Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 295(17): 5701-5716, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32184356

ABSTRACT

Lens proteins become increasingly cross-linked through nondisulfide linkages during aging and cataract formation. One mechanism that has been implicated in this cross-linking is glycation through formation of advanced glycation end products (AGEs). Here, we found an age-associated increase in stiffness in human lenses that was directly correlated with levels of protein-cross-linking AGEs. α-Crystallin in the lens binds to other proteins and prevents their denaturation and aggregation through its chaperone-like activity. Using a FRET-based assay, we examined the stability of the αA-crystallin-γD-crystallin complex for up to 12 days and observed that this complex is stable in PBS and upon incubation with human lens-epithelial cell lysate or lens homogenate. Addition of 2 mm ATP to the lysate or homogenate did not decrease the stability of the complex. We also generated complexes of human αA-crystallin or αB-crystallin with alcohol dehydrogenase or citrate synthase by applying thermal stress. Upon glycation under physiological conditions, the chaperone-client complexes underwent greater extents of cross-linking than did uncomplexed protein mixtures. LC-MS/MS analyses revealed that the levels of cross-linking AGEs were significantly higher in the glycated chaperone-client complexes than in glycated but uncomplexed protein mixtures. Mouse lenses subjected to thermal stress followed by glycation lost resilience more extensively than lenses subjected to thermal stress or glycation alone, and this loss was accompanied by higher protein cross-linking and higher cross-linking AGE levels. These results uncover a protein cross-linking mechanism in the lens and suggest that AGE-mediated cross-linking of α-crystallin-client complexes could contribute to lens aging and presbyopia.


Subject(s)
Aging , Lens, Crystalline/metabolism , Presbyopia/metabolism , alpha-Crystallin A Chain/metabolism , Adolescent , Adult , Aged , Glycation End Products, Advanced/analysis , Glycation End Products, Advanced/metabolism , Glycosylation , Humans , Lens, Crystalline/chemistry , Middle Aged , Protein Denaturation , Young Adult , alpha-Crystallin A Chain/chemistry , gamma-Crystallins/chemistry , gamma-Crystallins/metabolism
2.
Exp Eye Res ; 210: 108704, 2021 09.
Article in English | MEDLINE | ID: mdl-34302851

ABSTRACT

Advanced glycation end products (AGEs) accumulate with age in human lens capsules. AGEs in lens capsules potentiate the transforming growth factor beta-2-mediated mesenchymal transition of lens epithelial cells, which suggests that they play a role in posterior capsule opacification after cataract surgery. We measured AGEs by liquid chromatography-mass spectrometry in capsulorhexis specimens obtained during cataract surgery from nondiabetic and diabetic patients with and without established retinopathy. Our data showed that the levels of most AGEs (12 out of 13 measured) were unaltered in diabetic patients and diabetic patients with retinopathy compared to nondiabetic patients. There was one exception: glucosepane, which was significantly higher in diabetic patients, both with (6.85 pmol/µmol OH-proline) and without retinopathy (8.32 pmol/µmol OH-proline), than in nondiabetic patients (4.01 pmol/µmol OH-proline). Our study provides an explanation for the similar incidence of posterior capsule opacification between nondiabetic and diabetic cataract patients observed in several studies.


Subject(s)
Cataract/metabolism , Diabetic Retinopathy/metabolism , Glycation End Products, Advanced/metabolism , Lens Capsule, Crystalline/metabolism , Aged , Blood Glucose/metabolism , Capsulorhexis , Cataract/pathology , Chromatography, Liquid , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Diabetic Retinopathy/pathology , Female , Glycated Hemoglobin/metabolism , Humans , Lens Capsule, Crystalline/pathology , Male , Middle Aged , Tandem Mass Spectrometry
3.
Glycoconj J ; 38(3): 347-359, 2021 06.
Article in English | MEDLINE | ID: mdl-33245448

ABSTRACT

Proteins in the eye lens have negligible turnover and therefore progressively accumulate chemical modifications during aging. Carbonyls and oxidative stresses, which are intricately linked to one another, predominantly drive such modifications. Oxidative stress leads to the loss of glutathione (GSH) and ascorbate degradation; this in turn leads to the formation of highly reactive dicarbonyl compounds that react with proteins to form advanced glycation end products (AGEs). The formation of AGEs leads to the crosslinking and aggregation of proteins contributing to lens aging and cataract formation. To inhibit AGE formation, we developed a disulfide compound linking GSH diester and mercaptoethylguanidine, and we named it carboxitin. Bovine lens organ cultured with carboxitin showed higher levels of GSH and mercaptoethylguanidine in the lens nucleus. Carboxitin inhibited erythrulose-mediated mouse lens protein crosslinking, AGE formation and the formation of 3-deoxythreosone, a major ascorbate-derived AGE precursor in the human lens. Carboxitin inhibited the glycation-mediated increase in stiffness in organ-cultured mouse lenses measured using compressive mechanical strain. Delivery of carboxitin into the lens increases GSH levels, traps dicarbonyl compounds and inhibits AGE formation. These properties of carboxitin could be exploited to develop a therapy against the formation of AGEs and the increase in stiffness that causes presbyopia in aging lenses.


Subject(s)
Glutathione/analogs & derivatives , Glutathione/chemical synthesis , Lens, Crystalline/drug effects , Animals , Cattle , Glycation End Products, Advanced , Glycosylation , Lens, Crystalline/physiology , Mice , Mice, Inbred C57BL , Protein Binding , Tetroses/metabolism , Tumor Cells, Cultured
4.
Biochem Biophys Res Commun ; 533(4): 1352-1358, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33081971

ABSTRACT

The chaperone activity of α-crystallin is important for maintaining the transparency of the human lens. αB-crystallin (αBC) is a long-lived protein in the lens that accumulates chemical modifications during aging. The formation of advanced glycation end products (AGEs) through glycation is one such modification. αBC is a small heat shock protein that exhibits chaperone activity. We have previously shown that αBC-client protein complexes can undergo AGE-mediated interprotein cross-linking. Here, we demonstrate that short-term (1 h) exposure to elevated temperatures and methylglyoxal (MGO) during the chaperoning of client proteins by αBC promotes AGE-mediated interprotein cross-linking. Liquid chromatography/mass spectrometry (LC-MS/MS) analyses revealed the rapid formation of AGEs by MGO. Interestingly, we found that despite protein cross-linking, the chaperone activity of αBC increased during the transient elevation of temperature in the presence of MGO. Together, these results imply that transient and subtle elevation of temperature in the lens of the eye can promote protein cross-linking through AGEs, and if this phenomenon recurs over a period of many years, it could lead to early onset of presbyopia and age-related cataracts.


Subject(s)
Glycation End Products, Advanced/chemistry , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/metabolism , Arginine/analogs & derivatives , Arginine/chemistry , Arginine/metabolism , Cataract/metabolism , Citrate (si)-Synthase/chemistry , Citrate (si)-Synthase/metabolism , Cross-Linking Reagents/chemistry , Glycation End Products, Advanced/metabolism , Humans , Malate Dehydrogenase/chemistry , Malate Dehydrogenase/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Presbyopia/metabolism , Pyruvaldehyde/chemistry , Pyruvaldehyde/metabolism , Temperature , alpha-Crystallin B Chain/genetics
5.
Int J Mol Sci ; 21(2)2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31952342

ABSTRACT

Protein glycation is usually referred to as an array of non-enzymatic post-translational modifications formed by reducing sugars and carbonyl products of their degradation. The resulting advanced glycation end products (AGEs) represent a heterogeneous group of covalent adducts, known for their pro-inflammatory effects in mammals, and impacting on pathogenesis of metabolic diseases and ageing. In plants, AGEs are the markers of tissue ageing and response to environmental stressors, the most prominent of which is drought. Although water deficit enhances protein glycation in leaves, its effect on seed glycation profiles is still unknown. Moreover, the effect of drought on biological activities of seed protein in mammalian systems is still unstudied with respect to glycation. Therefore, here we address the effects of a short-term drought on the patterns of seed protein-bound AGEs and accompanying alterations in pro-inflammatory properties of seed protein in the context of seed metabolome dynamics. A short-term drought, simulated as polyethylene glycol-induced osmotic stress and applied at the stage of seed filling, resulted in the dramatic suppression of primary seed metabolism, although the secondary metabolome was minimally affected. This was accompanied with significant suppression of NF-kB activation in human SH-SY5Y neuroblastoma cells after a treatment with protein hydrolyzates, isolated from the mature seeds of drought-treated plants. This effect could not be attributed to formation of known AGEs. Most likely, the prospective anti-inflammatory effect of short-term drought is related to antioxidant effect of unknown secondary metabolite protein adducts, or down-regulation of unknown plant-specific AGEs due to suppression of energy metabolism during seed filling.


Subject(s)
Droughts , Metabolomics/methods , Pisum sativum/metabolism , Plant Proteins/metabolism , Protein Processing, Post-Translational , Seeds/metabolism , Antioxidants/metabolism , Cell Line, Tumor , Energy Metabolism , Gas Chromatography-Mass Spectrometry , Glycation End Products, Advanced/metabolism , Glycosylation , Humans , NF-kappa B/metabolism , Stress, Physiological
6.
Anal Chem ; 91(19): 12336-12343, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31503451

ABSTRACT

Recently discovered acylation by reactive acyl-CoA species is considered a novel regulatory mechanism in epigenetics and metabolism. Established analytical methods like Western blotting and proteomics fail to detect the plethora of acylation structures in a single analysis and lack the ability of absolute quantitation. In this paper, we developed an HPLC-MS/MS method for the simultaneous detection and quantitation of 14 acylated lysine species in biological samples. Extensive effort was invested into method validation resulting in recovery rates between 75 and 93% and levels of detection in the nanomolar range. Thus, we were able to quantitate 8 acylation structures in mouse liver, kidney, heart, and brain. Further enrichment by repetitive HPLC fractionation resulted in the quantitation of 6 additional acylation structures including 4 novel modifications: N6-acetoacetyl lysine, N6-isovaleryl lysine, N6-(2-methylbutyryl) lysine, and N6-tiglyl lysine.


Subject(s)
Acyl Coenzyme A/metabolism , Chromatography, High Pressure Liquid/methods , Proteins/metabolism , Tandem Mass Spectrometry/methods , Acylation , Animals , Hydrolysis , Lysine/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Proteins/chemistry
7.
Int J Mol Sci ; 20(24)2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31817246

ABSTRACT

Aging represents the accumulation of changes in an individual over time, encompassing physical, psychological, and social changes. Posttranslational modifications of proteins such as glycosylation, including sialylation or glycation, are proposed to be involved in this process, since they modulate a variety of molecular and cellular functions. In this study, we analyzed selected posttranslational modifications and the respective proteins on which they occur in young and old mouse brains. The expression of neural cell adhesion molecule (NCAM), receptor for advanced glycation endproducts (RAGE), as well as the carbohydrate-epitopes paucimannose and high-mannose, polysialic acid, and O-GlcNAc were examined. We demonstrated that mannose-containing glycans increased on glycoproteins in aged mouse brains and identified synapsin-1 as one major carrier of paucimannose in aged brains. In addition, we found an accumulation of so-called advanced glycation endproducts, which are generated by non-enzymatic reactions and interfere with protein function. Furthermore, we analyzed the expression of sialic acid and found also an increase during aging.


Subject(s)
Aging , Brain/metabolism , Glycoproteins/metabolism , N-Acetylneuraminic Acid/metabolism , Animals , Chromatography, High Pressure Liquid , Glycation End Products, Advanced/metabolism , Glycoproteins/analysis , Glycosylation , Male , Mannose/chemistry , Mannose/metabolism , Mass Spectrometry , Mice , N-Acetylneuraminic Acid/analysis , Neural Cell Adhesion Molecules/metabolism , Receptor for Advanced Glycation End Products/metabolism
8.
Glycoconj J ; 33(4): 499-512, 2016 08.
Article in English | MEDLINE | ID: mdl-27291759

ABSTRACT

Initially investigated as a color formation process in thermally treated foods, nowadays, the relevance of the Maillard reaction in vivo is generally accepted. Many chronic and age-related diseases such as diabetes, uremia, atherosclerosis, cataractogenesis and Alzheimer's disease are associated with Maillard derived advanced glycation endproducts (AGEs) and α-dicarbonyl compounds as their most important precursors in terms of reactivity and abundance. However, the situation in vivo is very challenging, because Maillard chemistry is paralleled by enzymatic reactions which can lead to both, increases and decreases in certain AGEs. In addition, mechanistic findings established under the harsh conditions of food processing might not be valid under physiological conditions. The present review critically discusses the relevant α-dicarbonyl compounds as central intermediates of AGE formation in vivo with a special focus on fragmentation pathways leading to formation of amide-AGEs.


Subject(s)
Glycation End Products, Advanced/metabolism , Maillard Reaction , Alzheimer Disease/metabolism , Animals , Atherosclerosis/metabolism , Cataract/metabolism , Diabetes Mellitus/metabolism , Humans , Uremia/metabolism
9.
Biochemistry ; 54(15): 2500-7, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25849437

ABSTRACT

In human lens proteins, advanced glycation endproducts (AGEs) originate from the reaction of glycating agents, e.g., vitamin C and glucose. AGEs have been considered to play a significant role in lens aging and cataract formation. Although several AGEs have been detected in the human lens, the contribution of individual glycating agents to their formation remains unclear. A highly sensitive liquid chromatography-tandem mass spectrometry multimethod was developed that allowed us to quantitate 21 protein modifications in normal and cataractous lenses, respectively. N(6)-Carboxymethyl lysine, N(6)-carboxyethyl lysine, N(7)-carboxyethyl arginine, methylglyoxal hydroimidazolone 1, and N(6)-lactoyl lysine were found to be the major Maillard protein modifications among these AGEs. The novel vitamin C specific amide AGEs, N(6)-xylonyl and N(6)-lyxonyl lysine, but also AGEs from glyoxal were detected, albeit in minor quantities. Among the 21 modifications, AGEs from the Amadori product (derived from the reaction of glucose and lysine) and methylglyoxal were dominant.


Subject(s)
Aging/metabolism , Cataract/metabolism , Eye Proteins/metabolism , Glycation End Products, Advanced/metabolism , Maillard Reaction , Protein Processing, Post-Translational , Adult , Aged , Aging/pathology , Cataract/pathology , Child , Female , Humans , Middle Aged
10.
J Biol Chem ; 289(41): 28676-88, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25164824

ABSTRACT

Maillard α-dicarbonyl compounds are known as central intermediates in advanced glycation end product (AGE) formation. Glucose is the primary source of energy for the human body, whereas l-threo-ascorbic acid (vitamin C) is an essential nutrient, involved in a variety of enzymatic reactions. Thus, the Maillard degradation of glucose and ascorbic acid is of major importance in vivo. To understand the complex mechanistic pathways of AGE formation, it is crucial to extend the knowledge on plasma concentrations of reactive key α-dicarbonyl compounds (e.g. 1-deoxyglucosone). With the present work, we introduce a highly sensitive LC-MS/MS multimethod for human blood plasma based on derivatization with o-phenylenediamine under acidic conditions. The impact of workup and reaction conditions, particularly of pH, was thoroughly evaluated. A comprehensive validation provided the limit of detection, limit of quantitation, coefficients of variation, and recovery rates. The method includes the α-dicarbonyls 1-deoxyglucosone, 3-deoxyglucosone, glucosone, Lederer's glucosone, dehydroascorbic acid, 2,3-diketogulonic acid, 1-deoxypentosone, 3-deoxypentosone, 3,4-dideoxypentosone, pentosone, 1-deoxythreosone, 3-deoxythreosone, threosone, methylglyoxal, glyoxal; the α-keto-carboxylic acids pyruvic acid and glyoxylic acid; and the dicarboxylic acid oxalic acid. The method was then applied to the analyses of 15 healthy subjects and 24 uremic patients undergoing hemodialysis. The comparison of the results revealed a clear shift in the product spectrum. In most cases, the plasma levels of target analytes were significantly higher. Thus, this is the first time that a complete spectrum of α-dicarbonyl compounds relevant in vivo has been established. The results provide further insights into the chemistry of AGE formation and will be helpful to find specific markers to differentiate between the various precursors of glycation.


Subject(s)
Aldehydes/blood , Carboxylic Acids/blood , Glycation End Products, Advanced/blood , Ketoses/blood , Metabolomics/methods , Uremia/blood , Adult , Aged , Aged, 80 and over , Case-Control Studies , Chromatography, Liquid/methods , Female , Humans , Hydrogen-Ion Concentration , Limit of Detection , Maillard Reaction , Male , Metabolomics/instrumentation , Middle Aged , Phenylenediamines/chemistry , Renal Dialysis , Reproducibility of Results , Tandem Mass Spectrometry/methods , Uremia/physiopathology , Uremia/therapy
11.
Eur J Clin Invest ; 45(12): 1333-40, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26519693

ABSTRACT

BACKGROUND: Haemodialysis patients suffer from chronic systemic inflammation and high incidence of cardiovascular disease. One cause for this may be the failure of diseased kidneys to eliminate immune mediators. Current haemodialysis treatment achieves insufficient elimination of proteins in the molecular weight range 15-45 kD. Thus, high cut-off dialysis might improve the inflammatory state. DESIGN: In this randomized crossover trial, 43 haemodialysis patients were treated for 3 weeks with high cut-off or high-flux dialysis. Inflammatory plasma mediators, monocyte subpopulation distribution and leucocyte gene expression were quantified. RESULTS: High cut-off dialysis supplemented by a low-flux filter did not influence the primary end-point, expression density of CD162 on monocytes. Nevertheless, treatment reduced multiple immune mediators in plasma. Such reduction proved - at least for some markers - to be a sustained effect over the interdialytic interval. Thus, for example, soluble TNF-receptor 1 concentration predialysis was reduced from median 13·3 (IQR 8·9-17·2) to 9·7 (IQR 7·5-13·2) ng/mL with high cut-off while remaining constant with high-flux treatment. The expression profile of multiple proinflammatory genes in leucocytes was significantly dampened. Treatment was well tolerated although albumin losses in high cut-off dialysis would be prohibitive against long-term use. CONCLUSIONS: The study shows for the first time that a dampening effect of high cut-off dialysis on systemic inflammation is achievable. Earlier studies had failed due to short study duration or insufficient dialysis efficacy. Removal of soluble mediators from the circulation influences cellular activation levels in leucocytes. Continued development of less albumin leaky membranes with similar cytokine elimination is justified.


Subject(s)
Kidney Failure, Chronic/therapy , Renal Dialysis/methods , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Cross-Over Studies , Humans , Male , Middle Aged , Muscle Cells/metabolism , Patient Safety , Prospective Studies , Treatment Outcome , Young Adult
12.
Reproduction ; 148(2): 169-78, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24821834

ABSTRACT

Diabetes mellitus (DM) during pregnancy is one of the leading causes of perinatal morbidity and birth defects. The mechanism by which maternal hyperglycemia, the major teratogenic factor, induces embryonic malformations remains unclear. Advanced glycation end products (AGEs) are known to accumulate during the course of DM and contribute to the development of diabetic complications. Employing a diabetic rabbit model, we investigated the influence of maternal hyperglycemia during the preimplantation period on AGE formation (pentosidine, argpyrimidine, and N(ϵ)-carboxymethyllysine (CML)) in the reproductive tract and the embryo itself. As a consequence of type 1 DM, the AGE levels in blood plasma increased up to 50%, correlating closely with an AGE accumulation in the endometrium of diabetic females. Embryos from diabetic mothers had increased protein-bound CML levels and showed enhanced fluorescent signals for AGE-specific fluorescence in the blastocyst cavity fluid (BCF). The quantification of CML by HPLC-mass spectrometry (MS/MS) showed a higher amount of soluble CML in the BCF of blastocysts from diabetic rabbits (0.26±0.05 µmol/l) compared with controls (0.18±0.02 µmol/l). The high amount of AGEs in blastocysts from diabetic mothers correlates positively with an increased AGER (receptor for AGE (RAGE)) mRNA expression. Our study gives alarming insights into the consequences of poorly controlled maternal diabetes for AGE formation in the embryo. Maternal hyperglycemia during the preimplantation period is correlated with an increase in AGE formation in the uterine environment and the embryo itself. This may influence the development of the embryo through increased AGE-mediated cellular stress by RAGEs.


Subject(s)
Blastocyst/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetes, Gestational/metabolism , Glycation End Products, Advanced/metabolism , Hyperglycemia/complications , Animals , Blastocyst/pathology , Cells, Cultured , Chromatography, High Pressure Liquid , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/pathology , Diabetes, Gestational/pathology , Female , Glycation End Products, Advanced/genetics , Hyperglycemia/physiopathology , Immunoenzyme Techniques , Male , Pregnancy , RNA, Messenger/genetics , Rabbits , Real-Time Polymerase Chain Reaction , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry
13.
Macromol Biosci ; 23(3): e2200487, 2023 03.
Article in English | MEDLINE | ID: mdl-36543753

ABSTRACT

The changes of technological properties of albumin-based hydrogels induced by increasing degrees of post-translational modification of the protein are reported. Maillard-type modification of amino acids arginine and lysine of albumin is achieved through glyoxal as an α-dicarbonyl compound. The degrees of modification are fine-tuned using different molar ratios of glyoxal. Hydrogels are thermally induced by heating highly concentrated precursor solutions above the protein's denaturation temperature. While the post-translational modifications are determined and quantified with mass spectrometry, continuous-wave (CW) electron paramagnetic resonance (EPR) spectroscopy shed light on the protein fatty acid binding capacity and changes thereof in solution and in the gel state. The viscoelastic behavior is characterized as a measure of the physical strength of the hydrogels. On the nanoscopic level, the modified albumins in low concentration solution reveal lower binding capacities with increasing degrees of modification. On the contrary, in the gel state, the binding capacity remains constant at all degrees of modifications. This indicates that the loss of fatty acid binding capacity for individual albumin molecules is partially compensated by new binding sites in the gel state, potentially formed by modified amino acids. Such, albumin glycation offers a fine-tuning method of technological and nanoscopic properties of these gels.


Subject(s)
Serum Albumin, Human , Serum Albumin , Humans , Serum Albumin/chemistry , Serum Albumin/metabolism , Maillard Reaction , Hydrogels , Glyoxal/chemistry , Lysine , Fatty Acids/chemistry
14.
J Biol Chem ; 286(52): 44350-6, 2011 Dec 30.
Article in English | MEDLINE | ID: mdl-22069309

ABSTRACT

The Maillard reaction in vivo entails alteration of proteins or free amino acids by non-enzymatic glycation or glycoxidation. The resulting modifications are called advanced glycation end products (AGEs) and play a prominent role in various pathologies, including normoglycemic uremia. Recently, we established a new class of lysine amide modifications in vitro. Now, human plasma levels of the novel amide-AGEs N(6)-acetyl lysine, N(6)-formyl lysine, N(6)-lactoyl lysine, and N(6)-glycerinyl lysine were determined by means of LC-MS/MS. They were significantly higher in uremic patients undergoing hemodialysis than in healthy subjects. Model reactions with N(1)-t-butoxycarbonyl-lysine under physiological conditions confirmed 1-deoxy-d-erythro-hexo-2,3-diulose as an immediate precursor. Because formation of N(6)-formyl lysine from glucose responded considerably to the presence of oxygen, glucosone was identified as another precursor. Comparison of the in vivo results with the model experiments enabled us to elucidate possible formation pathways linked to Maillard chemistry. The results strongly suggest a major participation of non-enzymatic Maillard mechanisms on amide-AGE formation pathways in vivo, which, in the case of N(6)-acetyl lysine, parallels enzymatic processes.


Subject(s)
Amides/blood , Glycation End Products, Advanced/blood , Lysine/analogs & derivatives , Lysine/blood , Maillard Reaction , Uremia/blood , Aged , Aged, 80 and over , Amides/analysis , Amides/chemistry , Female , Glucose/metabolism , Glycation End Products, Advanced/analysis , Glycation End Products, Advanced/chemistry , Humans , Lysine/analysis , Lysine/chemistry , Male , Middle Aged , Renal Dialysis , Uremia/therapy
15.
Anal Bioanal Chem ; 403(10): 2923-31, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22382856

ABSTRACT

High-fructose corn syrup (HFCS) is a widely used liquid sweetener produced from corn starch by hydrolysis and partial isomerization of glucose to fructose. During these processing steps, sugars can be considerably degraded, leading, for example, to the formation of reactive α-dicarbonyl compounds (α-DCs). The present study performed targeted screening to identify the major α-DCs in HFCS. For this purpose, α-DCs were selectively converted with o-phenylendiamine to the corresponding quinoxaline derivatives, which were analyzed by liquid chromatography with hyphenated diode array-tandem mass spectrometry (LC-DAD-MS/MS) detection. 3-Deoxy-D-erythro-hexos-2-ulose (3-deoxyglucosone), D-lyxo-hexos-2-ulose (glucosone), 3-deoxy-D-threo-hexos-2-ulose (3-deoxygalactosone), 1-deoxy-D-erythro-hexos-2,3-diulose (1-deoxyglucosone), 3,4-dideoxyglucosone-3-ene, methylglyoxal, and glyoxal were identified by enhanced mass spectra as well as MS/MS product ion spectra using the synthesized standards as reference. Addition of diethylene triamine pentaacetic acid and adjustment of the derivatization conditions ensured complete derivatization without de novo formation for all identified α-DCs in HFCS matrix except for glyoxal. Subsequently, a ultra-high performance LC-DAD-MS/MS method was established to quantify 3-deoxyglucosone, glucosone, 3-deoxygalactosone, 1-deoxyglucosone, 3,4-dideoxyglucosone-3-ene, and methylglyoxal in HFCS. Depending on the α-DC compound and concentration, the recovery ranged between 89.2% and 105.8% with a relative standard deviation between 1.9% and 6.5%. Subsequently, the α-DC profiles of 14 commercial HFCS samples were recorded. 3-Deoxyglucosone was identified as the major α-DC with concentrations up to 730 µg/mL HFCS. The total α-DC content ranged from 293 µg/mL to 1,130 µg/mL HFCS. Significantly different α-DC levels were not detected between different HFCS specifications, but between samples of various manufacturers indicating that the α-DC load is influenced by the production procedures.


Subject(s)
Food Analysis , Food Contamination/analysis , Fructose/chemistry , Glyoxal/analysis , Ketoses/analysis , Sweetening Agents/chemistry , Zea mays/chemistry , Chromatography, Liquid , Molecular Structure , Tandem Mass Spectrometry
16.
J Agric Food Chem ; 70(14): 4434-4444, 2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35348319

ABSTRACT

Short-chained α-hydroxycarbonyl compounds such as glycolaldehyde (GA) and its oxidized counterpart glyoxal (GX) are known as potent glycating agents. Here, a novel fluorescent lysine-lysine cross-link 1-(5-amino-5-carboxypentyl)-3-(5-amino-5-carboxy-pentylamino)pyridinium salt (meta-DLP) was synthesized and its structure unequivocally proven by 1H NMR, 13C-NMR attached proton test, and 2D NMR. Further characterization of chemical properties and mechanistic background was obtained in comparison to the known monovalent protein modification 2-ammonio-6-(3-oxidopyridinium-1-yl)hexanoate (OP-lysine). Identification and quantitation in various sugar incubations with N2-t-Boc-lysine revealed a novel alternative formation pathway for both advanced glycation end products (AGEs) by the interplay of both carbonyl compounds, GA and GX, which was confirmed by isotope labeling experiments. The concentration of pyridinium AGEs was about 1000-fold lower compared to the well-established N6-carboxymethyl lysine. However, pyridinium AGEs were shown to lead to the photosensitized generation of singlet oxygen in irradiation experiments, which was verified by the detection of 3,3'-(naphthalene-1,4-diyl)-dipropionate endoperoxide. Furthermore, meta-DLP was identified in hydrolyzed potato chip proteins by collision-induced dissociation mass spectrometry after HPLC enrichment.


Subject(s)
Glyoxal , Lysine , Acetaldehyde/analogs & derivatives , Glycation End Products, Advanced/chemistry , Glyoxal/chemistry , Lysine/chemistry , Maillard Reaction
17.
J Agric Food Chem ; 70(33): 10271-10283, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-35968682

ABSTRACT

In the present work, the contribution of lipid peroxidation on modifications of lysine and arginine residues of proteins was investigated. Lipid peroxidation had a major impact on malondialdehyde-derived protein modifications; however, the influence on glyoxal and methylglyoxal-derived modifications in flat wafers was negligible. Therefore, vegetable oils (either linseed oil, sunflower oil, or coconut oil) were added to respective batters, and flat wafers were baked (150 °C, 3-10 min). Analysis of malondialdehyde indicated oxidation in linseed wafers, which was supported by the direct quantitation of three malondialdehyde protein adducts in the range of 0.09-23.5 mg/kg after enzymatic hydrolysis. In contrast, levels of free glyoxal and methylglyoxal were independent of the type of oil added, which was in line with the analysis of 13 advanced glycation end products. Comprehensive incubations of 40 mM N2-t-Boc-lysine (100 mM phosphate buffer, pH 7.4) with either 10% oil or an equimolar concentration of carbohydrates led to magnitudes higher (103-105) amounts of N6-carboxymethyl lysine, N6-glycolyl lysine, and N6-carboxyethyl lysine in the latter. Furthermore, malondialdehyde exceeded glyoxal and methylglyoxal in incubations of pure oils at 150 °C by factors of 30 and 100, respectively.


Subject(s)
Glyoxal , Pyruvaldehyde , Carbohydrates , Glycation End Products, Advanced/chemistry , Lipid Peroxidation , Lysine/chemistry , Malondialdehyde , Proteins/metabolism , Pyruvaldehyde/chemistry
18.
J Agric Food Chem ; 70(9): 3033-3046, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35194998

ABSTRACT

Glycation significantly alters the physicochemical and biofunctional properties of proteins in foods and in vivo. In the present study, human serum albumin (HSA) as the major transporter of fatty acids was modified with glyoxal under physiological conditions. Reversibly albumin-bound glyoxal was removed, and advanced glycation end products were quantitated by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The total modification of protein-bound lysine and arginine residues reached up to 4.2 and 9.6%, respectively. The impact of these modifications on the transport capacity of long-chain fatty acids was characterized by spin-labeled fatty acid probes via electron paramagnetic resonance spectroscopy. With increasing degree of glycation, the equivalence of the seven binding sites of native HSA with a dissociation constant of 0.74 ± 0.09 µM was set off with only the three high-affinity sites 2, 4, and 5 remaining (0.46 ± 0.07 µM). The other four sites were shifted to low affinities with significantly higher dissociation constants (1.32 ± 0.35 µM). Tryptic peptide mapping enabled us to relate these findings to molecular changes at specific binding sites. Modification hotspots identified were lysine 351, 286, 159 and arginine 144, 485, 117. Further investigation of plasma protein samples of uremic patients vs healthy controls gave first insights into the in vivo situation.


Subject(s)
Serum Albumin, Human , Tandem Mass Spectrometry , Chromatography, Liquid , Fatty Acids , Glycation End Products, Advanced/chemistry , Glycosylation , Humans , Serum Albumin, Human/metabolism , Tandem Mass Spectrometry/methods
19.
Biochim Biophys Acta ; 1804(4): 829-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20026434

ABSTRACT

Human lens proteins (HLP) become chemically modified by kynurenines and advanced glycation end products (AGEs) during aging and cataractogenesis. We investigated the effects of kynurenines on AGE synthesis in HLP. We found that incubation with 5 mM ribose or 5 mM ascorbate produced significant quantities of pentosidine, and this was further enhanced in the presence of two different kynurenines (200-500 microM): N-formylkynurenine (Nfk) and kynurenine (Kyn). Another related compound, 3-hydroxykynurenine (3OH-Kyn), had disparate effects; low concentrations (10-200 microM) promoted pentosidine synthesis, but high concentrations (200-500 microM) inhibited it. 3OH-Kyn showed similar effects on pentosidine synthesis from Amadori-enriched HLP or ribated lysine. Chelex-100 treatment of phosphate buffer reduced pentosidine synthesis from Amadori-enriched HLP by approximately 90%, but it did not inhibit the stimulating effect of 3OH-Kyn and EDTA. 3OH-Kyn (100-500 microM) spontaneously produced copious amounts of H(2)O(2) (10-25 microM), but externally added H(2)O(2) had only a mild stimulating effect on pentosidine but had no effect on N(epsilon)-carboxymethyl lysine (CML) synthesis in HLP from ribose and ascorbate. Further, human lens epithelial cells incubated with ribose and 3OH-Kyn showed higher intracellular pentosidine than cells incubated with ribose alone. CML synthesis from glycating agents was inhibited 30 to 50% by 3OH-Kyn at concentrations of 100-500 microM. Argpyrimidine synthesis from 5mM methylglyoxal was slightly inhibited by all kynurenines at concentrations of 100-500 microM. These results suggest that AGE synthesis in HLP is modulated by kynurenines, and such effects indicate a mode of interplay between kynurenines and carbohydrates important for AGE formation during lens aging and cataract formation.


Subject(s)
Crystallins/metabolism , Glycation End Products, Advanced/biosynthesis , Kynurenine/pharmacology , Arginine/analogs & derivatives , Arginine/biosynthesis , Arginine/chemistry , Buffers , Cell Line , Crystallins/chemistry , Edetic Acid , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glycation End Products, Advanced/chemistry , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , In Vitro Techniques , Kynurenine/analogs & derivatives , Kynurenine/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/drug effects , Lens, Crystalline/metabolism , Lysine/analogs & derivatives , Lysine/biosynthesis , Lysine/chemistry , Maillard Reaction , Models, Biological , Resins, Synthetic
20.
Biochim Biophys Acta ; 1802(4): 432-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20085807

ABSTRACT

AlphaA-crystallin is a molecular chaperone; it prevents aggregation of denaturing proteins. We have previously demonstrated that upon modification by a metabolic alpha-dicarbonyl compound, methylglyoxal (MGO), alphaA-crystallin becomes a better chaperone. AlphaA-crystallin also assists in refolding of denatured proteins. Here, we have investigated the effect of mild modification of alphaA-crystallin by MGO (with 20-500 microM) on the chaperone function and its ability to refold denatured proteins. Under the conditions used, mildly modified protein contained mostly hydroimidazolone modifications. The modified protein exhibited an increase in chaperone function against thermal aggregation of beta(L)- and gamma-crystallins, citrate synthase (CS), malate dehydrogenase (MDH) and lactate dehydrogenase (LDH) and chemical aggregation of insulin. The ability of the protein to assist in refolding of chemically denatured beta(L)- and gamma-crystallins, MDH and LDH, and to prevent thermal inactivation of CS were unchanged after mild modification by MGO. Prior binding of catalytically inactive, thermally denatured MDH or the hydrophobic probe, 2-p-toluidonaphthalene-6-sulfonate (TNS) abolished the ability of alphaA-crystallin to assist in the refolding of denatured MDH. However, MGO modification of chaperone-null TNS-bound alphaA-crystallin resulted in partial regain of the chaperone function. Taken together, these results demonstrate that: 1) hydroimidazolone modifications are sufficient to enhance the chaperone function of alphaA-crystallin but such modifications do not change its ability to assist in refolding of denatured proteins, 2) the sites on the alphaA-crystallin responsible for the chaperone function and refolding are the same in the native alphaA-crystallin and 3) additional hydrophobic sites exposed upon MGO modification, which are responsible for the enhanced chaperone function, do not enhance alphaA-crystallin's ability to refold denatured proteins.


Subject(s)
Crystallins/chemistry , Imidazoles/chemistry , Molecular Chaperones/chemistry , Protein Folding , Crystallins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Chaperones/metabolism , Protein Denaturation , Protein Structure, Tertiary/physiology
SELECTION OF CITATIONS
SEARCH DETAIL