Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 38(1): 50-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23011206

ABSTRACT

Poly(ADP-ribose) polymerases (PARPs) are recognized as key regulators of cell survival or death. PARP-1 is essential to the repair of DNA single-strand breaks via the base excision repair pathway. The enzyme may be overactivated in response to inflammatory cues, thus depleting cellular energy pools and eventually causing cell death. Accordingly, PARP-1 inhibitors, acting by competing with its physiological substrate NAD(+), have been proposed to play a protective role in a wide range of inflammatory and ischemia/reperfusion-associated diseases. Recently, it has also been reported that PARP-1 regulates proinflammatory mediators, including cytokines, chemokines, adhesion molecules, and enzymes (e.g., iNOS). Furthermore, PARP-1 has been shown to act as a coactivator of NF-κB- and other transcription factors implicated in stress/inflammation, as AP-1, Oct-1, SP-1, HIF, and Stat-1. To further substantiate this hypothesis, we tested the biomolecular effects of PARP-1 inhibitors DPQ and PJ-34 on human glioblastoma cells, induced to a proinflammatory state with lipopolysaccharide and Interferon-γ. PARP-1 expression was evaluated by laser scanning confocal microscopy immunofluorescence (LSM); nitrite production, LDH release and cell viability were also determined. LSM of A-172, SNB-19 and CAS-1 cells demonstrated that DPQ and PJ-34 downregulate PARP-1 expression; they also cause a decrease of LDH release and nitrite production, while increasing cell viability. Similar effects were caused in all three cell lines by N-mono-methyl-arginine, a well known iNOS inhibitor, and by L-carnosine and trehalose, two antioxidant molecules. These results demonstrate that, similar to other well characterized drugs, DPQ and PJ-34 reduce cell inflammation and damage that follow PARP-1 overexpression, while they increase cell survival: this suggests their potential exploitation in clinical Medicine.


Subject(s)
Anti-Inflammatory Agents , Brain Neoplasms/pathology , Glioblastoma/pathology , Isoquinolines/pharmacology , Phenanthrenes/pharmacology , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Biomarkers/metabolism , Carnosine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Coloring Agents , Down-Regulation/drug effects , Fluorescent Antibody Technique , Humans , L-Lactate Dehydrogenase/biosynthesis , Microscopy, Confocal , Nitrites/metabolism , Poly (ADP-Ribose) Polymerase-1 , Tetrazolium Salts , Thiazoles , Trehalose/pharmacology
2.
Neurochem Res ; 35(12): 1880-915, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21080068

ABSTRACT

The widely accepted oxidative stress theory of aging postulates that aging results from accumulation of oxidative damage. A prediction of this theory is that, among species, differential rates of aging may be apparent on the basis of intrinsic differences in oxidative damage accrual. Although widely accepted, there is a growing number of exceptions to this theory, most contingently related to genetic model organism investigations. Proteins are one of the prime targets for oxidative damage and cysteine residues are particularly sensitive to reversible and irreversible oxidation. The adaptation and survival of cells and organisms requires the ability to sense proteotoxic insults and to coordinate protective cellular stress response pathways and chaperone networks related to protein quality control and stability. The toxic effects that stem from the misassembly or aggregation of proteins or peptides, in any cell type, are collectively termed proteotoxicity. Despite the abundance and apparent capacity of chaperones and other components of homeostasis to restore folding equilibrium, the cell appears poorly adapted for chronic proteotoxic stress which increases in cancer, metabolic and neurodegenerative diseases. Pharmacological modulation of cellular stress response pathways has emerging implications for the treatment of human diseases, including neurodegenerative disorders, cardiovascular disease, and cancer. A critical key to successful medical intervention is getting the dose right. Achieving this goal can be extremely challenging due to human inter-individual variation as affected by age, gender, diet, exercise, genetic factors and health status. The nature of the dose response in and adjacent to the therapeutic zones, over the past decade has received considerable advances. The hormetic dose-response, challenging long-standing beliefs about the nature of the dose-response in a lowdose zone, has the potential to affect significantly the design of pre-clinical studies and clinical trials as well as strategies for optimal patient dosing in the treatment of numerous diseases. Given the broad cytoprotective properties of the heat shock response there is now strong interest in discovering and developing pharmacological agents capable of inducing stress responses, including carnitines. This paper describes in mechanistic detail how hormetic dose responses are mediated for endogenous cellular defense pathways, including the possible signaling mechanisms by which the carnitine system, by interplaying metabolism, mitochondrial energetics and activation of critical vitagenes, modulates signal transduction cascades that confer cytoprotection against chronic degenerative damage associated to aging and neurodegenerative disorders.


Subject(s)
Aging/physiology , Carnitine/deficiency , Neurodegenerative Diseases/physiopathology , Stress, Physiological , Adaptation, Physiological , Humans , Mitochondria/physiology , Oxidation-Reduction
3.
Nat Rev Neurosci ; 8(10): 766-75, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17882254

ABSTRACT

At the end of the 1980s, it was clearly demonstrated that cells produce nitric oxide and that this gaseous molecule is involved in the regulation of the cardiovascular, immune and nervous systems, rather than simply being a toxic pollutant. In the CNS, nitric oxide has an array of functions, such as the regulation of synaptic plasticity, the sleep-wake cycle and hormone secretion. Particularly interesting is the role of nitric oxide as a Janus molecule in the cell death or survival mechanisms in brain cells. In fact, physiological amounts of this gas are neuroprotective, whereas higher concentrations are clearly neurotoxic.


Subject(s)
Central Nervous System/physiology , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/metabolism , Nitric Oxide/physiology , Animals , Central Nervous System/pathology , Humans , Neurodegenerative Diseases/prevention & control , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Nitric Oxide/therapeutic use , Nitric Oxide/toxicity
4.
J Neurosci Res ; 85(10): 2239-45, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17546663

ABSTRACT

The neuropeptide carnosine (beta-amyloid peptide aggregation has been demonstrated. Carnosine protection against peroxynitrite damage is particularly relevant, but until now there has been no evidence of any direct interaction with nitric oxide. In this study we examined the protection that carnosine provides against nitric oxide (NO)-induced cell death in primary rat astroglial cell cultures treated with lipopolysaccharide (LPS) and interferon gamma (INFgamma), a well-known neurotoxic proinflammatory condition. A correlation was found between cell protection and NO free-radical scavenging activity of carnosine. Moreover, by competitive spectrophotometric measurement and electrospray mass spectrometry analysis in cell-free experiments, we demonstrated a direct interaction of the dipeptide with NO. A comparison of carnosine with its homologues or derivatives (homocarnosine and carcinine) as well as with its amino acid constituents (L-histidine and beta-alanine) highlighted that only histidine showed significant scavenging activity. Therefore, carnosine shows direct NO-trapping ability and may be a valuable multifunctional molecule in the treatment of neurodegenerative disorders.


Subject(s)
Astrocytes/physiology , Carnosine/pharmacology , Cytoprotection , Free Radical Scavengers/pharmacology , Nitric Oxide/antagonists & inhibitors , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Carnosine/administration & dosage , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Free Radical Scavengers/administration & dosage , Histidine/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Neurotoxins/pharmacology , Rats , Rats, Wistar
5.
J Neurochem ; 101(3): 709-17, 2007 May.
Article in English | MEDLINE | ID: mdl-17241115

ABSTRACT

Increasing evidence suggests a critical role for oxidative and nitrosative stress in the pathogenesis of most important neurodegenerative disorders. Parkinson's disease (PD) is a neurodegenerative disease characterized by a severe depletion in number of dopaminergic cells of the substantia nigra (SN). Administration of L-DOPA (LD) is the more effective treatment for patients with PD. However, the vast majority of patients suffer LD-related complications, which represent the major problem in the clinical management of PD. In the present study, LD administration to rats resulted in a significant dose-dependent increase in Hsp70 synthesis which was specific for the SN. The amount of 70 kDa protein increased after 6 h treatment reaching the maximal induction after 24-48 h. Induction of Hsp70 in the SN was associated with a significant increase in constitutive Hsc70 and mitochondrial Hsp60 stress proteins, and with increased expression of mitochondrial complex I whereas no significant changes were found in the activity of complex IV. In the same experimental conditions, a significant decrease in reduced glutathione was observed, which was associated with an increased content of oxidized glutathione content as well as nitric oxide (NO) synthase activity, NO metabolites and nitrotyrosine immunoreactivity. Interestingly, Hsp70 induction, iNOS up-regulation and nitrotyrosine formation have been confirmed also in SN and striatum of rats treated with LD and carbidopa, this latter being an inhibitor of the peripheral DOPA decarboxylase. Our data are in favor of the importance of the heat shock signal pathway as a basic mechanism of defense against neurotoxicity elicited by free radical oxygen and nitrogen species produced in aging and neurodegenerative disorders.


Subject(s)
Dopamine Agents/pharmacology , Electron Transport Complex I/metabolism , Heat-Shock Proteins/metabolism , Levodopa/pharmacology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Substantia Nigra/drug effects , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Glutathione/metabolism , Male , Nitric Oxide Synthase/metabolism , Nitrites/metabolism , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Time Factors
6.
Cell Stress Chaperones ; 12(4): 299-306, 2007.
Article in English | MEDLINE | ID: mdl-18229449

ABSTRACT

Oxidative stress has been suggested to play a main role in the pathogenesis of type 2 diabetes mellitus and its complications. As a consequence of this increased oxidative status, a cellular-adaptive response occurs requiring functional chaperones, antioxidant production, and protein degradation. This study was designed to evaluate systemic oxidative stress and cellular stress response in patients suffering from type 2 diabetes-induced nephropathy and in age-matched healthy subjects. Systemic oxidative stress has been evaluated by measuring advanced glycation end-products (pentosidine), protein oxidation (protein carbonyls [DNPH]), and lipid oxidation (4-hydroxy-2-nonenal [HNE] and F2-isoprostanes) in plasma, lymphocytes, and urine, whereas the lymphocyte levels of the heat shock proteins (Hsps) heme oxygenase-1 (HO-1), Hsp70, and Hsp60 as well as thioredoxin reductase-1 (TrxR-1) have been measured to evaluate the systemic cellular stress response. We found increased levels of pentosidine (P < 0.01), DNPH (P < 0.05 and P < 0.01), HNE (P < 0.05 and P < 0.01), and F2-isoprostanes (P < 0.01) in all the samples from type 2 diabetic patients with nephropathy with respect to control group. This was paralleled by a significant induction of cellular HO-1, Hsp60, Hsp70, and TrxR-1 (P < 0.05 and P < 0.01). A significant upregulation of both HO-1 and Hsp70 has been detected also in lymphocytes from type 2 diabetic patients without uraemia. Significant positive correlations between DNPH and Hsp60, as well as between the degree of renal failure and HO-1 or Hsp70, also have been found in diabetic uremic subjects. In conclusion, patients affected by type 2 diabetes complicated with nephropathy are under condition of systemic oxidative stress, and the induction of Hsp and TrxR-1 is a maintained response in counteracting the intracellular pro-oxidant status.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Oxidative Stress , Aldehydes/blood , Arginine/analogs & derivatives , Arginine/blood , Arginine/urine , Chaperonin 60/metabolism , Diabetic Nephropathies/enzymology , F2-Isoprostanes/blood , F2-Isoprostanes/urine , Female , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response , Heme Oxygenase-1/metabolism , Humans , Lymphocytes/metabolism , Lysine/analogs & derivatives , Lysine/blood , Lysine/urine , Male , Middle Aged , Protein Carbonylation , Renal Insufficiency/pathology , Thioredoxin Reductase 1/metabolism
7.
Antioxid Redox Signal ; 8(11-12): 1975-86, 2006.
Article in English | MEDLINE | ID: mdl-17034343

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder with cognitive and memory decline, personality changes, and synapse loss. Increasing evidence indicates that factors such as oxidative and nitrosative stress, glutathione depletion, and impaired protein metabolism can interact in a vicious cycle, which is central to AD pathogenesis. In the present study, we demonstrate that brains of AD patients undergo oxidative changes classically associated with a strong induction of the so-called vitagenes, including the heat shock proteins (HSPs) heme oxygenase-1 (HO-1), HSP60, and HSP72, as well as thioredoxin reductase (TRXr). In inferior parietal brain of AD patients, a significant increase in the expression of HO-1 and TRXr was observed, whereas HO-2 expression was decreased, compared with controls. TRHr was not increased in AD cerebellum. Plasma GSH was decreased in AD patients, compared with the control group, and was associated with a significant increase in oxidative stress markers (i.e., GSSG, hydroxynonenal, protein carbonyl content, and nitrotyrosine). In AD lymphocytes, we observed an increased expression of inducible nitric oxide synthase, HO-1, Hsp72, HSP60, and TRXr. Our data support a role for nitrative stress in the pathogenesis of AD and indicate that the stress-responsive genes, such as HO-1 and TRXr, may represent important targets for novel cytoprotective strategies.


Subject(s)
Alzheimer Disease/metabolism , Homeostasis , Nitric Oxide/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/pathology , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Female , Humans , Male , Middle Aged , Nitrosation , Oxidation-Reduction , Oxidative Stress
8.
Neurobiol Aging ; 27(7): 1020-34, 2006 Jul.
Article in English | MEDLINE | ID: mdl-15996793

ABSTRACT

Age-related impairment of functionality of the central nervous system (CNS) is associated with increased susceptibility to develop many neurodegenerative diseases. Increased oxidative stress in the CNS of aged animals is manifested by increased protein oxidation, which is believed to contribute to the age-related learning and memory deficits. Glutamate dysregulation, mitochondrial dysfunction and impaired protein synthesis are observed in aged brains, along with increased protein oxidation. Interestingly, all of these age-related cellular alterations can be improved by caloric restriction (CR), which can also improve the plasticity and recovery of the CNS. Although the beneficial effects of CR on brains are well established, the mechanism(s) of its action remains unclear. In order to gain insight into the mechanism of CR in the brain, we located the brain regions that are benefited the most from reduced oxidative stress by CR. Along with other brain regions, striatum (ST) showed significantly decreased bulk protein carbonyl levels and hippocampus (HP) showed decreased bulk protein 3-nitrotyrosine (3-NT) levels in CR aged rats when compared to those of age matched controls. To determine which proteins were oxidatively modified in these brain regions, we used parallel proteomics approach to identify the proteins that are altered in oxidation and expression. The specific carbonyl levels of pyruvate kinase M2 (PKM2), alpha-enolase (ENO1), inositol monophosphatase (INSP1), and F1-ATPase Chain B (ATP-F1B) were significantly decreased in ST of aged CR rats. In contrast, the expression levels of phosphoglycerate kinase 1 (PKG1), inosine monophosphate cyclohydrolase (IMPCH) and F1-ATPase Chain A (ATP-F1A) were significantly increased in the ST of CR rats. In the hippocampus of CR rats, the specific 3-NT levels of malate dehydrogenase (MDH), phosphoglycerate kinase 1 (PKG1) and 14-3-3 zeta protein were significantly decreased and expression levels of DLP1 splice variant 1 (DLP1), mitochondrial aconitase (ACO2), dihydrolipoamide dehydrogenase (DLDH), neuroprotective peptide H3 (NPH3), and eukaryotic translation initiation factor 5A (eIF-5A) are increased. Moreover, an unnamed protein product (UNP1) with similar sequence to initiation factor 2 (IF-2) was decreased in the HP of CR rats. Our data support the hypothesis that CR induces a mild metabolic stress response by increasing the production of neurotrophic proteins, therefore, priming neurons against apoptosis. Moreover, our study shows that the improvement of glutamate dysregulation, mitochondrial dysfunction and protein synthesis by CR is, at least partially, due to the CR-mediated alteration of the oxidation or the expression of PKM2, ENO1, INSP1, ATP-F1B, PKG1, IMPCH, ATP-F1A MDH, PKG1 and 14-3-3 zeta protein, DLP1, ACO2, DLDH, NPH3, eIF-5A and UNP1. This study provides valuable insights into the mechanisms of the beneficial factors on brain aging by CR.


Subject(s)
Aging/metabolism , Brain/metabolism , Caloric Restriction , Glutamic Acid/metabolism , Mitochondria/metabolism , Nerve Tissue Proteins/biosynthesis , Neurodegenerative Diseases/metabolism , Aging/pathology , Animals , Brain/physiopathology , Down-Regulation/physiology , Energy Metabolism/physiology , Enzyme Activation/physiology , Enzymes/metabolism , Male , Nerve Growth Factors/biosynthesis , Neurodegenerative Diseases/physiopathology , Neurodegenerative Diseases/prevention & control , Oxidative Stress/physiology , Proteomics , Rats , Rats, Wistar , Up-Regulation/physiology
10.
Ital J Biochem ; 55(3-4): 263-82, 2006.
Article in English | MEDLINE | ID: mdl-17274531

ABSTRACT

There is increasing evidence that reactive oxygen species (ROS) are not only toxic but play an important role in cellular signaling and in the regulation of gene expression. A number of biochemical and physiologic stimuli, such as perturbation in redox status, expression of misfolded proteins, altered glyc(osyl)ation and glucose deprivation, overloading of products of polyunsaturated fatty acid peroxidation (Hydroxynonenals, HNE) or cholesterol oxidation and decomposition, can disrupt redox homeostasis, impose stress and subsequently lead to accumulation of unfolded or misfolded proteins in brain cells. Alzheimer's (AD), Parkinson's (PD), Huntington's disease (HD), Amyothrophic lateral sclerosis (ALS) and Friedreich ataxia (FRDA) are major neurological disorders associated with production of abnormal proteins and, as such, belong to the so called "protein conformational diseases". The Central Nervous System has evolved highly specific signaling pathways called the unfolded protein response to cope with the accumulation of unfolded or misfolded proteins. Recent discoveries of the mechanisms of cellular stress signaling have led to major new insights into the diverse processes that are regulated by cellular stress response. Thus, the pathogenic dysfunctional aggregation of proteins in non-native conformations is associated with metabolic derangements and excessive production of ROS. The brain response to detect and control metabolic or oxidative stress is accomplished by a complex network of "longevity assurance processes" integrated to the expression of genes termed vitagenes. Heat shock proteins are a highly conserved system responsible for the preservation and repair of correct protein conformation. Heme oxygenase-1, a inducible and redox-regulated enzyme, is currently considered as having an important role in cellular antioxidant defense. A neuroprotective effect, due to its heme degrading activity, and tissue-specific antioxidant effects due to its products CO and biliverdin, this latter being further reduced by biliverdin reductase in bilirubin is an emerging concept. There is a current interest in dietary compounds that can inhibit, retard or reverse the multi-stage pathophysiology of Alzheimer disease, with a chronic inflammatory response, brain injury and beta-amyloid associated pathology. Curcumin and ferulic acid, two powerful antioxidants, the first from the curry spice turmeric and the second a major constituent of fruit and vegetables, have emerged as strong inducers of the heat shock response. Food supplementation with curcumin and ferulic acid is considered a nutritional approach to reduce oxidative damage and amyloid pathology in Alzheimer disease. This review summarizes the complex regulation of cellular stress signaling and its relevance to human physiology and disease.


Subject(s)
Neurodegenerative Diseases/physiopathology , Reactive Oxygen Species/metabolism , Stress, Physiological/physiopathology , Animals , Antioxidants/physiology , Female , Free Radicals/metabolism , Heme Oxygenase (Decyclizing)/physiology , Humans , Longevity/physiology , Male , Nitric Oxide/physiology , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Oxidoreductases Acting on CH-CH Group Donors/physiology , Reactive Nitrogen Species/physiology , Signal Transduction , Thioredoxin-Disulfide Reductase/physiology , Thioredoxins/metabolism
11.
Exp Biol Med (Maywood) ; 228(5): 517-24, 2003 May.
Article in English | MEDLINE | ID: mdl-12709579

ABSTRACT

Increasing evidence supports the notion that reduction of cellular expression and activity of antioxidant proteins and the resulting increase of oxidative stress are fundamental causes in the aging processes and neurodegenerative diseases. In the present study, we evaluated, in the brains of young and aged rats, the gene expression profiles of two inducible proteins critically involved in the cellular defense against endogenous or exogenous oxidants: heme oxygenase-1 (HO-1) and manganese superoxide dismutase-2 (SOD-2). SOD-2 is an essential antioxidant and HO-1 has been reported to be very active in regulating cellular redox homeostasis. Deregulation of these enzymes has been extensively reported to play a crucial role in the pathogenesis of neurodegenerative disorders. To measure the regional distribution of HO-1 and SOD-2 transcript levels in the rat brain, we have developed a real time quantitative reverse transcription-polymerase chain reaction protocol. Although these two genes presented a highly dissimilar range of expression, with SOD-2 >HO-1, both transcripts were highly expressed in the cerebellum and the hippocampus, showing in a different scale a strikingly parallel distribution gradient. To further investigate the regional brain expression of these mRNAs, we performed in situ hybridization using specific riboprobes. In situ hybridization results showed that both transcripts were highly concentrated in the hippocampus, the cerebellum and some specific regions of the brain cortex. We have also quantified, by reverse transcription-polymerase chain reaction, the brain expression of HO-1 and SOD-2 mRNAs in middle aged (12 months) and aged (28 months) rats. We found that the hippocampus of aged rats presents a significant down regulation of SOD2 mRNA expression and a parallel upregulation of HO-1 mRNA compared with young (6 months) and middle-aged rats. Furthermore, in the cerebellum of the aged rats, we detected a parallel significant upregulation of both HO-1 and SOD-2 transcripts. These regional age-dependent differences may help to explain the increased susceptibility to oxidative damage in these two brain areas during aging.


Subject(s)
Aging/physiology , Brain/enzymology , Heme Oxygenase (Decyclizing)/metabolism , Homeostasis , Superoxide Dismutase/metabolism , Animals , Brain/physiology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , In Situ Hybridization , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Oxidation-Reduction , Rats , Rats, Wistar , Superoxide Dismutase/genetics
12.
Neurochem Res ; 28(2): 187-94, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12608693

ABSTRACT

A great deal of increasing evidence designs PARP as a multifunctional protein implicated in many cellular functions. Much interest is emerging to understand the precise mechanisms by which PARP mediates genome stabilization and protection against damage, as well as its involvement in cell death, either apoptotic or necrotic. Aside from the clearly established role of PARP hyperactivation in necrotic cell death, after excessive DNA damage and energy failure, it appears to be actively involved in the phenomenon of apoptosis. However, its exact role is still controversial. The identification of several enzymes sharing the poly(ADP-ribose) polymerase catalytic domain (PARPs), but with different features and subcellular localization, has opened a new perspective in the field of poly(ADP-ribosyl)ation. The picture of the role of PARP in the control of cell homeostasis became even more complex after demonstration of its implication in the regulation of gene transcription. The notion that energy failure is the sole mechanism by which PARP promotes cell death is therefore under reevaluation.


Subject(s)
Cell Death , Cell Survival , Oxidative Stress , Poly(ADP-ribose) Polymerases/metabolism , Animals , DNA Damage , Gene Expression Regulation
13.
Neurochem Res ; 27(4): 319-23, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11958534

ABSTRACT

In the present study, we investigated the effect of Ginkgo biloba extract, EGb 761, and one of its components, bilobalide, on gene expression of subunit 1 of mitochondrial NADH dehydrogenase (ND1) in PC12 cells. By Northern blot analysis we found a approximately 2-fold significant increase in NDI mRNA level, after 48 and 72 h exposure to 100 microg/ml EGb 761 and to 10 microg/ml bilobalide. We also evaluated, by oxygraphy measurements, mitochondrial respiration during state 3 and state 4. In cells treated with EGb 761 and bilobalide for 48 and 72 h, state 4 respiration was significantly decreased, and the respiratory control ratio was increased. These results provide evidence that EGb 761 and bilobalide exert their protective effects by up-regulating mitochondrial ND1 gene expression and by decreasing state 4 respiration, whose increase is thought to be responsible for oxidative damage.


Subject(s)
Antioxidants/pharmacology , Cyclopentanes/pharmacology , Diterpenes , Furans/pharmacology , Mitochondria/metabolism , NADH Dehydrogenase/genetics , Oxygen Consumption/drug effects , Plant Extracts/pharmacology , RNA, Messenger/genetics , Animals , Gene Expression Regulation, Neoplastic/drug effects , Ginkgo biloba , Ginkgolides , Kinetics , Mitochondria/drug effects , PC12 Cells , Pheochromocytoma , RNA, Messenger/drug effects , Rats , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...