Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Brain ; 147(5): 1710-1725, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38146639

ABSTRACT

Mitochondrial dysfunction is an early pathological feature of Alzheimer disease and plays a crucial role in the development and progression of Alzheimer's disease. Strategies to rescue mitochondrial function and cognition remain to be explored. Cyclophilin D (CypD), the peptidylprolyl isomerase F (PPIase), is a key component in opening the mitochondrial membrane permeability transition pore, leading to mitochondrial dysfunction and cell death. Blocking membrane permeability transition pore opening by inhibiting CypD activity is a promising therapeutic approach for Alzheimer's disease. However, there is currently no effective CypD inhibitor for Alzheimer's disease, with previous candidates demonstrating high toxicity, poor ability to cross the blood-brain barrier, compromised biocompatibility and low selectivity. Here, we report a new class of non-toxic and biocompatible CypD inhibitor, ebselen, using a conventional PPIase assay to screen a library of ∼2000 FDA-approved drugs with crystallographic analysis of the CypD-ebselen crystal structure (PDB code: 8EJX). More importantly, we assessed the effects of genetic and pharmacological blockade of CypD on Alzheimer's disease mitochondrial and glycolytic bioenergetics in Alzheimer's disease-derived mitochondrial cybrid cells, an ex vivo human sporadic Alzheimer's disease mitochondrial model, and on synaptic function, inflammatory response and learning and memory in Alzheimer's disease mouse models. Inhibition of CypD by ebselen protects against sporadic Alzheimer's disease- and amyloid-ß-induced mitochondrial and glycolytic perturbation, synaptic and cognitive dysfunction, together with suppressing neuroinflammation in the brain of Alzheimer's disease mouse models, which is linked to CypD-related membrane permeability transition pore formation. Thus, CypD inhibitors have the potential to slow the progression of neurodegenerative diseases, including Alzheimer's disease, by boosting mitochondrial bioenergetics and improving synaptic and cognitive function.


Subject(s)
Alzheimer Disease , Isoindoles , Mitochondria , Organoselenium Compounds , Peptidyl-Prolyl Isomerase F , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Peptidyl-Prolyl Isomerase F/metabolism , Animals , Mitochondria/drug effects , Mitochondria/metabolism , Mice , Humans , Cognition/drug effects , Azoles/pharmacology , Azoles/therapeutic use , Cyclophilins/metabolism , Cyclophilins/antagonists & inhibitors , Mice, Transgenic , Mice, Inbred C57BL , Male , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use
2.
Aging Cell ; 20(5): e13368, 2021 05.
Article in English | MEDLINE | ID: mdl-33951271

ABSTRACT

Mitochondrial dysfunction is one of the early pathological features of Alzheimer's disease (AD). Accumulation of cerebral and mitochondrial Aß links to mitochondrial and synaptic toxicity. We have previously demonstrated the mechanism by which presequence peptidase (PITRM1)-mediated clearance of mitochondrial Aß contributes to mitochondrial and cerebral amyloid pathology and mitochondrial and synaptic stress in adult transgenic AD mice overexpressing Aß up to 12 months old. Here, we investigate the effect of PITRM1 in an advanced age AD mouse model (up to 19-24 months) to address the fundamental unexplored question of whether restoration/gain of PITRM1 function protects against mitochondrial and synaptic dysfunction associated with Aß accumulation and whether this protection is maintained even at later ages featuring profound amyloid pathology and synaptic failure. Using newly developed aged PITRM1/Aß-producing AD mice, we first uncovered reduction in PITRM1 expression in AD-affected cortex of AD mice at 19-24 months of age. Increasing neuronal PITRM1 activity/expression re-established mitochondrial respiration, suppressed reactive oxygen species, improved synaptic function, and reduced loss of synapses even at advanced ages (up to 19-24 months). Notably, loss of PITRM1 proteolytic activity resulted in Aß accumulation and failure to rescue mitochondrial and synaptic function, suggesting that PITRM1 activity is required for the degradation and clearance of mitochondrial Aß and Aß deposition. These data indicate that augmenting PITRM1 function results in persistent life-long protection against Aß toxicity in an AD mouse model. Therefore, augmenting PITRM1 function may enhance Aß clearance in mitochondria, thereby maintaining mitochondrial integrity and ultimately slowing the progression of AD.


Subject(s)
Alzheimer Disease/enzymology , Metalloendopeptidases/metabolism , Mitochondria/enzymology , Neurons/enzymology , Synapses/metabolism , Aging , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/metabolism , Cognition , Disease Models, Animal , Female , Inflammation , Male , Metalloendopeptidases/genetics , Mice, Transgenic , Mitochondria/metabolism , Neurons/metabolism , Synapses/physiology
3.
Free Radic Biol Med ; 164: 429-438, 2021 02 20.
Article in English | MEDLINE | ID: mdl-33359687

ABSTRACT

Aging is a strong risk factor for brain dementia and cognitive decline. Age-related accumulation of metabolites such as advanced glycation end products (AGEs) could serve as danger signals to initiate and accelerate disease process and neurodegeneration. The underlying causes and consequences of cerebral AGEs accumulation remain largely unknown. Here, we comprehensively investigate age-related accumulation of AGEs and dicarbonyls, including methylglyoxal (MG), glyoxal (GO), and 3-deoxyglucosone (3-DG), and the effects of mitochondrial reactive oxygen species (ROS) on cerebral AGEs accumulation, mitochondrial function, and oxidative stress in the aging human and mouse brain. We demonstrate that AGEs, including arginine and lysine derived N(6)-carboxymethyl lysine (CML), Nε-(1-Carboxyethyl)-l-lysine (CEL), and methylglyoxal-derived hydroimidazolone-1 (MG-H1), were significantly elevated in the cerebral cortex and hippocampus with advanced age in mice. Accordingly, aging mouse and human brains revealed decrease in activities of mitochondrial respiratory chain complexes I & IV and ATP levels, and increased ROS. Notably, administration of mitoTEMPO (2-(2,2,6,6-Tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mTEMPO), a scavenger of mitochondrial ROS, not only suppressed ROS production but also reduced aged-induced accumulation of AGEs and dicarbonyls. mTEMPO treatment improved mitochondrial respiratory function and restored ATP levels. Our findings provide evidence linking age-related accumulation of toxic metabolites (AGEs) to mitochondrial oxidative stress. This highlights a novel mechanism by which AGEs-dependent signaling promotes carbonyl stress and sustained mitochondrial dysfunction. Eliminating formation and accumulation of AGEs may represent a new therapeutic avenue for combating cognitive decline and mitochondrial degeneration relevant to aging and neurodegenerative diseases including Alzheimer's disease.


Subject(s)
Glycation End Products, Advanced , Mitochondria , Animals , Arginine , Mice , Pyruvaldehyde , Reactive Oxygen Species
4.
J Alzheimers Dis ; 76(1): 165-178, 2020.
Article in English | MEDLINE | ID: mdl-32444539

ABSTRACT

BACKGROUND: Advanced glycation end products (AGEs) are an important risk factor for the development of cognitive decline in aging and late-onset neurodegenerative diseases including Alzheimer's disease. However, whether and how dietary AGEs exacerbate cognitive impairment and brain mitochondrial dysfunction in the aging process remains largely unknown. OBJECTIVE: We investigated the direct effects of dietary AGEs on AGE adducts accumulation, mitochondrial function, and cognitive performance in mice. METHODS: Mice were fed the AGE+ diet or AGE- diet. We examined levels of AGE adducts in serum and cerebral cortexes by immunodetection and immunohistochemistry, determined levels of reactive oxygen species by biochemical analysis, detected enzyme activity associated with mitochondrial respiratory chain complexes I & IV and ATP levels, and assessed learning and memory ability by Morris Water Maze and nesting behavior. RESULTS: Levels of AGE adducts (MG-H1 and CEL) were robustly increased in the serum and brain of AGE+ diet fed mice compared to the AGE- group. Furthermore, greatly elevated levels of reactive oxygen species, decreased activities of mitochondrial respiratory chain complexes I & IV, reduced ATP levels, and impaired learning and memory were evident in AGE+ diet fed mice compared to the AGE- group. CONCLUSION: These results indicate that dietary AGEs are important sources of AGE accumulation in vivo, resulting in mitochondrial dysfunction, impairment of energy metabolism, and subsequent cognitive impairment. Thus, reducing AGEs intake to lower accumulation of AGEs could hold therapeutic potential for the prevention and treatment of AGEs-induced mitochondrial dysfunction linked to cognitive decline.


Subject(s)
Cognition/physiology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Diet/adverse effects , Glycation End Products, Advanced/toxicity , Mitochondria/metabolism , Animals , Cognition/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Female , Glycation End Products, Advanced/administration & dosage , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Reactive Oxygen Species/metabolism
5.
Nat Commun ; 9(1): 2968, 2018 07 30.
Article in English | MEDLINE | ID: mdl-30061577

ABSTRACT

Endophilin A1 (EP) is a protein enriched in synaptic terminals that has been linked to Alzheimer's disease (AD). Previous in vitro studies have shown that EP can bind to a variety of proteins, which elicit changes in synaptic transmission of neurotransmitters and spine formation. Additionally, we previously showed that EP protein levels are elevated in AD patients and AD transgenic animal models. Here, we establish the in vivo consequences of upregulation of EP expression in amyloid-ß peptide (Aß)-rich environments, leading to changes in both long-term potentiation and learning and memory of transgenic animals. Specifically, increasing EP augmented cerebral Aß accumulation. EP-mediated signal transduction via reactive oxygen species (ROS)/p38 mitogen-activated protein (MAP) kinase contributes to Aß-induced mitochondrial dysfunction, synaptic injury, and cognitive decline, which could be rescued by blocking either ROS or p38 MAP kinase activity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Gene Expression Regulation , Adenosine Triphosphate/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Genetically Modified , Antioxidants/metabolism , Crosses, Genetic , Disease Models, Animal , Hippocampus/metabolism , Humans , Long-Term Potentiation , Mice , Mice, Transgenic , Mitochondria/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Peptide Fragments/metabolism , Reactive Oxygen Species/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Drug Discov Today ; 23(12): 1983-1989, 2018 12.
Article in English | MEDLINE | ID: mdl-30081095

ABSTRACT

The mitochondrial permeability transition pore (mPTP) has been considered a key contributor to cell death, inducing the process in several major neurodegenerative diseases. To date, the molecular nature of the mPTP remains confounding but its significance is universally acknowledged. Several targets have been screened and inhibition of mPTP has emerged as an attractive field for researchers. Nowadays, in silico-directed studies help to explore new small molecules targeting the mPTP to improve their drug-like properties and bioactivity. Here, we briefly summarize the role of mPTP in neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson disease (PD), and Huntington's disease (HD), and discusses current and future potential therapeutic targets.


Subject(s)
Mitochondrial Membrane Transport Proteins/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Animals , Cell Death/drug effects , Humans , Mitochondrial Permeability Transition Pore
7.
Methods Mol Biol ; 1779: 415-433, 2018.
Article in English | MEDLINE | ID: mdl-29886547

ABSTRACT

Mitochondrial and synaptic dysfunction is an early pathological feature of Alzheimer's disease (AD). Accumulation of amyloid beta-peptide (Aß) in mitochondria, particularly in synaptic mitochondria, potentiates and amplifies synaptic injury and disruption of synaptic transmission, leading to synaptic dysfunction and ultimately to synaptic failure. Thus, determination of the presence and levels of Aß in synaptic mitochondria associated with amyloid pathology is important for studying mitochondrial amyloid pathology. Here, we present a detailed methodology for the isolation of synaptic mitochondria from brain tissues and the determination of Aß levels in the isolated mitochondria as well as ultrastructural localization of synaptic mitochondrial Aß. These methods have been used successfully for the identification and characterization of Aß accumulation in synaptic mitochondria from mouse brains derived from transgenic AD mouse model. Additionally, we comprehensively discuss the sample preparation, experimental details, our unique procedures, optimization of parameters, and troubleshooting.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/isolation & purification , Brain/cytology , Mitochondria/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Centrifugation, Density Gradient , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Mitochondria/pathology , Mitochondria/ultrastructure , Synapses/metabolism , Synapses/pathology , Synapses/ultrastructure
8.
Hum Mol Genet ; 27(6): 1002-1014, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29329433

ABSTRACT

Receptor for Advanced Glycation End products (RAGE) has been implicated in amyloid ß-peptide (Aß)-induced perturbation relevant to the pathogenesis of Alzheimer's disease (AD). However, whether and how RAGE regulates Aß metabolism remains largely unknown. Aß formation arises from aberrant cleavage of amyloid pre-cursor protein (APP) by ß- and γ-secretase. To investigate whether RAGE modulates ß- and γ-secretase activity potentiating Aß formation, we generated mAPP mice with genetic deletion of RAGE (mAPP/RO). These mice displayed reduced cerebral amyloid pathology, inhibited aberrant APP-Aß metabolism by reducing ß- and γ-secretases activity, and attenuated impairment of learning and memory compared with mAPP mice. Similarly, RAGE signal transduction deficient mAPP mice (mAPP/DN-RAGE) exhibited the reduction in Aß40 and Aß42 production and decreased ß-and γ-secretase activity compared with mAPP mice. Furthermore, RAGE-deficient mAPP brain revealed suppression of activation of p38 MAP kinase and glycogen synthase kinase 3ß (GSK3ß). Finally, RAGE siRNA-mediated gene silencing or DN-RAGE-mediated signaling deficiency in the enriched human APP neuronal cells demonstrated suppression of activation of GSK3ß, accompanied with reduction in Aß levels and decrease in ß- and γ-secretases activity. Our findings highlight that RAGE-dependent signaling pathway regulates ß- and γ-secretase cleavage of APP to generate Aß, at least in part through activation of GSK3ß and p38 MAP kinase. RAGE is a potential therapeutic target to limit aberrant APP-Aß metabolism in halting progression of AD.


Subject(s)
Alzheimer Disease/metabolism , Receptor for Advanced Glycation End Products/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Memory/drug effects , Mice , Mice, Transgenic , Neurons/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Brain ; 140(12): 3233-3251, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29077793

ABSTRACT

Mitochondrial dysfunction and synaptic damage are early pathological features of the Alzheimer's disease-affected brain. Memory impairment in Alzheimer's disease is a manifestation of brain pathologies such as accumulation of amyloid-ß peptide and mitochondrial damage. The underlying pathogenic mechanisms and effective disease-modifying therapies for Alzheimer's disease remain elusive. Here, we demonstrate for the first time that decreased PTEN-induced putative kinase 1 (PINK1) expression is associated with Alzheimer's disease pathology. Restoring neuronal PINK1 function strikingly reduces amyloid-ß levels, amyloid-associated pathology, oxidative stress, as well as mitochondrial and synaptic dysfunction. In contrast, PINK1-deficient mAPP mice augmented cerebral amyloid-ß accumulation, mitochondrial abnormalities, impairments in learning and memory, as well as synaptic plasticity at an earlier age than mAPP mice. Notably, gene therapy-mediated PINK1 overexpression promotes the clearance of damaged mitochondria by augmenting autophagy signalling via activation of autophagy receptors (OPTN and NDP52), thereby alleviating amyloid-ß-induced loss of synapses and cognitive decline in Alzheimer's disease mice. Loss of PINK1 activity or blockade of PINK1-mediated signalling (OPTN or NDP52) fails to reverse amyloid-ß-induced detrimental effects. Our findings highlight a novel mechanism by which PINK1-dependent signalling promotes the rescue of amyloid pathology and amyloid-ß-mediated mitochondrial and synaptic dysfunctions in a manner requiring activation of autophagy receptor OPTN or NDP52. Thus, activation of PINK1 may represent a new therapeutic avenue for combating Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Protein Kinases/metabolism , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Autophagy , Brain/metabolism , Cell Cycle Proteins , Eye Proteins/metabolism , Female , Genetic Therapy , Humans , Male , Membrane Transport Proteins , Mice, Transgenic , Middle Aged , Nerve Tissue Proteins/metabolism , Oxidative Stress , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction
10.
J Alzheimers Dis ; 59(1): 223-239, 2017.
Article in English | MEDLINE | ID: mdl-28598851

ABSTRACT

Loss of synapse and synaptic dysfunction contribute importantly to cognitive impairment in Alzheimer's disease (AD). Mitochondrial dysfunction and oxidative stress are early pathological features in AD-affected brain. However, the effect of AD mitochondria on synaptogenesis remains to be determined. Using human trans-mitochondrial "cybrid" (cytoplasmic hybrid) neuronal cells whose mitochondria were transferred from platelets of patients with sporadic AD or age-matched non-AD subjects with relatively normal cognition, we provide the first evidence of mitochondrial dysfunction compromises synaptic development and formation of synapse in AD cybrid cells in response to chemical-induced neuronal differentiation. Compared to non-AD control cybrids, AD cybrid cells showed synaptic loss which was evidenced by a significant reduction in expression of two synaptic marker proteins: synaptophysin (presynaptic marker) and postsynaptic density protein-95, and neuronal proteins (MAP-2 and NeuN) upon neuronal differentiation. In parallel, AD-mediated synaptic deficits correlate to mitochondrial dysfunction and oxidative stress as well as activation of p38 MAP kinase. Notably, inhibition of p38 MAP kinase by pharmacological specific p38 inhibitor significantly increased synaptic density, improved mitochondrial function, and reduced oxidative stress. These results suggest that activation of p38 MAP kinase signaling pathway contributes to AD-mediated impairment in neurogenesis, possibly by inhibiting the neuronal differentiation. Our results provide new insight into the crosstalk of dysfunctional AD mitochondria to synaptic formation and maturation via activation of p38 MAP kinase. Therefore, blockade of p38 MAP kinase signal transduction could be a potential therapeutic strategy for AD by alleviating loss of synapses.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/pathology , Mitochondria/pathology , Mitochondrial Diseases/etiology , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation , Disks Large Homolog 4 Protein , Electron Transport Complex IV/metabolism , Female , Humans , Hybrid Cells , Male , Membrane Potential, Mitochondrial , Mitochondria/ultrastructure , Mitochondrial Diseases/pathology , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Rhodamines/metabolism , Synapses/metabolism , Synapses/pathology , Synaptophysin/metabolism
11.
Arterioscler Thromb Vasc Biol ; 37(8): 1536-1547, 2017 08.
Article in English | MEDLINE | ID: mdl-28642238

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/metabolism , Gene Deletion , Inflammation/metabolism , Ischemia/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Peripheral Arterial Disease/metabolism , Receptor for Advanced Glycation End Products/deficiency , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Antigens, Ly/metabolism , Blood Flow Velocity , Blood Glucose/metabolism , Cell Communication , Cells, Cultured , Cellular Microenvironment , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/physiopathology , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Glycation End Products, Advanced/metabolism , Inflammation/genetics , Inflammation/physiopathology , Ischemia/genetics , Ischemia/physiopathology , Macrophages/metabolism , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism , Muscle, Skeletal/metabolism , Peripheral Arterial Disease/genetics , Peripheral Arterial Disease/physiopathology , Phenotype , Receptor for Advanced Glycation End Products/genetics , Recovery of Function , Regional Blood Flow , Signal Transduction , Streptozocin , Time Factors
12.
Diabetes ; 63(6): 1948-65, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24520121

ABSTRACT

In mammals, changes in the metabolic state, including obesity, fasting, cold challenge, and high-fat diets (HFDs), activate complex immune responses. In many strains of rodents, HFDs induce a rapid systemic inflammatory response and lead to obesity. Little is known about the molecular signals required for HFD-induced phenotypes. We studied the function of the receptor for advanced glycation end products (RAGE) in the development of phenotypes associated with high-fat feeding in mice. RAGE is highly expressed on immune cells, including macrophages. We found that high-fat feeding induced expression of RAGE ligand HMGB1 and carboxymethyllysine-advanced glycation end product epitopes in liver and adipose tissue. Genetic deficiency of RAGE prevented the effects of HFD on energy expenditure, weight gain, adipose tissue inflammation, and insulin resistance. RAGE deficiency had no effect on genetic forms of obesity caused by impaired melanocortin signaling. Hematopoietic deficiency of RAGE or treatment with soluble RAGE partially protected against peripheral HFD-induced inflammation and weight gain. These findings demonstrate that high-fat feeding induces peripheral inflammation and weight gain in a RAGE-dependent manner, providing a foothold in the pathways that regulate diet-induced obesity and offering the potential for therapeutic intervention.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat , Inflammation/metabolism , Insulin Resistance , Liver/metabolism , Obesity/metabolism , Receptors, Immunologic/metabolism , Animals , Glucose Clamp Technique , Inflammation/genetics , Insulin Resistance/genetics , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Real-Time Polymerase Chain Reaction , Receptor for Advanced Glycation End Products , Weight Gain/genetics
13.
Diabetes ; 63(2): 761-74, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24186862

ABSTRACT

Sustained increases in glucose flux via the aldose reductase (AR) pathway have been linked to diabetic vascular complications. Previous studies revealed that glucose flux via AR mediates endothelial dysfunction and leads to lesional hemorrhage in diabetic human AR (hAR) expressing mice in an apoE(-/-) background. Our studies revealed sustained activation of Egr-1 with subsequent induction of its downstream target genes tissue factor (TF) and vascular cell adhesion molecule-1 (VCAM-1) in diabetic apoE(-/-)hAR mice aortas and in high glucose-treated primary murine aortic endothelial cells expressing hAR. Furthermore, we observed that flux via AR impaired NAD(+) homeostasis and reduced activity of NAD(+)-dependent deacetylase Sirt-1 leading to acetylation and prolonged expression of Egr-1 in hyperglycemic conditions. In conclusion, our data demonstrate a novel mechanism by which glucose flux via AR triggers activation, acetylation, and prolonged expression of Egr-1 leading to proinflammatory and prothrombotic responses in diabetic atherosclerosis.


Subject(s)
Aldehyde Reductase/metabolism , Early Growth Response Protein 1/metabolism , Gene Expression Regulation/physiology , Hyperglycemia/metabolism , Aldehyde Reductase/genetics , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Early Growth Response Protein 1/genetics , Endothelial Cells/physiology , Glucose/pharmacology , Humans , Mice , Mice, Transgenic , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
14.
Arterioscler Thromb Vasc Biol ; 33(8): 1779-87, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23766264

ABSTRACT

OBJECTIVE: Subjects with diabetes mellitus are at high risk for developing atherosclerosis through a variety of mechanisms. Because the metabolism of glucose results in production of activators of protein kinase C (PKC)ß, it was logical to investigate the role of PKCß in modulation of atherosclerosis in diabetes mellitus. APPROACH AND RESULTS: ApoE(-/-) and PKCß(-/-)/ApoE(-/-) mice were rendered diabetic with streptozotocin. Quantification of atherosclerosis, gene expression profiling, or analysis of signaling molecules was performed on aortic sinus or aortas from diabetic mice. Diabetes mellitus-accelerated atherosclerosis increased the level of phosphorylated extracellular signal-regulated kinase 1/2 and Jun-N-terminus kinase mitogen-activated protein kinases and augmented vascular expression of inflammatory mediators, as well as increased monocyte/macrophage infiltration and CD11c(+) cells accumulation in diabetic ApoE(-/-) mice, processes that were diminished in diabetic PKCß(-/-)/ApoE(-/-) mice. In addition, pharmacological inhibition of PKCß reduced atherosclerotic lesion size in diabetic ApoE(-/-) mice. In vitro, the inhibitors of PKCß and extracellular signal-regulated kinase 1/2, as well as small interfering RNA to Egr-1, significantly decreased high-glucose-induced expression of CD11c (integrin, alpha X 9 complement component 3 receptor 4 subunit]), chemokine (C-C motif) ligand 2, and interleukin-1ß in U937 macrophages. CONCLUSIONS: These data link enhanced activation of PKCß to accelerated diabetic atherosclerosis via a mechanism that includes modulation of gene transcription and signal transduction in the vascular wall, processes that contribute to acceleration of vascular inflammation and atherosclerosis in diabetes mellitus. Our results uncover a novel role for PKCß in modulating CD11c expression and inflammatory response of macrophages in the development of diabetic atherosclerosis. These findings support PKCß activation as a potential therapeutic target for prevention and treatment of diabetic atherosclerosis.


Subject(s)
Apolipoproteins E/immunology , Atherosclerosis/immunology , Diabetes Mellitus, Experimental/immunology , Protein Kinase C/immunology , Vasculitis/immunology , Animals , Aortitis/immunology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , CD11c Antigen/metabolism , Diabetic Angiopathies/genetics , Diabetic Angiopathies/immunology , Diabetic Angiopathies/metabolism , Disease Models, Animal , Gene Expression/immunology , Humans , Hyperglycemia/genetics , Hyperglycemia/immunology , Hyperglycemia/metabolism , Hyperlipidemias/genetics , Hyperlipidemias/immunology , Hyperlipidemias/metabolism , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/immunology , Protein Kinase C/genetics , Protein Kinase C beta , Signal Transduction/immunology , U937 Cells , Vasculitis/genetics , Vasculitis/metabolism
15.
Diabetes ; 62(3): 931-43, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23172920

ABSTRACT

Peripheral neuropathy and insensate limbs and digits cause significant morbidity in diabetic individuals. Previous studies showed that deletion of the receptor for advanced end-glycation products (RAGE) in mice was protective in long-term diabetic neuropathy. Here, we tested the hypothesis that RAGE suppresses effective axonal regeneration in superimposed acute peripheral nerve injury attributable to tissue-damaging inflammatory responses. We report that deletion of RAGE, particularly in diabetic mice, resulted in significantly higher myelinated fiber densities and conduction velocities consequent to acute sciatic nerve crush compared with wild-type control animals. Consistent with key roles for RAGE-dependent inflammation, reconstitution of diabetic wild-type mice with RAGE-null versus wild-type bone marrow resulted in significantly improved axonal regeneration and restoration of function. Diabetic RAGE-null mice displayed higher numbers of invading macrophages in the nerve segments postcrush compared with wild-type animals, and these macrophages in diabetic RAGE-null mice displayed greater M2 polarization. In vitro, treatment of wild-type bone marrow-derived macrophages with advanced glycation end products (AGEs), which accumulate in diabetic nerve tissue, increased M1 and decreased M2 gene expression in a RAGE-dependent manner. Blockade of RAGE may be beneficial in the acute complications of diabetic neuropathy, at least in part, via upregulation of regeneration signals.


Subject(s)
Diabetes Mellitus, Type 1/complications , Diabetic Neuropathies/metabolism , Nerve Regeneration , Receptors, Immunologic/metabolism , Sciatic Nerve/physiopathology , Sciatic Neuropathy/metabolism , Animals , Bone Marrow Transplantation , Cells, Cultured , Diabetic Neuropathies/immunology , Diabetic Neuropathies/pathology , Diabetic Neuropathies/prevention & control , Glycation End Products, Advanced/metabolism , Immunohistochemistry , Ligands , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Macrophages/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Crush/adverse effects , Neural Conduction , Organ Specificity , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Sciatic Nerve/immunology , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Sciatic Neuropathy/immunology , Sciatic Neuropathy/pathology , Sciatic Neuropathy/prevention & control
16.
Vascul Pharmacol ; 57(5-6): 160-7, 2012.
Article in English | MEDLINE | ID: mdl-22750165

ABSTRACT

The multi-ligand receptor RAGE was discovered on account of its ability to bind and transduce the cell stress-provoking signals of advanced glycation endproducts (AGEs). The finding that RAGE also bound pro-inflammatory molecules set the stage for linking RAGE and inflammation to the pathogenesis of diabetic macro- and microvascular complications. In this review, we focus on the roles of RAGE and its ligands in diabetes complications. We recount the findings from mice, rats, swine and human subjects suggesting that RAGE action potently contributes to vascular, inflammatory and end-organ stress and damage in types 1 and 2 diabetes. We detail the efforts to track ligands and RAGE in human subjects with diabetes to address if this axis may be a biomarker reflective of the state of the diabetic complications. Lastly, we suggest specific strategies to tackle AGE-ligand-RAGE interactions as potential therapeutic targets for diabetes and its complications.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Glycation End Products, Advanced/metabolism , Receptors, Immunologic/metabolism , Animals , Diabetes Complications/physiopathology , Diabetes Complications/therapy , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/therapy , Humans , Inflammation/physiopathology , Mice , Rats , Receptor for Advanced Glycation End Products , Signal Transduction , Swine
17.
Circ Res ; 110(10): 1279-93, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22511750

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Neointima/pathology , Oxidative Stress/physiology , Signal Transduction/physiology , Actin Cytoskeleton/physiology , Animals , Carrier Proteins/genetics , Cell Movement/physiology , Cells, Cultured , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , Formins , Glycation End Products, Advanced/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microtubules/physiology , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , Neointima/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism
18.
Amino Acids ; 42(4): 1151-61, 2012 Apr.
Article in English | MEDLINE | ID: mdl-20957395

ABSTRACT

The formation of advanced glycation endproducts (AGEs) occurs in diverse settings such as diabetes, aging, renal failure, inflammation and hypoxia. The chief cellular receptor for AGEs, RAGE, transduces the effects of AGEs via signal transduction, at least in part via processes requiring the RAGE cytoplasmic domain binding partner, diaphanous-1 or mDia1. Data suggest that RAGE perpetuates the inflammatory signals initiated by AGEs via multiple mechanisms. AGE-RAGE interaction stimulates generation of reactive oxygen species and inflammation--mechanisms which enhance AGE formation. Further, recent data in type 1 diabetic kidney reveal that deletion of RAGE prevents methylglyoxal accumulation, at least in part via RAGE-dependent regulation of glyoxalase-1, a major enzyme involved in methylglyoxal detoxification. Taken together, these considerations place RAGE in the center of biochemical and molecular stresses that characterize the complications of diabetes and chronic disease. Stopping RAGE-dependent signaling may hold the key to interrupting cycles of cellular perturbation and tissue damage in these disorders.


Subject(s)
Glycation End Products, Advanced/metabolism , Receptors, Immunologic/metabolism , Animals , Disease/etiology , Glycosylation , Humans , Receptor for Advanced Glycation End Products , Signal Transduction
19.
J Biol Chem ; 287(7): 5133-44, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22194616

ABSTRACT

The receptor for advanced glycation end products (RAGE) is a multiligand cell surface macromolecule that plays a central role in the etiology of diabetes complications, inflammation, and neurodegeneration. The cytoplasmic domain of RAGE (C-terminal RAGE; ctRAGE) is critical for RAGE-dependent signal transduction. As the most membrane-proximal event, mDia1 binds to ctRAGE, and it is essential for RAGE ligand-stimulated phosphorylation of AKT and cell proliferation/migration. We show that ctRAGE contains an unusual α-turn that mediates the mDia1-ctRAGE interaction and is required for RAGE-dependent signaling. The results establish a novel mechanism through which an extracellular signal initiated by RAGE ligands regulates RAGE signaling in a manner requiring mDia1.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Diabetes Complications/genetics , Diabetes Complications/metabolism , Formins , Humans , Inflammation/genetics , Inflammation/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Phosphorylation/physiology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor for Advanced Glycation End Products , Receptors, Immunologic/chemistry , Receptors, Immunologic/genetics
20.
J Gastrointest Surg ; 16(1): 104-12; discussion 112, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22052106

ABSTRACT

BACKGROUND: The receptor for advanced glycation end-products (RAGE) is implicated in pancreatic tumorigenesis. Activating Kras mutations and p16 inactivation are genetic abnormalities most commonly detected as pancreatic ductal epithelium progresses from intraepithelial neoplasia (PanIN) to adenocarcinoma (PDAC). OBJECTIVE: The aim of this study was to evaluate the effect of RAGE (or AGER) deletion on the development of PanIN and PDAC in conditional Kras ( G12D ) mice. MATERIALS AND METHODS: Pdx1-Cre; LSL-Kras ( G12D/+) mice were crossed with RAGE (-/-) mice to generate Pdx1-Cre; LSL-Kras ( G12D/+) ; RAGE (-/-) mice. Pdx1-Cre; LSL-Kras ( G12D/+); p16 ( Ink4a-/-) mice were crossed with RAGE (-/-) mice to generate Pdx1-Cre; LSL-Kras ( G12D/+); p16 ( Ink4a-/-); RAGE (-/-) mice. Pancreatic ducts were scored and compared to the relevant RAGE (+/+) controls. RESULTS: At 16 weeks of age, Pdx1-Cre; LSL-Kras ( G12D/+); RAGE (-/-) mice had significantly fewer high-grade PanIN lesions than Pdx1-Cre; LSL-Kras ( G12D/+); RAGE (+/+) controls. At 12 weeks of age, none of the Pdx1-Cre; LSL-Kras ( G12D/+); p16 ( Ink4a-/-); RAGE (-/-) mice had PDAC compared to a 45.5% incidence of PDAC in Pdx1-Cre; LSL-Kras ( G12D/+); p16 ( Ink4a-/-); RAGE (+/+) controls. Finally, Pdx1-Cre; LSL-Kras ( G12D/+); p16 ( Ink4a-/-); RAGE (-/-) mice also displayed markedly longer median survival. CONCLUSION: Loss of RAGE function inhibited the development of PanIN and progression to PDAC and significantly prolonged survival in these mouse models. Further work is needed to target the ligand-RAGE axis for possible early intervention and prophylaxis in patients at risk for developing pancreatic cancer.


Subject(s)
Adenocarcinoma/genetics , Carcinoma in Situ/genetics , Cell Transformation, Neoplastic/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Receptors, Immunologic/genetics , Adenocarcinoma/pathology , Animals , Carcinoma in Situ/pathology , Disease Progression , Gene Deletion , Kaplan-Meier Estimate , Mice , Models, Animal , Poisson Distribution , Receptor for Advanced Glycation End Products
SELECTION OF CITATIONS
SEARCH DETAIL
...