Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Neurotox Res ; 37(1): 210-226, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31422567

ABSTRACT

Exposure to environmental contaminants represents an important etiological factor in sporadic Parkinson's disease (PD). It has been reported that PD could arise from events that occur early in development and that lead to delayed adverse consequences in the nigrostriatal dopaminergic system at adult life. We investigated the occurrence of late nigrostriatal dopaminergic neurotoxicity induced by exposures to the pesticides paraquat (PQ) and maneb (MB) during the early postnatal period in mice, as well as whether the exposure to pesticides during development could enhance mice vulnerability to subsequent challenges. Male Swiss mice were exposed to a combination of 0.3 mg/kg PQ and 1.0 mg/kg MB (PQ + MB) from postnatal (PN) day 5 to 19. PN exposure to pesticides neither induced mortally nor modified motor-related parameters. However, PN pesticides exposure decreased the number of tyrosine hydroxylase (TH)- and dopamine transporter (DAT)-positive neurons in the substantia nigra pars compacta (SNpc), as well as reduced TH and DAT immunoreactivity in the striatum. A parallel group of animals developmentally exposed to the pesticides was re-challenged at 3 months of age with 10 mg/kg PQ plus 30 mg/kg MB (twice a week, 6 weeks). Mice exposed to pesticides at both periods (PN + adulthood) presented motor deficits and reductions in the number of TH- and DAT-positive neurons in the SNpc. These findings indicate that the exposure to PQ + MB during the early PN period can cause neurotoxicity in the mouse nigrostriatal dopaminergic system, rendering it more susceptible to a subsequent adult re-challenge with the same pesticides.


Subject(s)
Central Nervous System Sensitization/drug effects , Dopaminergic Neurons/drug effects , Maneb/toxicity , Paraquat/toxicity , Age Factors , Animals , Cell Count , Corpus Striatum/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/metabolism , Male , Mice , Motor Skills/drug effects , Pars Compacta/metabolism , Tyrosine 3-Monooxygenase/metabolism
2.
Mol Biol Rep ; 46(1): 751-762, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30511305

ABSTRACT

Selenium (Se) is an essential trace element for humans; its intake is needed to allow the proper synthesis of 25 different selenoproteins that are necessary to the normal functioning of several organs, including the brain. Accordingly, decreased Se levels have been associated with neurological disorders. In the present study, we investigated the potential beneficial effects of Se, as sodium selenite, against 3-nitropropionic acid (3-NP)-induced oxidative stress in primary cultures of mouse cortical neurons. 3-NP treatment caused a significant decrease in cellular viability, which was accompanied by decreases in mitochondrial complex II activity and reduced glutathione (GSH) content, as well as increases in reactive oxygen species (ROS) generation and oxidized glutathione (GSSG) levels. Sodium selenite pretreatment (6 days) attenuated 3-NP-induced decrease in cell viability. In addition, sodium selenite pretreatment significantly protected against 3-NP-induced increase in ROS generation and decrease in GSH/GSSG ratio. Of note, sodium selenite pretreatment did not change 3-NP-induced decrease of mitochondrial complex II activity, suggesting that Se modulates secondary events resultant from 3-NP-induced mitochondrial dyshomeostasis. In addition, sodium selenite pretreatment significantly increased glutathione peroxidase (GPx) activity. Our data provide insights into the mechanism of protection by sodium selenite, which is related, at least in part, to GPx induction.


Subject(s)
Cerebral Cortex/pathology , Neurons/pathology , Neuroprotective Agents/pharmacology , Nitro Compounds/toxicity , Oxidative Stress/drug effects , Propionates/toxicity , Sodium Selenite/pharmacology , Animals , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Glutathione Peroxidase/metabolism , MAP Kinase Signaling System/drug effects , Mice , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects
3.
Food Chem Toxicol ; 120: 603-615, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30086348

ABSTRACT

Fish and shellfish, which represent important sources of nutrients (i.e., n-3 fatty acids), can contain significant amounts of methylmercury (MeHg), a neurotoxic compound. We investigated the potential neuroprotective effects of perinatal treatment with dietary n-3 fatty acids against MeHg-induced neurotoxicity. Pregnant mice were divided in 4 groups: (i) Control; (ii) MeHg; (iii) n-3 enriched diet and (iv) n-3 enriched diet + MeHg. The treatments were performed from gestational day 1 to postnatal day 21. Twenty-four hours after treatments, motor-related behavioral tests, as well as the analyses of cerebellar biochemical, histological and immunohistochemical parameters related to neuronal and glial homeostasis, were performed. Maternal exposure to MeHg induced motor coordination impairment and cerebellar MeHg accumulation in the offspring and n-3 fatty acids treatment did not prevent these effects. The immunocontent of proteins related to synaptic homeostasis, glial fibrillary acidic protein immunostaining and morphology were not significantly altered in the pups perinatally exposed to MeHg and/or n-3 diet. The results indicate that perinatal exposure to MeHg causes motor coordination impairment even with no evident changes on the evaluated cerebellar biochemical and histological parameters. The performed exposure protocol was unable to show beneficial effects of n-3 fatty acids supplementation against MeHg-induced motor coordination.


Subject(s)
Behavior, Animal/drug effects , Cerebellum/drug effects , Fatty Acids, Omega-3/pharmacology , Maternal Exposure , Methylmercury Compounds/toxicity , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Animals , Cerebellum/metabolism , Cerebellum/physiopathology , Feeding Behavior/drug effects , Female , Glial Fibrillary Acidic Protein/metabolism , Homeostasis , Mice , Neuroglia/drug effects , Neurons/drug effects , Pregnancy
4.
Neurochem Res ; 43(3): 745-759, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29362970

ABSTRACT

Systemic inflammation triggered by lipopolysaccharide (LPS) administration disrupts blood-brain barrier (BBB) homeostasis in animal models. This event leads to increased susceptibility of several encephalic structures to potential neurotoxicants present in the bloodstream. In this study, we investigated the effects of alternate intraperitoneal injections of LPS on BBB permeability, social recognition memory and biochemical parameters in the striatum 24 h and 60 days after treatments. In addition, we investigated whether the exposure to a moderate neurotoxic dose of the herbicide paraquat could potentiate LPS-induced neurotoxicity. LPS administration caused a transient disruption of BBB integrity, evidenced by increased levels of exogenously administered sodium fluorescein in the striatum. Also, LPS exposure caused delayed impairment in social recognition memory (evaluated at day 38 after treatments) and increase in the striatal levels of 3-nitrotyrosine. These events were observed in the absence of significant changes in motor coordination and in the levels of tyrosine hydroxylase (TH) in the striatum and substantia nigra. PQ exposure, which caused a long-lasting decrease of striatal mitochondrial complex I activity, did not modify LPS-induced behavioral and striatal biochemical changes. The results indicate that systemic administration of LPS causes delayed social recognition memory deficit and striatal nitrosative stress in adult mice and that the coexposure to a moderately toxic dose of PQ did not magnify these events. In addition, PQ-induced inhibition of striatal mitochondrial complex I was also not magnified by LPS exposure, indicating the absence of synergic neurotoxic effects of LPS and PQ in this experimental model.


Subject(s)
Behavior, Animal/drug effects , Corpus Striatum/drug effects , Lipopolysaccharides/pharmacology , Nitrosative Stress/drug effects , Paraquat/toxicity , Animals , Corpus Striatum/metabolism , Male , Memory/drug effects , Mice , Neostriatum/drug effects , Neostriatum/metabolism , Neurotoxicity Syndromes/drug therapy , Substantia Nigra/drug effects , Substantia Nigra/metabolism
5.
Mol Neurobiol ; 54(2): 1513-1530, 2017 03.
Article in English | MEDLINE | ID: mdl-26852411

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by non-motor and motor disabilities. This study investigated whether succinobucol (SUC) could mitigate nigrostriatal injury caused by intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in mice. Moreover, the effects of SUC against MPTP-induced behavioral impairments and neurochemical changes were also evaluated. The quantification of tyrosine hydroxylase-positive (TH+) cells was also performed in primary mesencephalic cultures to evaluate the effects of SUC against 1-methyl-4-phenylpyridinium (MPP+) toxicity in vitro. C57BL/6 mice were treated with SUC (10 mg/kg/day, intragastric (i.g.)) for 30 days, and thereafter, animals received MPTP infusion (1 mg/nostril) and SUC treatment continued for additional 15 days. MPTP-infused animals displayed significant non-motor symptoms including olfactory and short-term memory deficits evaluated in the olfactory discrimination, social recognition, and water maze tasks. These behavioral impairments were accompanied by inhibition of mitochondrial NADH dehydrogenase activity (complex I), as well as significant decrease of TH and dopamine transporter (DAT) immunoreactivity in the substantia nigra pars compacta and striatum. Although SUC treatment did not rescue NADH dehydrogenase activity inhibition, it was able to blunt MPTP-induced behavioral impairments and prevented the decrease in TH and DAT immunoreactivities in substantia nigra (SN) and striatum. SUC also suppressed striatal astroglial activation and increased interleukin-6 levels in MPTP-intoxicated mice. Furthermore, SUC significantly prevented the loss of TH+ neurons induced by MPP+ in primary mesencephalic cultures. These results provide new evidence that SUC treatment counteracts early non-motor symptoms and neurodegeneration/neuroinflammation in the nigrostriatal pathway induced by intranasal MPTP administration in mice by modulating events downstream to the mitochondrial NADH dehydrogenase inhibition.


Subject(s)
Anticholesteremic Agents/therapeutic use , Corpus Striatum/drug effects , Parkinsonian Disorders/drug therapy , Probucol/analogs & derivatives , Substantia Nigra/drug effects , Animals , Anticholesteremic Agents/pharmacology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Female , Male , Memory Disorders/drug therapy , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Pregnancy , Probucol/pharmacology , Probucol/therapeutic use , Random Allocation , Rats , Rats, Sprague-Dawley , Smell/drug effects , Smell/physiology , Substantia Nigra/metabolism , Substantia Nigra/pathology
6.
Behav Brain Res ; 311: 31-38, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27185735

ABSTRACT

Although epidemiological studies have reported an association between hypercholesterolemia and mood disorders, there is a lack of data regarding depressive-like behavior in animal models of hypercholesterolemia. To address these questions, we assessed depressive-like behavior and hippocampal and cortical monoaminergic metabolism in three-month-old, low-density lipoprotein receptor knockout (LDLr(-/-)) and C57BL/6 wild-type mice. The LDLr(-/-) mice exhibited depressive-like behavior in the sucrose preference test, splash test, and tail suspension test. Increased monoamine oxidase (MAO) A and B activity was evidenced in the hippocampus of LDLr(-/-) mice. Furthermore, to address whether or not cholesterol modulates MAO activity, we exposed SH-SY5Y human neuroblastoma cells to human isolated low-density lipoprotein (LDL). Notably, LDL increased the activity of MAO-A and stimulated the reactive species generation in vitro. These findings indicate that depressive-like behavior in hypercholesterolemic mice is accompanied by alterations in the monoaminergic metabolism, providing new evidence about the association between hypercholesterolemia and depression.


Subject(s)
Depression/metabolism , Hypercholesterolemia/metabolism , Hypercholesterolemia/psychology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Hippocampus/metabolism , Humans , Lipoproteins, LDL/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Monoamine Oxidase/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics
7.
Behav Brain Res ; 305: 30-6, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26921691

ABSTRACT

Bradykinesia and hypokinesia represent well-known motor symptoms of Parkinson's disease (PD). While bradykinesia (slow execution of movements) is present in less affected PD patients and aggravates as the disease severity increases, hypokinesia (reduction of movement) seems to emerge prominently only in the more affected patients. Here we developed a model based on the central infusion of low dose (40µg) 6-hydroxydopamine (6-OHDA) in mice in an attempt to discriminate bradykinesia (accessed through forelimb inability) from hypokinesia (accessed through locomotor and exploratory activities). The potential beneficial effects of succinobucol against 6-OHDA-induced forelimb inability were also evaluated. One week after the beginning of treatment with succinobucol (i.p. injections, 10mg/kg/day), mice received a single i.c.v. infusion of 6-OHDA (40µg/site). One week after 6-OHDA infusion, general locomotor/exploratory activities (open field test), muscle strength (grid test), forelimb skill (single pellet task), as well as striatal biochemical parameters related to oxidative stress and cellular homeostasis (glutathione peroxidase, glutathione reductase and NADH dehydrogenases activities, lipid peroxidation and TH levels), were evaluated. 6-OHDA infusions did not change locomotor/exploratory activities and muscle strength, as well as the evaluated striatal biochemical parameters. However, 6-OHDA infusions caused significant reductions (50%) in the single pellet reaching task performance, which detects forelimb skill inability and can be used to experimentally identify bradykinesia. Succinobucol partially protected against 6-OHDA-induced forelimb inability. The decreased forelimb ability with no changes in locomotor/exploratory behavior indicates that our 6-OHDA-based protocol represents a useful tool to mechanistically study the dissociation of bradykinesia and hypokinesia in PD.


Subject(s)
Adrenergic Agents/administration & dosage , Forelimb/physiopathology , Hypokinesia/chemically induced , Hypokinesia/physiopathology , Oxidopamine/administration & dosage , Animals , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Histocompatibility Antigens Class I/metabolism , Hypokinesia/diagnosis , Injections, Intraventricular , Lipid Peroxidation/drug effects , Male , Mice , Muscle Strength/drug effects , Peptide Fragments/metabolism , Probucol/administration & dosage , Probucol/analogs & derivatives , Psychomotor Performance/drug effects , Thiobarbituric Acid Reactive Substances/metabolism
8.
Epilepsy Behav ; 55: 92-100, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26773677

ABSTRACT

Epilepsy is a brain function disorder characterized by unpredictable and recurrent seizures. The majority of patients with temporal lobe epilepsy (TLE), which is the most common type of epilepsy, have to live not only with seizures but also with behavioral alterations, including anxiety, psychosis, depression, and impaired cognitive functioning. The pilocarpine model has been recognized as an animal model of TLE. However, there are few studies addressing behavioral alterations in the maturation phase when evaluating the time course of the epileptogenic process after pilocarpine administration. Therefore, the present work was designed to analyze the neurobehavioral impairments of male adult Wistar rats during maturation and chronic phases in the pilocarpine model of epilepsy. Behavioral tests included: open-field tasks, olfactory discrimination, social recognition, elevated plus maze, and the forced swimming test. The main behavioral alterations observed in both maturation and chronic phases of the pilocarpine model were olfactory and short-term social memory deficits and decrease in the immobility time in the forced swimming test. Moreover, increased anxiety-like responses were only observed in the maturation phase. These findings indicate that early behavioral impairments can be observed in the pilocarpine model during the maturation phase, and these behavioral deficits also occur during the acquired epilepsy (chronic phase). Several of the neurobehavioral impairments that are associated with epilepsy in humans were observed in the pilocarpine-treated rats, thus, rendering this animal model a useful tool to study neuroprotective strategies as well as neurobiological and psychopathological mechanisms associated with epileptogenesis.


Subject(s)
Disease Models, Animal , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/psychology , Maze Learning/drug effects , Motor Activity/drug effects , Pilocarpine/toxicity , Animals , Anxiety/chemically induced , Anxiety/pathology , Anxiety/psychology , Epilepsy, Temporal Lobe/pathology , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Male , Maze Learning/physiology , Motor Activity/physiology , Rats , Rats, Wistar , Swimming/physiology , Swimming/psychology , Time Factors
9.
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
10.
Mol Neurobiol ; 53(2): 1280-1295, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25619973

ABSTRACT

Succinobucol (succinyl ester of probucol) is a lipid-lowering compound with anti-inflammatory and antioxidant properties. Recent experimental evidence has highlighted the potential neuroprotective effects of succinobucol. In the present study, cultured neuroblastoma (SH-SY5Y) cells were used to investigate mechanisms mediating the potential protective effect of succinobucol against mitochondrial metabolic impairment and oxidative stress induced by 3-nitropropionic acid (3-NP), a succinate dehydrogenase inhibitor that has been used in experimental models of the Huntington disease (HD). 3-NP decreased cellular viability after 24 h of incubation. This decline in cellular viability was preceded by (i) reduced mitochondrial complex II activity, (ii) increased reactive species generation, (iii) decreased mitochondrial membrane potential (ΔΨm), and (iv) diminished glutathione (GSH) levels. Succinobucol pretreatment (6 days) significantly prevented 3-NP-induced loss of cellular viability, generation of reactive oxygen species, and decrease of ΔΨm. However, succinobucol pretreatment did not protect against 3-NP-induced inhibition of mitochondrial complex II activity, pointing to the mitigation of secondary events resultant from mitochondrial complex II inhibition. Succinobucol pretreatment (6 days) significantly increased (50 %) the levels of GSH in SH-SY5Y cells, and this event was paralleled by significant increases in glutamate cysteine ligase messenger RNA (mRNA) expression and activity (GCL; the first enzyme in the GSH biosynthesis). The present findings are the first to show that succinobucol increases GSH levels via upregulation of GCL activity (possibly through the activation of the nuclear (erythroid-derived 2)-related factor (Nrf2)/antioxidant response element (ARE) pathway), displaying protective effects against mitochondrial dysfunction-derived oxidative stress.


Subject(s)
Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Hypolipidemic Agents/pharmacology , Mitochondria/metabolism , Oxidative Stress/drug effects , Probucol/analogs & derivatives , Up-Regulation/drug effects , Buthionine Sulfoximine/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione Peroxidase/metabolism , Humans , Hydroquinones/pharmacology , Mitochondria/drug effects , Nitro Compounds , Probucol/pharmacology , Propionates , Protective Agents/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
11.
Arch Toxicol ; 90(3): 647-60, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25618550

ABSTRACT

The organophosphorus (OP) pesticide malathion is a neurotoxic compound whose acute toxicity is primarily caused by the inhibition of acetylcholinesterase (AChE), leading to cholinergic syndrome-related symptoms. Some lines of evidence indicate that long-term exposure to low levels of OP may produce neuropsychiatric and/or neurobehavioral signs that do not necessarily involve the AChE inhibition. This study evaluated the effects of a repeated (15-day period) and low-dose malathion exposure on spatial memory and discrimination (object location task), as well as on biochemical parameters in the hippocampus of mice [AChE and mitochondrial chain complexes activities; levels of proapoptotic proteins (Bax and Bak) and cholinergic neuronal and astroglial markers (ChAT and GFAP, respectively)]. Malathion treatments (30 and 100 mg/kg, s.c.) did not affect the body weight of animals and caused no evident signs of cholinergic toxicity throughout the treatment, although the highest dose (100 mg/kg) was associated with inhibition of AChE activity. Malathion-exposed animals showed a significant impairment on spatial memory and discrimination, which was correlated with a decrease in the mitochondrial complex I activity in the hippocampus. Moreover, malathion increased the levels of proapoptotic proteins and induced astroglial activation. The results show that long-term malathion exposure, at a dose that does not affect hippocampal AChE activity (30 mg/kg), caused impaired spatial memory and discrimination in mice that was related to hippocampal mitochondrial dysfunctional, astrogliosis and apoptosis. When extrapolated to humans, such results shed light on noncholinergic mechanisms likely related to the neurobehavioral and cognitive deficits observed in individuals chronically exposed to this pesticide.


Subject(s)
Astrocytes/drug effects , Cognition Disorders/chemically induced , Hippocampus/drug effects , Insecticides/toxicity , Malathion/toxicity , Animals , Apoptosis/drug effects , Astrocytes/pathology , Cholinesterase Inhibitors/toxicity , Dose-Response Relationship, Drug , Hippocampus/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/pathology , Spatial Memory/drug effects , Toxicity Tests, Chronic/methods
12.
Biol Trace Elem Res ; 158(3): 399-409, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24723215

ABSTRACT

Manganese (Mn) exposure is related to industrial activities, where absorption by inhalation has high relevance. Manganism, a syndrome caused as a result of excessive accumulation of Mn in the central nervous system, has numerous symptoms similar to those seen in idiopathic Parkinson disease (IPD). Some of these symptoms, such as learning, memory, sensorial, and neurochemical changes, appear before the onset of motor deficits in both manganism and IPD. The aim of this study was to evaluate the possible neuroprotective effects of curcumin against behavioral deficits induced by Mn toxicity in young (2 months old) Swiss mice. We evaluated the effect of chronic inhalation of a Mn mixture [Mn(OAc)3 and MnCl2 (20:40 mM)], 1 h/session, three times a week, over a 14-week period on behavioral and neurochemical parameters. Curcumin was supplemented in the diet (500 or 1,500 ppm in food pellets). The Mn disrupted the motor performance evaluated in the single-pellet reach task, as well as the short- and long-term spatial memory evaluated in the step-down inhibitory avoidance task. Surprisingly, curcumin also produced similar deleterious effects in such behavioral tests. Moreover, the association of Mn plus curcumin significantly increased the levels of Mn and iron, and decreased the levels of dopamine and serotonin in the hippocampus. These alterations were not observed in the striatum. In conclusion, the current Mn treatment protocol resulted in mild deficits in motor and memory functions, resembling the early phases of IPD. Additionally, curcumin showed no beneficial effects against Mn-induced disruption of hippocampal metal and neurotransmitter homeostasis.


Subject(s)
Curcumin/pharmacology , Hippocampus/drug effects , Manganese/pharmacology , Metals/metabolism , Neurotransmitter Agents/metabolism , Acetates/administration & dosage , Acetates/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chlorides/administration & dosage , Chlorides/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Curcumin/administration & dosage , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Hippocampus/metabolism , Iron/metabolism , Male , Manganese/administration & dosage , Manganese/metabolism , Manganese Compounds/administration & dosage , Manganese Compounds/pharmacology , Memory/drug effects , Mice , Motor Activity/drug effects , Serotonin/metabolism
13.
J Alzheimers Dis ; 41(1): 43-60, 2014.
Article in English | MEDLINE | ID: mdl-24577472

ABSTRACT

Familial hypercholesterolemia is caused by inherited genetic abnormalities that directly or indirectly affect the function of the low-density lipoprotein (LDL) receptor. This condition is characterized by defective catabolism of LDL which results in increased plasma cholesterol concentrations and premature coronary artery disease. Nevertheless, there is increasing preclinical and clinical evidence indicating that familial hypercholesterolemia subjects show a particularly high incidence of mild cognitive impairment. Moreover, the LDL receptor (LDLr) has been implicated as the main central nervous system apolipoprotein E receptor that regulates amyloid deposition in distinct mouse models of ß-amyloidosis. In this regard, herein we hypothesized that the lack of LDLr would enhance the susceptibility to amyloid-ß-(Aß)-induced neurotoxicity in mice. Using the acute intracerebroventricular injection of aggregated Aß(1-40) peptide (400 pmol/mouse), a useful approach for the investigation of molecular mechanisms involved in Aß toxicity, we observed oxidative stress, neuroinflammation, and neuronal membrane damage within the hippocampus of C57BL/6 wild-type mice, which were associated with spatial reference memory and working memory impairments. In addition, our data show that LDLr knockout (LDLr(-/-)) mice, regardless of Aß treatment, displayed memory deficits and increased blood-brain barrier permeability. Nonetheless, LDLr(-/-) mice treated with Aß(1-40) peptide presented increased acetylcholinesterase activity, astrogliosis, oxidative imbalance, and cell permeability within the hippocampus in comparison with Aß(1-40)-treated C57BL/6 wild-type mice. Overall, the present study shows that the lack of LDLr increases the susceptibility to Aß-induced neurotoxicity in mice providing new evidence about the crosslink between familial hypercholesterolemia and cognitive impairment.


Subject(s)
Amyloidosis/physiopathology , Hippocampus/physiopathology , Memory Disorders/physiopathology , Prefrontal Cortex/physiopathology , Receptors, LDL/metabolism , Acetylcholinesterase/metabolism , Amyloid beta-Peptides , Amyloidosis/complications , Amyloidosis/pathology , Animals , Antioxidants/metabolism , Astrocytes/pathology , Astrocytes/physiology , Blood-Brain Barrier/physiopathology , Capillary Permeability/physiology , Cell Membrane Permeability/physiology , Disease Models, Animal , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/pathology , Memory Disorders/etiology , Memory Disorders/pathology , Memory, Short-Term/physiology , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation/physiology , Oxidative Stress/physiology , Peptide Fragments , Prefrontal Cortex/pathology , Receptors, LDL/genetics , Spatial Memory/physiology , Superoxide Dismutase/metabolism
14.
Biomed Res Int ; 2013: 537279, 2013.
Article in English | MEDLINE | ID: mdl-24350274

ABSTRACT

Organochalcogens, particularly ebselen, have been used in experimental and clinical trials with borderline efficacy. (PhSe)2 and (PhTe)2 are the simplest of the diaryl dichalcogenides and share with ebselen pharmacological properties. In view of the concerns with the use of mammals in studies and the great number of new organochalcogens with potential pharmacological properties that have been synthesized, it becomes important to develop screening protocols to select compounds that are worth to be tested in vivo. This study investigated the possible use of isolated human white cells as a preliminary model to test organochalcogen toxicity. Human leucocytes were exposed to 5-50 µM of ebselen, (PhSe)2, or (PhTe)2. All compounds were cytotoxic (Trypan's Blue exclusion) at the highest concentration tested, and Ebselen was the most toxic. Ebselen and (PhSe)2 were genotoxic (Comet Assay) only at 50 µM, and (PhTe)2 at 5-50 µM. Here, the acute cytotoxicity did not correspond with in vivo toxicity of the compounds. But the genotoxicity was in the same order of the in vivo toxicity to mice. These results indicate that in vitro genotoxicity in white blood cells should be considered as an early step in the investigation of potential toxicity of organochalcogens.


Subject(s)
Azoles/pharmacology , Benzene Derivatives/pharmacology , Leukocytes/drug effects , Mutagens/pharmacology , Organometallic Compounds/pharmacology , Organoselenium Compounds/pharmacology , Humans , Isoindoles , Mutagenicity Tests/methods
15.
PLoS One ; 8(6): e67658, 2013.
Article in English | MEDLINE | ID: mdl-23799154

ABSTRACT

Huntington's disease (HD) is an autosomal dominantly inherited neurodegenerative disease characterized by symptoms attributable to the death of striatal and cortical neurons. The molecular mechanisms mediating neuronal death in HD involve oxidative stress and mitochondrial dysfunction. Administration of 3-nitropropionic acid (3-NP), an irreversible inhibitor of the mitochondrial enzyme succinate dehydrogenase, in rodents has been proposed as a useful experimental model of HD. This study evaluated the effects of probucol, a lipid-lowering agent with anti-inflammatory and antioxidant properties, on the biochemical parameters related to oxidative stress, as well as on the behavioral parameters related to motor function in an in vivo HD model based on 3-NP intoxication in rats. Animals were treated with 3.5 mg/kg of probucol in drinking water daily for 2 months and, subsequently, received 3-NP (25 mg/kg i.p.) once a day for 6 days. At the end of the treatments, 3-NP-treated animals showed a significant decrease in body weight, which corresponded with impairment on motor ability, inhibition of mitochondrial complex II activity and oxidative stress in the striatum. Probucol, which did not rescue complex II inhibition, protected against behavioral and striatal biochemical changes induced by 3-NP, attenuating 3-NP-induced motor impairments and striatal oxidative stress. Importantly, probucol was able to increase activity of glutathione peroxidase (GPx), an enzyme important in mediating the detoxification of peroxides in the central nervous system. The major finding of this study was that probucol protected against 3-NP-induced behavioral and striatal biochemical changes without affecting 3-NP-induced mitochondrial complex II inhibition, indicating that long-term probucol treatment resulted in an increased resistance against neurotoxic events (i.e., increased oxidative damage) secondary to mitochondrial dysfunction. These data appeared to be of great relevance when extrapolated to human neurodegenerative processes involving mitochondrial dysfunction and indicates that GPx is an important molecular target involved in the beneficial effects of probucol.


Subject(s)
Antioxidants/pharmacology , Corpus Striatum/enzymology , Glutathione Peroxidase/metabolism , Huntington Disease/drug therapy , Oxidative Stress , Probucol/pharmacology , Animals , Antioxidants/therapeutic use , Catalase/metabolism , Corpus Striatum/drug effects , Drug Evaluation, Preclinical , Electron Transport Complex II/metabolism , Glutathione Reductase/metabolism , Humans , Huntington Disease/chemically induced , Huntington Disease/enzymology , Lipid Peroxidation , Male , Motor Activity/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitro Compounds , Probucol/therapeutic use , Propionates , Rats , Rats, Wistar , Rotarod Performance Test , Superoxide Dismutase/metabolism , Weight Loss/drug effects
16.
Chem Biol Interact ; 204(3): 191-9, 2013 Aug 25.
Article in English | MEDLINE | ID: mdl-23707192

ABSTRACT

The study evaluated whether a diet containing diphenyl diselenide (PhSe)2, a synthetic antioxidant, could reduce the biochemical alterations induced by chronic consumption of highly enriched fructose diet and/or hydrochlorothiazide (HCTZ). Rats were fed a control diet (CT) or a high fructose diet (HFD), supplemented with or not HCTZ (4.0g/kg) and/or (PhSe)2 (3ppm) for 18weeks. HFD intake increased significantly plasma glucose, fructosamine, triglycerides and cholesterol levels. (PhSe)2 supplementation significantly reduced triglycerides and cholesterol but could not restore them to control levels. The combination of HFD and HCTZ significantly altered plasma glucose, fructosamine, triglycerides and cholesterol levels which were not restore by (PhSe)2 supplementation. Lipid peroxidation, protein carbonyl formation, vitamin C level and catalase activity decreased after HFD, HCTZ or HFD plus HCTZ ingestion. Remarkably (PhSe)2 supplementation restored the oxidative stress parameters. HCTZ decreased renal superoxide dismutase (SOD) activity, which was restored to control levels by (PhSe)2. Furthermore, the association of HFD and HCTZ decreased plasma potassium levels and aggravated HCTZ-induced hypomagnesemia and hypertriglyceridemia. Here we provided evidence of the involvement of oxidative stress and metabolic disorders in a rat model of HFD associated or not with HTCZ. (PhSe)2 supplementation reduced the oxidative stress and this compound should be considered for the treatment of biochemical disturbances and oxidative stress in other animal models of metabolic disorders.


Subject(s)
Antioxidants/pharmacology , Benzene Derivatives/pharmacology , Diet , Dietary Supplements , Fructose/metabolism , Hydrochlorothiazide/metabolism , Organoselenium Compounds/pharmacology , Oxidative Stress/drug effects , Animals , Catalase/metabolism , Down-Regulation/drug effects , Male , Rats , Rats, Wistar
17.
Neurosci Lett ; 541: 193-8, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23470631

ABSTRACT

Epidemiological studies indicate that high midlife plasma cholesterol levels increases the risk of Alzheimer's disease. Moreover, middle-aged familial hypercholesterolemia (FH) subjects show a particularly high incidence of mild cognitive impairments (MCI). These evidence points to hypercholesterolemia as one of the modifiable risk factors focused on prevention/treatment of cognitive deterioration. The present study draws a comparison between pharmacological (lipid-lowering drug probucol) and non-pharmacological (voluntary running wheel, RW) approaches for the management of hypercholesterolemia and cognitive impairments associated with the low-density lipoprotein receptor-deficient (LDLr(-/-)) mice, a well-established rodent model of FH. We also investigated whether exposure to environmental enrichment (EE), a feasible option to increase physical activity in young mice cohort, from birth to adolescence (PN45) yields long-term behavioral changes in adult LDLr(-/-) mice (PN90). We observed that both probucol and RW significantly decreased total and non-HDL plasma cholesterol levels in LDLr(-/-) mice. Notably, only physical exercise mitigated the spatial memory deficits of LDLr(-/-) mice. In addition, we showed that exposure to EE from birth until the adolescence did not mitigate the spatial memory deficits of adult LDLr(-/-) mice in the object location task, although it induced persistent anxyolitic-like effects in the open field arena. Collectively, our results emphasize the advantages physical exercise, in comparison to lipid-lowering drugs, for the management of cognitive deficits associated with FH.


Subject(s)
Cognition Disorders/psychology , Hyperlipoproteinemia Type II/psychology , Physical Conditioning, Animal , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Cognition Disorders/etiology , Cognition Disorders/therapy , Founder Effect , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/drug therapy , Mice , Mice, Knockout , Motor Activity , Probucol/pharmacology , Probucol/therapeutic use , Receptors, LDL/genetics , Social Environment
18.
Neurochem Res ; 38(3): 660-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334712

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic nigrostriatal neurons. Although the etiology of the majority of human PD cases is unknown, experimental evidence points to oxidative stress as an early and causal event. Probucol is a lipid-lowering phenolic compound with anti-inflammatory and antioxidant properties that has been recently reported as protective in neurotoxicity and neurodegeneration models. This study was designed to investigate the effects of probucol on the vulnerability of striatal dopaminergic neurons to oxidative stress in a PD in vivo model. Swiss mice were treated with probucol during 21 days (11.8 mg/kg; oral route). Two weeks after the beginning of treatment, mice received a single intracerebroventricular (i.c.v.) infusion of 6-hydroxydopamine (6-OHDA). On the 21st day, locomotor performance, striatal oxidative stress-related parameters, and striatal tyrosine hydroxylase and synaptophysin levels, were measured as outcomes of toxicity. 6-OHDA-infused mice showed hyperlocomotion and a significant decrease in striatal tyrosine hydroxylase (TH) and synaptophysin levels. In addition, 6-OHDA-infused mice showed reduced superoxide dismutase activity and increased lipid peroxidation and catalase activity in the striatum. Notably, probucol protected against 6-OHDA-induced hyperlocomotion and striatal lipid peroxidation, catalase upregulation and decrease of TH levels. Overall, the present results show that probucol protects against 6-OHDA-induced toxicity in mice. These findings may render probucol as a promising molecule for further pharmacological studies on the search for disease-modifying treatment in PD.


Subject(s)
Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Probucol/therapeutic use , Animals , Catalase/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopaminergic Neurons/drug effects , Lipid Peroxidation/drug effects , Male , Mice , Motor Activity/drug effects , Oxidative Stress/drug effects , Oxidopamine , Tyrosine 3-Monooxygenase/metabolism
19.
J Mol Neurosci ; 49(1): 68-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23054587

ABSTRACT

Studies have demonstrated an association between stressful conditions and the onset of clinical depression. Considering the antidepressant-like properties of ascorbic acid in both experimental and clinical approaches, we evaluated the beneficial effect of this vitamin on restraint stress-induced behavioral and neurochemical alterations. Acute restraint stress caused a depressive-like behavior in the forced swimming test, accompanied by increased lipid peroxidation (cerebral cortex and hippocampus); increased superoxide dismutase (cerebral cortex and hippocampus), glutathione reductase (cerebral cortex), and glutathione peroxidase (cerebral cortex and hippocampus) activities; and elevated expression of Bcl-2 (hippocampus). Oral administration of ascorbic acid (1 mg/kg) or fluoxetine (10 mg/kg) 1 h before restraint stress prevented the stress-induced increase on immobility time in the forced swimming test. Moreover, this vitamin reduced lipid peroxidation to control levels and restored the activity of superoxide dismutase, glutathione reductase, and glutathione peroxidase. Ascorbic acid had no effect on the increased level of Bcl-2 induced by stress. Glutathione levels, glycogen synthase kinase-3ß phosphorylation, and Bax expression were not altered by stress or ascorbic acid administration. Besides reinforcing the antioxidant potential of ascorbic acid, our results support the notion that oxidative stress plays a role in the pathogenesis and treatment of stress-induced depression.


Subject(s)
Antioxidants/therapeutic use , Ascorbic Acid/therapeutic use , Depressive Disorder/drug therapy , Stress, Psychological/complications , Vitamins/therapeutic use , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Depressive Disorder/etiology , Female , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Reductase/genetics , Glutathione Reductase/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/enzymology , Hippocampus/metabolism , Lipid Metabolism/drug effects , Mice , Motor Activity/drug effects , Oxidation-Reduction/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Swimming , Transcription, Genetic/drug effects , Vitamins/pharmacology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
Exp Neurol ; 240: 112-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23142187

ABSTRACT

Experimental and epidemiological studies have shown the close relationship between stressful events, depression, and cognitive impairment. Folic acid has been reported to present antidepressant-like effects in both experimental and clinical approaches. However, the mechanisms mediating such effects are not understood. In the present study, we evaluated if folic acid administration to mice could protect against restraint stress-induced depressive-like behavior and cognitive deficit. Considering that oxidative stress has been pointed as a key event involved with depressive disorders, cerebrocortical and hippocampal oxidative stress-related parameters, such as the activities of antioxidant enzymes (mainly those related to the hydroperoxide-detoxifying system) and markers of lipid peroxidation, were also investigated. Restraint stress induced depressive-like behavior in the forced swimming test and memory impairment in the object recognition test, without altering locomotor activity of mice. Folic acid (50 mg/kg, p.o.) was able to prevent the stress-induced increase on immobility time in the forced swimming test, but did not prevent memory impairment. Moreover, restraint stress increased thiobarbituric acid reactive substance levels, and catalase, glutathione peroxidase and glutathione reductase activities in the cerebral cortex and hippocampus, and superoxide dismutase activity in the hippocampus. Folic acid treatment restored the activity of the antioxidant enzymes and reduced lipid peroxidation in the hippocampus. Glutathione, a non-enzymatic antioxidant, was not altered by stress and/or folic acid administration. Together, the results of the present work reinforce the notion that folic acid displays a specific antidepressant profile in the restraint stress paradigm that may be at least partly due to its antioxidant role.


Subject(s)
Antioxidants/adverse effects , Depressive Disorder, Major/prevention & control , Folic Acid/therapeutic use , Hippocampus/metabolism , Restraint, Physical/adverse effects , Stress, Psychological/etiology , Animals , Antidepressive Agents/therapeutic use , Antioxidants/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Mice , Oxidative Stress/drug effects , Oxidative Stress/physiology , Stress, Psychological/metabolism , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...