Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(7): e0235335, 2020.
Article in English | MEDLINE | ID: mdl-32628695

ABSTRACT

Diabetes is associated with a dramatic mortality rate due to its vascular complications. Chronic hyperglycemia in diabetes leads to enhanced glycation of erythrocytes and oxidative stress. Even though erythrocytes play a determining role in vascular complications, very little is known about how erythrocyte structure and functionality can be affected by glycation. Our objective was to decipher the impact of glycation on erythrocyte structure, oxidative stress parameters and capacity to interact with cultured human endothelial cells. In vitro glycated erythrocytes were prepared following incubation in the presence of different concentrations of glucose. To get insight into the in vivo relevance of our results, we compared these data to those obtained using red blood cells purified from diabetics or non-diabetics. We measured erythrocyte deformability, susceptibility to hemolysis, reactive oxygen species production and oxidative damage accumulation. Altered structures, redox status and oxidative modifications were increased in glycated erythrocytes. These modifications were associated with reduced antioxidant defence mediated by enzymatic activity. Enhanced erythrocyte phagocytosis by endothelial cells was observed when cultured with glycated erythrocytes, which was associated with increased levels of phosphatidylserine-likely as a result of an eryptosis phenomenon triggered by the hyperglycemic treatment. Most types of oxidative damage identified in in vitro glycated erythrocytes were also observed in red blood cells isolated from diabetics. These results bring new insights into the impact of glycation on erythrocyte structure, oxidative damage and their capacity to interact with endothelial cells, with a possible relevance to diabetes.


Subject(s)
Diabetes Mellitus, Type 2/blood , Erythrocytes/pathology , Glycation End Products, Advanced/metabolism , Reactive Oxygen Species/metabolism , Blood Glucose/metabolism , Cell Line , Coculture Techniques , Diabetes Mellitus, Type 2/pathology , Endothelial Cells , Eryptosis , Erythrocyte Deformability , Erythrocytes/metabolism , Glycated Hemoglobin/analysis , Healthy Volunteers , Hemolysis , Humans , Oxidative Stress , Primary Cell Culture
2.
Atherosclerosis ; 291: 87-98, 2019 12.
Article in English | MEDLINE | ID: mdl-31704555

ABSTRACT

BACKGROUND AND AIMS: Atherothrombotic plaques of type 2 diabetic (T2D) patients are characterized by an increased neovascularization and intraplaque hemorrhage. The clearance of erythrocytes may be carried out by vascular cells. We explored the potential of human endothelial cells to bind and phagocyte in vitro aged and/or glycated erythrocytes as well as erythrocytes obtained from diabetic patients. METHODS: Fresh, aged and glycated-aged erythrocytes from healthy volunteers and T2D patients were tested for their binding and phagocytosis capacity as well as the potential functional consequences on endothelial cells (viability, proliferation and wound healing capacity). Immunohistochemistry was also performed in human carotid atherothrombotic samples (from patients with or without T2D). RESULTS: Aging and glycation of erythrocytes induced phosphatidylserine (PS) exposure and oxidative stress leading to enhanced endothelial cell binding and engulfment. Phagocytosis by endothelial cells was more pronounced with aged and glycated erythrocytes than with fresh ones. Phagocytosis was enhanced with T2D versus healthy erythrocytes. Furthermore, endothelial wound healing potential was significantly blunted after exposure to glycated-aged versus fresh erythrocytes. Finally, we show that interactions between erythrocytes and endothelial cells and their potential phagocytosis may occur in vivo, in atherothrombotic conditions, in neovessels and in the luminal endothelial lining. CONCLUSIONS: Endothelial cells may play an important role in erythrocyte clearance in an atherothrombotic environment. Under diabetic conditions, erythrocyte glycation favors their engulfment by endothelial cells and may participate in endothelial dysfunction, thereby promoting vulnerable atherothrombotic plaques to rupture.


Subject(s)
Carotid Artery Diseases/metabolism , Cellular Senescence , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Erythrocytes/metabolism , Glycated Hemoglobin/metabolism , Phagocytosis , Carotid Artery Diseases/blood , Carotid Artery Diseases/pathology , Case-Control Studies , Cell Adhesion , Cell Line , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Endothelial Cells/pathology , Erythrocytes/pathology , Female , Humans , Male , Oxidative Stress , Plaque, Atherosclerotic , Rupture, Spontaneous , Wound Healing
3.
J Biol Chem ; 293(13): 4778-4791, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29414771

ABSTRACT

The long-acting glucagon-like peptide-1 analogue liraglutide has proven efficiency in the management of type 2 diabetes and also has beneficial effects on cardiovascular diseases. Liraglutide's protracted action highly depends on its capacity to bind to albumin via its palmitic acid part. However, in diabetes, albumin can undergo glycation, resulting in impaired drug binding. Our objective in this study was to assess the impact of human serum albumin (HSA) glycation on liraglutide affinity. Using fluorine labeling of the drug and 19F NMR, we determined HSA affinity for liraglutide in two glycated albumin models. We either glycated HSA in vitro by incubation with glucose (G25- or G100-HSA) or methylglyoxal (MGO-HSA) or purified in vivo glycated HSA from the plasma of diabetic patients with poor glycemic control. Nonglycated commercial HSA (G0-HSA) and HSA purified from plasma of healthy individuals served as controls. We found that glycation decreases affinity for liraglutide by 7-fold for G100-HSA and by 5-fold for MGO-HSA compared with G0-HSA. A similarly reduced affinity was observed for HSA purified from diabetic individuals compared with HSA from healthy individuals. Our results reveal that glycation significantly impairs HSA affinity to liraglutide and confirm that glycation contributes to liraglutide's variable therapeutic efficiency, depending on diabetes stage. Because diabetes is a progressive disease, the effect of glycated albumin on liraglutide affinity found here is important to consider when diabetes is managed with this drug.


Subject(s)
Glucagon-Like Peptide 1 , Liraglutide/chemistry , Serum Albumin, Human/chemistry , Diabetes Mellitus/blood , Glycosylation , Humans , Liraglutide/pharmacokinetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Serum Albumin, Human/metabolism
4.
Biomed Res Int ; 2017: 7989180, 2017.
Article in English | MEDLINE | ID: mdl-29362717

ABSTRACT

OBJECTIVES: Diabetes is a major health problem associated with hyperglycemia and chronically increased oxidative stress and enhanced formation of advanced glycation end-products (AGEs). The aim of this study was to determine whether oxidative plasma biomarkers in diabetic patients could be evidenced and associated with vascular complications. METHODS: Oxidative stress biomarkers such as thiols, ischemia-modified albumin (IMA), glycated albumin (GA), fructosamine, and AGEs were measured in 75 patients with poorly controlled type 2 diabetes (HbA1c > 7.5%) with (44) or without (31) vascular disease and in 31 nondiabetic controls. RESULTS: Most biomarkers of oxidation and glycation were significantly increased in diabetic patients in comparison with nondiabetics. Fructosamines, GA, IMA, and AGEs were positively correlated and levels of fluorescent AGEs were significantly increased in the plasma from patients presenting vascular complication. CONCLUSIONS: These results bring new evidence for the potential interest of glycated albumin, oxidative stress, and glycoxidation parameters in the monitoring of type 2 diabetic patients. Furthermore, it emphasizes fluorescent AGEs as a putative indicator for vascular event prediction in diabetic patients.


Subject(s)
Cardiovascular Diseases/etiology , Diabetes Mellitus, Type 2/complications , Glycation End Products, Advanced/metabolism , Biomarkers/blood , Biomarkers/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/metabolism , Case-Control Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Female , Fructosamine/metabolism , Glycated Hemoglobin/metabolism , Humans , Male , Middle Aged , Oxidation-Reduction , Oxidative Stress/physiology , Pilot Projects , Serum Albumin/metabolism , Serum Albumin, Human/metabolism , Glycated Serum Albumin
5.
Free Radic Biol Med ; 102: 133-148, 2017 01.
Article in English | MEDLINE | ID: mdl-27890722

ABSTRACT

Increased oxidative stress and advanced glycation end-product (AGE) formation are major contributors to the development of type 2 diabetes. Here plasma proteins e.g. albumin can undergo glycoxidation and play a key role in diabetes onset and related pathologies. However, despite recent progress linking albumin-AGE to increased oxidative stress and downstream effects, its action in metabolic organs such as the liver remains to be elucidated. The current study therefore investigated links between oxidative perturbations and biochemical/structural modifications of plasma albumin, and subsequent downstream effects in transgenic db/db mouse livers and HepG2 cells, respectively. Our data reveal increased oxidative stress biomarkers and lipid accumulation in plasma and livers of diabetic mice, together with albumin glycoxidation. Purified mouse albumin modifications resembled those typically found in diabetic patients, i.e. degree of glycation, carbonylation, AGE levels and in terms of chemical composition. Receptor for AGE expression and reactive oxygen species production were upregulated in db/db mouse livers, together with impaired proteolytic, antioxidant and mitochondrial respiratory activities. In parallel, acute exposure of HepG2 cells to glycated albumin also elicited intracellular free radical formation. Together this study demonstrates that AGE-modified albumin can trigger damaging effects on the liver, i.e. by increasing oxidative stress, attenuating antioxidant capacity, and by impairment of hepatic proteolytic and respiratory chain enzyme activities.


Subject(s)
Antigens, Neoplasm/genetics , Diabetes Mellitus, Type 2/metabolism , Liver/metabolism , Mitogen-Activated Protein Kinases/genetics , Oxidative Stress/genetics , Serum Albumin/metabolism , Animals , Antigens, Neoplasm/metabolism , Diabetes Mellitus, Type 2/pathology , Free Radicals/metabolism , Gene Expression Regulation , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/metabolism , Hep G2 Cells , Humans , Liver/pathology , Mice , Mice, Inbred NOD , Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Glycated Serum Albumin
6.
Adipocyte ; 5(4): 384-388, 2016.
Article in English | MEDLINE | ID: mdl-27994953

ABSTRACT

Chronic low grade inflammation is one of the major metabolic disorders in case of obesity and associated pathologies. By its important secretion function, the role of adipose tissue in this metabolic low grade inflammation is well known. Recently, it was demonstrated that the alarmin high mobility group box protein 1 (HMGB1) is involved in obesity-related pathologies by its increased serum levels in obese compared to normal weight individuals, and by its pro-inflammatory effects. However, the role of HMGB1 on adipocytes inflammation is poorly documented and we propose to investigate this point. Primary culture of human subcutaneous adipocytes were performed from human adipose tissue samples. Cells were treated with recombinant HMGB1 with/without anti-TLR4 antibody and inhibitors of NF-κB and P38 MAPK. Supernatants were collected for IL-6 and MCP-1 ELISA. HMGB1 initiates Toll-like receptor 4 (TLR4)-dependent activation of inflammation through the downstream NF-κB and P38 MAPK signaling pathway to upregulate the secretion of the pro-inflammatory cytokine IL-6. HMGB1 has pro-inflammatory effects on adipocytes. This reinforces the role of TLR4 in adipose tissue inflammation and antagonizing the HMGB1 inflammatory pathway could bring on new therapeutic targets to counteract obesity-associated pathologies.

7.
Biochim Biophys Acta ; 1830(6): 3532-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23376313

ABSTRACT

BACKGROUND: Albumin constitutes the most abundant circulating antioxidant and prevents oxidative damages. However, in diabetes, this plasmatic protein is exposed to several oxidative modifications, which impact on albumin antioxidant properties. METHODS: Most studies dealing on albumin antioxidant activities were conducted on in vitro modified protein. Here we tried to decipher whether reduced antioxidant properties of albumin could be evidenced in vivo. For this, we compared the antioxidant properties of albumin purified from diabetic patients to in vitro models of glycated albumin. RESULTS: Both in vivo and in vitro glycated albumins displayed impaired antioxidant activities in the free radical-induced hemolysis test. Surprisingly, the ORAC method (Oxygen Radical Antioxidant Capacity) showed an enhanced antioxidant activity for glycated albumin. Faced with this paradox, we investigated antioxidant and anti-inflammatory activities of our albumin preparations on cultured cells (macrophages and adipocytes). Reduced cellular metabolism and enhanced intracellular oxidative stress were measured in cells treated with albumin from diabetics. NF-kB -mediated gene induction was higher in macrophages treated with both type of glycated albumin compared with cells treated with native albumin. Anti inflammatory activity of native albumin is significantly impaired after in vitro glycation and albumin purified from diabetics significantly enhanced IL6 secretion by adipocytes. Expression of receptor for advanced glycation products is significantly enhanced in glycated albumin-treated cells. CONCLUSIONS AND GENERAL SIGNIFICANCE: Our results bring new evidences on the deleterious impairments of albumin important functions after glycation and emphasize the importance of in vivo model of glycation in studies relied to diabetes pathology.


Subject(s)
Antioxidants/metabolism , Diabetes Mellitus/metabolism , Models, Biological , Protein Processing, Post-Translational , Serum Albumin/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation , Humans , Male , NF-kappa B/metabolism , Oxidative Stress , Receptor for Advanced Glycation End Products , Receptors, Immunologic/biosynthesis
8.
Biochimie ; 94(9): 1960-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22627382

ABSTRACT

Albumin, the major circulating protein in blood, can undergo increased glycation in diabetes. One of the main properties of this plasma protein is its strong affinity to bind many therapeutic drugs, including warfarin and ketoprofen. In this study, we investigated whether or not there were any significant changes related to in vitro or in vivo glycation in the structural properties and the binding of human albumin to both therapeutic drugs. Structural parameters, including redox state and ketoamine contents of in vitro and in vivo glycated purified albumins, were investigated in parallel with their affinity for warfarin and ketoprofen. High-performance liquid chromatography was used to determine the free drug concentrations and dissociation constants according to the Scatchard method. An alternative method based on fluorescence spectroscopy was also used to assess drug-binding properties. Oxidation and glycation levels were found to be enhanced in albumin purified from diabetic patients or glycated with glucose or methylglyoxal, after determination of their ketoamine, free thiol, amino group and carbonyl contents. In parallel, significant impairments in the binding affinity of in vitro and in vivo glycated albumin, as indicated by the higher dissociation constant values and confirmed by higher free drug fractions, were observed. To a lesser extent, this alteration also significantly affected diabetic albumin affinity, indicated by a lower static quenching in fluorescence spectroscopy. This work provides useful information supporting in vivo diabetic albumin could be the best model of glycation for monitoring diabetic physiopathology and should be valuable to know if glycation of albumin could contribute to variability in drugs response during diabetes.


Subject(s)
Ketoprofen/metabolism , Serum Albumin/metabolism , Warfarin/metabolism , Adult , Female , Glycation End Products, Advanced , Humans , Male , Middle Aged , Protein Binding , Glycated Serum Albumin
SELECTION OF CITATIONS
SEARCH DETAIL
...