Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 389
Filter
Add more filters

Publication year range
1.
Eur J Immunol ; 50(8): 1174-1186, 2020 08.
Article in English | MEDLINE | ID: mdl-32250445

ABSTRACT

Staphylococcus aureus, a pathogen most frequently found in diabetic foot ulcer infection, was recently suggested as an intracellular pathogen. Autophagy in professional phagocytes like macrophages allows selective destruction of intracellular pathogens, and its dysfunction can increase the survival of internalized pathogens, causing infections to worsen and spread. Previous works have shown that S. aureus infections in diabetes appeared more severe and invasive, and coincided with the suppressed autophagy in dermal tissues of diabetic rat, but the exact mechanisms are unclear. Here, we demonstrated that accumulation of advanced glycation end products (AGEs) contributed to the diminished autophagy-mediated clearance of S. aureus in the macrophages differentiated from PMA-treated human monocytic cell line THP-1. Importantly, infected macrophages showed increased S. aureus containing autophagosome, but the subsequent fusion of S. aureus containing autophagosome and lysosome was suppressed in AGEs-pretreated cells, suggesting AGEs blocked the autophagic flux and enabled S. aureus survival and escape. At the molecular level, elevated lysosomal ARL8 expression in AGEs-treated macrophages was required for AGEs-mediated inhibition of autophagosome-lysosome fusion. Silencing ARL8 in AGEs-treated macrophages restored autophagic flux and increased S. aureus clearance. Our results therefore demonstrate a new mechanism, in which AGEs accelerate S. aureus immune evasion in macrophages by ARL8-dependent suppression of autophagosome-lysosome fusion and bactericidal capability.


Subject(s)
ADP-Ribosylation Factors/physiology , Glycation End Products, Advanced/physiology , Lysosomes/physiology , Macrophages/immunology , Phagocytosis , Staphylococcus aureus/immunology , Autophagosomes/physiology , Humans , Immune Evasion , THP-1 Cells , Up-Regulation
2.
Hepatology ; 67(2): 676-689, 2018 02.
Article in English | MEDLINE | ID: mdl-28865156

ABSTRACT

Cholangiopathies are a diverse group of progressive diseases whose primary cell targets are cholangiocytes. To identify shared pathogenesis and molecular connectivity among the three main human cholangiopathies (biliary atresia [BA], primary biliary cholangitis [PBC], and primary sclerosing cholangitis [PSC]), we built a comprehensive platform of published data on gene variants, gene expression, and functional studies and applied network-based analytics in the search for shared molecular circuits. Mining the data platform with largest connected component and interactome analyses, we validated previously reported associations and identified essential and hub genes. In addition to disease-specific modules, we found a substantial overlap of disease neighborhoods and uncovered a group of 34 core genes that are enriched for immune processes and abnormal intestine/hepatobiliary mouse phenotypes. Within this core, we identified a gene subcore containing signal transduction and activator of transcription 3, interleukin-6, tumor necrosis factor, and forkhead box P3 prominently placed in a regulatory connectome of genes related to cellular immunity and fibrosis. We also found substantial gene enrichment in the advanced glycation endproduct/receptor for advanced glycation endproducts (RAGE) pathway and showed that RAGE activation induced cholangiocyte proliferation. Conclusion: Human cholangiopathies share pathways enriched by immunity genes and a molecular connectome that links different pathogenic features of BA, PBC, and PSC. (Hepatology 2018;67:676-689).


Subject(s)
Bile Duct Diseases/genetics , Connectome , Animals , Bile Duct Diseases/etiology , Bile Duct Diseases/immunology , Biliary Atresia/genetics , Cholangitis, Sclerosing/genetics , Databases, Genetic , Forkhead Transcription Factors/physiology , Gene Regulatory Networks , Glycation End Products, Advanced/physiology , Humans , Interleukin-6/physiology , Mice , MicroRNAs/physiology , Receptor for Advanced Glycation End Products/physiology , STAT3 Transcription Factor/physiology
3.
Biosci Biotechnol Biochem ; 83(6): 1136-1145, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30822216

ABSTRACT

Advanced glycation end products (AGEs) are implicated in the development of diabetic complications via the receptor for AGEs (RAGE). We have reported that the 3-hydroxypyridinium (3HP)-containing AGEs derived from α-hydroxyaldehydes physically interact with RAGE and show cytotoxicity. Lactaldehyde (LA) is formed from a reaction between threonine and myeloperoxidase, but no LA-derived AGEs have been characterized. Here, we identify the structure and physiological effects of an AGE derived from LA. We isolated a novel 3HP derivative, 2-acetamido-6-(3-hydroxy-5-methyl-pyridin-1-ium-1-yl)hexanoate, named as N-acetyl-LAPL (lactaldehyde-derived pyridinium-type lysine adduct), from a mixture of LA with Nα-acetyl-L-lysine. LAPL was also detected in the LA-modified protein. LAPL elicited toxicity in PC12 neuronal cells, but the effect was suppressed by the soluble form of RAGE as a decoy receptor. Moreover, surface plasmon resonance-based analysis revealed that LAPL specifically binds to recombinant RAGE. These results indicate that LA generates an AGE containing the 3HP moiety and contributes to RAGE-dependent cytotoxicity. Abbreviations: AGEs: advanced glycation end products; RAGE: receptor for advanced glycation end products; 3HP: 3-hydroxypyridinium; LA: lactaldehyde; LAPL: lactaldehyde-derived pyridinium-type lysine adduct; BSA: bovine serum albumin; GLAP: glyceraldehyde-derived pyridinium; MPO: myeloperoxidase; HFBA: heptafluorobutyric acid; TFA: trifluoroacetic acid; HPLC: high performance liquid chromatography; LC-ESI-QTOF-MS: liquid chromatography-electrospray ionization-quadrupole time-of-flight-mass spectrometry; NMR: nuclear magnetic resonance; LA-BSA: lactaldehyde-modified bovine serum albumin; PBS: phosphate buffered saline, GST, glutathione S-transferase; SPR: surface plasmon resonance; OP-lysine: 2-ammonio-6-(3-oxidopyridinium-1-yl)hexanoate; GLO1: glyoxalase 1; MG, methylglyoxal.


Subject(s)
Aldehydes/chemistry , Glycation End Products, Advanced/chemistry , Glycation End Products, Advanced/physiology , Aldehydes/toxicity , Animals , Lysine/analogs & derivatives , Lysine/chemistry , Molecular Structure , Neurons/drug effects , PC12 Cells , Pyridinium Compounds/chemistry , Rats , Receptor for Advanced Glycation End Products/chemistry , Serum Albumin, Bovine/chemistry , Surface Properties
4.
Zhonghua Yan Ke Za Zhi ; 54(6): 475-480, 2018 Jun 11.
Article in Zh | MEDLINE | ID: mdl-29895121

ABSTRACT

Diabetic keratopathy is a common ocular complication of patients with a long-term history of diabetes and it will have a negative effect on the visual quality and function. A study reported that the incidence of diabetic keratopathy in diabetic patients ranged from 47% to 64%, but the precise underlying pathogenesis remains unclear. There is evidence that advanced glycation end products contribute substantially to the onset and progress of various diabetic complications and it is a key factor for the mechanism of the hyperglycemic memory. This review focuses primarily on the present research state and prospect of advanced glycation end products and their role in the pathological changes of the cornea. (Chin J Ophthalmol, 2018, 54: 475-480).


Subject(s)
Corneal Diseases , Diabetes Complications , Diabetes Mellitus , Glycation End Products, Advanced , Cornea , Corneal Diseases/physiopathology , Diabetes Complications/physiopathology , Glycation End Products, Advanced/physiology , Humans
5.
Microcirculation ; 24(3)2017 04.
Article in English | MEDLINE | ID: mdl-28129474

ABSTRACT

The disruption of endothelial integrity and the occurrence of angiogenesis in response to AGEs contribute greatly to micro- and macrovascular complications associated with DM. Among human dermal, brain, and retinal vascular ECs, activation of ERM, moesin, by phosphorylation of Thr-558 is involved in AGE-induced hyperpermeability and angiogenesis via the Rho and ROCK (Rho/ROCK) and p38 pathways. Src also plays an important role in AGE-induced endothelial barrier dysfunction by phosphorylating moesin, VE-cadherin, and FAK. Furthermore, recent studies have demonstrated that ROS serve as a key mediator of the AGE-induced endothelial response. ROS inhibition would greatly benefit ECs. This review focuses on the role of moesin in microvascular permeability and angiogenesis, and on the involvement of Src and ROS in endothelial barrier disruption.


Subject(s)
Endothelium, Vascular/physiopathology , Glycation End Products, Advanced/physiology , Microfilament Proteins/physiology , Capillary Permeability , Humans , Neovascularization, Pathologic , Reactive Oxygen Species/metabolism , src-Family Kinases/metabolism
6.
Mol Cell Biochem ; 434(1-2): 153-162, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28455790

ABSTRACT

Advanced glycation end products (AGEs) are formed from the non-enzymatic glycation reaction of reducing sugars or their metabolites with the free amino groups of several biomolecules and are known to play pathophysiological roles in various inflammatory diseases. In an earlier study, it was suggested that tumor necrosis factor-like weak inducer of apoptosis (TWEAK) has a unique role to regulate the tumor necrosis factor α (TNFα)-induced inflammatory response. In this study, we investigated the effect of the AGEs-TWEAK interaction on proinflammatory signaling responses in endothelial cells and the influence of AGEs on the cellular function of TWEAK in the inflammatory process. The effect of AGEs on the TWEAK/TNFα-induced gene expression of interleukin-8 (IL-8) was determined by real-time RT-PCR in endothelial-like EA.hy.926 cells. The pull-down assay was performed using recombinant His-tagged TWEAK and AGEs. The NF-κB activation was analyzed by Western blotting with canonical and non-canonical pathway-specific antibodies. AGEs dose-dependently inhibited TWEAK-induced IL-8 gene expression, whereas AGEs themselves had almost no effect on IL-8 expression. AGEs were found to bind directly to TWEAK in the pull-down assay. TNFα-induced IL-8 production and canonical NF-κB activation were suppressed by TWEAK pretreatment, whereas TWEAK-induced non-canonical NF-κB activation was enhanced by pretreatment. These effects induced by TWEAK pretreatment were abolished by the co-addition of AGEs. Our findings suggest that AGEs attenuate the function of TWEAK to regulate the TNFα-induced inflammatory responses, which provide important clues for understanding the significance of the AGEs-TWEAK interaction in inflammatory processes.


Subject(s)
Cytokine TWEAK/physiology , Glycation End Products, Advanced/physiology , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation Mediators/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Real-Time Polymerase Chain Reaction
7.
J Biol Chem ; 290(47): 28189-28199, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26472922

ABSTRACT

The interaction between advanced glycation end products (AGEs) and receptor of AGEs (RAGE) is associated with the development and progression of diabetes-associated osteoporosis, but the mechanisms involved are still poorly understood. In this study, we found that AGE-modified bovine serum albumin (AGE-BSA) induced a biphasic effect on the viability of hFOB1.19 cells; cell proliferation was stimulated after exposure to low dose AGE-BSA, but cell apoptosis was stimulated after exposure to high dose AGE-BSA. The low dose AGE-BSA facilitates proliferation of hFOB1.19 cells by concomitantly promoting autophagy, RAGE production, and the Raf/MEK/ERK signaling pathway activation. Furthermore, we investigated the effects of AGE-BSA on the function of hFOB1.19 cells. Interestingly, the results suggest that the short term effects of low dose AGE-BSA increase osteogenic function and decrease osteoclastogenic function, which are likely mediated by autophagy and the RAGE/Raf/MEK/ERK signal pathway. In contrast, with increased treatment time, the opposite effects were observed. Collectively, AGE-BSA had a biphasic effect on the viability of hFOB1.19 cells in vitro, which was determined by the concentration of AGE-BSA and treatment time. A low concentration of AGE-BSA activated the Raf/MEK/ERK signal pathway through the interaction with RAGE, induced autophagy, and regulated the proliferation and function of hFOB1.19 cells.


Subject(s)
Autophagy/physiology , Cell Proliferation/physiology , Glycation End Products, Advanced/physiology , MAP Kinase Signaling System , Osteoblasts/cytology , Receptor for Advanced Glycation End Products/physiology , raf Kinases/metabolism , Cell Line , Humans
8.
Biochim Biophys Acta ; 1850(9): 1898-904, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26028296

ABSTRACT

BACKGROUND: Receptor for advanced glycation end-products popularly known as RAGE is a cell surface immunoglobulin class of molecule, binds with multiple ligands and therefore considered as a multi-ligand receptor. Use of RAGE deficient mice (RAGE(-/-)) as well as established mouse models pertaining to inflammation-associated carcinogenesis such as that of chemically induced carcinogenesis and colitis associated cancer provides a direct genetic evidence for a likelihood novel role of RAGE in cancer, with respect to its ability to lead cancer cell proliferation and survival. Besides inflammation, interaction of RAGE with its various ligands enhances oxidative stress both in cancerous and noncancerous cells which further complicates the progression of cancers. SCOPE OF REVIEW: Till date, no single review article has discussed the mechanism of RAGE dependent complication of cancers, particularly the role of RAGE in cancer cell proliferation, angiogenesis, survival and anti-apoptosis needs to be discussed. MAJOR CONCLUSION: RAGE enhances the number of cancer cells by activating the cell cycle proteins (e.g., cyclin D1), anti-apoptotic proteins (e.g., BCl2), prosurvival (AKT) and autophagic proteins. Role of RAGE has also been detected in formation of new blood vessels (angiogenesis) in the cancer cells and activation of myeloid derived suppressor cells (MDSCs). GENERAL SIGNIFICANCE: This review article describes the role of RAGE in the complication of various types of cancers and the possible usefulness of RAGE dependent therapy to confront cancers in a stronger magnitude.


Subject(s)
Neoplasms/complications , Receptors, Immunologic/physiology , Animals , Apoptosis , Cell Proliferation , Cell Survival , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/physiology , Humans , Mice , Neoplasm Invasiveness , Receptor for Advanced Glycation End Products
9.
Neurochem Res ; 40(4): 713-22, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25596942

ABSTRACT

Recent studies revealed that dietary intake of docosahexaenoic acid (DHA) prevented diabetic retinopathy (DR), but the underlying mechanism was not fully understood. Retinal microglia are a specialized population of macrophages in retina. Considerable evidence has shown that microglia activation may trigger neuronal death and vascular dysfunction in DR. The aim of this study was to investigate the effects of DHA on advanced glycation end products (AGEs)-induced microglia activation using an in vitro microglia culture system, and concurrently to explore the mediating mechanisms. DHA inhibited AGEs-induced microglia activation and tumor necrosis factor α (TNFα) secretion. These effects of DHA were directly linked with suppression of nuclear factor-kappa B (NFκB) activity, as evident by the reduction of p-IκBα expression, p-NFκB p65 nucleus translocation, NFκB DNA binding activity, and the regulation of gene transcription (TNFα, IL-1ß, ICAM-1, and RAGE mRNA). Furthermore, DHA significantly increased phosphorylation of peroxisome proliferator-activated receptor-gamma (PPARγ), and combined with PPARγ stealth RNAi oligonucleotide, we confirmed that DHA inhibition of AGEs-induced microglia activation was partially through the PPARγ/NFκB pathway. Moreover, although AGEs incubation dramatically elevated expression of the cell surface receptor for AGEs (RAGE), DHA significantly inhibited RAGE and Src recruitment into lipid rafts. The AGEs-RAGE axis downstream signal transducers increased mitogen-activated protein kinase (p38 and JNK) phosphorylation. Taken together, DHA might inhibit AGEs-induced microglia activation via suppression of the PPARγ/NFκB pathway, and reduction of RAGE and AGEs/RAGE transducer recruitment into lipid rafts. These results provide a novel potential mechanism for the anti-inflammatory effects of DHA in DR prevention.


Subject(s)
Docosahexaenoic Acids/pharmacology , Glycation End Products, Advanced/physiology , Membrane Microdomains/metabolism , Microglia/drug effects , NF-kappa B/metabolism , PPAR gamma/metabolism , Receptor for Advanced Glycation End Products/metabolism , Retina/drug effects , Signal Transduction/drug effects , Animals , Microglia/cytology , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Retina/cytology , Retina/metabolism
10.
Arterioscler Thromb Vasc Biol ; 34(6): 1126-35, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24675660

ABSTRACT

In patients with diabetes mellitus, the ability of ischemic tissue to synchronize the molecular and cellular events leading to restoration of tissue perfusion in response to the atherosclerotic occlusion of a patent artery is markedly impaired. As a consequence, adverse tissue remodeling and the extent of ischemic injury are intensified, leading to increased morbidity and mortality. Growing evidence from preclinical and clinical studies has implicated alterations in hypoxia-inducible factor 1 levels in the abrogation of proangiogenic pathways, including vascular endothelial growth factor A/phosphoinositide 3' kinase/AKT/endothelial nitric oxide synthase and in the activation of antiangiogenic signals characterized by accumulation of advanced glycation end products, reactive oxygen species overproduction, and endoplasmic reticulum stress. In addition, the diabetic milieu shows a switch toward proinflammatory antiregenerative pathways. Finally, the mobilization, subsequent recruitment, and the proangiogenic potential of the different subsets of angiogenesis-promoting bone marrow-derived cells are markedly impaired in the diabetic environment. In this review, we will give an overview of the current understanding on the signaling molecules contributing to the diabetes mellitus-induced impairment of postischemic revascularization mainly in the setting of myocardial infarction or critical limb ischemia.


Subject(s)
Diabetic Angiopathies/physiopathology , Ischemia/physiopathology , Neovascularization, Physiologic , Animals , Bone Marrow Cells/physiology , Cell Movement , Endothelial Cells/cytology , Glycation End Products, Advanced/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Inflammation/physiopathology , MicroRNAs/physiology , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor A/physiology
11.
Biochim Biophys Acta ; 1833(1): 101-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23103427

ABSTRACT

RAGE (receptor for advanced glycation end-products) is a multiligand receptor of the immunoglobulin superfamily involved in inflammation, diabetes, atherosclerosis, nephropathy, neurodegeneration, and cancer. Advanced glycation end-products, high mobility group box-1 (amphoterin), ß-amyloid fibrils, certain S100 proteins, and DNA and RNA are RAGE ligands. Upon RAGE ligation, adaptor proteins (i.e., diaphanous-1, TIRAP, MyD88 and/or other as yet unidentified adaptors) associate with RAGE cytoplasmic domain resulting in signaling. However, RAGE activation may not be restricted to pathological statuses, the receptor being involved in tissue homeostasis and regeneration/repair upon acute injury, and in resolution of inflammation. RAGE effects are strongly dependent on the cell type and the context, which may condition therapeutic strategies aimed at reducing RAGE signaling.


Subject(s)
Homeostasis/genetics , Receptors, Immunologic/physiology , Regeneration/genetics , Wound Healing/genetics , Animals , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacology , Glycation End Products, Advanced/physiology , Homeostasis/drug effects , Homeostasis/physiology , Humans , Models, Biological , Receptor for Advanced Glycation End Products , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Regeneration/drug effects , Regeneration/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Wound Healing/drug effects , Wound Healing/physiology
12.
Lab Invest ; 94(5): 503-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24614199

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a major risk factor for hepatic fibrogenesis. NASH is often found in diabetic patients with hyperglycemia. Hyperglycemia induces non-enzymatic glycation of proteins, yielding advanced glycation end-products (AGEs). Effects of AGEs are mainly mediated by two categories of cytoplasmic membrane receptors. Receptor for AGEs (RAGE) is associated with increased oxidative stress and inflammation, whereas AGE receptor-1 (AGE-R1) is involved in detoxification and clearance of AGEs. Activation of hepatic stellate cells (HSC) is crucial to the development of hepatic fibrosis. We recently reported that AGEs stimulated HSC activation likely by inhibiting gene expression of AGE-R1 and inducing gene expression of RAGE in HSC, which were eliminated by the antioxidant curcumin. This study is to test our hypothesis that curcumin eliminates the effects of AGEs on the divergent regulation of the two receptors of AGEs in HSC by interrupting the AGE-caused activation of leptin signaling, leading to the inhibition of HSC activation. We observed herein that AGEs activated leptin signaling by inducing gene expression of leptin and its receptor in HSC. Like AGEs, leptin differentially regulated gene expression of RAGE and AGE-R1. Curcumin eliminated the effects of AGEs in HSC by interrupting leptin signaling and activating transcription factor NF-E2 p45-related factor 2 (Nrf2), leading to the elevation of cellular glutathione and the attenuation of oxidative stress. In conclusions, curcumin eliminated the effects of AGEs on the divergent regulation of gene expression of RAGE and AGE-R1 in HSC by interrupting the AGE-caused activation of leptin signaling, leading to the inhibition of HSC activation.


Subject(s)
Antineoplastic Agents/therapeutic use , Curcumin/therapeutic use , Glycation End Products, Advanced/antagonists & inhibitors , Hepatic Stellate Cells/drug effects , Leptin/antagonists & inhibitors , Receptors, Immunologic/metabolism , Animals , Antineoplastic Agents/pharmacology , Cells, Cultured , Chromones , Curcuma , Curcumin/pharmacology , Gene Expression Regulation/drug effects , Glycation End Products, Advanced/physiology , Hepatic Stellate Cells/metabolism , Leptin/metabolism , Leptin/physiology , Liver Cirrhosis/prevention & control , Male , Mice , Mice, Inbred C57BL , Morpholines , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products , Receptors, Leptin/metabolism , Tyrphostins
13.
Am J Pathol ; 182(1): 132-41, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23159951

ABSTRACT

Advanced glycation end products (AGEs) and their receptor (RAGE) play a role in diabetic nephropathy. Asymmetric dimethylarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, contributes to diabetic nephropathy. We have found that glucagon-like peptide-1 (GLP-1) inhibits the AGE-induced inflammatory reactions in endothelial cells. However, effects of GLP-1 on the AGE-RAGE-ADMA axis are unknown. This study examined the effects of GLP-1 on reactive oxygen species (ROS) generation, gene expression of protein arginine methyltransfetase-1 (PRMT-1), an enzyme that mainly generates ADMA, and ADMA levels in human proximal tubular cells. Streptozotocin-induced diabetic rats received continuous i.p. infusion of 0.3 µg of vehicle or 1.5 µg of the GLP-1 analog exendin-4 per kilogram of body weight for 2 weeks. We further investigated whether and how exendin-4 treatment reduced ADMA levels and renal damage in streptozotocin-induced diabetic rats. GLP-1 inhibited the AGE-induced RAGE and PRMT-1 gene expression, ROS, and ADMA generation in tubular cells, which were blocked by small-interfering RNAs raised against GLP-1 receptor. Exendin-4 treatment decreased gene expression of Rage, Prmt-1, Icam-1, and Mcp-1 and ADMA level; reduced urinary excretions of 8-hydroxy-2'-deoxyguanosine and albumin; and improved histopathologic changes of the kidney in diabetic rats. Our present study suggests that GLP-1 receptor agonist may inhibit the AGE-RAGE-mediated ADMA generation by suppressing PRMT-1 expression via inhibition of ROS generation, thereby protecting against the development and progression of diabetic nephropathy.


Subject(s)
Arginine/analogs & derivatives , Diabetes Mellitus, Experimental/metabolism , Kidney/metabolism , Peptides/pharmacology , Protein-Arginine N-Methyltransferases/biosynthesis , Receptors, Glucagon/agonists , Venoms/pharmacology , Animals , Arginine/biosynthesis , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Drug Evaluation, Preclinical/methods , Exenatide , Gene Expression Regulation, Enzymologic/drug effects , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Glycation End Products, Advanced/physiology , Humans , Hypertrophy/prevention & control , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Kidney Glomerulus/pathology , Kidney Tubules/metabolism , Macrophages/pathology , Male , Peptides/therapeutic use , Protein-Arginine N-Methyltransferases/genetics , RNA, Messenger/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Receptors, Glucagon/metabolism , Receptors, Immunologic/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Venoms/therapeutic use
14.
Blood ; 119(25): 6136-44, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22431576

ABSTRACT

Diabetes mellitus has been associated with platelet hyperreactivity, which plays a central role in the hyperglycemia-related prothrombotic phenotype. The mechanisms responsible for this phenomenon are not established. In the present study, we investigated the role of CD36, a class-B scavenger receptor, in this process. Using both in vitro and in vivo mouse models, we demonstrated direct and specific interactions of platelet CD36 with advanced glycation end products (AGEs) generated under hyperglycemic conditions. AGEs bound to platelet CD36 in a specific and dose-dependent manner, and binding was inhibited by the high-affinity CD36 ligand NO(2)LDL. Cd36-null platelets did not bind AGE. Using diet- and drug-induced mouse models of diabetes, we have shown that cd36-null mice had a delayed time to the formation of occlusive thrombi compared with wild-type (WT) in a FeCl(3)-induced carotid artery injury model. Cd36-null mice had a similar level of hyperglycemia and a similar level of plasma AGEs compared with WT mice under this condition, but WT mice had more AGEs incorporated into thrombi. Mechanistic studies revealed that CD36-dependent JNK2 activation is involved in this prothrombotic pathway. Therefore, the results of the present study couple vascular complications in diabetes mellitus with AGE-CD36-mediated platelet signaling and hyperreactivity.


Subject(s)
Asymptomatic Diseases , Blood Platelets/metabolism , CD36 Antigens/metabolism , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/physiology , Thrombosis/etiology , Animals , Blood Platelets/drug effects , CD36 Antigens/genetics , CD36 Antigens/physiology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/blood , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Diet, Atherogenic , Glycation End Products, Advanced/pharmacology , Hyperglycemia/blood , Hyperglycemia/etiology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Platelet Aggregation/genetics , Platelet Aggregation/physiology , Protein Binding , Streptozocin , Thrombosis/metabolism , Thrombosis/pathology
15.
Hum Reprod ; 29(1): 125-34, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24256989

ABSTRACT

STUDY QUESTION: Do advanced glycation end products (AGE) and the receptor for advanced glycation end products (RAGE) affect the cells of the human ovarian follicle? SUMMARY ANSWER: AGE accumulate on the surface of ovarian granulosa-lutein (GL) cells and monocytes by binding to RAGE and other receptors with possible functional effects on these cells. WHAT IS KNOWN ALREADY: AGE and RAGE are expressed in granulosa and theca cells, as well as in luteinized cells derived from the ovary. STUDY DESIGN, SIZE, DURATION: In this prospective cohort study, human follicle fluid-derived cells were isolated from aspirates of ovarian follicles of women who underwent assisted reproduction treatment. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immunofluorescence microscopy and multi-colour flow cytometry were used to determine the presence of AGE and RAGE on the surface of follicular fluid-derived cells and to characterize downstream effects of RAGE activation. MAIN RESULTS AND THE ROLE OF CHANCE: GL cells and ovarian monocytes were found to contain AGE and RAGE and to bind AGE-bovine serum albumin (BSA) in correlation with the patients' chronological age. AGE-BSA and BSA failed to induce significantly the cleavage of caspase-3, phosphorylation of nuclear factor-κB or the binding of annexin V (the latter was marginally increased). AGE-fibronectin was found to induce detachment of cultured GL cells in vitro. LIMITATIONS, REASONS FOR CAUTION: The impact of AGE and RAGE in the ovary, shown here in cells in culture, remains to be affirmed in clinical settings. WIDER IMPLICATIONS OF THE FINDINGS: The ligands of RAGE and their effects in the ovary remain uncertain but this study implies that AGEs in the form of structural long-lived extracellular matrix proteins, rather than soluble AGEs, may play a role in the decline of ovarian function during ageing. STUDY FUNDING/COMPETING INTEREST(S): The project was funded by the Norwegian Resource Centre for Women's Health, Oslo University Hospital. The authors have no conflicts of interests.


Subject(s)
Glycation End Products, Advanced/physiology , Ovary/physiology , Receptors, Immunologic/physiology , Female , Follicular Fluid/cytology , Glycation End Products, Advanced/metabolism , Humans , Ovary/growth & development , Prospective Studies , Receptor for Advanced Glycation End Products , Serum Albumin, Bovine/metabolism
16.
Hum Reprod ; 29(1): 135-45, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24173721

ABSTRACT

STUDY QUESTION: Do advanced glycation end products (AGEs) and their receptors play a role in female reproduction? SUMMARY ANSWER: AGEs might contribute to the etiology of polycystic ovary syndrome (PCOS) and infertility. WHAT IS KNOWN ALREADY: The endogenous AGEs are produced in the body by chemical reactions. Exogenous sources of AGEs are diet and smoking. AGEs have been proposed to be among the main intermediaries involved in several diseases, such as metabolic syndrome, type 2 diabetes mellitus, cardiovascular disease, ovarian aging, inflammation, neurodegenerative disorders and PCOS. STUDY DESIGN, SIZE, DURATION: A systematic review was performed for all available basic science and clinical peer-reviewed articles published in PubMed from 1987 to date. Abstracts of annual meetings of the Endocrine Society and American Society for Reproductive Medicine were also reviewed. PARTICIPANTS/MATERIALS, SETTING, METHODS: A total of 275 publications and scientific abstracts were identified from the initial search. Sixty-two papers and four published scientific abstracts were selected for full review. The main outcomes were the regulatory effects of AGEs on: (i) granulosa cells, adipocyte physiology, obesity and insulin resistance in women with PCOS and in polycystic ovary animal models and (ii) infertility and measures of ovarian reserve. MAIN RESULTS AND THE ROLE OF CHANCE: There is an intricate relationship between the AGE-RAGE (receptor for AGEs) system and some aspects of PCOS, such as granulosa cell dysfunction, adipocyte pathophysiology, obesity and insulin resistance. Additionally, irregular ovarian AGE signaling might in part explain the abnormal ovarian histology observed in women with PCOS. The ovarian dysfunction due to AGEs in women without PCOS suggests a role for the AGE-RAGE system in the ovarian follicular environment, and might relate to assisted reproduction technology outcome and measures of ovarian reserve. LIMITATIONS, REASONS FOR CAUTION: The body of literature currently available limits these findings. The results obtained from granulosa cell lines and animal models may not fully extrapolate to humans. WIDER IMPLICATIONS OF THE FINDINGS: This review underscores a critical need to unveil the exact mechanistic actions of AGEs in reproductive physiology and more specifically the hypothalamic-pituitary-ovarian axis. AGE inhibitors might present an emerging therapeutic approach with significant applications in the context of PCOS and infertility. STUDY FUNDING/COMPETING INTEREST(S): American Society for Reproductive Medicine New Investigator Award and University of Vermont College of Medicine Internal Funds. No competing interests.


Subject(s)
Glycation End Products, Advanced/physiology , Polycystic Ovary Syndrome/etiology , Reproduction/drug effects , Adipocytes/drug effects , Adipocytes/physiology , Animals , Female , Granulosa Cells/metabolism , Humans , Insulin Resistance/physiology , Metabolic Syndrome/physiopathology , Obesity/metabolism , Polycystic Ovary Syndrome/physiopathology , Reproduction/physiology
17.
Amino Acids ; 46(2): 267-77, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23588491

ABSTRACT

Research on the impact of Maillard reaction products (MRPs) on microorganisms has been reported in the literature for the last 60 years. In the current study, the impact of an MRP-rich medium on the growth of three strains of Escherichia coli was measured by comparing two classic methods for studying the growth of bacteria (plate counting and optical density at 600 nm) and by tracing MRP utilisation. Early stage and advanced MRPs in the culture media were assessed by quantifying furosine and N (ε) -carboxymethyllysine (CML) levels, respectively, using chromatographic methods. These measures were performed prior to and during bacterial growth to estimate the potential use of these MRPs by Escherichia coli CIP 54.8. Glucose and lysine, the two MRP precursors used in the MRP-rich medium, were also quantified by chromatographic means. Compared to control media, increased lag phases and decreased growth rates were observed in the MRP-rich medium for two out of the three Escherichia coli strains tested. In contrast, one strain isolated from the faeces of a piglet fed on a MRP-rich diet was not influenced by the presence of MRPs in the medium. Overall, CML as well as the products obtained by the thermal degradation of glucose and lysine, regardless of the Maillard reaction, did not affect the growth of the three strains tested. In addition, no degradation of fructoselysine or CML was found in the presence of Escherichia coli CIP 54.8.


Subject(s)
Escherichia coli/metabolism , Glycation End Products, Advanced/physiology , Culture Media/chemistry , Escherichia coli/growth & development , Gastrointestinal Tract/microbiology , Glycation End Products, Advanced/chemistry , Humans
18.
Minerva Endocrinol ; 39(3): 167-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25068304

ABSTRACT

Obesity affects ovarian function, one of the main regulators of female fertility. Tissue levels of the proinflammatory advanced glycation end-products (AGEs) and their receptors (RAGE) are elevated in obesity. AGEs are key contributors to perturbations in the ovarian microenvironment. On this basis, the present review focuses on clinical and experimental studies supporting the role of AGE-RAGE system as a contributor to obesity-related ovarian dysfunction. Particular emphasis has been given to changes in AGEs, RAGE and the anti-inflammatory soluble receptor (sRAGE) levels in obesity state and following dietary interventions (high-fat diet and weight loss). Ovarian sensitivity, in particular granulosa cell function and oocyte meiosis, to the pro-inflammatory AGE-RAGE system as well as the relationship of follicular fluid AGEs and sRAGE to in vitro fertilization outcome are also discussed. Overall, obesity, with its alterations in the AGE-RAGE system, can disrupt the ovarian microenvironment potentially compromising oocyte competence and fertility. This review underscores a critical need to uncover the mechanistic actions of AGE-RAGE system in obesity-related ovarian dysfunction. Clinical and basic studies focusing on elucidating the patterns of accumulation and role of the AGE-RAGE system in human ovarian follicles are key steps in understanding their contribution to the health of human oocytes and embryos.


Subject(s)
Glycation End Products, Advanced/physiology , Infertility, Female/etiology , Obesity/complications , Ovary/physiopathology , Anovulation/etiology , Anovulation/physiopathology , Anti-Mullerian Hormone/blood , Cellular Microenvironment , Diet, Western/adverse effects , Female , Fertilization in Vitro , Follicular Fluid/metabolism , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/pharmacokinetics , Humans , Infertility, Female/metabolism , Infertility, Female/physiopathology , Inflammation , Obesity/metabolism , Obesity/physiopathology , Oxidative Stress , Pregnancy , Pregnancy Outcome , Receptor for Advanced Glycation End Products/physiology , Solubility
19.
Annu Rev Physiol ; 72: 219-46, 2010.
Article in English | MEDLINE | ID: mdl-20148674

ABSTRACT

Obesity induces an insulin-resistant state in adipose tissue, liver, and muscle and is a strong risk factor for the development of type 2 diabetes mellitus. Insulin resistance in the setting of obesity results from a combination of altered functions of insulin target cells and the accumulation of macrophages that secrete proinflammatory mediators. At the molecular level, insulin resistance is promoted by a transition in macrophage polarization from an alternative M2 activation state maintained by STAT6 and PPARs to a classical M1 activation state driven by NF-kappaB, AP1, and other signal-dependent transcription factors that play crucial roles in innate immunity. Strategies focused on inhibiting the inflammation/insulin resistance axis that otherwise preserve essential innate immune functions may hold promise for therapeutic intervention.


Subject(s)
Inflammation/pathology , Insulin Resistance/physiology , Macrophages/physiology , Adipose Tissue/pathology , Adipose Tissue/physiopathology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/physiology , Humans , Inflammation/genetics , Inflammation/physiopathology , Insulin Resistance/genetics , Liver/pathology , Macrophage Activation/physiology , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Obesity/physiopathology , PPAR gamma/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Toll-Like Receptor 4/physiology
20.
Ann Pharm Fr ; 72(6): 400-8, 2014 Nov.
Article in French | MEDLINE | ID: mdl-25438650

ABSTRACT

Advanced glycation end products (AGE) result from a chemical reaction between the carbonyl group of reducing sugar and the nucleophilic NH2 of a free amino acid or a protein; lysine and arginine being the main reactive amino acids on proteins. Following this first step, a molecular rearrangement occurs, rearrangement of Amadori resulting to the formation of Maillard products. Glycation can cause the clouding of the lens by inducing reactions crosslinking proteins. Specialized receptors (RAGE, Galectin 3…) bind AGE. The binding to the receptor causes the formation of free radicals, which have a deleterious effect because they are powerful oxidizing agents, but also play the role of intracellular messenger, altering the cell functions. This is especially true at the level of endothelial cells: the attachment of AGE to RAGE receptor causes an increase in vascular permeability. AGE binding to endothelium RAGE and to monocytes-macrophages, led to the production of cytokines, growth factors, to the expression of adhesion molecules, and the production of procoagulant activity. Diabetic retinopathy is related to excessive secretion of vascular growth factor (vascular endothelial growth factor [VEGF]). AGE-RAGE receptor binding causes the synthesis and secretion of VEGF. Increased permeability, facilitation of leukocyte migration, the production of reactive oxygen species, cytokines and VEGF suggest that the AGE could be an element of a cascade of reactions responsible for the diabetic angiopathy and vascular damages observed during aging and chronic renal failure. Balanced diet or some drugs can limit the deleterious effect of AGE.


Subject(s)
Glycation End Products, Advanced/metabolism , Glycation End Products, Advanced/genetics , Glycation End Products, Advanced/physiology , Humans , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL