Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(19)2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33019603

RESUMEN

We addressed the involvement of the receptor for advanced glycation end products (RAGE) in the impairment of the cellular cholesterol efflux elicited by glycated albumin. Albumin was isolated from type 1 (DM1) and type 2 (DM2) diabetes mellitus (HbA1c > 9%) and non-DM subjects (C). Moreover, albumin was glycated in vitro (AGE-albumin). Macrophages from Ager null and wild-type (WT) mice, or THP-1 transfected with siRNA-AGER, were treated with C, DM1, DM2, non-glycated or AGE-albumin. The cholesterol efflux was reduced in WT cells exposed to DM1 or DM2 albumin as compared to C, and the intracellular lipid content was increased. These events were not observed in Ager null cells, in which the cholesterol efflux and lipid staining were, respectively, higher and lower when compared to WT cells. In WT, Ager, Nox4 and Nfkb1, mRNA increased and Scd1 and Abcg1 diminished after treatment with DM1 and DM2 albumin. In Ager null cells treated with DM-albumin, Nox4, Scd1 and Nfkb1 were reduced and Jak2 and Abcg1 increased. In AGER-silenced THP-1, NOX4 and SCD1 mRNA were reduced and JAK2 and ABCG1 were increased even after treatment with AGE or DM-albumin. RAGE mediates the deleterious effects of AGE-albumin in macrophage cholesterol efflux.


Asunto(s)
HDL-Colesterol/sangre , LDL-Colesterol/sangre , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Macrófagos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/genética , Adulto , Animales , Estudios de Casos y Controles , Línea Celular , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Hemoglobina Glucada/genética , Hemoglobina Glucada/metabolismo , Productos Finales de Glicación Avanzada/sangre , Productos Finales de Glicación Avanzada/farmacología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/deficiencia , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Albúmina Sérica Humana/metabolismo , Albúmina Sérica Humana/farmacología , Células THP-1 , Triglicéridos/sangre
2.
JCI Insight ; 5(13)2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32641587

RESUMEN

Despite advances in lipid-lowering therapies, people with diabetes continue to experience more limited cardiovascular benefits. In diabetes, hyperglycemia sustains inflammation and preempts vascular repair. We tested the hypothesis that the receptor for advanced glycation end-products (RAGE) contributes to these maladaptive processes. We report that transplantation of aortic arches from diabetic, Western diet-fed Ldlr-/- mice into diabetic Ager-/- (Ager, the gene encoding RAGE) versus WT diabetic recipient mice accelerated regression of atherosclerosis. RNA-sequencing experiments traced RAGE-dependent mechanisms principally to the recipient macrophages and linked RAGE to interferon signaling. Specifically, deletion of Ager in the regressing diabetic plaques downregulated interferon regulatory factor 7 (Irf7) in macrophages. Immunohistochemistry studies colocalized IRF7 and macrophages in both murine and human atherosclerotic plaques. In bone marrow-derived macrophages (BMDMs), RAGE ligands upregulated expression of Irf7, and in BMDMs immersed in a cholesterol-rich environment, knockdown of Irf7 triggered a switch from pro- to antiinflammatory gene expression and regulated a host of genes linked to cholesterol efflux and homeostasis. Collectively, this work adds a new dimension to the immunometabolic sphere of perturbations that impair regression of established diabetic atherosclerosis and suggests that targeting RAGE and IRF7 may facilitate vascular repair in diabetes.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Inflamación/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Macrófagos/metabolismo , Animales , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 37(8): 1536-1547, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28642238

RESUMEN

OBJECTIVE: Diabetic subjects are at higher risk of ischemic peripheral vascular disease. We tested the hypothesis that advanced glycation end products (AGEs) and their receptor (RAGE) block angiogenesis and blood flow recovery after hindlimb ischemia induced by femoral artery ligation through modulation of immune/inflammatory mechanisms. APPROACH AND RESULTS: Wild-type mice rendered diabetic with streptozotocin and subjected to unilateral femoral artery ligation displayed increased accumulation and expression of AGEs and RAGE in ischemic muscle. In diabetic wild-type mice, femoral artery ligation attenuated angiogenesis and impaired blood flow recovery, in parallel with reduced macrophage content in ischemic muscle and suppression of early inflammatory gene expression, including Ccl2 (chemokine [C-C motif] ligand-2) and Egr1 (early growth response gene-1) versus nondiabetic mice. Deletion of Ager (gene encoding RAGE) or transgenic expression of Glo1 (reduces AGEs) restored adaptive inflammation, angiogenesis, and blood flow recovery in diabetic mice. In diabetes mellitus, deletion of Ager increased circulating Ly6Chi monocytes and augmented macrophage infiltration into ischemic muscle tissue after femoral artery ligation. In vitro, macrophages grown in high glucose display inflammation that is skewed to expression of tissue damage versus tissue repair gene expression. Further, macrophages grown in high versus low glucose demonstrate blunted macrophage-endothelial cell interactions. In both settings, these adverse effects of high glucose were reversed by Ager deletion in macrophages. CONCLUSIONS: These findings indicate that RAGE attenuates adaptive inflammation in hindlimb ischemia; underscore microenvironment-specific functions for RAGE in inflammation in tissue repair versus damage; and illustrate that AGE/RAGE antagonism may fill a critical gap in diabetic peripheral vascular disease.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Angiopatías Diabéticas/metabolismo , Eliminación de Gen , Inflamación/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Enfermedad Arterial Periférica/metabolismo , Receptor para Productos Finales de Glicación Avanzada/deficiencia , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Antígenos Ly/metabolismo , Velocidad del Flujo Sanguíneo , Glucemia/metabolismo , Comunicación Celular , Células Cultivadas , Microambiente Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/fisiopatología , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Productos Finales de Glicación Avanzada/metabolismo , Inflamación/genética , Inflamación/fisiopatología , Isquemia/genética , Isquemia/fisiopatología , Macrófagos/metabolismo , Ratones Noqueados , Ratones Transgénicos , Monocitos/metabolismo , Músculo Esquelético/metabolismo , Enfermedad Arterial Periférica/genética , Enfermedad Arterial Periférica/fisiopatología , Fenotipo , Receptor para Productos Finales de Glicación Avanzada/genética , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal , Estreptozocina , Factores de Tiempo
4.
Front Cell Neurosci ; 10: 117, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27242430

RESUMEN

The etiology of amyotrophic lateral sclerosis (ALS), a fatal motor neuron disorder characterized by progressive muscle weakness and spasticity, remains largely unknown. Approximately 5-10% of cases are familial, and of those, 15-20% are associated with mutations in the gene encoding Cu/Zn superoxide dismutase (SOD1). Mutations of the SOD1 gene interrupt cellular homeostasis and contribute to cellular toxicity evoked by the presence of altered SOD1, along with other toxic species, such as advanced glycation end products (AGEs). AGEs trigger activation of their chief cell surface receptor, RAGE (receptor for advanced glycation end products), and induce RAGE-dependent cellular stress and inflammation in neurons, thereby affecting their function and leading to apoptosis. Here, we show for the first time that the expression of RAGE is higher in the SOD1 transgenic mouse model of ALS vs. wild-type mouse spinal cord. We tested whether pharmacological blockade of RAGE may delay the onset and progression of disease in this mouse model. Our findings reveal that treatment of SOD1 transgenic mice with soluble RAGE (sRAGE), a natural competitor of RAGE that sequesters RAGE ligands and blocks their interaction with cell surface RAGE, significantly delays the progression of ALS and prolongs life span compared to vehicle treatment. We demonstrate that in sRAGE-treated SOD1 transgenic animals at the final stage of the disease, a significantly higher number of neurons and lower number of astrocytes is detectable in the spinal cord. We conclude that RAGE antagonism may provide a novel therapeutic strategy for ALS intervention.

5.
Protein Eng Des Sel ; 28(11): 501-6, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26243887

RESUMEN

The catalytic bioscavenger phosphotriesterase (PTE) is experimentally an effective antidote for organophosphate poisoning. We are interested in the molecular engineering of this enzyme to confer additional functionality, such as improved in vivo longevity. To this aim, we developed PTE cysteine mutants with free sulfhydryls to allow macromolecular attachments to the protein. A library of PTE cysteine mutants were assessed for efficiency in hydrolysing the toxic pesticide metabolite paraoxon, and screened for attachment with a sulfhydryl-reactive small molecule, fluorescein 5-maleimide (F5M), to examine cysteine availability. We established that the newly incorporated cysteines were readily available for labelling, with R90C, E116C and S291C displaying the highest affinity for binding with F5M. Next, we screened for efficiency in attaching a large macromolecule, a 30 000 Da polyethylene glycol (PEG) molecule. Using a solid-phase PEGylation strategy, we found the E116C mutant to be the best single-mutant candidate for attachment with PEG30. Kinetic activity of PEGylated E116C, with paraoxon as substrate, displayed activity approaching that of the unPEGylated wild-type. Our findings demonstrate, for the first time, an efficient cysteine mutation and subsequent method for sulfhydryl-specific macromolecule attachment to PTE.


Asunto(s)
Cisteína/química , Organofosfatos/metabolismo , Hidrolasas de Triéster Fosfórico/química , Hidrolasas de Triéster Fosfórico/metabolismo , Cisteína/genética , Cisteína/metabolismo , Fluoresceínas/química , Fluoresceínas/metabolismo , Cinética , Modelos Moleculares , Mutación , Organofosfatos/análisis , Paraoxon/análisis , Paraoxon/metabolismo , Hidrolasas de Triéster Fosfórico/genética , Ingeniería de Proteínas
6.
Diabetes ; 64(12): 4046-60, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26253613

RESUMEN

Diabetes exacerbates cardiovascular disease, at least in part through suppression of macrophage cholesterol efflux and levels of the cholesterol transporters ATP binding cassette transporter A1 (ABCA1) and ABCG1. The receptor for advanced glycation end products (RAGE) is highly expressed in human and murine diabetic atherosclerotic plaques, particularly in macrophages. We tested the hypothesis that RAGE suppresses macrophage cholesterol efflux and probed the mechanisms by which RAGE downregulates ABCA1 and ABCG1. Macrophage cholesterol efflux to apolipoprotein A1 and HDL and reverse cholesterol transport to plasma, liver, and feces were reduced in diabetic macrophages through RAGE. In vitro, RAGE ligands suppressed ABCG1 and ABCA1 promoter luciferase activity and transcription of ABCG1 and ABCA1 through peroxisome proliferator-activated receptor-γ (PPARG)-responsive promoter elements but not through liver X receptor elements. Plasma levels of HDL were reduced in diabetic mice in a RAGE-dependent manner. Laser capture microdissected CD68(+) macrophages from atherosclerotic plaques of Ldlr(-/-) mice devoid of Ager (RAGE) displayed higher levels of Abca1, Abcg1, and Pparg mRNA transcripts versus Ager-expressing Ldlr(-/-) mice independently of glycemia or plasma levels of total cholesterol and triglycerides. Antagonism of RAGE may fill an important therapeutic gap in the treatment of diabetic macrovascular complications.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Colesterol/metabolismo , Angiopatías Diabéticas/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Macrófagos/metabolismo , Receptor para Productos Finales de Glicación Avanzada/agonistas , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Transporte Biológico , Línea Celular , Células Cultivadas , Angiopatías Diabéticas/sangre , Angiopatías Diabéticas/inmunología , Angiopatías Diabéticas/patología , Productos Finales de Glicación Avanzada/sangre , Humanos , Ligandos , Lipoproteínas/antagonistas & inhibidores , Lipoproteínas/genética , Lipoproteínas/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones Noqueados , PPAR gamma/genética , PPAR gamma/metabolismo , Placa Aterosclerótica/sangre , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Regiones Promotoras Genéticas , Receptor para Productos Finales de Glicación Avanzada/sangre , Receptor para Productos Finales de Glicación Avanzada/genética , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
7.
Front Cell Neurosci ; 9: 485, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26733811

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disorder of largely unknown pathogenesis. Recent studies suggest that enhanced oxidative stress and neuroinflammation contribute to the progression of the disease. Mounting evidence implicates the receptor for advanced glycation end-products (RAGE) as a significant contributor to the pathogenesis of certain neurodegenerative diseases and chronic conditions. It is hypothesized that detrimental actions of RAGE are triggered upon binding to its ligands, such as AGEs (advanced glycation end products), S100/calgranulin family members, and High Mobility Group Box-1 (HMGB1) proteins. Here, we examined the expression of RAGE and its ligands in human ALS spinal cord. Tissue samples from age-matched human control and ALS spinal cords were tested for the expression of RAGE, carboxymethyllysine (CML) AGE, S100B, and HMGB1, and intensity of the immunofluorescent and immunoblotting signals was assessed. We found that the expression of both RAGE and its ligands was significantly increased in the spinal cords of ALS patients versus age-matched control subjects. Our study is the first report describing co-expression of both RAGE and its ligands in human ALS spinal cords. These findings suggest that further probing of RAGE as a mechanism of neurodegeneration in human ALS is rational.

8.
Int J Mol Sci ; 14(10): 19891-910, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24084731

RESUMEN

Oxidative stress is a central mechanism by which the receptor for advanced glycation endproducts (RAGE) mediates its pathological effects. Multiple experimental inquiries in RAGE-expressing cultured cells have demonstrated that ligand-RAGE interaction mediates generation of reactive oxygen species (ROS) and consequent downstream signal transduction and regulation of gene expression. The primary mechanism by which RAGE generates oxidative stress is via activation of NADPH oxidase; amplification mechanisms in the mitochondria may further drive ROS production. Recent studies indicating that the cytoplasmic domain of RAGE binds to the formin mDia1 provide further support for the critical roles of this pathway in oxidative stress; mDia1 was required for activation of rac1 and NADPH oxidase in primary murine aortic smooth muscle cells treated with RAGE ligand S100B. In vivo, in multiple distinct disease models in animals, RAGE action generates oxidative stress and modulates cellular/tissue fate in range of disorders, such as in myocardial ischemia, atherosclerosis, and aneurysm formation. Blockade or genetic deletion of RAGE was shown to be protective in these settings. Indeed, beyond cardiovascular disease, evidence is accruing in human subjects linking levels of RAGE ligands and soluble RAGE to oxidative stress in disorders such as doxorubicin toxicity, acetaminophen toxicity, neurodegeneration, hyperlipidemia, diabetes, preeclampsia, rheumatoid arthritis and pulmonary fibrosis. Blockade of RAGE signal transduction may be a key strategy for the prevention of the deleterious consequences of oxidative stress, particularly in chronic disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Productos Finales de Glicación Avanzada/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Animales , Humanos
9.
Neurosci Res ; 74(2): 144-55, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22902553

RESUMEN

Proteolipid protein (PLP) and its alternatively spliced isoform DM20 comprise ∼50% of central nervous system (CNS) myelin protein. The two proteins are identical in sequence except for the presence of a 35 amino sequence within the intracellular loop of PLP that is absent in DM20. In this work, we compared the expression of PLP/DM20 in transfected cells, oligodendrocytes and brain. In all 3 tissues, PLP exists as both a monomer and a disulfide-linked dimer; in contrast, DM20 is found mainly as a monomer. PLP dimers were increased by both chemical crosslinking and incubation with hydrogen peroxide, and were mediated by a cysteine at amino acid 108, located within the proximal intracellular loop of both PLP and DM20. The PLP-specific sequence thus influences the accessibility of this cysteine to chemical modification, perhaps as a result of altering protein structure. Consistent with these findings, several mutant PLPs known to cause Pelizaeus-Merzbacher disease form predominantly disulfide-linked, high molecular weight aggregates in transfected COS7 cells that are arrested in the ER and are associated with increased expression of CHOP, a part of the cellular response to unfolded proteins. In contrast, the same mutations in DM20 accumulate fewer high molecular weight disulfide-linked species that are expressed at the cell surface, and are not associated with increased CHOP. Taken together, these data suggest that mutant PLP multimerization, mediated in part by way of cysteine 108, may be part of the pathogenesis of Pelizaeus-Merzbacher disease.


Asunto(s)
Cisteína/química , Proteína Proteolipídica de la Mielina/química , Enfermedad de Pelizaeus-Merzbacher/genética , Secuencia de Aminoácidos , Animales , Encéfalo/metabolismo , Células COS , Membrana Celular/metabolismo , Células Cultivadas/metabolismo , Chlorocebus aethiops , Reactivos de Enlaces Cruzados/farmacología , Cistina/química , Dimerización , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/metabolismo , Conformación Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Factor de Transcripción CHOP/biosíntesis , Factor de Transcripción CHOP/genética , Transfección , Respuesta de Proteína Desplegada
10.
Circ Res ; 110(10): 1279-93, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22511750

RESUMEN

RATIONALE: The mammalian diaphanous-related formin (mDia1), governs microtubule and microfilament dynamics while functioning as an effector for Rho small GTP-binding proteins during key cellular processes such as adhesion, cytokinesis, cell polarity, and morphogenesis. The cytoplasmic domain of the receptor for advanced glycation endproducts binds to the formin homology 1 domain of mDia1; mDia1 is required for receptor for advanced glycation endproducts ligand-induced cellular migration in transformed cells. OBJECTIVE: Because a key mechanism in vascular remodeling is the induction of smooth muscle cell migration, we tested the role of mDia1 in this process. METHODS AND RESULTS: We report that endothelial denudation injury to the murine femoral artery significantly upregulates mDia1 mRNA transcripts and protein in the injured vessel, particularly in vascular smooth muscle cells within the expanding neointima. Loss of mDia1 expression significantly reduces pathological neointimal expansion consequent to injury. In primary murine aortic smooth muscle cells, mDia1 is required for receptor for advanced glycation endproducts ligand-induced membrane translocation of c-Src, which leads to Rac1 activation, redox phosphorylation of AKT/glycogen synthase kinase 3ß, and consequent smooth muscle cell migration. CONCLUSIONS: We conclude that mDia1 integrates oxidative and signal transduction pathways triggered, at least in part, by receptor for advanced glycation endproducts ligands, thereby regulating pathological neointimal expansion.


Asunto(s)
Proteínas Portadoras/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/patología , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Citoesqueleto de Actina/fisiología , Animales , Proteínas Portadoras/genética , Movimiento Celular/fisiología , Células Cultivadas , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Arteria Femoral/patología , Forminas , Productos Finales de Glicación Avanzada/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Microtúbulos/fisiología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , Neointima/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...