Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
2.
Mol Neurobiol ; 55(11): 8538-8549, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29564809

ABSTRACT

Kynurenic acid (KYNA) and quinolinic acid (QUIN) are metabolites produced in the degradation of tryptophan and have important neurological activities. KYNA/QUIN ratio changes are known to be associated with central nervous system disorders, such Alzheimer, Parkinson, and Huntington diseases. In the present study, we investigate the ability of KYNA in prevent the first events preceding QUIN-induced neurodegeneration in striatal slices of rat. We evaluated the protective effect of KYNA on oxidative status (reactive oxygen species production, antioxidant enzymes activities, lipid peroxidation, nitrite levels, protein and DNA damage, and iNOS immunocontent), mitochondrial function (mitochondrial mass, membrane potential, and respiratory chain enzymes), and Na+,K+-ATPase in striatal slices of rats treated with QUIN. Since QUIN alters the levels of Nrf2, we evaluated the influence of KYNA protection on this parameter. Striatal slices from 30-day-old Wistar rats were preincubated with KYNA (100 µM) for 15 min, followed by incubation with 100-µM QUIN for 30 min. Results showed that KYNA prevented the increase of ROS production caused by QUIN and restored antioxidant enzyme activities and the protein and lipid damage, as well as the Nrf2 levels. KYNA also prevented the effects of QUIN on mitochondrial mass and mitochondrial membrane potential, as well as the decrease in the activities of complex II, SDH, and Na+,K+-ATPase. We suggest that KYNA prevents changes in Nrf2 levels, oxidative imbalance, and mitochondrial dysfunction caused by QUIN in striatal slices. This study elucidates some of the protective effects of KYNA against the damage caused by QUIN toxicity.


Subject(s)
Corpus Striatum/pathology , Kynurenic Acid/pharmacology , NF-E2-Related Factor 2/metabolism , Quinolinic Acid/toxicity , Animals , Antioxidants/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Fluoresceins/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Oxidation-Reduction , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Sulfhydryl Compounds/metabolism
3.
Mol Cell Endocrinol ; 470: 281-294, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29155306

ABSTRACT

In the present study we provide evidence that 3,3',5'-triiodothyronine (reverse T3, rT3) restores neurochemical parameters induced by congenital hypothyroidism in rat hippocampus. Congenital hypothyroidism was induced by adding 0.05% propylthiouracil in the drinking water from gestation day 8 and continually up to lactation day 15. In the in vivo rT3 exposure, hypothyroid 12-day old pups were daily injected with rT3 (50 ng/kg body weight) or saline until day 14. In the ex vivo rT3 treatment, hippocampal slices from 15-day-old hypothyroid pups were incubated for 30 min with or without rT3 (1 nM). We found that ex vivo and/or in vivo exposure to rT3 failed in restoring the decreased 14C-glutamate uptake; however, restored the phosphorylation of glial fibrillary acidic protein (GFAP), 45Ca2+ influx, aspartate transaminase (AST), glutamine synthetase (GS) and gamma-glutamate transferase (GGT) activities, as well as glutathione (GSH) levels in hypothyroid hippocampus. In addition, rT3 improved 14C-2-deoxy-D-glucose uptake and lactate dehydrogenase (LDH) activity. Receptor agonists/antagonists (RGD peptide and AP-5), kinase inhibitors of p38MAPK, ERK1/2, CaMKII, PKA (SB239063, PD98059, KN93 and H89, respectively), L-type voltage-dependent calcium channel blocker (nifedipine) and intracellular calcium chelator (BAPTA-AM) were used to determine the mechanisms of the nongenomic rT3 action on GGT activity. Using molecular docking analysis, we found rT3 interaction with αvß3 integrin receptors, nongenomically activating signaling pathways (PKA, CaMKII, p38MAPK) that restored GGT activity. We provide evidence that rT3 is an active TH metabolite and our results represent an important contribution to elucidate the nonclassical mechanism of action of this metabolite in hypothyroidism.


Subject(s)
Hippocampus/enzymology , Hypothyroidism/enzymology , Integrin alphaVbeta3/metabolism , Signal Transduction , Triiodothyronine, Reverse/pharmacology , Animals , Biological Transport/drug effects , Calcium/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Glutathione/metabolism , Homeostasis/drug effects , Hypothyroidism/pathology , L-Lactate Dehydrogenase/metabolism , Models, Biological , Molecular Docking Simulation , Phosphorylation/drug effects , Rats, Wistar , Receptors, Glutamate/metabolism , Signal Transduction/drug effects , Transaminases/metabolism
4.
Mol Neurobiol ; 55(6): 5362-5376, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28936789

ABSTRACT

It has been shown that synergistic toxic effects of quinolinic acid (QUIN) and glutaric acid (GA), both in isolated nerve endings and in vivo conditions, suggest the contribution of these metabolites to neurodegeneration. However, this synergism still requires a detailed characterization of the mechanisms involved in cell damage during its occurrence. In this study, the effects of subtoxic concentrations of QUIN and/or GA were tested in neuronal cultures, co-cultures (neuronal cells + astrocytes), and mixed cultures (neuronal cells + astrocytes + microglia) from rat cortex and striatum. The exposure of different cortical and striatal cell cultures to QUIN + GA resulted in cell death and stimulated different markers of oxidative stress, including reactive oxygen species (ROS) formation; changes in the activity of antioxidant enzymes such as superoxide dismutase, catalase, and glutathione peroxidase; and depletion of endogenous antioxidants such as -SH groups and glutathione. The co-incubation of neuronal cultures with QUIN + GA plus the N-methyl-D-aspartate antagonist MK-801 prevented cell death but not ROS formation, whereas the antioxidant melatonin reduced both parameters. Our results demonstrated that QUIN and GA can create synergistic scenarios, inducing toxic effects on some parameters of cell viability via the stimulation of oxidative damage. Therefore, it is likely that oxidative stress may play a major causative role in the synergistic actions exerted by QUIN + GA in a variety of cell culture conditions involving the interaction of different neural types.


Subject(s)
Glutarates/toxicity , Models, Biological , Neurons/metabolism , Oxidative Stress , Quinolinic Acid/toxicity , Animals , Antioxidants/metabolism , Catalase/metabolism , Cell Survival/drug effects , Cerebral Cortex/pathology , Coculture Techniques , Dizocilpine Maleate/pharmacology , Female , Gliosis/metabolism , Gliosis/pathology , Glutarates/administration & dosage , Glutathione/metabolism , Melatonin/pharmacology , Neostriatum/pathology , Neurites/drug effects , Neurites/metabolism , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Quinolinic Acid/administration & dosage , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
5.
Neurotox Res ; 34(1): 147-163, 2018 07.
Article in English | MEDLINE | ID: mdl-29124681

ABSTRACT

The brain of patients affected by Alzheimer's disease (AD) develops progressive neurodegeneration linked to the formation of proteins aggregates. However, their single actions cannot explain the extent of brain damage observed in this disorder, and the characterization of co-adjuvant involved in the early toxic processes evoked in AD is essential. In this line, quinolinic acid (QUIN) and homocysteine (Hcy) appear to be involved in the AD neuropathogenesis. Herein, we investigate the effects of QUIN and Hcy on early toxic events in cortical neurons and astrocytes. Exposure of primary cortical cultures to these neurometabolites for 24 h induced concentration-dependent neurotoxicity. In addition, QUIN (25 µM) and Hcy (30 µM) triggered ROS production, lipid peroxidation, diminished of Na+,K+-ATPase activity, and morphologic alterations, culminating in reduced neuronal viability by necrotic cell death. In astrocytes, QUIN (100 µM) and Hcy (30 µM) induced caspase-3-dependent apoptosis and morphologic alterations through oxidative status imbalance. To establish specific mechanisms, we preincubated cell cultures with different protective agents. The combined toxicity of QUIN and Hcy was attenuated by melatonin and Trolox in neurons and by NMDA antagonists and glutathione in astrocytes. Cellular death and morphologic alterations were prevented when co-culture was treated with metabolites, suggesting the activation of protector mechanisms dependent on soluble factors and astrocyte and neuron communication through gap junctions. These findings suggest that early damaging events involved in AD can be magnified by synergistic toxicity of the QUIN and Hcy. Therefore, this study opens new possibilities to elucidate the molecular mechanisms of neuron-astrocyte interactions and their role in neuroprotection against QUIN and Hcy.


Subject(s)
Astrocytes/drug effects , Cerebral Cortex/cytology , Homocysteine/pharmacology , Neurons/drug effects , Neurotoxins/pharmacology , Quinolinic Acid/pharmacology , Analysis of Variance , Animals , Annexin A5/metabolism , Astrocytes/ultrastructure , Cells, Cultured , Coculture Techniques , Drug Synergism , Embryo, Mammalian , Female , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/ultrastructure , Nerve Tissue Proteins/metabolism , Neurons/ultrastructure , Pregnancy , Rats , Sodium-Potassium-Exchanging ATPase/metabolism , Thiobarbituric Acid Reactive Substances
6.
Mol Neurobiol ; 55(5): 4362-4372, 2018 May.
Article in English | MEDLINE | ID: mdl-28647871

ABSTRACT

Cytoskeletal proteins are increasingly recognized as having important roles as a target of the action of different neurotoxins. In the last years, several works of our group have shown that quinolinic acid (QUIN) was able to disrupt the homeostasis of the cytoskeleton of neural cells and this was associated with cell dysfunction and neurodegeneration. QUIN is an excitotoxic metabolite of tryptophan metabolism and its accumulation is associated with several neurodegenerative diseases. In the present review, we provide a comprehensive view of the actions of QUIN upstream of glutamate receptors, eliciting kinase/phosphatase signaling cascades that disrupt the homeostasis of the phosphorylation system associated with intermediate filament proteins of astrocytes and neurons. We emphasize the critical role of calcium in these actions and the evidence that misregulated cytoskeleton takes part of the cell response to the injury resulting in neurodegeneration in different brain regions, disrupted cell signaling in acute tissue slices, and disorganized cytoskeleton with altered cell morphology in primary cultures. We also discuss the interplay among misregulated cytoskeleton, oxidative stress, and cell-cell contact through gap junctions mediating the quinolinic acid injury in rat brain. The increasing amount of cross talks identified between cytoskeletal proteins and cellular signaling cascades reinforces the exciting possibility that cytoskeleton could be a new target in the neurotoxicity of QUIN and further studies will be necessary to develop strategies to protect the cytoskeleton and counteracts the cytotoxicity of this metabolite.


Subject(s)
Cytoskeleton/metabolism , Neurotoxins/toxicity , Quinolinic Acid/toxicity , Animals , Cytoskeleton/drug effects , Disease Models, Animal , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology
7.
Mol Neurobiol ; 55(6): 5111-5124, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28840509

ABSTRACT

Kynurenic acid (KYNA) is a neuroactive metabolite of tryptophan known to modulate a number of mechanisms involved in neural dysfunction. Although its activity in the brain has been widely studied, the effect of KYNA counteracting the actions of quinolinic acid (QUIN) remains unknown. The present study aims at describing the ability of 100 µM KYNA preventing cytoskeletal disruption provoked by QUIN in astrocyte/neuron/microglia mixed culture. KYNA totally preserved cytoskeletal organization, cell morphology, and redox imbalance in mixed cultures exposed to QUIN. However, KYNA partially prevented morphological alteration in isolated primary astrocytes and failed to protect the morphological alterations of neurons caused by QUIN exposure. Moreover, KYNA prevented QUIN-induced microglial activation and upregulation of ionized calcium-binding adapter molecule 1 (Iba-1) and partially preserved tumor necrosis factor-α (TNF-α) level in mixed cultures. TNF-α level was also partially preserved in astrocytes. In addition to the mechanisms dependent on redox imbalance and microglial activation, KYNA prevented downregulation of connexin-43 and the loss of functionality of gap junctions (GJs), preserving cell-cell contact, cytoskeletal organization, and cell morphology in QUIN-treated cells. Furthermore, the toxicity of QUIN targeting the cytoskeleton of mixed cultures was not prevented by the N-methyl-D-aspartate (NMDA) antagonist MK-801. We suggest that KYNA protects the integrity of the cytoskeleton of mixed cultures by complex mechanisms including modulating microglial activation preventing oxidative imbalance and misregulated GJs leading to disrupted cytoskeleton in QUIN-treated cells. This study contributed to elucidate the molecular basis of KYNA protection against QUIN toxicity.


Subject(s)
Corpus Striatum/pathology , Cytoskeleton/metabolism , Kynurenic Acid/pharmacology , Quinolinic Acid/toxicity , Animals , Antioxidants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Communication/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Dizocilpine Maleate/pharmacology , Gap Junctions/drug effects , Gap Junctions/metabolism , Microglia/drug effects , Microglia/metabolism , Models, Biological , N-Methylaspartate/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
Mol Neurobiol ; 55(2): 980-988, 2018 02.
Article in English | MEDLINE | ID: mdl-28084592

ABSTRACT

In the current study, we verified the effects of maternal hypermethioninemia on the number of neurons, apoptosis, nerve growth factor, and brain-derived neurotrophic factor levels, energy metabolism parameters (succinate dehydrogenase, complex II, and cytochrome c oxidase), expression and immunocontent of Na+,K+-ATPase, edema formation, inflammatory markers (tumor necrosis factor-alpha and interleukin-6), and mitochondrial hydrogen peroxide levels in the encephalon from the offspring. Pregnant Wistar rats were divided into two groups: the first one received saline (control) and the second group received 2.68 µmol methionine/g body weight by subcutaneous injections twice a day during gestation (approximately 21 days). After parturition, pups were killed at the 21st day of life for removal of encephalon. Neuronal staining (anti-NeuN) revealed a reduction in number of neurons, which was associated to decreased nerve growth factor and brain-derived neurotrophic factor levels. Maternal hypermethioninemia also reduced succinate dehydrogenase and complex II activities and increased expression and immunocontent of Na+,K+-ATPase alpha subunits. These results indicate that maternal hypermethioninemia may be a predisposing factor for damage to the brain during the intrauterine life.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain/metabolism , Energy Metabolism/physiology , Glycine N-Methyltransferase/deficiency , Nerve Growth Factors/metabolism , Neurons/metabolism , Prenatal Exposure Delayed Effects/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Metabolism, Inborn Errors/chemically induced , Animals , Cell Count , Female , Glycine N-Methyltransferase/metabolism , Methionine , Oxidation-Reduction , Pregnancy , Rats , Rats, Wistar
9.
Mol Neurobiol ; 55(5): 4098-4106, 2018 May.
Article in English | MEDLINE | ID: mdl-28593435

ABSTRACT

Hypoxanthine is the major purine involved in the salvage pathway of purines in the brain. High levels of hypoxanthine are characteristic of Lesch-Nyhan Disease. Since hypoxanthine is a purine closely related to ATP formation, the aim of this study was to investigate the effect of intrastriatal hypoxanthine administration on neuroenergetic parameters (pyruvate kinase, succinate dehydrogenase, complex II, cytochrome c oxidase, and ATP levels) and mitochondrial function (mitochondrial mass and membrane potential) in striatum of rats. We also evaluated the effect of cell death parameters (necrosis and apoptosis). Wistar rats of 60 days of life underwent stereotactic surgery and were divided into two groups: control (infusion of saline 0.9%) and hypoxanthine (10 µM). Intrastriatal hypoxanthine administration did not alter pyruvate kinase activity, but increased succinate dehydrogenase and complex II activities and diminished cytochrome c oxidase activity and immunocontent. Hypoxanthine injection decreased the percentage of cells with mitochondrial membrane label and increased mitochondrial membrane potential labeling. There was a decrease in the number of live cells and an increase in the number of apoptotic cells by caused hypoxanthine. Our findings show that intrastriatal hypoxanthine administration altered neuroenergetic parameters, and caused mitochondrial dysfunction and cell death by apoptosis, suggesting that these processes may be associated, at least in part, with neurological symptoms found in patients with Lesch-Nyhan Disease.


Subject(s)
Aging/pathology , Corpus Striatum/pathology , Energy Metabolism , Hypoxanthine/pharmacology , Animals , Cell Death/drug effects , Creatine Kinase/metabolism , Electron Transport Complex IV/metabolism , Hypoxanthine/administration & dosage , Male , Mitochondria/drug effects , Mitochondria/metabolism , Pyruvate Kinase/metabolism , Rats, Wistar , Succinate Dehydrogenase/metabolism
10.
Metab Brain Dis ; 33(3): 693-704, 2018 06.
Article in English | MEDLINE | ID: mdl-29288365

ABSTRACT

Although methylphenidate (MPH) is ubiquitously prescribed to children and adolescents, the consequences of chronic utilization of this psychostimulant are poorly understood. In this study, we investigated the effects of MPH on cytoskeletal homeostasis and lipid content in rat hippocampus. Wistar rats received intraperitoneal injections of MPH (2.0 mg/kg) or saline solution (controls), once a day, from the 15th to the 44th day of age. Results showed that MPH provoked hypophosphorylation of glial fibrillary acidic protein (GFAP) and reduced its immunocontent. Middle and high molecular weight neurofilament subunits (NF-M, NF-H) were hypophosphorylated by MPH on KSP repeat tail domains, while NFL, NFM and NFH immunocontents were not altered. MPH increased protein phosphatase 1 (PP1) and 2A (PP2A) immunocontents. MPH also decreased the total content of ganglioside and phospholipid, as well as the main brain gangliosides (GM1, GD1a, and GD1b) and the major brain phospholipids (sphingomyelin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, and phosphatidylserine). Total cholesterol content was also reduced in the hippocampi of juvenile rats treated with MPH. These results provide evidence that disruptions of cytoskeletal and lipid homeostasis in hippocampus of juvenile rats are triggers by chronic MPH treatment and present a new basis for understanding the effects and consequences associated with chronic use of this psychostimulant during the development of the central nervous system.


Subject(s)
Cytoskeleton/drug effects , Hippocampus/drug effects , Homeostasis/drug effects , Lipid Metabolism/drug effects , Methylphenidate/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Lipids , Male , Rats, Wistar
11.
Neuromolecular Med ; 19(2-3): 345-356, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689355

ABSTRACT

The objective of study was to investigate changes caused by ovariectomy (OVX) on aversive and non-aversive memories, as well as on cytoskeleton phosphorylating system and on vitamin D receptor (VDR) immunocontent in hippocampus. The neuroprotective role of vitamin D was also investigated. Ninety-day-old female Wistar rats were divided into four groups: SHAM, OVX, VITAMIN D and OVX + VITAMIN D; 30 days after the OVX, vitamin D supplementation (500 IU/kg), by gavage, for 30 days was started. Results showed that OVX impaired short-term and long-term recognition, and long-term aversive memories. OVX altered hippocampal cytoskeleton phosphorylating system, evidenced by the hyperphosphorylation of glial fibrillary acidic protein (GFAP), low molecular weight neurofilament subunit (NFL), medium molecular weight neurofilament subunit (NFM) and high molecular weight neurofilament subunit (NFH), and increased the immunocontent of c-Jun N-terminal protein kinases (JNK), Ca2+/calmodulin-dependent protein kinase II (PKCaMII) and of the sites phosphorylated lysine-serine-proline (KSP) repeats, Ser55 and Ser57. Vitamin D reversed the effects caused by OVX on cytoskeleton in hippocampus, but it was not able to reverse the effects on memory.


Subject(s)
Cholecalciferol/therapeutic use , Cytoskeleton/drug effects , Hippocampus/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Ovariectomy/adverse effects , Animals , Avoidance Learning/drug effects , Cholecalciferol/pharmacology , Cytoskeletal Proteins/metabolism , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Female , Hippocampus/metabolism , Hippocampus/pathology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Phosphorylation , Protein Processing, Post-Translational/drug effects , Random Allocation , Rats , Rats, Wistar
12.
Toxicology ; 379: 1-11, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28137618

ABSTRACT

Diphenylditelluride (PhTe)2 is a neurotoxin that disrupts cytoskeletal homeostasis. We are showing that different concentrations of (PhTe)2 caused hypophosphorylation of glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits (NFL, NFM and NFH) and altered actin organization in co-cultured astrocytes and neurons from cerebral cortex of rats. These mechanisms were mediated by N-methyl-d-aspartate (NMDA) receptors without participation of either L-type voltage-dependent calcium channels (L-VDCC) or metabotropic glutamate receptors. Upregulated Ca2+ influx downstream of NMDA receptors activated Ca2+-dependent protein phosphatase 2B (PP2B) causing hypophosphorylation of astrocyte and neuron IFs. Immunocytochemistry showed that hypophosphorylated intermediate filaments (IF) failed to disrupt their organization into the cytoskeleton. However, phalloidin-actin-FITC stained cytoskeleton evidenced misregulation of actin distribution, cell spreading and increased stress fibers in astrocytes. ßIII tubulin staining showed that neurite meshworks are not altered by (PhTe)2, suggesting greater susceptibility of astrocytes than neurons to (PheTe)2 toxicity. These findings indicate that signals leading to IF hypophosphorylation fail to disrupt the cytoskeletal IF meshwork of interacting astrocytes and neurons in vitro however astrocyte actin network seems more susceptible. Our findings support that intracellular Ca2+ is one of the crucial signals that modulate the action of (PhTe)2 in co-cultured astrocytes and neurons and highlights the cytoskeleton as an end-point of the neurotoxicity of this compound. Cytoskeletal misregulation is associated with cell dysfunction, therefore, the understanding of the molecular mechanisms mediating the neurotoxicity of this compound is a matter of increasing interest since tellurium compounds are increasingly released in the environment.


Subject(s)
Astrocytes/drug effects , Benzene Derivatives/toxicity , Cell Communication , Cytoskeleton/drug effects , Neurons/drug effects , Organometallic Compounds/toxicity , Animals , Astrocytes/physiology , Calcium/metabolism , Coculture Techniques , Neurons/physiology , Phosphorylation , Rats
13.
Mol Neurobiol ; 54(10): 7796-7807, 2017 12.
Article in English | MEDLINE | ID: mdl-27844288

ABSTRACT

The study of the long-term neurological consequences of early exposure with methylphenidate (MPH) is very important since this psychostimulant has been widely misused by children and adolescents who do not meet full diagnostic criteria for ADHD. The aim of this study was to examine the effect of early chronic exposure with MPH on amino acids profile, glutamatergic and Na+,K+-ATPase homeostasis, as well as redox and energy status in the hippocampus of juvenile rats. Wistar male rats received intraperitoneal injections of MPH (2.0 mg/kg) or saline solution (controls), once a day, from the 15th to the 45th day of age. Results showed that MPH altered amino acid profile in the hippocampus, decreasing glutamine levels. Glutamate uptake and Na+,K+-ATPase activity were decreased after chronic MPH exposure in the hippocampus of rats. No changes were observed in the immunocontents of glutamate transporters (GLAST and GLT-1), and catalytic subunits of Na+,K+-ATPase (α1, α2, and α3), as well as redox status. Moreover, MPH provoked a decrease in ATP levels in the hippocampus of chronically exposed rats, while citrate synthase, succinate dehydrogenase, respiratory chain complexes activities (II, II-III, and IV), as well as mitochondrial mass and mitochondrial membrane potential were not altered. Taken together, our results suggest that chronic MPH exposure at early age impairs glutamate uptake and Na+,K+-ATPase activity probably by decreasing in ATP levels observed in rat hippocampus.


Subject(s)
Adenosine Triphosphate/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Methylphenidate/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Age Factors , Animals , Central Nervous System Stimulants/pharmacology , Male , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors
14.
Mol Neurobiol ; 54(4): 2790-2800, 2017 05.
Article in English | MEDLINE | ID: mdl-27013467

ABSTRACT

Hypoxanthine, the major oxypurine metabolite involved in purine's salvage pathway in the brain, is accumulated in Lesch-Nyhan disease, an inborn error of metabolism of purine. The purpose of this study was to investigate the effects of hypoxanthine intrastriatal administration on infant and young adult rats submitted to stereotactic surgery. We analyzed the effect of hypoxanthine on neuroinflammatory parameters, such as cytokine levels, immunocontent of NF-κB/p65 subunit, iNOS immunocontent, nitrite levels, as well as IBA1 and GFAP immunocontent in striatum of infant and young adult rats. We also evaluate some oxidative parameters, including reactive species production, superoxide dismutase, catalase, glutathione peroxidase activities, as well as DNA damage. Wistar rats of 21 and 60 days of life underwent stereotactic surgery and were divided into two groups: control (infusion of saline 0.9 %) and hypoxanthine (10 µM). Intrastriatal administration of hypoxanthine increased IL-6 levels in striatum of both ages of rats tested, while TNF-α increased only in 21-day-old rats. Hypoxanthine also increased nuclear immunocontent of NF-κB/p65 subunit in striatum of both ages of rats. Nitrite levels were decreased in striatum of 21-day-old rats; however, the immunocontent of iNOS was increased in striatum of hypoxanthine groups. Microglial and astrocyte activation was seen by the increase in IBA1 and GFAP immunocontent, respectively, in striatum of infant rats. All oxidative parameters were altered, suggesting a strong neurotoxic hypoxanthine role on oxidative stress. According to our results, hypoxanthine intrastriatal administration increases neuroinflammatory parameters perhaps through oxidative misbalance, suggesting that this process may be involved, at least in part, to neurological disorders found in patients with Lesch-Nyhan disease.


Subject(s)
Corpus Striatum/metabolism , Corpus Striatum/pathology , Hypoxanthine/administration & dosage , Hypoxanthine/pharmacology , Inflammation/metabolism , Inflammation/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Corpus Striatum/drug effects , Cytokines/metabolism , Cytosol/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats, Wistar
15.
Mol Neurobiol ; 54(8): 5752-5767, 2017 10.
Article in English | MEDLINE | ID: mdl-27660266

ABSTRACT

In the present work, we focused on mechanisms of methylmercury (MeHg) toxicity in primary astrocytes and neurons of rats. Cortical astrocytes and neurons exposed to 0.5-5 µM MeHg present a link among morphological alterations, glutathione (GSH) depletion, glutamate dyshomeostasis, and cell death. Disrupted neuronal cytoskeleton was assessed by decreased neurite length and neurite/neuron ratio. Astrocytes presented reorganization of actin and glial fibrillary acidic protein (GFAP) networks and reduced cytoplasmic area. Glutamate uptake and Na+K+ATPase activity in MeHg-treated astrocytes were preserved; however, downregulated EAAC1-mediated glutamate uptake was associated with impaired Na+K+ATPase activity in neurons. Oxidative imbalance was found in astrocytes and neurons through increased 2'7'-dichlorofluorescein (DCF) production and misregulated superoxide dismutase (SOD), catalase (CAT), and glutathione reductase (GPX) activities. Glutathione (GSH) levels were downregulated in both astrocytes and neurons. MeHg reduced neuronal viability and induced caspase 3-dependent apoptosis together with downregulated PI3K/Akt pathway. In astrocytes, necrotic death was associated with increased TNF-α and JNK/MAPK activities. Cytoskeletal remodeling and cell death were fully prevented in astrocytes and neurons by GSH, but not melatonin or Trolox supplementation. These findings support a role for depleted GSH in the cytotoxicity of MeHg leading to disruption of the cytoskeleton and cell death. Moreover, in neurons, glutamate antagonists also prevented cytoskeletal disruption and neuronal death. We propose that cytoskeleton is an end point in MeHg cytotoxicity. Oxidative imbalance and glutamate mechanisms mediate MeHg cytoskeletal disruption and apoptosis in neurons. Otherwise, redox imbalance and glutamate-independent mechanisms disrupted the cytoskeleton and induced necrosis in MeHg-exposed astrocyte.


Subject(s)
Astrocytes/drug effects , Cytoskeleton/drug effects , Methylmercury Compounds/pharmacology , Neurons/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Female , Neurons/drug effects , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism
16.
Mol Neurobiol ; 54(6): 4127-4137, 2017 08.
Article in English | MEDLINE | ID: mdl-27324790

ABSTRACT

Classical galactosemia is an inborn error of carbohydrate metabolism in which patients accumulate high concentration of galactose in the brain. The most common treatment is a galactose-restricted diet. However, even treated patients develop several complications. One of the most common symptoms is motor coordination impairment, including affected gait, balance, and speech, as well as tremor and ataxia. In the present study, we investigated the effects of intracerebroventricular galactose administration on motor coordination, as well as on histological and biochemical parameters in cerebellum of adult rats. Wistar rats received 5 µL of galactose (4 mM) or saline by intracerebroventricular injection. The animals performed the beam walking test at 1 and 24 h after galactose administration. Histological and biochemical parameters were performed 24 h after the injections. The results showed motor coordination impairment at 24 h after galactose injection. Galactose also decreased the number of cells in the molecular and granular layers of the cerebellum. The immunohistochemistry results suggest that the cell types lost by galactose are neurons and astrocytes in the spinocerebellum and neurons in the cerebrocerebellum. Galactose increased active caspase-3 immunocontent and acetylcholinesterase activity, decreased acetylcholinesterase immunocontent, glutathione, and BDNF levels, as well as caused protein and DNA damage. Our results suggest that galactose induces histological and biochemical changes in cerebellum, which can be associated with motor coordination impairment.


Subject(s)
Cerebellum/pathology , Cerebellum/physiopathology , Galactose/pharmacology , Motor Activity/drug effects , Acetylcholinesterase/metabolism , Animals , Antigens, Nuclear/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Caspase 3/metabolism , Cell Count , Cerebellum/drug effects , DNA Damage , Galactose/administration & dosage , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Injections, Intraventricular , Male , Nerve Tissue Proteins/metabolism , Rats, Wistar , Sulfhydryl Compounds/metabolism
17.
Mol Neurobiol ; 54(6): 4201-4216, 2017 08.
Article in English | MEDLINE | ID: mdl-27324900

ABSTRACT

Although the use, and misuse, of methylphenidate is increasing in childhood and adolescence, there is little information about the consequences of this psychostimulant chronic use on brain and behavior during development. The aim of the present study was to investigate hippocampus biochemical, histochemical, and behavioral effects of chronic methylphenidate treatment to juvenile rats. Wistar rats received intraperitoneal injections of methylphenidate (2.0 mg/kg) or an equivalent volume of 0.9 % saline solution (controls), once a day, from the 15th to the 45th day of age. Results showed that chronic methylphenidate administration caused loss of astrocytes and neurons in the hippocampus of juvenile rats. BDNF and pTrkB immunocontents and NGF levels were decreased, while TNF-α and IL-6 levels, Iba-1 and caspase 3 cleaved immunocontents (microglia marker and active apoptosis marker, respectively) were increased. ERK and PKCaMII signaling pathways, but not Akt and GSK-3ß, were decreased. SNAP-25 was decreased after methylphenidate treatment, while GAP-43 and synaptophysin were not altered. Both exploratory activity and object recognition memory were impaired by methylphenidate. These findings provide additional evidence that early-life exposure to methylphenidate can have complex effects, as well as provide new basis for understanding of the biochemical and behavioral consequences associated with chronic use of methylphenidate during central nervous system development.


Subject(s)
Astrocytes/pathology , Behavior, Animal/drug effects , Hippocampus/pathology , Methylphenidate/toxicity , Neurons/pathology , Animals , Antigens, Nuclear/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cytokines/metabolism , Exploratory Behavior/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Maze Learning/drug effects , Memory/drug effects , Models, Biological , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Recognition, Psychology , Signal Transduction , Synaptosomal-Associated Protein 25/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
Biochim Biophys Acta ; 1863(12): 3001-3014, 2016 12.
Article in English | MEDLINE | ID: mdl-27663072

ABSTRACT

QUIN is a glutamate agonist playing a role in the misregulation of the cytoskeleton, which is associated with neurodegeneration in rats. In this study, we focused on microglial activation, FGF2/Erk signaling, gap junctions (GJs), inflammatory parameters and redox imbalance acting on cytoskeletal dynamics of the in QUIN-treated neural cells of rat striatum. FGF-2/Erk signaling was not altered in QUIN-treated primary astrocytes or neurons, however cytoskeleton was disrupted. In co-cultured astrocytes and neurons, QUIN-activated FGF2/Erk signaling prevented the cytoskeleton from remodeling. In mixed cultures (astrocyte, neuron, microglia), QUIN-induced FGF-2 increased level failed to activate Erk and promoted cytoskeletal destabilization. The effects of QUIN in mixed cultures involved redox imbalance upstream of Erk activation. Decreased connexin 43 (Cx43) immunocontent and functional GJs, was also coincident with disruption of the cytoskeleton in primary astrocytes and mixed cultures. We postulate that in interacting astrocytes and neurons the cytoskeleton is preserved against the insult of QUIN by activation of FGF-2/Erk signaling and proper cell-cell interaction through GJs. In mixed cultures, the FGF-2/Erk signaling is blocked by the redox imbalance associated with microglial activation and disturbed cell communication, disrupting the cytoskeleton. Thus, QUIN signal activates differential mechanisms that could stabilize or destabilize the cytoskeleton of striatal astrocytes and neurons in culture, and glial cells play a pivotal role in these responses preserving or disrupting a combination of signaling pathways and cell-cell interactions. Taken together, our findings shed light into the complex role of the active interaction of astrocytes, neurons and microglia in the neurotoxicity of QUIN.


Subject(s)
Astrocytes/drug effects , Cytoskeleton/drug effects , Excitatory Amino Acid Agonists/toxicity , Microglia/drug effects , Quinolinic Acid/toxicity , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/drug effects , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Gene Expression Regulation , MAP Kinase Signaling System , Microglia/cytology , Microglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Pregnancy , Primary Cell Culture , Rats , Rats, Wistar
19.
Biochim Biophys Acta ; 1860(11 Pt A): 2510-2520, 2016 11.
Article in English | MEDLINE | ID: mdl-27475002

ABSTRACT

BACKGROUND: Diphenylditelluride (PhTe)2 is a potent neurotoxin disrupting the homeostasis of the cytoskeleton. METHODS: Cultured astrocytes and neurons were incubated with (PhTe)2, receptor antagonists and enzyme inhibitors followed by measurement of the incorporation of [32P]orthophosphate into intermediate filaments (IFs). RESULTS: (PhTe)2 caused hyperphosphorylation of glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits (NFL, NFM and NFH) from primary astrocytes and neurons, respectively. These mechanisms were mediated by N-methyl-d-aspartate (NMDA) receptors, L-type voltage-dependent calcium channels (L-VDCCs) as well as metabotropic glutamate receptors upstream of phospholipase C (PLC). Upregulated Ca(2+) influx activated protein kinase A (PKA) and protein kinase C (PKC) in astrocytes causing hyperphosphorylation of GFAP and vimentin. Hyperphosphorylated (IF) together with RhoA-activated stress fiber formation, disrupted the cytoskeleton leading to altered cell morphology. In neurons, the high intracellular Ca(2+) levels activated the MAPKs, Erk and p38MAPK, beyond PKA and PKC, provoking hyperphosphorylation of NFM, NFH and NFL. CONCLUSIONS: Our findings support that intracellular Ca(2+) is one of the crucial signals that modulate the action of (PhTe)2 in isolated cortical astrocytes and neurons modulating the response of the cytoskeleton against the insult. GENERAL SIGNIFICANCE: Cytoskeletal misregulation is associated with neurodegeneration. This compound could be a valuable tool to induce molecular changes similar to those found in different pathologies of the brain.


Subject(s)
Actin Cytoskeleton/metabolism , Astrocytes/drug effects , Benzene Derivatives/pharmacology , Calcium Signaling , Neurons/drug effects , Organometallic Compounds/pharmacology , Animals , Astrocytes/metabolism , Benzene Derivatives/toxicity , Cells, Cultured , Neurons/metabolism , Organometallic Compounds/toxicity , Rats , Rats, Wistar
20.
Int J Dev Neurosci ; 50: 1-6, 2016 May.
Article in English | MEDLINE | ID: mdl-26948151

ABSTRACT

Tissue accumulation of galactose is a hallmark in classical galactosemia. Cognitive deficit is a symptom of this disease which is poorly understood. The aim of this study was to investigate the effects of intracerebroventricular administration of galactose on memory (inhibitory avoidance and novel object recognition tasks) of adult rats. We also investigated the effects of galactose on acetylcholinesterase (AChE) activity, immunocontent and gene expression in hippocampus and cerebral cortex. Wistar rats received a single injection of galactose (4mM) or saline (control). For behavioral parameters, galactose was injected 1h or 24h previously to the testing. For biochemical assessment, animals were decapitated 1h, 3h or 24h after galactose or saline injection; hippocampus and cerebral cortex were dissected. Results showed that galactose impairs the memory formation process in aversive memory (inhibitory avoidance task) and recognition memory (novel object recognition task) in rats. The activity of AChE was increased, whereas the gene expression of this enzyme was decreased in hippocampus, but not in cerebral cortex. These findings suggest that these changes in AChE may, at least in part, to lead to memory impairment caused by galactose. Taken together, our results can help understand the etiopathology of classical galactosemia.


Subject(s)
Acetylcholinesterase/metabolism , Avoidance Learning/drug effects , Galactose/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Memory Disorders/chemically induced , Acetylcholinesterase/genetics , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Exploratory Behavior/drug effects , Hippocampus/drug effects , Hippocampus/enzymology , Inhibition, Psychological , Injections, Intraventricular , Male , Rats , Rats, Wistar , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL