classical enzymatic reaction to produce NO will be maladjusted. The NO3/NO2/NO path can compensate for the dysregulation of the classical way. Still, the NO3/NO2/NO pathway is disrupted when nitrate intake is inadequate, when antibacterial mouthwash/antibiotics are used, and when antacid therapy is used. These two pathways can compensate for each other, but when both systems fail, the NO-based signal will be suppressed entirely, eventually leading to disease. More studies are supporting the benefits of NRB on human whole-body health. From this review, we have discussed the influence of NRB on human systemic health through different disease models (Fig. 6).

Fig. 6
figure 6

Potential role of NRB in human systemic health and disease through the NO3/NO2/NO pathway

Digestive system

Oral-related diseases

There is a lack of effective treatment for salivary gland dysfunction caused by radiotherapy for head and neck tumors (HNT). Feng et al. [125] discovered that nitrate supplementation in the diet effectively prevented radiation-induced parotid hypofunction in miniature pigs. Nitrate is a dose-dependent way to maintain the function of irradiated parotid gland tissue, thus preventing radiation-induced damage to the parotid gland. Nitrate addition to diet was shown to increase the expression of sialin, a nitrate transporter, resulting in a positive feedback loop between nitrate and sialin. It can also stimulate the proliferation of human parotid cells (hPGCs) through the EGFR–AKT–MAPK signaling pathway. Furthermore, it is noteworthy that radiation treatment induces a hypoxic and acidic environment in the salivary gland. Under these circumstances, NO has been synthesized through the NO3/NO2/NO exogenous pathway, in which NRB is essential in reducing nitrate. Adding nitrate to the diet leads to the production of NO. It decreases hypoxia by inducing a long-term increase in blood flow and increasing glandular micro vascularization, facilitating salivary production by glandular vesicle cells [126]. Furthermore, nitrate-mediated NO generation can enhance the expression of sialin and upregulate the EGFR–AKT–MAPK signaling pathway. This signaling pathway can promote cell proliferation, maintaining cell survival and preventing apoptosis [127, 128]. Therefore, the NO produced by DN through the NO3/NO2/NO pathway mediated by NRB may be a mechanism for avoiding xerostomia caused by radiotherapy. Recent studies have shown that radiotherapy and chemotherapy in patients with oral cancer (OCC) and oropharyngeal carcinoma (OPC) lead to oral ecological disorders and specific deletion of bacteria regulating the NO3/NO2/NO pathway in the intestinal salivary circulation [9]. Among them, Neisseria, Haemophilus, Porphyromonas, Clostridium and Cilium disappeared, while Lactobacillus increased, indicating that radiotherapy may lead to NO homeostasis imbalance. This also correlates with the significantly down-regulated oral metabolomic profile of NO-related precursors, regulators, or catalysts (e.g., aspartate, phenylalanine, l-ornithine, l-proline, xanthine, tyrosine, and glycine) in saliva samples from patients after radiotherapy. The decrease in the abundance of these NRB may lead to complications related to NO deficiency, such as xerostomia and local inflammation. Therefore, supplementation of NRB and DN in patients with head and neck radiotherapy and chemotherapy may be a new, safe, and effective method for treating radiation-induced xerostomia. We suggest that future studies explore the likelihood of oral microflora transplantation (OMT) and dietary interventions to reintroduce beneficial microorganisms in the OC of patients after head and neck radiation therapy to improve quality of life.

Esophagus and GT-related diseases

Considerable evidence suggests that the microbiota has a crucial role in esophageal cancer [129, 130]. Li and his team [11] obtained matched pairs of saliva and esophageal brush samples from 276 subjects who underwent upper gastrointestinal endoscopy, using 16S rRNA analysis and next-generation sequencing technologies to study esophageal microbes. They found that compared with the normal group, the microbial diversity of saliva and cell brush samples decreased with the progression of the disease, and the nitrate reductase function of salivary floras in patients with esophageal squamous cell carcinoma (ESCC) decreased. It was also found that the part of nitrate reductase in matched esophageal brush samples from the same patient had the same change. The microflora in saliva and esophageal cancer cell brush samples are different, but the function of nitrate reductase is weakened, indicating that the bacteria with nitrate reductase can become a sensitive and specific clinical diagnostic marker for ESCC. In addition, it is suggested that NRB play a significant role in regulating the balance of esophageal microorganisms in patients with ESCC, which may be one of the critical mechanisms of the role of esophageal microorganisms in oesophagal cancer.

In addition to the esophagus, NRB can also be found in the stomach and may be closely associated with human health [86, 89, 131, 132]. A research team has, for the first time, published a report on the composition of gastric microflora during the development of gastric cancer. They found NRB (including Neisseria, Clostridium, and staphylococci) in the stomach and believed these bacteria were potential candidates associated with gastric cancer. However, the actual role of these bacteria in the development of gastric cancer has not been evaluated [86]. In addition, there is preliminary evidence that DN can protect the stomach by inducing gastric mucosal vasodilation, improving gastric mucosal blood flow, and promoting gastric mucus production. Recent studies suggest that these observations may be caused by NRB reducing nitrate through the NO3/NO2/NO pathway to produce NO in the human stomach [93, 133, 134]. Therefore, the specific effect of NRB on the stomach is worthy of our in-depth study.

DN can alter the gut microbial ecology system [75, 135, 136]. Hu and his colleagues [137] have demonstrated that DN supplements prevent colitis from upregulating carcinogenic pathways implicated in colorectal cancer development (e.g., activation of p53), indicating that nitrate may regulate inflammation by reshaping gut microbes. Previous studies have found reduced bacterial diversity and increased bacterial instability in patients with IBD compared to healthy individuals [138,139,140], confirming the changes in the microbiota of patients with IBD. However, the pathogenesis of IBD is still not fully understood, and a primary cause may be linked to the unbalance of intestinal bacteria. To investigate the mechanistic details leading to the dysbiosis of the intestinal bacteria in IBD, investigators assessed the role of nitrate in a mouse model of dextran sulfate sodium salt (DSS)-induced colitis [137]. The results revealed that nitrate addition to the diet maintained colonic consistency, improved colonic length, increased microvascular density, modulated serum Th cells, and decreased apoptosis rate in colonic epithelial cells, indicating that nitrate supplementation could reduce experimental colitis in mice [28, 137]. This suggests that nitrate can significantly ameliorate DSS-induced colitis by reducing the inflammatory response, decreasing apoptosis of colitis cells, improving intestinal blood flow, and activating the NO3/NO2/NO pathway to regulate intestinal floras [28, 141, 142]. Therefore, one of the pathogenic mechanisms of IBD may be that l-arginine-dependent endogenous NO synthesis is inhibited under inflammation, while exogenous dietary nitrate activates the NO3/NO2/NO pathway under the action of NRB to restore the balance of intestinal floras.

Cardiovascular system

Hypertension and pulmonary hypertension

Although we have improved the way we diagnose and image cardiovascular diseases (CD) at an early stage, with many new drugs approved for the treatment of CVD (such as hypertension, etc.), hypertension still puts us at risk of heart disease and stroke, which is the leading cause of death around the world [143]. Although the pathophysiological aspects of hypertension have been intensively studied in these decades, its incidence and prevalence have not decreased significantly. It is assessed that even with active drug therapy for hypertension, only approximately 50% of patients have their BP under control [25, 143]. At present, looking for new targets to prevent and treat hypertension is still the direction of our efforts. In 2006, the first study showed that inorganic nitrate reduced diastolic BP (3.7 mmHg) in healthy subjects 3 days after taking sodium nitrate (0.1 mmol/kg) [144]. Nitrate was later found to affect systolic blood pressure in a similar group of subjects [145,146,147]. A subsequent study validated previous claims that healthy volunteers reduced systolic and diastolic BP by 10.4 mmHg and 8 mmHg, respectively, after taking 500 ml of beetroot juice [147]. Interest in the effects of nitrate on cardiovascular function has been raised by these studies showing the potential of DN to lower BP. In recent years, it has been indicated that imbalances in the oral microbial community can negatively impact cardiovascular and metabolic health. Nitrate can produce NO in the human gastrointestinal tract through the NO3/NO2/NO pathway mediated by NRB, which may be one of the essential mechanisms of nitrate lowering BP in patients [12,13,14, 41, 148]. Recently, a review has elucidated the role of NO in the fight against CD, including hypertension, among others [149]. Evidence has shown that hypertension can be offset by drugs that improve NO signaling or restore NO bioavailability (for example, angiotensin-converting enzyme (ACE) inhibitors promote elevated levels of bradykinin, which activates the bradykinin B2 receptor in endothelial cells and eNOS, which increases NO production) [108]. Carlström et al. [150] further described the effects of dietary nitrate on various organ systems through the NO3/NO2/NO pathway under the action of NRB and discussed the potential mechanism of dietary inorganic nitrate reducing BP.

Small reductions in systolic BP in groups of people can remarkably decrease the risk of hypertension and mortality from CD (e.g., stroke). Therefore, exploring the effect of NRB on systemic blood pressure is significant. Evidence from studies has revealed that untreated hypercholesterolemic patients who consumed beetroot juice rich in nitrate for 6 weeks increased the abundance of NRB in the microflora of saliva while improving brachial flow-mediated dilation and reducing platelet monocyte aggregation [151]. More recently, Vanhatalo and his colleagues [14] showed in a clinical study that supplementation with nitrate-rich beet juice for 10 days similarly increased the abundance of NRB (Rhodobacter spp. and Neisseria spp.) in the salivary microbiota in both older (70–79 years) and younger (18–22 years) healthy subjects. They also found that acute (10 days) nitrate supplementation only reduced BP in healthy older adults, not in healthy younger adults. The current research results show that the oral microbial community is plastic and changes with the change of dietary inorganic nitrate intake, and the diet-induced changes in the oral microbial community are related to NO homeostasis and vascular health index. It has been shown that antimicrobial mouthwash reduces the number of NRB in the OC and can impair the antihypertensive and vasoprotective effects of l-arginine [152]. To explore the potential relationship between NRB and systemic BP, Petersson et al. [153] gave rats antibacterial mouthwash twice a day and supplemented nitrate drinking water simultaneously. They found that the simultaneous use of mouthwash significantly reduced the number of oral NRB compared with rats fed only nitrate drinking water, offsetting the drop in systemic BP caused by nitrate supplementation. More recently, data from various sets of animal models and humans have also revealed that antibacterial mouthwash can decrease the concentration of nitrite in the mouth and plasma while increasing BP by 2–3 mmHg [152,153,154,155,156]. Senkus et al. [83] performed an in-depth dissection of eight published studies between 2009 and 2016 (including five human crossover studies and three animal control studies). The data indicated that applying antibacterial mouthwash negatively affects saliva and plasma nitrate/nitrite concentrations, accompanied by increased BP. A review has elaborated that the disturbance of NO homeostasis by antibacterial mouthwash may cause an increased risk of cardiovascular mortality [53]. These findings demonstrated that DN supplementation increased the abundance of NRB, and that salivary nitrate can modulate cardiovascular function through the bioactivation of oral commensal bacteria. In contrast, overuse of antibacterial mouthwash may diminish the biological activity of DN. Goh et al. [78] pointed out that oral NRB can benefit the modulation of BP. Exploiting the NRA of specific symbiotic bacteria to make the NO3/NO2/NO pathway a potential system for maintaining NO bioavailability requires far-reaching and truly transformative research. Future longitudinal studies will strengthen the evaluation of the relationship between NRB exposure and hypertension, predict biomarkers of cardiometabolic risk and clinical disease progression, achieve early prevention of disease risk, and will provide information for the evolution of further intervention research manipulating oral NRB.

To explore the deeper mechanisms of the NO3/NO2/NO pathway to lower BP, Guimarães et al. [18] administered DN supplementation to angiotensin II (Ang II)-induced hypertensive rats and found that inorganic nitrate treatment not only reduced oxidative stress by promoting the NO3/NO2/NO pathway but also resulted in the suppression of sympathetic nerve activity (SNA) in this animal model or even normalized it, ultimately reducing BP in this animal model. They found that enhanced SNA boosts the progression of the disease, including hypertension, and raises the risk of adverse complications. A novel strategy to inhibit SNA may be of immense value in preventing or treating hypertension. Promoting the NO3/NO2/NO pathway through DN supplementation is a new nutritional and pharmacological approach to SNA inhibition in which NRB play an irreplaceable key role. However, this trial did not clarify that acute and chronic supplementation with inorganic nitrate can suppress SNA in hypertensive patients.

Recent studies have shown that S-nitrosation is impaired in hypertension, and increasing this modification may be an effective antihypertensive strategy. It is worth noting that NO plays a vital role in activating guanylate cyclase (GC), especially through the S-nitrosation of proteins. Nitrosylation affects G protein-coupled receptor (GPCR)-mediated signaling and can alter the affinity of angiotensin II for angiotensin II type 1 receptor and β-blocker transport [108]. These evidence suggests that this NO-mediated retro-translational modification may be closely related to vascular regulation and that NRB is a crucial contributor to NO production. Thus, NRB may regulate protein S-nitrosation through the NO3/NO2/NO pathway and thus effectively counteract hypertension.

In addition, it has been shown that when brassica vegetables rich in thioglucosides are consumed dietary (e.g., cauliflower and broccoli), mustard enzymes can convert thioglucosides to thiocyanate and increase serum thiocyanate levels [108]. Eating vegetables rich in nitrate can lower BP, but this effect will be eliminated when eating vegetables rich in nitrate and thiocyanate simultaneously. To investigate how this thiocyanate affects the antihypertensive effect of nitrate, Dewhurst-Trigg and his colleagues [157] found that consumption of thiocyanate-rich vegetables did not impact salivary nitrate intake but may inhibit the activity and metabolism of NRB, thereby affecting the capacity for nitrate conversion to nitrite in the OC. In addition, smoking increases the cycle level of thiocyanate, because cyanide in cigarette smoke is easily converted to thiocyanate through a desulphurization reaction catalysed by thiosulfate and 3-mercaptopyruvate. The study found that compared with non-smokers, smokers increased and decreased the concentration of nitrate in plasma and saliva while higher levels of thiocyanate in plasma and saliva. These phenomena are related to eliminating antihypertensive effects after smoking dietary nitrate in smokers. Both dietary intakes of thiocyanate-rich vegetables and smoking impair nitrate metabolism and the antihypertensive effect on this anion. It was recently demonstrated that smokers significantly impaired the conversion from nitrate to nitrite in the OC, suggesting that altered activity of oral NRB may be an essential pathophysiological mechanism.

Chronic gestational hypertension is significantly associated with poor outcomes in pregnancy, raising the risk of preeclampsia [158, 159], fetal growth restriction [160], and preterm birth. Dietary supplementation with nitrate has been shown to restore NO balance in the body, improve endothelial dysfunction, and lower BP. A clinical study has demonstrated a potential effect of DN (beet juice) supplementation on BP in hypertensive pregnant women [161]. The NO3/NO2/NO pathway process primarily involves the activity of bacterial nitrate reductase. It is hypothesized that variations in the effects of nitrate supplementation are associated with differences in individual microbiomes. Further studies must confirm the relationship between chronic gestational hypertension and NRB. Future trials should explore and assess the beneficial influence of probiotic supplementation on the prognosis of pregnant women with hypertension. Probiotic interventions and supplementation with DN may suggest a safe and effective way to treat hypertensive disorders of pregnancy.

Moreover, pulmonary arterial hypertension (PAH) is a vascular disease in which mechanical obstruction increases in mean pulmonary arterial pressure [162]. Endothelial dysfunction, inflammatory and immune responses, and abnormal extracellular matrix function play a key role in PAH. PAH has vascular endothelial dysfunction and low NO bioavailability. Studies have shown that low-dose inhalation of NO (lasting for 4 h at 20 ppm concentration and then for 20 h at 6 ppm concentration) can improve oxygenation in PAH newborns without affecting the whole body and reduce systolic BP in nine PAH newborns [163, 164]. Subsequently, Roberts and his team [165] confirmed that inhaled NO increased systemic oxygen levels through a multicenter randomized controlled study of full-term and near-term infants with PAH. In addition to in vitro NO supplementation, restoration of NO bioavailability through dietary supplementation with NRB may be a dietary therapy for PAH.

As discussed earlier, the NRB-mediated NO3/NO2/NO pathway performs a vital role in preventing the progression of CD, for example, by reducing arterial stiffness, improving endothelial function, and reducing the risk of CD [12]. In addition, a recent study by feeding nitrate-added drinking water to mice with chronic ischemia in the hind limbs for 2 weeks found increased mobilization of CD34(+)/Flk-1(+) cells and migration of bone-marrow-derived (BMD)CD31(+)/CD45(−) cells to the site of ischemia, correlating with enhanced revascularization [166, 167]. These BMD endothelial progenitor cells (EPC) are activated in response to vascular stress and injury and are engaged in angiogenesis and restoration. The regenerative effect of DN can be abrogated using an antibacterial mouthwash, suggesting the importance of NRB in the NO3/NO2/NO pathway. Subsequently, the same acute mobilization of EPC was observed in the same studies after nitrate supplementation in humans [167,168,169]. These findings reveal that NRB may protect vascular function by mediating the NO3/NO2/NO pathway to affect the release and migration of various BMD cells.

However, due to the short duration and small sample size of the completed studies, the proof of the long-term effects of DN on BP in patients at increased cardiovascular risk is still not conclusive at this point. Therefore, future researchers need to use more accurate methods to evaluate the benefits and potential mechanisms of NRB in CD, such as hypertension, in clinical trials with a larger scale (> 300 participants) and longer DN supplementation time (> 12 months). The NO3/NO2/NO pathway determines NO homeostasis in cardiovascular health and disease. NRB plays a vital part in this pathway, providing a new target for the treatment of hypertension. We can modify CD patients’ prognosis through DN and probiotic supplementation. The findings of Goh et al. [78] support the role of oral NRB having a beneficial effect on BP modulation and insulin resistance (IR), which is one of the first studies to directly test the priori established hypothesis that oral NRB influences cardiometabolic outcomes. Future longitudinal studies will strengthen the estimation of the potential of NRB as biomarkers for predicting cardiometabolic risk and clinical disease progression and provide information for the evolution of future intervention studies that may manipulate oral nitrate-reducing capacity.

Heart disease

According to the most recent statistics on factors associated with heart disease reported annually by the American Heart Organs Association and the National Institutes of Health, diet is one-factor affecting heart health [170]. Nitrate is a potential dietary supplement for lowering BP. NRB acts as a key “driver” for lowering BP in patients through the NO3/NO2/NO pathway, with few studies investigating its effects on cardiac function. Under normal conditions, the decrease in BP due to vasodilation usually causes an increase in pressure reflex activity and heart rate, but with DN supplementation, only a drop in BP and no change in heart rate was observed [171, 172]. There are two explanations for this result: first, the effect of NO3/NO2/NO pathway mediated by NRB on BP is not enough to cause an increase in baroreflex activity and heart rate; second, NRB may have a direct or indirect inhibitory effect on baroreflex. Beetroot juice can be used as a natural nitrate supplement to increase the concentration of nitrite in plasma under the action of NRB [173]. In isolated and perfused Langendorff rat heart models, it has been shown that the increase of plasma nitrite concentration has a cGMP/PKG-dependent negative inotropic and muscular tone effect, characterized by a decrease in left ventricular relaxation and BP. This suggests that NRB may play an essential role in cardiac negative myodynamia and muscular tension.

In addition, studies have shown that the mechanisms of coronary dysfunction mainly involve inflammation, extensive and microvascular spasms, abnormal clotting and endothelial dysfunction [174, 175]. Endothelial dysfunction is a primary causative mechanism in patients with coronary heart disease risk factors. Endothelial dysfunction breaks down the endothelial l-NOS–NO pathway, contributing to a diminution in NO production, which leads to a decrease in endothelium-dependent diastolic effects and an increased risk of developing coronary artery disease, suggesting a critical function of NO in the regulation of endothelial function. Kanno et al. [176] similarly indicated that NO is vital in regulating cardiac function. They noted that in a healthy heart, NO produced by the endogenous NOS pathway reduces basal muscle force and plays a crucial role in safeguarding the myocardium from systolic/diastolic dysfunction, remodeling, and arrhythmias in the failing heart. Exogenous nitrate can produce NO through the NO3/NO2/NO pathway under the action of NRB, which is an independent alternative to the imbalance of endogenous pathways in a pathological state. Mechanisms by which NO acts include: (1) activation of soluble guanylate cyclase to produce cyclic guanosine monophosphate (cGMP) and relax vascular smooth muscle [177]; (2) increasing platelet cGMP levels to enhance anti-platelet agglutination and antithrombotic effects [178]; (3) decreases venous return and left ventricular end-diastolic pressure to diminish myocardial oxygen demand while increasing blood flow to the subendocardial; and (4) dilates coronary stenosis and increases collateral blood flow to directly increase myocardial oxygen supply [149, 179]. It is indicated that nitrate induces vasodilation of vascular smooth muscle through the NO3/NO2/NO pathway under the action of NRB, decreasing cardiac load and thus reducing left ventricular wall tension and end-diastolic pressure. It is a vital process in patients with stable ischemic heart disease (SIHD) as it not only reduces the load on the heart but also decreases the oxygen demand of the myocardium [180]. One study showed that nitrate could reduce myocardial ischemia and ischemic pain and increase exercise tolerance in stable and unstable angina pectoris. After acute myocardial infarction, nitrate can reduce ventricular dilatation, reducing pulmonary congestion and mitral regurgitation [181]. Besides, nitrate can also be used prophylactically before exercise, as they may improve exercise tolerance and avoid exercise-induced angina attacks [182, 183]. There is no doubt that nitrate have tremendous therapeutic potential in common cardiac diseases, such as angina pectoris. Currently, nitrate distributes coronary blood flow to ischemic areas through the NO3/NO2/NO pathway, thus improving local tissue hypoxia caused by illness as one of the many mechanisms to increase coronary perfusion. NRB, as the critical bacteria of nitrate metabolism to produce NO, few studies have clarified the potential mechanism of NRB in the recovery of coronary artery blood flow and reperfusion. The non-enzymatic and non-oxygen-dependent entero-salivary pathway is the primary mode of NO production, but an antibacterial mouthwash influences the effects produced by this pathway. A patient with angina pectoris reported in a case that had relief symptoms after discontinuation of antibacterial antiseptic mouthwash [184]. Therefore, we estimate that the NO3/NO2/NO path depends on the action of NRB to restore NO homeostasis and thus improve coronary perfusion.

Heart failure with reduced ejection fraction (HFrEF) is a fatal and disabling disease that is a significant public health concern. This disease is thought to be partially associated with the poor bioavailability of NO [181]. DN performs an essential part in treating conditions, including HFrEF, as a new source of human NO through intestinal salivary circulation and under the action of NRB. Some investigators have directly studied myocardial tissue from patients with HfrEF and found reduced cGMP levels, protein kinase G activity, and nitrite concentrations [185, 186]. Recent studies have shown that the impairment of coronary microvascular structure and function in HFpEF is primarily associated with decreased NO–cGMP bioavailability. When the organism is in a disease state, it leads to local microenvironmental hypoxia, which reduces endogenous NO production. We need new NO source pathways to restore NO homeostasis in the body [187,188,189]. The NO3/NO2/NO path is another strategy to enhance the NO–cGMP signaling pathway. Thus, NRB may serve as a suitable means to improve the efficiency of cardiac and peripheral NO signaling production at the early stages of the disease, thereby reducing the risk of disease progression to advanced settings. In addition, it has been shown that nitrite improves skeletal muscle mitochondrial efficiency, insulin sensitivity, and glucose uptake [190,191,192], so acute administration of inorganic nitrate treatment may enhance muscle strength in patients with HfrEF [192]. NRB is expected to be a probiotic to improve exercise capacity in patients with chronic HFpEF.

Endocrine system

II diabetes mellitus and metabolic syndrome

Type 2 diabetes (T2D) and metabolic syndrome (MS) are chronic non-communicable diseases with high prevalence and rapid growth rates worldwide [193, 194]. MS is like T2D in clinical signs and is also predominantly insulin resistant. Patients with MS may also have T2D, a major manifestation of disorders of the body’s metabolism of protein, fat, carbohydrate, and other substances [195]. A WHO report predicts that by 2040, the number of adults with T2D will exceed 650 million worldwide [15]. These diseases require long-term control and treatment, are costly to treat, and are correlated with an increased risk of death. An unhealthy diet (e.g., high in fat, sugar, etc.) is important in the increased incidence of T2D and MS, which are often associated with oxidative stress, impaired NO signaling, and CD [196, 197]. Oral microflora disorder is also considered to be related to the occurrence and development of T2D and MS [78, 198,199,200]. Among them, NRB, which have a significant part in the metabolic homeostasis of oral microorganisms, are of interest, because they are involved in a major part of NO production (NO3/NO2/NO pathway) [201], supplying an alternative systemic source of NO. NO is considered to be a signal molecule closely related to carbohydrate metabolism [202], so the imbalance of NRB in oral microorganisms is a risk factor for the impairment of carbohydrate metabolism and the occurrence and development of T2D and MS [78]. Under hyperglycemia, the impairment of the l-NOS–NO pathway leads to the decrease of NO synthesis and bioavailability. Some studies have shown that increased blood glucose and advanced glycation end products (AGEs) can down-regulate the expression/activity [203] of eNOS through inflammation [204], and redox pathway [205].T2D can lead to elevated cytokine levels (e.g., TNF-α) and further downregulate eNOS expression [206]. This all leads to reduced endogenous NO synthesis and reduced bioavailability. In contrast, DN supplementation can compensate for the disturbance of the impaired enzyme-dependent pathway by promoting the NRB-dependent dietary NO3/NO2/NO pathway. Huang et al. [207] showed that mice deficient in eNOS evolve an MS-like phenotype with age. Carlstrom et al. [208] further showed that compared to controls, long-term dietary supplementation of nitrate decreased visceral fat accumulation, body weight gain, circulating triglycerides, and glycated hemoglobin (HbA1c) levels in mice and reversed the MS profile and prodromal diabetic phenotype in eNOS-deficient mice compared to controls. To investigate the relationship between T2D and MS and nitrate metabolism, Ohtake and his colleagues [209] found in a group of postmenopausal MS mouse models evoked by ovariectomy and a high-fat diet that these mice had lower circulating nitrate and nitrite levels compared to the corresponding controls and developed obesity, visceral adipocyte hypertrophy, and insulin resistance (IR), which would be avoided by nitrite treatment. Nyström and others [210] further pointed out that nitrite has dual stimulating effects on islet function, including indirect enhancement (increasing islet blood flow and redistribution through microcirculation) and direct insulin-promoting effect on β-cells. The insulin-promoting effect of nitrite is cGMP-dependent and involves the formation of active nitrogen and oxygen. In contrast, most nitrite in humans are produced by the reduction of DN by NRB. In addition, Khalifi et al. [211] investigated the effect of DN on glucose tolerance and lipids in a rat model of T2D induced by streptozotocin and nicotinamide injections. The results revealed that plasma nitrate and nitrite content decreased in these mice but recovered after nitrate supplementation, reducing hyperglycemia, and increasing blood lipid and glucose tolerance. Gheibi et al. [212] similarly found that obese T2D rats showed improved glucose tolerance, IR, and dyslipidemia after 2 months of DN supplementation compared to controls. These beneficial effects were correlated with increased GLUT4 expression in insulin-sensitive tissues and reduced gluconeogenesis, inflammation, and oxidative stress. In addition, Li et al. [213] found that DN supplementation attenuated the elevation of circulating triglycerides, total cholesterol, low-density lipoprotein (LDL) cholesterol, and high-density lipoprotein (HDL) cholesterol induced by dietary interventions (high-fat and high-fructose diets). In summary, inorganic nitrate/nitrite supplementation showed good therapeutic effects in animal models of T2D and MS, as widely reported in other literature as [39, 213,214,215]. These anions increased insulin secretion from beta cells [210, 216] and improved peripheral glucose utilization [217, 218], visceral fat accumulation, and circulating levels of triglycerides. We hypothesize that endogenous NO deficiency may be responsible for MS and T2D and that NRB performs a critical part in the alternative pathway (NO3/NO2/NO pathway). Joshipua et al. [219] studied the effects of oral disinfectant rinses on more than 900 people over 3 years. The participants who used the mouthwash regularly were found to have a 55% higher risk of developing prediabetes/diabetes than those who used it infrequently. The authors did not examine the underlying mechanism of this process, but it may be that the use of antiseptic mouthwash blocked the action of NRB. In human clinical trials, nitrate and nitrite were ineffective in improving metabolic disorders [220,221,222]. Gilchrist and his colleagues [223] found that while dietary supplementation with nitrate caused a noticeable rise in circulating nitrite in patients with T2D, it did not improve islet function in patients with T2D. The cause of the paucity of effect was thought to be linked to the fact that T2D patients were being treated with bisphosphonates. The mechanisms of action of inorganic nitrate and biguanides share some striking similarities in many respects [224], leading to no significant effect of DN supplementation in patients already receiving biguanides. Among these, the interaction of nitrate and biguanides with the host microbiota may be central to the underlying mechanism [225,226,227]. Studies have shown that the high relative abundance of NRB in human OC is associated with insulin resistance and reduced risk of prediabetes [78]. Both animal and human trials have shown that alterations in the oral microbiota of patients with T2D lead to reduced nitrate reduction in the OC, decreased NO bioavailability, and the development of IR. In contrast, inorganic nitrate can modulate the oral microbiota by raising the number of health-related NRB and reducing the abundance of Prevotella and Weyongococcus spp., thereby increasing NO production and improving NO utilization. In addition, dietary epidemiological research has revealed that increased intake of nitrate-rich vegetables can convey weight loss and anti-diabetic effects and prevent the development of T2D [228, 229]. A review has discussed in detail how inorganic nitrate can improve the oral microbial community of patients with T2D and make it plays a probiotic role [230]. Further understanding of the pathophysiological mechanisms of T2D and MS will help develop new preventive and therapeutic strategies. Future animal studies or clinical trials will assess the potential beneficial effects of dietary supplementation with NRB in patients with metabolic diseases, including T2D. Combining the effects of the interaction of NRB with nitrate, restoring the oral microbiota of patients with T2D or MS to a state comparable to health is a state-of-the-art approach [41], which could enhance systemic NO production and offer an alternative way in the presence of impaired enzyme-dependent endogenous pathways. These findings may offer innovative nutritional prevention and treatment strategies for T2D and MS.

Nonalcoholic fatty liver

Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, called hepatic steatosis or fatty liver, and is closely related to overweight, obesity, and MS [231, 232]. NAFLD can be reversed with weight loss and movement, which can also develop into severe diseases, including non-alcoholic steatohepatitis, fibrosis, and liver cirrhosis [233, 234]. Recent studies have shown that steatosis can be avoided by simple dietary approaches in rodent and human models of MS [209, 235]. In the latest trial, DN was proven to modify vascular function in patients with hypercholesterolemia [151]. Sonoda et al. [16] further found that high nitrate levels in diet positively affect liver steatosis associated with metabolic syndrome. DN provides fuel for the NO3/NO2/NO pathway, which can reverse many characteristics of metabolic syndrome and liver steatosis in high-fat diet mice (whether combined with NO synthase inhibitor (l-NAME) or not) [6, 236, 237]. Many scholars believe impaired NO bioavailability and signaling may be a candidate mechanism for hepatic steatosis [28, 238]. Lázár et al. [239] noted that the beneficial effects of nitrate metabolism could be attributed to its reduction of nitrite followed by further generation of NO species and activation of soluble guanylate cyclase [114, 240]. Furthermore, Cordero-Herrera et al.’s [15] in vivo disease model and in vitro studies using HepG2 cells and primary human hepatocyte spheres show that stimulating the NO3/NO2/NO pathway may slow down the evolution of hepatic steatosis by activating AMP-activated protein kinase (AMPK) signal transduction and reducing NOX-derived oxidative stress. These beneficial effects of nitrate were not present in germ-free mice, suggesting a central part of the host microbiota in the bioactivation of nitrate, which is necessary for DN to avoid steatosis. These findings may have implications for developing new strategies for the prevention and treatment of hepatic steatosis associated with metabolic disorders based on NRB.

Neuro system

Sympathetic excitation is accompanied by the increase of AngII signal and oxidative stress and impairs the bioavailability of NO [18, 19]. DN supplementation may influence the body's systemic health by modulating sympathetic activity and thus reducing vascular tone, regulating BP, and altering mental behaviors. These beneficial impacts of DN may be mediated through the inhibitory effect of the NO3/NO2/NO pathway on sympathetic excitation, in which NRB play an important role [18, 241]. Guimarães et al. [18] first demonstrated that long-term nitrate supplementation inhibited or restored regular sympathetic activity in an animal model of angiotensin-converting enzyme II-induced hypertension. However, this experiment did not directly verify the role of NRB but instead emphasized the significance of NRB in the regulation of NO homeostasis and signaling via the NO3/NO2/NO pathway. NO has a significant function in regulating synaptogenesis and neurotransmission in the central and peripheral nervous system [242]. There is evidence that basal NO production and NO bioavailability are reduced in diabetic patients [17]. Oghbaei et al. [243] used a ureazotocin-induced diabetic male rat model to demonstrate that long-term supplementation with DN can affect testicular function and structure in these rats through the hypothalamic–pituitary–gonadal axis, thereby improving fertility parameters. Subsequently, Keyhanmanesh et al. [244] also demonstrated the beneficial therapeutic effect of DN supplementation on testicular damage in streptozotocin-induced diabetic male rats. These findings are correlated with the increase in miR-34b and decrease in p53 mRNA, of which the role of NRB has not been revealed. We speculate that NRB may function as an essential part of this process. In addition, García-Jaramillo et al. [245] pointed out that DN supplementation altered the metabolomic profile of the zebrafish brain and led to mild anxiety-like behaviors. They suggest that DN supplementation can deplete brain metabolites (e.g., reduction of γ-aminobutyric acid and its precursor glutamine) by modulating neural activity, a process in which NO plays a vital role. We hypothesize that NRB is closely related to the operation of regulation of neural activity in humans, pending further experimental evidence. In addition to regulating nerve activity, DN can improve cognitive function by regulating blood flow to the brain and reducing response times in neuropsychological tests [246, 247]. Vanhatalo et al. [13] found that after DN supplementation, the microbiome module associated with pro-inflammatory metabolism decreased, and the microbial module with NRA increased and suggested that this microbial module alteration may be related to improvements in age-induced cognitive impairment. The relationship between NRB and cognitive function still needs further experimental validation.

Respiratory system

Bronchopulmonary dysplasia

Bronchopulmonary dysplasia (BPD) is the most common severe respiratory complication in preterm infants and contributes significantly to the mortality of preterm infants [22, 248]. Currently, we lack treatments to block the progression of the disease as well as biomarkers to predict BPD. Despite the complexity of the aetiology and pathogenesis of BPD (which remains to be elucidated), recent studies demonstrated a relationship between the microecological environment of the digestive system (including the oral, respiratory, and intestinal tracts) and BPD. Among them, the correlation microbiome mediates NRA that regulates NO bioavailability and signaling, and defective NO bioavailability can lead to an increased incidence of BPD. Therefore, the bacteria with NRA in the OC may be the key to predicting and treating BPD [77, 249, 250]. In a single-center prospective cohort study, Gentle et al. [251] found that NRA peaked at 29 weeks of gestational age; when infants were categorized and compared by whether they had BPD, those with BPD had noticeably lower NRA at 29 weeks of gestational age than those without BPD. High NRA was associated with a lower incidence of BPD. This suggests that oral microflora and NRA may play a role in the occurrence of BPD in very premature infants. The difference in NRA mediated by oral microflora may provide a non-invasive biomarker for the development of BPD and has the potential for targeted therapy. In addition, studies have shown that significant changes in respiratory tract microecology (such as abnormal microbial diversity and differences in evolutionary patterns) have taken place in premature infants before the occurrence of BPD [252, 253]. Similarly, Wagner et al. [254] noted that the respiratory tract of infants with severe BPD has a higher weekly detectable microbial load than that of infants with mild BPD (with a lower colonization rate of Staphylococcus in the respiratory tract of infants with severe BPD). This suggests that the colonization pattern of respiratory microflora in premature infants may be a marker for predicting the severity of BPD. A review has elucidated in some detail the relationship between respiratory microbes and BPD and their primary mechanisms of action in the pathogenesis of BPD, suggesting that intervention of respiratory microecology with probiotics holds promise to become a new policy in the therapy of BPD [22]. Furthermore, there is growing evidence that the respiratory and gut microecological environments can interact, affecting the development of BPD. Dysregulation of the respiratory and gut microbiota can cause immune disorders and exacerbate the consequences of disease by stimulating inflammatory processes [255, 256]. Therefore, maintaining respiratory and intestinal microecological balance can significantly improve BPD. Studies have shown that there are also bacteria with NRA in the respiratory tract and intestinal tract. These bacteria are essential in regulating the microecological balance of the respiratory tract and intestinal tract [257, 258]. Therefore, we hypothesize that NRB may be highly promising targets for predicting and treating BPD. At present, NO and NO-producing precursors have been evaluated as potential BPD predictive markers and prevention methods, but the relationship between nitrate-reducing bacteria and BPD and the role of nitrate-reducing bacteria in predicting and treating BPD need to be further studied.

Urology systems

Chronic kidney disease

Chronic kidney disease (CKD) is a primary health problem affecting 8–16% of the global population [259]. Studies have shown that the microflora of patients with CKD is impaired (biological imbalance), increasing the number of potentially pathogenic and pro-inflammatory bacteria that can produce uremic toxins that facilitate the progression of CKD and decrease the production of enzymatic NO [24]. It has been confirmed in most animal experiments that the bioavailability of NO in patients with CKD is reduced. Since the decreased bioavailability of NO and increased oxidative stress are the key to the occurrence and development of renal disease [260], the therapeutic targets for NO may be beneficial [261,262,263]. In several disease models, DN supplementation has increased NO production and reduced endothelial dysfunction [145, 264]. In addition, supplementation with nitrite has been demonstrated to prevent or minimize renal damage directly by increasing NO production but has not been shown to improve the prognosis of patients with CKD [265, 266]. Similarly, we can also prevent or reduce renal impairment by increasing NO production through DN supplementation. Different ways of stimulating the NO3/NO2/NO pathway have been presented to avoid renal ischemia–reperfusion damage. Nitrate supplementation can improve renal injury in ischemia–reperfusion animal model [267, 268]. Two weeks of DN supplementation (1 mmol/kg/day) enhanced glomerular perfusion and filtration, prevented a decrease in glomerular filtration rate and attenuated glomerular and tubular injury in mice 2 weeks after ischemic renal injury [269]. Mechanistically, these renal protective effects associated with nitrate supplementation have been linked to reduced oxidative stress and increased NO bioavailability. A review detailed that the enhanced NO3/NO2/NO pathway may perform an antioxidant role through different targets and cellular mechanisms (including regulation of mitochondrial function, reduction of ROS produced by NOX and XO, and restoration of eNOS function) [25]. Although NRB is the only human symbiotic bacteria in the NO3/NO2/NO pathway, few studies clarify the relationship between NRB and CKD. As the impact of microorganisms on human health is gradually revealed, the microbiota can regulate the nitrogen cycle in various ways (e.g., through nitrification and denitrification, regulation of reduction and oxidation reactions, and through biological and chemical reactions) and play a crucial role in NO production [21, 270, 271]. One review has elaborated on how microorganisms regulate the nitrogen cycle [24]. Therefore, the effect of NRB on CKD may be a topic worth exploring in depth. In conclusion, the nitrogen cycle is an essential component of CKD prevention. Improper regulation of any part of the nitrogen cycle may promote the development of CKD. Regulating the nitrogen cycle by developing interventions that target NO production (e.g., supplementing NRB to regulate the microbiota) and thereby increasing NO production has the potential to reduce the risk of CKD. So far, few studies have fully described the role of NRB in the oral, esophageal or gastrointestinal microflora of patients with CKD. Therefore, we should focus on exploring this aspect in future studies and experiments, which may provide promising innovative ideas for predicting and treating CKD.

Summary and discussion

NRB perform a vital part in human health and diseases, because human beings lack the enzyme mechanism of reducing nitrate to nitrite, so we can only rely on NRB to reduce nitrate and make the NO3/NO2/NO pathway play a substitute role in the inactivation of the classical NOS pathway, thus restoring the balance of NO. In addition, NRB function as a critical regulator in the human floras. NRB have many beneficial effects on the body by restoring NO balance through the NO3/NO2/NO pathway or by regulating floras balance, including improving the hypofunction of salivary glands due to radiotherapy for HNT, regulating oral floras, protecting the stomach by improving gastric MBF and gastric mucus production, regulating intestinal floras, improving hypertension and pulmonary hypertension, alleviating heart disease, diabetes, MS, fatty liver, bronchopulmonary dysplasia, and chronic kidney disease. This review comprehensively summarizes the potential relationship between NRB and human health and conditions, an aspect that has received little attention and has not been summarized. NRB may be the key to the production of NO by nitrate metabolism and affect human health and may also be important bacteria for human health regulation. Our follow-up study can use these bacteria as a breakthrough to study the potential mechanism between nitrate metabolism, health, and disease. Therefore, this article accumulates a theoretical foundation for further studies on the effects of NRB on human health and diseases and stimulates more thoughts. The relationship between the function of microorganisms and the balance of human health and disease is becoming increasingly clear. Among them, studying the effects of bacteria with nitrate reductase on human systemic health and disease is still at an early stage. With the accelerated evolution of high-throughput sequencing and bioinformatics technologies, comprehensive microbiome characterization of NRB in different disease populations has become possible. According to the current research, the related studies on the relationship between NRB and human health and diseases are based on digestive tract floras (including the oral cavity, esophagus and gastrointestinal tract). This suggests a close relationship between NRB in our digestive tract and human health, which is worthy of in-depth excavation. Furthermore, NRB can influence the health of other body organs through the oral–intestinal axis, which can provide innovative ideas for disease diagnosis and treatment, such as finding biomarkers for specific diagnoses of diseases and treating-related conditions through probiotics by screening the changes of NRB in the digestive tract. In addition, the beneficial effect of NRB on the human body may be a protective mechanism of disease state or ageing, which not only indicates that NRB may be a potential therapeutic target but also may be a beneficial flora for improving age-related diseases, which needs to be further confirmed by more clinical trials. Recently, the modulation of the gut microbiota by NRB as beneficial commensals has attracted attention because of the potential effect of these floras on a variety of conditions and also as supplements to improve exercise performance [272, 273]. Using diet or supplements to change the microflora to increase the conversion of nitrate to nitrite, to increase the production and utilization of NO signal, which opens a new way in the field of sports performance. The use of high-throughput sequencing technology and biological information technology to understand the bacteria with nitrate reductase activity has been deepened, and a large number of data and information have been obtained. How to effectively transform big biological data into a clinical diagnosis and treatment method with practical application value and then provide effective personalized medical services for patients, there are still many problems to be solved. The growing development of metagenomics and high-throughput sequencing technologies significantly extended the human knowledge of the association between NRB and systemic diseases. Understanding the specific mechanism of NRB affecting human health and diseases and controlling NO3/NO2/NO metabolism is of great significance for preventing and treating human systemic diseases.

Availability of data and materials

This review does not involve statistics, and all the pictures are original.

References

  1. Armet AM, Deehan EC, O’Sullivan AF, Mota JF, Field CJ, Prado CM, et al. Rethinking healthy eating in light of the gut microbiome. Cell Host Microbe. 2022;30(6):764–85.

    PubMed  CAS  Google Scholar 

  2. Attaye I, Warmbrunn MV, Boot ANAF, van der Wolk SC, Hutten BA, Daams JG, et al. A systematic review and meta-analysis of dietary interventions modulating gut microbiota and cardiometabolic diseases-striving for new standards in microbiome studies. Gastroenterology. 2022;162(7):1911–32.

    PubMed  CAS  Google Scholar 

  3. Jukic Peladic N, Dell’Aquila G, Carrieri B, Maggio M, Cherubini A, Orlandoni P. Potential role of probiotics for inflammaging: a narrative review. Nutrients. 2021;13(9):2919.

    PubMed  PubMed Central  CAS  Google Scholar 

  4. Michels N, Zouiouich S, Vanderbauwhede B, Vanacker J, Indave Ruiz BI, Huybrechts I. Human microbiome and metabolic health: an overview of systematic reviews. Obes Rev. 2022;23(4): e13409.

    PubMed  Google Scholar 

  5. Peng X, Cheng L, You Y, Tang C, Ren B, Li Y, et al. Oral microbiota in human systematic diseases. Int J Oral Sci. 2022;14(1):14.

    PubMed  PubMed Central  CAS  Google Scholar 

  6. Vazquez-Torres A, Baumler AJ. Nitrate, nitrite and nitric oxide reductases: from the last universal common ancestor to modern bacterial pathogens. Curr Opin Microbiol. 2016;29:1–8.

    PubMed  CAS  Google Scholar 

  7. Lundberg JO, Weitzberg E. Nitric oxide signaling in health and disease. Cell. 2022;185(16):2853–78.

    PubMed  CAS  Google Scholar 

  8. Wang S, El-Fahmawi A, Christian DA, Fang Q, Radaelli E, Chen L, et al. Infection-induced intestinal dysbiosis is mediated by macrophage activation and nitrate production. MBio. 2019;10(3):e00935-e1019.

    PubMed  PubMed Central  CAS  Google Scholar 

  9. Lim Y, Tang KD, Karpe AV, Beale DJ, Totsika M, Kenny L, et al. Chemoradiation therapy changes oral microbiome and metabolomic profiles in patients with oral cavity cancer and oropharyngeal cancer. Head Neck. 2021;43(5):1521–34.

    PubMed  Google Scholar 

  10. Nasseri-Moghaddam S, Nokhbeh-Zaeem H, Saniee P, Pedramnia S, Sotoudeh M, Malekzadeh R. Oral nitrate reductase activity and erosive gastro-esophageal reflux disease: a nitrate hypothesis for GERD pathogenesis. Dig Dis Sci. 2012;57(2):413–8.

    PubMed  CAS  Google Scholar 

  11. Li Z, Dou L, Zhang Y, He S, Zhao D, Hao C, et al. Characterization of the oral and esophageal microbiota in esophageal precancerous lesions and squamous cell carcinoma. Front Cell Infect Microbiol. 2021;11: 714162.

    PubMed  PubMed Central  CAS  Google Scholar 

  12. Blekkenhorst LC, Bondonno NP, Liu AH, Ward NC, Prince RL, Lewis JR, et al. Nitrate, the oral microbiome, and cardiovascular health: a systematic literature review of human and animal studies. Am J Clin Nutr. 2018;107(4):504–22.

    PubMed  Google Scholar 

  13. Vanhatalo A, L’Heureux JE, Kelly J, Blackwell JR, Wylie LJ, Fulford J, et al. Network analysis of nitrate-sensitive oral microbiome reveals interactions with cognitive function and cardiovascular health across dietary interventions. Redox Biol. 2021;41: 101933.

    PubMed  PubMed Central  CAS  Google Scholar 

  14. Vanhatalo A, Blackwell JR, L’Heureux JE, Williams DW, Smith A, van der Giezen M, et al. Nitrate-responsive oral microbiome modulates nitric oxide homeostasis and blood pressure in humans. Free Radical Biol Med. 2018;124:21–30.

    CAS  Google Scholar 

  15. Cordero-Herrera I, Kozyra M, Zhuge Z, McCann Haworth S, Moretti C, Peleli M, et al. AMP-activated protein kinase activation and NADPH oxidase inhibition by inorganic nitrate and nitrite prevent liver steatosis. Proc Natl Acad Sci USA. 2019;116(1):217–26.

    PubMed  CAS  Google Scholar 

  16. Sonoda K, Kono Y, Kitamori K, Ohtake K, Shiba S, Kasono K, et al. Beneficial effects of dietary nitrite on a model of nonalcoholic steatohepatitis induced by high-fat/high-cholesterol diets in SHRSP5/Dmcr rats: a preliminary study. Int J Mol Sci. 2022;23(6):2931.

    PubMed  PubMed Central  CAS  Google Scholar 

  17. Ikonomidis I, Pavlidis G, Tsoumani M, Kousathana F, Katogiannis K, Tsilivarakis D, et al. Endothelial dysfunction is associated with decreased nitric oxide bioavailability in dysglycaemic subjects and first-degree relatives of type 2 diabetic patients. J Clin Med. 2022;11(12):3299.

    PubMed  PubMed Central  CAS  Google Scholar 

  18. Guimarães DD, Cruz JC, Carvalho-Galvão A, Zhuge Z, Marques SM, Naves LM, et al. Dietary nitrate reduces blood pressure in rats with angiotensin II-induced hypertension via mechanisms that involve reduction of sympathetic hyperactivity. Hypertension (Dallas, Tex: 1979). 2019;73(4):839–48.

    PubMed  Google Scholar 

  19. Sharma NM, Haibara AS, Katsurada K, Liu X, Patel KP. Central angiotensin II-protein inhibitor of neuronal nitric oxide synthase (PIN) axis contribute to neurogenic hypertension. Nitric Oxide Biol Chem. 2020;94:54–62.

    CAS  Google Scholar 

  20. Kakavandi NR, Hasanvand A, Ghazi-Khansari M, Sezavar AH, Nabizadeh H, Parohan M. Maternal dietary nitrate intake and risk of neural tube defects: a systematic review and dose-response meta-analysis. Food Chem Toxicol. 2018;118:287–93.

    PubMed  CAS  Google Scholar 

  21. Morou-Bermúdez E, Torres-Colón JE, Bermúdez NS, Patel RP, Joshipura KJ. Pathways linking oral bacteria, nitric oxide metabolism, and health. J Dent Res. 2022;101(6):623–31.

    PubMed  PubMed Central  Google Scholar 

  22. Sun T, Yu H, Fu J. Respiratory tract microecology and bronchopulmonary dysplasia in preterm infants. Front Pediatr. 2021;9: 762545.

    PubMed  PubMed Central  Google Scholar 

  23. Soodaeva S, Klimanov I, Kubysheva N, Popova N, Batyrshin I. The state of the nitric oxide cycle in respiratory tract diseases. Oxid Med Cell Longev. 2020;2020:4859260.

    PubMed  PubMed Central  Google Scholar 

  24. Briskey D, Tucker PS, Johnson DW, Coombes JS. Microbiota and the nitrogen cycle: implications in the development and progression of CVD and CKD. Nitric Oxide. 2016;57:64–70.

    PubMed  CAS  Google Scholar 

  25. Carlstrom M, Montenegro MF. Oxidative stress and compromised nitric oxide signaling in cardiorenal disease therapeutic value of stimulating the nitrate-nitrite-nitric oxide pathway. J Intern Med. 2019;285(1):2–18.

    PubMed  CAS  Google Scholar 

  26. Cosola C, Sabatino A, di Bari I, Fiaccadori E, Gesualdo L. Nutrients, nutraceuticals, and xenobiotics affecting renal health. Nutrients. 2018;10(7):808.

    PubMed  PubMed Central  Google Scholar 

  27. Kobayashi J. Effect of diet and gut environment on the gastrointestinal formation of N-nitroso compounds: a review. Nitric Oxide. 2018;73:66–73.

    PubMed  CAS  Google Scholar 

  28. Ma L, Hu L, Jin L, Wang J, Li X, Wang W, et al. Rebalancing glucolipid metabolism and gut microbiome dysbiosis by nitrate-dependent alleviation of high-fat diet-induced obesity. BMJ Open Diabetes Res Care. 2020;8(1): e001255.

    PubMed  PubMed Central  Google Scholar 

  29. Rocha BS, Laranjinha J. Nitrate from diet might fuel gut microbiota metabolism: minding the gap between redox signaling and inter-kingdom communication. Free Radic Biol Med. 2020;149:37–43.

    PubMed  CAS  Google Scholar 

  30. Tiso M, Schechter AN. Nitrate reduction to nitrite, nitric oxide and ammonia by gut bacteria under physiological conditions. PLoS ONE. 2015;10(3): e0119712.

    PubMed  PubMed Central  Google Scholar 

  31. Qu XM, Wu ZF, Pang BX, Jin LY, Qin LZ, Wang SL. From nitrate to nitric oxide: the role of salivary glands and oral bacteria. J Dent Res. 2016;95(13):1452–6.

    PubMed  CAS  Google Scholar 

  32. Shannon OM, Gregory S, Siervo M. Dietary nitrate, aging and brain health: the latest evidence. Curr Opin Clin Nutr Metab Care. 2022;25:393–400.

    PubMed  Google Scholar 

  33. Babateen AM, Shannon OM, O’Brien GM, Okello E, Smith E, Olgacer D, et al. Incremental doses of nitrate-rich beetroot juice do not modify cognitive function and cerebral blood flow in overweight and obese older adults: a 13-week pilot randomised clinical trial. Nutrients. 2022;14(5):1052.

    PubMed  PubMed Central  CAS  Google Scholar 

  34. Piknova B, Schechter AN, Park JW, Vanhatalo A, Jones AM. Skeletal muscle nitrate as a regulator of systemic nitric oxide homeostasis. Exerc Sport Sci Rev. 2022;50(1):2.

    PubMed  Google Scholar 

  35. Brunetta HS, Petrick HL, Momken I, Handy RM, Pignanelli C, Nunes EA, et al. Nitrate consumption preserves HFD-induced skeletal muscle mitochondrial ADP sensitivity and lysine acetylation: a potential role for SIRT1. Redox Biol. 2022;52: 102307.

    PubMed  PubMed Central  CAS  Google Scholar 

  36. Ashworth A, Cutler C, Farnham G, Liddle L, Burleigh M, Rodiles A, et al. Dietary intake of inorganic nitrate in vegetarians and omnivores and its impact on blood pressure, resting metabolic rate and the oral microbiome. Free Radic Biol Med. 2019;138:63–72.

    PubMed  CAS  Google Scholar 

  37. Alzahrani HS, Jackson KG, Hobbs DA, Lovegrove JA. The role of dietary nitrate and the oral microbiome on blood pressure and vascular tone. Nutr Res Rev. 2021;34(2):222–39.

    PubMed  CAS  Google Scholar 

  38. Rosier BT, Takahashi N, Zaura E, Krom BP, MartÍnez-Espinosa RM, van Breda SGJ, et al. The importance of nitrate reduction for oral health. J Dent Res. 2022;101(8):887–97.

    PubMed  CAS  Google Scholar 

  39. Lundberg JO, Carlström M, Weitzberg E. Metabolic effects of dietary nitrate in health and disease. Cell Metab. 2018;28(1):9–22.

    PubMed  CAS  Google Scholar 

  40. González-Soltero R, Bailén M, de Lucas B, Ramírez-Goercke MI, Pareja-Galeano H, Larrosa M. Role of oral and gut microbiota in dietary nitrate metabolism and its impact on sports performance. Nutrients. 2020;12(12):3611.

    PubMed  PubMed Central  Google Scholar 

  41. Pignatelli P, Fabietti G, Ricci A, Piattelli A, Curia MC. How periodontal disease and presence of nitric oxide reducing oral bacteria can affect blood pressure. Int J Mol Sci. 2020;21(20):7538.

    PubMed  PubMed Central  CAS  Google Scholar 

  42. Said Abasse K, Essien EE, Abbas M, Yu X, Xie W, Sun J, et al. Association between dietary nitrate, nitrite intake, and site-specific cancer risk: a systematic review and meta-analysis. Nutrients. 2022;14(3):666.

    PubMed  PubMed Central  CAS  Google Scholar 

  43. Moazeni M, Heidari Z, Golipour S, Ghaisari L, Sillanpää M, Ebrahimi A. Dietary intake and health risk assessment of nitrate, nitrite, and nitrosamines: a Bayesian analysis and Monte Carlo simulation. Environ Sci Pollut Res Int. 2020;27(36):45568–80.

    PubMed  CAS  Google Scholar 

  44. Tannenbaum SR, Correa P. Nitrate and gastric cancer risks. Nature. 1985;317(6039):675–6.

    PubMed  CAS  Google Scholar 

  45. Tricker AR, Preussmann R. Carcinogenic N-nitrosamines in the diet: occurrence, formation, mechanisms and carcinogenic potential. Mutat Res. 1991;259(3–4):277–89.

    PubMed  CAS  Google Scholar 

  46. Tricker AR, Pfundstein B, Theobald E, Preussmann R, Spiegelhalder B. Mean daily intake of volatile N-nitrosamines from foods and beverages in West Germany in 1989–1990. Food Chem Toxicol. 1991;29(11):729–32.

    PubMed  CAS  Google Scholar 

  47. Alexander J, Benford DJ, Cockburn A, Cravedi J-P, Dogliotti E, Domenico Ad, et al., editors. Nitrate in vegetables scientific opinion of the panel on contaminants in the food chain 12008.

  48. Ohara M, Suyama T. Nitrate reductase activity during the embryonal development of the frog. Nature. 1952;169(4294):285–6.

    PubMed  CAS  Google Scholar 

  49. Akiyama K, Kimura A, Suzuki H, Takeyama Y, Gluckman TL, Terhakopian A, et al. Production of oxidative products of nitric oxide in infarcted human heart. J Am Coll Cardiol. 1998;32(2):373–9.

    PubMed  CAS  Google Scholar 

  50. Burleigh MC, Liddle L, Monaghan C, Muggeridge DJ, Sculthorpe N, Butcher JP, et al. Salivary nitrite production is elevated in individuals with a higher abundance of oral nitrate-reducing bacteria. Free Radic Biol Med. 2018;120:80–8.

    PubMed  CAS  Google Scholar 

  51. Daims H, Lucker S, Wagner M. A new perspective on microbes formerly known as nitrite-oxidizing bacteria. Trends Microbiol. 2016;24(9):699–712.

    PubMed  PubMed Central  CAS  Google Scholar 

  52. Duncan C, Dougall H, Johnston P, Green S, Brogan R, Leifert C, et al. Chemical generation of nitric oxide in the mouth from the enterosalivary circulation of dietary nitrate. Nat Med. 1995;1(6):546–51.

    PubMed  CAS  Google Scholar 

  53. Blot S. Antiseptic mouthwash, the nitrate-nitrite-nitric oxide pathway, and hospital mortality: a hypothesis generating review. Intensive Care Med. 2021;47(1):28–38.

    PubMed  CAS  Google Scholar 

  54. Flores M, Toldra F. Chemistry, safety, and regulatory considerations in the use of nitrite and nitrate from natural origin in meat products—invited review. Meat Sci. 2021;171: 108272.

    PubMed  CAS  Google Scholar 

  55. Shannon OM, Allen JD, Bescos R, Burke L, Clifford T, Easton C, et al. Dietary inorganic nitrate as an ergogenic aid: an expert consensus derived via the modified Delphi technique. Sports Med (Auckland, NZ). 2022;52:2537–58.

    Google Scholar 

  56. Kondo H, Akoumianakis I, Badi I, Akawi N, Kotanidis CP, Polkinghorne M, et al. Effects of canagliflozin on human myocardial redox signalling: clinical implications. Eur Heart J. 2021;42(48):4947–60.

    PubMed  PubMed Central  CAS  Google Scholar 

  57. Qin C, Bian X-L, Wu H-Y, Xian J-Y, Lin Y-H, Cai C-Y, et al. Prevention of the return of extinguished fear by disrupting the interaction of neuronal nitric oxide synthase with its carboxy-terminal PDZ ligand. Mol Psychiatry. 2021;26(11):6506–19.

    PubMed  CAS  Google Scholar 

  58. Wong NF, Xu-Friedman MA. Induction of activity-dependent plasticity at auditory nerve synapses. J Neurosci. 2022;42(32):6211–20.

    PubMed  PubMed Central  CAS  Google Scholar 

  59. Degjoni A, Campolo F, Stefanini L, Venneri MA. The NO/cGMP/PKG pathway in platelets: the therapeutic potential of PDE5 inhibitors in platelet disorders. J Thromb Haemost. 2022. https://doi.org/10.1111/jth.15844.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Qian Y, Kumar R, Chug MK, Massoumi H, Brisbois EJ. Therapeutic delivery of nitric oxide utilizing saccharide-based materials. ACS Appl Mater Interfaces. 2021. https://doi.org/10.1021/acsami.1c10964.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Tran DL, Le Thi P, Lee SM, Hoang Thi TT, Park KD. Multifunctional surfaces through synergistic effects of heparin and nitric oxide release for a highly efficient treatment of blood-contacting devices. J Control Release. 2021;329:401–12.

    PubMed  CAS  Google Scholar 

  62. Liu T, Schroeder H, Power GG, Blood AB. A physiologically relevant role for NO stored in vascular smooth muscle cells: a novel theory of vascular NO signaling. Redox Biol. 2022;53: 102327.

    PubMed  PubMed Central  CAS  Google Scholar 

  63. Durgin BG, Wood KC, Hahn SA, McMahon B, Baust JJ, Straub AC. Smooth muscle cell CYB5R3 preserves cardiac and vascular function under chronic hypoxic stress. J Mol Cell Cardiol. 2022;162:72–80.

    PubMed  CAS  Google Scholar 

  64. Jones T, Dunn EL, Macdonald JH, Kubis H-P, McMahon N, Sandoo A. The effects of beetroot juice on blood pressure, microvascular function and large-vessel endothelial function: a randomized, double-blind, placebo-controlled pilot study in healthy older adults. Nutrients. 2019;11(8):1792.

    PubMed  PubMed Central  CAS  Google Scholar 

  65. Benjamim CJR, Porto AA, Valenti VE, Sobrinho ACDS, Garner DM, Gualano B, et al. Nitrate derived from beetroot juice lowers blood pressure in patients with arterial hypertension: a systematic review and meta-analysis. Front Nutr. 2022;9: 823039.

    PubMed  PubMed Central  Google Scholar 

  66. Zamani H, de Joode MEJR, Hossein IJ, Henckens NFT, Guggeis MA, Berends JE, et al. The benefits and risks of beetroot juice consumption: a systematic review. Crit Rev Food Sci Nutr. 2021;61(5):788–804.

    PubMed  CAS  Google Scholar 

  67. Craig JC, Broxterman RM, Smith JR, Allen JD, Barstow TJ. Effect of dietary nitrate supplementation on conduit artery blood flow, muscle oxygenation, and metabolic rate during handgrip exercise. J Appl Physiol (1985). 2018;125(2):254–62.

    PubMed  CAS  Google Scholar 

  68. Brookes ZLS, Belfield LA, Ashworth A, Casas-Agustench P, Raja M, Pollard AJ, et al. Effects of chlorhexidine mouthwash on the oral microbiome. J Dent. 2021;113: 103768.

    PubMed  CAS  Google Scholar 

  69. Rosier BT, Moya-Gonzalvez EM, Corell-Escuin P, Mira A. Isolation and characterization of nitrate-reducing bacteria as potential probiotics for oral and systemic health. Front Microbiol. 2020;11: 555465.

    PubMed  PubMed Central  Google Scholar 

  70. Walker MY, Pratap S, Southerland JH, Farmer-Dixon CM, Lakshmyya K, Gangula PR. Role of oral and gut microbiome in nitric oxide-mediated colon motility. Nitric Oxide Biol Chem. 2018;73:81–8.

    CAS  Google Scholar 

  71. Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining the normal bacterial flora of the oral cavity. J Clin Microbiol. 2005;43(11):5721–32.

    PubMed  PubMed Central  Google Scholar 

  72. Kilian M, Chapple ILC, Hannig M, Marsh PD, Meuric V, Pedersen AML, et al. The oral microbiome—an update for oral healthcare professionals. Br Dent J. 2016;221(10):657–66.

    PubMed  CAS  Google Scholar 

  73. Krishnan K, Chen T, Paster BJ. A practical guide to the oral microbiome and its relation to health and disease. Oral Dis. 2017;23(3):276–86.

    PubMed  CAS  Google Scholar 

  74. Li H, Duncan C, Townend J, Killham K, Smith LM, Johnston P, et al. Nitrate-reducing bacteria on rat tongues. Appl Environ Microbiol. 1997;63(3):924–30.

    PubMed  PubMed Central  CAS  Google Scholar 

  75. Liddle L, Burleigh MC, Monaghan C, Muggeridge DJ, Sculthorpe N, Pedlar CR, et al. Variability in nitrate-reducing oral bacteria and nitric oxide metabolites in biological fluids following dietary nitrate administration: an assessment of the critical difference. Nitric Oxide Biol Chem. 2019;83:1–10.

    CAS  Google Scholar 

  76. Hyde ER, Luk B, Cron S, Kusic L, McCue T, Bauch T, et al. Characterization of the rat oral microbiome and the effects of dietary nitrate. Free Radical Biol Med. 2014;77:249–57.

    CAS  Google Scholar 

  77. Hyde ER, Andrade F, Vaksman Z, Parthasarathy K, Jiang H, Parthasarathy DK, et al. Metagenomic analysis of nitrate-reducing bacteria in the oral cavity: implications for nitric oxide homeostasis. PLoS ONE. 2014;9(3): e88645.

    PubMed  PubMed Central  Google Scholar 

  78. Goh CE, Trinh P, Colombo PC, Genkinger JM, Mathema B, Uhlemann A-C, et al. Association between nitrate-reducing oral bacteria and cardiometabolic outcomes: results from ORIGINS. J Am Heart Assoc. 2019;8(23): e013324.

    PubMed  PubMed Central  Google Scholar 

  79. Jones AM, Vanhatalo A, Seals DR, Rossman MJ, Piknova B, Jonvik KL. Dietary nitrate and nitric oxide metabolism: mouth, circulation, skeletal muscle, and exercise performance. Med Sci Sports Exerc. 2021;53(2):280–94.

    PubMed  CAS  Google Scholar 

  80. Hansen TH, Kern T, Bak EG, Kashani A, Allin KH, Nielsen T, et al. Impact of a vegan diet on the human salivary microbiota. Sci Rep. 2018;8(1):5847.

    PubMed  PubMed Central  Google Scholar 

  81. Ahmed KA, Kim K, Ricart K, Van Der Pol W, Qi X, Bamman MM, et al. Potential role for age as a modulator of oral nitrate reductase activity. Nitric Oxide Biol Chem. 2021;108:1–7.

    CAS  Google Scholar 

  82. Tribble GD, Angelov N, Weltman R, Wang B-Y, Eswaran SV, Gay IC, et al. Frequency of tongue cleaning impacts the human tongue microbiome composition and enterosalivary circulation of nitrate. Front Cell Infect Microbiol. 2019;9:39.

    PubMed  PubMed Central  CAS  Google Scholar 

  83. Senkus KE, Crowe-White KM. Influence of mouth rinse use on the enterosalivary pathway and blood pressure regulation: a systematic review. Crit Rev Food Sci Nutr. 2020;60(17):2874–86.

    PubMed  Google Scholar 

  84. Macfarlane S, Furrie E, Macfarlane GT, Dillon JF. Microbial colonization of the upper gastrointestinal tract in patients with Barrett’s esophagus. Clin Infect Dis. 2007;45(1):29–38.

    PubMed  Google Scholar 

  85. Pajecki D, Zilberstein B, Cecconello I, Dos Santos MAA, Yagi OK, Gama-Rodrigues JJ. Larger amounts of nitrite and nitrate-reducing bacteria in megaesophagus of Chagas’ disease than in controls. J Gastrointest Surg. 2007;11(2):199–203.

    PubMed  CAS  Google Scholar 

  86. Park CH, Lee JG, Lee AR, Eun CS, Han DS. Network construction of gastric microbiome and organization of microbial modules associated with gastric carcinogenesis. Sci Rep. 2019;9(1):12444.

    PubMed  PubMed Central  Google Scholar 

  87. Sanduleanu S, Jonkers D, De Bruïne A, Hameeteman W, Stockbrügger RW. Double gastric infection with Helicobacter pylori and non-Helicobacter pylori bacteria during acid-suppressive therapy: increase of pro-inflammatory cytokines and development of atrophic gastritis. Aliment Pharmacol Ther. 2001;15(8):1163–75.

    PubMed  CAS  Google Scholar 

  88. Mowat C, Williams C, Gillen D, Hossack M, Gilmour D, Carswell A, et al. Omeprazole, Helicobacter pylori status, and alterations in the intragastric milieu facilitating bacterial N-nitrosation. Gastroenterology. 2000;119(2):339–47.

    PubMed  CAS  Google Scholar 

  89. Jo HJ, Kim J, Kim N, Park JH, Nam RH, Seok Y-J, et al. Analysis of gastric microbiota by pyrosequencing: minor role of bacteria other than Helicobacter pylori in the gastric carcinogenesis. Helicobacter. 2016;21(5):364–74.

    PubMed  CAS  Google Scholar 

  90. Conley MN, Roberts C, Sharpton TJ, Iwaniec UT, Hord NG. Increasing dietary nitrate has no effect on cancellous bone loss or fecal microbiome in ovariectomized rats. Mol Nutr Food Res. 2017;61(5):1600372.

    PubMed  PubMed Central  Google Scholar 

  91. Rocha BS, Correia MG, Pereira A, Henriques I, Da Silva GJ, Laranjinha J. Inorganic nitrate prevents the loss of tight junction proteins and modulates inflammatory events induced by broad-spectrum antibiotics: a role for intestinal microbiota? Nitric Oxide Biol Chem. 2019;88:27–34.

    CAS  Google Scholar 

  92. Petersson J, Jädert C, Phillipson M, Borniquel S, Lundberg JO, Holm L. Physiological recycling of endogenous nitrate by oral bacteria regulates gastric mucus thickness. Free Radical Biol Med. 2015;89:241–7.

    CAS  Google Scholar 

  93. Petersson J, Phillipson M, Jansson EA, Patzak A, Lundberg JO, Holm L. Dietary nitrate increases gastric mucosal blood flow and mucosal defense. Am J Physiol Gastrointest Liver Physiol. 2007;292(3):G718–24.

    PubMed  CAS  Google Scholar 

  94. Mohamed NI, Suddek GM, El-Kashef DH. Molsidomine alleviates acetic acid-induced colitis in rats by reducing oxidative stress, inflammation and apoptosis. Int Immunopharmacol. 2021;99: 108005.

    PubMed  CAS  Google Scholar 

  95. Florin TH, Neale G, Cummings JH. The effect of dietary nitrate on nitrate and nitrite excretion in man. Br J Nutr. 1990;64(2):387–97.

    PubMed  CAS  Google Scholar 

  96. Dellavalle CT, Xiao Q, Yang G, Shu X-O, Aschebrook-Kilfoy B, Zheng W, et al. Dietary nitrate and nitrite intake and risk of colorectal cancer in the Shanghai Women’s Health Study. Int J Cancer. 2014;134(12):2917–26.

    PubMed  CAS  Google Scholar 

  97. Bradbury KE, Appleby PN, Key TJ. Fruit, vegetable, and fiber intake in relation to cancer risk: findings from the European Prospective Investigation into Cancer and Nutrition (EPIC). Am J Clin Nutr. 2014;100(Suppl 1):394S-S398.

    PubMed  CAS  Google Scholar 

  98. Machha A, Schechter AN. Inorganic nitrate: a major player in the cardiovascular health benefits of vegetables? Nutr Rev. 2012;70(6):367–72.

    PubMed  Google Scholar 

  99. Gangolli SD, van den Brandt PA, Feron VJ, Janzowsky C, Koeman JH, Speijers GJ, et al. Nitrate, nitrite and N-nitroso compounds. Eur J Pharmacol. 1994;292(1):1–38.

    PubMed  CAS  Google Scholar 

  100. Hord NG, Tang Y, Bryan NS. Food sources of nitrates and nitrites: the physiologic context for potential health benefits. Am J Clin Nutr. 2009;90(1):1–10.

    PubMed  CAS  Google Scholar 

  101. van Velzen AG, Sips AJAM, Schothorst RC, Lambers AC, Meulenbelt J. The oral bioavailability of nitrate from nitrate-rich vegetables in humans. Toxicol Lett. 2008;181(3):177–81.

    PubMed  Google Scholar 

  102. Vasco E, Dias MG, Oliveira L. The first harmonised total diet study in Portugal: nitrate occurrence and exposure assessment. Food Chem. 2022;392: 133152.

    PubMed  CAS  Google Scholar 

  103. Ma L, Hu L, Feng X, Wang S. Nitrate and nitrite in health and disease. Aging Dis. 2018;9(5):938–45.

    PubMed  PubMed Central  Google Scholar 

  104. Karwowska M, Kononiuk A. Nitrates/nitrites in food-risk for nitrosative stress and benefits. Antioxidants (Basel). 2020;9(3):241.

    PubMed  CAS  Google Scholar 

  105. DeMartino AW, Kim-Shapiro DB, Patel RP, Gladwin MT. Nitrite and nitrate chemical biology and signalling. Br J Pharmacol. 2019;176(2):228–45.

    PubMed  CAS  Google Scholar 

  106. Kevil CG, Kolluru GK, Pattillo CB, Giordano T. Inorganic nitrite therapy: historical perspective and future directions. Free Radic Biol Med. 2011;51(3):576–93.

    PubMed  PubMed Central  CAS  Google Scholar 

  107. Alderton WK, Cooper CE, Knowles RG. Nitric oxide synthases: structure, function and inhibition. Biochem J. 2001;357(Pt 3):593–615.

    PubMed  PubMed Central  CAS  Google Scholar 

  108. Oliveira-Paula GH, Pinheiro LC, Tanus-Santos JE. Mechanisms impairing blood pressure responses to nitrite and nitrate. Nitric Oxide. 2019;85:35–43.

    PubMed  CAS  Google Scholar 

  109. Rajapakse NW, Giam B, Kuruppu S, Head GA, Kaye DM. Impaired l-arginine-nitric oxide pathway contributes to the pathogenesis of resistant hypertension. Clin Sci (Lond). 2019;133(20):2061–7.

    PubMed  CAS  Google Scholar 

  110. Belzer V, Hanani M. Nitric oxide as a messenger between neurons and satellite glial cells in dorsal root ganglia. Glia. 2019;67(7):1296–307.

    PubMed  Google Scholar 

  111. Boccellino M, Galasso G, Ambrosio P, Stiuso P, Lama S, Di Zazzo E, et al. H9c2 cardiomyocytes under hypoxic stress: biological effects mediated by sentinel downstream targets. Oxid Med Cell Longev. 2021;2021:6874146.

    PubMed  PubMed Central  Google Scholar 

  112. Schiffer TA, Lundberg JO, Weitzberg E, Carlström M. Modulation of mitochondria and NADPH oxidase function by the nitrate-nitrite-NO pathway in metabolic disease with focus on type 2 diabetes. Biochim Biophys Acta. 2020;1866(8): 165811.

    CAS  Google Scholar 

  113. Carlstrom M, Montenegro MF. Therapeutic value of stimulating the nitrate-nitrite-nitric oxide pathway to attenuate oxidative stress and restore nitric oxide bioavailability in cardiorenal disease. J Intern Med. 2019;285(1):2–18.

    PubMed  CAS  Google Scholar 

  114. Lundberg JO, Gladwin MT, Weitzberg E. Strategies to increase nitric oxide signalling in cardiovascular disease. Nat Rev Drug Discovery. 2015;14(9):623–41.

    PubMed  CAS  Google Scholar 

  115. Lee DY, Lee SY, Jo C, Yoon Y, Jeong JY, Hur SJ. Effect on health from consumption of meat and meat products. J Anim Sci Technol. 2021;63(5):955–76.

    PubMed  PubMed Central  CAS  Google Scholar 

  116. Hezel MP, Liu M, Schiffer TA, Larsen FJ, Checa A, Wheelock CE, et al. Effects of long-term dietary nitrate supplementation in mice. Redox Biol. 2015;5:234–42.

    PubMed  PubMed Central  CAS  Google Scholar 

  117. Bryan NS, Alexander DD, Coughlin JR, Milkowski AL, Boffetta P. Ingested nitrate and nitrite and stomach cancer risk: an updated review. Food Chem Toxicol. 2012;50(10):3646–65.

    PubMed  CAS  Google Scholar 

  118. McNally B, Griffin JL, Roberts LD. Dietary inorganic nitrate: from villain to hero in metabolic disease? Mol Nutr Food Res. 2016;60(1):67–78.

    PubMed  CAS  Google Scholar 

  119. Godfrey S, Labhasetwar P, Wate S, Pimpalkar S. How safe are the global water coverage figures? Case study from Madhya Pradesh. India Environ Monit Assess. 2011;176(1–4):561–74.

    PubMed  Google Scholar 

  120. Verheijen FW, Verbeek E, Aula N, Beerens CE, Havelaar AC, Joosse M, et al. A new gene, encoding an anion transporter, is mutated in sialic acid storage diseases. Nat Genet. 1999;23(4):462–5.

    PubMed  CAS  Google Scholar 

  121. Qin L, Liu X, Sun Q, Fan Z, Xia D, Ding G, et al. Sialin (SLC17A5) functions as a nitrate transporter in the plasma membrane. Proc Natl Acad Sci USA. 2012;109(33):13434–9.

    PubMed  PubMed Central  CAS  Google Scholar 

  122. Reimer RJ. SLC17: a functionally diverse family of organic anion transporters. Mol Aspects Med. 2013;34(2–3):350–9.

    PubMed  PubMed Central  CAS  Google Scholar 

  123. Lundberg JO. Nitrate transport in salivary glands with implications for NO homeostasis. Proc Natl Acad Sci USA. 2012;109(33):13144–5.

    PubMed  PubMed Central  CAS  Google Scholar 

  124. Hezel MP, Weitzberg E. The oral microbiome and nitric oxide homoeostasis. Oral Dis. 2015;21(1):7–16.

    PubMed  CAS  Google Scholar 

  125. Feng X, Wu Z, Xu J, Xu Y, Zhao B, Pang B, et al. Dietary nitrate supplementation prevents radiotherapy-induced xerostomia. Elife. 2021;10: e70710.

    PubMed  PubMed Central  CAS  Google Scholar 

  126. Lundberg JO, Weitzberg E, Gladwin MT. The nitrate-nitrite-nitric oxide pathway in physiology and therapeutics. Nat Rev Drug Discov. 2008;7(2):156–67.

    PubMed  CAS  Google Scholar 

  127. Sabbah DA, Hajjo R, Sweidan K. Review on epidermal growth factor receptor (EGFR) structure, signaling pathways, interactions, and recent updates of EGFR inhibitors. Curr Top Med Chem. 2020;20(10):815–34.

    PubMed  CAS  Google Scholar 

  128. Zhang X, Tang N, Hadden TJ, Rishi AK. Akt, FoxO and regulation of apoptosis. Biochem Biophys Acta. 2011;1813(11):1978–86.

    PubMed  CAS  Google Scholar 

  129. Shen W, Tang D, Wan P, Peng Z, Sun M, Guo X, et al. Identification of tissue-specific microbial profile of esophageal squamous cell carcinoma by full-length 16S rDNA sequencing. Appl Microbiol Biotechnol. 2022;106(8):3215–29.

    PubMed  CAS  Google Scholar 

  130. Kovaleva O, Podlesnaya P, Rashidova M, Samoilova D, Petrenko A, Mochalnikova V, et al. Prognostic significance of the microbiome and stromal cells phenotype in esophagus squamous cell carcinoma. Biomedicines. 2021;9(7):743.

    PubMed  PubMed Central  CAS  Google Scholar 

  131. Wallace JL, Miller MJ. Nitric oxide in mucosal defense: a little goes a long way. Gastroenterology. 2000;119(2):512–20.

    PubMed  CAS  Google Scholar 

  132. Lanas A, Bajador E, Serrano P, Fuentes J, Carreño S, Guardia J, et al. Nitrovasodilators, low-dose aspirin, other nonsteroidal antiinflammatory drugs, and the risk of upper gastrointestinal bleeding. N Engl J Med. 2000;343(12):834–9.

    PubMed  CAS  Google Scholar 

  133. Jansson EA, Petersson J, Reinders C, Sobko T, Björne H, Phillipson M, et al. Protection from nonsteroidal anti-inflammatory drug (NSAID)-induced gastric ulcers by dietary nitrate. Free Radical Biol Med. 2007;42(4):510–8.

    CAS  Google Scholar 

  134. Jin L, Qin L, Xia D, Liu X, Fan Z, Zhang C, et al. Active secretion and protective effect of salivary nitrate against stress in human volunteers and rats. Free Radical Biol Med. 2013;57:61–7.

    CAS  Google Scholar 

  135. Eriksson KE, Yang T, Carlström M, Weitzberg E. Organ uptake and release of inorganic nitrate and nitrite in the pig. Nitric Oxide Biol Chem. 2018;75:16–26.

    CAS  Google Scholar 

  136. Lundberg JO, Govoni M. Inorganic nitrate is a possible source for systemic generation of nitric oxide. Free Radical Biol Med. 2004;37(3):395–400.

    CAS  Google Scholar 

  137. Hu L, Jin L, Xia D, Zhang Q, Ma L, Zheng H, et al. Nitrate ameliorates dextran sodium sulfate-induced colitis by regulating the homeostasis of the intestinal microbiota. Free Radical Biol Med. 2020;152:609–21.

    CAS  Google Scholar 

  138. Dicksved J, Halfvarson J, Rosenquist M, Järnerot G, Tysk C, Apajalahti J, et al. Molecular analysis of the gut microbiota of identical twins with Crohn’s disease. ISME J. 2008;2(7):716–27.

    PubMed  CAS  Google Scholar 

  139. Nishikawa J, Kudo T, Sakata S, Benno Y, Sugiyama T. Diversity of mucosa-associated microbiota in active and inactive ulcerative colitis. Scand J Gastroenterol. 2009;44(2):180–6.

    PubMed  CAS  Google Scholar 

  140. Walker AW, Sanderson JD, Churcher C, Parkes GC, Hudspith BN, Rayment N, et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2011;11:7.

    PubMed  PubMed Central  Google Scholar 

  141. Björne H, Weitzberg E, Lundberg JO. Intragastric generation of antimicrobial nitrogen oxides from saliva–physiological and therapeutic considerations. Free Radical Biol Med. 2006;41(9):1404–12.

    Google Scholar 

  142. Jädert C, Phillipson M, Holm L, Lundberg JO, Borniquel S. Preventive and therapeutic effects of nitrite supplementation in experimental inflammatory bowel disease. Redox Biol. 2014;2:73–81.

    PubMed  Google Scholar 

  143. Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med. 2006;3(11): e442.

    PubMed  PubMed Central  Google Scholar 

  144. Larsen FJ, Ekblom B, Sahlin K, Lundberg JO, Weitzberg E. Effects of dietary nitrate on blood pressure in healthy volunteers. N Engl J Med. 2006;355(26):2792–3.

    PubMed  CAS  Google Scholar 

  145. Kapil V, Milsom AB, Okorie M, Maleki-Toyserkani S, Akram F, Rehman F, et al. Inorganic nitrate supplementation lowers blood pressure in humans: role for nitrite-derived NO. Hypertension (Dallas, Tex: 1979). 2010;56(2):274–81.

    PubMed  CAS  Google Scholar 

  146. Vanhatalo A, Bailey SJ, Blackwell JR, DiMenna FJ, Pavey TG, Wilkerson DP, et al. Acute and chronic effects of dietary nitrate supplementation on blood pressure and the physiological responses to moderate-intensity and incremental exercise. Am J Physiol Regul Integr Comp Physiol. 2010;299(4):R1121–31.

    PubMed  CAS  Google Scholar 

  147. Webb AJ, Patel N, Loukogeorgakis S, Okorie M, Aboud Z, Misra S, et al. Acute blood pressure lowering, vasoprotective, and antiplatelet properties of dietary nitrate via bioconversion to nitrite. Hypertension (Dallas, Tex: 1979). 2008;51(3):784–90.

    PubMed  CAS  Google Scholar 

  148. Koch CD, Gladwin MT, Freeman BA, Lundberg JO, Weitzberg E, Morris A. Enterosalivary nitrate metabolism and the microbiome: Intersection of microbial metabolism, nitric oxide and diet in cardiac and pulmonary vascular health. Free Radical Biol Med. 2017;105:48–67.

    CAS  Google Scholar 

  149. Bryan NS. Functional nitric oxide nutrition to combat cardiovascular disease. Curr Atheroscler Rep. 2018;20(5):21.

    PubMed  Google Scholar 

  150. Carlstrom M, Lundberg JO, Weitzberg E. Mechanisms underlying blood pressure reduction by dietary inorganic nitrate. Acta Physiol (Oxf). 2018;224(1): e13080.

    PubMed  CAS  Google Scholar 

  151. Velmurugan S, Gan JM, Rathod KS, Khambata RS, Ghosh SM, Hartley A, et al. Dietary nitrate improves vascular function in patients with hypercholesterolemia: a randomized, double-blind, placebo-controlled study. Am J Clin Nutr. 2016;103(1):25–38.

    PubMed  CAS  Google Scholar 

  152. Batista RIM, Nogueira RC, Ferreira GC, Oliveira-Paula GH, Damacena-Angelis C, Pinheiro LC, et al. Antiseptic mouthwash inhibits antihypertensive and vascular protective effects of L-arginine. Eur J Pharmacol. 2021;907: 174314.

    PubMed  CAS  Google Scholar 

  153. Petersson J, Carlström M, Schreiber O, Phillipson M, Christoffersson G, Jägare A, et al. Gastroprotective and blood pressure lowering effects of dietary nitrate are abolished by an antiseptic mouthwash. Free Radical Biol Med. 2009;46(8):1068–75.

    CAS  Google Scholar 

  154. Babateen AM, Shannon OM, Mathers JC, Siervo M. Validity and reliability of test strips for the measurement of salivary nitrite concentration with and without the use of mouthwash in healthy adults. Nitric Oxide Biol Chem. 2019;91:15–22.

    CAS  Google Scholar 

  155. Bescos R, Ashworth A, Cutler C, Brookes ZL, Belfield L, Rodiles A, et al. Effects of chlorhexidine mouthwash on the oral microbiome. Sci Rep. 2020;10(1):5254.

    PubMed  PubMed Central  CAS  Google Scholar 

  156. Sundqvist ML, Lundberg JO, Weitzberg E. Effects of antiseptic mouthwash on resting metabolic rate: a randomized, double-blind, crossover study. Nitric Oxide Biol Chem. 2016;61:38–44.

    CAS  Google Scholar 

  157. Dewhurst-Trigg R, Yeates T, Blackwell JR, Thompson C, Linoby A, Morgan PT, et al. Lowering of blood pressure after nitrate-rich vegetable consumption is abolished with the co-ingestion of thiocyanate-rich vegetables in healthy normotensive males. Nitric Oxide Biol Chem. 2018;74:39–46.

    CAS  Google Scholar 

  158. Huppertz B. Placental origins of preeclampsia: challenging the current hypothesis. Hypertension (Dallas, Tex: 1979). 2008;51(4):970–5.

    PubMed  CAS  Google Scholar 

  159. Bramham K, Parnell B, Nelson-Piercy C, Seed PT, Poston L, Chappell LC. Chronic hypertension and pregnancy outcomes: systematic review and meta-analysis. BMJ. 2014;348: g2301.

    PubMed  PubMed Central  Google Scholar 

  160. Chappell LC, Enye S, Seed P, Briley AL, Poston L, Shennan AH. Adverse perinatal outcomes and risk factors for preeclampsia in women with chronic hypertension: a prospective study. Hypertension (Dallas, Tex: 1979). 2008;51(4):1002–9.

    PubMed  CAS  Google Scholar 

  161. Ormesher L, Myers JE, Chmiel C, Wareing M, Greenwood SL, Tropea T, et al. Effects of dietary nitrate supplementation, from beetroot juice, on blood pressure in hypertensive pregnant women: a randomised, double-blind, placebo-controlled feasibility trial. Nitric Oxide Biol Chem. 2018;80:37–44.

    CAS  Google Scholar 

  162. Guignabert C, Tu L, Girerd B, Ricard N, Huertas A, Montani D, et al. New molecular targets of pulmonary vascular remodeling in pulmonary arterial hypertension: importance of endothelial communication. Chest. 2015;147(2):529–37.

    PubMed  Google Scholar 

  163. Kinsella JP, Neish SR, Ivy DD, Shaffer E, Abman SH. Clinical responses to prolonged treatment of persistent pulmonary hypertension of the newborn with low doses of inhaled nitric oxide. J Pediatr. 1993;123(1):103–8.

    PubMed  CAS  Google Scholar 

  164. Kinsella JP, Neish SR, Shaffer E, Abman SH. Low-dose inhalation nitric oxide in persistent pulmonary hypertension of the newborn. Lancet. 1992;340(8823):819–20.

    PubMed  CAS  Google Scholar 

  165. Roberts JD, Fineman JR, Morin FC, Shaul PW, Rimar S, Schreiber MD, et al. Inhaled nitric oxide and persistent pulmonary hypertension of the newborn. The inhaled nitric oxide study group. N Engl J Med. 1997;336(9):605–10.

    PubMed  CAS  Google Scholar 

  166. Hendgen-Cotta UB, Luedike P, Totzeck M, Kropp M, Schicho A, Stock P, et al. Dietary nitrate supplementation improves revascularization in chronic ischemia. Circulation. 2012;126(16):1983–92.

    PubMed  CAS  Google Scholar 

  167. Thum T, Fraccarollo D, Thum S, Schultheiss M, Daiber A, Wenzel P, et al. Differential effects of organic nitrates on endothelial progenitor cells are determined by oxidative stress. Arterioscler Thromb Vasc Biol. 2007;27(4):748–54.

    PubMed  CAS  Google Scholar 

  168. Hung H-C, Joshipura KJ, Jiang R, Hu FB, Hunter D, Smith-Warner SA, et al. Fruit and vegetable intake and risk of major chronic disease. J Natl Cancer Inst. 2004;96(21):1577–84.

    PubMed  Google Scholar 

  169. Joshipura KJ, Ascherio A, Manson JE, Stampfer MJ, Rimm EB, Speizer FE, et al. Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA. 1999;282(13):1233–9.

    PubMed  CAS  Google Scholar 

  170. Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, et al. Heart disease and stroke statistics-2022 update: a report from the American Heart Association. Circulation. 2022;145(8):e153–639.

    PubMed  Google Scholar 

  171. Notay K, Incognito AV, Millar PJ. Acute beetroot juice supplementation on sympathetic nerve activity: a randomized, double-blind, placebo-controlled proof-of-concept study. Am J Physiol Heart Circ Physiol. 2017;313(1):H59–65.

    PubMed  Google Scholar 

  172. Bock JM, Ueda K, Schneider AC, Hughes WE, Limberg JK, Bryan NS, et al. Inorganic nitrate supplementation attenuates peripheral chemoreflex sensitivity but does not improve cardiovagal baroreflex sensitivity in older adults. Am J Physiol Heart Circ Physiol. 2018;314(1):H45–51.

    PubMed  Google Scholar 

  173. Pellegrino D, Shiva S, Angelone T, Gladwin MT, Tota B. Nitrite exerts potent negative inotropy in the isolated heart via eNOS-independent nitric oxide generation and cGMP-PKG pathway activation. Biochem Biophys Acta. 2009;1787(7):818–27.

    PubMed  CAS  Google Scholar 

  174. Cannon RO, Epstein SE. “Microvascular angina” as a cause of chest pain with angiographically normal coronary arteries. Am J Cardiol. 1988;61(15):1338–43.

    PubMed  Google Scholar 

  175. Kaski JC, Rosano GM, Collins P, Nihoyannopoulos P, Maseri A, Poole-Wilson PA. Cardiac syndrome X: clinical characteristics and left ventricular function. Long-term follow-up study. J Am Coll Cardiol. 1995;25(4):807–14.

    PubMed  CAS  Google Scholar 

  176. Kanno S, Lee PC, Zhang Y, Ho C, Griffith BP, Shears LL, et al. Attenuation of myocardial ischemia/reperfusion injury by superinduction of inducible nitric oxide synthase. Circulation. 2000;101(23):2742–8.

    PubMed  CAS  Google Scholar 

  177. Emdin M, Aimo A, Castiglione V, Vergaro G, Georgiopoulos G, Saccaro LF, et al. Targeting cyclic guanosine monophosphate to treat heart failure: JACC review topic of the week. J Am Coll Cardiol. 2020;76(15):1795–807.

    PubMed  CAS  Google Scholar 

  178. Raubenheimer K, Hickey D, Leveritt M, Fassett R, Ortiz de Zevallos Munoz J, Allen JD, et al. Acute effects of nitrate-rich beetroot juice on blood pressure, hemostasis and vascular inflammation markers in healthy older adults: a randomized, placebo-controlled crossover study. Nutrients. 2017;9(11):1270.

    PubMed  PubMed Central  Google Scholar 

  179. Mónica FZ, Bian K, Murad F. The endothelium-dependent nitric oxide-cGMP pathway. Adv Pharmacol. 2016;77:1–27.

    PubMed  Google Scholar 

  180. Padala SK, Lavelle MP, Sidhu MS, Cabral KP, Morrone D, Boden WE, et al. Antianginal therapy for stable ischemic heart disease: a contemporary review. J Cardiovasc Pharmacol Ther. 2017;22(6):499–510.

    PubMed  CAS  Google Scholar 

  181. Reddy YNV, Lewis GD, Shah SJ, LeWinter M, Semigran M, Davila-Roman VG, et al. INDIE-HFpEF (inorganic nitrite delivery to improve exercise capacity in heart failure with preserved ejection fraction): rationale and design. Circ Heart Fail. 2017;10(5): e003862.

    PubMed  PubMed Central  CAS  Google Scholar 

  182. Münzel T, Daiber A. Inorganic nitrite and nitrate in cardiovascular therapy: a better alternative to organic nitrates as nitric oxide donors? Vascul Pharmacol. 2018;102:1–10.

    PubMed  Google Scholar 

  183. Thadani U. Secondary preventive potential of nitrates in ischaemic heart disease. Eur Heart J. 1996;17(Suppl F):30–6.

    PubMed  Google Scholar 

  184. Steitieh D, Amin N. Angina pectoris worsened by mouthwash. Proc (Bayl Univ Med Cent). 2019;32(4):570–1.

    PubMed  Google Scholar 

  185. van Heerebeek L, Hamdani N, Falcão-Pires I, Leite-Moreira AF, Begieneman MPV, Bronzwaer JGF, et al. Low myocardial protein kinase G activity in heart failure with preserved ejection fraction. Circulation. 2012;126(7):830–9.

    PubMed  Google Scholar 

  186. Franssen C, Chen S, Unger A, Korkmaz HI, De Keulenaer GW, Tschöpe C, et al. Myocardial microvascular inflammatory endothelial activation in heart failure with preserved ejection fraction. JACC Heart Fail. 2016;4(4):312–24.

    PubMed  Google Scholar 

  187. Mohammed SF, Hussain S, Mirzoyev SA, Edwards WD, Maleszewski JJ, Redfield MM. Coronary microvascular rarefaction and myocardial fibrosis in heart failure with preserved ejection fraction. Circulation. 2015;131(6):550–9.

    PubMed  Google Scholar 

  188. van Empel VPM, Mariani J, Borlaug BA, Kaye DM. Impaired myocardial oxygen availability contributes to abnormal exercise hemodynamics in heart failure with preserved ejection fraction. J Am Heart Assoc. 2014;3(6): e001293.

    PubMed  PubMed Central  Google Scholar 

  189. Srivaratharajah K, Coutinho T, deKemp R, Liu P, Haddad H, Stadnick E, et al. Reduced myocardial flow in heart failure patients with preserved ejection fraction. Circ Heart Fail. 2016;9(7): e002562.

    PubMed  Google Scholar 

  190. Lai Y-C, Tabima DM, Dube JJ, Hughan KS, Vanderpool RR, Goncharov DA, et al. SIRT3-AMP-activated protein kinase activation by nitrite and metformin improves hyperglycemia and normalizes pulmonary hypertension associated with heart failure with preserved ejection fraction. Circulation. 2016;133(8):717–31.

    PubMed  PubMed Central  CAS  Google Scholar 

  191. Larsen FJ, Schiffer TA, Borniquel S, Sahlin K, Ekblom B, Lundberg JO, et al. Dietary inorganic nitrate improves mitochondrial efficiency in humans. Cell Metab. 2011;13(2):149–59.

    PubMed  CAS  Google Scholar 

  192. Coggan AR, Leibowitz JL, Spearie CA, Kadkhodayan A, Thomas DP, Ramamurthy S, et al. Acute dietary nitrate intake improves muscle contractile function in patients with heart failure: a double-blind, placebo-controlled, randomized trial. Circ Heart Failure. 2015;8(5):914–20.

    PubMed  CAS  Google Scholar 

  193. Davidson KW, Barry MJ, Mangione CM, Cabana M, Caughey AB, Davis EM, et al. Screening for prediabetes and type 2 diabetes: US preventive services task force recommendation statement. JAMA. 2021;326(8):736–43.

    PubMed  Google Scholar 

  194. Bozkurt B, Aguilar D, Deswal A, Dunbar SB, Francis GS, Horwich T, et al. Contributory risk and management of comorbidities of hypertension, obesity, diabetes mellitus, hyperlipidemia, and metabolic syndrome in chronic heart failure: a scientific statement from the american heart association. Circulation. 2016;134(23):e535–78.

    PubMed  Google Scholar 

  195. He S, Wang J, Zhang X, Qian X, Yan S, Wang W, et al. Long-term influence of type 2 diabetes and metabolic syndrome on all-cause and cardiovascular death, and microvascular and macrovascular complications in Chinese adults—a 30-year follow-up of the Da Qing Diabetes Study. Diabetes Res Clin Pract. 2022;191: 110048.

    PubMed  CAS  Google Scholar 

  196. Carter P, Gray LJ, Troughton J, Khunti K, Davies MJ. Fruit and vegetable intake and incidence of type 2 diabetes mellitus: systematic review and meta-analysis. BMJ. 2010;341: c4229.

    PubMed  PubMed Central  Google Scholar 

  197. Liese AD, Nichols M, Sun X, D’Agostino RB, Haffner SM. Adherence to the DASH Diet is inversely associated with incidence of type 2 diabetes: the insulin resistance atherosclerosis study. Diabetes Care. 2009;32(8):1434–6.

    PubMed  PubMed Central  Google Scholar 

  198. Long J, Cai Q, Steinwandel M, Hargreaves MK, Bordenstein SR, Blot WJ, et al. Association of oral microbiome with type 2 diabetes risk. J Periodontal Res. 2017;52(3):636–43.

    PubMed  PubMed Central  CAS  Google Scholar 

  199. Wang R-R, Xu Y-S, Ji M-M, Zhang L, Li D, Lang Q, et al. Association of the oral microbiome with the progression of impaired fasting glucose in a Chinese elderly population. J Oral Microbiol. 2019;11(1):1605789.

    PubMed  PubMed Central  CAS  Google Scholar 

  200. Graves DT, Corrêa JD, Silva TA. The oral microbiota is modified by systemic diseases. J Dent Res. 2019;98(2):148–56.

    PubMed  CAS  Google Scholar 

  201. Lundberg JO, Weitzberg E, Cole JA, Benjamin N. Nitrate, bacteria and human health. Nat Rev Microbiol. 2004;2(7):593–602.

    PubMed  CAS  Google Scholar 

  202. Bahadoran Z, Mirmiran P, Ghasemi A. Role of nitric oxide in insulin secretion and glucose metabolism. Trends Endocrinol Metab. 2020;31(2):118–30.

    PubMed  CAS  Google Scholar 

  203. Chu S, Bohlen HG. High concentration of glucose inhibits glomerular endothelial eNOS through a PKC mechanism. Am J Physiol Renal Physiol. 2004;287(3):F384–92.

    PubMed  CAS  Google Scholar 

  204. Cosentino F, Eto M, De Paolis P, van der Loo B, Bachschmid M, Ullrich V, et al. High glucose causes upregulation of cyclooxygenase-2 and alters prostanoid profile in human endothelial cells: role of protein kinase C and reactive oxygen species. Circulation. 2003;107(7):1017–23.

    PubMed  CAS  Google Scholar 

  205. Geraldes P, King GL. Activation of protein kinase C isoforms and its impact on diabetic complications. Circ Res. 2010;106(8):1319–31.

    PubMed  PubMed Central  CAS  Google Scholar 

  206. Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front Physiol. 2019;10:1607.

    PubMed  Google Scholar 

  207. Huang PL. eNOS, metabolic syndrome and cardiovascular disease. Trends Endocrinol Metab. 2009;20(6):295–302.

    PubMed  PubMed Central  CAS  Google Scholar 

  208. Carlström M, Larsen FJ, Nyström T, Hezel M, Borniquel S, Weitzberg E, et al. Dietary inorganic nitrate reverses features of metabolic syndrome in endothelial nitric oxide synthase-deficient mice. Proc Natl Acad Sci USA. 2010;107(41):17716–20.

    PubMed  PubMed Central  Google Scholar 

  209. Ohtake K, Ehara N, Chiba H, Nakano G, Sonoda K, Ito J, et al. Dietary nitrite reverses features of postmenopausal metabolic syndrome induced by high-fat diet and ovariectomy in mice. Am J Physiol Endocrinol Metab. 2017;312(4):E300–8.

    PubMed  Google Scholar 

  210. Nyström T, Ortsäter H, Huang Z, Zhang F, Larsen FJ, Weitzberg E, et al. Inorganic nitrite stimulates pancreatic islet blood flow and insulin secretion. Free Radical Biol Med. 2012;53(5):1017–23.

    Google Scholar 

  211. Khalifi S, Rahimipour A, Jeddi S, Ghanbari M, Kazerouni F, Ghasemi A. Dietary nitrate improves glucose tolerance and lipid profile in an animal model of hyperglycemia. Nitric Oxide Biol Chem. 2015;44:24–30.

    CAS  Google Scholar 

  212. Gheibi S, Jeddi S, Carlström M, Gholami H, Ghasemi A. Effects of long-term nitrate supplementation on carbohydrate metabolism, lipid profiles, oxidative stress, and inflammation in male obese type 2 diabetic rats. Nitric Oxide Biol Chem. 2018;75:27–41.

    CAS  Google Scholar 

  213. Li T, Lu X, Sun Y, Yang X. Effects of spinach nitrate on insulin resistance, endothelial dysfunction markers and inflammation in mice with high-fat and high-fructose consumption. Food Nutr Res. 2016;60:32010.

    PubMed  Google Scholar 

  214. Ghasemi A, Jeddi S. Anti-obesity and anti-diabetic effects of nitrate and nitrite. Nitric Oxide Biol Chem. 2017;70:9–24.

    CAS  Google Scholar 

  215. Bahadoran Z, Ghasemi A, Mirmiran P, Azizi F, Hadaegh F. Beneficial effects of inorganic nitrate/nitrite in type 2 diabetes and its complications. Nutr Metab (Lond). 2015;12:16.

    PubMed  Google Scholar 

  216. Gheibi S, Bakhtiarzadeh F, Jeddi S, Farrokhfall K, Zardooz H, Ghasemi A. Nitrite increases glucose-stimulated insulin secretion and islet insulin content in obese type 2 diabetic male rats. Nitric Oxide Biol Chem. 2017;64:39–51.

    Google Scholar 

  217. Jiang H, Torregrossa AC, Potts A, Pierini D, Aranke M, Garg HK, et al. Dietary nitrite improves insulin signaling through GLUT4 translocation. Free Radical Biol Med. 2014;67:51–7.

    CAS  Google Scholar 

  218. Roberts LD, Ashmore T, Kotwica AO, Murfitt SA, Fernandez BO, Feelisch M, et al. Inorganic nitrate promotes the browning of white adipose tissue through the nitrate-nitrite-nitric oxide pathway. Diabetes. 2015;64(2):471–84.

    PubMed  CAS  Google Scholar 

  219. Joshipura KJ, Muñoz-Torres FJ, Morou-Bermudez E, Patel RP. Over-the-counter mouthwash use and risk of pre-diabetes/diabetes. Nitric Oxide Biol Chem. 2017;71:14–20.

    CAS  Google Scholar 

  220. Mills CE, Govoni V, Faconti L, Casagrande M-L, Morant SV, Crickmore H, et al. A randomised, factorial trial to reduce arterial stiffness independently of blood pressure: Proof of concept? The VaSera trial testing dietary nitrate and spironolactone. Br J Clin Pharmacol. 2020;86(5):891–902.

    PubMed  PubMed Central  CAS  Google Scholar 

  221. Shepherd AI, Gilchrist M, Winyard PG, Jones AM, Hallmann E, Kazimierczak R, et al. 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 Radical Biol Med. 2015;86:200–8.

    CAS  Google Scholar 

  222. Greenway FL, Predmore BL, Flanagan DR, Giordano T, Qiu Y, Brandon A, et al. Single-dose pharmacokinetics of different oral sodium nitrite formulations in diabetes patients. Diabetes Technol Ther. 2012;14(7):552–60.

    PubMed  PubMed Central  CAS  Google Scholar 

  223. Gilchrist M, Winyard PG, Aizawa K, Anning C, Shore A, Benjamin N. Effect of dietary nitrate on blood pressure, endothelial function, and insulin sensitivity in type 2 diabetes. Free Radical Biol Med. 2013;60:89–97.

    CAS  Google Scholar 

  224. Pernicova I, Korbonits M. Metformin–mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol. 2014;10(3):143–56.

    PubMed  CAS  Google Scholar 

  225. Bauer PV, Duca FA, Waise TMZ, Rasmussen BA, Abraham MA, Dranse HJ, et al. Metformin alters upper small intestinal microbiota that impact a glucose-SGLT1-sensing glucoregulatory pathway. Cell Metab. 2018;27(1):101–17.

    PubMed  CAS  Google Scholar 

  226. de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, Velásquez-Mejía EP, Carmona JA, Abad JM, et al. Metformin is associated with higher relative abundance of mucin-degrading akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut. Diabetes Care. 2017;40(1):54–62.

    PubMed  Google Scholar 

  227. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528(7581):262–6.

    PubMed  PubMed Central  CAS  Google Scholar 

  228. Appel LJ, Moore TJ, Obarzanek E, Vollmer WM, Svetkey LP, Sacks FM, et al. A clinical trial of the effects of dietary patterns on blood pressure. DASH Collaborative Research Group. N Engl J Med. 1997;336(16):1117–24.

    PubMed  CAS  Google Scholar 

  229. Ahluwalia A, Gladwin M, Coleman GD, Hord N, Howard G, Kim-Shapiro DB, et al. Dietary nitrate and the epidemiology of cardiovascular disease: report from a national heart, lung, and blood institute workshop. J Am Heart Assoc. 2016;5(7): e003402.

    PubMed  PubMed Central  Google Scholar 

  230. Bahadoran Z, Mirmiran P, Carlstrom M, Ghasemi A. Inorganic nitrate: a potential prebiotic for oral microbiota dysbiosis associated with type 2 diabetes. Nitric Oxide. 2021;116:38–46.

    PubMed  CAS  Google Scholar 

  231. Bilson J, Sethi JK, Byrne CD. Non-alcoholic fatty liver disease: a multi-system disease influenced by ageing and sex, and affected by adipose tissue and intestinal function. Proc Nutr Soc. 2022;81(2):146–61.

    PubMed  CAS  Google Scholar 

  232. Long MT, Noureddin M, Lim JK. AGA clinical practice update: diagnosis and management of nonalcoholic fatty liver disease in lean individuals: expert review. Gastroenterology. 2022;163(3):764-774.e1.

    PubMed  Google Scholar 

  233. Watanabe S, Hashimoto E, Ikejima K, Uto H, Ono M, Sumida Y, et al. Evidence-based clinical practice guidelines for nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. J Gastroenterol. 2015;50(4):364–77.

    PubMed  Google Scholar 

  234. Cusi K, Isaacs S, Barb D, Basu R, Caprio S, Garvey WT, et al. American Association of Clinical Endocrinology clinical practice guideline for the diagnosis and management of nonalcoholic fatty liver disease in primary care and endocrinology clinical settings: co-sponsored by the american association for the study of liver diseases (AASLD). Endocr Pract. 2022;28(5):528–62.

    PubMed  Google Scholar 

  235. Wang H, Hu L, Li L, Wu X, Fan Z, Zhang C, et al. Inorganic nitrate alleviates the senescence-related decline in liver function. Sci China Life Sci. 2018;61(1):24–34.

    PubMed  CAS  Google Scholar 

  236. Aluko EO, Omobowale TO, Oyagbemi AA, Adejumobi OA, Ajibade TO, Fasanmade AA. Reduction in nitric oxide bioavailability shifts serum lipid content towards atherogenic lipoprotein in rats. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2018;101:792–7.

  237. Sato I, Yamamoto S, Kakimoto M, Fujii M, Honma K, Kumazaki S, et al. Suppression of nitric oxide synthase aggravates non-alcoholic steatohepatitis and atherosclerosis in SHRSP5/Dmcr rat via acceleration of abnormal lipid metabolism. Pharmacol Rep. 2022;74(4):669–83.

    PubMed  CAS  Google Scholar 

  238. Eccleston HB, Andringa KK, Betancourt AM, King AL, Mantena SK, Swain TM, et al. Chronic exposure to a high-fat diet induces hepatic steatosis, impairs nitric oxide bioavailability, and modifies the mitochondrial proteome in mice. Antioxid Redox Signal. 2011;15(2):447–59.

    PubMed  PubMed Central  CAS  Google Scholar 

  239. Lázár Z, Mészáros M, Bikov A. The nitric oxide pathway in pulmonary arterial hypertension: pathomechanism, biomarkers and drug targets. Curr Med Chem. 2020;27(42):7168–88.

    PubMed  Google Scholar 

  240. Litvinova L, Atochin DN, Fattakhov N, Vasilenko M, Zatolokin P, Kirienkova E. Nitric oxide and mitochondria in metabolic syndrome. Front Physiol. 2015;6:20.

    PubMed  PubMed Central  Google Scholar 

  241. Abdulla MH, Johns EJ. The role of brain angiotensin II (type 2) receptors and nitric oxide in the renal sympathoinhibitory response to acute volume expansion in conscious rats. J Hypertens. 2017;35(2):338–47.

    PubMed  CAS  Google Scholar 

  242. Kiss JP. Role of nitric oxide in the regulation of monoaminergic neurotransmission. Brain Res Bull. 2000;52(6):459–66.

    PubMed  CAS  Google Scholar 

  243. Oghbaei H, Alipour MR, Hamidian G, Ahmadi M, Ghorbanzadeh V, Keyhanmanesh R. Two months sodium nitrate supplementation alleviates testicular injury in streptozotocin-induced diabetic male rats. Exp Physiol. 2018;103(12):1603–17.

    PubMed  CAS  Google Scholar 

  244. Keyhanmanesh R, Hamidian G, Alipour MR, Oghbaei H. Beneficial treatment effects of dietary nitrate supplementation on testicular injury in streptozotocin-induced diabetic male rats. Reprod Biomed Online. 2019;39(3):357–71.

    PubMed  CAS  Google Scholar 

  245. García-Jaramillo M, Beaver LM, Truong L, Axton ER, Keller RM, Prater MC, et al. Nitrate and nitrite exposure leads to mild anxiogenic-like behavior and alters brain metabolomic profile in zebrafish. PLoS ONE. 2020;15(12): e0240070.

    PubMed  PubMed Central  Google Scholar 

  246. Wightman EL, Haskell-Ramsay CF, Thompson KG, Blackwell JR, Winyard PG, Forster J, et al. Dietary nitrate modulates cerebral blood flow parameters and cognitive performance in humans: a double-blind, placebo-controlled, crossover investigation. Physiol Behav. 2015;149:149–58.

    PubMed  CAS  Google Scholar 

  247. Gilchrist M, Winyard PG, Fulford J, Anning C, Shore AC, Benjamin N. Dietary nitrate supplementation improves reaction time in type 2 diabetes: development and application of a novel nitrate-depleted beetroot juice placebo. Nitric Oxide Biol Chem. 2014;40:67–74.

    CAS  Google Scholar 

  248. Yang K, Dong W. Perspectives on probiotics and bronchopulmonary dysplasia. Front Pediatr. 2020;8: 570247.

    PubMed  PubMed Central  Google Scholar 

  249. Ruiz L, Bacigalupe R, García-Carral C, Boix-Amoros A, Argüello H, Silva CB, et al. Microbiota of human precolostrum and its potential role as a source of bacteria to the infant mouth. Sci Rep. 2019;9(1):8435.

    PubMed  PubMed Central  Google Scholar 

  250. Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker NM. Antibiotic treatment at delivery shapes the initial oral microbiome in neonates. Sci Rep. 2017;7:43481.

    PubMed  PubMed Central  Google Scholar 

  251. Gentle SJ, Ahmed KA, Yi N, Morrow CD, Ambalavanan N, Lal CV, et al. Bronchopulmonary dysplasia is associated with reduced oral nitrate reductase activity in extremely preterm infants. Redox Biol. 2021;38: 101782.

    PubMed  CAS  Google Scholar 

  252. Ambalavanan N, Cotten CM, Page GP, Carlo WA, Murray JC, Bhattacharya S, et al. Integrated genomic analyses in bronchopulmonary dysplasia. J Pediatr. 2015;166(3):531–7.

    PubMed  CAS  Google Scholar 

  253. Klinger G, Sokolover N, Boyko V, Sirota L, Lerner-Geva L, Reichman B. Perinatal risk factors for bronchopulmonary dysplasia in a national cohort of very-low-birthweight infants. Am J Obstet Gynecol. 2013;208(2):115.e1-115.e9.

    PubMed  Google Scholar 

  254. Wagner BD, Sontag MK, Harris JK, Miller JI, Morrow L, Robertson CE, et al. Airway microbial community turnover differs by BPD severity in ventilated preterm infants. PLoS ONE. 2017;12(1): e0170120.

    PubMed  PubMed Central  Google Scholar 

  255. Chen S-M, Lin C-P, Jan M-S. Early gut microbiota changes in preterm infants with bronchopulmonary dysplasia: a pilot case-control study. Am J Perinatol. 2021;38(11):1142–9.

    PubMed  Google Scholar 

  256. Marsland BJ, Trompette A, Gollwitzer ES. The gut-lung axis in respiratory disease. Ann Am Thorac Soc. 2015;12(Suppl 2):S150–6.

    PubMed  Google Scholar 

  257. Kolpen M, Kragh KN, Bjarnsholt T, Line L, Hansen CR, Dalbøge CS, et al. Denitrification by cystic fibrosis pathogens—Stenotrophomonas maltophilia is dormant in sputum. Int J Med Microbiol. 2015;305(1):1–10.

    PubMed  CAS  Google Scholar 

  258. Kolpen M, Kühl M, Bjarnsholt T, Moser C, Hansen CR, Liengaard L, et al. Nitrous oxide production in sputum from cystic fibrosis patients with chronic Pseudomonas aeruginosa lung infection. PLoS ONE. 2014;9(1): e84353.

    PubMed  PubMed Central  Google Scholar 

  259. Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382(9888):260–72.

    PubMed  Google Scholar 

  260. Baylis C. Nitric oxide deficiency in chronic kidney disease. Am J Physiol Renal Physiol. 2008;294(1):F1–9.

    PubMed  CAS  Google Scholar 

  261. Tumur Z, Niwa T. Oral sorbent AST-120 increases renal NO synthesis in uremic rats. J Ren Nutr. 2008;18(1):60–4.

    PubMed  Google Scholar 

  262. Kuczmarski JM, Martens CR, Kim J, Lennon-Edwards SL, Edwards DG. Cardiac function is preserved following 4 weeks of voluntary wheel running in a rodent model of chronic kidney disease. J Appl Physiol (1985). 2014;117(5):482–91.

    PubMed  CAS  Google Scholar 

  263. Al Suleimani YM, Al Za’abi M, Ramkumar A, Al Mahruqi AS, Tageldin MH, Nemmar A, et al. Influence of treatment with gum acacia on renal vascular responses in a rat model of chronic kidney disease. Eur Rev Med Pharmacol Sci. 2015;19(3):498–506.

    PubMed  CAS  Google Scholar 

  264. Sindler AL, Fleenor BS, Calvert JW, Marshall KD, Zigler ML, Lefer DJ, et al. Nitrite supplementation reverses vascular endothelial dysfunction and large elastic artery stiffness with aging. Aging Cell. 2011;10(3):429–37.

    PubMed  CAS  Google Scholar 

  265. Okamoto M, Tsuchiya K, Kanematsu Y, Izawa Y, Yoshizumi M, Kagawa S, et al. Nitrite-derived nitric oxide formation following ischemia-reperfusion injury in kidney. Am J Physiol Renal Physiol. 2005;288(1):F182–7.

    PubMed  CAS  Google Scholar 

  266. Tripatara P, Patel NSA, Webb A, Rathod K, Lecomte FMJ, Mazzon E, et al. Nitrite-derived nitric oxide protects the rat kidney against ischemia/reperfusion injury in vivo: role for xanthine oxidoreductase. J Am Soc Nephrol. 2007;18(2):570–80.

    PubMed  CAS  Google Scholar 

  267. Tsuchiya K, Tomita S, Ishizawa K, Abe S, Ikeda Y, Kihira Y, et al. Dietary nitrite ameliorates renal injury in L-NAME-induced hypertensive rats. Nitric Oxide Biol Chem. 2010;22(2):98–103.

    CAS  Google Scholar 

  268. Tatematsu S, Wakino S, Kanda T, Homma K, Yoshioka K, Hasegawa K, et al. Role of nitric oxide-producing and -degrading pathways in coronary endothelial dysfunction in chronic kidney disease. J Am Soc Nephrol. 2007;18(3):741–9.

    PubMed  CAS  Google Scholar 

  269. Yang T, Zhang X-M, Tarnawski L, Peleli M, Zhuge Z, Terrando N, et al. Dietary nitrate attenuates renal ischemia-reperfusion injuries by modulation of immune responses and reduction of oxidative stress. Redox Biol. 2017;13:320–30.

    PubMed  PubMed Central  CAS  Google Scholar 

  270. Silva KVC, Costa BD, Gomes AC, Saunders B, Mota JF. Factors that moderate the effect of nitrate ingestion on exercise performance in adults: a systematic review with meta-analyses and meta-regressions. Adv Nutr. 2022;13:1866–81.

    PubMed  PubMed Central  Google Scholar 

  271. Goh CE, Bohn B, Marotz C, Molinsky R, Roy S, Paster BJ, et al. Nitrite generating and depleting capacity of the oral microbiome and cardiometabolic risk: results from ORIGINS. J Am Heart Assoc. 2022;11(10): e023038.

    PubMed  PubMed Central  CAS  Google Scholar 

  272. Cerdá B, Pérez M, Pérez-Santiago JD, Tornero-Aguilera JF, González-Soltero R, Larrosa M. Gut microbiota modification: another piece in the puzzle of the benefits of physical exercise in health? Front Physiol. 2016;7:51.

    PubMed  PubMed Central  Google Scholar 

  273. Sorrenti V, Fortinguerra S, Caudullo G, Buriani A. Deciphering the Role of polyphenols in sports performance: from nutritional genomics to the gut microbiota toward phytonutritional epigenomics. Nutrients. 2020;12(5):1265.

    PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

Thanks to all the authors who have contributed to this article. Thanks to the Guangzhou Science and Technology Project and the Guangdong Science and Technology Program for their support.

Funding

This work was supported by the Science and Technology Project of Guangzhou City (201802020018) and the key area Project of Guangdong University (2023ZDZX2009).

Author information

Authors and Affiliations

Authors

Contributions

HL, YH, and MH wrote the manuscript; BJ was responsible for the overall conception, supervision, editing and management of this manuscript; MW, YM, WC, YC and ZT made the figures and document management. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Bo Jia.

Ethics declarations

Ethics approval and consent to participate

This review does not directly address studies of human participants or animal experiments. No moral approval or informed consent is required.

Consent for publication

All the participating authors of this article agreed to publish it.

Competing interests

The authors report no conflicts of interest in this work.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, H., Huang, Y., Huang, M. et al. From nitrate to NO: potential effects of nitrate-reducing bacteria on systemic health and disease. Eur J Med Res 28, 425 (2023). https://doi.org/10.1186/s40001-023-01413-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-023-01413-y

Keywords