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










Publication year range
1.
Bioorg Chem ; 116: 105315, 2021 11.
Article in English | MEDLINE | ID: mdl-34496319

ABSTRACT

Chalcones and their derivatives have been described as promising compounds with antiproliferative activity against leukemic cells. This study aimed to investigate the cytotoxic effect of three synthetic chalcones derived from 1-naphthylacetophenone (F07, F09, and F10) in acute leukemia cell lines (K562 and Jurkat) and examine the mechanisms of cell death induced by these compounds. The three compounds were cytotoxic to K562 and Jurkat cells, with IC50 values ranging from 1.03 to 31.66 µM. Chalcones induced intrinsic and extrinsic apoptosis, resulting in activation of caspase-3 and DNA fragmentation. F07, F09, and F10 were not cytotoxic to human peripheral blood mononuclear cells, did not produce any significant hemolytic activity, and did not affect platelet aggregation after ADP stimulation. These results, combined with calculations of molecular properties, suggest that chalcones F07, F09, and F10 are promising molecules for the development of novel antileukemic drugs.


Subject(s)
Acetophenones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Acetophenones/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chalcones/chemical synthesis , Chalcones/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship
2.
J Pharm Pharm Sci ; 24: 23-36, 2021.
Article in English | MEDLINE | ID: mdl-33735604

ABSTRACT

BACKGROUND: Current therapies for acute leukemias (ALs) are associated with severe adverse reactions and high relapse rates, which makes the search for new antileukemic agents a necessity. Therefore, the aim of this study was to evaluate the effects of a new sulfonamide, S1, in AL cells K562 and Jurkat. METHODS: The cytotoxic activity of S1 was assessed using MTT method. The involvement of apoptosis in the mechanism of cell death was assessed by flow cytometry and fluorescence microscopy. RESULTS: Our results demonstrated that S1 induced morphological changes suggestive of apoptosis in both K562 and Jurkat cells. Additionally, S1 was not cytotoxic to normal erythrocytes and mononuclear cells and had a highly selective cytotoxicity for AL lineages. The mechanisms of cell death induced by S1 in K562 cells involves cell cycle arrest at G2/M phase and the activation of both extrinsic and intrinsic apoptosis, with an increased FasR and AIF expression and the loss of mitochondrial potential. As for Jurkat, we observed cell cycle blockade at G0/G1 phase, phosphatidylserine exposure and the involvement of intrinsic apoptosis only, with mitochondrial potential loss and a reduced expression of Survivin.  Although sulfonamide S1 did not altered Bcl-2 and Bax expression in AL cell lines, it was able to activate caspase-3 in K562 cells. CONCLUSION: Our results suggest that sulfonamide S1 may be a promising candidate for the development of new drugs for the treatment of ALs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzene Derivatives/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzene Derivatives/chemical synthesis , Benzene Derivatives/chemistry , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Jurkat Cells , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Molecular Structure , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
3.
Neurochem Res ; 45(12): 2868-2883, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32968860

ABSTRACT

Recent evidence suggests that young rodents submitted to high fructose (FRU) diet develop metabolic, and cognitive dysfunctions. However, it remains unclear whether these detrimental effects of FRU intake can also be observed in middle-aged mice. Nine months-old C57BL/6 female mice were fed with water (Control) or 10% FRU in drinking water during 12 weeks. After that, metabolic, and neurochemical alterations were evaluated, focusing on neurotransmitters, and antioxidant defenses. Behavioral parameters related to motor activity, memory, anxiety, and depression were also evaluated. Mice consuming FRU diet displayed increased water, and caloric intake, resulting in weight gain, which was partially compensated due to decreased food pellet intake. FRU fed animals displayed increased plasma glucose, and cholesterol levels, which was not observed in overnight-fasted animals. Superoxide dismutase (SOD), and catalase (CAT) activities were markedly decreased in the prefrontal cortex of animals receiving FRU diet, while glutathione peroxidase (GPx) slightly increased. Liver (lower GPx), striatum (higher SOD and lower CAT), and hippocampus (no changes) were less impacted. No changes were observed in glutathione reductase, and thioredoxin reductase activities, two ancillary enzymes for peroxide detoxification. FRU intake did not alter serotonin, dopamine, and norepinephrine levels in the hippocampus, prefrontal cortex, and striatum. No significant alterations were observed in working, and short-term spatial memory; and in anxiety- and depressive-like behaviors in animals treated with FRU. Increased locomotor activity was observed in FRU-fed middle-aged mice, as evaluated in the open field, elevated plus-maze, Y maze, and object location tasks. Overall, these results demonstrate that high FRU consumption can disturb antioxidant defenses, and increase locomotor activity in middle-aged mice, open the opportunity for further studies to address the underlying mechanisms related to these findings.


Subject(s)
Catalase/metabolism , Fructose/pharmacology , Locomotion/drug effects , Superoxide Dismutase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Elevated Plus Maze Test , Female , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Open Field Test/drug effects
4.
Inflammation ; 41(6): 2060-2067, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30030654

ABSTRACT

In this study, we evaluated the effects of high-intensity swimming in an experimental model of acute lung injury (ALI) induced by lipopolysaccharide (LPS) on lung inflammation and antioxidant defenses. Balb/C male mice were submitted to exercise (30 min/day, 5 days/week, for a period of 3 weeks) prior to LPS instillation in the lung. Twenty-four hours after delivery of LPS (10 µg/animal), mice were euthanized and bronchoalveolar fluid (BALF) was obtained for cell counting and analysis of cytokines by ELISA. Lung tissue was used to evaluate antioxidant defenses. LPS instillation resulted in an increase in total and mononuclear cells, IL-1ß, TNF-α, and IL-6 in BALF. LPS instillation also altered IL-10 and IL-ra levels in BALF and induced antioxidant defenses (glutathione, superoxide dismutase, catalase, and glutathione peroxidase) in the lung. Protein carbonyl increased in the LPS-treated animals. High-intensity swimming prevented all these changes induced by LPS. Significance: Therefore, this experimental protocol of high-intensity swimming showed a protective effect on ALI, decreasing inflammatory processes and preventing disturbances in antioxidant defenses into the lungs.


Subject(s)
Antioxidants/metabolism , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Physical Conditioning, Animal/physiology , Pneumonia/metabolism , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Male , Mice , Mice, Inbred BALB C , Pneumonia/chemically induced , Swimming
5.
Toxicol In Vitro ; 42: 273-280, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28461233

ABSTRACT

Thiol homeostasis has a critical role in the maintenance of proper cellular functions and survival, being coordinated by the action of several reductive enzymes, including glutathione (GSH)/glutathione reductase (GR) and thioredoxin (Trx)/thioredoxin reductase (TrxR) systems. Here, we investigated the effects of the GR inhibitor 2-acetylamino-3-[4-(2-acetylamino-2-carboxyethylsulfanylthiocarbonylamino)phenylthiocarbamoylsulfanyl]propionic acid (2-AAPA) on the activity of thiol reductases (GR and TrxR), redox balance and mitochondrial function of A172 glioblastoma cells. 2-AAPA inhibited cell GR (IC50=6.7µM) and TrxR (IC50=8.7µM). A significant decrease in the cellular ability to decompose cumene hydroperoxide was observed and associated to a greater susceptibility to this peroxide. The redox state of peroxiredoxins (Prx1, Prx2 and Prx3) was markedly shifted to dimer 30min after treatment with 100µM 2-AAPA, an event preceding 2-AAPA-induced decrease in cell viability. Furthermore, mitochondrial function was also severely impaired, leading to a decrease in the respiratory control ratio, reserve capacity, and ATP synthesis-coupled respiration, as well as an increase in mitochondrial membrane potential. Our results indicate that inhibition of GR and TrxR activities, disruption of the ability to detoxify peroxides, increased oxidation of Prxs, as well as compromised mitochondrial function represent early events mediating 2-AAPA toxicity to A172 glioblastoma cells.


Subject(s)
Acetylcysteine/analogs & derivatives , Antineoplastic Agents/pharmacology , Glutathione Reductase/antagonists & inhibitors , Thiocarbamates/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Acetylcysteine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glutathione Reductase/metabolism , Humans , Hydrogen Peroxide/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Peroxiredoxins/metabolism , Thioredoxin-Disulfide Reductase/metabolism
6.
Neurotox Res ; 32(3): 340-350, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28478530

ABSTRACT

Thioredoxin (Trx) and glyoxalase (Glo) systems have been suggested to be molecular targets of methylglyoxal (MGO). This highly reactive endogenous compound has been associated with the development of neurodegenerative pathologies and cell death. In the present study, the glutathione (GSH), Trx, and Glo systems were investigated to understand early events (0.5-3 h) that may determine cell fate. It is shown for the first time that MGO treatment induces an increase in glutathione reductase (GR) protein in hippocampal slices (1 h) and HT22 nerve cells (0.5 and 2.5 h). Thioredoxin interacting protein (Txnip), thioredoxin reductase (TrxR), Glo1, and Glo2 were markedly increased (2- to 4-fold) in hippocampal slices and 1.2- to 1.3-fold in HT22 cells. This increase in protein levels in hippocampal slices was followed by a corresponding increase in GR, TrxR, and Glo1 activities, but not in HT22 cells. In these cells, GR and TrxR activities were decreased by MGO. This result is in agreement with the idea that MGO can affect the Trx/TrxR reducing system, and now we show that GR and Txnip can also be affected by MGO. Impairment in the GR or TrxR reducing capacity can impair peroxide removal by glutathione peroxidase and peroxiredoxin, as both peroxidases depend on reduced GSH and Trx, respectively. In this regard, inhibition of GR and TrxR by 2-AAPA or auranofin, respectively, potentiated MGO toxicity in differentiated SH-SY5Y cells. Overall, MGO not only triggers a clear defense response in hippocampal slices and HT22 cells but also impairs the Trx/TrxR and GSH/GR reducing couples in HT22 cells. The increased MGO toxicity caused by inhibition of GR and TrxR with specific inhibitors, or their inhibition by MGO treatment, supports the notion that both reducing systems are relevant molecular targets of MGO.


Subject(s)
Cell Survival/physiology , Glutathione Reductase/metabolism , Pyruvaldehyde/toxicity , Thioredoxins/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Female , Glutathione/metabolism , Hippocampus/enzymology , Humans , Mice , Neurons/enzymology , Neuroprotection/physiology , Pyruvaldehyde/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Tissue Culture Techniques
7.
Behav Brain Res ; 320: 365-373, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27825895

ABSTRACT

Pramipexole (PPX), a dopamine D2/3 receptor preferring agonist, is currently in use for the treatment of Parkinson's disease symptoms and restless legs syndrome. Recently, anti-inflammatory properties of PPX have been shown in an autoimmune model of multiple sclerosis, and case reports indicate PPX ameliorates depressive symptoms. Since peripheral inflammation is known to induce depression-like behavior in rodents, we assessed the potential antidepressant effect of PPX in an inflammatory model of depression induced by LPS. Repeated (daily for 7days, 1mg/kg, i.p.), but not acute (1h before LPS) treatment with PPX abolished the depression-like behavior induced by LPS (0.1mg/kg, i.p.) in the forced swim test, and the anhedonic behavior in the splash test. Interestingly, PPX per se decreased interleukin 1ß levels and reversed LPS-induced increase in its content in mice hippocampus⋅ Repeated PPX treatment also prevented the increase in hippocampal levels of the 3-nitrotyrosine protein adducts induced by LPS. Haloperidol (0.2mg/kg, i.p.) and sulpiride (50mg/kg, i.p.) were unable to prevent the antidepressant-like effect of PPX in LPS-treated mice. Altogether, these results suggest that the observed antidepressant-like effect of PPX in LPS-treated mice may be dependent on its anti-inflammatory properties and may not be related to dopamine D2 receptor activation.


Subject(s)
Benzothiazoles/therapeutic use , Depression/drug therapy , Depression/etiology , Dopamine Agonists/therapeutic use , Inflammation/complications , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/metabolism , Illness Behavior/physiology , Inflammation/chemically induced , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Locomotion/drug effects , Malondialdehyde/metabolism , Mice , Pramipexole , Swimming/psychology , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
8.
Cell Mol Neurobiol ; 36(6): 1015-1022, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26749581

ABSTRACT

α-Tocopheryl phosphate (αTP) is a phosphorylated form of α-tocopherol. Since it is phosphorylated in the hydroxyl group that is essential for the antioxidant property of α-tocopherol, we hypothesized that αTP would modulate the antioxidant system, rather than being an antioxidant agent per se. α-TP demonstrated antioxidant activity in vitro against iron-induced oxidative stress in a mitochondria-enriched fraction preparation treated with 30 or 100 µM α-TP. However, this effect was not observed ex vivo with mitochondrial-enriched fraction from mice treated with an intracerebroventricular injection of 0.1 or 1 nmol/site of αTP. Two days after treatment (1 nmol/site αTP), peroxiredoxin 2 (Prx2) and glutathione reductase (GR) expression and GR activity were decreased in cerebral cortex and hippocampus. Glutathione content, glutathione peroxidase, and thioredoxin reductase activities were not affected by αTP. In conclusion, the persistent decrease in GR and Prx2 protein content is the first report of an in vivo effect of αTP on protein expression in the mouse brain, potentially associated to a novel and biologically relevant function of this naturally occurring compound.


Subject(s)
Antioxidants/pharmacology , Brain/drug effects , Glutathione Reductase/metabolism , Oxidative Stress/drug effects , Peroxiredoxins/metabolism , alpha-Tocopherol/analogs & derivatives , Animals , Antioxidants/metabolism , Brain/metabolism , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , alpha-Tocopherol/pharmacology
9.
PLoS One ; 10(9): e0137273, 2015.
Article in English | MEDLINE | ID: mdl-26332044

ABSTRACT

Studies have reported that exposure to diesel exhaust particles (DEPs) induces lung inflammation and increases oxidative stress, and both effects are susceptible to changes via regular aerobic exercise in rehabilitation programs. However, the effects of exercise on lungs exposed to DEP after the cessation of exercise are not clear. Therefore, the aim of this study was to evaluate the effects of high-intensity swimming on lung inflammation and oxidative stress in mice exposed to DEP concomitantly and after exercise cessation. Male Swiss mice were divided into 4 groups: Control (n = 12), Swimming (30 min/day) (n = 8), DEP (3 mg/mL-10 µL/mouse) (n = 9) and DEP+Swimming (n = 8). The high-intensity swimming was characterized by an increase in blood lactate levels greater than 1 mmoL/L between 10th and 30th minutes of exercise. Twenty-four hours after the final exposure to DEP, the anesthetized mice were euthanized, and we counted the number of total and differential inflammatory cells in the bronchoalveolar fluid (BALF), measured the lung homogenate levels of IL-1ß, TNF-α, IL-6, INF-Ï«, IL-10, and IL-1ra using ELISA, and measured the levels of glutathione, non-protein thiols (GSH-t and NPSH) and the antioxidant enzymes catalase and glutathione peroxidase (GPx) in the lung. Swimming sessions decreased the number of total cells (p<0.001), neutrophils and lymphocytes (p<0.001; p<0.05) in the BALF, as well as lung levels of IL-1ß (p = 0.002), TNF-α (p = 0.003), IL-6 (p = 0.0001) and IFN-Ï« (p = 0.0001). However, the levels of IL-10 (p = 0.01) and IL-1ra (p = 0.0002) increased in the swimming groups compared with the control groups, as did the CAT lung levels (p = 0.0001). Simultaneously, swimming resulted in an increase in the GSH-t and NPSH lung levels in the DEP group (p = 0.0001 and p<0.002). We concluded that in this experimental model, the high-intensity swimming sessions decreased the lung inflammation and oxidative stress status during DEP-induced lung inflammation in mice.


Subject(s)
Disease Models, Animal , Oxidative Stress , Pneumonia/physiopathology , Swimming , Vehicle Emissions/toxicity , Animals , Bronchoalveolar Lavage Fluid , Catalase/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Glutathione/metabolism , Inflammation Mediators/metabolism , Lung/drug effects , Lung/enzymology , Lung/metabolism , Male , Mice , Pneumonia/chemically induced , Sulfhydryl Compounds/metabolism
10.
Biomed Res Int ; 2014: 723060, 2014.
Article in English | MEDLINE | ID: mdl-24689052

ABSTRACT

Traumatic brain injury (TBI) is frequently associated with abnormal blood-brain barrier function, resulting in the release of factors that can be used as molecular biomarkers of TBI, among them GFAP, UCH-L1, S100B, and NSE. Although many experimental studies have been conducted, clinical consolidation of these biomarkers is still needed to increase the predictive power and reduce the poor outcome of TBI. Interestingly, several of these TBI biomarkers are oxidatively modified to carbonyl groups, indicating that markers of oxidative stress could be of predictive value for the selection of therapeutic strategies. Some drugs such as corticosteroids and progesterone have already been investigated in TBI neuroprotection but failed to demonstrate clinical applicability in advanced phases of the studies. Dietary antioxidants, such as curcumin, resveratrol, and sulforaphane, have been shown to attenuate TBI-induced damage in preclinical studies. These dietary antioxidants can increase antioxidant defenses via transcriptional activation of NRF2 and are also known as carbonyl scavengers, two potential mechanisms for neuroprotection. This paper reviews the relevance of redox biology in TBI, highlighting perspectives for future studies.


Subject(s)
Antioxidants/metabolism , Biomarkers/metabolism , Brain Injuries/metabolism , Brain Injuries/pathology , Oxidative Stress , Animals , Brain Injuries/physiopathology , Humans , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
11.
Behav Brain Res ; 237: 176-84, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23018126

ABSTRACT

The olfactory bulbectomy (OB) animal model of depression is a well-established model that is capable of detecting antidepressant activity following chronic drug therapy, and the surgery results in behavioral and biochemical changes that are reminiscent of various symptoms of depression. In the present study, we investigated the degree to which 14 days of p.o. administration of the classic antidepressant fluoxetine (10mg/kg) were able to reverse OB-induced changes in behavior (namely, hyperactivity in the open-field test and reduced motivational and self-care behaviors in the splash test) and in the activation of hippocampal cell signaling pathways that are thought to be involved in synaptic plasticity. OB caused significant increases in ERK1 and CREB (Ser(133)) phosphorylation and in the expression of BDNF immunocontent, all of which were prevented by fluoxetine administration. Moreover, fluoxetine administration also caused a significant decrease in ERK2 phosphorylation in mice that had undergone OB. Neither Akt nor GSK-3ß phosphorylation was altered in any experimental condition. In conclusion, the present study shows that OB can induce significant behavioral changes that are accompanied by the activation of hippocampal signaling pathways, namely the ERK1/CREB/BDNF pathway, which is involved in the synaptic plasticity. Conversely, fluoxetine prevented these OB-induced behavioral changes and avoided the activation of ERK1/CREB/BDNF in the hippocampus. Taken together, our results extend the data from the existing literature regarding OB-induced behavioral and neurochemical changes, and suggest a possible underlying mechanism that can account for the antidepressant effect of fluoxetine in this model.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Fluoxetine/pharmacology , Hippocampus/cytology , Neuronal Plasticity/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Analysis of Variance , Anhedonia/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein/metabolism , Disease Models, Animal , Exploratory Behavior/drug effects , Female , Food Preferences/drug effects , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hyperkinesis/drug therapy , Hyperkinesis/etiology , MAP Kinase Signaling System/drug effects , Mice , Olfaction Disorders/complications , Olfaction Disorders/etiology , Olfactory Bulb/surgery , Oncogene Protein v-akt/metabolism
12.
Free Radic Biol Med ; 51(1): 69-77, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21440059

ABSTRACT

Cells are endowed with several overlapping peroxide-degrading systems whose relative importance is a matter of debate. In this study, three different sources of neural cells (rat hippocampal slices, rat C6 glioma cells, and mouse N2a neuroblastoma cells) were used as models to understand the relative contributions of individual peroxide-degrading systems. After a pretreatment (30 min) with specific inhibitors, each system was challenged with either H2O2 or cumene hydroperoxide (CuOOH), both at 100 µM. Hippocampal slices, C6 cells, and N2a cells showed a decrease in the H2O2 decomposition rate (23-28%) by a pretreatment with the catalase inhibitor aminotriazole. The inhibition of glutathione reductase (GR) by BCNU (1,3-bis(2-chloroethyl)-1-nitrosourea) significantly decreased H2O2 and CuOOH decomposition rates (31-77%). Inhibition of catalase was not as effective as BCNU at decreasing cell viability (MTT assay) and cell permeability or at increasing DNA damage (comet test). Impairing the thioredoxin (Trx)-dependent peroxiredoxin (Prx) recycling by thioredoxin reductase (TrxR) inhibition with auranofin neither potentiated peroxide toxicity nor decreased the peroxide-decomposition rate. The results indicate that neural peroxidatic systems depending on Trx/TrxR for recycling are not as important as those depending on GSH/GR. Dimer formation, which leads to Prx2 inactivation, was observed in hippocampal slices and N2a cells treated with H2O2, but not in C6 cells. However, Prx-SO3 formation, another form of Prx inactivation, was observed in all neural cell types tested, indicating that redox-mediated signaling pathways can be modulated in neural cells. These differences in Prx2 dimerization suggest specific redox regulation mechanisms in glia-derived (C6) compared to neuron-derived (N2a) cells and hippocampal slices.


Subject(s)
Catalase/metabolism , Glutathione/metabolism , Neurons/metabolism , Peroxides/metabolism , Peroxiredoxins/metabolism , Animals , Benzene Derivatives/pharmacology , Catalase/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage , Glutathione Reductase/antagonists & inhibitors , Glutathione Reductase/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Mice , Neurons/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism
13.
Behav Brain Res ; 209(2): 249-59, 2010 Jun 19.
Article in English | MEDLINE | ID: mdl-20144659

ABSTRACT

This study investigated the antidepressant potential of alpha-tocopherol, the most active and abundant form of vitamin E, in the forced swim test (FST) and tail suspension test (TST). The acute oral treatment with alpha-tocopherol at the doses of 30 and 100mg/kg reduced the immobility time in the FST and in the TST. A single i.c.v. administration of alpha-tocopheryl phosphate, a water-soluble analogue of alpha-tocopherol, also reduced the immobility time in the FST (0.1 and 1 nmol/site) and in the TST (0.1 nmol/site). In addition, the long-term treatment (28 days) with alpha-tocopherol (10mg/kg, p.o.) significantly reduced the immobility time in the FST. Moreover, a subeffective dose of alpha-T (10mg/kg, p.o.) potentiated the effect of fluoxetine (10mg/kg, p.o.) in the FST. The long-term treatment with alpha-T was able to increase the glutathione (GSH) antioxidant defense system, while the acute treatment was not. The long-term treatment with alpha-tocopherol (10mg/kg) increased the GSH levels in the hippocampus and in the prefrontal cortex and increased the glutathione peroxidase and glutathione reductase activity in the hippocampus (10mg/kg) and in the prefrontal cortex (10-100mg/kg). The long-term treatment with fluoxetine (10mg/kg, p.o.), a positive control, was also able to increase the GSH levels in the hippocampus, but failed to alter the activity of both enzymes. Besides the specific antidepressant-like effect, long-term, but not the acute treatment with alpha-T, especially in the doses that produced an antidepressant-like effect (10mg/kg), improved the antioxidant defenses in the mouse hippocampus and prefrontal cortex, two structures closely implicated in the pathophysiology of depression.


Subject(s)
Depressive Disorder/drug therapy , Immobility Response, Tonic/drug effects , alpha-Tocopherol/analogs & derivatives , alpha-Tocopherol/pharmacology , Analysis of Variance , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Exploratory Behavior/drug effects , Female , Fluoxetine/pharmacology , Glutathione/analysis , Hindlimb Suspension , Hippocampus/chemistry , Hippocampus/drug effects , Mice , Motor Activity/drug effects , Prefrontal Cortex/chemistry , Prefrontal Cortex/drug effects , Swimming , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...