Next Article in Journal
SLC6A4 DNA Methylation Levels and Serum Kynurenine/Tryptophan Ratio in Eating Disorders: A Possible Link with Psychopathological Traits?
Next Article in Special Issue
Skin Autofluorescence Mirrors Surrogate Parameters of Vascular Aging: An Enable Study
Previous Article in Journal
Selective Daily Mobility Bias in the Community Food Environment: Case Study of Greater Hartford, Connecticut
Previous Article in Special Issue
Alcoholic Liver Disease Is Associated with Elevated Plasma Levels of Novel Advanced Glycation End-Products: A Preliminary Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advanced Glycation End-Products and Their Effects on Gut Health

by
Kate Phuong-Nguyen
,
Bryony A. McNeill
,
Kathryn Aston-Mourney
and
Leni R. Rivera
*
IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Deakin University, Geelong 3220, Australia
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(2), 405; https://doi.org/10.3390/nu15020405
Submission received: 1 December 2022 / Revised: 20 December 2022 / Accepted: 11 January 2023 / Published: 13 January 2023

Abstract

:
Dietary advanced glycation end-products (AGEs) are a heterogeneous group of compounds formed when reducing sugars are heated with proteins, amino acids, or lipids at high temperatures for a prolonged period. The presence and accumulation of AGEs in numerous cell types and tissues are known to be prevalent in the pathology of many diseases. Modern diets, which contain a high proportion of processed foods and therefore a high level of AGE, cause deleterious effects leading to a multitude of unregulated intracellular and extracellular signalling and inflammatory pathways. Currently, many studies focus on investigating the chemical and structural aspects of AGEs and how they affect the metabolism and the cardiovascular and renal systems. Studies have also shown that AGEs affect the digestive system. However, there is no complete picture of the implication of AGEs in this area. The gastrointestinal tract is not only the first and principal site for the digestion and absorption of dietary AGEs but also one of the most susceptible organs to AGEs, which may exert many local and systemic effects. In this review, we summarise the current evidence of the association between a high-AGE diet and poor health outcomes, with a special focus on the relationship between dietary AGEs and alterations in the gastrointestinal structure, modifications in enteric neurons, and microbiota reshaping.

1. Introduction

Advanced glycation end-products (AGEs) are a group of chemically heterogeneous compounds that are formed through the Maillard reaction when reducing sugars react non-enzymatically with amino groups in proteins, nucleic acids, and lipids [1]. These reactions are irreversible, resulting in the accumulation of AGEs in the blood and tissues over time. In humans, the accumulation of AGEs over the lifespan contributes to the normal age-related physiological decline and is directly involved in the pathogenesis of degenerative musculoskeletal conditions in older people, such as osteoporosis [2], osteoarthritis [3,4], and sarcopenia [5,6,7]. However, an increase in AGE accumulation related to modern dietary practices has been implicated in the dramatic rise in the prevalence of non-communicable diseases in younger people, such as diabetes [8,9,10], kidney disease [10,11,12], and mental health conditions [13,14,15,16]. AGEs in the body can be broadly divided into two groups, i.e., endogenous and exogenous, reflecting their origin. Endogenous AGEs are formed in the body as part of normal metabolic processes. As endogenous AGE production is directly related to blood glucose concentration, their production is accelerated in hyperglycaemic conditions such as diabetes and insulin resistance [17,18]. Exogenous AGEs are derived from external sources, primarily the diet, and are present in particularly high concentrations in foods which have been cooked at high temperatures, are highly processed, or have been prepared for long-term storage [19].
Although the negative health effects of AGEs have been well described in most body systems, their effects on the gastrointestinal tract have been largely overlooked. Within the gut, there is a strong body of evidence to indicate that AGEs negatively affect the gut and overall body health directly, through their effects on the gut architecture, as well as indirectly, through their interactions with the gut microbiota. This review will focus on the effects of AGEs on gastrointestinal structure, function, and microbiome, as well as identify knowledge gaps in the current literature.

2. Formation of Advanced Glycation End-Products (AGEs)

The Maillard reaction is a complex reaction that occurs when the free amino groups of proteins, lipids, or nucleic acids are heated in the presence of reducing sugars (glucose, lactose, fructose, and maltose). This condensation reaction leads to the formation of an unstable Schiff base (early glycation product), which spontaneously undergoes a process of intramolecular degradation, known as the Amadori rearrangement, resulting in the formation of Amadori products (intermediate glycation products) [20,21,22]. The chemical arrangement of the Amadori products is more stable than that of Schiff bases, although this reaction is reversible as these products are highly unsaturated, making them susceptible to polymerisation [23,24]. In the final stage, the Amadori products undergo irreversible chemical rearrangements including oxidation, dehydration, enolisation, cyclisation, and fragmentation to produce diverse classes of reactive intermediates including reactive AGE precursors [3,25]. These reactive AGE precursors can interact with protein-bound lysine or arginine, leading to the formation of AGEs (Figure 1).
While the majority of AGEs are produced by the non-enzymatic Maillard reaction [26,27], some AGEs are formed via alternative pathways such as autoxidation of glucose and peroxidation of lipids [28,29,30,31,32], or via the polyol pathway. In this pathway, glucose is converted first to sorbitol by the enzyme aldose reductase and then to fructose by the action of sorbitol dehydrogenase. After that, metabolites of fructose (such as fructose-3-phosphate) are converted into α-oxaldehydes, which give rise to several reactive dicarbonyl derivatives (reactive AGE precursors), including glyoxal, methylglyoxal, and 3-deoxyglucasone [33], which bind to intracellular and extracellular proteins and DNA to modify them, producing AGEs.

3. Sources of AGEs

3.1. Endogenous AGEs

Endogenous AGEs are formed intracellularly and extracellularly in all tissues and body fluids, when the normal sugar metabolism in the cells and circulating glucose form covalent adducts with plasma proteins through the process of glycation, which undergo multiple steps of the Maillard reactions described in the previous section, to produce AGEs [26,34]. The rate of endogenous AGE production is determined by genetics [35,36], age [37,38], and circulating glucose concentration [39].

3.2. Dietary-Derived (Exogenous) AGEs

There has been increasing evidence supporting the significant contribution of AGEs derived from exogenous sources, primarily diets, to the accumulation of AGEs in tissues and their circulation throughout the body. These exogenous AGEs are hence recognised as a major contributing source to the total body pool of AGEs [40,41,42]. The AGE content of the diet can vary significantly, depending on the types of foods consumed and their preparation methods. Food of animal origin and those which have been exposed to high heat or alkaline conditions during preparation are particularly high in AGEs [29,40]. High processing temperatures are required for ensuring food safety, as well as to increase the flavour and appearance of food. Consequently, dietary AGE consumption is particularly high in the modern Western diet [43,44,45]. In contrast, food prepared using cooking methods which use lower heat, shorter cooking time, and higher humidity (e.g., boiling, steaming) are associated with much lower AGE concentrations [19,40,46,47,48].
In addition to dietary AGE consumption, exposure to tobacco products can be a significant source of exogenous AGEs. Cured tobacco and tobacco smoke contain highly reactive glycation products leading to the formation of AGEs [49], which is reflected by higher circulating concentrations of AGEs in smokers compared with non-smokers [50,51,52].
In summary, the AGE concentration in the blood and tissue is dependent on dietary intake, genetics, age, and circulating glucose concentration. Furthermore, there is growing evidence that AGEs derived from multiple sources may act synergistically to contribute to a range of negative health outcomes [43]. A high total body AGE content, regardless of whether it is endogenous or dietary in origin, is associated with a wide range of negative health outcomes and with increased chronic disease burden affecting numerous sites in the body, including the gastrointestinal system.

4. AGEs, Oxidative Stress, and Inflammation in the Gut

Studies have shown that the accumulation of AGEs within the gut promotes an influx of macrophages [53,54] triggering a local inflammatory response characterised by high concentrations of pro-inflammatory cytokines and reactive oxygen species. Inflammatory bowel disease (IBD), an umbrella term which encompasses both ulcerative colitis and Crohn’s disease, is a potentially debilitating condition that presents with a range of gastrointestinal symptoms including diarrhoea, abdominal pain, and rectal bleeding [55]. Although the causes of IBD are multifactorial [56], there are currently limited studies looking at the direct effects of AGEs in IBD. However, there is evidence indicating the effects of AGEs in the worsening pathogenesis of related diseases (such as diabetes), particularly in increased inflammation in the gut, implying a mounting body of evidence that AGEs are a contributing factor to the development of IBD [57,58,59,60].

4.1. The Role of AGEs in the Pathophysiology of IBD

There is an established relationship between AGE and its receptor, the receptor for advanced glycation end-products (RAGE), signalling and IBD, primarily in the form of correlational data. For example, a study by Kato et al. [61] indicated a direct correlation between the concentration of the AGE pentosidine and that of u8-OHdG, a biomarker of oxidative DNA damage and tissue damage, in samples of inflamed gastrointestinal tissue from human subjects with IBD [61]. Furthermore, biopsy samples from people with IBD showed that RAGE-mediated NF-kB activation was higher in areas of inflamed tissue than in areas of non-inflamed gut tissue [62,63]. Of note, the accuracy of the ELISA method used by Kato et al. [61] and Andrassy et al. [62] has been questioned for the reliability of AGE measurement, considering the potential detection of additional contaminants [64,65]. Moreover, the association between RAGEs and ulcerative colitis reported by Andrassy et al. [62] is based on findings from a very small sample, indicating that further research in this area is warranted. Recently, a large prospective cohort study reported that the risk of developing IBD was positively correlated with the intake of ultra-processed foods [66,67]. Although this study did not specifically assess the AGE content of the diet, consumption of soft drinks, refined sweetened foods, and processed meats was associated with an increased hazard ratio for the development of IBD [66]. Given that these food groups either have a high AGE content (processed meats [19]) or promote the formation of AGEs through the endogenous pathway (soft drinks [68]), the total AGE (endogenous and exogenous) intake is considered higher with the ultra-processed diet [19,69]. Collectively, the data from these studies indicate that there is a potential relationship between AGE/RAGE accumulation in the gastrointestinal tract and the inflammatory symptoms of IBD.
Alongside the available human data, studies using relevant animal models have provided experimental support for an association between AGEs and IBD and provide an important insight into the mechanisms by which AGEs act to promote inflammation and oxidative stress in the gut. Importantly, animal models provide an opportunity to study the changes which occur in the gut prior to the onset of clinical disease. In this regard, Bramhall et al. [70] found that upregulated RAGE expression is a marker of colitis susceptibility, using the AKR mouse model of IBD. Another mouse model, the RAGE-knockout mouse, is protected from developing enterocolitis and colitis in the presence of the AGEs indomethacin, dextran sulphate sodium (DSS), and trinitrobenzene sulfonic acid (TNBS) [71]. Additionally, it is well established that upregulated NF-κB signalling is associated with the development of IBD [72,73,74,75] and gastrointestinal tumorigenesis [76]. Remarkably, a study by Nass et al. [77] indicated that transgenic mice expressing the firefly reporter gene under the control of an NF-κB-responsive promoter showed luciferase activity in the gut after injection of AGE-BSA. This implies a significant induction of NF-κB signalling in the gut after the ingestion of dietary AGEs proposedly due to an elevated AGE/RAGE interaction [77]. However, it should also be noted that AGE is only one of many RAGE pro-inflammatory ligands (such as S100 proteins and HMGB1), some of which bind to RAGE with stronger affinities in comparison to AGEs [78]. Particularly, increased interaction between RAGE and S100 proteins (S100A, S100A9, and S100A12) is associated with increased inflammation [79,80], contributing to worsen the pathogenesis of IBD [81,82,83,84,85,86]. Similarly, HMGB1 is another ligand binding to RAGE [80] and known to be a biomarker of IBD in numerous animal studies [87,88,89] and human studies [90,91]. There is also evidence that AGEs increase the expression of S100 proteins [92] and the activation of HMGB1 signals [93] to induce increased inflammation. These findings lend further support to the potential role of a direct AGE/RAGE interaction and an indirect interaction of AGEs and other RAGE ligands contributing to the pathogenesis of IBD. However, more studies (such as on S100 and HMGB1 knockout models) are required to have a better understanding of the important involvement of AGEs in the development of IBD.
The manipulation of dietary AGE intake and the examination of the associated effects on gastrointestinal structure and function have also been valuable for examining the relationship between AGE intake and IBD. Overall, the findings of these studies indicate that a high dietary AGE intake is associated with an increase in oxidative stress and the development of a pro-inflammatory environment consistent with IBD. For example, macrophage infiltration, inflammation, and increased oxidative stress within the colon have been reported in several studies in which rats and mice were fed a high-AGE diet [53,54,94]. Additionally, an in vitro study by van der Lugt et al. exposing human macrophage-like cells to dietary AGEs led to a significant induction of TNF-alpha secretion, which was later shown to be reduced by the addition of a RAGE antagonist (carboxymethyllysine antibodies), further supporting a relationship between high AGE intake and inflammation [64]. Moreover, there have been some reports suggesting that a high AGEs intake might directly worsen inflammation. For example, studies indicated that high consumption of dietary AGEs from an extensively hydrolysed formula by infants resulted in not only an elevated circulation of the body’s AGE pool [95] but also a significant increase in intestinal permeability compared to the consumption of a conventional formula or breast milk [96]. To date, the current literature has questioned many findings from animal and human studies due to the unreliability of the methods used for AGEs measurements. Most studies in the past predominantly measured AGE levels in foods using ELISA [61,62,97,98,99,100,101,102,103] instead of ultraperformance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS), which is currently considered the gold standard for AGE measurements [19,104]. In addition, many studies focused on producing high-AGE and low-AGE diets using high and low baking (heat-treated) methods or changing the cooking technique (boiling, poaching instead of grilling, frying) to adjust the AGE concentrations in foods. It is worth noting that changing the cooking method to alter the AGE content may lead to alterations in nutrients and caloric intake, which can potentially contribute to the effects of AGEs in IBD [99,100,101,103,105,106,107,108]

4.2. Dietary Approaches to Reducing AGE-Related Inflammation in the Gut

Given that AGE/RAGE signalling contributes to the onset and severity of inflammatory conditions of the gut, attenuating triggers of RAGE activation and/or lowering AGE accumulation have been proposed as potential treatments for IBD [71]. While there are not many reports directly investigating the immediate effects of AGEs consumption in the context of IBD, the effect of dietary AGEs in diabetes has been widely investigated [109,110]. There has been increasing evidence indicating that diabetes drives the endogenous formation of AGEs and promotes intestinal barrier disturbance and dysfunction [54,110,111,112]. As studies indicate the beneficial effects of low-AGE diets in improving diabetic complications, this might be proposed as a strategic treatment to improve IBD. For example, a human study by Luévano-Contreras et al. [103] suggested that human subjects who consumed a low-AGE diet instead of the standard diet (recommended by the American Diabetes Association) for 6 weeks had significantly lower circulating concentrations of inflammatory markers and oxidative stress. Moreover, a meta-analysis conducted by Baye et al. [113] also indicated the positive effects of low-AGE diets in reducing inflammation and oxidative stress, regardless of the diabetes status. Collectively, the data from these studies suggest diets low in AGE content might be a viable strategy for reducing inflammation in the context of IBD.
Although the majority of studies indicate pro-inflammatory effects of AGEs in the gut, it is possible that specific AGEs can be anti-inflammatory, but further research is required. For example, a human study by Dittrich et al. [114] reported that the consumption of certain high-AGE-containing foods (bread crust, dark beer, and coffee) is associated with the protective effects of increased oxidative resistance of human low-density lipoprotein in plasma. These findings highlight the potential diverse effects of AGEs and other Maillard reaction products. However, in the context of IBD, the available evidence suggests potential detrimental effects of AGEs. Nevertheless, more studies with validated methodologies (such as UPLC–MS/MS) are needed to verify the existing knowledge base of AGE measurements (which were previously detected using ELISA) and AGE implications on gut health.

5. AGEs and the Gut Barrier

The intestinal barrier is specialised to maintain homeostasis in the internal environment and to fulfil two seemingly opposing roles: (1) to maintain a peaceful co-existence with numerous intestinal symbionts without provoking chronic inflammation and (2) to provide measured inflammatory and defence mechanisms as part of the immune response [115]. The intestinal barrier is composed of a single layer of enterocytes with several intermolecular protein structures between each cell, including tight junction proteins. Tight junction proteins (e.g., claudins, occludins, tricellulin, and zona occludens family) are found towards the apical end of the enterocyte and function as a seal between neighbouring enterocytes to maintain the barrier integrity. Adherens junction proteins, in particular, E-cadherin, also play an important role in maintaining barrier integrity [116,117].
To date, there has been increasing evidence suggesting a high AGEs intake might be causative to elevated intestinal disruption, increased intestinal permeability, and high levels of inflammatory markers in the gut. AGEs and RAGE are highly expressed in the epithelial cells of the gastrointestinal tract [57,58,59]. In streptozotocin-induced diabetic rats, the detection of AGEs and RAGE significantly increased in the small and large intestines [57,58]. Furthermore, previous studies of diabetic rats showed that brush border membrane fluidity is decreased by oxidative damage and is correlated with an increase in AGE in the duodenum (28%), jejunum (94%), and ileum (58%) [118]. The increased expression of RAGE might enhance the ligation of AGEs to RAGE, consequently accelerating the localised production of reactive oxygen species (ROS). Increased expression of RAGE and ROS is known to be closely involved in disrupting the integrity of intercellular tight junctions, which can compromise the integrity of the gut barrier, leading to increased gut permeability [119]. ROS production may also increase inflammation in intestinal epithelial cells and contribute to increased gut permeability [120].
Elevated levels of AGEs have been directly implicated in increased gut permeability both in vitro and in vivo. Using an in vitro model of the human intestinal epithelium, Guibourdenche et al. [121] demonstrated reduced tight junction and mucin gene expression in the intestinal mucosa following a 6 h exposure to AGEs (carboxymethyllysine and acrylamide), with no significant changes in paracellular intestinal permeability measured by changes in FITC-Dextran efflux. In other studies by Shi et al. [122,123], the treatment of small intestinal epithelial cells (IEC-6 cells) with glycated caseinate hydrolysates led to a reduced expression of tight junction proteins and increased intestinal permeability, measured by changes in transepithelial resistance. The discrepancy in the changes in intestinal permeability may be due to the different methods of measurement (FITC-dextran efflux versus transepithelial electrical resistance), different cell models, different treatment times, as well as various AGE concentrations.
Recently, Snelson et al. [54] showed that rats fed a high AGE diet for 24 weeks had altered expression of tight junction proteins in the jejunum (reduced occludin and claudin-1; increased claudin-5), ileum (increased claudin-1 and claudin-5; reduced occludin), and colon (increased claudin-1 and claudin-5; reduced occludin). Additionally, a study by Qu et al. [124] investigated the changes in colonic permeability in rats fed a high-AGE diet. A histological examination of the colon revealed significant alterations in the colonic structure of rats fed a high AGE diet for 18 weeks, including crypt loss and distortion, reduced goblet cells, increased dysplasia, and mucosal thickening with no obvious inflammatory infiltration. The expression levels of the tight junction proteins occludin and zonula occludens-1 were also significantly decreased, suggesting increased colonic permeability [124]. Of note, in both humans and rats, an increase in serum LPS was reported following the consumption of a high-AGE diet. Specifically, Pendya et al. [125] indicated a 71% increase in plasma lipopolysaccharide (LPS) in human subjects consuming a high-AGE diet after only 4 weeks, and the previously mentioned rat study by Snelson et al. [54] also noted an increase in serum LPS after 24 weeks on a high-AGE diet. In this study, the increase in LPS was reversed by treatment with alagebrium (an AGE inhibitor) and the C5aR1 inhibitor (complement signalling inhibitor), indicating that AGE signalling may mediate this increase in LPS. However, the levels of plasma LPS in the animal study by Qu et al. [124] were modestly but not significantly increased in rats after 18 weeks of consuming a high-AGE diet. LPS are bacterial glycolipids found on the surface of most Gram-negative bacteria in the gut [126,127] and are known to be macrophage activators triggering cellular signals for chronic inflammation [128,129]. It is established that an increased level of LPS is correlated with increased intestinal permeability and epithelial damage [127]. Overall, these findings highlight the pro-inflammatory effects of AGEs in the gut and their pathogenic actions, altering the gut barrier structure and promoting gut leakiness.

6. AGEs and Enteric Neurons

The enteric nervous system (ENS) is composed of nerve plexuses, which include neurons, axons, and enteric glial cells [130]. In the intestine, most neurons are found in two ganglionated plexuses, the myenteric plexus, which is mainly involved in coordinating motility, and the submucosal plexus, which is largely involved in the control of mucosal function and blood flow [131]. There are approximately twenty different types of neurons including motor neurons, interneurons, and intrinsic primary afferent neurons (also referred to as intrinsic sensory neurons) [131]. These enteric neurons substantially form circuits capable of autonomic reflex activity to predominantly direct the functions of the gut [132,133]. Therefore, any loss or damage to enteric neurons may result in gastrointestinal dysfunction [134,135] and contribute to the pathology of many gastrointestinal diseases [136]. To date, there has been increasing evidence suggesting that an excessive AGEs intake might lead to the loss of enteric neurons and an alteration in nitrergic signalling in the ENS.
RAGE has been shown to be expressed in myenteric [58,137] and submucosal [58] neurons in the oesophagus and intestine. The expression of RAGE in the ENS is also enhanced in diabetic conditions, suggesting the involvement of the AGE/RAGE interaction in gastrointestinal-related diabetic neuropathy [58].
There is also good evidence that AGEs alter the nitrergic signalling in the ENS. Nitric oxide (NO) is a free radical involved in numerous biological functions, including vasodilation [138], transmission from inhibitory neurons to the gut muscle, modulation of neurotransmission, inhibition of platelet aggregation, and inhibition of smooth muscle proliferation [138,139]. NO is synthesised from L-arginine by the activity of nitric oxide synthases (NOS) [138,140,141]. All three isoforms of NOS have been identified in the intestine: neuronal NOS (nNOS/NOS1), endothelial NOS (eNOS/NOS3), and inducible NOS (iNOS/NOS2) [141]. NO produced by nNOS is a transmitter of the inhibitory neurons supplying the muscle of the gastrointestinal tract. A rat study by Korenaga et al. [137] indicated that AGEs suppress the expression of nNOS in vitro via RAGE. In a follow-up study, the authors showed a significant reduction in nNOS expression in the myenteric plexus of the duodenum of diabetic rats, which was reversed by inhibiting the formation of AGE using aminoguanidine (a compound established to block the formation of AGEs) and ALT-711 (a compound breaking the cross-links of AGEs) [137,142]. In line with this finding, Voukali et al. [143] demonstrated that exposure to high levels of AGEs only resulted in decreased nNOS-positive neurons in the myenteric plexus, with no effect on vasointestinal peptide- or calbindin-positive neurons.
The literature regarding the mechanisms of action of the AGE/RAGE interaction in the ENS is limited, despite the central role that the ENS plays in coordinating and regulating gut function. Furthermore, most studies have only focused on changes in nNOS expression in the myenteric plexus, while very little is known about what happens to other subtypes of enteric neurons in the myenteric and submucosal plexuses which are affected by AGEs. As discussed above, there is a correlation between high levels of AGEs and increased gut permeability. Therefore, it is possible that damage to the enteric neurons is linked to increased gut permeability, given that the enteric neurons are involved in the regulation of the intestinal barrier function [144].

7. AGEs and the Gut Microbiota

The gastrointestinal tract is host to trillions of microorganisms, collectively known as the gut microbiota. A substantial body of evidence supports the pivotal role that the gut microbiota plays in maintaining host health. A large proportion of dietary AGEs cannot be absorbed in the small intestine, and as a result, these compounds pass through to the large intestine where they may be partially degraded and metabolised by the gut microbiota [145]. It is well established that excessive AGE consumption can lead to deleterious health outcomes; therefore, it is important to understand how AGEs affect the gut microbiota composition, given that these compounds are abundantly present in the modern diet.

7.1. Human Studies

There are limited studies in humans investigating the effects of AGEs on the gut microbiome composition. One of the early human studies conducted by Seiquer et al. [146] randomised the trialing effects of a high-AGE diet on the gut microbiota composition in adolescents for 2 weeks. Their study indicated that adolescents consuming a high-AGE diet had a significantly lower level of Lactobacilli. The reduction in the relative abundance of Lactobacilli is not likely to be beneficial, because this is a Gram-positive lactic acid bacterium [147] known to play key roles in protecting the intestinal barrier [148] and in maintaining microbiota homeostasis [149,150]. Yacoub et al. [101] conducted a randomised open-label-controlled trial, looking at the association between the gut microbiota and the consumption of dietary AGEs in patients with end-stage renal disease. Their study indicated that a one-month dietary AGE restriction was associated with a decline in the relative abundance of Prevotella copri and Bifidobacterium animalis and with an increase in the relative abundance of Alistipes indistinctus, Clostridium citroniae, Clostridium hathewayi, and Ruminociccus gauvreauii [101,102,151,152]. The increase in Alistipes indistinctus is not likely beneficial, as it is recognised to be highly pathogenic due to its strong association with colitis and tumorigenesis in mice [153]. Further, the decrease in Prevotella copri may also be detrimental, as this is known to generate high levels of short-chain fatty acids (SCFAs) [102,124,149,150,154,155,156]. SCFAs are recognised to exert multiple health benefits, in particular improved intestinal barrier function [155,156,157]. Recently, a randomised controlled trial conducted by Linkens et al. [158] with a large number of participants who were abdominally obese but otherwise healthy investigated the effect of low and high AGEs intakes for 4 weeks on the gut microbiota composition. While the researchers indicated no differences in microbial diversity, richness, and overall microbiota composition between the two dietary treatments, their PERMANOVA analysis revealed that a 4-week dietary AGE restriction significantly reduced the relative abundance of Anaerostipes and Oscillibacte and increased the relative abundance of Tyzzerella, Family_XIII_UCG-001. The reduction in the relative abundance of Anaerostipes and Oscillibacter is likely to be unfavourable because Anaerostipes is a SCFA-producing genus [159] and Oscillibacter is associated with increased insulin sensitivity [160]. In contrast, the enrichment of Tyzzerella and Family_XIII_UCG-001 is identified to be associated with chronic intestinal inflammation and an increased risk of irritable bowel syndrome [161] and IBD development [162,163]. Together, the gut microbiota dysbiosis observed in these studies might suggest potential roles of dietary AGEs in markedly reshaping the microbiota profile; however, consuming a low-AGE diet in such a short period might not show protective effects in improving the gut microbiome. Moreover, these results also indicate that multiple environmental conditions and/or co-morbidities could have taken place beyond the dietary AGE consumption to reduce gut microbiota dysbiosis and improve gut health.

7.2. Animal Studies

There are numerous animal studies that investigated diets that are high in AGEs and their effects on the gut microbiota composition. Although there are conflicting findings, many studies found reduced gut microbiota diversity and richness [102,151,152], a reduced level of butyrate-producing bacteria, [102,151,157,164], and increased levels of Desulfobrivio [102,124,154], Lachnospiraceae [157] and Dubosiella [165] following exposure to high-AGE diets.
To investigate the relationship between AGE intake and the gut microbiota, Wang et al. [102] exposed mice to two high-AGE diets, i.e., a heat-treated diet and a diet enriched with exogenous AGEs for 24 weeks, and assessed the microbiome composition. This study found a reduced abundance of Bacteroidales_S24-7, Bacteroidaceae, Porphyromonadaceae, Odoribacteraceae, Lachnospiraceae, Rikenellaceae, and Erysipelotrichaceae and an increased abundance of Desulfovibrionaceae in mice which were fed high-AGE diets. They also looked at co-abundance groups (CAGs), which aggregate individual microbiome members into functional ecological units. Microbiome members were grouped together if they utilised similar resources or worked together as a functional group. The authors found that CAG1/2/3/4/5 was decreased in mice fed exogenous and dietary AGEs. Interestingly, these CAGs contained Operational Taxonomic Units from Bacteroidales_S24-7, Ruminococcaceae, and Lachnospiraceae, which are butyrate-producing bacteria. Butyrate is the major energy source of enterocytes, and a reduction in butyrate-producing bacteria can be associated with impaired epithelial barrier function and increased inflammatory and oxidative stress [102]. In addition, Qu et al. [124] showed that high-AGE diets in rats resulted in a dramatic loss of beneficial bacteria, including Ruminococcaceae, Lachnospiraceae, Alloprevotella, and Butyrivibrio. Further, an increased AGE content led to enhanced protein fermentation, as evidenced by the elevated concentration of branched-chain fatty acids (BCFAs) and ammonia in the colon. Colonic protein fermentation is deemed to be detrimental to host health due to toxic and harmful products such as phenolic, sulphur, indoxyl sulphate, and ammonia, implicating adverse effects on gut health by diminishing the energy supply to colonocytes [124]. Together, these findings highlight the pivotal role of AGEs in markedly altering the gut microbiota homeostasis and in promoting the microbiota dysbiosis.

8. Conclusions

Dietary AGEs are highly prevalent in the modern diet, with increased total endogenous and exogenous AGE formation contributing to the pathogenesis of many diseases. In this review, we provided evidence of the deleterious effects of AGEs on the gastrointestinal tract, specifically, their contribution to markedly altering the gut structure leading to increased intestinal permeability and reduced expression of enteric neurons, as well as to reshaping the microbiota composition (Figure 2). The crosstalk between AGEs and the gut signifies important influences in local and systemic effects on overall human health. However, little is known about the molecular mechanisms in the gut involved in absorbing the dietary AGEs and the potential intestinal niches influencing the gut structure and function. Additionally, reducing the AGE intake may be beneficial to improve gut health, given how prevalent AGEs are in the modern diet. Thus, more human studies with validated analytical methods (such as UPCL–MS/MS or a better gold standard for AGE measurements in the future) are needed to support the existing knowledge base of high AGE exposure to the gut, which is primarily derived from animal studies, and to determine the long-term effects of a low AGE consumption in people experiencing gastrointestinal disorders and their co-morbidities.

Author Contributions

Conceptualization, K.P.-N. and L.R.R.; writing—original draft preparation, K.P.-N., L.R.R. and B.A.M.; writing—review and editing, K.P.-N., L.R.R., B.A.M. and K.A.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Singh, R.; Barden, A.; Mori, T.; Beilin, L. Advanced glycation end-products: A review. Diabetologia 2001, 44, 129–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ge, W.; Jie, J.; Yao, J.; Li, W.; Cheng, Y.; Lu, W. Advanced glycation end products promote osteoporosis by inducing ferroptosis in osteoblasts. Mol. Med. Rep. 2022, 25, 140. [Google Scholar] [CrossRef] [PubMed]
  3. Galliera, E.; Marazzi, M.G.; Gazzaruso, C.; Gallotti, P.; Coppola, A.; Montalcini, T.; Pujia, A.; Romanelli, M.M.C. Evaluation of circulating sRAGE in osteoporosis according to BMI, adipokines and fracture risk: A pilot observational study. Immun. Ageing 2017, 14, 13. [Google Scholar] [CrossRef] [Green Version]
  4. Nakano, M.; Nakamura, Y.; Suzuki, T.; Miyazaki, A.; Takahashi, J.; Saito, M.; Shiraki, M. Pentosidine and carboxymethyl-lysine associate differently with prevalent osteoporotic vertebral fracture and various bone markers. Sci. Rep. 2020, 10, 22090. [Google Scholar] [CrossRef]
  5. Sun, K.; Semba, R.D.; Fried, L.P.; Schaumberg, D.A.; Ferrucci, L.; Varadhan, R. Elevated Serum Carboxymethyl-Lysine, an Advanced Glycation End Product, Predicts Severe Walking Disability in Older Women: The Women’s Health and Aging Study I. J. Aging Res. 2012, 2012, 586385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Eguchi, Y.; Toyoguchi, T.; Inage, K.; Fujimoto, K.; Orita, S.; Suzuki, M.; Kanamoto, H.; Abe, K.; Norimoto, M.; Umimura, T.; et al. Advanced glycation end products are associated with sarcopenia in older women: Aging marker dynamics. J. Women Aging 2021, 33, 328–340. [Google Scholar] [CrossRef] [PubMed]
  7. Yabuuchi, J.; Ueda, S.; Yamagishi, S.-I.; Nohara, N.; Nagasawa, H.; Wakabayashi, K.; Matsui, T.; Yuichiro, H.; Kadoguchi, T.; Otsuka, T.; et al. Association of advanced glycation end products with sarcopenia and frailty in chronic kidney disease. Sci. Rep. 2020, 10, 17467. [Google Scholar] [CrossRef] [PubMed]
  8. Jiang, T.; Zhang, Y.; Dai, F.; Liu, C.; Hu, H.; Zhang, Q. Advanced glycation end products and diabetes and other metabolic indicators. Diabetol. Metab. Syndr. 2022, 14, 104. [Google Scholar] [CrossRef]
  9. Indyk, D.; Bronowicka-Szydełko, A.; Gamian, A.; Kuzan, A. Advanced glycation end products and their receptors in serum of patients with type 2 diabetes. Sci. Rep. 2021, 11, 13264. [Google Scholar] [CrossRef]
  10. Koska, J.; Gerstein, H.C.; Beisswenger, P.J.; Reaven, P.D. Advanced Glycation End Products Predict Loss of Renal Function and High-Risk Chronic Kidney Disease in Type 2 Diabetes. Diabetes Care 2022, 45, 684–691. [Google Scholar] [CrossRef]
  11. Vlassara, H.; Uribarri, J. Advanced Glycation End Products (AGE) and Diabetes: Cause, Effect, or Both? Curr. Diabetes Rep. 2014, 14, 453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Nishad, R.; Tahaseen, V.; Kavvuri, R.; Motrapu, M.; Singh, A.K.; Peddi, K.; Pasupulati, A.K. Advanced-Glycation End-Products Induce Podocyte Injury and Contribute to Proteinuria. Front. Med. 2021, 8, 685447. [Google Scholar] [CrossRef] [PubMed]
  13. Miyashita, M.; Yamasaki, S.; Ando, S.; Suzuki, K.; Toriumi, K.; Horiuchi, Y.; Yoshikawa, A.; Imai, A.; Nagase, Y.; Miyano, Y.; et al. Fingertip advanced glycation end products and psychotic symptoms among adolescents. Schizophrenia 2021, 7, 37. [Google Scholar] [CrossRef] [PubMed]
  14. Kobori, A.; Miyashita, M.; Miyano, Y.; Suzuki, K.; Toriumi, K.; Niizato, K.; Oshima, K.; Imai, A.; Nagase, Y.; Yoshikawa, A.; et al. Advanced glycation end products and cognitive impairment in schizophrenia. PLoS ONE 2021, 16, e0251283. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, J.; Mooldijk, S.S.; Licher, S.; Waqas, K.; Ikram, M.K.; Uitterlinden, A.G.; Zillikens, M.C. Assessment of Advanced Glycation End Products and Receptors and the Risk of Dementia. JAMA Netw. Open 2021, 4, e2033012. [Google Scholar] [CrossRef]
  16. van Dooren, F.E.P.; Pouwer, F.; Schalkwijk, C.G.; Sep, S.J.S.; Stehouwer, C.D.A.; Henry, R.M.A.; Dagnelie, P.C.; Schaper, N.C.; van der Kallen, C.J.H.; Koster, A.; et al. Advanced Glycation End Product (AGE) Accumulation in the Skin is Associated with Depression: The Maastricht Study. Depress. Anxiety 2016, 34, 59–67. [Google Scholar] [CrossRef]
  17. Schmidt, A.M.; Du Yan, S.; Wautier, J.-L.; Stern, D. Activation of Receptor for Advanced Glycation End Products. Circ. Res. 1999, 84, 489–497. [Google Scholar] [CrossRef] [Green Version]
  18. Schleicher, E.D.; Wagner, E.; Nerlich, A.G. Increased accumulation of the glycoxidation product N(epsilon)-(carboxymethyl)lysine in human tissues in diabetes and aging. J. Clin. Investig. 1997, 99, 457–468. [Google Scholar] [CrossRef]
  19. Scheijen, J.L.; Clevers, E.; Engelen, L.; Dagnelie, P.C.; Brouns, F.; Stehouwer, C.D.; Schalkwijk, C.G. Analysis of advanced glycation endproducts in selected food items by ultra-performance liquid chromatography tandem mass spectrometry: Presentation of a dietary AGE database. Food Chem. 2016, 190, 1145–1150. [Google Scholar] [CrossRef]
  20. A Finot, P.; Magnenat, E. Metabolic transit of early and advanced Maillard products. Prog. Food Nutr. Sci. 1981, 5, 193–207. [Google Scholar]
  21. Hodge, J.E. Dehydrated Foods, Chemistry of Browning Reactions in Model Systems. J. Agric. Food Chem. 1953, 1, 928–943. [Google Scholar] [CrossRef]
  22. Zhang, Q.; Ames, J.M.; Smith, R.; Baynes, J.W.; Metz, T.O. A Perspective on the Maillard Reaction and the Analysis of Protein Glycation by Mass Spectrometry: Probing the Pathogenesis of Chronic Disease. J. Proteome Res. 2008, 8, 754–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Harohally, N.V.; Srinivas, S.M.; Umesh, S. ZnCl2-mediated practical protocol for the synthesis of Amadori ketoses. Food Chem. 2014, 158, 340–344. [Google Scholar] [CrossRef]
  24. Wu, C.-H.; Huang, S.-M.; Lin, J.-A.; Yen, G.-C. Inhibition of advanced glycation endproduct formation by foodstuffs. Food Funct. 2011, 2, 224–234. [Google Scholar] [CrossRef] [PubMed]
  25. Thornalley, P.J.; Yurek-George, A.; Argirov, O.K. Kinetics and mechanism of the reaction of aminoguanidine with the α-oxoaldehydes glyoxal, methylglyoxal, and 3-deoxyglucosone under physiological conditions. Biochem. Pharmacol. 2000, 60, 55–65. [Google Scholar] [CrossRef] [PubMed]
  26. Ott, C.; Jacobs, K.; Haucke, E.; Santos, A.N.; Grune, T.; Simm, A. Role of advanced glycation end products in cellular signaling. Redox Biol. 2014, 2, 411–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Deluyker, D.; Evens, L.; Bito, V. Advanced glycation end products (AGEs) and cardiovascular dysfunction: Focus on high molecular weight AGEs. Amino Acids 2017, 49, 1535–1541. [Google Scholar] [CrossRef] [PubMed]
  28. Fu, M.-X.; Requena, J.R.; Jenkins, A.J.; Lyons, T.J.; Baynes, J.W.; Thorpe, S.R. The Advanced Glycation End Product, Nepsilon-(Carboxymethyl)lysine, Is a Product of both Lipid Peroxidation and Glycoxidation Reactions. J. Biol. Chem. 1996, 271, 9982–9986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Nowotny, K.; Jung, T.; Höhn, A.; Weber, D.; Grune, T.; Nowotny, K.; Jung, T.; Höhn, A.; Weber, D.; Grune, T. Advanced Glycation End Products and Oxidative Stress in Type 2 Diabetes Mellitus. Biomolecules 2015, 5, 194–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Dhar, I.; Prasad, K. Oxidative Stress as a Mechanism of Added Sugar-Induced Cardiovascular Disease. Int. J. Angiol. 2014, 23, 217–226. [Google Scholar] [CrossRef] [Green Version]
  31. Thornalley, P.J.; Langborg, A.; Minhas, H.S. Formation of glyoxal, methylglyoxal and 3-deoxyglucosone in the glycation of proteins by glucose. Biochem. J. 1999, 344, 109–116. [Google Scholar] [CrossRef] [PubMed]
  32. Wells-Knecht, K.J.; Brinkmann, E.; Baynes, J.W. Characterization of an Imidazolium Salt Formed from Glyoxal and N.alpha.-Hippuryllysine: A Model for Maillard Reaction Crosslinks in Proteins. J. Org. Chem. 1995, 60, 6246–6247. [Google Scholar] [CrossRef]
  33. Lorenzi, M. The Polyol Pathway as a Mechanism for Diabetic Retinopathy: Attractive, Elusive, and Resilient. Exp. Diabetes Res. 2007, 2007, 61038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kellow, N.J.; Savige, G.S. Dietary advanced glycation end-product restriction for the attenuation of insulin resistance, oxidative stress and endothelial dysfunction: A systematic review. Eur. J. Clin. Nutr. 2013, 67, 239–248. [Google Scholar] [CrossRef] [Green Version]
  35. Leslie, R.D.G.; Beyan, H.; Sawtell, P.; Boehm, B.O.; Spector, T.D.; Snieder, H. Level of an Advanced Glycated End Product Is Genetically Determined. Diabetes 2003, 52, 2441–2444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Pugliese, G.; Pricci, F.; Iacobini, C.; Leto, G.; Amadio, L.; Barsotti, P.; Frigeri, L.; Hsu, D.K.; Vlassara, H.; Liu, F.-T.; et al. Accelerated diabetic glomerulopathy in galectin-3/AGE receptor 3 knockout mice. FASEB J. 2001, 15, 2471–2479. [Google Scholar] [CrossRef] [Green Version]
  37. Gkogkolou, P.; Böhm, M. Advanced glycation end products. Dermato-Endocrinol. 2012, 4, 259–270. [Google Scholar] [CrossRef] [Green Version]
  38. Much, G.; Thome, J.; Foley, P.; Schinzel, R.; Riederer, P. AGEs in aging and Alzheimers disease. Brain Res. Rev. 1997, 23, 6. [Google Scholar]
  39. Khan, N.; Bakshi, K.S.; Jaggi, A.S.; Singh, N. Ameliorative Potential of Spironolactone in Diabetes Induced Hyperalgesia in Mice. YAKUGAKU ZASSHI 2009, 129, 593–599. [Google Scholar] [CrossRef] [Green Version]
  40. Uribarri, J.; Woodruff, S.; Goodman, S.; Cai, W.; Chen, X.; Pyzik, R.; Yong, A.; Striker, G.E.; Vlassara, H. Advanced Glycation End Products in Foods and a Practical Guide to Their Reduction in the Diet. J. Am. Diet. Assoc. 2010, 110, 911–916.e12. [Google Scholar] [CrossRef] [Green Version]
  41. Uribarri, J.; Del Castillo, M.D.; De La Maza, M.P.; Filip, R.; Gugliucci, A.; Luevano-Contreras, C.; Macías-Cervantes, M.H.; Bastos, D.H.M.; Medrano, A.; Menini, T.; et al. Dietary Advanced Glycation End Products and Their Role in Health and Disease. Adv. Nutr. Int. Rev. J. 2015, 6, 461–473. [Google Scholar] [CrossRef] [Green Version]
  42. Hechtman, L. 209—Polycystic Ovary Syndrome (PCOS). In Textbook of Natural Medicine, 5th ed.; Pizzorno, J.E., Murray, M.T., Eds.; Churchill Livingstone: St. Louis, MO, USA, 2020; pp. 1694–1706.e7. [Google Scholar]
  43. Delgado-Andrade, C. Carboxymethyl-lysine: Thirty years of investigation in the field of AGE formation. Food Funct. 2015, 7, 46–57. [Google Scholar] [CrossRef]
  44. Bettiga, A.; Fiorio, F.; Di Marco, F.; Trevisani, F.; Romani, A.; Porrini, E.; Salonia, A.; Montorsi, F.; Vago, R. The Modern Western Diet Rich in Advanced Glycation End-Products (AGEs): An Overview of Its Impact on Obesity and Early Progression of Renal Pathology. Nutrients 2019, 11, 1748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Cordain, L.; Eaton, S.B.; Sebastian, A.; Mann, N.; Lindeberg, S.; Watkins, B.A.; O’Keefe, J.H.; Brand-Miller, J. Origins and evolution of the Western diet: Health implications for the 21st century. Am. J. Clin. Nutr. 2005, 81, 341–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Helou, C.; Gadonna-Wideham, P.; Robert, N.; Branlard, G.; Thebault, J.; Librere, S.; Jacquot, S.; Mardon, J.; Piquet-Pissaloux, A.; Chapron, S.; et al. The impact of raw materials and baking conditions on Maillard reaction products, thiamine, folate, phytic acid and minerals in white bread. Food Funct. 2016, 7, 2498–2507. [Google Scholar] [CrossRef] [PubMed]
  47. Hull, G.L.; Woodside, J.V.; Ames, J.M.; Cuskelly, G.J. Nε-(carboxymethyl)lysine content of foods commonly consumed in a Western style diet. Food Chem. 2012, 131, 170–174. [Google Scholar] [CrossRef]
  48. Pérez-Burillo, S.; Rajakaruna, S.; Pastoriza, S.; Paliy, O.; Rufián-Henares, J. Bioactivity of food melanoidins is mediated by gut microbiota. Food Chem. 2020, 316, 126309. [Google Scholar] [CrossRef]
  49. Cerami, C.; Founds, H.; Nicholl, I.; Mitsuhashi, T.; Giordano, D.; Vanpatten, S.; Lee, A.; Al-Abed, Y.; Vlassara, H.; Bucala, R.; et al. Tobacco smoke is a source of toxic reactive glycation products. Proc. Natl. Acad. Sci. USA 1997, 94, 13915–13920. [Google Scholar] [CrossRef] [Green Version]
  50. Berg, T.J.; Snorgaard, O.; Faber, J.; A Torjesen, P.; Hildebrandt, P.; Mehlsen, J.; Hanssen, K.F. Serum levels of advanced glycation end products are associated with left ventricular diastolic function in patients with type 1 diabetes. Diabetes Care 1999, 22, 1186–1190. [Google Scholar] [CrossRef]
  51. Gopal, P.; Reynaert, N.L.; Scheijen, J.L.J.M.; Engelen, L.; Schalkwijk, C.G.; Franssen, F.M.; Wouters, E.F.; Rutten, E.P. Plasma advanced glycation end-products and skin autofluorescence are increased in COPD. Eur. Respir. J. 2013, 43, 430–438. [Google Scholar] [CrossRef] [Green Version]
  52. Nicholl, I.D.; Stitt, A.W.; Moore, J.E.; Ritchie, A.J.; Archer, D.B.; Bucala, R. Increased Levels of Advanced Glycation Endproducts in the Lenses and Blood Vessels of Cigarette Smokers. Mol. Med. 1998, 4, 594–601. [Google Scholar] [CrossRef] [Green Version]
  53. Kehm, R.; Rückriemen, J.; Weber, D.; Deubel, S.; Grune, T.; Höhn, A. Endogenous advanced glycation end products in pancreatic islets after short-term carbohydrate intervention in obese, diabetes-prone mice. Nutr. Diabetes 2019, 9, 9. [Google Scholar] [CrossRef]
  54. Snelson, M.; Tan, S.M.; Clarke, R.E.; de Pasquale, C.; Thallas-Bonke, V.; Nguyen, T.-V.; Penfold, S.A.; Harcourt, B.E.; Sourris, K.C.; Lindblom, R.S.; et al. Processed foods drive intestinal barrier permeability and microvascular diseases. Sci. Adv. 2021, 7, eabe4841. [Google Scholar] [CrossRef] [PubMed]
  55. Hendrickson, B.A.; Gokhale, R.; Cho, J.H. Clinical Aspects and Pathophysiology of Inflammatory Bowel Disease. Clin. Microbiol. Rev. 2002, 15, 79–94. [Google Scholar] [CrossRef] [Green Version]
  56. Loddo, I.; Romano, C. Inflammatory Bowel Disease: Genetics, Epigenetics, and Pathogenesis. Front. Immunol. 2015, 6, 551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Chen, P.; Zhao, J.; Gregersen, H. Up-Regulated Expression of Advanced Glycation End-Products and Their Receptor in the Small Intestine and Colon of Diabetic Rats. Dig. Dis. Sci. 2011, 57, 48–57. [Google Scholar] [CrossRef]
  58. Chen, P.-M.; Gregersen, H.; Zhao, J.-B. Advanced glycation end-product expression is upregulated in the gastrointestinal tract of type 2 diabetic rats. World J. Diabetes 2015, 6, 662–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Ling, X.; Nagai, R.; Sakashita, N.; Takeya, M.; Horiuchi, S.; Takahashi, K. Immunohistochemical Distribution and Quantitative Biochemical Detection of Advanced Glycation End Products in Fetal to Adult Rats and in Rats with Streptozotocin-Induced Diabetes. Lab. Investig. 2001, 81, 845–861. [Google Scholar] [CrossRef] [Green Version]
  60. Sparvero, L.J.; Asafu-Adjei, D.; Kang, R.; Tang, D.; Amin, N.; Im, J.; Rutledge, R.; Lin, B.; A Amoscato, A.; Zeh, H.J.; et al. RAGE (Receptor for Advanced Glycation Endproducts), RAGE Ligands, and their role in Cancer and Inflammation. J. Transl. Med. 2009, 7, 17. [Google Scholar] [CrossRef] [Green Version]
  61. Kato, S.; Itoh, K.; Ochiai, M.; Iwai, A.; Park, Y.; Hata, S.; Takeuchi, K.; Ito, M.; Imaki, J.; Miura, S.; et al. Increased pentosidine, an advanced glycation end-product, in urine and tissue reflects disease activity in inflammatory bowel diseases. J. Gastroenterol. Hepatol. 2008, 23, S140–S145. [Google Scholar] [CrossRef]
  62. Andrassy, M.; Igwe, J.; Autschbach, F.; Volz, C.; Remppis, A.; Neurath, M.F.; Schleicher, E.; Humpert, P.M.; Wendt, T.; Liliensiek, B.; et al. Posttranslationally Modified Proteins as Mediators of Sustained Intestinal Inflammation. Am. J. Pathol. 2006, 169, 1223–1237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Ciccocioppo, R. Role of the advanced glycation end products receptor in Crohn’s disease inflammation. World J. Gastroenterol. 2013, 19, 8269–8281. [Google Scholar] [CrossRef]
  64. van der Lugt, T.; Weseler, A.R.; Gebbink, W.A.; Vrolijk, M.F.; Opperhuizen, A.; Bast, A. Dietary Advanced Glycation Endproducts Induce an Inflammatory Response in Human Macrophages in Vitro. Nutrients 2018, 10, 1868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Tessier, F. The Maillard reaction in the human body. The main discoveries and factors that affect glycation. Pathol. Biol. 2010, 58, 214–219. [Google Scholar] [CrossRef]
  66. Narula, N.; Wong, E.C.L.; Dehghan, M.; Mente, A.; Rangarajan, S.; Lanas, F.; Lopez-Jaramillo, P.; Rohatgi, P.; Lakshmi, P.V.M.; Varma, R.P.; et al. Association of ultra-processed food intake with risk of inflammatory bowel disease: Prospective cohort study. BMJ 2021, 374, n1554. [Google Scholar] [CrossRef]
  67. Chen, X.; Zhang, Z.; Yang, H.; Qiu, P.; Wang, H.; Wang, F.; Zhao, Q.; Fang, J.; Nie, J. Consumption of ultra-processed foods and health outcomes: A systematic review of epidemiological studies. Nutr. J. 2020, 19, 86. [Google Scholar] [CrossRef]
  68. Ottum, M.S.; Mistry, A.M. Advanced glycation end-products: Modifiable environmental factors profoundly mediate insulin resistance. J. Clin. Biochem. Nutr. 2015, 57, 1–12. [Google Scholar] [CrossRef] [Green Version]
  69. Boșca, A.B.; Mihu, C.M.; Ilea, A. Advanced Glycation End Products as Biomarkers in Nutrition; Springer: Berlin/Heidelberg, Germany, 2022; pp. 1–23. [Google Scholar] [CrossRef]
  70. Bramhall, M.; Rich, K.; Chakraborty, A.; Logunova, L.; Han, N.; Wilson, J.; McLaughlin, J.; Brass, A.; Cruickshank, S.M. Differential Expression of Soluble Receptor for Advanced Glycation End-products in Mice Susceptible or Resistant to Chronic Colitis. Inflamm. Bowel Dis. 2019, 26, 360–368. [Google Scholar] [CrossRef] [Green Version]
  71. Body-Malapel, M.; Djouina, M.; Waxin, C.; Langlois, A.; Gower-Rousseau, C.; Zerbib, P.; Schmidt, A.-M.; Desreumaux, P.; Boulanger, E.; Vignal, C. The RAGE signaling pathway is involved in intestinal inflammation and represents a promising therapeutic target for Inflammatory Bowel Diseases. Mucosal Immunol. 2019, 12, 468–478. [Google Scholar] [CrossRef]
  72. McDaniel, D.; Eden, K.; Ringel, V.M.; Allen, I.C. Emerging Roles for Noncanonical NF-κB Signaling in the Modulation of Inflammatory Bowel Disease Pathobiology. Inflamm. Bowel Dis. 2016, 22, 2265–2279. [Google Scholar] [CrossRef] [Green Version]
  73. Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
  74. Rogler, G.; Brand, K.; Vogl, D.; Page, S.; Hofmeister, R.; Andus, T.; Knuechel, R.; Baeuerle∥, P.A.; Schölmerich, J.; Gross, V. Nuclear factor κB is activated in macrophages and epithelial cells of inflamed intestinal mucosa. Gastroenterology 1998, 115, 357–369. [Google Scholar] [CrossRef] [PubMed]
  75. Schreiber, S.; Nikolaus, S.; Hampe, J. Activation of nuclear factor κB in inflammatory bowel disease. Gut 1998, 42, 477–484. [Google Scholar] [CrossRef] [PubMed]
  76. Peng, C.; Ouyang, Y.; Lu, N.; Li, N. The NF-κB Signaling Pathway, the Microbiota, and Gastrointestinal Tumorigenesis: Recent Advances. Front. Immunol. 2020, 11, 1387. [Google Scholar] [CrossRef]
  77. Nass, N.; Bayreuther, K.; Simm, A. Systemic activation of NF-κB driven luciferase activity in transgenic mice fed advanced glycation end products modified albumin. Glycoconj. J. 2017, 34, 157–161. [Google Scholar] [CrossRef] [PubMed]
  78. Leclerc, E.; Fritz, G.; Vetter, S.W.; Heizmann, C.W. Binding of S100 proteins to RAGE: An update. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2009, 1793, 993–1007. [Google Scholar] [CrossRef] [Green Version]
  79. Zwadlo, G.; Brüggen, J.; Gerhards, G.; Schlegel, R.; Sorg, C. Two calcium-binding proteins associated with specific stages of myeloid cell differentiation are expressed by subsets of macrophages in inflammatory tissues. Clin. Exp. Immunol. 1988, 72, 510–515. [Google Scholar]
  80. Xia, C.; Braunstein, Z.; Toomey, A.C.; Zhong, J.; Rao, X. S100 Proteins As an Important Regulator of Macrophage Inflammation. Front. Immunol. 2018, 8, 1908. [Google Scholar] [CrossRef] [Green Version]
  81. Foell, D.; Wittkowski, H.; Ren, Z.; Turton, J.; Pang, G.; Daebritz, J.; Ehrchen, J.; Heidemann, J.; Borody, T.; Roth, J.; et al. Phagocyte-specific S100 proteins are released from affected mucosa and promote immune responses during inflammatory bowel disease. J. Pathol. 2008, 216, 183–192. [Google Scholar] [CrossRef]
  82. Leach, S.T.; Yang, Z.; Messina, I.; Song, C.; Geczy, C.L.; Cunningham, A.M.; Day, A.S. Serum and mucosal S100 proteins, calprotectin (S100A8/S100A9) and S100A12, are elevated at diagnosis in children with inflammatory bowel disease. Scand. J. Gastroenterol. 2007, 42, 1321–1331. [Google Scholar] [CrossRef] [PubMed]
  83. Cirillo, C.; Sarnelli, G.; Esposito, G.; Grosso, M.; Petruzzelli, R.; Izzo, P.; Calì, G.; D’Armiento, F.P.; Rocco, A.; Naradone, G.; et al. Increased mucosal nitric oxide production in ulcerative colitis is mediated in part by the enteroglial-derived S100B protein. Neurogastroenterol. Motil. 2009, 21, 1209-e112. [Google Scholar] [CrossRef] [PubMed]
  84. Esposito, G.; Cirillo, C.; Sarnelli, G.; De Filippis, D.; D’Armiento, F.P.; Rocco, A.; Nardone, G.; Petruzzelli, R.; Grosso, M.; Izzo, P.; et al. Enteric Glial-Derived S100B Protein Stimulates Nitric Oxide Production in Celiac Disease. Gastroenterology 2007, 133, 918–925. [Google Scholar] [CrossRef]
  85. Cirillo, C. S100B protein in the gut: The evidence for enteroglial-sustained intestinal inflammation. World J. Gastroenterol. 2011, 17, 1261–1266. [Google Scholar] [CrossRef]
  86. Srikrishna, G.; Turovskaya, O.; Shaikh, R.; Newlin, R.; Foell, D.; Murch, S.; Kronenberg, M.; Freeze, H.H. Carboxylated glycans mediate colitis through activation of NF-κB. J. Immunol. 2005, 175, 5412–5422. [Google Scholar] [CrossRef] [Green Version]
  87. Palone, F.; Vitali, R.; Cucchiara, S.; Pierdomenico, M.; Negroni, A.; Aloi, M.; Nuti, F.; Felice, C.; Armuzzi, A.; Stronati, L. Role of HMGB1 as a Suitable Biomarker of Subclinical Intestinal Inflammation and Mucosal Healing in Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2014, 20, 1448–1457. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, X.; Li, L.; Khan, M.N.; Shi, L.; Wang, Z.; Zheng, F.; Gong, F.; Fang, M. HMGB1 exacerbates experimental mouse colitis by enhancing innate lymphoid cells 3 inflammatory responses via promoted IL-23 production. J. Endotoxin Res. 2016, 22, 696–705. [Google Scholar] [CrossRef]
  89. Stavely, R.; Sahakian, L.; Filippone, R.T.; Stojanovska, V.; Bornstein, J.C.; Sakkal, S.; Nurgali, K. Oxidative Stress-Induced HMGB1 Translocation in Myenteric Neurons Contributes to Neuropathy in Colitis. Biomolecules 2022, 12, 1831. [Google Scholar] [CrossRef]
  90. Chen, Y.; Wu, D.; Sun, L. Clinical Significance of High-Mobility Group Box 1 Protein (HMGB1) and Nod-Like Receptor Protein 3 (NLRP3) in Patients with Ulcerative Colitis. J. Pharmacol. Exp. Ther. 2020, 26, e919530. [Google Scholar] [CrossRef] [PubMed]
  91. Palone, F.; Vitali, R.; Cucchiara, S.; Mennini, M.; Armuzzi, A.; Pugliese, D.; D’incà, R.; Barberio, B.; Stronati, L. Fecal HMGB1 Reveals Microscopic Inflammation in Adult and Pediatric Patients with Inflammatory Bowel Disease in Clinical and Endoscopic Remission. Inflamm. Bowel Dis. 2016, 22, 2886–2893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Nakajima, Y.; Inagaki, Y.; Kido, J.; Nagata, T. Advanced glycation end products increase expression of S100A8 and A9viaRAGE-MAPK in rat dental pulp cells. Oral Dis. 2014, 21, 328–334. [Google Scholar] [CrossRef] [PubMed]
  93. Watanabe, M.; Toyomura, T.; Tomiyama, M.; Wake, H.; Liu, K.; Teshigawara, K.; Takahashi, H.; Nishibori, M.; Mori, S. Advanced glycation end products (AGEs) synergistically potentiated the proinflammatory action of lipopolysaccharide (LPS) and high mobility group box-1 (HMGB1) through their direct interactions. Mol. Biol. Rep. 2020, 47, 7153–7159. [Google Scholar] [CrossRef]
  94. Shangari, N.; Depeint, F.; Furrer, R.; Bruce, W.R.; Popovic, M.; Zheng, F.; O’Brien, P.J. A thermolyzed diet increases oxidative stress, plasma α-aldehydes and colonic inflammation in the rat. Chem.-Biol. Interact. 2007, 169, 100–109. [Google Scholar] [CrossRef] [PubMed]
  95. Šebeková, K.; Saavedra, G.; Zumpe, C.; Somoza, V.; Klenovicsová, K.; Birlouez-Aragon, I. Plasma Concentration and Urinary Excretion of Nɛ-(Carboxymethyl)lysine in Breast Milk- and Formula-fed Infants. Ann. N. Y. Acad. Sci. 2008, 1126, 177–180. [Google Scholar] [CrossRef]
  96. Siljander, H.; Jason, E.; Ruohtula, T.; Selvenius, J.; Koivusaari, K.; Salonen, M.; Ahonen, S.; Honkanen, J.; Ilonen, J.; Vaarala, O.; et al. Effect of Early Feeding on Intestinal Permeability and Inflammation Markers in Infants with Genetic Susceptibility to Type 1 Diabetes: A Randomized Clinical Trial. J. Pediatr. 2021, 238, 305–311.e3. [Google Scholar] [CrossRef]
  97. Goldberg, T.; Cai, W.; Peppa, M.; Dardaine, V.; Baliga, B.S.; Uribarri, J.; Vlassara, H. Advanced glycoxidation end products in commonly consumed foods. J. Am. Diet. Assoc. 2004, 104, 1287–1291. [Google Scholar] [CrossRef] [PubMed]
  98. Uribarri, J.; Cai, W.; Peppa, M.; Goodman, S.; Ferruci, L.; Striker, G.; Vlassara, H. Circulating glycotoxins and dietary advanced glycation endproducts: Two links to inflammatory response, oxidative stress, and aging. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2007, 62, 427–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Vlassara, H.; Cai, W.; Tripp, E.; Pyzik, R.; Yee, K.; Goldberg, L.; Tansman, L.; Chen, X.; Mani, V.; Fayad, Z.A.; et al. Oral AGE restriction ameliorates insulin resistance in obese individuals with the metabolic syndrome: A randomised controlled trial. Diabetologia 2016, 59, 2181–2192. [Google Scholar] [CrossRef] [Green Version]
  100. Uribarri, J.; Cai, W.; Ramdas, M.; Goodman, S.; Pyzik, R.; Chen, X.; Zhu, L.; Striker, G.E.; Vlassara, H. Restriction of Advanced Glycation End Products Improves Insulin Resistance in Human Type 2 Diabetes. Diabetes Care 2011, 34, 1610–1616. [Google Scholar] [CrossRef] [Green Version]
  101. Yacoub, R.; Nugent, M.; Cai, W.; Nadkarni, G.N.; Chaves, L.D.; Abyad, S.; Honan, A.M.; Thomas, S.A.; Zheng, W.; Valiyaparambil, S.A.; et al. Advanced glycation end products dietary restriction effects on bacterial gut microbiota in peritoneal dialysis patients; a randomized open label controlled trial. PLoS ONE 2017, 12, e0184789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Wang, J.; Cai, W.; Yu, J.; Liu, H.; He, S.; Zhu, L.; Xu, J. Dietary Advanced Glycation End Products Shift the Gut Microbiota Composition and Induce Insulin Resistance in Mice. Diabetes Metab. Syndr. Obesity Targets Ther. 2022, 2022, 427–437. [Google Scholar] [CrossRef]
  103. Luévano-Contreras, C.; Garay-Sevilla, M.E.; Wrobel, K.; Malacara, J.M.; Wrobel, K. Dietary advanced glycation end products restriction diminishes inflammation markers and oxidative stress in patients with type 2 diabetes mellitus. J. Clin. Biochem. Nutr. 2013, 52, 22–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Linkens, A.M.; Houben, A.J.; Niessen, P.M.; Wijckmans, N.E.; de Goei, E.E.; Eynde, M.D.V.D.; Scheijen, J.L.; van den Waarenburg, M.P.; Mari, A.; Berendschot, T.T.; et al. A 4-week high-AGE diet does not impair glucose metabolism and vascular function in obese individuals. J. Clin. Investig. 2022, 7, e156950. [Google Scholar] [CrossRef] [PubMed]
  105. Mark, A.B.; Poulsen, M.W.; Andersen, S.; Andersen, J.M.; Bak, M.J.; Ritz, C.; Holst, J.J.; Nielsen, J.; de Courten, B.; Dragsted, L.O.; et al. Consumption of a Diet Low in Advanced Glycation End Products for 4 Weeks Improves Insulin Sensitivity in Overweight Women. Diabetes Care 2013, 37, 88–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Ott, B.; Skurk, T.; Hastreiter, L.; Lagkouvardos, I.; Fischer, S.; Büttner, J.; Kellerer, T.; Clavel, T.; Rychlik, M.; Haller, D.; et al. Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women. Sci. Rep. 2017, 7, 11955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Adolph, T.E.; Zhang, J. Diet fuelling inflammatory bowel diseases: Preclinical and clinical concepts. Gut 2022, 71, 2574–2586. [Google Scholar] [CrossRef]
  108. Sbierski-Kind, J.; Grenkowitz, S.; Schlickeiser, S.; Sandforth, A.; Friedrich, M.; Kunkel, D.; Glauben, R.; Brachs, S.; Mai, K.; Thürmer, A.; et al. Effects of caloric restriction on the gut microbiome are linked with immune senescence. Microbiome 2022, 10, 57. [Google Scholar] [CrossRef]
  109. Singh, V.P.; Bali, A.; Singh, N.; Jaggi, A.S. Advanced Glycation End Products and Diabetic Complications. Korean J. Physiol. Pharmacol. 2014, 18, 1–14. [Google Scholar] [CrossRef] [Green Version]
  110. Snelson, M.; Lucut, E.; Coughlan, M.T. The Role of AGE-RAGE Signalling as a Modulator of Gut Permeability in Diabetes. Int. J. Mol. Sci. 2022, 23, 1766. [Google Scholar] [CrossRef]
  111. Foell, D.; Kucharzik, T.; Kraft, M.; Vogl, T.; Sorg, C.; Domschke, W.; Roth, J. Neutrophil derived human S100A12 (EN-RAGE) is strongly expressed during chronic active inflammatory bowel disease. Gut 2003, 52, 847–853. [Google Scholar] [CrossRef] [Green Version]
  112. Thaiss, C.A.; Levy, M.; Grosheva, I.; Zheng, D.; Soffer, E.; Blacher, E.; Braverman, S.; Tengeler, A.C.; Barak, O.; Elazar, M.; et al. Hyperglycemia drives intestinal barrier dysfunction and risk for enteric infection. Science 2018, 359, 1376–1383. [Google Scholar] [CrossRef] [Green Version]
  113. Baye, E.; Kiriakova, V.; Uribarri, J.; Moran, L.J.; de Courten, B. Consumption of diets with low advanced glycation end products improves cardiometabolic parameters: Meta-analysis of randomised controlled trials. Sci. Rep. 2017, 7, 2266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Dittrich, R.; Dragonas, C.; Kannenkeril, D.; Hoffmann, I.; Mueller, A.; Beckmann, M.W.; Pischetsrieder, M. A diet rich in Maillard reaction products protects LDL against copper induced oxidation ex vivo, a human intervention trial. Food Res. Int. 2009, 42, 1315–1322. [Google Scholar] [CrossRef]
  115. Bischoff, S.C.; Barbara, G.; Buurman, W.; Ockhuizen, T.; Schulzke, J.-D.; Serino, M.; Tilg, H.; Watson, A.; Wells, J.M. Intestinal permeability—A new target for disease prevention and therapy. BMC Gastroenterol. 2014, 14, 189. [Google Scholar] [CrossRef] [Green Version]
  116. Buckley, A.; Turner, J.R. Cell Biology of Tight Junction Barrier Regulation and Mucosal Disease. Cold Spring Harb. Perspect. Biol. 2017, 10, a029314. [Google Scholar] [CrossRef]
  117. Assimakopoulos, S.F.; Papageorgiou, I.; Charonis, A. Enterocytes’ tight junctions: From molecules to diseases. World J. Gastrointest. Pathophysiol. 2011, 2, 123–137. [Google Scholar] [CrossRef]
  118. Bhor, V.; Sivakami, S. Regional variations in intestinal brush border membrane fluidity and function during diabetes and the role of oxidative stress and non-enzymatic glycation. Mol. Cell. Biochem. 2003, 252, 125–132. [Google Scholar] [CrossRef]
  119. Kim, K.A.; Jung, J.H.; Kang, I.G.; Choi, Y.S.; Kim, S.T. ROS Is Involved in Disruption of Tight Junctions of Human Nasal Epithelial Cells Induced by HRV16. Laryngoscope 2018, 128, E393–E401. [Google Scholar] [CrossRef] [PubMed]
  120. Kellow, N.; Coughlan, M.T. Effect of diet-derived advanced glycation end products on inflammation. Nutr. Rev. 2015, 73, 737–759. [Google Scholar] [CrossRef]
  121. Guibourdenche, M.; Haug, J.; Chevalier, N.; Spatz, M.; Barbezier, N.; Gay-Quéheillard, J.; Anton, P.M. Food Contaminants Effects on an In Vitro Model of Human Intestinal Epithelium. Toxics 2021, 9, 135. [Google Scholar] [CrossRef]
  122. Shi, J.; Zhao, X.-H. Influence of the Maillard-type caseinate glycation with lactose on the intestinal barrier activity of the caseinate digest in IEC-6 cells. Food Funct. 2019, 10, 2010–2021. [Google Scholar] [CrossRef]
  123. Shi, J.; Fu, Y.; Zhao, X.; Lametsch, R. Glycation sites and bioactivity of lactose-glycated caseinate hydrolysate in lipopolysaccharide-injured IEC-6 cells. J. Dairy Sci. 2021, 104, 1351–1363. [Google Scholar] [CrossRef]
  124. Qu, W.; Yuan, X.; Zhao, J.; Zhang, Y.; Hu, J.; Wang, J.; Li, J. Dietary advanced glycation end products modify gut microbial composition and partially increase colon permeability in rats. Mol. Nutr. Food Res. 2017, 61, 1700118. [Google Scholar] [CrossRef]
  125. Pendyala, S.; Walker, J.M.; Holt, P.R. A High-Fat Diet Is Associated With Endotoxemia That Originates From the Gut. Gastroenterology 2012, 142, 1100–1101.e2. [Google Scholar] [CrossRef] [Green Version]
  126. Bertani, B.; Ruiz, N. Function and Biogenesis of Lipopolysaccharides. EcoSal Plus 2018, 8. [Google Scholar] [CrossRef]
  127. Candelli, M.; Franza, L.; Pignataro, G.; Ojetti, V.; Covino, M.; Piccioni, A.; Gasbarrini, A.; Franceschi, F. Interaction between Lipopolysaccharide and Gut Microbiota in Inflammatory Bowel Diseases. Int. J. Mol. Sci. 2021, 22, 6242. [Google Scholar] [CrossRef]
  128. Kitaura, A.; Nishinaka, T.; Hamasaki, S.; Hatipoglu, O.F.; Wake, H.; Nishibori, M.; Mori, S.; Nakao, S.; Takahashi, H. Advanced glycation end-products reduce lipopolysaccharide uptake by macrophages. PLoS ONE 2021, 16, e0245957. [Google Scholar] [CrossRef]
  129. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef]
  130. Furness, J.B.; Nguyen, T.V.; Nurgali, K.; Shimizu, Y. The Enteric Nervous System and Its Extrinsic Connections. In Textbook of Gastroenrerology; Wiley: Hoboken, NJ, USA, 2008; pp. 15–39. [Google Scholar] [CrossRef]
  131. Furness, J.B. The Enteric Nervous System; John Wiley & Sons: Hoboken, NJ, USA, 2008. [Google Scholar]
  132. Furness, J.B. Enteric Nervous System. In Encyclopedia of Neuroscience; Binder, M.D., Hirokawa, N., Windhorst, U., Eds.; Springer: Berlin/Heidelberg, Germany, 2009; pp. 1122–1125. [Google Scholar]
  133. Furness, J.B.; Callaghan, B.P.; Rivera, L.R.; Cho, H.-J. The Enteric Nervous System and Gastrointestinal Innervation: Integrated Local and Central Control. In Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Health and Disease; Springer: Berlin, Germany, 2014; Volume 817, pp. 39–71. [Google Scholar] [CrossRef]
  134. Aube, A.-C.; Cabarrocas, J.; Bauer, J.; Philippe, D.; Aubert, P.; Doulay, F.; Liblau, R.; Galmiche, J.P.; Neunlist, M. Changes in enteric neurone phenotype and intestinal functions in a transgenic mouse model of enteric glia disruption. Gut 2006, 55, 630–637. [Google Scholar] [CrossRef] [Green Version]
  135. Schemann, M.; Michel, K.; Ceregrzyn, M.; Zeller, F.; Seidl, S.; Bischoff, S.C. Human mast cell mediator cocktail excites neurons in human and guinea-pig enteric nervous system. Neurogastroenterol. Motil. 2004, 17, 281–289. [Google Scholar] [CrossRef]
  136. De Giorgio, R.; Bianco, F.; Latorre, R.; Caio, G.; Clavenzani, P.; Bonora, E. Enteric neuropathies: Yesterday, Today and Tomorrow. Enteric Nerv. Syst. 2016, 891, 123–133. [Google Scholar] [CrossRef]
  137. Korenaga, K.; Micci, M.-A.; Taglialatela, G.; Pasricha, P.J. Suppression of nNOS expression in rat enteric neurones by the receptor for advanced glycation end-products. Neurogastroenterol. Motil. 2006, 18, 392–400. [Google Scholar] [CrossRef]
  138. Moro, M.; Cárdenas, A.; Hurtado, O.; Leza, J.; Lizasoain, I. Role of nitric oxide after brain ischaemia. Cell Calcium 2004, 36, 265–275. [Google Scholar] [CrossRef] [PubMed]
  139. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Love, S. Oxidative Stress in Brain Ischemia. Brain Pathol. 2006, 9, 119–131. [Google Scholar] [CrossRef]
  141. Mallick, I.H.; Yang, W.; Winslet, M.C.; Seifalian, A.M. REVIEW: Ischemia–Reperfusion Injury of the Intestine and Protective Strategies Against Injury. Dig. Dis. Sci. 2004, 49, 1359–1377. [Google Scholar] [CrossRef]
  142. Jeyabal, P.V.S.; Kumar, R.; Gangula, P.R.R.; Micci, M.-A.; Pasricha, P.J. Inhibitors of advanced glycation end-products prevent loss of enteric neuronal nitric oxide synthase in diabetic rats. Neurogastroenterol. Motil. 2008, 20, 253–261. [Google Scholar] [CrossRef]
  143. Voukali, E.; Shotton, H.R.; Lincoln, J. Selective responses of myenteric neurons to oxidative stress and diabetic stimuli. Neurogastroenterol. Motil. 2011, 23, 964-e411. [Google Scholar] [CrossRef]
  144. Sharkey, K.A. Emerging roles for enteric glia in gastrointestinal disorders. J. Clin. Investig. 2015, 125, 918–925. [Google Scholar] [CrossRef] [Green Version]
  145. Hellwig, M.; Bunzel, D.; Huch, M.; Franz, C.M.A.P.; Kulling, S.E.; Henle, T. Stability of Individual Maillard Reaction Products in the Presence of the Human Colonic Microbiota. J. Agric. Food Chem. 2015, 63, 6723–6730. [Google Scholar] [CrossRef]
  146. Seiquer, I.; Rubio, L.A.; Peinado, M.J.; Delgado-Andrade, C.; Navarro, M.P. Maillard reaction products modulate gut microbiota composition in adolescents. Mol. Nutr. Food Res. 2014, 58, 1552–1560. [Google Scholar] [CrossRef]
  147. Tannock, G.W. A Special Fondness for Lactobacilli. Appl. Environ. Microbiol. 2004, 70, 3189–3194. [Google Scholar] [CrossRef] [Green Version]
  148. Qin, D.; Ma, Y.; Wang, Y.; Hou, X.; Yu, L. Contribution of Lactobacilli on Intestinal Mucosal Barrier and Diseases: Perspectives and Challenges of Lactobacillus casei. Life 2022, 12, 1910. [Google Scholar] [CrossRef] [PubMed]
  149. Flint, H.J.; Bayer, E.A.; Rincon, M.T.; Lamed, R.; White, B.A. Polysaccharide utilization by gut bacteria: Potential for new insights from genomic analysis. Nat. Rev. Microbiol. 2008, 6, 121–131. [Google Scholar] [CrossRef] [PubMed]
  150. Yeoh, Y.K.; Sun, Y.; Ip, L.Y.T.; Wang, L.; Chan, F.K.L.; Miao, Y.; Ng, S.C. Prevotella species in the human gut is primarily comprised of Prevotella copri, Prevotella stercorea and related lineages. Sci. Rep. 2022, 12, 9055. [Google Scholar] [CrossRef]
  151. Qu, W.; Nie, C.; Zhao, J.; Ou, X.; Zhang, Y.; Yang, S.; Bai, X.; Wang, Y.; Wang, J.; Li, J. Microbiome–Metabolomics Analysis of the Impacts of Long-Term Dietary Advanced-Glycation-End-Product Consumption on C57BL/6 Mouse Fecal Microbiota and Metabolites. J. Agric. Food Chem. 2018, 66, 8864–8875. [Google Scholar] [CrossRef]
  152. Marungruang, N.; Fåk, F.; Tareke, E. Heat-treated high-fat diet modifies gut microbiota and metabolic markers in apoe−/− mice. Nutr. Metab. 2016, 13, 22. [Google Scholar] [CrossRef] [Green Version]
  153. Moschen, A.R.; Gerner, R.R.; Wang, J.; Klepsch, V.; E Adolph, T.; Reider, S.J.; Hackl, H.; Pfister, A.; Schilling, J.; Moser, P.L.; et al. Lipocalin 2 Protects from Inflammation and Tumorigenesis Associated with Gut Microbiota Alterations. Cell Host Mircrobe 2016, 19, 455–469. [Google Scholar] [CrossRef] [Green Version]
  154. Mao, Z.; Ren, Y.; Zhang, Q.; Dong, S.; Han, K.; Feng, G.; Wu, H.; Zhao, Y. Glycated fish protein supplementation modulated gut microbiota composition and reduced inflammation but increased accumulation of advanced glycation end products in high-fat diet fed rats. Food Funct. 2019, 10, 3439–3451. [Google Scholar] [CrossRef]
  155. Fung, K.Y.C.; Cosgrove, L.; Lockett, T.; Head, R.; Topping, D.L. A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br. J. Nutr. 2012, 108, 820–831. [Google Scholar] [CrossRef] [Green Version]
  156. Topping, D.L.; Fukushima, M.; Bird, A.R. Resistant starch as a prebiotic and synbiotic: State of the art. Proc. Nutr. Soc. 2003, 62, 171–176. [Google Scholar] [CrossRef]
  157. Mastrocola, R.; Collotta, D.; Gaudioso, G.; Le Berre, M.; Cento, A.; Ferreira, G.A.; Chiazza, F.; Verta, R.; Bertocchi, I.; Manig, F.; et al. Effects of Exogenous Dietary Advanced Glycation End Products on the Cross-Talk Mechanisms Linking Microbiota to Metabolic Inflammation. Nutrients 2020, 12, 2497. [Google Scholar] [CrossRef] [PubMed]
  158. Linkens, A.M.A.; van Best, N.; Niessen, P.M.; Wijckmans, N.E.G.; de Goei, E.E.C.; Scheijen, J.L.J.M.; van Dongen, M.C.J.M.; van Gool, C.C.J.A.W.; de Vos, W.M.; Houben, A.J.H.M.; et al. A 4-Week Diet Low or High in Advanced Glycation Endproducts Has Limited Impact on Gut Microbial Composition in Abdominally Obese Individuals: The deAGEing Trial. Int. J. Mol. Sci. 2022, 23, 5328. [Google Scholar] [CrossRef] [PubMed]
  159. Huda, M.N.; Kim, M.; Bennett, B.J. Modulating the Microbiota as a Therapeutic Intervention for Type 2 Diabetes. Front. Endocrinol. 2021, 12, 632335. [Google Scholar] [CrossRef]
  160. Cui, J.; Ramesh, G.; Wu, M.; Jensen, E.T.; Crago, O.; Bertoni, A.G.; Gao, C.; Hoffman, K.L.; Sheridan, P.A.; Wong, K.E.; et al. Butyrate-Producing Bacteria and Insulin Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes 2022, 71, 2438–2446. [Google Scholar] [CrossRef] [PubMed]
  161. Zhu, S.; Liu, S.; Li, H.; Zhang, Z.; Zhang, Q.; Chen, L.; Zhao, Y.; Chen, Y.; Gu, J.; Min, L.; et al. Identification of Gut Microbiota and Metabolites Signature in Patients With Irritable Bowel Syndrome. Front. Cell. Infect. Microbiol. 2019, 9, 346. [Google Scholar] [CrossRef] [Green Version]
  162. Dai, L.; Tang, Y.; Zhou, W.; Dang, Y.; Sun, Q.; Tang, Z.; Zhu, M.; Ji, G. Gut Microbiota and Related Metabolites Were Disturbed in Ulcerative Colitis and Partly Restored After Mesalamine Treatment. Front. Pharmacol. 2021, 11, 620724. [Google Scholar] [CrossRef]
  163. Olaisen, M.; Flatberg, A.; Granlund, A.V.B.; Røyset, E.S.; Martinsen, T.C.; Sandvik, A.K.; Fossmark, R. Bacterial Mucosa-associated Microbiome in Inflamed and Proximal Noninflamed Ileum of Patients With Crohn’s Disease. Inflamm. Bowel Dis. 2020, 27, 12–24. [Google Scholar] [CrossRef]
  164. Wu, Y.; Dong, L.; Song, Y.; Wu, Y.; Zhang, Y.; Wang, S. Preventive effects of polysaccharides from Physalis alkekengi L. on dietary advanced glycation end product-induced insulin resistance in mice associated with the modulation of gut microbiota. Int. J. Biol. Macromol. 2022, 204, 204–214. [Google Scholar] [CrossRef]
  165. van Dongen, K.C.; Linkens, A.M.; Wetzels, S.M.; Wouters, K.; Vanmierlo, T.; van de Waarenburg, M.P.; Scheijen, J.L.; de Vos, W.M.; Belzer, C.; Schalkwijk, C.G. Dietary advanced glycation endproducts (AGEs) increase their concentration in plasma and tissues, result in inflammation and modulate gut microbial composition in mice; evidence for reversibility. Food Res. Int. 2021, 147, 110547. [Google Scholar] [CrossRef]
Figure 1. Overview of the Maillard reaction leading to the formation of advanced glycation end-products (AGEs). The Maillard reaction occurs when a carbonyl group is exposed to an amine, leading to a Schiff base formation, followed by Amadori rearrangement and formation of Amadori products. The Amadori products undergo irreversible oxidation, dehydration, enolisation, cyclisation, and fragmentation that lead to the formation of reactive intermediate AGE precursors. Reactive AGE precursors interact with protein-bound lysine or arginine to form AGEs, which can be classified as crosslinking or non-crosslinking.
Figure 1. Overview of the Maillard reaction leading to the formation of advanced glycation end-products (AGEs). The Maillard reaction occurs when a carbonyl group is exposed to an amine, leading to a Schiff base formation, followed by Amadori rearrangement and formation of Amadori products. The Amadori products undergo irreversible oxidation, dehydration, enolisation, cyclisation, and fragmentation that lead to the formation of reactive intermediate AGE precursors. Reactive AGE precursors interact with protein-bound lysine or arginine to form AGEs, which can be classified as crosslinking or non-crosslinking.
Nutrients 15 00405 g001
Figure 2. Overview of the effects of dietary AGEs on the gut. AGEs are ligands for various cell surface receptors, of which the receptor for AGEs (RAGE) is the main one and has been extensively studied. RAGE is expressed in the intestinal epithelium and the enteric neurons of the submucosal plexus and myenteric plexus. There has been growing interest indicating the potential roles of endogenous and exogenous AGEs that may act synergistically to accelerate the pathogenetic effects associated with AGEs. AN excessive dietary AGE intake is associated with alterations in the gut structure leading to enhanced gut barrier dysfunction, changes in the expression of enteric neurons, microbial dysbiosis, and inflammation (Created with BioRender.com).
Figure 2. Overview of the effects of dietary AGEs on the gut. AGEs are ligands for various cell surface receptors, of which the receptor for AGEs (RAGE) is the main one and has been extensively studied. RAGE is expressed in the intestinal epithelium and the enteric neurons of the submucosal plexus and myenteric plexus. There has been growing interest indicating the potential roles of endogenous and exogenous AGEs that may act synergistically to accelerate the pathogenetic effects associated with AGEs. AN excessive dietary AGE intake is associated with alterations in the gut structure leading to enhanced gut barrier dysfunction, changes in the expression of enteric neurons, microbial dysbiosis, and inflammation (Created with BioRender.com).
Nutrients 15 00405 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Phuong-Nguyen, K.; McNeill, B.A.; Aston-Mourney, K.; Rivera, L.R. Advanced Glycation End-Products and Their Effects on Gut Health. Nutrients 2023, 15, 405. https://doi.org/10.3390/nu15020405

AMA Style

Phuong-Nguyen K, McNeill BA, Aston-Mourney K, Rivera LR. Advanced Glycation End-Products and Their Effects on Gut Health. Nutrients. 2023; 15(2):405. https://doi.org/10.3390/nu15020405

Chicago/Turabian Style

Phuong-Nguyen, Kate, Bryony A. McNeill, Kathryn Aston-Mourney, and Leni R. Rivera. 2023. "Advanced Glycation End-Products and Their Effects on Gut Health" Nutrients 15, no. 2: 405. https://doi.org/10.3390/nu15020405

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop