Next Article in Journal
Reply to Gurney, T.; Ronca, F. Comment on “Hack et al. Effect of Guarana (Paullinia cupana) on Cognitive Performance: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 434”
Next Article in Special Issue
Advanced Glycation End Products (AGEs) in Diet and Skin in Relation to Stool Microbiota: The Rotterdam Study
Previous Article in Journal
Comment on Hack et al. Effect of Guarana (Paullinia cupana) on Cognitive Performance: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 434
Previous Article in Special Issue
Skin Autofluorescence Mirrors Surrogate Parameters of Vascular Aging: An Enable Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Blood and Tissue Advanced Glycation End Products as Determinants of Cardiometabolic Disorders Focusing on Human Studies

Department of Food and Nutrition, Institute of Agriculture and Life Science, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Gyeongsangnam-do, Republic of Korea
Nutrients 2023, 15(8), 2002; https://doi.org/10.3390/nu15082002
Submission received: 4 April 2023 / Revised: 18 April 2023 / Accepted: 20 April 2023 / Published: 21 April 2023

Abstract

:
Cardiometabolic disorders are characterised by a cluster of interactive risk determinants such as increases in blood glucose, lipids and body weight, as well as elevated inflammation and oxidative stress and gut microbiome changes. These disorders are associated with onset of type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). T2DM is strongly associated with CVD. Dietary advanced glycation end products (dAGEs) attributable from modern diets high in sugar and/or fat, highly processed foods and high heat-treated foods can contribute to metabolic etiologies of cardiometabolic disorders. This mini review aims to determine whether blood dAGEs levels and tissue dAGEs levels are determinants of the prevalence of cardiometabolic disorders through recent human studies. ELISA (enzyme-linked immunosorbent assay), high-performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS) for blood dAGEs measurement and skin auto fluorescence (SAF) for skin AGEs measurement can be used. Recent human studies support that a diet high in AGEs can negatively influence glucose control, body weight, blood lipid levels and vascular health through the elevated oxidative stress, inflammation, blood pressure and endothelial dysfunction compared with a diet low in AGEs. Limited human studies suggested a diet high in AGEs could negatively alter gut microbiota. SAF could be considered as one of the predictors affecting risks for cardiometabolic disorders. More intervention studies are needed to determine how dAGEs are associated with the prevalence of cardiometabolic disorders through gut microbiota changes. Further human studies are conducted to find the association between CVD events, CVD mortality and total mortality through SAF measurement, and a consensus on whether tissue dAGEs act as a predictor of CVD is required.

1. Introduction

Human studies showed the association between plasma concentrations of advanced glycation end products (AGEs) and cardiometabolic disorders [1,2,3]. The association between higher blood circulating AGEs levels and higher insulin resistance was observed [2,3], while other studies [4,5,6] showed the association between lower blood circulating AGEs levels and higher insulin resistance. Moreover, AGEs accumulation in tissues can increase risk of cardiovascular disease (CVD), especially in subjects with diabetes mellitus and coronary artery disease [7].
Several papers [8,9] have reviewed the purpose of dietary advanced glycation end products (dAGEs) on cardiometabolic disorders. Clarke et al. 2016 [8] conducted a systematic review of 12 randomised controlled trials (11 publications [10,11,12,13,14,15,16,17,18,19,20]) to examine dAGEs on risk for chronic disease. They concluded that AGEs high in diet appeared to increase risk for chronic disease attributable from oxidative stress and inflammation. Luévano-Contreras et al. 2017 [9] concluded that dAGEs contributed to risk for cardiometabolic disorders by endogenously interacting with AGEs. A diet high in N-epsilon-(Carboxymethyl) lysine (CML) elevated endothelial dysfunction and arterial aging. Subjects with type 2 diabetes mellitus (T2DM) or higher risk for cardiometabolic disorders who consumed high dAGEs were more likely to have increased oxidative stress and markers of inflammation and endothelial dysfunction [9].
Therefore, the aims of this mini review are as follows: (a) to examine the effect of dietary blood AGEs levels on cardiometabolic disorders based on recent human studies; (b) to examine the role of skin AGEs levels on cardiometabolic disorders.

2. Advanced Glycation End Products

2.1. AGEs Formation

AGEs are stable, irreversible end products generated from nonenzymatic reaction between reducing sugars with amino groups or lipids or nucleic acids through the Maillard reaction forming Schiff base/Amadori products which can be promoted by high temperature, resulting in features of yellow-brown colour [21,22]. AGEs can be endogenously formed but can be exogenously sourced from diet (highly processed, high temperature-treated foods), pollution and smoking [23,24,25,26]. The high-temperature cooking techniques such as dry heat (e.g., roasting, grilling, baking and deep-frying) can lead to the Maillard reaction [27]. The quantity of AGEs consumed is obviously determined by the type of food, cooking method and degree of processing in industry [24,28]. Foods high in lipid and protein (e.g., particularly cheese, butter and red meat) when treated by dry heat contain a high quantity of AGEs [24,29]. Food items processed in industry (e.g., baked, bakery products) contain considerably high AGEs [30], while whole grains, milk, fruits and vegetables are relatively low in AGEs [24].
The endogenous formation of AGEs is involved in several pathways (non-enzymatic Maillard reaction, polyol pathway and lipid peroxidation) [31,32,33]. AGEs can be endogenously produced as products of Schiff base/Amadori products or/and through the polyol pathway [32]. A number of chemical compounds can be generated through the Maillard reaction. These chemical compounds are classified into reactive intermediates and stable end products of AGEs [32]. The Maillard reaction can occur; the early step includes condensation resulting from non-enzymatic reaction between carbonyl groups of reducing sugars (aldose or ketose) and amine groups of nucleic acids, proteins (free arginine, lysine, cysteine, histidine and tryptophan), or lipids, leading to endogenous formation of reversible intermediate products such as Schiff’s base, and then Amadori rearrangement and subsequent glycoxidation for AGEs production [34]. AGEs are a group of heterogeneous, cross-link formation through glycation process. The AGEs formation can be promoted in persistent hyperglycaemia (e.g., in diabetes mellitus) [35]. Elevated glucose levels are associated with oxidative stress, glucose auto oxidation and activated polyol pathway [36,37]. Hyperglycaemia condition promotes lipid peroxidation leading to production of advanced lipid peroxidation end products (ALEs) [38,39]. In addition, the Namiki pathway (aldimins) and the Wolff pathway (auto-oxidation of carbonyl compounds or monosaccharides) through reactive oxygen species (ROS) or transition metals can be involved in AGEs production [40]. Dicarbonyl compounds (highly reactive molecules of protein cross-links) such as glyoxal, methylglyoxal (MG) and 3-deoxyglucosone can be produced through oxidative degradation or auto-oxidation of Amadori products [41]. Several AGEs are not exclusively produced through the Maillard reaction. Especially, CML (glucose-lysine or threose-lysine) can be generated through oxidative degradation of Amadori products or direct reaction of glyoxal with lysine and lipid peroxidation [42,43].

2.2. Characteristics of AGEs

The properties and chemical structures of AGEs (e.g., fluorescent, cross-linking and weights) can be determined through several pathways and/or various AGEs precursors involved in AGEs production. AGEs can be classified with several groups based on their sources, precursors non-fluorescent or fluorescent, non-cross-linked or cross-linked, molecular weight, receptor for AGEs (RAGE) ligands and physiological importance (toxic or non-toxic) [34]. Pentosidine is a fluorescent glycoxidation product and protein–protein cross-linked AGEs. Argpyrimidine is a fluorescent and non-cross-linked AGE. N-epsilon-(1-carboxyethyl) lysine (CEL), CML and pyrraline are non-fluorescent and non-cross-linked AGEs. Glyoxyl-lysine dimer (GOLD), methylglyoxal-lysine dimer (MOLD) and 3-deoxyglucosone-lysine dimer (DOLD) are non-fluorescent and cross-linked AGEs [34].
AGEs less than 12 kDa are classified as low weight molecules. AGEs greater than 12 kDa are regarded as large weight molecules. High-molecular AGEs are known as protein-binding proteins, and small-molecular AGEs are known as free proteins or peptide-binding molecules. Low-molecular-weight AGEs can become high molecules by heat treatment [44,45].
The extent of dAGEs absorption depends on their chemical structures, molecular weights and hydrophobicity (e.g., pyrraline and argpyrimidine). Highly hydrophobic AGEs easily pass through the basolateral membrane compared with hydrophilic AGEs (e.g., CML, CEL and MG-H1) [34,46,47]. Approximately one-third of AGEs are excreted in the urine. However, less than 5% of AGEs are excreted in patients with diabetes [48].
Circulating AGEs contents in the whole body can be attributed by the balance between endogenous AGEs synthesis, AGEs tissue accumulation, exogenous AGEs absorption from food and AGEs clearance in the kidney [7,24,49,50]. Plasma AGEs levels have been used as biomarkers to examine the incidence of cardiometabolic disorders.
Several potential treatment options for AGEs-mediated cardiometabolic disorders are recognised as follows: (a) AGEs formation inhibition [e.g., aminoguanidine, 2, 3-diamino-phenazine, tenilsetam, pyridoxamine, arginine, 2-isopropylidenehydrazono-4-oxo-thiazolidin-5-ylacetanidide (OPB-9195); (b) breaking down Maillard cross-links [e.g., 4, 5-dimethyl-3-phenacylthiozolium chloride (ALT-711), N-(2-acetamidoethyl) hydrazine carboximidamide hydrochloride (ALT-946), N-phenacetylthiazolium bromide (PTB) and alagebrium]; (c) RAGE inhibitors (e.g., anti-RAGE antibodies, statins and curcumin); (d) redox regulators (e.g., aldose reductase inhibitors, angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers); (e) antioxidants (e.g., vitamin C, vitamin E, α-tocopherol, taurine, niacinamide, oxerutin pyridoxal, lipoic acid, N-acetylcysteine and epigallocatechin-3-gallat) [51,52,53,54,55,56,57,58,59].

2.3. AGEs Measurements

Various AGEs quantification techniques have been developed, including biochemical and immunohistochemical methods to measure pentosidine and CML. ELISA (enzyme-linked immunosorbent assay) utilising AGEs-recognising antibodies (Ac) have been designed [60,61]. Moreover, high-performance liquid chromatography (HPLC), liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS) can determine plasma concentrations of AGEs according to the fluorescence intensity [62,63,64]. Skin auto fluorescence (SAF) can assess AGEs in skin [65,66,67].

3. Dietary Patterns Influence on Carbonyl Stress or Blood AGEs Levels

Certain diets low in AGEs may attenuate a total AGEs level in the body contributing to a lower risk of cardiometabolic disorders. In observational studies [48,68], a diet high in AGEs (AGEs, CML, CEL and MG-H1) was associated with higher concentrations of CML, CEL and MG-H1 in plasma and urine [48]. Maasen et al.al. 2019 [68] conducted a cross-sectional Diabetes and Atherosclerosis Maastricht (CODAM) study of 574 subjects with a moderately increased risk of T2DM and CVD, in order to examine the effect of carbohydrate quality [glycaemic index (GI)] and quantity [glycaemic load (GL)] in a habitual diet on dicarbonyls and AGEs. They found the association between higher GL diet and higher levels of free urinary MG-H1 [68].
In randomised controlled trials (RCTs) [69,70], Delgado-Andrade et al. 2011 [69] examined the influence of Millard reaction products (MRP) diet on CML (one of the most biologically active MRP), consumption and excretion comparing a MRP-high diet with a MRP-low diet in 20 healthy male adolescents aged 11–14 years. A MRP-high diet (high in processed browning food: chocolates, corn flakes, baked products, fried and breaded foods) elevated CML levels and CML faecal excretion compared with a MRP-low diet, indicating that dietary CML levels affect CML absorption and CML faecal excretion [69]. Brinkley et al. 2020 [70] showed the association between higher dietary protein intake (≥1.2 g/kg/d) and over 5–10% higher levels of CML and soluble RAGE (sRAGE), compared with lower protein intakes in older subjects aged mean 73 years [70].
The Mediterranean diet is a dietary pattern rich in whole grains, legumes, nuts, vegetables, fruits and olive oil including a moderate amount of seafood, fish, dairy and red wine, and less amount of processed meat and red meat. This Mediterranean diet constitutes low in saturated fat, animal protein and high in monounsaturated fatty acids (MUFAs), antioxidants and fibre, and proper balance between omega-6 and omega-3 fatty acid [71,72]. Mediterranean diets [73,74] or Mediterranean-type diets [rich in MUFAs [75] with a low AGEs content have been shown to reduce circulating AGEs, decrease oxidative stress and have anti-inflammatory effects. Gutierrez-Mariscal et al. 2020 [73] observed that a 5-year intake of a Mediterranean diet decreased blood AGEs levels (especially, MG levels), which was associated with improved beta cell functionality index assessed by the disposition index (DI) in newly diagnosed T2DM subjects with coronary heart disease (CHD), compared with intake of a low-fat diet. This indicated that a Mediterranean diet could play a T2DM remission role through enhanced beta cell function attributable to reduced circulating AGEs levels. Lopez-Moreno et al. 2016 [74] conducted a RCT of 20 subjects aged over 65 years in a crossover design. Subjects underwent three 4-week isocaloric diet interventions. A meal test was performed at the end of each 4-week intervention. Three isocaloric diets included the Mediterranean diet, saturated fat diet and omega-3 diet consisting of low in fat and high in carbohydrate. The Mediterranean diet was comprised of 38% fat [24% MUFA from virgin olive oil; 10% saturated fatty acids (SFA); 4% polyunsaturated fatty acids (PUFA)]. The saturated fat diet was comprised of 38% fat (12% MUFA; 22% SFA; 4% PUFA). The omega-3 diet was comprised of 28% fat (10% SFA; 12% MUFA; 8% PUFA with 2% alpha-linolenic acid). The Mediterranean diet reduced levels of sMG, sCML and RAGE mRNA (oxidative stress and inflammation inducer) and increased levels of AGE receptor 1 (AGER1: AGEs clearance with AGEs endocytic uptake and degradation), and GloxI (antioxidant defence marker) in the fasting and postprandial states, compared with the SFA diet or the omega-3 diet [74]. Lopez-Moreno et al. 2017 [75] conducted a RCT in 75 subjects with metabolic syndrome. Subjects with metabolic syndrome were randomly allocated to one of four isoenergetic diets for 12 weeks. Four diets are as follows: (a) a high-fat, saturated fatty acid-rich diet [38% fat (16% SFA)]; (b) a high MUFA diet [38% fat (20% MUFA from olive oil)]; (c) low-fat, high-complex carbohydrate-rich diets with placebo capsule [1.2 g/day of control high-oleic sunflower seed oil capsules (placebo)]; (d) low-fat, high-complex carbohydrate-rich diets with a 1.24 g/day of n-3 capsule (long-chain (n-3) polyunsaturated fatty acid [ratio of 1.4 eicosapentaenoic acid (EPA):1 docosahexaenoic acid (DHA)]. Meal tests representing each intervention diet were performed at the end of each 12-week diet. Reductions in blood levels of AGEs and RAGE mRNA and increases in blood mRNA levels of AGER1and GloxI were observed after a 12-week high MUFA diet, compared with other diets. These findings indicated a low-AGEs diet rich MUFA could alter AGEs metabolism by increasing AGER1 which can decrease AGEs levels in the intracellular and extracellular spaces as an antioxidant [75].

4. Dietary AGEs Influence on Glucose Homeostasis

AGEs can cause pancreatic β cell failure by suppressing insulin production and secretion, causing abnormal glycaemic control and eventually inducing the onset of T2DM [2,76].
Uribarri et al. 2015 [77] conducted a cross-sectional study in a total of 269 obese subjects aged over 50 years with or without metabolic syndrome. Obese subjects with metabolic syndrome showed increased serum AGEs levels compared with obese subjects without metabolic syndrome. Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), leptin, tumour necrosis factor α (TNF-α) and RAGE were associated with AGEs levels, while SIRT1, AGER1, glyoxalase-I and adiponectin were inversely associated with AGEs levels. The AGEs intake was higher in obese subjects with metabolic syndrome than in obese subjects without metabolic syndrome. These elevated AGEs levels were positively associated with dAGEs.
Acute studies [78,79,80] evaluated the effects of AGE meals on glycaemic control. Poulsen et al. 2014 [78] conducted an acute study of 19 healthy overweight subjects in randomised crossover design. They examined postprandial changes of satiety, inflammation and endothelial activation comparing a high-AGEs meal with a low-AGEs meal. The higher glycaemic response was observed after a high-AGEs meal than after a low-AGEs meal. A high-AGEs meal also elevated levels of urinary isoprostanes. No differences in visual analogue scale (VAS) scores for hunger, satiety, fullness and prospective food intake were observed between meals. No differences in blood levels of total ghrelin, glucagonlike peptide-1 (GLP-1) and peptide YY (PYY) were observed between meals. Blood levels of high-sensitivity C-reactive protein (hs-CRP), TNF-α, interleukin 6 (IL-6) and vascular cell adhesion molecule-1 (VCAM-1) did not differ between meals [78]. Schiekofer et al. 2006 [79] investigated acute effects of a casein meal with low- or high- AGEs contents on transcription factor nuclear factor-kappaB (NF-κB) in peripheral blood mononuclear cells (PBMC) of healthy subjects, which was independent of the AGEs content of the casein meal [79]. Negrean et al. 2007 [80] found higher postprandial levels of endothelial activation markers after a high-AGEs meal compared with a low-AGEs meal in subjects with T2DM. The higher flow-mediated dilation (FMD) for microvascular function was observed after a high-AGEs meal than after a low-AGEs meal even though both meals lowered FMD. Markers of endothelial dysfunction [plasma E-selectin, intracellular adhesion molecule 1 (ICAM-1) and VCAM-1] increased 2 h or/and 4 h after a high-AGEs meal compared with 2 h or/and 4 h after a low-AGEs meal. There were no significant differences in glucose, insulin, lipids [triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C)], inflammatory markers (hs-CRP, IL-6 and TNF-α) [80].
Meta-analyses of RCTs [81,82] examined the dAGEs on glycaemic control. A recent meta-analysis of 13 RCTs [10,11,12,13,15,16,17,19,83,84,85,86,87] conducted by Sohouli et al. 2020 [81] showed the favourable effect of a low-AGEs diet on metabolic syndrome risk factors such as insulin resistance, fasting insulin, TC and LDL-C. A low-AGEs diet showed significantly decreased concentrations of HOMA-IR [weighted mean difference (WMD) −1.204; 95% confidence interval (CI) −2.057, −0.358; p = 0.006] and fasting insulin (WMD −5.472 μU/mL; 95% CI −9.718, −1.234 μU/mL; p = 0.011), compared with a high-AGEs diet in quantitative analysis [81]. In lipid profile changes, a low-AGEs diet showed significantly decreased concentrations of TC (WMD −5.486 mg/dL; 95% CI: −10.222, −0.747 mg/dL; p = 0.023) and LDL (WMD −6.263 mg/dL; 95% CI −11.659, −0.866 mg/dL; p = 0.023), compared with a high-AGEs diet [81]. In a meta-analysis of 17 RCTs (22 publications [10,11,12,13,14,15,16,17,19,20,69,78,80,83,86,88,89,90,91,92,93,94]), Bay et al. 2017 [82] examined cardiometabolic risk by dAGEs in subjects with or without T2DM. Decreases in insulin resistance, TC and LDL-C were observed after a low-AGEs diet. Weight, fasting glucose, 2-h glucose and insulin, haemoglobin A1C, HDL-C or blood pressure (BP) were not changed after a low-AGEs diet. Especially, in subjects with T2DM, a low-AGEs diet led to reduced fasting insulin, TNF-α, VCAM-1, 8-isoprostane, leptin, circulating AGEs and receptor for AGEs. Adiponectin and sirtuin-1 were increased [82].
Several low-AGEs diet interventions of obese subjects showed improved insulin sensitivity [13,89,95]. Mark et al. 2014 [13] showed that a low-AGEs diet improved HOMA-IR after 4 weeks in 37 overweight women [13]. In a double-blind, randomised crossover trial of 20 healthy overweight subjects conducted by de Courten et al. 2016 [89], a low-AGEs diet improved insulin sensitivity as assessed by a hyper-insulinemic euglycaemic clamp and an intravenous glucose tolerance test, compared with a high-AGEs diet, even though no difference in body weight or insulin secretion was observed between diets. Two diets were isoenergetic- and macronutrient-matched diets with differences in AGEs contents [89]. Goudarzi et al. 2020 [95] examined the effects of a restricted AGEs diet on glucose metabolism, lipid profiles, oxidative stress (malondialdehyde) and inflammation (TNF-α and hs-CRP) in a RCT. Forty overweight subjects with metabolic syndrome completed either a calorie restriction and regular AGEs (R-AGEs) diet or a calorie restriction and a low-AGEs (L-AGEs) diet for 8 weeks. The L-AGEs diet significantly decreased CML, fasting glucose, fasting insulin, HOMA-IR, TNF-α and MDA compared with the R-AGEs diet. No significant difference in hs-CRP was seen between the two diets [95]. However, Linkens et al. 2022 [96] found no effect of a 4-week AGEs diet on glucose control and vascular function in 73 abdominally obese subjects (mean age 52 years; mean body mass index (BMI) 30.6 ± 4.0 kg/m2) with a double-blind parallel design of the intervention. A diet low in AGEs did not alter glucose, HbA1c, insulin, C-peptide, insulin sensitivity, secretion and clearance, micro- and macrovascular function (measured by FMD), inflammation, fatty liver index, estimated glomerular filtration rate (eGFR), lipid profiles (HDL-C, LDL-C and TG), oxidative stress and DNA glycation, compared with a diet high in AGEs. The gold standard ultra-performance LC–tandem mass spectrometry (UPLC-MS/MS) was used to assess AGEs and dicarbonyls in food, plasma and urine. Insulin sensitivity, secretion and clearance were assessed by a combined hyperinsulinemic-euglycaemic and hyperglycaemic clamp. No differences in urinary and plasma CML, CEL and Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1) were observed between a low-AGEs diet group and a high-AGEs diet group [96].
A low-AGEs diet intervention of healthy subjects showed improved insulin sensitivity [97]. In an intervention of healthy subjects conducted by Birlouez-Aragon et al. 2010 [97], a high-heat-treated diet for 1 month significantly decreased insulin sensitivity, plasma levels of omega-3 fatty acids, vitamin C and vitamin E compared with a steamed diet. Moreover, reductions in TC, TG and HDL-C were observed after a high-heat-treated diet compared with a steamed diet [97]. A low-AGEs diet intervention of subjects with T2DM showed improved insulin sensitivity [17]. An AGEs-restricted diet for 4 months reduced HOMA-IR, insulin, leptin, TNF-α, NF-κB, p65 acetylation, serum AGEs and 8-isoprostanes in subjects with T2DM, compared with a standard diet [17].
In summary, given the recent intervention findings, a low-AGEs diet in healthy subjects, obese subjects and diabetic subjects appears to improve glucose homeostasis.

5. Dietary AGEs Influence on Body Weight

In epidemiological studies [Physical Activity, Nutrition, Alcohol, Cessation of smoking, eating out of home in relation to Anthropometry (PANACEA) study and a sub-cohort of the European Prospective Investigation into Cancer and Nutrition (EPIC) study] with 255,170 European subjects aged 25–70 years, the association between higher dietary AGEs intake and marginal greater weight gain was observed during the 5-year follow-up period. Especially, a high-CEL diet showed a 10% greater body weight increase compared with a low-CEL diet [98].
In a meta-analysis of 13 RCTs [10,11,13,16,17,19,83,84,85,86,87,89,99] conducted by the same research team of Sohouli et al. 2020 [100], a low-AGEs diet showed reductions in BMI (WMD −0.3 kg/m2; 95% CI −0.52, −0.09; p = 0.005; I2 = 55.8%), weight (WMD −0.83 kg; 95% CI: −1.55; −0.10; p = 0.026; I2 = 67.0%) and leptin (WMD −19.85 ng/mL; 95% CI −29.88, −9.82; p < 0.001; I2 = 81.8%), compared with a high-AGEs diet. Moreover, a low-AGEs diet showed significantly increased concentrations of adiponectin (WMD 5.50 µg/mL; 95% CI 1.33, 9.67; p = 0.010; I2 = 90.6%), compared with a high-AGEs diet.
Dicarbonyl stress is attributable from the abnormal accumulation of dicarbonyl compounds, which consequently promotes the development of cardiometabolic disorders including obesity by elevating protein and DNA modification [101,102,103,104]. Maessen et al. 2016 [105] examined whether a very low-calorie diet (VLCD) or Roux-en-Y gastric bypass (RYGB) decreased α-dicarbonyl stress in lean (n = 12) or obese women without (n = 27) and with T2DM (n = 27). Four groups [obese glucose-tolerant (NGT)-gastric banding (GB), NGT-RYGB, T2DM-RYGB and T2DM-VLCD] underwent mixed meal tests at baseline and 3 weeks at the end of intervention. Fasting and postprandial plasma α-dicarbonyl levels were higher in obese females with T2DM than in obese females without T2DM. Obese women with T2DM had significantly decreased postprandial α-dicarbonyl and fasting plasma α-dicarbonyls concentration after a 3-week VLCD [105]. Van den 2021 [106] conducted a weight loss intervention. Postprandial dicarbonyls [iAUC of MGO, GO and 3-deoxyglucosone (3-DG)] were decreased after an 8-week weight loss diet as compared with a habitual diet in abdominally obese men.
In summary, given the recent intervention findings, a low-AGEs diet appears to benefit body weight management.

6. Dietary AGEs Influence on Oxidative Stress and Inflammation

AGEs with oxidant properties elevate oxidative stress and inflammatory responses by stimulating RAGE in the AGE formation process, leading to the risk of cardiometabolic disorders [107,108,109,110,111].
Overweight and obese subjects (BMI 26–39 kg/m2) were randomly allocated to either a low-AGEs diet or a high-AGEs diet for 2 weeks in the intervention with a randomised crossover design. Two diets (9 MJ/day a diet) were isocaloric and macronutrient matched diets. A low-AGEs (3302 kU CML) diet improved inflammatory profiles [monocyte chemoattractant protein-1 (MCP-1) and macrophage migration inhibitory factor (MIF)] and renal function, compared with a high-AGEs (14,090 kU CML) diet [11]. Vlassara et al. 2016 [83] conducted a RCT of 100 obese subjects with the metabolic syndrome comparing a low-AGEs diet with a regular-AGEs diet for 1 year. After a low-AGEs diet compared with a regular-AGEs diet, HOMA-IR, fasting plasma insulin and insulin levels at 120 min of an oral glucose tolerance test (OGTT) were decreased. Body weight was slightly reduced. AGEs, oxidative stress (8-isoprostanes) and inflammation (TNF-α and VCAM-1) and RAGE mRNA decreased. The protective factors including mRNA levels of sirtuin 1, glyoxalase I and AGER1 increased. No differences in fasting blood glucose, HbA1c or blood glucose at 60 and 120 min of the OGTT were observed between two diets. Di Pino et al. 2016 [84] observed significantly decreased lipid levels of TC, apolipoprotein B (Apo B) and LDL-C after a 24-week low-AGEs diet compared with a standard-AGEs diet in 62 pre-diabetic subjects. Reductions in hs-CRP and intima-media thickness were observed after a low-AGEs diet, compared with the baseline of a low-AGEs diet. No change in arterial stiffness was observed in a within-diet group and a between-diet group. Semba et al. 2014 [15] performed a randomised, parallel-arm, controlled trial of 24 healthy subjects aged 50–69 years for 6 weeks. No differences in peripheral arterial tonometry, serum and urine CML, inflammatory mediators (IL-6, hs-CRP, VCAM1 and TNF-α receptors I and II), sRAGE and endogenous secretory receptors for AGEs were observed.
CML, one of primary dAGEs, stable and chemically inert AGEs, are unable to directly interact with tissue protein [112,113]. CML from a diet can be deposited in all tissues but adipose tissues. Kidneys, ileum, colon and lungs are organs with a higher deposition of CML, whereas the heart, liver and muscle are organs with a lower deposition of CML [114].
In vitro and in vivo studies showed the association-elevated inflammation and RAGE and CML-RAGE axis in obesity, consequently elevating insulin resistance [115,116]. In the Cohort on Diabetes and Atherosclerosis Maastricht Study and Hoorn Study performed by Gaens et al. 2015 [117], subjects with central obesity showed lower plasma levels of RAGE-mediated CML compared with lean subjects. The inverse association between RAGE-mediated CML and low-grade inflammation (LGI) scores was observed. These findings indicated in part “AGEs trapping (or AGEs build up)” in adipose tissue resulting in decreased levels of circulating plasma AGEs [117]. Ruix et al. 2021 [118] demonstrated that AGE/RAGE/diaphanous 1 (DIAPH1) axis in the immunometabolic pathophysiology of obesity was associated with insulin resistance partly throughout the expression and activity of this axis in abdominal subcutaneous (SAT) adipose tissue in obese subjects without T2DM [118]. CML does not always bind to RAGE for inflammatory response leading to cardiometabolic disorders, which indicates that dAGEs can promote inflammatory signalling cascade via a RAGE-independent pathway [112].
In summary, dietary AGEs are involved in increased oxidative stress and inflammation responses, which is associated with vascular dysfunction.

7. Dietary AGEs Influence on Vascular Function

AGEs deposited in tissues and urine have been observed in cardiometabolic disorders and aging [119,120,121]. Two mechanisms of how AGEs contribute to tissue damage of cardiac or vascular dysfunction have been postulated: (a) AGEs can promote cardiac or vascular dysfunction by inducing inflammation and oxidative stress through receptors (Receptor-dependent way) [119]; (b) AGEs can elevate vascular and myocardial stiffness by cross-linking of elastin and collagen, consequently leading to vascular stiffness and cardiac fibrosis (Receptor-independent way) [122].
The RAGE, which is the most known human AGE receptor, is a multi-ligand type I cell surface receptor of the immunoglobulin (Ig) superfamily composed of three extracellular immunoglobulin domains, C1, C2 and V. Other AGEs receptors include scavenger receptors (e.g., stabilin-1, stabilin-2, SR-AI, SR-BI, OxLDL receptor 1, FEEL-1 and FEEL-2) and AGE-Rs (e.g., AGE-R1, AGE -R2 and AGE-R3) [123,124]. The expression of RAGE was found on different cells (endothelial cell, smooth muscle cells, chondrocytes, dendritic cell, fibroblasts, monocytes, microphages, T-lymphocytes, neuronal cell, glia cells and keratinocytes) [125,126,127,128,129,130]. Binding of AGEs to RAGE induces different intracellular signalling pathways, leading to elevated oxidative stress by increased intracellular ROS production with activation of NADPH oxidase, and activation of p21(ras)-dependent mitogen-activated protein kinase (MAPK) pathways triggering upregulation of NF-κB, its target genes and inflammation. The elevated levels of circulating cytokines (IL-1, IL-6 and TNF-α) contribute to a persistent inflammatory state [40,107,131,132,133].
Obesity was associated with decreased plasma levels of sRAGE leading to atherosclerosis risk [117,134,135,136,137,138,139,140,141,142]. The sAGE comprised of the ligand-binding site acts as a decoy receptor that inhibits the attachment of AGEs to RAGE [143]. Inhibition of AGE-RAGE binding can attenuate the development of atherosclerosis [143,144,145,146,147].
Rodríguez-Morteraa et al. 2019 [148] showed the effect of sRAGE on vascular function in a cross-sectional study of adolescents aged 15–19 years (33 obesity and 33 normal weight). sRAGE was associated with low-FMD and arterial stiffness index (Iβ). CML and AGEs were associated with atherogenic index (AI) [148].
AGEs accumulation sourced from food is associated with chronic inflammation, leading to risk of cardiometabolic disorders (especially, diabetic complication: risk of atherosclerosis in subjects with T2DM over about 10 years) [149]. The reactive dicarbonyl compounds, including glyoxal, MG and 3-deoxyglucosone, which are precursors of the primary quantitative AGEs, can directly influence extracellular matrix (ECM) modification [43,150]. The dAGEs can modify ECM. AGEs can be produced on proteins (collagen, laminin, elastin and vitronectin), and lipids in the ECM, which cause modification of ECM and stimulate increased stiffness [51,151,152,153]. The AGEs build-up on proteins in the ECM produces cross-links that can entrap other molecules [154]. AGE cross-linking on type I collagen and elastin can expand the area of ECM, leading to elevated vascular stiffness [51,151]. Moreover, AGEs build-up on proteins in the ECM can promote lipid-linked AGEs formation in subjects with or without T2DM [152]. Glycation of LDL-C in subjects with T2DM can decrease endothelial nitric oxide (NO) production and LDL-C clearance [153].
In the population-based cohort Maastricht Study of 2792 participants aged 60 years (26% T2DM) by Cordova et al. 2020 [155], the association between higher habitual intake of dicarbonyls methylglyoxal (MGO) and lower grade inflammation was observed after full adjustment, which was inversely associated with hs-CRP and TNF-α. Moreover, the association between higher dietary MGO intake and impaired retinal venular dilation was observed after full adjustment [155]. Linkens et al. 2022 [156] investigated the association between dietary AGEs intake and generalised microvascular function in a cross-sectional study (Maastricht Study) of 3144 participants aged 60 years. They determined dAGEs intake by combining the consumption of food items within the FFQ with their UPLC-MS/MS dietary AGEs database [30,156]. Microvascular function was evaluated with the retina [flicker light-induced arteriolar dilation, flicker light-induced venular dilation, central retinal arteriolar equivalent (CRAE) and central retinal venular equivalent (CRVE), the plasma endothelial dysfunction biomarkers [soluble VCAM-1, soluble ICAM-1, soluble E-selectin and von Willebrand factor(vWf)], skin (heat-induced skin hyperaemic response) and urine (24-h albuminuria). They did not find overall association between intakes of CML, CEL and MG-H1 and generalised microvascular function even though a higher CEL intake was associated with flicker light-induced venular dilation (β percentage change over baseline: 0.14; 95% CI 0.02, 0.26) and a lower plasma biomarker z score (β −0.04 SD; 95% CI −0.08, −0.00 SD) [156].
Linkens et al. 2021 [157] found intake of the dAGEs (CML, CEL and MG-H1) was not significantly associated with arterial stiffness as assessed by carotid–femoral pulse wave velocity (cfPWV), carotid distensibility coefficient (DC), and carotid Young’s elastic modulus (YEM) in the Maastricht Study [157].
In summary, the development of cardiometabolic disorders could be lowered by attenuating AGEs levels and RAGE expression and enhancing levels of sRAGE. The AGEs load can be reduced with high-AGEs diet restriction, low-AGEs cooking methods (low-temperature, boiling and steaming) and stopping smoking.

8. Dietary AGEs Influence on Gut Microbiome

Most of the dAGEs ingested orally are absorbed in the small intestine via the gastric tract and transported to the vascular system, while their remains are exported to the urine. The extent of dAGEs absorption depends on their chemical structures, molecular weights and hydrophobicity (e.g., pyrraline and argpyrimidine). Highly hydrophobic AGEs easily pass through the basolateral membrane compared with hydrophilic AGEs (e.g., CML, CEL and MG-H1) [34]. Non-absorbed AGEs are transported into the lower gut, where some are digested by gut microbiota, and the remains are exported via the faeces [46].
Western-style diets high in red meat, animal fat and/or simple sugar and low in fibre appear to negatively modify gut microbiota composition [158,159,160]. Several RCTs showed the effect of AGEs on gut bacterial microbiota [90,99,161]. Twenty male adolescents aged mean 12 years underwent a brown diet high in MRP content and a white diet low in MRP content in a two-week crossover design. A brown-diet group showed a reduction of enterobacteria correlated with increased intakes of hydroxymethylfurfural (HMF) and CML. Lactobacilli numbers were negatively associated with dietary advanced MRP such as hydroxymethylfurfural and CML. Bifidobacteria counts were negatively associated with Amadori compounds intake [90].
In a randomised, open label trial of 20 peritoneal dialysis patients, an AGEs-low diet for 1 month decreased Prevotella copri (considered a good bacteria [162]) and Bifidobacterium animalis compared with an AGEs-high diet. Moreover, an AGEs-low diet for 1 month increased Alistipes indistinctus (pathogenic causing colitis and site-specific tumours [163]), Clostridium hatewayi, Clostridium citroniae and Ruminococcus gauvreauii compared with an AGEs-high diet [99]. Linkens et al. 2022 [161] conducted a RCT with a double-blind parallel design to examine effects of 4-week isocaloric and macronutrient-matched AGEs diet on the gut microbial composition in 70 abdominally obese subjects (mean age 52 years; mean BMI 30.6 ± 4.0 kg/m2). They found the limited effects of dAGEs on microbiota composition. No difference in the Shannon index for microbial richness and diversity was observed comparing a low-AGEs diet with a high-AGEs diet. No difference in the overall microbial composition was observed comparing a low-AGEs diet with a high-AGEs diet. However, a low-AGEs diet decreased only Anaerostipes spp. abundance (one of 15 most abundant taxa in healthy individuals) compared with a high-AGEs diet. Moreover, the association between 3-deoxyglucosone (3-DG) intake and an abundance of several genera was observed [161].
In summary, limited studies showed dietary AGEs could play a potential role in disrupting gut microbiome and immune system. More interventions are required to clarify underlying mechanisms of action addressing the role of dAGEs in gut dysbiosis.

9. Skin AGEs as a Predictor of Cardiometabolic Disorders

The SAF is a non-invasive recent method to measure AGEs (Figure 1). The association between SAF and the quantity of certain AGEs in skin biopsies has been shown [65,66,67]. The levels of AGEs may markedly differ between tissue and plasma measurements. AGEs can be strongly deposited within the tissues (especially, dermal collagen), which can be measured through SAF considering certain AGEs have fluorescent properties. The SAF assessed by a Scout DS device showed clinically higher advantages than fasting glucose and HbA1c in subjects with undiagnosed abnormal glucose levels [164,165].
Meta-analysis conducted by Cavero-Redondo et al. 2018 [50] indicated that chronic accumulation SAF produced by enzymatic glycation in skin could be biomarkers of all-cause mortality and CVD mortality in subjects with diabetes and cardiovascular and/or renal diseases. This study showed the association between higher SAF and higher CVD mortality [hazard ratio (HR) 2.06; 95% CI 1.58, 2.67; I2 = 34.7%; p = 0.163] or higher all-cause mortality (HR 1.91; 95% CI 1.42, 2.56; I2 = 60.8%; p = 0.0.18) [50].
In a prospective cohort study with a 5-year follow up study period, the associations between higher SAF and elevated risk for all-cause mortality and elevated risk for fatal or nonfatal major adverse CVD events (MACE) were observed in subjects with peripheral artery disease [166].
Chen et al. 2022 [7] conducted a meta-analysis of 14 [1,73,166,167,168,169,170,171,172,173,174,175,176,177] prospective studies to examine if skin AGEs are associated with MACE. Skin AGEs were assessed by SAF. The significant associations between skin AGEs and elevated fatal CVD (HR 1.88; 95% CI 1.30, 2.70) and nonfatal CVD (HR 1.40; 95% CI 1.12, 1.74) were observed. In subjects with diabetes (HR 1.88; 95% CI 1.31, 2.69) and kidney disease (HR 1.50; 95% CI 1.16, 1.94), skin AGEs was associated with elevated MACE. This meta-analysis found the association between increased AGEs assessed by SAF and increased CVD events indicating skin AGEs could become a predictor of CVD events [7].
Several studies showed no association between skin AGEs and CVD events or death [73,168,169,170,171], while some studies showed the association skin AGEs and CVD events or death [177,178,179,180,181]. Waateringe et al. 2019 [177] conducted the Dutch Lifelines Cohort Prospective Study of 72,880 general non-diabetic population with a 4-year follow-up. They found the association between SAF and incident of T2DM, CVD and death independent of risk factors age, sex, waist circumference, metabolic syndrome, smoking and blood glucose levels. Especially, SAF was observed as the strong predictor for incident T2DM and/or CVD in subjects aged over 35 years.
Studies [182,183] showed higher SAF in subjects with metabolic syndrome compared to subjects without metabolic syndrome. SAF levels were independent of the presence of metabolic syndrome, waist circumference, impaired fasting glucose levels and BP.

10. Conclusions

A diet high in AGEs induces cardiometabolic disorders through increasing blood glucose and blood lipid concentrations, oxidative stress, inflammatory response, BP and body weight, compared with a diet low in AGEs. In addition, a diet high in AGEs may negatively change the gut microbiota and promote prevalence of cardiometabolic disorders compared with a diet low in AGEs. Further human studies using SAF assessments are needed to clarify the association between dAGEs in tissues and obesity, T2DM, CVD events, CVD mortality and total mortality.

Funding

This research was funded by the National Research Foundation of Korea (NRF), grant number NRF-2022R1F1A1063108. The NRF had no role in the study design, data analysis or writing of this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yozgatli, K.; Lefrandt, J.D.; Noordzij, M.J.; Oomen, P.H.N.; Brouwer, T.; Jager, J.; Castro Cabezas, M.; Smit, A.J. Accumulation of advanced glycation end products is associated with macrovascular events and glycaemic control with microvascular complications in Type 2 diabetes mellitus. Diabet. Med. A J. Br. Diabet. Assoc. 2018; Online ahead of print. [Google Scholar] [CrossRef]
  2. Tan, K.C.; Shiu, S.W.; Wong, Y.; Tam, X. Serum advanced glycation end products (AGEs) are associated with insulin resistance. Diabetes/Metab. Res. Rev. 2011, 27, 488–492. [Google Scholar] [CrossRef] [PubMed]
  3. Tahara, N.; Yamagishi, S.; Matsui, T.; Takeuchi, M.; Nitta, Y.; Kodama, N.; Mizoguchi, M.; Imaizumi, T. Serum levels of advanced glycation end products (AGEs) are independent correlates of insulin resistance in nondiabetic subjects. Cardiovasc. Ther. 2012, 30, 42–48. [Google Scholar] [CrossRef]
  4. Sebeková, K.; Krivošíková, Z.; Gajdoš, M. Total plasma Nε-(carboxymethyl)lysine and sRAGE levels are inversely associated with a number of metabolic syndrome risk factors in non-diabetic young-to-middle-aged medication-free subjects. Clin. Chem. Lab. Med. 2014, 52, 139–149. [Google Scholar] [CrossRef]
  5. Accacha, S.; Rosenfeld, W.; Jacobson, A.; Michel, L.; Schnurr, F.J.; Shelov, S.; Ten, S.; Boucher-Berry, C.; Carey, D.E.; Speiser, P.W.; et al. Plasma advanced glycation end products (AGEs), receptors for AGEs and their correlation with inflammatory markers in middle school-age children. Horm. Res. Paediatr. 2013, 80, 318–327. [Google Scholar] [CrossRef] [PubMed]
  6. Sebeková, K.; Somoza, V.; Jarcusková, M.; Heidland, A.; Podracká, L. Plasma advanced glycation end products are decreased in obese children compared with lean controls. Int. J. Pediatr. Obes. IJPO Off. J. Int. Assoc. Study Obes. 2009, 4, 112–118. [Google Scholar] [CrossRef] [PubMed]
  7. Chen, Q.; Huang, Q.; Liu, W.; Zhou, X. Advanced glycation end products via skin autofluorescence as a new biomarker for major adverse cardiovascular events: A meta-analysis of prospective studies. Nutr. Metab. Cardiovasc. Dis. NMCD 2022, 32, 1083–1092. [Google Scholar] [CrossRef]
  8. Clarke, R.E.; Dordevic, A.L.; Tan, S.M.; Ryan, L.; Coughlan, M.T. Dietary Advanced Glycation End Products and Risk Factors for Chronic Disease: A Systematic Review of Randomised Controlled Trials. Nutrients 2016, 8, 125. [Google Scholar] [CrossRef]
  9. Luévano-Contreras, C.; Gómez-Ojeda, A.; Macías-Cervantes, M.H.; Garay-Sevilla, M.E. Dietary Advanced Glycation End Products and Cardiometabolic Risk. Curr. Diabetes Rep. 2017, 17, 63. [Google Scholar] [CrossRef]
  10. Cai, W.; He, J.C.; Zhu, L.; Peppa, M.; Lu, C.; Uribarri, J.; Vlassara, H. High levels of dietary advanced glycation end products transform low-density lipoprotein into a potent redox-sensitive mitogen-activated protein kinase stimulant in diabetic patients. Circulation 2004, 110, 285–291. [Google Scholar] [CrossRef]
  11. Harcourt, B.E.; Sourris, K.C.; Coughlan, M.T.; Walker, K.Z.; Dougherty, S.L.; Andrikopoulos, S.; Morley, A.L.; Thallas-Bonke, V.; Chand, V.; Penfold, S.A.; et al. Targeted reduction of advanced glycation improves renal function in obesity. Kidney Int. 2011, 80, 190–198. [Google Scholar] [CrossRef]
  12. 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]
  13. 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 2014, 37, 88–95. [Google Scholar] [CrossRef]
  14. Peppa, M.; Uribarri, J.; Cai, W.; Lu, M.; Vlassara, H. Glycoxidation and inflammation in renal failure patients. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2004, 43, 690–695. [Google Scholar] [CrossRef] [PubMed]
  15. Semba, R.D.; Gebauer, S.K.; Baer, D.J.; Sun, K.; Turner, R.; Silber, H.A.; Talegawkar, S.; Ferrucci, L.; Novotny, J.A. Dietary intake of advanced glycation end products did not affect endothelial function and inflammation in healthy adults in a randomized controlled trial. J. Nutr. 2014, 144, 1037–1042. [Google Scholar] [CrossRef] [PubMed]
  16. Uribarri, J.; Cai, W.; Pyzik, R.; Goodman, S.; Chen, X.; Zhu, L.; Ramdas, M.; Striker, G.E.; Vlassara, H. Suppression of native defense mechanisms, SIRT1 and PPARγ, by dietary glycoxidants precedes disease in adult humans; relevance to lifestyle-engendered chronic diseases. Amino Acids 2014, 46, 301–309. [Google Scholar] [CrossRef] [PubMed]
  17. 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: Potential role of AGER1 and SIRT1. Diabetes Care 2011, 34, 1610–1616. [Google Scholar] [CrossRef]
  18. Uribarri, J.; Peppa, M.; Cai, W.; Goldberg, T.; Lu, M.; Baliga, S.; Vassalotti, J.A.; Vlassara, H. Dietary glycotoxins correlate with circulating advanced glycation end product levels in renal failure patients. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2003, 42, 532–538. [Google Scholar] [CrossRef]
  19. Vlassara, H.; Cai, W.; Crandall, J.; Goldberg, T.; Oberstein, R.; Dardaine, V.; Peppa, M.; Rayfield, E.J. Inflammatory mediators are induced by dietary glycotoxins, a major risk factor for diabetic angiopathy. Proc. Natl. Acad. Sci. USA 2002, 99, 15596–15601. [Google Scholar] [CrossRef]
  20. Vlassara, H.; Cai, W.; Goodman, S.; Pyzik, R.; Yong, A.; Chen, X.; Zhu, L.; Neade, T.; Beeri, M.; Silverman, J.M.; et al. Protection against loss of innate defenses in adulthood by low advanced glycation end products (AGE) intake: Role of the antiinflammatory AGE receptor-1. J. Clin. Endocrinol. Metab. 2009, 94, 4483–4491. [Google Scholar] [CrossRef]
  21. John, W.G.; Lamb, E.J. The Maillard or browning reaction in diabetes. Eye 1993, 7 Pt 2, 230–237. [Google Scholar] [CrossRef]
  22. Singh, R.; Barden, A.; Mori, T.; Beilin, L. Advanced glycation end-products: A review. Diabetologia 2001, 44, 129–146. [Google Scholar] [CrossRef]
  23. Garay-Sevilla, M.E.; Rojas, A.; Portero-Otin, M.; Uribarri, J. Dietary AGEs as Exogenous Boosters of Inflammation. Nutrients 2021, 13, 2802. [Google Scholar] [CrossRef]
  24. 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. [Google Scholar] [CrossRef] [PubMed]
  25. 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]
  26. Nicholl, I.D.; Bucala, R. Advanced glycation endproducts and cigarette smoking. Cell. Mol. Biol. 1998, 44, 1025–1033. [Google Scholar] [PubMed]
  27. Inan-Eroglu, E.; Ayaz, A.; Buyuktuncer, Z. Formation of advanced glycation endproducts in foods during cooking process and underlying mechanisms: A comprehensive review of experimental studies. Nutr. Res. Rev. 2020, 33, 77–89. [Google Scholar] [CrossRef] [PubMed]
  28. O’Brien, J.; Morrissey, P.A. Nutritional and toxicological aspects of the Maillard browning reaction in foods. Crit. Rev. Food Sci. Nutr. 1989, 28, 211–248. [Google Scholar] [CrossRef]
  29. 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]
  30. Scheijen, J.; Clevers, E.; Engelen, L.; Dagnelie, P.C.; Brouns, F.; Stehouwer, C.D.A.; 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]
  31. Thornalley, P.J. Pharmacology of methylglyoxal: Formation, modification of proteins and nucleic acids, and enzymatic detoxification—A role in pathogenesis and antiproliferative chemotherapy. Gen. Pharmacol. 1996, 27, 565–573. [Google Scholar] [CrossRef]
  32. Henning, C.; Glomb, M.A. Pathways of the Maillard reaction under physiological conditions. Glycoconj. J. 2016, 33, 499–512. [Google Scholar] [CrossRef] [PubMed]
  33. Hamada, Y.; Araki, N.; Koh, N.; Nakamura, J.; Horiuchi, S.; Hotta, N. Rapid formation of advanced glycation end products by intermediate metabolites of glycolytic pathway and polyol pathway. Biochem. Biophys. Res. Commun. 1996, 228, 539–543. [Google Scholar] [CrossRef] [PubMed]
  34. Twarda-Clapa, A.; Olczak, A.; Białkowska, A.M.; Koziołkiewicz, M. Advanced Glycation End-Products (AGEs): Formation, Chemistry, Classification, Receptors, and Diseases Related to AGEs. Cells 2022, 11, 1312. [Google Scholar] [CrossRef] [PubMed]
  35. Khalid, M.; Petroianu, G.; Adem, A. Advanced Glycation End Products and Diabetes Mellitus: Mechanisms and Perspectives. Biomolecules 2022, 12, 542. [Google Scholar] [CrossRef]
  36. Chung, S.S.; Ho, E.C.; Lam, K.S.; Chung, S.K. Contribution of polyol pathway to diabetes-induced oxidative stress. J. Am. Soc. Nephrol. JASN 2003, 14, S233–S236. [Google Scholar] [CrossRef]
  37. Fiorentino, T.V.; Prioletta, A.; Zuo, P.; Folli, F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr. Pharm. Des. 2013, 19, 5695–5703. [Google Scholar] [CrossRef]
  38. Dias, I.H.; Griffiths, H.R. Oxidative stress in diabetes—Circulating advanced glycation end products, lipid oxidation and vascular disease. Ann. Clin. Biochem. 2014, 51, 125–127. [Google Scholar] [CrossRef]
  39. Moldogazieva, N.T.; Mokhosoev, I.M.; Mel’nikova, T.I.; Porozov, Y.B.; Terentiev, A.A. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxidative Med. Cell. Longev. 2019, 2019, 3085756. [Google Scholar] [CrossRef]
  40. Ott, C.; Jacobs, K.; Haucke, E.; Navarrete Santos, A.; Grune, T.; Simm, A. Role of advanced glycation end products in cellular signaling. Redox Biol. 2014, 2, 411–429. [Google Scholar] [CrossRef]
  41. 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 Pt 1, 109–116. [Google Scholar] [CrossRef]
  42. 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]
  43. Kamalvand, G.; Ali-Khan, Z. Immunolocalization of lipid peroxidation/advanced glycation end products in amyloid A amyloidosis. Free Radic. Biol. Med. 2004, 36, 657–664. [Google Scholar] [CrossRef] [PubMed]
  44. Gerdemann, A.; Lemke, H.D.; Nothdurft, A.; Heidland, A.; Münch, G.; Bahner, U.; Schinzel, R. Low-molecular but not high-molecular advanced glycation end products (AGEs) are removed by high-flux dialysis. Clin. Nephrol. 2000, 54, 276–283. [Google Scholar] [PubMed]
  45. Thomas, M.C.; Forbes, J.M.; MacIsaac, R.; Jerums, G.; Cooper, M.E. Low-molecular weight advanced glycation end products: Markers of tissue AGE accumulation and more? Ann. N. Y. Acad. Sci. 2005, 1043, 644–654. [Google Scholar] [CrossRef]
  46. Manig, F.; Hellwig, M.; Pietz, F.; Henle, T. Studies about the Dietary Impact on “Free” Glycation Compounds in Human Saliva. Foods 2022, 11, 2112. [Google Scholar] [CrossRef]
  47. Hellwig, M.; Geissler, S.; Matthes, R.; Peto, A.; Silow, C.; Brandsch, M.; Henle, T. Transport of free and peptide-bound glycated amino acids: Synthesis, transepithelial flux at Caco-2 cell monolayers, and interaction with apical membrane transport proteins. Chembiochem A Eur. J. Chem. Biol. 2011, 12, 1270–1279. [Google Scholar] [CrossRef]
  48. Scheijen, J.; Hanssen, N.M.J.; van Greevenbroek, M.M.; Van der Kallen, C.J.; Feskens, E.J.M.; Stehouwer, C.D.A.; Schalkwijk, C.G. Dietary intake of advanced glycation endproducts is associated with higher levels of advanced glycation endproducts in plasma and urine: The CODAM study. Clin. Nutr. 2018, 37, 919–925. [Google Scholar] [CrossRef]
  49. Willemsen, S.; Hartog, J.W.; Heiner-Fokkema, M.R.; van Veldhuisen, D.J.; Voors, A.A. Advanced glycation end-products, a pathophysiological pathway in the cardiorenal syndrome. Heart Fail. Rev. 2012, 17, 221–228. [Google Scholar] [CrossRef]
  50. Cavero-Redondo, I.; Soriano-Cano, A.; Álvarez-Bueno, C.; Cunha, P.G.; Martínez-Hortelano, J.A.; Garrido-Miguel, M.; Berlanga-Macías, C.; Martínez-Vizcaíno, V. Skin Autofluorescence-Indicated Advanced Glycation End Products as Predictors of Cardiovascular and All-Cause Mortality in High-Risk Subjects: A Systematic Review and Meta-analysis. J. Am. Heart Assoc. 2018, 7, e009833. [Google Scholar] [CrossRef]
  51. Bakris, G.L.; Bank, A.J.; Kass, D.A.; Neutel, J.M.; Preston, R.A.; Oparil, S. Advanced glycation end-product cross-link breakers. A novel approach to cardiovascular pathologies related to the aging process. Am. J. Hypertens. 2004, 17, 23s–30s. [Google Scholar] [CrossRef]
  52. Nagai, R.; Murray, D.B.; Metz, T.O.; Baynes, J.W. Chelation: A fundamental mechanism of action of AGE inhibitors, AGE breakers, and other inhibitors of diabetes complications. Diabetes 2012, 61, 549–559. [Google Scholar] [CrossRef] [PubMed]
  53. Reddy, V.P.; Beyaz, A. Inhibitors of the Maillard reaction and AGE breakers as therapeutics for multiple diseases. Drug Discov. Today 2006, 11, 646–654. [Google Scholar] [CrossRef]
  54. Voziyan, P.A.; Khalifah, R.G.; Thibaudeau, C.; Yildiz, A.; Jacob, J.; Serianni, A.S.; Hudson, B.G. Modification of proteins in vitro by physiological levels of glucose: Pyridoxamine inhibits conversion of Amadori intermediate to advanced glycation end-products through binding of redox metal ions. J. Biol. Chem. 2003, 278, 46616–46624. [Google Scholar] [CrossRef] [PubMed]
  55. Candido, R.; Forbes, J.M.; Thomas, M.C.; Thallas, V.; Dean, R.G.; Burns, W.C.; Tikellis, C.; Ritchie, R.H.; Twigg, S.M.; Cooper, M.E.; et al. A breaker of advanced glycation end products attenuates diabetes-induced myocardial structural changes. Circ. Res. 2003, 92, 785–792. [Google Scholar] [CrossRef] [PubMed]
  56. Wu, X.; Monnier, V.M. Enzymatic deglycation of proteins. Arch. Biochem. Biophys. 2003, 419, 16–24. [Google Scholar] [CrossRef] [PubMed]
  57. Ishibashi, Y.; Yamagishi, S.; Matsui, T.; Ohta, K.; Tanoue, R.; Takeuchi, M.; Ueda, S.; Nakamura, K.; Okuda, S. Pravastatin inhibits advanced glycation end products (AGEs)-induced proximal tubular cell apoptosis and injury by reducing receptor for AGEs (RAGE) level. Metab. Clin. Exp. 2012, 61, 1067–1072. [Google Scholar] [CrossRef] [PubMed]
  58. Miyata, T.; van Ypersele de Strihou, C.; Ueda, Y.; Ichimori, K.; Inagi, R.; Onogi, H.; Ishikawa, N.; Nangaku, M.; Kurokawa, K. Angiotensin II receptor antagonists and angiotensin-converting enzyme inhibitors lower in vitro the formation of advanced glycation end products: Biochemical mechanisms. J. Am. Soc. Nephrol. JASN 2002, 13, 2478–2487. [Google Scholar] [CrossRef]
  59. Mustata, G.T.; Rosca, M.; Biemel, K.M.; Reihl, O.; Smith, M.A.; Viswanathan, A.; Strauch, C.; Du, Y.; Tang, J.; Kern, T.S.; et al. Paradoxical effects of green tea (Camellia sinensis) and antioxidant vitamins in diabetic rats: Improved retinopathy and renal mitochondrial defects but deterioration of collagen matrix glycoxidation and cross-linking. Diabetes 2005, 54, 517–526. [Google Scholar] [CrossRef]
  60. Bronowicka-Szydełko, A.; Krzystek-Korpacka, M.; Gacka, M.; Pietkiewicz, J.; Jakobsche-Policht, U.; Gamian, A. Association of Novel Advanced Glycation End-Product (AGE10) with Complications of Diabetes as Measured by Enzyme-Linked Immunosorbent Assay. J. Clin. Med. 2021, 10, 4499. [Google Scholar] [CrossRef]
  61. Nakayama, H.; Makita, Z.; Kato, M.; Taneda, S.; Yoshida, H.; Yanagisawa, K.; Nakagawa, S. Quantitative enzyme-linked immunosorbent assay (ELISA) for non-enzymatically glycated serum protein. J. Immunol. Methods 1987, 99, 95–100. [Google Scholar] [CrossRef]
  62. Maciel, E.; da Silva, R.N.; Simões, C.; Melo, T.; Ferreira, R.; Domingues, P.; Domingues, M.R. Liquid chromatography-tandem mass spectrometry of phosphatidylserine advanced glycated end products. Chem. Phys. Lipids 2013, 174, 1–7. [Google Scholar] [CrossRef]
  63. Nagai, R.; Shirakawa, J.; Ohno, R.; Hatano, K.; Sugawa, H.; Arakawa, S.; Ichimaru, K.; Kinoshita, S.; Sakata, N.; Nagai, M. Antibody-based detection of advanced glycation end-products: Promises vs. limitations. Glycoconj. J. 2016, 33, 545–552. [Google Scholar] [CrossRef] [PubMed]
  64. Milkovska-Stamenova, S.; Schmidt, R.; Frolov, A.; Birkemeyer, C. GC-MS Method for the Quantitation of Carbohydrate Intermediates in Glycation Systems. J. Agric. Food Chem. 2015, 63, 5911–5919. [Google Scholar] [CrossRef] [PubMed]
  65. Meerwaldt, R.; Links, T.; Graaff, R.; Thorpe, S.R.; Baynes, J.W.; Hartog, J.; Gans, R.; Smit, A. Simple noninvasive measurement of skin autofluorescence. Ann. N. Y. Acad. Sci. 2005, 1043, 290–298. [Google Scholar] [CrossRef]
  66. Mulder, D.J.; Water, T.V.; Lutgers, H.L.; Graaff, R.; Gans, R.O.; Zijlstra, F.; Smit, A.J. Skin autofluorescence, a novel marker for glycemic and oxidative stress-derived advanced glycation endproducts: An overview of current clinical studies, evidence, and limitations. Diabetes Technol. Ther. 2006, 8, 523–535. [Google Scholar] [CrossRef] [PubMed]
  67. Larsson, M.; Favilla, R.; Strömberg, T. Assessment of advanced glycated end product accumulation in skin using auto fluorescence multispectral imaging. Comput. Biol. Med. 2017, 85, 106–111. [Google Scholar] [CrossRef] [PubMed]
  68. Maasen, K.; van Greevenbroek, M.M.J.; Scheijen, J.; van der Kallen, C.J.H.; Stehouwer, C.D.A.; Schalkwijk, C.G. High dietary glycemic load is associated with higher concentrations of urinary advanced glycation endproducts: The Cohort on Diabetes and Atherosclerosis Maastricht (CODAM) Study. Am. J. Clin. Nutr. 2019, 110, 358–366. [Google Scholar] [CrossRef]
  69. Delgado-Andrade, C.; Tessier, F.J.; Niquet-Leridon, C.; Seiquer, I.; Pilar Navarro, M. Study of the urinary and faecal excretion of Nε-carboxymethyllysine in young human volunteers. Amino Acids 2012, 43, 595–602. [Google Scholar] [CrossRef]
  70. Brinkley, T.E.; Semba, R.D.; Kritchevsky, S.B.; Houston, D.K. Dietary protein intake and circulating advanced glycation end product/receptor for advanced glycation end product concentrations in the Health, Aging, and Body Composition Study. Am. J. Clin. Nutr. 2020, 112, 1558–1565. [Google Scholar] [CrossRef]
  71. Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet; a Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef]
  72. Morris, L.; Bhatnagar, D. The Mediterranean diet. Curr. Opin. Lipidol. 2016, 27, 89–91. [Google Scholar] [CrossRef]
  73. Gerrits, E.G.; Lutgers, H.L.; Smeets, G.H.; Groenier, K.H.; Smit, A.J.; Gans, R.O.; Bilo, H.J. Skin autofluorescence: A pronounced marker of mortality in hemodialysis patients. Nephron Extra 2012, 2, 184–191. [Google Scholar] [CrossRef] [PubMed]
  74. Lopez-Moreno, J.; Quintana-Navarro, G.M.; Delgado-Lista, J.; Garcia-Rios, A.; Delgado-Casado, N.; Camargo, A.; Perez-Martinez, P.; Striker, G.E.; Tinahones, F.J.; Perez-Jimenez, F.; et al. Mediterranean Diet Reduces Serum Advanced Glycation End Products and Increases Antioxidant Defenses in Elderly Adults: A Randomized Controlled Trial. J. Am. Geriatr. Soc. 2016, 64, 901–904. [Google Scholar] [CrossRef] [PubMed]
  75. Lopez-Moreno, J.; Quintana-Navarro, G.M.; Camargo, A.; Jimenez-Lucena, R.; Delgado-Lista, J.; Marin, C.; Tinahones, F.J.; Striker, G.E.; Roche, H.M.; Perez-Martinez, P.; et al. Dietary fat quantity and quality modifies advanced glycation end products metabolism in patients with metabolic syndrome. Mol. Nutr. Food Res. 2017, 61, 1601029. [Google Scholar] [CrossRef] [PubMed]
  76. 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]
  77. Uribarri, J.; Cai, W.; Woodward, M.; Tripp, E.; Goldberg, L.; Pyzik, R.; Yee, K.; Tansman, L.; Chen, X.; Mani, V.; et al. Elevated serum advanced glycation endproducts in obese indicate risk for the metabolic syndrome: A link between healthy and unhealthy obesity? J. Clin. Endocrinol. Metab. 2015, 100, 1957–1966. [Google Scholar] [CrossRef]
  78. Poulsen, M.W.; Bak, M.J.; Andersen, J.M.; Monošík, R.; Giraudi-Futin, A.C.; Holst, J.J.; Nielsen, J.; Lauritzen, L.; Larsen, L.H.; Bügel, S.; et al. Effect of dietary advanced glycation end products on postprandial appetite, inflammation, and endothelial activation in healthy overweight individuals. Eur. J. Nutr. 2014, 53, 661–672. [Google Scholar] [CrossRef]
  79. Schiekofer, S.; Franke, S.; Andrassy, M.; Chen, J.; Rudofsky, G.; Schneider, J.G.; von Eynatten, M.; Wendt, T.; Morcos, M.; Kientsch-Engel, R.; et al. Postprandial mononuclear NF-kappaB activation is independent of the AGE-content of a single meal. Exp. Clin. Endocrinol. Diabetes 2006, 114, 160–167. [Google Scholar] [CrossRef]
  80. Negrean, M.; Stirban, A.; Stratmann, B.; Gawlowski, T.; Horstmann, T.; Götting, C.; Kleesiek, K.; Mueller-Roesel, M.; Koschinsky, T.; Uribarri, J.; et al. Effects of low- and high-advanced glycation endproduct meals on macro- and microvascular endothelial function and oxidative stress in patients with type 2 diabetes mellitus. Am. J. Clin. Nutr. 2007, 85, 1236–1243. [Google Scholar] [CrossRef]
  81. Sohouli, M.H.; Fatahi, S.; Sharifi-Zahabi, E.; Santos, H.O.; Tripathi, N.; Lari, A.; Pourrajab, B.; Kord-Varkaneh, H.; Găman, M.A.; Shidfar, F. The Impact of Low Advanced Glycation End Products Diet on Metabolic Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2021, 12, 766–776. [Google Scholar] [CrossRef]
  82. 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]
  83. 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] [PubMed]
  84. Di Pino, A.; Currenti, W.; Urbano, F.; Mantegna, C.; Purrazzo, G.; Piro, S.; Purrello, F.; Rabuazzo, A.M. Low advanced glycation end product diet improves the lipid and inflammatory profiles of prediabetic subjects. J. Clin. Lipidol. 2016, 10, 1098–1108. [Google Scholar] [CrossRef] [PubMed]
  85. Baye, E.; de Courten, M.P.; Walker, K.; Ranasinha, S.; Earnest, A.; Forbes, J.M.; de Courten, B. Effect of dietary advanced glycation end products on inflammation and cardiovascular risks in healthy overweight adults: A randomised crossover trial. Sci. Rep. 2017, 7, 4123. [Google Scholar] [CrossRef] [PubMed]
  86. Macías-Cervantes, M.H.; Rodríguez-Soto, J.M.; Uribarri, J.; Díaz-Cisneros, F.J.; Cai, W.; Garay-Sevilla, M.E. Effect of an advanced glycation end product-restricted diet and exercise on metabolic parameters in adult overweight men. Nutrition 2015, 31, 446–451. [Google Scholar] [CrossRef] [PubMed]
  87. Tantalaki, E.; Piperi, C.; Livadas, S.; Kollias, A.; Adamopoulos, C.; Koulouri, A.; Christakou, C.; Diamanti-Kandarakis, E. Impact of dietary modification of advanced glycation end products (AGEs) on the hormonal and metabolic profile of women with polycystic ovary syndrome (PCOS). Hormones 2014, 13, 65–73. [Google Scholar] [CrossRef] [PubMed]
  88. Uribarri, J.; Peppa, M.; Cai, W.; Goldberg, T.; Lu, M.; He, C.; Vlassara, H. Restriction of dietary glycotoxins reduces excessive advanced glycation end products in renal failure patients. J. Am. Soc. Nephrol. JASN 2003, 14, 728–731. [Google Scholar] [CrossRef] [PubMed]
  89. de Courten, B.; de Courten, M.P.; Soldatos, G.; Dougherty, S.L.; Straznicky, N.; Schlaich, M.; Sourris, K.C.; Chand, V.; Scheijen, J.L.; Kingwell, B.A.; et al. Diet low in advanced glycation end products increases insulin sensitivity in healthy overweight individuals: A double-blind, randomized, crossover trial. Am. J. Clin. Nutr. 2016, 103, 1426–1433. [Google Scholar] [CrossRef]
  90. 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]
  91. Seiquer, I.; Ruiz-Roca, B.; Mesías, M.; Muñoz-Hoyos, A.; Galdó, G.; Ochoa, J.J.; Navarro, M.P. The antioxidant effect of a diet rich in Maillard reaction products is attenuated after consumption by healthy male adolescents. In vitro and in vivo comparative study. J. Sci. Food Agric. 2008, 88, 1245–1252. [Google Scholar]
  92. Stirban, A.; Kotsi, P.; Franke, K.; Strijowski, U.; Cai, W.; Götting, C.; Tschoepe, D. Acute macrovascular dysfunction in patients with type 2 diabetes induced by ingestion of advanced glycated β-lactoglobulins. Diabetes Care 2013, 36, 1278–1282. [Google Scholar] [CrossRef] [PubMed]
  93. Stirban, A.; Negrean, M.; Götting, C.; Uribarri, J.; Gawlowski, T.; Stratmann, B.; Kleesiek, K.; Koschinsky, T.; Vlassara, H.; Tschoepe, D. Dietary advanced glycation endproducts and oxidative stress: In vivo effects on endothelial function and adipokines. Ann. N. Y. Acad. Sci. 2008, 1126, 276–279. [Google Scholar] [CrossRef] [PubMed]
  94. Stirban, A.; Negrean, M.; Stratmann, B.; Götting, C.; Salomon, J.; Kleesiek, K.; Tschoepe, D. Adiponectin decreases postprandially following a heat-processed meal in individuals with type 2 diabetes: An effect prevented by benfotiamine and cooking method. Diabetes Care 2007, 30, 2514–2516. [Google Scholar] [CrossRef]
  95. Goudarzi, R.; Sedaghat, M.; Hedayati, M.; Hekmatdoost, A.; Sohrab, G. Low advanced Glycation end product diet improves the central obesity, insulin resistance and inflammatory profiles in Iranian patients with metabolic syndrome: A randomized clinical trial. J. Diabetes Metab. Disord. 2020, 19, 1129–1138. [Google Scholar] [CrossRef] [PubMed]
  96. Linkens, A.M.; Houben, A.J.; Niessen, P.M.; Wijckmans, N.E.; de Goei, E.E.; Van den Eynde, M.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. JCI Insight 2022, 7, e156950. [Google Scholar] [CrossRef]
  97. Birlouez-Aragon, I.; Saavedra, G.; Tessier, F.J.; Galinier, A.; Ait-Ameur, L.; Lacoste, F.; Niamba, C.N.; Alt, N.; Somoza, V.; Lecerf, J.M. A diet based on high-heat-treated foods promotes risk factors for diabetes mellitus and cardiovascular diseases. Am. J. Clin. Nutr. 2010, 91, 1220–1226. [Google Scholar] [CrossRef] [PubMed]
  98. Cordova, R.; Knaze, V.; Viallon, V.; Rust, P.; Schalkwijk, C.G.; Weiderpass, E.; Wagner, K.H.; Mayen-Chacon, A.L.; Aglago, E.K.; Dahm, C.C.; et al. Dietary intake of advanced glycation end products (AGEs) and changes in body weight in European adults. Eur. J. Nutr. 2020, 59, 2893–2904. [Google Scholar] [CrossRef]
  99. 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]
  100. Sohouli, M.H.; Sharifi-Zahabi, E.; Lari, A.; Fatahi, S.; Shidfar, F. The impact of low advanced glycation end products diet on obesity and related hormones: A systematic review and meta-analysis. Sci. Rep. 2020, 10, 22194. [Google Scholar] [CrossRef]
  101. Nigro, C.; Raciti, G.A.; Leone, A.; Fleming, T.H.; Longo, M.; Prevenzano, I.; Fiory, F.; Mirra, P.; D’Esposito, V.; Ulianich, L.; et al. Methylglyoxal impairs endothelial insulin sensitivity both in vitro and in vivo. Diabetologia 2014, 57, 1485–1494. [Google Scholar] [CrossRef]
  102. Nin, J.W.; Jorsal, A.; Ferreira, I.; Schalkwijk, C.G.; Prins, M.H.; Parving, H.H.; Tarnow, L.; Rossing, P.; Stehouwer, C.D. Higher plasma levels of advanced glycation end products are associated with incident cardiovascular disease and all-cause mortality in type 1 diabetes: A 12-year follow-up study. Diabetes Care 2011, 34, 442–447. [Google Scholar] [CrossRef] [PubMed]
  103. Tikellis, C.; Pickering, R.J.; Tsorotes, D.; Huet, O.; Cooper, M.E.; Jandeleit-Dahm, K.; Thomas, M.C. Dicarbonyl stress in the absence of hyperglycemia increases endothelial inflammation and atherogenesis similar to that observed in diabetes. Diabetes 2014, 63, 3915–3925. [Google Scholar] [CrossRef] [PubMed]
  104. Hanssen, N.M.; Beulens, J.W.; van Dieren, S.; Scheijen, J.L.; van der A, D.L.; Spijkerman, A.M.; van der Schouw, Y.T.; Stehouwer, C.D.; Schalkwijk, C.G. Plasma advanced glycation end products are associated with incident cardiovascular events in individuals with type 2 diabetes: A case-cohort study with a median follow-up of 10 years (EPIC-NL). Diabetes 2015, 64, 257–265. [Google Scholar] [CrossRef] [PubMed]
  105. Maessen, D.E.; Hanssen, N.M.; Lips, M.A.; Scheijen, J.L.; Willems van Dijk, K.; Pijl, H.; Stehouwer, C.D.; Schalkwijk, C.G. Energy restriction and Roux-en-Y gastric bypass reduce postprandial α-dicarbonyl stress in obese women with type 2 diabetes. Diabetologia 2016, 59, 2013–2017. [Google Scholar] [CrossRef]
  106. Van den Eynde, M.D.G.; Kusters, Y.; Houben, A.; Scheijen, J.; van Duynhoven, J.; Fazelzadeh, P.; Joris, P.J.; Plat, J.; Mensink, R.P.; Hanssen, N.M.J.; et al. Diet-induced weight loss reduces postprandial dicarbonyl stress in abdominally obese men: Secondary analysis of a randomized controlled trial. Clin. Nutr. 2021, 40, 2654–2662. [Google Scholar] [CrossRef]
  107. Wautier, M.P.; Chappey, O.; Corda, S.; Stern, D.M.; Schmidt, A.M.; Wautier, J.L. Activation of NADPH oxidase by AGE links oxidant stress to altered gene expression via RAGE. Am. J. Physiol. Endocrinol. Metab. 2001, 280, E685–E694. [Google Scholar] [CrossRef]
  108. Lee, H.W.; Gu, M.J.; Kim, Y.; Lee, J.Y.; Lee, S.; Choi, I.W.; Ha, S.K. Glyoxal-Lysine Dimer, an Advanced Glycation End Product, Induces Oxidative Damage and Inflammatory Response by Interacting with RAGE. Antioxidants 2021, 10, 1486. [Google Scholar] [CrossRef]
  109. Cepas, V.; Manig, F.; Mayo, J.C.; Hellwig, M.; Collotta, D.; Sanmartino, V.; Carrocera-Pumarino, R.; Collino, M.; Henle, T.; Sainz, R.M. In Vitro Evaluation of the Toxicological Profile and Oxidative Stress of Relevant Diet-Related Advanced Glycation End Products and Related 1,2-Dicarbonyls. Oxidative Med. Cell. Longev. 2021, 2021, 9912240. [Google Scholar] [CrossRef]
  110. Kong, X.; Lu, A.L.; Yao, X.M.; Hua, Q.; Li, X.Y.; Qin, L.; Zhang, H.M.; Meng, G.X.; Su, Q. Activation of NLRP3 Inflammasome by Advanced Glycation End Products Promotes Pancreatic Islet Damage. Oxidative Med. Cell. Longev. 2017, 2017, 9692546. [Google Scholar] [CrossRef]
  111. Son, S.; Hwang, I.; Han, S.H.; Shin, J.S.; Shin, O.S.; Yu, J.W. Advanced glycation end products impair NLRP3 inflammasome-mediated innate immune responses in macrophages. J. Biol. Chem. 2017, 292, 20437–20448. [Google Scholar] [CrossRef]
  112. Buetler, T.M.; Leclerc, E.; Baumeyer, A.; Latado, H.; Newell, J.; Adolfsson, O.; Parisod, V.; Richoz, J.; Maurer, S.; Foata, F.; et al. N(epsilon)-carboxymethyllysine-modified proteins are unable to bind to RAGE and activate an inflammatory response. Mol. Nutr. Food Res. 2008, 52, 370–378. [Google Scholar] [CrossRef] [PubMed]
  113. Reddy, S.; Bichler, J.; Wells-Knecht, K.J.; Thorpe, S.R.; Baynes, J.W. N epsilon-(carboxymethyl)lysine is a dominant advanced glycation end product (AGE) antigen in tissue proteins. Biochemistry 1995, 34, 10872–10878. [Google Scholar] [CrossRef] [PubMed]
  114. Tessier, F.J.; Niquet-Léridon, C.; Jacolot, P.; Jouquand, C.; Genin, M.; Schmidt, A.M.; Grossin, N.; Boulanger, E. Quantitative assessment of organ distribution of dietary protein-bound (13) C-labeled N(ɛ) -carboxymethyllysine after a chronic oral exposure in mice. Mol. Nutr. Food Res. 2016, 60, 2446–2456. [Google Scholar] [CrossRef] [PubMed]
  115. Song, F.; Hurtado del Pozo, C.; Rosario, R.; Zou, Y.S.; Ananthakrishnan, R.; Xu, X.; Patel, P.R.; Benoit, V.M.; Yan, S.F.; Li, H.; et al. RAGE regulates the metabolic and inflammatory response to high-fat feeding in mice. Diabetes 2014, 63, 1948–1965. [Google Scholar] [CrossRef] [PubMed]
  116. Gaens, K.H.; Goossens, G.H.; Niessen, P.M.; van Greevenbroek, M.M.; van der Kallen, C.J.; Niessen, H.W.; Rensen, S.S.; Buurman, W.A.; Greve, J.W.; Blaak, E.E.; et al. Nε-(carboxymethyl)lysine-receptor for advanced glycation end product axis is a key modulator of obesity-induced dysregulation of adipokine expression and insulin resistance. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
  117. Gaens, K.H.; Ferreira, I.; van de Waarenburg, M.P.; van Greevenbroek, M.M.; van der Kallen, C.J.; Dekker, J.M.; Nijpels, G.; Rensen, S.S.; Stehouwer, C.D.; Schalkwijk, C.G. Protein-Bound Plasma Nε-(Carboxymethyl)lysine Is Inversely Associated With Central Obesity and Inflammation and Significantly Explain a Part of the Central Obesity-Related Increase in Inflammation: The Hoorn and CODAM Studies. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 2707–2713. [Google Scholar] [CrossRef]
  118. Ruiz, H.H.; Nguyen, A.; Wang, C.; He, L.; Li, H.; Hallowell, P.; McNamara, C.; Schmidt, A.M. AGE/RAGE/DIAPH1 axis is associated with immunometabolic markers and risk of insulin resistance in subcutaneous but not omental adipose tissue in human obesity. Int. J. Obes. 2021, 45, 2083–2094. [Google Scholar] [CrossRef]
  119. Grossin, N.; Auger, F.; Niquet-Leridon, C.; Durieux, N.; Montaigne, D.; Schmidt, A.M.; Susen, S.; Jacolot, P.; Beuscart, J.B.; Tessier, F.J.; et al. Dietary CML-enriched protein induces functional arterial aging in a RAGE-dependent manner in mice. Mol. Nutr. Food Res. 2015, 59, 927–938. [Google Scholar] [CrossRef]
  120. Aronson, D. Cross-linking of glycated collagen in the pathogenesis of arterial and myocardial stiffening of aging and diabetes. J. Hypertens. 2003, 21, 3–12. [Google Scholar] [CrossRef]
  121. Goldin, A.; Beckman, J.A.; Schmidt, A.M.; Creager, M.A. Advanced glycation end products: Sparking the development of diabetic vascular injury. Circulation 2006, 114, 597–605. [Google Scholar] [CrossRef]
  122. Hegab, Z.; Gibbons, S.; Neyses, L.; Mamas, M.A. Role of advanced glycation end products in cardiovascular disease. World J. Cardiol. 2012, 4, 90–102. [Google Scholar] [CrossRef] [PubMed]
  123. Teissier, T.; Boulanger, É. The receptor for advanced glycation end-products (RAGE) is an important pattern recognition receptor (PRR) for inflammaging. Biogerontology 2019, 20, 279–301. [Google Scholar] [CrossRef] [PubMed]
  124. Matsumoto, S.; Yoshida, T.; Murata, H.; Harada, S.; Fujita, N.; Nakamura, S.; Yamamoto, Y.; Watanabe, T.; Yonekura, H.; Yamamoto, H.; et al. Solution structure of the variable-type domain of the receptor for advanced glycation end products: New insight into AGE-RAGE interaction. Biochemistry 2008, 47, 12299–12311. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, Y.; Wang, H.; Piper, M.G.; McMaken, S.; Mo, X.; Opalek, J.; Schmidt, A.M.; Marsh, C.B. sRAGE induces human monocyte survival and differentiation. J. Immunol. 2010, 185, 1822–1835. [Google Scholar] [CrossRef]
  126. Pollreisz, A.; Hudson, B.I.; Chang, J.S.; Qu, W.; Cheng, B.; Papapanou, P.N.; Schmidt, A.M.; Lalla, E. Receptor for advanced glycation endproducts mediates pro-atherogenic responses to periodontal infection in vascular endothelial cells. Atherosclerosis 2010, 212, 451–456. [Google Scholar] [CrossRef]
  127. Tian, J.; Avalos, A.M.; Mao, S.Y.; Chen, B.; Senthil, K.; Wu, H.; Parroche, P.; Drabic, S.; Golenbock, D.; Sirois, C.; et al. Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat. Immunol. 2007, 8, 487–496. [Google Scholar] [CrossRef] [PubMed]
  128. Liu, Y.; Liang, C.; Liu, X.; Liao, B.; Pan, X.; Ren, Y.; Fan, M.; Li, M.; He, Z.; Wu, J.; et al. AGEs increased migration and inflammatory responses of adventitial fibroblasts via RAGE, MAPK and NF-kappaB pathways. Atherosclerosis 2010, 208, 34–42. [Google Scholar] [CrossRef]
  129. Nah, S.S.; Choi, I.Y.; Yoo, B.; Kim, Y.G.; Moon, H.B.; Lee, C.K. Advanced glycation end products increases matrix metalloproteinase-1, -3, and -13, and TNF-alpha in human osteoarthritic chondrocytes. FEBS Lett. 2007, 581, 1928–1932. [Google Scholar] [CrossRef]
  130. Zhu, P.; Ren, M.; Yang, C.; Hu, Y.X.; Ran, J.M.; Yan, L. Involvement of RAGE, MAPK and NF-κB pathways in AGEs-induced MMP-9 activation in HaCaT keratinocytes. Exp. Dermatol. 2012, 21, 123–129. [Google Scholar] [CrossRef]
  131. Bierhaus, A.; Humpert, P.M.; Morcos, M.; Wendt, T.; Chavakis, T.; Arnold, B.; Stern, D.M.; Nawroth, P.P. Understanding RAGE, the receptor for advanced glycation end products. J. Mol. Med. 2005, 83, 876–886. [Google Scholar] [CrossRef]
  132. Kierdorf, K.; Fritz, G. RAGE regulation and signaling in inflammation and beyond. J. Leukoc. Biol. 2013, 94, 55–68. [Google Scholar] [CrossRef] [PubMed]
  133. Gao, Z.Q.; Yang, C.; Wang, Y.Y.; Wang, P.; Chen, H.L.; Zhang, X.D.; Liu, R.; Li, W.L.; Qin, X.J.; Liang, X.; et al. RAGE upregulation and nuclear factor-kappaB activation associated with ageing rat cardiomyocyte dysfunction. Gen. Physiol. Biophys. 2008, 27, 152–158. [Google Scholar] [PubMed]
  134. D’Adamo, E.; Giannini, C.; Chiavaroli, V.; de Giorgis, T.; Verrotti, A.; Chiarelli, F.; Mohn, A. What is the significance of soluble and endogenous secretory receptor for advanced glycation end products in liver steatosis in obese prepubertal children? Antioxid. Redox Signal. 2011, 14, 1167–1172. [Google Scholar] [CrossRef]
  135. Schmidt, A.M.; Stern, D. Atherosclerosis and diabetes: The RAGE connection. Curr. Atheroscler. Rep. 2000, 2, 430–436. [Google Scholar] [CrossRef] [PubMed]
  136. Yoon, S.J.; Park, S.; Park, C.; Chang, W.; Cho, D.K.; Ko, Y.G.; Choi, D.; Kwon, H.M.; Jang, Y.; Chung, N. Association of soluble receptor for advanced glycation end-product with increasing central aortic stiffness in hypertensive patients. Coron. Artery Dis. 2012, 23, 85–90. [Google Scholar] [CrossRef]
  137. Koyama, H.; Nishizawa, Y. AGEs/RAGE in CKD: Irreversible metabolic memory road toward CVD? Eur. J. Clin. Investig. 2010, 40, 623–635. [Google Scholar] [CrossRef]
  138. Guclu, M.; Ali, A.; Eroglu, D.U.; Büyükuysal, S.O.; Cander, S.; Ocak, N. Serum Levels of sRAGE Are Associated with Body Measurements, but Not Glycemic Parameters in Patients with Prediabetes. Metab. Syndr. Relat. Disord. 2016, 14, 33–39. [Google Scholar] [CrossRef]
  139. Dozio, E.; Briganti, S.; Delnevo, A.; Vianello, E.; Ermetici, F.; Secchi, F.; Sardanelli, F.; Morricone, L.; Malavazos, A.E.; Corsi Romanelli, M.M. Relationship between soluble receptor for advanced glycation end products (sRAGE), body composition and fat distribution in healthy women. Eur. J. Nutr. 2017, 56, 2557–2564. [Google Scholar] [CrossRef]
  140. Rowisha, M.; El-Batch, M.; El Shikh, T.; El Melegy, S.; Aly, H. Soluble receptor and gene polymorphism for AGE: Relationship with obesity and cardiovascular risks. Pediatr. Res. 2016, 80, 67–71. [Google Scholar] [CrossRef]
  141. Miranda, E.R.; Somal, V.S.; Mey, J.T.; Blackburn, B.K.; Wang, E.; Farabi, S.; Karstoft, K.; Fealy, C.E.; Kashyap, S.; Kirwan, J.P.; et al. Circulating soluble RAGE isoforms are attenuated in obese, impaired-glucose-tolerant individuals and are associated with the development of type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 2017, 313, E631–E640. [Google Scholar] [CrossRef]
  142. Zaki, M.; Kamal, S.; Kholousi, S.; El-Bassyouni, H.T.; Yousef, W.; Reyad, H.; Mohamed, R.; Basha, W.A. Serum soluble receptor of advanced glycation end products and risk of metabolic syndrome in Egyptian obese women. EXCLI J. 2017, 16, 973–980. [Google Scholar] [CrossRef] [PubMed]
  143. Steenbeke, M.; De Bruyne, S.; De Buyzere, M.; Lapauw, B.; Speeckaert, R.; Petrovic, M.; Delanghe, J.R.; Speeckaert, M.M. The role of soluble receptor for advanced glycation end-products (sRAGE) in the general population and patients with diabetes mellitus with a focus on renal function and overall outcome. Crit. Rev. Clin. Lab. Sci. 2021, 58, 113–130. [Google Scholar] [CrossRef] [PubMed]
  144. Park, L.; Raman, K.G.; Lee, K.J.; Lu, Y.; Ferran, L.J., Jr.; Chow, W.S.; Stern, D.; Schmidt, A.M. Suppression of accelerated diabetic atherosclerosis by the soluble receptor for advanced glycation endproducts. Nat. Med. 1998, 4, 1025–1031. [Google Scholar] [CrossRef] [PubMed]
  145. Wautier, J.L.; Zoukourian, C.; Chappey, O.; Wautier, M.P.; Guillausseau, P.J.; Cao, R.; Hori, O.; Stern, D.; Schmidt, A.M. Receptor-mediated endothelial cell dysfunction in diabetic vasculopathy. Soluble receptor for advanced glycation end products blocks hyperpermeability in diabetic rats. J. Clin. Investig. 1996, 97, 238–243. [Google Scholar] [CrossRef]
  146. Bucciarelli, L.G.; Wendt, T.; Qu, W.; Lu, Y.; Lalla, E.; Rong, L.L.; Goova, M.T.; Moser, B.; Kislinger, T.; Lee, D.C.; et al. RAGE blockade stabilizes established atherosclerosis in diabetic apolipoprotein E-null mice. Circulation 2002, 106, 2827–2835. [Google Scholar] [CrossRef]
  147. Ha, C.H.; Kim, S.; Chung, J.; An, S.H.; Park, S.; Choi, D.; Kwon, K. Inhibitory effect of soluble RAGE in disturbed flow-induced atherogenesis. Int. J. Mol. Med. 2013, 32, 373–380. [Google Scholar] [CrossRef]
  148. Rodríguez-Mortera, R.; Luevano-Contreras, C.; Solorio-Meza, S.; Gómez-Ojeda, A.; Caccavello, R.; Bains, Y.; Gugliucci, A.; Garay-Sevilla, M.E. Soluble Receptor for Advanced Glycation End Products and Its Correlation with Vascular Damage in Adolescents with Obesity. Horm. Res. Paediatr. 2019, 92, 28–35. [Google Scholar] [CrossRef]
  149. Saremi, A.; Howell, S.; Schwenke, D.C.; Bahn, G.; Beisswenger, P.J.; Reaven, P.D. Advanced Glycation End Products, Oxidation Products, and the Extent of Atherosclerosis During the VA Diabetes Trial and Follow-up Study. Diabetes Care 2017, 40, 591–598. [Google Scholar] [CrossRef]
  150. Rabbani, N.; Xue, M.; Thornalley, P.J. Dicarbonyls and glyoxalase in disease mechanisms and clinical therapeutics. Glycoconj. J. 2016, 33, 513–525. [Google Scholar] [CrossRef]
  151. Gautieri, A.; Passini, F.S.; Silván, U.; Guizar-Sicairos, M.; Carimati, G.; Volpi, P.; Moretti, M.; Schoenhuber, H.; Redaelli, A.; Berli, M.; et al. Advanced glycation end-products: Mechanics of aged collagen from molecule to tissue. Matrix Biol. J. Int. Soc. Matrix Biol. 2017, 59, 95–108. [Google Scholar] [CrossRef]
  152. Bucala, R.; Makita, Z.; Vega, G.; Grundy, S.; Koschinsky, T.; Cerami, A.; Vlassara, H. Modification of low density lipoprotein by advanced glycation end products contributes to the dyslipidemia of diabetes and renal insufficiency. Proc. Natl. Acad. Sci. USA 1994, 91, 9441–9445. [Google Scholar] [CrossRef] [PubMed]
  153. Posch, K.; Simecek, S.; Wascher, T.C.; Jürgens, G.; Baumgartner-Parzer, S.; Kostner, G.M.; Graier, W.F. Glycated low-density lipoprotein attenuates shear stress-induced nitric oxide synthesis by inhibition of shear stress-activated L-arginine uptake in endothelial cells. Diabetes 1999, 48, 1331–1337. [Google Scholar] [CrossRef] [PubMed]
  154. Schmidt, A.M.; Hasu, M.; Popov, D.; Zhang, J.H.; Chen, J.; Yan, S.D.; Brett, J.; Cao, R.; Kuwabara, K.; Costache, G.; et al. Receptor for advanced glycation end products (AGEs) has a central role in vessel wall interactions and gene activation in response to circulating AGE proteins. Proc. Natl. Acad. Sci. USA 1994, 91, 8807–8811. [Google Scholar] [CrossRef] [PubMed]
  155. Maasen, K.; Eussen, S.; Dagnelie, P.C.; Houben, A.; Webers, C.A.B.; Schram, M.T.; Berendschot, T.; Stehouwer, C.D.A.; Opperhuizen, A.; van Greevenbroek, M.M.J.; et al. Habitual intake of dietary methylglyoxal is associated with less low-grade inflammation: The Maastricht Study. Am. J. Clin. Nutr. 2022, 116, 1715–1728. [Google Scholar] [CrossRef]
  156. Linkens, A.M.A.; Houben, A.; Kroon, A.A.; Schram, M.T.; Berendschot, T.; Webers, C.A.B.; van Greevenbroek, M.; Henry, R.M.A.; de Galan, B.; Stehouwer, C.D.A.; et al. Habitual intake of dietary advanced glycation end products is not associated with generalized microvascular function-the Maastricht Study. Am. J. Clin. Nutr. 2022, 115, 444–455. [Google Scholar] [CrossRef]
  157. Linkens, A.M.; Eussen, S.J.; Houben, A.J.; Kroon, A.A.; Schram, M.T.; Reesink, K.D.; Dagnelie, P.C.; Henry, R.M.; van Greevenbroek, M.; Wesselius, A.; et al. Habitual Intake of Dietary Advanced Glycation End Products Is Not Associated with Arterial Stiffness of the Aorta and Carotid Artery in Adults: The Maastricht Study. J. Nutr. 2021, 151, 1886–1893. [Google Scholar] [CrossRef]
  158. Hold, G.L. Western lifestyle: A ‘master’ manipulator of the intestinal microbiota? Gut 2014, 63, 5–6. [Google Scholar] [CrossRef]
  159. Agus, A.; Denizot, J.; Thévenot, J.; Martinez-Medina, M.; Massier, S.; Sauvanet, P.; Bernalier-Donadille, A.; Denis, S.; Hofman, P.; Bonnet, R.; et al. Western diet induces a shift in microbiota composition enhancing susceptibility to Adherent-Invasive E. coli infection and intestinal inflammation. Sci. Rep. 2016, 6, 19032. [Google Scholar] [CrossRef]
  160. Martinez, K.B.; Leone, V.; Chang, E.B. Western diets, gut dysbiosis, and metabolic diseases: Are they linked? Gut Microbes 2017, 8, 130–142. [Google Scholar] [CrossRef]
  161. Linkens, A.M.A.; van Best, N.; Niessen, P.M.; Wijckmans, N.E.G.; de Goei, E.E.C.; Scheijen, J.; van Dongen, M.; van Gool, C.; de Vos, W.M.; Houben, A.; 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]
  162. 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]
  163. Moschen, A.R.; Gerner, R.R.; Wang, J.; Klepsch, V.; Adolph, T.E.; 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 Microbe 2016, 19, 455–469. [Google Scholar] [CrossRef] [PubMed]
  164. Maynard, J.D.; Rohrscheib, M.; Way, J.F.; Nguyen, C.M.; Ediger, M.N. Noninvasive type 2 diabetes screening: Superior sensitivity to fasting plasma glucose and A1C. Diabetes Care 2007, 30, 1120–1124. [Google Scholar] [CrossRef]
  165. Fokkens, B.T.; van Waateringe, R.P.; Mulder, D.J.; Wolffenbuttel, B.H.R.; Smit, A.J. Skin autofluorescence improves the Finnish Diabetes Risk Score in the detection of diabetes in a large population-based cohort: The LifeLines Cohort Study. Diabetes Metab. 2018, 44, 424–430. [Google Scholar] [CrossRef] [PubMed]
  166. de Vos, L.C.; Mulder, D.J.; Smit, A.J.; Dullaart, R.P.; Kleefstra, N.; Lijfering, W.M.; Kamphuisen, P.W.; Zeebregts, C.J.; Lefrandt, J.D. Skin autofluorescence is associated with 5-year mortality and cardiovascular events in patients with peripheral artery disease. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 933–938. [Google Scholar] [CrossRef]
  167. Meerwaldt, R.; Lutgers, H.L.; Links, T.P.; Graaff, R.; Baynes, J.W.; Gans, R.O.; Smit, A.J. Skin autofluorescence is a strong predictor of cardiac mortality in diabetes. Diabetes Care 2007, 30, 107–112. [Google Scholar] [CrossRef] [PubMed]
  168. Lutgers, H.L.; Gerrits, E.G.; Graaff, R.; Links, T.P.; Sluiter, W.J.; Gans, R.O.; Bilo, H.J.; Smit, A.J. Skin autofluorescence provides additional information to the UK Prospective Diabetes Study (UKPDS) risk score for the estimation of cardiovascular prognosis in type 2 diabetes mellitus. Diabetologia 2009, 52, 789–797. [Google Scholar] [CrossRef]
  169. Fraser, S.D.; Roderick, P.J.; McIntyre, N.J.; Harris, S.; McIntyre, C.W.; Fluck, R.J.; Taal, M.W. Skin autofluorescence and all-cause mortality in stage 3 CKD. Clin. J. Am. Soc. Nephrol. CJASN 2014, 9, 1361–1368. [Google Scholar] [CrossRef]
  170. Siriopol, D.; Hogas, S.; Veisa, G.; Mititiuc, I.; Volovat, C.; Apetrii, M.; Onofriescu, M.; Busila, I.; Oleniuc, M.; Covic, A. Tissue advanced glycation end products (AGEs), measured by skin autofluorescence, predict mortality in peritoneal dialysis. Int. Urol. Nephrol. 2015, 47, 563–569. [Google Scholar] [CrossRef]
  171. Shardlow, A.; McIntyre, N.J.; Kolhe, N.V.; Nellums, L.B.; Fluck, R.J.; McIntyre, C.W.; Taal, M.W. The association of skin autofluorescence with cardiovascular events and all-cause mortality in persons with chronic kidney disease stage 3: A prospective cohort study. PLoS Med. 2020, 17, e1003163. [Google Scholar] [CrossRef]
  172. Arsov, S.; Trajceska, L.; van Oeveren, W.; Smit, A.J.; Dzekova, P.; Stegmayr, B.; Sikole, A.; Rakhorst, G.; Graaff, R. Increase in skin autofluorescence and release of heart-type fatty acid binding protein in plasma predicts mortality of hemodialysis patients. Artif. Organs 2013, 37, E114–E122. [Google Scholar] [CrossRef] [PubMed]
  173. Furuya, F.; Shimura, H.; Takahashi, K.; Akiyama, D.; Motosugi, A.; Ikegishi, Y.; Haraguchi, K.; Kobayashi, T. Skin autofluorescence is a predictor of cardiovascular disease in chronic kidney disease patients. Ther. Apher. Dial. 2015, 19, 40–44. [Google Scholar] [CrossRef] [PubMed]
  174. Kimura, H.; Tanaka, K.; Kanno, M.; Watanabe, K.; Hayashi, Y.; Asahi, K.; Suzuki, H.; Sato, K.; Sakaue, M.; Terawaki, H.; et al. Skin autofluorescence predicts cardiovascular mortality in patients on chronic hemodialysis. Ther. Apher. Dial. 2014, 18, 461–467. [Google Scholar] [CrossRef] [PubMed]
  175. Vélayoudom-Céphise, F.L.; Rajaobelina, K.; Helmer, C.; Nov, S.; Pupier, E.; Blanco, L.; Hugo, M.; Farges, B.; Astrugue, C.; Gin, H.; et al. Skin autofluorescence predicts cardio-renal outcome in type 1 diabetes: A longitudinal study. Cardiovasc. Diabetol. 2016, 15, 127. [Google Scholar] [CrossRef]
  176. Blanc-Bisson, C.; Velayoudom-Cephise, F.L.; Cougnard-Gregoire, A.; Helmer, C.; Rajaobelina, K.; Delcourt, C.; Alexandre, L.; Blanco, L.; Mohammedi, K.; Monlun, M.; et al. Skin autofluorescence predicts major adverse cardiovascular events in patients with type 1 diabetes: A 7-year follow-up study. Cardiovasc. Diabetol. 2018, 17, 82. [Google Scholar] [CrossRef]
  177. van Waateringe, R.P.; Fokkens, B.T.; Slagter, S.N.; van der Klauw, M.M.; van Vliet-Ostaptchouk, J.V.; Graaff, R.; Paterson, A.D.; Smit, A.J.; Lutgers, H.L.; Wolffenbuttel, B.H.R. Skin autofluorescence predicts incident type 2 diabetes, cardiovascular disease and mortality in the general population. Diabetologia 2019, 62, 269–280. [Google Scholar] [CrossRef]
  178. Raposeiras-Roubín, S.; Rodiño-Janeiro, B.K.; Paradela-Dobarro, B.; Grigorian-Shamagian, L.; García-Acuña, J.M.; Aguiar-Souto, P.; Jacquet-Hervet, M.; Reino-Maceiras, M.V.; González-Juanatey, J.R.; Álvarez, E. Fluorescent advanced glycation end products and their soluble receptor: The birth of new plasmatic biomarkers for risk stratification of acute coronary syndrome. PLoS ONE 2013, 8, e74302. [Google Scholar] [CrossRef]
  179. Raposeiras-Roubín, S.; Rodino-Janeiro, B.K.; Paradela-Dobarro, B.; Almansour, H.; Grigorian-Shamagian, L.; Reino-Maceiras, M.V.; García-Acuña, J.M.; González-Juanatey, J.R.; Álvarez, E. Advanced glycation end-products as long-term predictors of death and reinfarction after an acute coronary syndrome. Biomark. Med. 2015, 9, 209–216. [Google Scholar] [CrossRef]
  180. Jensen, L.J.; Lindberg, S.; Hoffmann, S.; Iversen, A.Z.; Pedersen, S.H.; Møgelvang, R.; Galatius, S.; Flyvbjerg, A.; Jensen, J.S.; Bjerre, M. Dynamic changes in sRAGE levels and relationship with cardiac function in STEMI patients. Clin. Biochem. 2015, 48, 297–301. [Google Scholar] [CrossRef]
  181. Choi, L.S.; Ahmed, K.; Kim, Y.S.; Yim, J.E. Skin accumulation of advanced glycation end products and cardiovascular risk in Korean patients with type 2 diabetes mellitus. Heliyon 2022, 8, e09571. [Google Scholar] [CrossRef]
  182. den Engelsen, C.; van den Donk, M.; Gorter, K.J.; Salomé, P.L.; Rutten, G.E. Advanced glycation end products measured by skin autofluorescence in a population with central obesity. Derm.-Endocrinol. 2012, 4, 33–38. [Google Scholar] [CrossRef] [PubMed]
  183. van Waateringe, R.P.; Slagter, S.N.; van Beek, A.P.; van der Klauw, M.M.; van Vliet-Ostaptchouk, J.V.; Graaff, R.; Paterson, A.D.; Lutgers, H.L.; Wolffenbuttel, B.H.R. Skin autofluorescence, a non-invasive biomarker for advanced glycation end products, is associated with the metabolic syndrome and its individual components. Diabetol. Metab. Syndr. 2017, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  184. Yamagishi, S.; Fukami, K.; Matsui, T. Evaluation of tissue accumulation levels of advanced glycation end products by skin autofluorescence: A novel marker of vascular complications in high-risk patients for cardiovascular disease. Int. J. Cardiol. 2015, 185, 263–268. [Google Scholar] [CrossRef] [PubMed]
  185. Da Moura Semedo, C.; Webb, M.; Waller, H.; Khunti, K.; Davies, M. Skin autofluorescence, a non-invasive marker of advanced glycation end products: Clinical relevance and limitations. Postgrad. Med. J. 2017, 93, 289–294. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Description of the working principle and operating procedure of skin auto fluorescence (SAF) reader [184,185]. (A) Interior of a SAF reader is comprised of a scanner, an illumination window, a spectrometer, a result monitor and a fibre probe. (B) The working principle and operating procedure of advanced glycation end products (AGEs) measurement by SAF reader are as follows: (1) Measurement should be undertaken in a semi-dark environment. A reader is switched on by pressing power button; (2) A subject’s forearm should be placed on the scanner; (3) Subject’s information including age is entered into the reader, and then a run button is pressed. UV light from light source is illuminated with a maximum wavelength of 300–420 nm upon about 1 cm2 of skin, when the shutter is opened. This UV light activates autofluorescent AGEs in the skin of a subject’s forearm through the illumination window. The emitted light from the skin is transferred through the fibre probe. A spectrometer detects 420–600 nm fluorescence from light. The autofluorescence of the activated AGEs is assessed by the ratio between the emitted light intensity (420–600 nm) and the excitation light intensity (300–420 nm) multiplied by 100 on the integrated spectrometer; (4) AGEs scores (expressed in arbitrary units) from the spectrometer are reported as SAF value through the monitor.
Figure 1. Description of the working principle and operating procedure of skin auto fluorescence (SAF) reader [184,185]. (A) Interior of a SAF reader is comprised of a scanner, an illumination window, a spectrometer, a result monitor and a fibre probe. (B) The working principle and operating procedure of advanced glycation end products (AGEs) measurement by SAF reader are as follows: (1) Measurement should be undertaken in a semi-dark environment. A reader is switched on by pressing power button; (2) A subject’s forearm should be placed on the scanner; (3) Subject’s information including age is entered into the reader, and then a run button is pressed. UV light from light source is illuminated with a maximum wavelength of 300–420 nm upon about 1 cm2 of skin, when the shutter is opened. This UV light activates autofluorescent AGEs in the skin of a subject’s forearm through the illumination window. The emitted light from the skin is transferred through the fibre probe. A spectrometer detects 420–600 nm fluorescence from light. The autofluorescence of the activated AGEs is assessed by the ratio between the emitted light intensity (420–600 nm) and the excitation light intensity (300–420 nm) multiplied by 100 on the integrated spectrometer; (4) AGEs scores (expressed in arbitrary units) from the spectrometer are reported as SAF value through the monitor.
Nutrients 15 02002 g001
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

Kim, Y. Blood and Tissue Advanced Glycation End Products as Determinants of Cardiometabolic Disorders Focusing on Human Studies. Nutrients 2023, 15, 2002. https://doi.org/10.3390/nu15082002

AMA Style

Kim Y. Blood and Tissue Advanced Glycation End Products as Determinants of Cardiometabolic Disorders Focusing on Human Studies. Nutrients. 2023; 15(8):2002. https://doi.org/10.3390/nu15082002

Chicago/Turabian Style

Kim, Yoona. 2023. "Blood and Tissue Advanced Glycation End Products as Determinants of Cardiometabolic Disorders Focusing on Human Studies" Nutrients 15, no. 8: 2002. https://doi.org/10.3390/nu15082002

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