Open Access

Role of gut microbiota in the modulation of the health effects of advanced glycation end‑products (Review)

  • Authors:
    • Michael Aschner
    • Anatoly V. Skalny
    • Viktor A. Gritsenko
    • Olga L. Kartashova
    • Abel Santamaria
    • Joao B.T. Rocha
    • Demetrios A. Spandidos
    • Irina P. Zaitseva
    • Aristidis Tsatsakis
    • Alexey A. Tinkov
  • View Affiliations

  • Published online on: April 11, 2023     https://doi.org/10.3892/ijmm.2023.5247
  • Article Number: 44
  • Copyright: © Aschner et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present review was to summarize the potential interactive effects between the gut microbiota and advanced glycation end‑product (AGE) accumulation and toxicity in the host, and to reveal potential the mediatory effects of the gut microbiota on AGE‑related health effects. The existing data demonstrate that dietary AGEs can have a significant impact on the richness and diversity of the gut microbiota, although the particular effect is dependent on the type of species, as well as the exposure dose. In addition, the gut microbiota may metabolize dietary AGEs. It has been also demonstrated that the characteristics of the gut microbiota, including its richness and relative abundance of certain taxa, is tightly associated with AGE accumulation in the host organism. In turn, a bilateral interplay between AGE toxicity and the modulation of the gut microbiota may contribute to pathogenesis of ageing and diabetes‑associated diseases. Bacterial endotoxin lipopolysaccharide appears as the molecule that mediates the interactions between the gut microbiota and AGE toxicity, specifically via the modulation of the receptor for AGE signaling. Therefore, it is proposed that the modulation of the gut microbiota using probiotics or other dietary interventions may have a significant impact on AGE‑induced glycative stress and systemic inflammation.

1. Introduction

Advanced glycation end-products (AGEs) are highly heterogeneous group of chemical species formed through non-enzymatic reactions of glucose or other carbohydrates with proteins and other biomolecules (1). AGEs are formed due to condensation between the carbonyl group of a reducing sugar and free amine group of proteins, lipids or nucleic acids with the irreversible formation of end-products (2). Depending on the molecules involved in glycation, AGEs have been classified into three groups as follows: i) Glycated proteins [e.g., glycated hemoglobin (HbA1c), ApoB100, crystallin, etc.]; ii) low molecular weight AGEs [pyrraline, carboxyethyl lysine (CEL), carboxymethyl lysine (CML), pentosidine, imidazole]; iii) AGEs formed by modification with a particular glycating agent [glucose (AGE-1), glyceraldehyde (AGE-2), glycolaldehyde (AGE-3), methylglyoxal (AGE-4), glyoxal (AGE-5), 3-deoxyglucosone (AGE-6) and acetaldehyde (AA-AGE)] (3). AGEs have also been classified as fluorescent (pentosidine, methylglyoxal-lysine dimer) and non-fluorescent (CML, CEL and pyrraline) (1).

AGEs are formed both endogenously and exogenously through a number of mechanisms (Fig. 1). One of the mechanisms of AGE formation termed the Maillard reaction involves a series of non-enzymatic reactions with the formation of a Schiff base and its subsequent rearrangement into a more stable Amadori product. AGEs are also formed through the interaction of reactive carbonyl species, including glyoxal or methylglyoxal with protein amino acid residues (4). At the same time, multiple other mechanisms may also contribute to the formation of AGEs (5,6).

Endogenously formed AGEs are generated at high amounts in diabetes mellitus due to insulin resistance and persistent hyperglycemia (7). AGEs impart toxic effects to cells through the induction of oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction, apoptosis and inflammation dysregulation (8,9). Excessive AGE formation along with its toxicity in diabetes mellitus is considered a potential mechanism linking diabetes with other metabolic disorders (10). In addition to diabetes and metabolic syndrome (11), AGEs have been shown to be involved in the pathogenesis of a variety of other diseases, including neurodegeneration (12), cancer (13), osteoporosis (14), infertility (15), chronic kidney disease (16) and aging (17,18). Correspondingly, the results of a recent meta-analysis demonstrated a significant association between circulating AGEs and their soluble receptor levels and both all-cause and cardiovascular mortality (19).

Dietary AGE intake also significantly contributes to AGE accumulation and toxicity (20). Western diets which are based on highly processed and heat-treated foods are known to contain high levels of AGEs (21). Given these associations, AGEs are considered a potential link between the modern diet and adverse health outcomes (22).

It has been proposed that the modulation of the gut microbiota significantly contributes to the effect of AGEs on human health (23), and mediates the differences observed between the effects of low and high molecular mass glycation products in the organism (24). However, the existing data are inconsistent and the potential contribution of the gut microbiota in the modulation of AGE-induced toxicity and glycation stress, as well as the health effects of AGE accumulation are unclear. Therefore, the aim of the present review was to summarize and discuss the potential interactive effects between the gut microbiota and AGE accumulation and toxicity in the host, as well as to reveal the potential mediatory effects of the gut microbiota on AGE-related health effects.

2. Bacterial AGE metabolism

The existing data demonstrate that the gut microbiota may be considered as a source of AGEs. Specifically, it has been demonstrated that Escherichia coli (E. coli) cultures release AGEs during growth (25). Such an effect may be mediated by the bacterial secretion of methylglyoxal (MGO), which is considered as a reactive carbonyl species and an AGE precursor (4). High MGO-producing activity has been demonstrated for Proteus spp. (26), E. coli (27), Pediococcus acidilactici and other bacteria (28).

MGO is formed in bacterial cells as a product of multiple metabolic processes (29), although its overaccumulation with a subsequent increase in AGE formation has been shown to exert toxic effects (30) and limit bacterial growth (31).

The main source of MGO is glucose catabolism, including both enzymatic and non-enzymatic reactions. The key mechanism of MGO synthesis is the transformation of dihydroxyacetone phosphate catalyzed by methylglyoxal synthase (MgsA). In turn, non-enzymatic MGO formation may result from the fragmentation of triosephosphates via phosphoenediolate intermediate (29).

The toxicity of MGO for bacterial cells is associated with its high reactivity and modification of nucleic acids and proteins, resulting in AGE formation (30). Specifically, it has been demonstrated that MGO exposure is toxic to both Gram-positive (Bacillus subtilis and Staphylococcus aureus) and Gram-negative (Pseudomonas aeruginosa and E. coli) bacteria, inhibiting their growth and inducing structural alterations in bacterial fimbriae and flagella (31).

To overcome the toxic effects of increasing MGO concentrations and subsequent cell death (32), MGO decomposition is strictly regulated by a number of mechanisms involving glyoxalases and NAD-dependent enzymes (Fig. 2). Glyoxalases I and II catalyze the glutathione (GSH)-dependent conversion of MGO to D-lactate through the formation of S-D-lactoylglutathione, whereas glyoxalase III catalyzes this conversion without consuming GSH. The NAD-dependent enzymes, glycerol dehydrogenase (GldA) and aldehyde dehydrogenase (AldA), catalyze the transformation of MGO to D-lactate through D-lactaldehyde (30). The resulting D-lactate may be subsequently transformed to pyruvate or excreted (33).

In addition to MGO, which is considered a precursor of AGEs, existing data demonstrate that gut bacteria can metabolize other AGEs. Specifically, it has been demonstrated that the human gut microbiota is able to degrade Maillard reaction products with a substantial reduction at 24 h observed for fructosyllysine (100%) > carboxymethyllysine (41%) > pyrraline (20%), but not maltosine (34). A recent study by Bui et al (35) demonstrated that the gut microbiota is capable of anaerobic carboxymethyllysine degradation to carboxymethylated biogenic amines and 11 carboxylic acids with Oscillibacter and Cloacibacillus evryensis being the potential responsible taxa. It has been demonstrated that adult fecal microbiota and particularly Intestinimonas spp. can convert Nε-fructosyllysine to butyrate, whereas such a property in bacteria isolated from 3-4-month-old infants was dependent on the type of feeding. Specifically, the microbiota of breast-fed infants was unable to degrade Nε-fructosyllysine, whereas that of formula-fed infants possessed a Nε-fructosyllysine-converting ability due to the presence of this AGE in infant formulas following thermal exposure, thus being indicative of the adaptation of microbiota metabolism to dietary compounds (36). In addition, E. coli has been shown to metabolize CML with the formation of three metabolites, N-carboxymethylcadaverine, N-carboxymethylaminopentanoic acid and the N-carboxymet hyl-Δ1-piperideinium ion, although the particular end-product may be strain-specific (37).

It has also been demonstrated that bacterial metabolites can modulate AGE toxicity. The existing data demonstrate that gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) can increase the production of AGEs in the aorta, thus promoting arterial stiffening (38), and underlying the role of TMAO and AGEs in the progression of cardiovascular and chronic kidney diseases (39).

3. The impact of dietary AGE exposure on gut microbiota characteristics and host metabolism

Diet has a significant impact on the biodiversity and metabolism of the gut microbiota (40). Correspondingly, the results of a recent systematic review demonstrated that the characteristics of dietary protein, including protein glycation can modulate gut biodiversity, although significant inconsistencies still exist (41).

Adverse effects of dietary AGEs on the gut microbiota

Consistent with the overall understanding of AGEs as perilous molecules, several studies have demonstrated that the dietary intake of AGEs induces the dysfunction of the gut microbiota along with unfavorable effects in the host organism. Specifically, in a human study, it was demonstrated that the increased exposure to glycated BSA significantly affected the colonic microbiota sampled from the feces of both healthy subjects and patients with ulcerative colitis, with a profound decrease in beneficial bacteria (eubacteria and bifidobacteria) and an increased abundance of more hazardous phyla (clostridia, bacteroides, sulfate-reducing bacteria) (42). The detailed study by Seiquer et al (43) revealed the significant association between AGEs intake and gut microbiota composition both in humans and rats. Specifically, the relative abundance of lactobacteria was inversely associated with dietary hydroxymethylfurfural and carboxymethyl-lysine, whereas the relative numbers of bifidobacterial were inversely associated with the intake of Amadori compounds in adolescents. Similarly, in rats, the dietary intake of the Amadori compounds, hydroxymethylfurfural and carboxymethyl-lysine, was negatively associated with both total bacteria and lactobacteria (43).

In a laboratory study, it was demonstrated that AGE-rich food significantly aggravated alterations in gut microbiota biodiversity in a time-dependent manner, being more profound at 18 weeks of exposure, as compared to 6 and 12 weeks. Furthermore, it was shown that the high-AGE group ws characterized by a significant decrease in the abundance of Ruminococcaceae, Lachnospiraceae, Alloprevotella, Mollicutes, Christensenellaceae, Treponema, Prevotellaceae, Sphaerochaeta, Elusimicrobium, Butyrivibrio and Anaeroplasma, whereas that of Oscillibacter, Allobaculum, Anaerotruncus, Barnesiella, Fusicatenibacter and Veillonellaceae was elevated (44). Feeding rats with a heat-treated diet rich in AGEs resulted in a significant increase in the relative abundance of Parabacteroides, Alloprevotella, Helicobacter, Ruminococcaceae_UCG-014, and unclassified Rhodospirillaceae, whereas populations of Desulfovibrio, Rikenellaceae, Lachnospiraceae and Alistipes, were significantly reduced, being associated with perturbations of microbial metabolites and certain metabolic pathways, including impaired carbohydrate and amino acid metabolism (45). Correspondingly, in another study, the administration of an AGE-rich diet, obtained by heating the standard chow, to C57BL/6 mice resulted in a significant increase in circulating and tissue AGEs levels, systemic inflammation, and the alteration of the gut microbiota mainly characterized by the increased abundance of Clostridium_sensu_stricto_1, Turicibacter and Parasutterella, and in particular, Dubosiella at the genera level, as well as the increased abundance of Clostridiaceae_1, Erysipelotrichaceae and Burkholderiaceae families (46).

Dietary AGE-induced effects on gut microbiota have also been linked to several diseases. Specifically, a previous study demonstrated that feeding C57BL/6 mice with an AGE-rich diet significantly increased systemic AGE (CML) levels, protein glycosylation, receptor for AGEs (RAGE) expression in the ileum and submandibular glands, as well as complex alterations in gut microbiota composition. AGE intake significantly increased Lawsonia, Parabacteroides and Ruminococcus abundance, whereas the relative numbers of Lactobacillus, Prevotella, Anaerostipes and Candidatus Arthromitus were significantly reduced, altogether being associated with impaired insulin signal transduction (47). It has been also demonstrated that feeding rats with casein glycated with methylglyoxal 5-hydro-5-methylimidazolone significantly affects the intestinal microbiota, which may at least in part be responsible for a reduction in systemic gastric inhibitory polypeptide and glucagon-like peptide-1 levels, consistent with an altered incretin-insulin axis, as well as with the overproduction of the pro-inflammatory cytokines, interleukin (IL)-1β and IL-17 and plasminogen activator inhibitor-1 (48).

Potentially beneficial effects of dietary AGEs on the gut microbiota

In contrast to the aforementioned findings, a number of studies have demonstrated that administration of dietary AGEs can afford beneficial effects, both to the gut microbiota and host metabolism. It is notable that the majority of results demonstrating the positive influence of AGEs on gut microbiota were attributable to administration of glycated fish proteins. Specifically, the glycation of dietary grass carp myofibrillar proteins resulting in increased furosine levels in glycoconjugates significantly increased gut microbiota biodiversity and butyrate production that was positively associated with the relative abundance of Mitsuokella, Lachnospiraceae_UCG-004, Sutterella, Salinimicrobium, Fodinibius and Nitriliruptor, being inversely associated with that of Enterococcus, Dorea, Escherichia-Shigella and Phascolarctobacterium, thus being indicative of the potential beneficial effects of protein glycation on gut health (49). Correspondingly, another study demonstrated that the administration of glycated fish protein to rats significantly increased the relative abundance of Allobaculum, Akkermansia, Turicibacter and Lactobacillus animalis, and reduced that of Escherichia-Shigella, Fusobacterium and Erysipelatoclostridium in the caecum, altogether being associated with the increased production of butyrate from fructoselysine (50). Similar findings were obtained in another study, where the administration of fish peptide glycated with galactooligosaccharide resulted in increased abundance of the Veillonellaceae, Prevotellaceae and Coriobacteriaceae families, and increased the abundance of the genera, Anaerovibrio, Collinsella, Prevotella_9, as well as reduced Alloprevotella, Holdemanella, Escherichia-Schigella and Streptococcus when compared to the control rats (51). Increasing fish protein glycation through heating for 24 or 48 h with its subsequent in vitro fermentation in a model of human distal colon significantly increased the relative abundance of Lactococcus in parallel with a decrease in Bacteroides. Moreover, the exposure of gut bacteria to glycated fish protein heated for 48 h resulted in a greater abundance of Holdemania, Streptococcus, Enterococcus and Lactobacillus, as well as a reduction of Parabacteroides when compared to a less glycated protein (24 h of heating). These changes, and particularly a decrease in Bacteroides, Dialister and Parabacteroides, were associated with reduced ammonia and indole production (52). The intake of glycated fish protein in high-fat fed rats also decreased relative abundance of Ruminiclostridium and Desulfovibrio, as well as dose-dependent effects, including increased Ruminococcus and Roseburia abundance in low glycated protein diet and decreased Helicobacter and Lachnospiraceae upon high-dose glycated protein intake. Moreover, the observed AGE-induced modulation of gut microbiota composition was associated with a significant reduction of systemic proinflammatory cytokine (IL-1β and IL-6) levels and lipid profile improvement, thus indicative of the potential beneficial effects on gut and metabolism (53).

It has been also demonstrated that the glycation of the milk proteins, β-lactoglobulin and casein, significantly increased fermentability of the proteins by Lactobacillus and Bifidobacterium thus promoting their growth, although the effect was more profound at the initial steps of Maillard reaction (54). β-lactoglobulin-galactose conjugate was also shown to promote Clostridium coccoides-Eubacterium rectal group growth, as well as increase bacterial acetate production (55). Notably, the modification of β-lactoglobulin by glycation and ultrasonication has been shown to reduce milk protein allergenicity, which may be mediated by lower digestibility, modification of allergenic epitopes on the protein molecule, as well as modulation of gut microbiota composition (56).

The administration of glycated whey proteins to aged male non-obese diabetic mice with autoimmune prostatitis significantly increased mice survival, reduced prostatic inflammation, as well as an increased abundance of Allobaculum, Anaerostipes, Bacteroides, Parabacteroides and Prevotella and reduced abundance Adlercreutzia and Roseburia, whereas the population of Bacteroides acidifaciens significantly correlated with the observed effects, indicative of the role of gut microbiota modulation in protective effects of glycated whey proteins (57). Similarly, a beneficial effect on the gut microbiota was demonstrated for glycated pea protein which increased Bacteroides, Lactobacillus/Enterococcus and Bifidobacterium growth as well as short chain fatty acids (SCFAs), acetate, propionate, lactate, and butyrate production (58).

The dietary restriction of AGEs was shown to reduce systemic CML and MG levels, as well as affect the gut microbiota by increasing the relative abundance of Alistipes indistinctus, Clostridium citroniae, Clostridium hathewayi, and Ruminococcus gauvreauii, in parallel with a decrease in Prevotella copri and Bifidobacterium animalis in peritoneal dialysis patients (59). Concomitantly, in another study, a reduction in dietary AGEs intake did not have a significant effect on the most abundant gut bacteria in healthy obese subjects. As compared to the group with a high AGE intake, the low dietary AGE group was characterized by a greater abundance of Tyzzerella, Family_XII_UCG-001 and Christensenellaceae_R-7 Group, as well as a lower abundance of Negativibacillus, Oscillibacter and Anaerostipes (60).

An insight into the distinct effects of various AGEs on the gut microbiota

As clearly detailed in previous studies, the effects of dietary AGEs on the gut microbiota vary significantly, depending on their characteristics, including both the dose, source and chemical properties. The study by Cao et al (61) proposed that the observed inconsistencies in the reported effects of AGE intake on the gut microbiota and overall health were dependent on the dose of the glycated protein in the diet. The intake of low levels of glycated fish proteins for 15 weeks in mice resulted not only in an increased abundance of the butyrate-producing bacteria, Lachnospiraceae and Allobaculum, but also increased intestinal tight junction protein expression (occludin and Zonula occludens-1), reduced pro-inflammatory cytokine production (IL-1β and IL-6) and improved insulin sensitivity. By contrast, inverse effects were observed upon exposure to high-dose glycated fish protein in parallel with a reduction in Bifidobacterium and Lactobacillus abundance (61). It has been posited that free AGEs, such as carboxymethyllysine have detrimental effects on the composition and functions of the gut microbiota, whereas bound AGEs have a more beneficial effect, although certain detrimental effects may be also observed (62). It was hypothesized that the positive effects of glycated proteins, namely fish proteins on the gut microbiota are due to the use of such proteins as a slow fermentable protein source or carbonyl donors providing additional energy to gut microbiota (46). In addition, free and protein-bound AGEs in the diet have distinct effects on the gut microbiota due to differences in digestibility (63). In addition to the AGE species, the molecular weight of the ligand has a significant impact on digestibility. Specifically, in a previous study, in a model of glyoxal-glycated casein digests, CML was degraded predominantly in the low molecular weight fraction (38.7%) followed by medium (21.7%) and high molecular weight fractions (9.6%) which may be mediated by the lower activity of proteases (64).

4. Involvement of AGE-gut microbiota interplay in disease pathogenesis

Type 2 diabetes mellitus

The understanding of the pathogenesis of diabetes mellitus sheds light onto the toxicological effects of AGEs and their role in metabolic diseases (10). Although the dysfunction of the gut microbiota has been known to play a significant role in diabetogenesis (65), the potential interplay between gut microbiota dysfunction and excessive protein glycation in diabetes has been studied only recently. Wu et al (66) demonstrated that dietary AGE exposure significantly affected the gut microbiota, with an irreversible increase in Bacteroidetes populations and a decrease in Firmicutes abundance at the phylum level, whereas at the genera level, a high-AGE diet stimulated Helicobacter, Bacteroides, Rikenella, Alistipes, Bifidobacterium, Candidatus Saccharimonas, Faecalibaculum, Clostridiales, Erysipelatoclostridium and Intestinimonas, and decreased unidentified Lachnospiraceae, Roseburia, Oscillibacter, Anaerotruncus, Blautia, Mucispirillum, Angelakisella, Lachnoclostridium, Lachnospira, Ruminiclostridium, Acetatifactor, and Desulfovibrio. These perturbations were shown to contribute to diabetes pathogenesis through the modulation of glyceraldehyde and pyruvate production with the subsequent aggravation of insulin resistance and other alterations in carbohydrate and lipid metabolism, as well as inflammation due to higher systemic lipopolysaccharide (LPS) levels (66). A comparative analysis demonstrated that despite a significant increase in circulating AGEs and the induction of insulin resistance in mice exposed to both an AGE-rich diet or purified methylglyoxal-bovine serum protein (exogenous AGE), profound alterations of intestinal permeability and microbiota structure were observed only upon exogenous AGE intake. A high AGE intake was shown to reduce the abundance of Bacteroidales_S24-7, Bacteroidaceae, Porphyromonadaceae, Odoribacteraceae, Lachnospiraceae, Rikenellaceae, and Erysipelotrichaceae in parallel with an increase in Desulfovibrionaceae abundance (67). It was proposed that a decrease in butyrate production by butyrate-producing bacteria may promote the impairment of the intestinal epithelial barrier and induce inflammation, thus contributing to systemic insulin resistance (67). The observed increase in Desulfovibrio abundance generally corresponds to the early observed positive association between these bacteria with blood glucose indices (68) and Parkinson's disease (69), although the results of the Guangdong Gut Microbiome Project demonstrated that the abundance of Desulfovibrio may be inversely associated with body mass index and triglyceride levels (70). Moreover, earlier findings in diabetic db/db mice exposed to high levels of dietary AGEs demonstrated in a significant increase in gut permeability, as well as an elevation of the Firmicutes-to-Bacteroidetes ratio, altogether being associated with kidney damage and albuminuria, whereas the administration of resistant starch ameliorated these effects (71). It is also notable that the formation of Maillard reaction products with subsequent protein aggregation in bacterial species shares certain similarity to that observed in Parkinson's and Alzheimer's disease (72). Therefore, preliminary data demonstrate that AGE-induced alterations in the gut microbiota can contribute to the aggravation of insulin resistance through a number of mechanisms, including the impairment of the intestinal epithelial barrier and subsequent increase in circulating LPS levels.

Ageing-associated diseases

Recent studies have demonstrated that alterations in the gut microbiota, as well as increased levels of AGEs are associated with aging, contributing to the development of age-related diseases (73,74). Age-related changes in the gut microbiota characterized by a reduced Firmicutes-to-Bacteroidetes ratio at the phylum level, as well as by the increased abundance of Turibacter, Alloprevotella, Parasutterella, Bifidobacterium, Macellibacteroides, Alistipes sensu stricto 1, Peptostreptococcaceae incertae sedis and Parabacteroides, and the lower abundance of Pantoea, Anoxybacillus, Lachnospiraceae incertae sedis, Cutrobacterium and Acetatifactor at the genera level, were shown to contribute to the accumulation of N6-carboxymethyllysine in microglia and subsequent oxidative stress and mitochondrial dysfunction by increasing intestinal permeability, whereas germ-free mice brain microglia were characterized by lower oxidative stress and mitochondrial damage (75). In corroboration, the translocation of fecal microbiota from aged to young rats impaired cognition, induced synaptic dysfunction, along with oxidative stress and inflammation, which may be at least partially mediated by an increased AGE production and RAGE expression (76). Correspondingly, the antibiotic treatment of 5xFAD mice, a model of Alzheimer's disease that is known to be age-related, resulted in a significant decrease in intestinal Lactobacillaceae abundance, being also associated with reduced hippocampal plaque formation, antidiabetic effect and decreased RAGE expression (77). Taken together, even these limited data demonstrate that age-related alterations of the gut microbiota may contribute to AGE accumulation, particularly in brain tissues, indicating the gut microbiota-AGE interplay in age-related neurodegeneration.

Other diseases

A recent study in ethanol-fed mice demonstrated an increased abundance of Bacteroidetes and a decrease in Firmicutes numbers, which was associated with an elevation in AGE and RAGE levels in colonic tissues, and considered a potential mechanism of ethanol-related colorectal cancer pathogenesis (78).

5. Effects of gut microbiota modulation on AGE metabolism and toxicity

Probiotics

Several studies have demonstrated that the modulation of the gut microbiota using probiotics is also associated with reduced AGE accumulation and toxicity, thus also supporting the role of the gut microbiota in AGE toxicity. Specifically, in a previous study, in a model of Alzheimer's disease, the modulation of the gut microbiota through the administration of SLAB51 probiotic significantly reduced brain AGE accumulation and tau phosphorylation, and also improved insulin signaling through the Akt/AMPK pathway (79). In another laboratory in vivo study, the administration of probiotic Lactobacillus paraplantarum BGCG11 significantly reduced AGE accumulation in parallel with inhibiting hyperglycemia, oxidative stress, DNA damage, liver and kidney fibrosis in rats with streptozotocin-induced diabetes (80). In another study, the administration of the commercial probiotic, Protexin®, in Cd-exposed rats significantly reduced the serum MGO levels, as well as decreased tissue Cd accumulation and Cd-induced oxidative stress (81). Lactococcus lactis KF140 supplementation has also been shown to reduce serum CML levels and hepatic CML accumulation that may be at least partially mediated by activity of bacteria-derived β-galactosidase (82).

In addition to probiotics, it has been demonstrated that treatment with prebiotics may also modulate AGE accumulation and toxicity. Specifically, the administration of the prebiotic, resistant dextrin, has been shown to significantly reduce carboxymethyl lysine, soluble RAGE, as well as several other cardiometabolic risk factors in adult women (83).

At the same time, additional studies, including clinical trials are required to address the impact of microbiota modulation by probiotics and prebiotics on AGE toxicity and RAGE signaling, as well as the clinical validity of these interventions.

Phytochemicals

Polyphenols have also been shown to have a significant beneficial effect on gut microbiota and glycative stress, and these effects appear partially interrelated (84). Specifically, the administration of Physalis alkekengi L. fruit polysaccharide to AGE-fed mice was shown to modulate gut microbiota by increasing the abundance of Rikenallaceae, Alistipes, Nocardiaceae, Rhodococus, Bacilli, Lactobacillaceae, Bacteroidaceae and Burkholderiaceae, improving the Bacteroidetes/Firmicutes ratio, as well as decreasing LPS production. Treatment with Physalis alkekengi polysaccharide also improved the bacterial production of SCFAs, namely acetic and propionic acids, which may at least partially mediate the treatment-induced reduction of insulin resistance (85). It has also been demonstrated that the administration of quercetin to AGE-fed mice significantly ameliorated cognitive dysfunction through the reduction of tau phosphorylation, cathepsin B and neuroinflammation, as well as increased gut microbiota biodiversity and reduced the abundance of Verrucomicrobia phylum, and Blautia and Anaerotruncus genera (86). In addition, it has been proposed that an increase in Lactobacteria and particularly Bifidobacteria by Geranium dielsianum extract may at least partially mediate antiglycative effect of the extract (87). These data demonstrate that the protective effects of phytochemicals against AGE toxicity are mediated by its influence on the composition of the gut microbiota.

6. Role of lipopolysaccharide in the interplay between microbiota and AGE toxicity

LPS, also known as endotoxin, is a cell wall component of Gram-negative bacteria. In the human gut microbiota, Bacteroidetes, and to a lesser extent, Proteobacteria phyla, are considered as key sources of LPS production (88). LPS mediates a substantial part of the effects of altered microbiota on the host, including the regulation of systemic inflammation (89).

Several studies have demonstrated the significant effect of LPS on cellular production and the accumulation of AGEs or their precursors. Specifically, a previous study demonstrated that the stimulation of RAW264.7 murine macrophages with LPS resulted in a significant increase in intracellular methylglyoxal generation upon high-glucose conditions in parallel with HIF-1 downregulation and pyroptosis (90). A similar effect was observed in LPS-stimulated J774A.1 macrophages and N11 microglia (91). Correspondingly, long-term LPS treatment was shown to increase aortal AGE accumulation (92). In turn, in rat aortic smooth muscle cells, MGO treatment was shown to inhibit LPS-stimulated inducible nitric oxide synthase expression by inhibiting Akt phosphorylation that may be involved in diabetic vascular dysfunction (93). These findings generally corroborate recent data obtained by Kitaura et al (94), demonstrating that AGEs may reduce LPS uptake by RAW264.7 macrophages, which may be at least partially mediated by RAGE activation, resulting in altered immune response in diabetes (94).

LPS is a potent pro-inflammatory agent that induces an inflammatory response through a number of mechanisms. It has been shown that AGEs potentiate the pro-inflammatory effects of LPS on gingival fibroblasts under high-glucose conditions, as evidenced by an elevated IL-8 secretion (95). The potentiation of the pro-inflammatory effects of LPS and AGEs may be mediated by the activation of mitogen-activated protein kinases and NF-κB activation in endothelial cells (96).

One of the mechanisms underlying the pro-inflammatory effects of LPS and its impact on NF-κB signaling is the modulation of RAGE signaling (97) (Fig. 3). The activation of RAGE signaling has been shown to mediate certain effects of glucotoxicity, as well as the toxic effects of environmental factors (98,99).

Given the role of AGEs as ligands for RAGE, the modulation of RAGE signal transduction by LPS may also be considered as one of the aspects of microbiota-AGE interplay. Specifically, in human umbilical vein endothelial cells, LPS treatment has been shown to increase both RAGE expression and NF-κB activation, as well as p65 nuclear translocation, whereas anti-RAGE antibody ameliorated the LPS-induced NF-κB activation and subsequent endothelial barrier dysfunction, thus being indicative of the role of RAGE in the LPS-induced inflammatory reaction (100). Anti-RAGE antibody has also been shown to reduce LPS-induced acute lung injury in a neonatal rat model (101). Concomitantly, tje inhibition of NF-κB signaling significantly decreases LPS-induced RAGE expression in alveolar type I epithelial cells, whereas RAGE knockdown inhibits both basal and LPS-induced NF-κB activation (102).

A previous study demonstrated that LPS may directly bind RAGE with a subsequent NF-κB-dependent inflammatory response in a murine model of septic shock, whereas the injection of soluble RAGE significantly reduced LPS-induced proinflammatory cytokine expression and tissue damage (103). It has been proposed that the ratio between cell surface RAGE and soluble RAGE (sRAGE) may significantly mediate inflammatory response to bacterial molecules (103). Additional research has demonstrated that the direct interaction between LPS, high mobility group box 1 (HMGB1) and AGEs results in the formation of a triplet complex and subsequent increase in HMGB1 mobility, altogether leading to increased TNF-α mRNA expression in RAW264.7 macrophages through Toll-like receptor 4 (TLR4) and RAGE activation (104).

Another study demonstrated that the inhibition of RAGE signaling thwarted the LPS-induced upregulation of HMGB1 and IL-6 expression through a NF-κB-mediated mechanism (105). Accordingly, the inhibition of RAGE activation has been considered as one of the mechanisms underlying the protective effects of certain agents including β-caryophyllene and perindopril against LPS-induced liver injury (106) and amyloidogenesis (107), respectively. In agreement with this, the phytochemicals, icariin and icaritin, have been found to significantly reduce LPS-induced hippocampal neuroinflammation through the downregulation of HMGB1-RAGE signaling (108). Concomitantly, preconditioning with HMGB1 has been shown to induce LPS tolerance in a RAGE-dependent manner (109).

In addition to the role of RAGE signaling in LPS-induced inflammation, this mechanism may also trigger the adverse effects of LPS on the cytoskeleton and tight junction proteins. Specifically, RAGE signaling has been shown to be involved in LPS-induced cytoskeletal alterations in mouse pulmonary microvascular endothelial cells, as demonstrated in RAGE-knockout pulmonary microvascular endothelial cells, which did not develop F-actin rearrangement and stress fiber formation upon LPS stimulation (110). In another study, RAGE-deficient mice were also found to be resistant to LPS-induced leukocyte infiltration and proinflammatory cytokine secretion, as well as alteration of lung Zonula occludens-1, sodium-potassium ATPase (Na, K-ATPase), and epithelial sodium channel expression. Moreover, in patients with infection-induced acute respiratory distress syndrome, bronchial alveolar lavage fluid sRAGE levels were increased, being associated with pro-inflammatory cytokine levels and pulmonary vascular permeability (111). In line with these observations, it was previously demonstrated that the inhibition of RAGE signaling not only reduced pro-inflammatory cytokine expression in a model of LPS-induced acute lung injury, but also prevented the downregulation of claudin-2 and occludin expression (112). While discussing the role of AGE signaling and LPS in the alteration of cell contacts, it is important to note that glycated caseinate hydrolysate has been shown to possess significantly lower barrier-protective effects in LPS-exposed intestinal IEC-6 cells as compared to unmodified caseinate hydrolysate (113).

The activation of LPS-RAGE signaling has also been shown to mediate carcinogenesis. Specifically, the upregulation of HMGB1/RAGE signaling has been found to be responsible for LPS-induced inflammation and the subsequent malignant transformation of normal cervical epithelial cells (114). Moreover, a previous study demonstrated that the breast tumor microbiota was enriched with Gram-negative bacteria producing LPS. In vitro LPS treatment was shown to upregulate S100A7 expression in breast cancer cells, resulting in the upregulation of RAGE expression along with a reduced TLR4 expression that may contribute to tumor growth progression (115).

At the same time, it has been proposed that RAGE signaling may mediate the inflammatory response to bacteria through reactions to other bacterial molecules than LPS (116).

Taken together, these findings demonstrate that AGEs can modulate the pro-inflammatory effects of bacterial LPS, that is normally released by gut microbiota, whereas the pro-inflammatory signals of LPS are mediated by RAGE activation, which is also activated by AGEs.

7. Conclusions and future perspectives

The existing data demonstrate a bilateral association between gut microbiota and the effects of AGEs. Such an association may be summarized by the following aspects: i) Dietary AGEs may have a significant impact on the richness and diversity of the gut microbiota; ii) the gut microbiota may metabolize dietary AGEs; iii) the composition of the gut microbiota is tightly associated with AGE accumulation in the host organism; iv) certain effects of AGE accumulation in the organism may be mediated by the modulation of the gut microbiota; v) the alteration of the gut microbiota may mediate the development of comorbidities associated with ageing and diabetes; vi) LPS may be considered as the molecule mediating the association between the gut microbiota and AGEs, and particularly, RAGE signaling; vii) dietary interventions aimed at the improvement of the gut microbiota may exert protective effects against AGEs toxicity. Given a mutual interaction between AGE toxicity and dysbiosis, it can be hypothesized that exposure to dietary AGEs may induce gut dysbiosis that further promotes AGE production, thus composing a vicious circle involved in disease pathogenesis. This vicious circle may be involved in the development of opportunistic infections and systemic inflammation in diabetic patients characterized by high levels of endogenous AGE. Therefore, the modulation of the gut microbiota with probiotics or other nutrients may be considered as a potential protective strategy against AGE-induced glycative stress and systemic inflammation. However, further studies on the molecular aspects of the interaction between gut microbiota and AGE metabolism and toxicity are required.

Availability of data and materials

Not applicable.

Authors' contributions

MA and AAT were involved in the conceptualization of the study. MA, AVS, VAG, OLK, AS, JBTR, IPZ, AT and AAT were involved in the investigation of the literature and in the curation of data for inclusion in the review. VAG, OLK, IPZ and AAT were involved in the writing and preparation of the original draft. MA, AVS, AS, JBTR, DAS and AT were involved in the writing, reviewing and editing of the manuscript. AAT was involved in visualization. MA, AVS, AS, JBTR, DAS and AT supervised the study. AAT was involved in funding acquisition. All authors have read and agreed to the published version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

DAS is the Editor-in-Chief for the journal, but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article. The other authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was performed with the financial support of the Russian Science Foundation (project No. 20-16-00078).

References

1 

Perrone A, Giovino A, Benny J and Martinelli F: Advanced glycation end products (AGEs): Biochemistry, signaling, analytical methods, and epigenetic effects. Oxid Med Cell Longev. 2020:38181962020. View Article : Google Scholar : PubMed/NCBI

2 

Twarda-Clapa A, Olczak A, Białkowska AM and Koziołkiewicz M: Advanced glycation end-products (AGEs): Formation, chemistry, classification, receptors, and diseases related to AGEs. Cells. 11:13122022. View Article : Google Scholar : PubMed/NCBI

3 

Kuzan A: Toxicity of advanced glycation end products (Review). Biomed Rep. 14:462021. View Article : Google Scholar : PubMed/NCBI

4 

Aaseth J, Skalny AV, Roos PM, Alexander J, Aschner M and Tinkov AA: Copper, iron, selenium and lipo-glycemic dysmetabolism in Alzheimer's disease. Int J Mol Sci. 22:94612021. View Article : Google Scholar : PubMed/NCBI

5 

Luevano-Contreras C, Garay-Sevilla ME and Chapman-Novakofski K: Role of dietary advanced glycation end products in diabetes mellitus. J Evid Based Complementary Altern Med. 18:50–66. 2013. View Article : Google Scholar

6 

Song Q, Liu J, Dong L, Wang X and Zhang X: Novel advances in inhibiting advanced glycation end product formation using natural compounds. Biomed Pharmacother. 140:1117502021. View Article : Google Scholar : PubMed/NCBI

7 

Vlassara H and Uribarri J: Advanced glycation end products (AGE) and diabetes: Cause, effect, or both? Curr Diab Rep. 14:4532014. View Article : Google Scholar :

8 

Uribarri J, del Castillo MD, de la Maza MP, Filip R, Gugliucci A, Luevano-Contreras C, Macías-Cervantes MH, Markowicz Bastos DH, Medrano A, Menini T, et al: Dietary advanced glycation end products and their role in health and disease. Adv Nutr. 6:461–473. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Sruthi CR and Raghu KG: Advanced glycation end products and their adverse effects: The role of autophagy. J Biochem Mol Toxicol. 35:e227102021. View Article : Google Scholar : PubMed/NCBI

10 

Ruiz HH, Ramasamy R and Schmidt AM: Advanced glycation end products: Building on the concept of the 'common soil' in metabolic disease. Endocrinology. 161:bqz0062020. View Article : Google Scholar

11 

Margina D, Gradinaru D, Manda G, Neagoe I and Ilie M: Membranar effects exerted in vitro by polyphenols-quercetin, epigallocatechin gallate and curcumin-on HUVEC and Jurkat cells, relevant for diabetes mellitus. Food Chem Toxicol. 61:86–93. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Chrysanthou M, Miro Estruch I, Rietjens IMCM, Wichers HJ and Hoppenbrouwers T: In vitro methodologies to study the role of advanced glycation end products (AGEs) in neurodegeneration. Nutrients. 14:3632022. View Article : Google Scholar : PubMed/NCBI

13 

Khan H, Khan MS and Ahmad S: The in vivo and in vitro approaches for establishing a link between advanced glycation end products and lung cancer. J Cell Biochem. 119:9099–9109. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Ge W, Jie J, Yao J, Li W, Cheng Y and Lu W: Advanced glycation end products promote osteoporosis by inducing ferroptosis in osteoblasts. Mol Med Rep. 25:1402022. View Article : Google Scholar : PubMed/NCBI

15 

Zhu JL, Cai YQ, Long SL, Chen Z and Mo ZC: The role of advanced glycation end products in human infertility. Life Sci. 255:1178302020. View Article : Google Scholar : PubMed/NCBI

16 

Bettiga A, Fiorio F, Di Marco F, Trevisani F, Romani A, Porrini E, Salonia A, Montorsi F and 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. 11:17482019. View Article : Google Scholar : PubMed/NCBI

17 

Moskalev A, Stambler I and Caruso C: Innate and adaptive immunity in aging and longevity: The foundation of resilience. Aging Dis. 11:1363–1373. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Borsa C, Gradinaru D, Margina D, Prada G and Peña C: Receptor of advanced glycation end products and cardiovascular risk in elderly with type 2 diabetes mellitus. J Biol Res. 90:81–86. 2017.

19 

Sharifi-Zahabi E, Sharafabad FH, Abdollahzad H, Malekahmadi M and Rad NB: Circulating advanced glycation end products and their soluble receptors in relation to all-cause and cardiovascular mortality: A systematic review and meta-analysis of prospective observational studies. Adv Nutr. 12:2157–2171. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Nowotny K, Schröter D, Schreiner M and Grune T: Dietary advanced glycation end products and their relevance for human health. Ageing Res Rev. 47:55–66. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Inan-Eroglu E, Ayaz A and Buyuktuncer Z: Formation of advanced glycation endproducts in foods during cooking process and underlying mechanisms: A comprehensive review of experimental studies. Nutr Res Rev. 33:77–89. 2020. View Article : Google Scholar

22 

Gill V, Kumar V, Singh K, Kumar A and Kim JJ: Advanced glycation end products (AGEs) may be a striking link between modern diet and health. Biomolecules. 9:8882019. View Article : Google Scholar : PubMed/NCBI

23 

Lin JA, Wu CH and Yen GC: Perspective of advanced glycation end products on human health. J Agric Food Chem. 66:2065–2070. 2018. View Article : Google Scholar : PubMed/NCBI

24 

van Dongen KCW, Kappetein L, Miro Estruch I, Belzer C, Beekmann K and Rietjens IMCM: Differences in kinetics and dynamics of endogenous versus exogenous advanced glycation end products (AGEs) and their precursors. Food Chem Toxicol. 164:1129872022. View Article : Google Scholar : PubMed/NCBI

25 

Srebreva LN, Stoynev GA and Ivanov IG: Evidence for excretion of glycation agents from E. coli cells during growth. Biotechnol Biotechnol Equip. 23:1068–1071. 2009. View Article : Google Scholar

26 

Baskaran S, Rajan DP and Balasubramanian KA: Formation of methylglyoxal by bacteria isolated from human faeces. J Med Microbiol. 28:211–215. 1989. View Article : Google Scholar : PubMed/NCBI

27 

Zhao C, Dong H, Zhang Y and Li Y: Discovery of potential genes contributing to the biosynthesis of short-chain fatty acids and lactate in gut microbiota from systematic investigation in E. coli. NPJ Biofilms Microbiomes. 5:192019. View Article : Google Scholar : PubMed/NCBI

28 

Popkov VA, Zharikova AA, Demchenko EA, Andrianova NV, Zorov DB and Plotnikov EY: Gut microbiota as a source of uremic toxins. Int J Mol Sci. 23:4832022. View Article : Google Scholar : PubMed/NCBI

29 

Chakraborty S, Karmakar K and Chakravortty D: Cells producing their own nemesis: Understanding methylglyoxal metabolism. IUBMB Life. 66:667–678. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Lee C and Park C: Bacterial responses to glyoxal and methylglyoxal: reactive electrophilic species. Int J Mol Sci. 18:1692017. View Article : Google Scholar : PubMed/NCBI

31 

Rabie E, Serem JC, Oberholzer HM, Gaspar AR and Bester MJ: How methylglyoxal kills bacteria: An ultrastructural study. Ultrastruct Pathol. 40:107–111. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Booth IR, Ferguson GP, Miller S, Li C, Gunasekera B and Kinghorn S: Bacterial production of methylglyoxal: A survival strategy or death by misadventure? Biochem Soc Trans. 31:1406–1408. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Ferguson GP, Tötemeyer S, MacLean MJ and Booth IR: Methylglyoxal production in bacteria: Suicide or survival? Arch Microbiol. 170:209–218. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Hellwig M, Bunzel D, Huch M, Franz CMAP, Kulling SE and Henle T: Stability of individual maillard reaction products in the presence of the human colonic microbiota. J Agric Food Chem. 63:6723–6730. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Bui TPN, Troise AD, Fogliano V and de Vos WM: Anaerobic degradation of N-ε-carboxymethyllysine, a major glycation end-product, by human intestinal bacteria. J Agric Food Chem. 67:6594–6602. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Bui TPN, Troise AD, Nijsse B, Roviello GN, Fogliano V and de Vos WM: Intestinimonas-like bacteria are important butyrate producers that utilize Nε-fructosyllysine and lysine in formula-fed infants and adults. J Funct Foods. 70:1039742020. View Article : Google Scholar

37 

Hellwig M, Auerbach C, Müller N, Samuel P, Kammann S, Beer F, Gunzer F and Henle T: Metabolization of the advanced glycation end product N-ε-carboxymethyllysine (CML) by different probiotic E. coli Strains. J Agric Food Chem. 67:1963–1972. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Brunt VE, Casso AG, Gioscia-Ryan RA, Sapinsley ZJ, Ziemba BP, Clayton ZS, Bazzoni AE, VanDongen NS, Richey JJ, Hutton DA, et al: Gut microbiome-derived metabolite trimethylamine N-oxide induces aortic stiffening and increases systolic blood pressure with aging in mice and humans. Hypertension. 78:499–511. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Taguchi K, Fukami K, Elias BC and Brooks CR: Dysbiosis-related advanced glycation endproducts and trimethylamine N-oxide in chronic kidney disease. Toxins (Basel). 13:3612021. View Article : Google Scholar : PubMed/NCBI

40 

Scott KP, Gratz SW, Sheridan PO, Flint HJ and Duncan SH: The influence of diet on the gut microbiota. Pharmacol Res. 69:52–60. 2013. View Article : Google Scholar

41 

Wu S, Bhat ZF, Gounder RS, Mohamed Ahmed IA, Al-Juhaimi FY, Ding Y and Bekhit AEA: Effect of dietary protein and processing on gut microbiota-A systematic review. Nutrients. 14:4532022. View Article : Google Scholar : PubMed/NCBI

42 

Mills DJS, Tuohy KM, Booth J, Buck M, Crabbe MJC, Gibson GR and Ames JM: Dietary glycated protein modulates the colonic microbiota towards a more detrimental composition in ulcerative colitis patients and non-ulcerative colitis subjects. J Appl Microbiol. 105:706–714. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Seiquer I, Rubio LA, Peinado MJ, Delgado-Andrade C and Navarro MP: Maillard reaction products modulate gut microbiota composition in adolescents. Mol Nutr Food Res. 58:1552–1560. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Qu W, Yuan X, Zhao J, Zhang Y, Hu J, Wang J and Li J: Dietary advanced glycation end products modify gut microbial composition and partially increase colon permeability in rats. Mol Nutr Food Res. 61:2017. View Article : Google Scholar : PubMed/NCBI

45 

Qu W, Nie C, Zhao J, Ou X, Zhang Y, Yang S, Bai X, Wang Y, Wang J and Li J: Microbiome-metabolomics analysis of the impacts of long-term dietary advanced-glycation-end-product consumption on C57BL/6 mouse fecal microbiota and metabo-lites. J Agric Food Chem. 66:8864–8875. 2018. View Article : Google Scholar : PubMed/NCBI

46 

van Dongen KCW, Linkens AMA, Wetzels SMW, Wouters K, Vanmierlo T, van de Waarenburg MPH, Scheijen JLJM, de Vos WM, Belzer C and Schalkwijk CG: 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. 147:1105472021. View Article : Google Scholar : PubMed/NCBI

47 

Mastrocola R, Collotta D, Gaudioso G, Le Berre M, Cento AS, Ferreira Alves G, 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. 12:24972020. View Article : Google Scholar : PubMed/NCBI

48 

Gaudioso G, Collotta D, Chiazza F, Mastrocola R, Cento A, Fava F, Aragno M, Collino M and Tuohy K: Advanced glycation end products (AGEs) in metabolic disease: Linking diet, inflammation and microbiota. Proc Nutr Soc. 79:E3682020. View Article : Google Scholar

49 

Han K, Yao Y, Dong S, Jin S, Xiao H, Wu H and Zeng M: Chemical characterization of the glycated myofibrillar proteins from grass carp (Ctenopharyngodon idella) and their impacts on the human gut microbiota in vitro fermentation. Food Funct. 8:1184–1194. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Han K, Jin W, Mao Z, Dong S, Zhang Q, Yang Y, Chen B, Wu H and Zeng M: Microbiome and butyrate production are altered in the gut of rats fed a glycated fish protein diet. J Funct Foods. 47:423–433. 2018. View Article : Google Scholar

51 

Jin W, Han K, Dong S, Yang Y, Mao Z, Su M and Zeng M: Modifications in gut microbiota and fermentation metabolites in the hindgut of rats after the consumption of galactooligosaccharide glycated with a fish peptide. Food Funct. 9:2853–2864. 2018. View Article : Google Scholar : PubMed/NCBI

52 

Yang Y, Wu H, Dong S, Jin W, Han K, Ren Y and Zeng M: Glycation of fish protein impacts its fermentation metabolites and gut microbiota during in vitro human colonic fermentation. Food Res Int. 113:189–196. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Mao Z, Ren Y, Zhang Q, Dong S, Han K, Feng G, Wu H and 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. 10:3439–3451. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Corzo-Martínez M, Ávila M, Moreno FJ, Requena T and Villamiel M: Effect of milk protein glycation and gastrointestinal digestion on the growth of bifidobacteria and lactic acid bacteria. Int J Food Microbiol. 153:420–427. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Corzo-Martínez M, Hernandez-Hernandez O, Villamiel M, Rastall RA and Moreno FJ: In vitro bifidogenic effect of Maillard-type milk protein-galactose conjugates on the human intestinal microbiota. Int Dairy J. 31:127–131. 2013. View Article : Google Scholar

56 

Shao YH, Zhang Y, Zhang L, Liu J and Tu ZC: Mechanism of reduction in allergenicity and altered human intestinal microbiota of digested β-lactoglobulin modified by ultrasonic pretreatment combined with glycation. J Agric Food Chem. 69:14004–14012. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Chen Y, Guo KM, Nagy T and Guo TL: Chronic oral exposure to glycated whey proteins increases survival of aged male NOD mice with autoimmune prostatitis by regulating the gut microbiome and anti-inflammatory responses. Food Funct. 11:153–162. 2020. View Article : Google Scholar :

58 

Świątecka D, Narbad A, Ridgway KP and Kostyra H: The study on the impact of glycated pea proteins on human intestinal bacteria. Int J Food Microbiol. 145:267–272. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Yacoub R, Nugent M, Cai W, Nadkarni GN, Chaves LD, Abyad S, Honan AM, Thomas SA, Zheng W, Valiyaparambil SA, 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. 12:e01847892017. View Article : Google Scholar : PubMed/NCBI

60 

Linkens AMA, van Best N, Niessen PM, Wijckmans NEG, de Goei EEC, Scheijen JLJM, van Dongen MCJM, van Gool CCJAW, de Vos WM, Houben AJHM, 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. 23:53282022. View Article : Google Scholar : PubMed/NCBI

61 

Cao C, Tang M, Zhao N, Dong S and Wu H: Effects of fish protein with glycation extent on gut microbiota and colonic barrier function in mice fed a high-fat diet. J Funct Foods. 85:1046362021. View Article : Google Scholar

62 

Yuan X, Nie C, Liu H, Ma Q, Peng B, Zhang M, Chen Z and Li J: Comparison of metabolic fate, target organs, and microbiota interactions of free and bound dietary advanced glycation end products. Crit Rev Food Sci Nutr. Oct 26–2021.Epub ahead of print. View Article : Google Scholar

63 

Zhao D, Sheng B, Wu Y, Li H, Xu D, Nian Y, Mao S, Li C, Xu X and Zhou G: Comparison of free and bound advanced glycation end products in food: A review on the possible influence on human health. J Agric Food Chem. 67:14007–14018. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Xu D, Li L, Zhang X, Yao H, Yang M, Gai Z, Li B and Zhao D: Degradation of peptide-bound maillard reaction products in gastrointestinal digests of glyoxal-glycated casein by human colonic microbiota. J Agric Food Chem. 67:12094–12104. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Gurung M, Li Z, You H, Rodrigues R, Jump DB, Morgun A and Shulzhenko N: Role of gut microbiota in type 2 diabetes pathophysiology. EBioMedicine. 51:1025902020. View Article : Google Scholar : PubMed/NCBI

66 

Wu Y, Zong M, Wu H, He D, Li L, Zhang X, Zhao D and Li B: Dietary advanced glycation end-products affects the progression of early diabetes by intervening in carbohydrate and lipid metabolism. Mol Nutr Food Res. 66:e22000462022. View Article : Google Scholar : PubMed/NCBI

67 

Wang J, Cai W, Yu J, Liu H, He S, Zhu L and Xu J: Dietary advanced glycation end products shift the gut microbiota composition and induce insulin resistance in mice. Diabetes Metab Syndr Obes. 15:427–437. 2022. View Article : Google Scholar : PubMed/NCBI

68 

Su L, Hong Z, Zhou T, Jian Y, Xu M, Zhang X, Zhu X and Wang J: Health improvements of type 2 diabetic patients through diet and diet plus fecal microbiota transplantation. Sci Rep. 12:11522022. View Article : Google Scholar : PubMed/NCBI

69 

Murros KE, Huynh VA, Takala TM and Saris PEJ: Desulfovibrio bacteria are associated with Parkinson's disease. Front. Cell Infect Microbiol. 11:6526172021. View Article : Google Scholar

70 

Chen YR, Jing QL, Chen FL, Zheng H, Chen LD and Yang ZC: Desulfovibrio is not always associated with adverse health effects in the guangdong gut microbiome project. PeerJ. 9:e120332021. View Article : Google Scholar : PubMed/NCBI

71 

Snelson M, Tan SM, Sourris K, Thallas-Bonke V, Ziemann M, El-Osta S, Cooper M and Coughlan M: SAT-301 resistant starch ameliorates advanced glycation endproduct-induced gut dysbiosis and albuminuria in a mouse model of type 2 diabetes. Kidney Int Rep. 4(Suppl): S1342019. View Article : Google Scholar

72 

Reddy VP, Aryal P and Darkwah EK: Advanced glycation end products in health and disease. Microorganisms. 10:18482022. View Article : Google Scholar : PubMed/NCBI

73 

Semba RD, Nicklett EJ and Ferrucci L: Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci. 65:963–975. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Maynard C and Weinkove D: The gut microbiota and ageing. Subcell Biochem. 90:351–371. 2018. View Article : Google Scholar

75 

Mossad O, Batut B, Yilmaz B, Dokalis N, Mezö C, Nent E, Nabavi LS, Mayer M, Maron FJM, Buescher JM, et al: Gut microbiota drives age-related oxidative stress and mitochondrial damage in microglia via the metabolite N6-carboxymethyllysine. Nat Neurosci. 25:295–305. 2022. View Article : Google Scholar : PubMed/NCBI

76 

Li Y, Ning L, Yin Y, Wang R, Zhang Z, Hao L, Wang B, Zhao X, Yang X, Yin L, et al: Age-related shifts in gut microbiota contribute to cognitive decline in aged rats. Aging (Albany NY). 12:7801–7817. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Guilherme MDS, Nguyen VTT, Reinhardt C and Endres K: Impact of gut microbiome manipulation in 5xFAD mice on Alzheimer's disease-like pathology. Microorganisms. 9:8152021. View Article : Google Scholar : PubMed/NCBI

78 

Ohira H, Tsuruya A, Oikawa D, Nakagawa W, Mamoto R, Hattori M, Waki T, Takahashi S, Fujioka Y and Nakayama T: Alteration of oxidative-stress and related marker levels in mouse colonic tissues and fecal microbiota structures with chronic ethanol administration: Implications for the pathogenesis of ethanol-related colorectal cancer. PLoS One. 16:e02465802021. View Article : Google Scholar : PubMed/NCBI

79 

Bonfili L, Cecarini V, Gogoi O, Berardi S, Scarpona S, Angeletti M, Rossi G and Eleuteri AM: Gut microbiota manipulation through probiotics oral administration restores glucose homeostasis in a mouse model of Alzheimer's disease. Neurobiol Aging. 87:35–43. 2020. View Article : Google Scholar

80 

Mihailović M, Živković M, Jovanović JA, Tolinački M, Sinadinović M, Rajić J, Uskoković A, Dinić S, Grdović N, Golić N and Vidaković M: Oral administration of probiotic Lactobacillus paraplantarum BGCG11 attenuates diabetes-induced liver and kidney damage in rats. J Funct Foods. 38:427–437. 2017. View Article : Google Scholar

81 

Al-Enazi AMM, Virk P, Hindi A, Awad MA, Elobeid M and Qindeel R: Protective effect of probiotic bacteria and its nanoformulation against cadmium-induced oxidative stress in male Wistar rat. J King Saud Univ Sci. 32:3045–3051. 2020. View Article : Google Scholar

82 

Park HY, Lee HB, Lee SY, Oh MJ, Ha SK, Do E, Lee HHL, Hur J, Lee KW, Nam MH, et al: Lactococcus lactis KF140 reduces dietary absorption of Nε-(Carboxymethyl)lysine in rats and humans via β-galactosidase activity. Front Nutr. 9:9162622022. View Article : Google Scholar

83 

Farhangi MA, Dehghan P and Namazi N: Prebiotic supple-mentation modulates advanced glycation end-products (AGEs), soluble receptor for AGEs (sRAGE), and cardiometabolic risk factors through improving metabolic endotoxemia: A randomized-controlled clinical trial. Eur J Nutr. 59:3009–3021. 2020. View Article : Google Scholar

84 

Li Y, Peng Y, Shen Y, Zhang Y, Liu L and Yang X: Dietary polyphenols: Regulate the advanced glycation end products-RAGE axis and the microbiota-gut-brain axis to prevent neurodegenerative diseases. Crit Rev Food Sci Nutr. May 19–2022.Epub ahead of print.

85 

Wu Y, Dong L, Song Y, Wu Y, Zhang Y and 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. 204:204–214. 2022. View Article : Google Scholar : PubMed/NCBI

86 

Yang S, Zhou H, Wang G, Zhong XH, Shen QL, Zhang XJ, Li RY, Chen LH, Zhang YH and Wan Z: Quercetin is protective against short-term dietary advanced glycation end products intake induced cognitive dysfunction in aged ICR mice. J Food Biochem. 44:e131642020. View Article : Google Scholar : PubMed/NCBI

87 

Yonei Y, Ikeda T, Ogawa H, Yagi N, Takabe W, Ito M and Morii H: Anti-glycation and improvement microbiota by Geranium dielsianum extract: Relation to health problems in athletes. Glycative Stress Res. 6:31–38. 2019.

88 

Di Lorenzo F, De Castro C, Silipo A and Molinaro A: Lipopolysaccharide structures of Gram-negative populations in the gut microbiota and effects on host interactions. FEMS Microbiol Rev. 43:257–272. 2019. View Article : Google Scholar : PubMed/NCBI

89 

Mohr AE, Crawford M, Jasbi P, Fessler S and Sweazea KL: Lipopolysaccharide and the gut microbiota: Considering structural variation. FEBS Lett. 596:849–875. 2022. View Article : Google Scholar : PubMed/NCBI

90 

Aki T, Funakoshi T, Noritake K, Unuma K and Uemura K: Extracellular glucose is crucially involved in the fate decision of LPS-stimulated RAW264.7 murine macrophage cells. Sci Rep. 10:105812020. View Article : Google Scholar : PubMed/NCBI

91 

Dhananjayan K, Gunawardena D, Hearn N, Sonntag T, Moran C, Gyengesi E, Srikanth V and Münch G: Activation of macrophages and microglia by interferon-γ and lipopolysaccharide increases methylglyoxal production: A new mechanism in the development of vascular complications and cognitive decline in type 2 diabetes mellitus? J Alzheimers Dis. 59:467–479. 2017. View Article : Google Scholar

92 

Ko YH, Tsai MS, Lee PH, Liang JT and Chang KC: Methylprednisolone stiffens aortas in lipopolysaccharide-induced chronic inflammation in rats. PLoS One. 8:e696362013. View Article : Google Scholar : PubMed/NCBI

93 

Shamsaldeen YA, Alsugoor MH, Lione LA and Benham CD: Dysfunction in nitric oxide synthesis in streptozotocin treated rat aorta and role of methylglyoxal. Eur J Pharmacol. 842:321–328. 2019. View Article : Google Scholar

94 

Kitaura A, Nishinaka T, Hamasaki S, Hatipoglu OF, Wake H, Nishibori M, Mori S, Nakao S and Takahashi H: Advanced glycation end-products reduce lipopolysaccharide uptake by macrophages. PLoS One. 16:e02459572021. View Article : Google Scholar : PubMed/NCBI

95 

Chiu HC, Fu MM, Yang TS, Fu E, Chiang CY, Tu HP, Chin YT, Lin FG and Shih KC: Effect of high glucose, Porphyromonas gingivalis lipopolysaccharide and advanced glycation end-products on production of interleukin-6/-8 by gingival fibroblasts. J Periodontal Res. 52:268–276. 2017. View Article : Google Scholar

96 

Liu J, Zhao S, Tang J, Li Z, Zhong T, Liu Y, Chen D, Zhao M, Li Y, Gong X, et al: Advanced glycation end products and lipopolysaccharide synergistically stimulate proinflammatory cytokine/chemokine production in endothelial cells via activation of both mitogen-activated protein kinases and nuclear factor-kappaB. FEBS J. 276:4598–4606. 2009. View Article : Google Scholar : PubMed/NCBI

97 

Yamamoto Y and Yamamoto H: Interaction of receptor for advanced glycation end products with advanced oxidation protein products induces podocyte injury. Kidney Int. 82:733–735. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Pinkas A, Cunha Martins A Jr and Aschner M: C. elegans-an emerging model to study metal-induced RAGE-related pathologies. Int J Environ Res Public Health. 15:14072018. View Article : Google Scholar : PubMed/NCBI

99 

Lawes M, Pinkas A, Frohlich BA, Iroegbu JD, Ijomone OM and Aschner M: Metal-induced neurotoxicity in a RAGE-expressing C. elegans model. Neurotoxicology. 80:71–75. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Wang L, Wu J, Guo X, Huang X and Huang Q: RAGE plays a role in LPS-induced NF-κB activation and endothelial hyperpermeability. Sensors (Basel). 17:7222017. View Article : Google Scholar

101 

Li Y, Wu R, Tian Y, Yu M, Tang Y, Cheng H and Tian Z: RAGE/NF-κB signaling mediates lipopolysaccharide induced acute lung injury in neonate rat model. Int J Clin Exp Med. 8:13371–13376. 2015.

102 

Li Y, Wu R, Zhao S, Cheng H, Ji P, Yu M and Tian Z: RAGE/NF-κB pathway mediates lipopolysaccharide-induced inflammation in alveolar type I epithelial cells isolated from neonate rats. Inflammation. 37:1623–1629. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Yamamoto Y, Harashima A, Saito H, Tsuneyama K, Munesue S, Motoyoshi S, Han D, Watanabe T, Asano M, Takasawa S, et al: Septic shock is associated with receptor for advanced glycation end products ligation of LPS. J Immunol. 186:3248–3257. 2011. View Article : Google Scholar : PubMed/NCBI

104 

Watanabe M, Toyomura T, Tomiyama M, Wake H, Liu K, Teshigawara K, Takahashi H, Nishibori M and 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. 47:7153–7159. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Huang J, Xiong T, Zhang Z, Tan Y and Guo L: Inhibition of the receptor for advanced glycation inhibits lipopolysaccharide-mediated high mobility group protein B1 and Interleukin-6 synthesis in human gingival fibroblasts through the NF-κB signaling pathway. Arch Oral Biol. 105:81–87. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Cho HI, Hong JM, Choi JW, Choi HS, Hwan Kwak J, Lee DU, Kook Lee S and Lee SM: β-Caryophyllene alleviates D-galactosamine and lipopolysaccharide-induced hepatic injury through suppression of the TLR4 and RAGE signaling pathways. Eur J Pharmacol. 764:613–621. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Goel R, Bhat SA, Hanif K, Nath C and Shukla R: Perindopril attenuates lipopolysaccharide-induced amyloidogenesis and memory impairment by suppression of oxidative stress and RAGE activation. ACS Chem Neurosci. 7:206–217. 2016. View Article : Google Scholar

108 

Liu L, Zhao Z, Lu L, Liu J, Sun J, Wu X and Dong J: Icariin and icaritin ameliorated hippocampus neuroinflammation via inhibiting HMGB1-related pro-inflammatory signals in lipopolysaccharide-induced inflammation model in C57BL/6 J mice. Int Immunopharmacol. 68:95–105. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Aneja RK, Tsung A, Sjodin H, Gefter JV, Delude RL, Billiar TR and Fink MP: Preconditioning with high mobility group box 1 (HMGB1) induces lipopolysaccharide (LPS) tolerance. J Leukoc Biol. 84:1326–1334. 2008. View Article : Google Scholar : PubMed/NCBI

110 

Zhou XY, Zhang WJ, Huang QB and Guo XH: Role of RAGE in lipopolysaccharide-induced cytoskeletal changes in mouse pulmonary microvascular endothelial cells. Nan Fang Yi Ke Da Xue Xue Bao. 35:6–11. 2015.In Chinese. PubMed/NCBI

111 

Wang H, Wang T, Yuan Z, Cao Y, Zhou Y, He J, Shen Y, Zeng N, Dai L, Wen F and Chen L: Role of receptor for advanced glycation end products in regulating lung fluid balance in lipopolysaccharide-induced acute lung injury and infection-related acute respiratory distress syndrome. Shock. 50:472–482. 2018. View Article : Google Scholar

112 

Li J, Wang K, Huang B, Li R, Wang X, Zhang H, Tang H and Chen X: The receptor for advanced glycation end products mediates dysfunction of airway epithelial barrier in a lipopolysaccharides-induced murine acute lung injury model. Int Immunopharmacol. 93:1074192021. View Article : Google Scholar : PubMed/NCBI

113 

Shi J, Fu Y, Zhao XH and Lametsch R: Glycation sites and bioactivity of lactose-glycated caseinate hydrolysate in lipopolysaccharide-injured IEC-6 cells. J Dairy Sci. 104:1351–1363. 2021. View Article : Google Scholar

114 

You L, Cui H, Zhao F, Sun H, Zhong H, Zhou G and Chen X: Inhibition of HMGB1/RAGE axis suppressed the lipopolysaccharide (LPS)-induced vicious transformation of cervical epithelial cells. Bioengineered. 12:4995–5003. 2021. View Article : Google Scholar : PubMed/NCBI

115 

Wilkie T, Verma AK, Zhao H, Charan M, Ahirwar DK, Kant S, Pancholi V, Mishra S and Ganju RK: Lipopolysaccharide from the commensal microbiota of the breast enhances cancer growth: Role of S100A7 and TLR4. Mol Oncol. 16:1508–1522. 2022. View Article : Google Scholar :

116 

Ramsgaard L, Englert JM, Manni ML, Milutinovic PS, Gefter J, Tobolewski J, Crum L, Coudriet GM, Piganelli J, Zamora R, et al: Lack of the receptor for advanced glycation end-products attenuates E. coli pneumonia in mice. PLoS One. 6:e201322011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2023
Volume 51 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Aschner M, Skalny AV, Gritsenko VA, Kartashova OL, Santamaria A, Rocha JB, Spandidos DA, Zaitseva IP, Tsatsakis A, Tinkov AA, Tinkov AA, et al: Role of gut microbiota in the modulation of the health effects of advanced glycation end‑products (Review). Int J Mol Med 51: 44, 2023
APA
Aschner, M., Skalny, A.V., Gritsenko, V.A., Kartashova, O.L., Santamaria, A., Rocha, J.B. ... Tinkov, A.A. (2023). Role of gut microbiota in the modulation of the health effects of advanced glycation end‑products (Review). International Journal of Molecular Medicine, 51, 44. https://doi.org/10.3892/ijmm.2023.5247
MLA
Aschner, M., Skalny, A. V., Gritsenko, V. A., Kartashova, O. L., Santamaria, A., Rocha, J. B., Spandidos, D. A., Zaitseva, I. P., Tsatsakis, A., Tinkov, A. A."Role of gut microbiota in the modulation of the health effects of advanced glycation end‑products (Review)". International Journal of Molecular Medicine 51.5 (2023): 44.
Chicago
Aschner, M., Skalny, A. V., Gritsenko, V. A., Kartashova, O. L., Santamaria, A., Rocha, J. B., Spandidos, D. A., Zaitseva, I. P., Tsatsakis, A., Tinkov, A. A."Role of gut microbiota in the modulation of the health effects of advanced glycation end‑products (Review)". International Journal of Molecular Medicine 51, no. 5 (2023): 44. https://doi.org/10.3892/ijmm.2023.5247