Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biomol NMR ; 78(1): 61-72, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38114873

RESUMO

Reducing sugars can spontaneously react with free amines in protein side chains leading to posttranslational modifications (PTMs) called glycation. In contrast to glycosylation, glycation is a non-enzymatic modification with consequences on the overall charge, solubility, aggregation susceptibility and functionality of a protein. Glycation is a critical quality attribute of therapeutic monoclonal antibodies. In addition to glucose, also disaccharides like maltose can form glycation products. We present here a detailed NMR analysis of the Amadori product formed between proteins and maltose. For better comparison, data collection was done under denaturing conditions using 7 M urea-d4 in D2O. The here presented correlation patterns serve as a signature and can be used to identify maltose-based glycation in any protein that can be denatured. In addition to the model protein BSA, which can be readily glycated, we present data of the biotherapeutic abatacept containing maltose in its formulation buffer. With this contribution, we demonstrate that NMR spectroscopy is an independent method for detecting maltose-based glycation, that is suited for cross-validation with other methods.


Assuntos
Reação de Maillard , Maltose , Maltose/química , Ressonância Magnética Nuclear Biomolecular , Proteínas/metabolismo , Espectroscopia de Ressonância Magnética
2.
J Mol Model ; 29(3): 81, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36856830

RESUMO

CONTEXT: The Maillard reaction is a high-temperature reaction of amino acids and carbohydrates to produce macromolecular substances such as melanoidins and intermediate reducing ketones, aldehydes, and volatile compounds. At present, only very limited researches involved the reaction mechanisms of Maillard reaction, which causes a lot of confusion in understanding numerous food processes. The detailed calculations of Maillard reaction are urgently needed. METHODS: The density functional theory (DFT) method (M06-2X/6-311G*) was used to deeply explore the specific mechanism of the primary and intermediate stages of Maillard reaction for a selected model system. RESULTS: The results show that the basic reaction processes in primary stage are the formation of Schiff-base by the condensation of amino and carbonyl groups, and then, Schiff-base tautomerization twice through proton transfer to generate Amadori rearrangement products. In the intermediate stage, two main reaction paths, 1-2 and 2-3 enolization, were comprehensively investigated. The first route finally generates 5-hydroxymethylfurfural through isomerization, dehydration, hydrolysis, elimination, and condensation, and the second route products dicarbonyl compounds through isomerization and elimination and then Strecker degradation forms aldehydes through condensation, decarboxylation, hydrolysis, and elimination. The results show that both paths are involved in complex reactions, some are lower barrier reactions, and some higher barrier reactions. An important aspect is that water catalysis is critical in all of these reactions; it is present in most processes. Our study deepens the understanding of the Maillard reaction from molecular level and facilitate the regulation of some harmful products.

3.
Pharm Res ; 40(6): 1341-1353, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36510116

RESUMO

OBJECTIVE: Glycation is a non-enzymatic and spontaneous post-translational modification (PTM) generated by the reaction between reducing sugars and primary amine groups within proteins. Because glycation can alter the properties of proteins, it is a critical quality attribute of therapeutic monoclonal antibodies (mAbs) and should therefore be carefully monitored. The most abundant product of glycation is formed by glucose and lysine side chains resulting in fructoselysine after Amadori rearrangement. In proteomics, which routinely uses a combination of chromatography and mass spectrometry to analyze PTMs, there is no straight-forward way to distinguish between glycation products of a reducing monosaccharide and an additional hexose within a glycan, since both lead to a mass difference of 162 Da. METHODS: To verify that the observed mass change is indeed a glycation product, we developed an approach based on 2D NMR spectroscopy spectroscopy and full-length protein samples denatured using high concentrations of deuterated urea. RESULTS: The dominating ß-pyranose form of the Amadori product shows a characteristic chemical shift correlation pattern in 1H-13C HSQC spectra suited to identify glucose-induced glycation. The same pattern was observed in spectra of a variety of artificially glycated proteins, including two mAbs, as well as natural proteins. CONCLUSION: Based on this unique correlation pattern, 2D NMR spectroscopy can be used to unambiguously identify glucose-induced glycation in any protein of interest. We provide a robust method that is orthogonal to MS-based methods and can also be used for cross-validation.


Assuntos
Anticorpos Monoclonais , Glucose , Reação de Maillard , Processamento de Proteína Pós-Traducional , Espectroscopia de Ressonância Magnética
4.
Curr Protein Pept Sci ; 23(11): 773-781, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36177615

RESUMO

BACKGROUND: Glycation of immunoglobulin-G (IgG) molecules with monosaccharides may cause significant structural disability, thus resulting in their loss of function. The accumulation of AGEs formed from glycation plays an important role in the aliments associated with metabolic diseases. Therefore, excess sugar in plasma interferes with the functioning of IgG and may contribute to a wide range of diabetes-associated complications. The long-term formation of these heterogeneous AGEs may accumulate and affect plasma proteins, especially long-lived proteins. In this study, we analyze immunoglobulin-G (IgG) glycation with 2'-deoxyribose (deoxyribose) instigated modification in IgG structure and AGEs formation. METHODS: This study aims to glycate IgG from varying concentrations of pentose sugar, 2'-deoxyribose (deoxyribose). Various physicochemical methods and techniques characterized post glycation of IgG, both the native and its glycated analogue. The glycated protein will be assessed for its stability and perturbations by UV-VIS., fluorescence and FT-IR spectroscopic techniques. Moreover, the early glycation product will be done by NBT assay, and other biochemical parameters like HMF, carbonyl content and thioflavin-T assays were also performed to see the biochemical changes induced in the glycated IgG macromolecule. RESULTS: Glycation of protein macromolecules generates stable early glycation products (Amadori products). Later, these Amadori products involved a series of chemical reactions to form more stable advanced glycation end products (AGEs). Our experimental study results could validate the modification in IgG structure and AGEs formation. CONCLUSION: The formation of IgG-AGEs from glycation of IgG with deoxyribose could exert cellular toxicity, and might initiates secondary complications of diabetes. Therefore, this study emphasized the glycation reaction of IgG from deoxyribose, which has not been reported yet.


Assuntos
Complicações do Diabetes , Reação de Maillard , Humanos , Imunoglobulina G/química , Glicosilação , Açúcares , Desoxirribose , Pentoses , Espectroscopia de Infravermelho com Transformada de Fourier , Produtos Finais de Glicação Avançada/metabolismo
5.
Toxicol In Vitro ; 72: 105078, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33429044

RESUMO

Fructoselysine is formed upon heating during processing of food products, and being a key intermediate in advanced glycation end product formation considered to be potentially hazardous to human health. Human gut microbes can degrade fructoselysine to yield the short chain fatty acid butyrate. However, quantitative information on these biochemical reactions is lacking, and interindividual differences therein are not well established. Anaerobic incubations with pooled and individual human fecal slurries were optimized and applied to derive quantitative kinetic information for these biochemical reactions. Of 16 individuals tested, 11 were fructoselysine metabolizers, with Vmax, Km and kcat-values varying up to 14.6-fold, 9.5-fold, and 4.4-fold, respectively. Following fructoselysine exposure, 10 of these 11 metabolizers produced significantly increased butyrate concentrations, varying up to 8.6-fold. Bacterial taxonomic profiling of the fecal samples revealed differential abundant taxa for these reactions (e.g. families Ruminococcaceae, Christenellaceae), and Ruminococcus_1 showed the strongest correlation with fructoselysine degradation and butyrate production (ρ ≥ 0.8). This study highlights substantial interindividual differences in gut microbial degradation of fructoselysine. The presented method allows for quantification of gut microbial degradation kinetics for foodborne xenobiotics, and interindividual differences therein, which can be used to refine prediction of internal exposure.


Assuntos
Fezes/microbiologia , Lisina/análogos & derivados , Adulto , Variação Biológica da População , Ácidos Graxos Voláteis/metabolismo , Feminino , Microbioma Gastrointestinal/genética , Humanos , Lisina/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Ribossômico 16S , Adulto Jovem
6.
J Poult Sci ; 57(1): 63-66, 2020 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-32174766

RESUMO

Eighty 14-d-old single-comb White Leghorn male chicks were divided into 16 groups with five birds each. Fructosyl-valine, which is a valine-glucose-Amadori product, was intravenously (2,250 nmol/kg body weight) or orally (300 µmol/kg body weight) administered to chicks. Blood samples were collected 15, 30, 60, 120, 180, 360, 720 and 1440 min after administration. Plasma concentrations of fructosyl-valine were measured by using a liquid chromatography / mass spectrometry (LC/MS). The time course change in plasma fructosyl-valine concentration showed an exponential curve, as y=a+be-λt. The half-life of plasma fructosyl-valine was calculated by the following equation: (loge2)/λ. When fructosyl-valine was injected intravenously, the highest value for plasma fructosyl-valine concentration was observed 15 min after administration. When injected intravenously, the half-life of plasma fructosyl-valine was calculated to be 231 min. When fructosyl-valine was administered orally to chicks, the highest value for plasma fructosyl-valine concentration was observed 180 min after administration. When administered orally, the half-life of plasma fructosyl-valine was calculated to be 277 min. We conclude that the half-life of fructosyl-valine in plasma was approximately 4 h, which is longer than that of glycated tryptophan.

7.
Hemoglobin ; 43(1): 42-49, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31060394

RESUMO

Nonenzymatic glycation (NEG) of human hemoglobin (Hb A) consists of initial non covalent, reversible steps involving glucose and amino acid residues, which may also involve effector reagent(s) in the formation of labile Hb A1c (the conjugate acid of the Schiff base). Labile Hb A1c can then undergo slow, largely irreversible, formation of stable Hb A1c (the Amadori product). Stable Hb A1c is measured to assess diabetic progression after labile Hb A1c removal. This study aimed to increase the understanding of the distinctions between labile and stable Hb A1c from a mechanistic perspective in the presence of 2,3-bisphosphoglycerate (2,3-BPG). 2,3-Bisphosphoglycerate is an effector reagent that reversibly binds in the Hb A1c pocket and modestly enhances overall NEG rate. The deprotonation of C2 on labile Hb A1c in the formation of the Amadori product was previously proposed to be rate-limiting. Computational chemistry was used here to identify the mechanism(s) by which 2,3-BPG facilitates the deprotonation of C2 on labile Hb A1c. 2,3-Bisphosphoglycerate is capable of abstracting protons on C2 and the α-nitrogen of labile Hb A1c and can also deprotonate water and/or amino acid residues, therefore preparing these secondary reagents to deprotonate labile Hb A1c. Parallel reactions not leading to an Amadori product were found that include formation of the neutral Schiff base, dissociation of glucose from the protein, and cyclic glycosylamine formation. These heretofore under appreciated parallel reactions may help explain both the selective removal of labile from stable Hb A1c and the slow rate of NEG.


Assuntos
2,3-Difosfoglicerato/metabolismo , Hemoglobinas Glicadas/metabolismo , 2,3-Difosfoglicerato/química , Aminoácidos/química , Glucose/química , Glucose/metabolismo , Hemoglobinas Glicadas/química , Glicosilação , Humanos , Ligação Proteica , Multimerização Proteica
8.
J Agric Food Chem ; 66(44): 11806-11811, 2018 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-30336014

RESUMO

In this study, α-dicarbonyl compounds consisting of a backbone with six carbon atoms resulting from the Maillard reaction of d-fructose with γ-aminobutyric acid were determined. The reaction was carried out under mild reaction conditions at 50 °C and water contents between 0 and 90%. A thus far unknown α-dicarbonyl compound was found as the main product in the first 24 h at water contents below 50%. After isolation of its stable quinoxaline derivative, it was possible to identify the compound as 2-deoxy-d- glycero-hexo-3,4-diulose (2-deoxyglucosone). For the first time, the four C6-α-dicarbonyl compounds, 1-deoxyglucosone, 2-deoxyglucosone, 3-deoxyglucosone, and 4-deoxyglucosone, could be identified in the Maillard reaction of a hexose at the same time. This indicates the formation of a 2,3-eneaminol from the Schiff base of d-fructose and the formation of 2-amino-2-deoxy-3-ketose as an alternative to the Heyns product.


Assuntos
Desoxiglucose/química , Frutose/química , Ácido gama-Aminobutírico/química , Cinética , Reação de Maillard , Estrutura Molecular
9.
J Bacteriol ; 199(22)2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28847920

RESUMO

Salmonella enterica can utilize fructose-asparagine (F-Asn) as a source of carbon and nitrogen. This capability has been attributed to five genes in the fra locus. Previously, we determined that mutations in fraB (deglycase), fraD (kinase), or fraA (transporter) eliminated the ability of Salmonella to grow on F-Asn, while a mutation in fraE allowed partial growth. We hypothesized that FraE, a putative periplasmic fructose-asparaginase, converts F-Asn to NH4 + and fructose-aspartate (F-Asp). FraA could then transport F-Asp into the cytoplasm for subsequent catabolism. Here, we report that growth of the fraE mutant on F-Asn is caused by a partially redundant activity provided by AnsB, a periplasmic asparaginase. Indeed, a fraE ansB double mutant is unable to grow on F-Asn. Moreover, biochemical assays using periplasmic extracts of mutants that express only FraE or AnsB confirmed that each of these enzymes converts F-Asn to F-Asp and NH4 + However, FraE does not contribute to growth on asparagine. We tested and confirmed the hypothesis that a fraE ansB mutant can grow on F-Asp, while mutants lacking fraA, fraD, or fraB cannot. This finding provides strong evidence that FraA transports F-Asp but not F-Asn from the periplasm to the cytoplasm. Previously, we determined that F-Asn is toxic to a fraB mutant due to the accumulation of the FraB substrate, 6-phosphofructose-aspartate (6-P-F-Asp). Here, we found that, as expected, a fraB mutant is also inhibited by F-Asp. Collectively, these findings contribute to a better understanding of F-Asn utilization by Salmonella IMPORTANCE Salmonella is able to utilize fructose-asparagine (F-Asn) as a nutrient. We recently reported that the disruption of a deglycase enzyme in the F-Asn utilization pathway inhibits the growth of Salmonella in mice and recognized this pathway as a novel and specific drug target. Here, we characterize the first step in the pathway wherein FraE hydrolyzes F-Asn to release NH4 + and F-Asp in the periplasm of the cell. A fraE mutant continues to grow slowly on F-Asn due to asparaginase activity encoded by ansB.

10.
Appl Physiol Nutr Metab ; 42(10): 1054-1063, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28618238

RESUMO

Sweeteners have replaced the natural sugars in the food and beverage industry because of many reasons, such as hyperglycemia and cost. Saccharin, sucralose, aspartame and acesulfame-K are the most commonly used sweeteners. In the present study, the abovementioned artificial sweeteners were used to assess their glycating properties by established methods such as browning, fructosamine assay, determination of carbonyl content, protein aggregation, and measurement of fluorescence. Amadori and advanced glycation end products (AGEs) are formed as a result of the interaction between carbonyl groups of reducing sugars and amino groups of proteins and other macromolecules during glycation. The objective of this study was to investigate the influence of artificial sweeteners on the formation of AGEs and protein oxidation in an in vitro model of glucose-mediated protein glycation. The results indicated that the abovementioned artificial sweeteners do not enhance the process of glycation. On the other hand, acesulfame-K was found to have antiglycating potential as it caused decreased formation of Amadori products and AGEs. Further studies are essential in the characterization of Amadori products and AGEs produced as a result of interaction between sweeteners and proteins, which are interfered with by sweeteners. This study is significant in understanding the probable role of artificial sweeteners in the process of glycation and the subsequent effect on macromolecular alteration.


Assuntos
Produtos Finais de Glicação Avançada/metabolismo , Carbonilação Proteica/efeitos dos fármacos , Edulcorantes/farmacologia , Tiazinas/farmacologia , Dano ao DNA/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Frutosamina/metabolismo , Glucose/farmacologia , Glicosilação , Reação de Maillard , Oxirredução , Agregados Proteicos , Espectrometria de Fluorescência
11.
J Poult Sci ; 54(3): 242-246, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-32908432

RESUMO

Glycation is a chemical reaction in which reducing sugars bind non-enzymatically to compounds containing amino groups. Avian species like chickens are hyperglycemic animals and have high body temperature compared to mammalian species, which enables avian species to accelerate the glycation of proteins and amino acids with glucose. Although varying dietary crude protein (CP) levels alter plasma concentrations of proteins and amino acids, the influence of varying CP levels on the glycation of plasma proteins and amino acids has not been studied so far. In the present study, therefore, glycation of albumin, tryptophan and valine in the plasma of chickens fed diets with varying CP levels (0, 10, 20, 40 and 60%) was examined. At the end of the experimental period, blood samples were collected and plasma concentrations of glycoalbumin, glycated tryptophan (tryptophan-Amadori product and (1R, 3S) - 1 - (D - gluco - 1, 2, 3, 4, 5 - pentahydroxypentyl) - 1, 2, 3, 4 - tetrahydro - ß - carboline - 3 - carboxylic acid (PHP-THßC)), and valine-Amadori product were measured. Although plasma albumin concentration was reduced along with the decrease in dietary CP levels from 20% to 0%, glycoalbumin in the plasma was increased under such dietary conditions. Similar increase in the ratios of tryptophan-Amadori product to tryptophan and valine-Amadori product to valine in the plasma of chickens fed a protein-free diet was observed. These results suggest that dietary protein deficiency might enhance the non-enzymatic glycation of plasma proteins and amino acids in chickens.

12.
Food Chem ; 221: 489-495, 2017 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27979232

RESUMO

Thermal treatment preserves the microbiological safety of milk, but also induces Maillard reactions modifying for example proteins. The purpose of this study was evaluating the influence of consumer behaviors (storage and heating) on protein glycation degrees in bovine milk products. Lactosylation and hexosylation sites were identified in ultra-high temperature (UHT), lactose-free pasteurized, and lactose-free UHT milk (ULF) and infant formula (IF) using tandem mass spectrometry (electron transfer dissociation). Overall, 303 lactosylated and 199 hexosylated peptides were identified corresponding to 170 lactosylation (31 proteins) and 117 hexosylation sites (25 proteins). In quantitative terms, storage increased lactosylation up to fourfold in UHT and IF and hexosylation up to elevenfold in ULF and threefold in IF. These levels increased additionally twofold when the stored samples were heated (40°C). In conclusion, storage and heating appear to influence protein glycation levels in milk at similar or even higher degrees than industrial processing.


Assuntos
Armazenamento de Alimentos/normas , Temperatura Alta , Lactose/análise , Proteínas do Leite/análise , Leite/química , Animais , Bovinos , Armazenamento de Alimentos/métodos , Glicosilação , Temperatura Alta/efeitos adversos , Humanos , Lactente , Fórmulas Infantis/análise , Lactose/metabolismo , Reação de Maillard , Leite/metabolismo , Proteínas do Leite/metabolismo , Pasteurização , Peptídeos/análise , Peptídeos/metabolismo , Espectrometria de Massas em Tandem/métodos
13.
Food Chem ; 220: 87-92, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27855939

RESUMO

To investigate the reactivity of free guanidine and arginine in the formation of imidazolinone derivatives, model systems of guanidine or arginine/glucose or 13[C-6]-glucose were heated in aqueous solutions at110°C for 3h and the residues were analyzed by ESI/qTOF/MS using MS/MS and isotope labeling techniques. The analysis of the data indicated that guanidine and arginine formed both covalent and non-covalent interaction products. Covalent interactions included Amadori rearrangement at the α-nitrogen with glucose and imidazolinone formation with 3-deoxy-glucosone at the guanidine side-chain. Non-covalent interactions, such as self-interaction and interaction with free guanidine or arginine and glucose, were also observed. Guanidine underwent three sequential Amadori rearrangements and the free and mono-glycated guanidine also formed imidazolinone derivatives and their corresponding dehydration products and at the same time exhibiting various non-covalent interactions. On the other hand, arginine formed free Amadori product, free imidazolinone and Amadori-derived imidazolinone derivative in addition to methylglyoxal-derived hydroimidazolones.


Assuntos
Arginina/química , Glucose/química , Guanidina/química , Imidazóis/química , Aldeído Pirúvico/química , Espectrometria de Massas em Tandem
14.
Food Chem ; 215: 463-9, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27542499

RESUMO

To explore the interaction of nucleosides and nucleobases in the context of the Maillard reaction and to identify the selectivity of purine nitrogen atoms towards various electrophiles, model systems composed of adenine or adenosine, glycine, ribose and/or 2-furanmethanol (with and without copper) were studied in aqueous solutions heated at 110°C for 2h and subsequently analyzed by ESI/qTOF/MS/MS in addition to isotope labelling techniques. The results indicated that ribose selectively formed mono-ribosylated N(6) adenine, but in the presence of (Ade)2Cu complex the reaction mixture generated mono-, di- and tri-substituted sugar complexes and their hydrolysis products of mono-ribosylated N(6) and N(9) adenine adducts and di-ribosylated N(6,9) adenine. Furthermore, the reaction of 2-furanmethanol with adenine in the presence of ribose generated kinetin and its isomer, while its reaction with adenosine generated kinetin riboside, as confirmed by comparing the MS/MS profiles of these adducts to those of commercial standards.


Assuntos
Adenina/química , Adenosina/química , Cinetina/química , Reação de Maillard , Nitrogênio/química , Ribose/química , Espectrometria de Massas em Tandem
15.
J Agric Food Chem ; 64(38): 7234-43, 2016 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-27594145

RESUMO

The Maillard reaction is important for beer color and flavor, but little is known about the occurrence of individual glycated amino acids in beer. Therefore, seven Maillard reaction products (MRPs), namely, fructosyllysine, maltulosyllysine, pyrraline, formyline, maltosine, MG-H1, and argpyrimidine, were synthesized and quantitated in different types of beer (Pilsner, dark, bock, wheat, and nonalcoholic beers) by HPLC-ESI-MS/MS in the multiple reaction monitoring mode through application of the standard addition method. Free MRPs were analyzed directly. A high molecular weight fraction was isolated by dialysis and hydrolyzed enzymatically prior to analysis. Maltulosyllysine was quantitated for the first time in food. The most important free MRPs in beer are fructosyllysine (6.8-27.0 mg/L) and maltulosyllysine (3.7-21.8 mg/L). Beer contains comparatively high amounts of late-stage free MRPs such as pyrraline (0.2-1.6 mg/L) and MG-H1 (0.3-2.5 mg/L). Minor amounts of formyline (4-230 µg/L), maltosine (6-56 µg/L), and argpyrimidine (0.1-4.1 µg/L) were quantitated. Maltulosyllysine was the most significant protein-bound MRP, but both maltulosyllysine and fructosyllysine represent only 15-60% of the total protein-bound lysine-derived Amadori products. Differences in the patterns of protein-bound and free individual MRPs and the ratios between them were identified, which indicate differences in their chemical, biochemical, and microbiological stabilities during the brewing process.


Assuntos
Cerveja/análise , Manipulação de Alimentos , Reação de Maillard , Aminoácidos/análise , Lisina/análogos & derivados , Lisina/análise , Norleucina/análogos & derivados , Norleucina/análise , Ornitina/análogos & derivados , Ornitina/análise , Piridonas/análise , Pirimidinas/análise , Pirróis/análise , Espectrometria de Massas em Tandem
16.
J Photochem Photobiol B ; 163: 345-51, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27614245

RESUMO

Amadori product is an important and stable intermediate, which is produced during glycation process. It is a marker of hyperglycemia in diabetes mellitus, and its accumulation in the body contributes to microvascular complication of diabetes including diabetic nephropathy and retinopathy. In this study, the effect of acetoacetate on the formation of Amadori products and biophysical properties of human serum albumin (HSA), after incubation with glucose, was investigated using various methods. These included circular dichroism (CD), Fourier transform infrared (FTIR) spectroscopy, and UV-visible and fluorescence spectroscopy. Our results indicated that the production of Amadori products in HSA incubated with glucose (GHSA) was increased in the presence of acetoacetate. We also detected alterations in the secondary and tertiary structure of GHSA, which was increased in the presence of acetoacetate. These changes were attributed to the formation of covalent bonds between the carbonyl group of acetoacetate and the nucleophilic groups (lysine residues) of HSA. Thus, acetoacetate can enhance the production of Amadori products through formation of covalent bonds with biomaterials.


Assuntos
Acetoacetatos/farmacologia , Albumina Sérica/metabolismo , Glucose/metabolismo , Glicosilação/efeitos dos fármacos , Humanos , Estrutura Secundária de Proteína , Albumina Sérica/química
17.
Molecules ; 21(2): 251, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26907243

RESUMO

Accumulation of Advanced Glycation Endproducts (AGEs) in body tissues plays a major role in the development of diabetic complications. Here, the inhibitory effect of bioactive metabolites isolated from fruit hulls of Garcinia mangostana on AGE formation was investigated through bio-guided approach using aminoguanidine (AG) as a positive control. Including G. mangostana total methanol extract (GMT) in the reaction mixture of bovine serum albumin (BSA) and glucose or ribose inhibited the fluorescent and non-fluorescent AGEs formation in a dose dependent manner. The bioassay guided fractionation of GMT revealed isolation of four bioactive constituents from the bioactive fraction; which were identified as: garcimangosone D (1), aromadendrin-8-C-glucopyranoside (2), epicatechin (3), and 2,3',4,5',6-pentahydroxybenzophenone (4). All the tested compounds significantly inhibited fluorescent and non-fluorescent AGEs formation in a dose dependent manner whereas compound 3 (epicatechin) was found to be the most potent. In search for the level of action, addition of GMT, and compounds 2-4 inhibited fructosamine (Amadori product) and protein aggregation formation in both glucose and ribose. To explore the mechanism of action, it was found that addition of GMT and only compound (3) to reaction mixture increased protein thiol in both glucose and ribose while compounds 1, 2 and 4 only increased thiol in case of ribose. In conclusion, phenolic compounds 1-4 inhibited AGEs formation at the levels of Amadori product and protein aggregation formation through saving protein thiol.


Assuntos
Garcinia mangostana/química , Produtos Finais de Glicação Avançada/antagonistas & inibidores , Agregação Patológica de Proteínas/tratamento farmacológico , Compostos de Sulfidrila/química , Animais , Benzofenonas/química , Benzofenonas/isolamento & purificação , Benzofenonas/uso terapêutico , Catequina/química , Catequina/isolamento & purificação , Catequina/uso terapêutico , Bovinos , Flavonoides/química , Flavonoides/isolamento & purificação , Flavonoides/uso terapêutico , Glucose/química , Glucosídeos/química , Glucosídeos/isolamento & purificação , Glucosídeos/uso terapêutico , Produtos Finais de Glicação Avançada/química , Extratos Vegetais/química , Soroalbumina Bovina/química
18.
J Poult Sci ; 53(2): 153-156, 2016 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-32908378

RESUMO

Amadori products are non-enzymatically formed by binding carbonyl groups and amino groups. Glycated amino acids generated by reacting amino acid and glucose are also in a group of Amadori products of which the transport and metabolism have been investigated mainly in mammals but not in avians. In the present study, therefore, we examined whether dietary fructosyl-valine, which is one of the glycated amino acids, orally administrated to chickens can be incorporated into blood or not. Fructosyl-valine was orally administrated to the chicken and blood samples were collected at 0, 20, 40, 60, 120 and 180 min after administration. Plasma concentration of fructosyl-valine was measured by using LC/MS. The plasma concentration of fructosyl-valine was increased by passing time from 0 to 180 min after administration, and no change was observed in the control group. Conclusively, it was clarified that fructosyl-valine orally administrated to the chicken could be absorbed from gastrointestinal tract and incorporated into blood.

19.
J Agric Food Chem ; 63(30): 6723-30, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26186075

RESUMO

Maillard reaction products (MRPs) are taken up in substantial amounts with the daily diet, but the majority are not transported across the intestinal epithelium. The aim of this study was to obtain first insights into the stability of dietary MRPs in the presence of the intestinal microbiota. Four individual MRPs, namely, N-ε-fructosyllysine (FL), N-ε-carboxymethyllysine (CML), pyrraline (PYR), and maltosine (MAL), were anaerobically incubated with fecal suspensions from eight human volunteers at 37 °C for up to 72 h. The stability of the MRPs was measured by HPLC with UV and MS/MS detections. The Amadori product FL could no longer be detected after 4 h of incubation. Marked interindividual differences were observed for CML metabolism: Depending on the individual, at least 40.7 ± 1.5% of CML was degraded after 24 h of incubation, and the subjects could thus be tentatively grouped into fast and slow metabolizers of this compound. PYR was degraded by 20.3 ± 4.4% during 24 h by all subjects. The concentration of MAL was not significantly lowered in the presence of fecal suspensions. In no case could metabolites be identified and quantified by different mass spectrometric techniques. This is the first study showing that the human colonic microbiota is able to degrade selected glycated amino acids and possibly use them as a source of energy, carbon, and/or nitrogen.


Assuntos
Bactérias/metabolismo , Colo/microbiologia , Microbioma Gastrointestinal , Lisina/análogos & derivados , Norleucina/análogos & derivados , Piridonas/química , Pirróis/química , Adulto , Colo/metabolismo , Feminino , Humanos , Lisina/química , Lisina/metabolismo , Reação de Maillard , Masculino , Pessoa de Meia-Idade , Norleucina/química , Norleucina/metabolismo , Piridonas/metabolismo , Pirróis/metabolismo
20.
J Pept Sci ; 21(1): 17-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25420700

RESUMO

Cleavage reactions at backbone loci are one of the consequences of oxidation of proteins and peptides. During α-amidation, the Cα -N bond in the backbone is cleaved under formation of an N-terminal peptide amide and a C-terminal keto acyl peptide. On the basis of earlier works, a facilitation of α-amidation by the thioether group of adjacent methionine side chains was proposed. This reaction was characterized by using benzoyl methionine and benzoyl alanyl methionine as peptide models. The decomposition of benzoylated amino acids (benzoyl-methionine, benzoyl-alanine, and benzoyl-methionine sulfoxide) to benzamide in the presence of different carbohydrate compounds (reducing sugars, Amadori products, and reductones) was studied during incubation for up to 48 h at 80 °C in acetate-buffered solution (pH 6.0). Small amounts of benzamide (0.3-1.5 mol%) were formed in the presence of all sugars and from all benzoylated species. However, benzamide formation was strongly enhanced, when benzoyl methionine was incubated in the presence of reductones and Amadori compounds (3.5-4.2 mol%). The reaction was found to be intramolecular, because α-amidation of a similar 4-methylbenzoylated amino acid was not enhanced in the presence of benzoyl-methionine and carbohydrate compounds. In the peptide benzoyl-alanyl-methionine, α-amidation at the methionine residue is preferred over α-amidation at the benzoyl peptide bond. We propose here a mechanism for the enhancement of α-amidation at methionine residues.


Assuntos
Alanina/análogos & derivados , Alanina/química , Metionina/análogos & derivados , Metionina/química , Amidas/química , Benzamidas/química , Cinética , Modelos Químicos , Oxirredução , Proteólise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA