Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Neurobiol Aging ; 33(1): 196.e13-27, 2012 Jan.
Article in English | MEDLINE | ID: mdl-20638753

ABSTRACT

Although the pathogenesis of sporadic Alzheimer disease (AD) is not clearly understood, it is likely dependent on several age-related factors. Diabetes is a risk factor for AD, and multiple mechanisms connecting the 2 diseases have been proposed. Hyperglycemia enhances the formation of advanced glycation end products (AGEs) that result from the auto-oxidation of glucose and fructose. The interaction of AGEs with their receptor, named RAGE, elicits the formation of reactive oxygen species that are also believed to be an early event in AD pathology. To investigate a functional link between the disorders diabetes and AD, the effect of 2 AGEs, pentosidine and glyceraldehydes-derived pyridinium (GLAP), was studied on BACE1 expression both in vivo, in streptozotocin treated rats, and in vitro in differentiated neuroblastoma cells. We showed that pentosidine and GLAP were able to upregulate BACE1 expression through their binding with RAGE and the consequent activation of NF-κB. In addition, both pentosidine and GLAP were found to be increased in the brain in sporadic AD patients. Our findings demonstrate that activation of the AGEs/RAGE axis, by upregulating the key enzyme for amyloid-ß production, provides a pathologic link between diabetes mellitus and AD.


Subject(s)
Amyloid Precursor Protein Secretases/physiology , Aspartic Acid Endopeptidases/physiology , Glycation End Products, Advanced/physiology , NF-kappa B/physiology , Receptors, Immunologic/physiology , Signal Transduction/genetics , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animals , Diabetes Mellitus, Experimental , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Risk Factors , Signal Transduction/physiology , Up-Regulation
2.
ScientificWorldJournal ; 9: 781-91, 2009 Aug 11.
Article in English | MEDLINE | ID: mdl-19705038

ABSTRACT

While it is well established that stroke and cerebral hypoperfusion are risk factors for Alzheimer's disease (AD), the molecular link between ischemia/hypoxia and amyloid precursor protein (APP) processing has only been recently established. Here we review the role of the release of reactive oxygen species (ROS) by the mitochondrial electron chain in response to hypoxia, providing evidence that hypoxia fosters the amyloidogenic APP processing through a biphasic mechanism that up-regulates Beta-secretase activity, which involves an early release of ROS and an activation of HIF-1Alpha.


Subject(s)
Alzheimer Disease/pathology , Hypoxia/pathology , Oxidative Stress , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/biosynthesis , Aspartic Acid Endopeptidases/biosynthesis , Electron Transport , Enzyme Induction , Humans , Hypoxia/metabolism , Reactive Oxygen Species/metabolism
3.
Free Radic Biol Med ; 47(7): 1067-74, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19616615

ABSTRACT

This study concentrated on the initial events triggering the development of nonalcoholic fatty liver disease induced by a high-fat plus fructose (HF-F) diet and on the possibility of delaying nonalcoholic fatty liver disease progression by adding dehydroepiandrosterone (DHEA) to the diet. Sterol regulatory element binding protein-1c (SREBP-1c) activation plays a crucial role in the progression of nonalcoholic fatty liver disease induced by an HF-F diet. This study investigated the protective effects of DHEA, a compound of physiological origin with multitargeted antioxidant properties, against the induction of SREBP-1c and on liver insulin resistance in rats fed an HF-F diet, which mimics a typical unhealthy Western diet. An HF-F diet, fortified or not with DHEA (0.01%, w/w), was administered for 15 weeks to male Wistar rats. After HF-F the liver showed unbalanced oxidative status, fatty infiltration, hepatic insulin resistance, and inflammation. The addition of DHEA to the diet reduced both activation of oxidative-stress-dependent pathways and expression of SREBP-1c and partially restored the expression of liver X-activated receptor-alpha and insulin receptor substrate-2 genes. DHEA supplementation of the HF-F diet reduced de novo lipogenesis and delayed progression of nonalcoholic fatty liver disease, demonstrating a relationship between oxidative stress and nonalcoholic fatty liver disease via SREBP-1c.


Subject(s)
Dietary Fats/pharmacology , Fatty Liver/metabolism , Fructose/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Cell Death , Dehydroepiandrosterone/pharmacology , Dietary Fats/adverse effects , Fatty Liver/pathology , Hyperlipidemias/pathology , Insulin Resistance , Male , Oxidative Stress , Rats , Rats, Wistar
4.
J Neurochem ; 108(4): 1045-56, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19196431

ABSTRACT

While it is well established that stroke and cerebral hypoperfusion are both significant risk factors for Alzheimer's disease, the molecular link between ischemia and amyloid precursor protein processing has only been recently established. Specifically, hypoxia significantly increases beta-site APP cleaving enzyme (BACE1) gene transcription through the over-expression of hypoxia inducible factor 1alpha, resulting in increased BACE1 secretase activity and amyloid-beta production. In this study, we significantly extend these findings both in vitro, in differentiated SK-N-BE neuroblastoma cells, and in vivo, in rats subjected to cerebral ischemia, showing that hypoxia up-regulates BACE1 expression through a biphasic mechanism. The early post-hypoxic up-regulation of BACE1 depends on the production of reactive oxygen species mediated by the sudden interruption of the mitochondrial electron transport chain, while the later expression of BACE1 is caused by hypoxia inducible factor 1alpha activation. The involvement of reactive oxygen species released by mitochondria in the BACE1 up-regulation was confirmed by the complete protection exerted by complex I inhibitors such as rotenone and diphenyl-phenylen iodonium. Moreover, the oxidative stress-mediated up-regulation of BACE1 is mediated by c-jun N terminal kinase pathway as demonstrated by the protection exerted by the silencing of c-jun N-terminal kinase isoforms 1 and 2. Our study strengthens the hypothesis that oxidative stress is a basic common mechanism of amyloid-beta accumulation.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Ischemia, Brain/metabolism , Oxidative Stress/physiology , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Brain/metabolism , Brain/physiopathology , Cell Line, Tumor , Disease Models, Animal , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Humans , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/physiopathology , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Uncoupling Agents/pharmacology , Up-Regulation/physiology
5.
Neurobiol Aging ; 30(10): 1563-73, 2009 Oct.
Article in English | MEDLINE | ID: mdl-18255190

ABSTRACT

The activity of beta-secretase (BACE1), the endo-protease essential for the production of amyloid beta (Abeta) peptides, is increased in brain of late-onset sporadic Alzheimer's disease (AD), and oxidative stress is the potential cause of this event. Oxidative stress up-regulates the expression and the activity of BACE1 in cellular and animal models, through a mechanism that involves the increase of gamma-secretase cleavage on APP and the activation of c-jun N-terminal kinase/activator protein 1 (JNK/AP1) pathway. We further characterized the cellular pathways that control BACE1 expression under oxidative stress. We investigated the involvement of extracellular signal regulated MAP kinase (ERK1/2) pathway in the regulation of BACE1 expression, since it has been recently shown that ERK1/2 is an endogenous regulator of the gamma-secretase activity. We found that ERK1/2 pathway negatively modulates BACE1 expression and activity. Moreover, we observed that conditions that abrogate the gamma-secretase activity favor the activation of signalling pathways that promote cell survival, such as ERK1/2 and the serine/threonine kinase Akt/protein kinase B (Akt). These data suggest that the positive or negative cellular responses to oxidative stress parallel the activities of the beta- and the gamma-secretase. ERK1/2 and JNK pathways are involved in this bipartite response, which can lead to neurodegeneration or neuroprotection depending on the cellular and environmental conditions or cooperation with other signalling pathways such as Akt cascade.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidative Stress/physiology , Cell Line, Tumor , Cell Survival/physiology , Humans , MAP Kinase Signaling System , Neurons/physiology , Phosphorylation , Presenilins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism
6.
Shock ; 30(3): 299-307, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18323734

ABSTRACT

The serine/threonine glycogen synthase kinase 3beta (GSK-3beta) is abundant in the central nervous system, particularly in the hippocampus, and plays a pivotal role in the pathophysiology of a number of diseases, including neurodegeneration. This study was designed to investigate the effects of GSK-3beta inhibition against I/R injury in the rat hippocampus. Transient cerebral ischemia (30 min) followed by 1 h of reperfusion significantly increased generation of reactive oxygen species and modulated superoxide dismutase activity; 24 h of reperfusion evoked apoptosis (determined as mitochondrial cytochrome c release and Bcl-2 and caspase-9 expression), resulted in high plasma levels of TNF-alpha and increased expression of cyclooxygenase-2, inducible nitric oxide synthase, and intercellular adhesion molecule-1. The selective GSK-3beta inhibitor, 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), was administered before and after ischemia or during reperfusion alone to assess its potential as prophylactic or therapeutic strategy. Prophylactic or therapeutic administration of TDZD-8 caused the phosphorylation (Ser(9)) and hence inactivation of GSK-3beta. Infarct volume and levels of S100B protein, a marker of cerebral injury, were reduced by TDZD-8. This was associated with a significant reduction in markers of oxidative stress, apoptosis, and the inflammatory response resulting from cerebral I/R. These beneficial effects were associated with a reduction of I/R-induced activation of the mitogen-activated protein kinases JNK1/2 and p38 and nuclear factor-kappaB. The present study demonstrates that TDZD-8 protects the brain against I/R injury by inhibiting GSK-3beta activity. Collectively, our data may contribute to focus the role of GSK-3beta in cerebral I/R.


Subject(s)
Brain Ischemia/drug therapy , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hippocampus/drug effects , Reperfusion Injury/pathology , Thiadiazoles/pharmacology , Animals , Antioxidants/metabolism , Brain/pathology , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Male , Mitochondria/metabolism , Models, Biological , Nerve Growth Factors/biosynthesis , Phosphorylation , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , S100 Calcium Binding Protein beta Subunit , S100 Proteins/biosynthesis
7.
Endocrinology ; 149(1): 380-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17901230

ABSTRACT

Diabetic cardiomyopathy is characterized by myocyte loss and myocardial fibrosis, leading to decreased elasticity and impaired contractile function. The study examines the downstream signaling whereby oxidative stress, induced by hyperglycemia, leads to myocardial fibrosis and impaired contractile function in the left ventricle of diabetic rats. It also examines the effects of dehydroepiandrosterone (DHEA), which prevents the oxidative damage induced by hyperglycemia in experimental models. DHEA was administered for 6 wk in the diet [0.02%, wt/wt)] to rats with streptozotocin-induced diabetes. Oxidative balance, advanced glycated end products (AGEs) and AGE receptors, transcription factors nuclear factor-kappaB and activator protein-1, and profibrogenic growth factors (connective tissue growth factor and TGFbeta1) were determined in the left ventricle of treated and untreated streptozotocin-diabetic rats. Structural and ultrastructural changes, and the contractile force developed by electrically driven papillary muscles, under basal conditions and after stimulation with isoproterenol, were also evaluated. Oxidative stress induced by hyperglycemia increased AGEs and AGE receptors and triggered a cascade of signaling, eventually leading to interstitial fibrosis. DHEA treatment, by improving oxidative balance, counteracted the enhanced AGE receptor activation and increase of profibrogenic factors and restored tissue levels of collagen I, collagen IV, and fibronectin to those of control animals. Moreover, DHEA completely restored the contractility of isolated papillary muscle. Oxidative stress led to cardiac fibrosis, the most important pathogenetic factor of the heart's impaired functional integrity in diabetes. Structural and ultrastructural changes and impairment of muscle function induced by experimental diabetes were minimized by DHEA treatment.


Subject(s)
Diabetes Mellitus, Experimental/complications , Heart Diseases/etiology , Myocardium/pathology , Oxidative Stress/physiology , Animals , Connective Tissue Growth Factor , Dehydroepiandrosterone/pharmacology , Dehydroepiandrosterone/therapeutic use , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Drug Evaluation, Preclinical , Fibrosis , Gene Expression Regulation/drug effects , Glycation End Products, Advanced/metabolism , Heart Diseases/drug therapy , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Myocardium/ultrastructure , Oxidative Stress/drug effects , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Receptors, Immunologic/metabolism , Streptozocin , Transcription Factor AP-1/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
J Neurochem ; 104(3): 683-95, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18005001

ABSTRACT

Sequential cleavages of the beta-amyloid precursor protein cleaving enzyme 1 (BACE1) by beta-secretase and gamma-secretase generate the amyloid beta-peptides, believed to be responsible of synaptic dysfunction and neuronal cell death in Alzheimer's disease (AD). Levels of BACE1 are increased in vulnerable regions of the AD brain, but the underlying mechanism is unknown. Here we show that oxidative stress (OS) stimulates BACE1 expression by a mechanism requiring gamma-secretase activity involving the c-jun N-terminal kinase (JNK)/c-jun pathway. BACE1 levels are increased in response to OS in normal cells, but not in cells lacking presenilins or amyloid precursor protein. Moreover, BACE1 is induced in association with OS in the brains of mice subjected to cerebral ischaemia/reperfusion. The OS-induced BACE1 expression correlates with an activation of JNK and c-jun, but is absent in cultured cells or mice lacking JNK. Our findings suggest a mechanism by which OS induces BACE1 transcription, thereby promoting production of pathological levels of amyloid beta in AD.


Subject(s)
Amyloid Precursor Protein Secretases/pharmacology , Amyloid beta-Protein Precursor/metabolism , Oxidative Stress/physiology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/drug effects , Animals , Aspartic Acid Endopeptidases/metabolism , Cells, Cultured , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Feedback/drug effects , Feedback/physiology , Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Infarction, Middle Cerebral Artery/physiopathology , MAP Kinase Kinase 4/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Presenilins/deficiency , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors , Transfection/methods
9.
Endocrinology ; 147(12): 5967-74, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16935841

ABSTRACT

Oxidative stress plays a key role in the pathogenesis of diabetic cardiomyopathy, which is characterized by myocyte loss and fibrosis, finally resulting in heart failure. The study looked at the downstream signaling whereby oxidative stress leads to reduced myocardial contractility in the left ventricle of diabetic rats and the effects of dehydroepiandrosterone (DHEA), which production is suppressed in the failing heart and prevents the oxidative damage induced by hyperglycemia in several experimental models. DHEA was given orally at a dose of 4 mg/rat per day for 21 d to rats with streptozotocin (STZ)-induced diabetes and genetic diabetic-fatty (ZDF) rats. Oxidative balance, advanced glycated end products (AGEs) and AGE receptors, cardiac myogenic factors, and myosin heavy-chain gene expression were determined in the left ventricle of treated and untreated STZ-diabetic rats and ZDF rats. Oxidative stress induced by chronic hyperglycemia increased AGE and AGE receptors and led to activation of the pleoitropic transcription factor nuclear factor-kappaB. Nuclear factor-kappaB activation triggered a cascade of signaling, which finally led to the switch in the cardiac myosin heavy-chain (MHC) gene expression from the alpha-MHC isoform to the beta-MHC isoform. DHEA treatment, by preventing the activation of the oxidative pathways induced by hyperglycemia, counteracted the enhanced AGE receptor activation in the heart of STZ-diabetic rats and ZDF rats and normalized downstream signaling, thus avoiding impairment of the cardiac myogenic factors, heart autonomic nervous system and neural crest derivatives (HAND) and myogenic enhancer factor-2, and the switch in MHC gene expression, which are the early events in diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Myocardium/chemistry , Oxidative Stress/physiology , Transcription Factors/analysis , Animals , Dehydroepiandrosterone/pharmacology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Glycation End Products, Advanced/blood , Heart/drug effects , Heart Ventricles/chemistry , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/metabolism , Organ Specificity , Oxidative Stress/drug effects , Rats , Rats, Wistar , Rats, Zucker , Receptor for Advanced Glycation End Products , Receptors, Immunologic/analysis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Free Radic Biol Med ; 41(2): 202-12, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16814100

ABSTRACT

The amyloid cascade hypothesis suggests that the insoluble and fibrillar form of beta-amyloid (A beta) may play a primary pathogenic role in Alzheimer disease at the molecular level. However, neither the rate of dementia nor the extent of neuronal change seems to correlate with the levels of amyloidotic plaques (i.e., aggregated/fibrillar A beta). Recent evidence suggests, however, that neurotoxicity may be exerted also by rather small soluble aggregates of A beta, including oligomers. To characterize the mechanisms underlying toxicity mediated by the various aggregation states of A beta peptides is then a major goal of research. In this work we investigated the effects of fibrillar, prefibrillar, and oligomeric A beta(1-42) on the induction of oxidative stress, cell death, and BACE-1 expression in NT2 neuronal cells. We found that prefibrillar and oligomeric A beta(1-42) resulted in a more dramatic increase in the oxidative stress markers 4-hydroxynonenal and hydrogen peroxide compared to fibrillar A beta(1-42). Moreover, increased oxidative stress levels also resulted in a more rapid and significant induction of both apoptotic and necrotic neuronal cell death. Accordingly, fibrillar A beta(1-42), but not the soluble nonfibrillar forms, was the only condition able to up-regulate BACE-1 expression and activity.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , Oxidative Stress , Peptide Fragments/metabolism , Base Sequence , Cell Death , Cell Line , DNA Primers , Humans , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction
11.
Free Radic Biol Med ; 41(4): 579-89, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16863991

ABSTRACT

This study investigated the effects of the selective peroxisome proliferator-activated receptor-alpha (PPAR-alpha) agonist WY14643 on ischemia/reperfusion (I/R) injury in the rat hippocampus. Transient cerebral ischemia (30 min), followed by 1-24 h reperfusion, significantly increased the generation of reactive oxygen species, nitric oxide (NO), and lipid peroxidation end-products, as well as markedly reducing levels of the endogenous antioxidant glutathione. Reperfusion for 3-6 h led to increased expression of the proteins heme oxygenase-1 (HO-1), cyclooxygenase-2 (COX-2), inducible NO synthase (iNOS), and intercellular adhesion molecule-1 (ICAM-1). Pretreatment with WY14643 suppressed oxidative stress and expression of HO-1, iNOS, and ICAM-1, but had no effect on COX-2. These effects are due to suppression of the activation of p38 mitogen-activated protein kinase and nuclear factor-kappaB. The PPAR-alpha antagonist MK886 abolished the beneficial effects of WY14643. The levels of S100B protein, a marker of cerebral injury used in stroke trials to monitor injury, were high in the hippocampus of rats exposed to I/R, but markedly reduced by WY14643. We propose that WY14643 protects the brain against excessive oxidative stress and inflammation and may thus be useful in treating stroke.


Subject(s)
Brain Ischemia/metabolism , Oxidative Stress , PPAR alpha/agonists , Pyrimidines/pharmacology , Reperfusion Injury/metabolism , Animals , Blotting, Western , Cyclooxygenase 2/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipid Peroxidation , MAP Kinase Signaling System/drug effects , Male , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar
12.
Neurobiol Aging ; 27(11): 1588-94, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16464517

ABSTRACT

Clusterin/apolipoprotein J is a multifunctional protein up-regulated during various pathophysiological states. Since oxidative stress plays an important role in brain aging, and in many neurodegenerative disorders, to further understand the mechanistic underpinnings of clusterin expression, in this study, we examined clusterin expression in human neuroblastoma cells under conditions of increased production of reactive oxygen species and lipid peroxidation. Specifically, we analyzed clusterin mRNA and protein levels in human neuroblastoma IMR-32 and SH-SY5Y cells following exposure to sub-lethal amounts of iron-ascorbate to induce an increase in reactive oxygen species generation and lipid peroxidation. Under such conditions, we observed a time-dependent up-regulation of clusterin protein and mRNA levels, detected by immunoblot analysis and RT-PCR, respectively. Given the known roles of clusterin, the results of the present study support the notion that an increase in clusterin expression may be a physiological defence mounted to reduce cell damage and maintain cell viability during periods of increased oxidative stress.


Subject(s)
Clusterin/metabolism , Lipid Peroxidation , Neurons/metabolism , Oxidative Stress , Up-Regulation , Alzheimer Disease , Ascorbic Acid/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Clusterin/genetics , Ferrous Compounds/pharmacology , Humans , Hydrogen Peroxide/metabolism , Oxidants/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
13.
Eur J Pharmacol ; 530(1-2): 70-80, 2006 Jan 13.
Article in English | MEDLINE | ID: mdl-16386242

ABSTRACT

Agonists of the peroxisome proliferator-activated receptor-gamma (PPAR-gamma) exert protective effects in several models of ischemia/reperfusion injury, but their role in stroke is less clear. The study investigates the effects of two PPAR-gamma agonists, rosiglitazone and pioglitazone, on oxidative stress and inflammatory response induced by ischemia/reperfusion in the rat hippocampus. Common carotid artery occlusion for 30 min followed by 1 h reperfusion resulted in a significant increase in the generation of reactive oxygen species, nitric oxide and the end products of lipid peroxidation as well as markedly reduced endogenous antioxidant glutathione levels and up-regulated superoxide dismutase activity. Western blot analysis showed that ischemia/reperfusion lead to an increase in cyclooxygenase-2 (COX-2) expression, as well activating p38 and p42/44 mitogen-activated protein kinases (MAPKs) and nuclear factor-kappaB (NF-kappaB). Pre-treatment with either rosiglitazone or pioglitazone significantly reduced oxidative stress, COX-2 protein expression and activation of MAPKs and NF-kappaB. Taken together, the results provide convincing evidence that PPAR-gamma agonists exert protective effects in a rat model of mild forebrain ischemia/reperfusion injury by inhibiting oxidative stress and excessive inflammatory response.


Subject(s)
Brain Ischemia/physiopathology , Hippocampus/drug effects , Inflammation/drug therapy , Oxidative Stress/drug effects , PPAR gamma/agonists , Animals , Brain Ischemia/drug therapy , Cyclooxygenase 2/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Inflammation/physiopathology , Injections, Intravenous , Lipid Peroxidation/drug effects , Male , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Oxidative Stress/physiology , Pioglitazone , Rats , Rats, Wistar , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Rosiglitazone , Signal Transduction/drug effects , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use
14.
Endocrinology ; 146(12): 5561-7, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16166220

ABSTRACT

Diabetics have at least twice the risk of stroke and may show performance deficit in a wide range of cognitive domains. The mechanisms underlying this gradually developing end-organ damage may involve both vascular changes and direct damage to neuronal cells as a result of overproduction of superoxide by the respiratory chain and consequent oxidative stress. The study aimed to assess the role of oxidative stress on the aldose reductase-polyol pathway, on advanced glycated end-product (AGE)/AGE-receptor interaction, and on downstream signaling in the hippocampus of streptozotocin-treated rats. Data show that, in diabetic rats, levels of prooxidant compounds increase, whereas levels of antioxidant compounds fall. Receptor for AGE and galectin-3 content and polyol flux increase, whereas glyceraldehyde-3-phosphate dehydrogenase activity is impaired. Moreover, nuclear factor kappaB (p65) transcription factor levels and S-100 protein are increased in the hippocampus cytosol, suggesting that oxidative stress triggers the cascade of events that finally leads to neuronal damage. Dehydroepiandrosterone, the most abundant hormonal steroid in the blood, has been reported to possess antioxidant properties. When dehydroepiandrosterone was administered to diabetic rats, the improved oxidative imbalance and the marked reduction of AGE receptors paralleled the reduced activation of nuclear factor kappaB and the reduction of S-100 levels, reinforcing the suggestion that oxidative stress plays a role in diabetes-related neuronal damage.


Subject(s)
Antioxidants/pharmacology , Brain/metabolism , Diabetes Mellitus, Experimental/metabolism , Receptors, Immunologic/metabolism , Up-Regulation/drug effects , Aldehyde Reductase/metabolism , Animals , Glycation End Products, Advanced/metabolism , Hippocampus/metabolism , Male , NF-kappa B/metabolism , Nerve Growth Factors/metabolism , Oxidative Stress , Rats , Rats, Wistar , Receptor for Advanced Glycation End Products , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism
15.
J Hepatol ; 43(5): 791-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16085334

ABSTRACT

BACKGROUND/AIMS: In liver cirrhosis atrial natriuretic peptide (ANP) decreases portal vascular resistance and tributary flow. The enzyme neutral endopeptidase (NEP) degrades ANP and bradykinin and generates endothelin-1 from big-endothelin. We determined the effects of NEP inhibition by candoxatrilat on hormonal status, liver function and arterial and portal pressures in rats with CCl4-induced cirrhosis. METHODS: Two groups of seven control rats received 1 ml 5% glucose solution alone or containing 10 mg/kg candoxatrilat; three groups of 10 ascitic cirrhotic rats received placebo, 5 or 10 mg/kg candoxatrilat. NEP protein concentration and immunostaining were analyzed in normal and cirrhotic livers. RESULTS: In cirrhotic rats 10 mg/kg candoxatrilat significantly increased steady-state indocyanine green clearance (a parameter reflecting liver plasma flow) (P<0.01), decreased portal pressure (P<0.01), had no effect on arterial pressure and plasma renin activity but increased ANP plasma levels (P<0.05) and urinary excretions (P<0.01) of ANP and cGMP. In the cytosol fraction of rat cirrhotic livers a 280% increase in NEP content was found (P<0.01), chiefly localized in desmin-positive myofibroblast-like cells of fibrous septa. CONCLUSIONS: Candoxatrilat has few effects on systemic hemodynamics and hormonal status; its portal hypotensive action depends on effects exerted on intrahepatic vascular resistance.


Subject(s)
Hypertension, Portal/drug therapy , Liver Cirrhosis, Experimental/enzymology , Neprilysin , Vascular Resistance/physiology , Animals , Arginine Vasopressin/metabolism , Atrial Natriuretic Factor/metabolism , Blood Pressure/drug effects , Blood Pressure/physiology , Carbon Tetrachloride/toxicity , Cyclic GMP/metabolism , Cyclohexanecarboxylic Acids/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , Cytokines/metabolism , Endothelin-1/metabolism , Humans , Liver/cytology , Liver/enzymology , Liver Cirrhosis, Experimental/chemically induced , Male , Neprilysin/antagonists & inhibitors , Neprilysin/metabolism , Portal Vein/drug effects , Portal Vein/physiology , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use , Rats , Rats, Wistar , Vascular Resistance/drug effects
16.
J Neurochem ; 92(3): 628-36, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15659232

ABSTRACT

4-Hydroxynonenal (HNE), an aldehydic product of lipid peroxidation, up-regulates expression of the beta-site APP cleaving enzyme (BACE-1), an aspartyl protease responsible for the beta-secretase cleavage of amyloid precursor protein (AbetaPP), and results in increased levels of amyloid beta (Abeta) peptide. The mechanisms underlying this remain unclear but are of fundamental importance because prevention of BACE-1 up-regulation is viewed as an important therapeutic strategy. In this study, we exposed NT(2) neurons to a range of HNE concentrations (0.5-5 microm) that elicited an up-regulation of BACE-1 expression, a significant increase in intracellular and secreted levels of Abeta peptides as well as apoptosis involving poly-ADP ribose polymerase cleavage and activation of caspase 3. To delineate the molecular events involved in HNE-mediated BACE-1 activation, we investigated the involvement of stress-activated protein kinases (SAPK), signal transducers and activators of transcription (STAT) and serine-threonine kinase B/phosphatidylinositol phosphate 3 kinase (Akt/PtdIns3K). Using specific pharmacological inhibitors, our results show that activation of c-Jun N-terminal kinases and p38(MAPK.), but not STAT or Akt/PtdIns3K, pathways mediate the HNE-dependent up-regulation of BACE-1 expression. Therefore, HNE, an oxidative stress mediator detected in vivo in the brains of Alzheimer's disease patients, may play a pathogenetic role in Alzheimer's disease by selectively activating SAPK pathways and BACE-1 that regulate the proteolytic processing of AbetaPP.


Subject(s)
Aldehydes/pharmacology , Endopeptidases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Signal Transduction/drug effects , Up-Regulation/drug effects , Amyloid Precursor Protein Secretases , Apoptosis/drug effects , Aspartic Acid Endopeptidases , Cell Line , DNA-Binding Proteins/metabolism , Endopeptidases/genetics , Enzyme Activators/pharmacology , Humans , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , STAT1 Transcription Factor , Trans-Activators/metabolism , Up-Regulation/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Free Radic Biol Med ; 38(2): 215-25, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15607904

ABSTRACT

4-Hydroxynonenal (HNE), a product of lipid peroxidation, inhibits proliferation of several tumor cells. The p53 tumor suppressor protein plays a critical role in cell cycle control, by inducing p21 expression, and in apoptosis, by inducing bax expression. Recently, two other proteins with many p53-like properties, TAp73 (p73) and TAp63 (p63), have been discovered. SK-N-BE human neuroblastoma cells express the three p53 family proteins and can be used for the study of their induction. We investigated HNE action in the control of proliferation, differentiation, and apoptosis in SK-N-BE cells and the HNE effect on the expression of p53, p63, p73, p21, bax, and G1 cyclins. Retinoic acid (RA) was used as a positive control. HNE inhibited cell proliferation without inducing differentiation; it decreased S-phase cells and increased the number of apoptotic cells. RA reduced the proportion of S-phase cells and did not induce apoptosis. HNE increased p53, p73, p63, p21, and bax expression at different time points. HNE reduced cyclin D2 expression and the phosphorylation of pRb protein. Our results demonstrated that HNE inhibits SK-N-BE cell proliferation by increasing the expression of p53 family proteins and p53 target proteins which modulate cell cycle progression and apoptosis.


Subject(s)
Aldehydes/pharmacology , Neuroblastoma/metabolism , Tumor Suppressor Protein p53/physiology , Apoptosis , Blotting, Western , Cell Cycle , Cell Cycle Proteins/biosynthesis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinase Inhibitor p21 , Cysteine Proteinase Inhibitors/pharmacology , DNA-Binding Proteins/metabolism , Flow Cytometry , Genes, Tumor Suppressor , HL-60 Cells , Humans , Lipid Metabolism , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Peroxides/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trans-Activators/metabolism , Transcription Factors , Tretinoin/metabolism , Tumor Protein p73 , Tumor Suppressor Proteins , bcl-2-Associated X Protein
18.
Diabetes ; 53(4): 1082-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047625

ABSTRACT

Alongside increased proteolysis, the inability to repair damaged skeletal muscle is a characteristic feature of uncontrolled diabetes. This study evaluates the role of oxidative stress in muscle-specific gene regulatory regions and myosin chain synthesis in streptozotocin (STZ)-induced diabetic and ZDF rats. In the gastrocnemius muscle of diabetic rats, prooxidant compounds were seen to increase while antioxidant levels fell. Myogenic regulatory factors--Myo, myogenin, and Jun D--were also reduced, and muscle enhancer factor (MEF)-1 DNA binding activity was impaired. Moreover, synthesis of muscle creatine kinase and both heavy and light chains of myosin were impaired, suggesting that oxidative stress triggers the cascade of events that leads to impaired muscle repair. Dehydroepiandrosterone has been reported to possess antioxidant properties. When it was administered to diabetic rats, in addition to an improved oxidative imbalance there was a recovery of myogenic factors, MEF-1 DNA binding activity, synthesis of muscle creatine kinase, and myosin light and heavy chains. Vitamin E administration to STZ-induced diabetic rats reverses oxidative imbalance and improves muscle gene transcription, reinforcing the suggestion that oxidative stress may play a role in diabetes-related impaired muscle repair.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Muscle, Skeletal/physiopathology , Oxidative Stress/physiology , Animals , Dehydroepiandrosterone/pharmacology , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Rats , Rats, Wistar , Vitamin E/pharmacology
19.
J Hepatol ; 40(1): 60-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14672615

ABSTRACT

BACKGROUND/AIMS: 4-Hydroxynonenal (HNE) is a putative pro-fibrogenic product of oxidative stress able to elicit apoptosis and cytotoxicity in several cell types. This study has been performed to evaluate its 'in vivo' levels in injured liver and whether HNE may induce apoptosis and/or affect selected phenotypic responses in activated human hepatic stellate cells (HSC/MF). METHODS/RESULTS: During the development of acute liver injury induced by CCl(4), liver tissue HNE levels were in the range 0.5-10 microM, as shown by high performance liquid chromatography analysis. Cultured human HSC/MF, developed cytotoxicity only if exposed to very high HNE concentrations (25-50 microM) without any sign of induction of classic, caspase-dependent apoptosis, as assessed by evaluating morphology and biochemical parameters of cell death. HNE, at non-cytotoxic doses, up-regulated procollagen type I and tissue inhibitor of metalloproteinases-1 gene expression and/or protein synthesis without significantly affecting chemotaxis (wound healing and haptotaxis assay), matrix metalloproteinases 1 and 2 mRNA expression and activity as well as basal DNA synthesis. CONCLUSIONS: HNE, at concentrations compatible with those detected in vivo, does not elicit HSC/MF classic apoptosis but, rather, may act as a potent pro-fibrogenic stimulus for the expression of genes involved in excess extracellular matrix deposition and proposed as survival signals for HSC/MF.


Subject(s)
Aldehydes/pharmacology , Liver Cirrhosis/chemically induced , Liver/drug effects , Actins/metabolism , Acute Disease , Aldehydes/administration & dosage , Aldehydes/metabolism , Animals , Apoptosis , Carbon Tetrachloride , Cell Death , Cells, Cultured , Chemical and Drug Induced Liver Injury , Cytoskeleton/drug effects , Dose-Response Relationship, Drug , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression/drug effects , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Diseases/metabolism , Liver Diseases/pathology , Liver Diseases/physiopathology , Male , Osmolar Concentration , Phenotype , Rats , Rats, Wistar , Signal Transduction
20.
Kidney Int ; 64(3): 836-43, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12911533

ABSTRACT

BACKGROUND: The pathogenesis of ischemia/reperfusion (I/R) involves generation of reactive oxygen and nitrogen species. This in vivo study investigates the effect of dehydroepiandrosterone (DHEA), a physiologic steroid with antioxidant properties, on oxidative balance and renal dysfunctions induced by monolateral I/R. METHODS: Normal and DHEA-treated rats (4 mg/day x 21 days, orally) were subjected to monolateral renal I/R (30 minutes/6 hours). The oxidative state was determined by measuring hydrogen peroxide level and activities of glutathione-peroxidase, catalase, and superoxide dismutase. Tumor necrosis factor-alpha (TNF-alpha) and nitric oxide production and inducible nitric oxide synthase (iNOS) levels were also measured. Hydroxynonenal content was used to probe lipid peroxidation. Functional parameters determined were creatinine levels and Na/K-ATPase activity. Immunohistochemical and morphologic studies were also performed. RESULTS: A markedly pro-oxidant state was evident in the kidney of rats subjected to I/R. Both hydrogen peroxide and reactive nitrogen species (nitric oxide and iNOS) increased, whereas antioxidants decreased. Oxidant species induce TNF-alpha increase, which, in turn, produces lipoperoxidative processes, as documented by the increased hydroxynonenal (HNE) level. As final result, impaired renal functionality, hydropic degeneration, and vacuolization of proximal convolute tubules were observed in kidneys of I/R rats. DHEA pretreatment improved the parameters considered. CONCLUSION: I/R induces oxidative stress and consequently damages the proximal convolute renal tubules. Rats supplemented with DHEA and subjected to I/R had reduced pro-oxidant state, oxidative damage, and improved renal functionality, indicating an attenuation of oxidative injury and dysfunctions mediated by I/R.


Subject(s)
Antioxidants/pharmacology , Dehydroepiandrosterone/pharmacology , Kidney/physiopathology , Oxidative Stress , Renal Circulation , Reperfusion Injury/physiopathology , Animals , Immunohistochemistry , Kidney/drug effects , Kidney/pathology , Male , Rats , Rats, Wistar , Reperfusion Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...