Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
J Neurosci Res ; 93(2): 268-84, 2015 Feb.
Article En | MEDLINE | ID: mdl-25306914

Quinolinic acid (QUIN) is an endogenous metabolite of the kynurenine pathway involved in several neurological disorders. Among the several mechanisms involved in QUIN-mediated toxicity, disruption of the cytoskeleton has been demonstrated in striatally injected rats and in striatal slices. The present work searched for the actions of QUIN in primary striatal neurons. Neurons exposed to 10 µM QUIN presented hyperphosphorylated neurofilament (NF) subunits (NFL, NFM, and NFH). Hyperphosphorylation was abrogated in the presence of protein kinase A and protein kinase C inhibitors H89 (20 µM) and staurosporine (10 nM), respectively, as well as by specific antagonists to N-methyl-D-aspartate (50 µM DL-AP5) and metabotropic glutamate receptor 1 (100 µM MPEP). Also, intra- and extracellular Ca(2+) chelators (10 µM BAPTA-AM and 1 mM EGTA, respectively) and Ca(2+) influx through L-type voltage-dependent Ca(2+) channel (10 µM verapamil) are implicated in QUIN-mediated effects. Cells immunostained for the neuronal markers ßIII-tubulin and microtubule-associated protein 2 showed altered neurite/neuron ratios and neurite outgrowth. NF hyperphosphorylation and morphological alterations were totally prevented by conditioned medium from QUIN-treated astrocytes. Cocultured astrocytes and neurons interacted with one another reciprocally, protecting them against QUIN injury. Cocultured cells preserved their cytoskeletal organization and cell morphology together with unaltered activity of the phosphorylating system associated with the cytoskeleton. This article describes cytoskeletal disruption as one of the most relevant actions of QUIN toxicity in striatal neurons in culture with soluble factors secreted by astrocytes, with neuron-astrocyte interaction playing a role in neuroprotection.


Astrocytes/physiology , Cell Communication/physiology , Corpus Striatum/cytology , Cytoskeleton/metabolism , Homeostasis/drug effects , Neurons/drug effects , Quinolinic Acid/pharmacology , Animals , Animals, Newborn , Astrocytes/chemistry , Cell Communication/drug effects , Cell Survival/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Phosphorylation/drug effects , Pregnancy , Rats , Rats, Wistar , Valine/analogs & derivatives , Valine/pharmacology
2.
Metab Brain Dis ; 28(3): 429-38, 2013 Sep.
Article En | MEDLINE | ID: mdl-23378107

Carbonyl compounds such as methylglyoxal (MGO) seem to play an important role in complications resulting from diabetes mellitus, in aging and neurodegenerative disorders. In this study, we are showing, that MGO is able to suppress cell viability and induce apoptosis in the cerebral cortex and hippocampus of neonatal rats ex-vivo. These effects are partially related with ROS production, evaluated by DCFH-DA assay. Coincubation of MGO and reduced glutathione (GSH) or Trolox (vitamin E) totally prevented ROS production but only partially prevented the MGO-induced decreased cell viability in the two brain structures, as evaluated by the MTT assay. Otherwise, L-NAME, a nitric oxide (NO) inhibitor, partially prevented ROS production in the two structures but partially prevented cytotoxicity in the hippocampus. Pharmacological inhibition of Erk, has totally attenuated MGO-induced ROS production and cytotoxicity, suggesting that MEK/Erk pathway could be upstream of ROS generation and cell survival. Otherwise, p38MAPK and JNK failed to prevent ROS generation but induced decreased cell survival consistent with ROS-independent mechanisms. We can propose that Erk, p38MAPK and JNK are involved in the cytotoxicity induced by MGO through different signaling pathways. While Erk could be an upstream effector of ROS generation, p38MAPK and JNK seem to be associated with ROS-independent cytotoxicity in neonatal rat brain. The cytotoxic damage progressed to apoptotic cell death at MGO concentration higher than those described for adult brain, suggesting that the neonatal brain is resistant to MGO-induced cell death. The consequences of MGO-induced brain damage early in life, remains to be clarified. However, it is feasible that high MGO levels during cortical and hippocampal development could be, at least in part, responsible for the impairment of cognitive functions in adulthood.


Brain/pathology , Mitogen-Activated Protein Kinases/metabolism , Oxidative Stress/drug effects , Pyruvaldehyde/toxicity , Animals , Animals, Newborn , Annexin A5/metabolism , Antioxidants/pharmacology , Blotting, Western , Brain/drug effects , Brain/enzymology , Cell Survival/drug effects , Coloring Agents , Fluorescent Dyes , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , MAP Kinase Kinase 4/metabolism , Nerve Tissue Proteins/metabolism , Pyruvaldehyde/antagonists & inhibitors , Pyruvaldehyde/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Tetrazolium Salts , Thiazoles , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Exp Cell Res ; 319(3): 89-104, 2013 Feb 01.
Article En | MEDLINE | ID: mdl-23142028

Hyperprolinemia is an inherited disorder of proline (Pro) metabolism and patients affected by this disease may present neurological manifestations. However, the mechanisms of neural excitotoxicity elicited by hyperprolinemia are far from being understood. Considering the pivotal role of cytoskeletal remodeling in several neurodegenerative pathologies and the potential links between cytoskeleton, reactive oxygen species production and cell death, the aim of the present work was to study the effects of Pro on astrocyte and neuron cytoskeletal remodeling and the possible oxidative stress involvement. Pro induced a shift of actin cytoskeleton in stress fibers together with increased RhoA immunocontent and ERK1/2 phosphorylation/activation in cortical astrocytes. Unlike astrocytes, results evidenced little susceptibility of neuron cytoskeleton remodeling, since Pro-treated neurons presented unaltered neuritogenesis. We observed increased hydrogen peroxide production characterizing oxidative stress together with decreased superoxide dismutase (SOD) and catalase (CAT) activities in cortical astrocytes after Pro treatment, while glutathione peroxidase (GSHPx) activity remained unaltered. However, coincubation with Pro and Trolox/melatonin prevented decreased SOD and CAT activities in Pro-treated astrocytes. Accordingly, these antioxidants were able to prevent the remodeling of the actin cytoskeleton, RhoA increased levels and ERK1/2 phosphorylation in response to high Pro exposure. Taken together, these findings indicated that the cytoskeleton of cortical astrocytes, but not of neurons in culture, is a target to Pro and such effects could be mediated, at least in part, by redox imbalance, RhoA and ERK1/2 signaling pathways. The vulnerability of astrocyte cytoskeleton may have important implications for understanding the effects of Pro in the neurotoxicity linked to inborn errors of Pro metabolism.


Astrocytes/drug effects , Cerebral Cortex/drug effects , Cytoskeleton/drug effects , Oxidative Stress/drug effects , Proline/pharmacology , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Animals, Newborn , Antioxidants/metabolism , Astrocytes/metabolism , Astrocytes/physiology , Astrocytes/ultrastructure , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/physiology , Embryo, Mammalian , Oxidative Stress/physiology , Proline/adverse effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
4.
Neurotoxicology ; 34: 175-88, 2013 Jan.
Article En | MEDLINE | ID: mdl-23182946

In the present report 15 day-old rats were injected with 0.3µmol of diphenyl ditelluride (PhTe)(2)/kg body weight and parameters of neurodegeneration were analyzed in slices from cerebellum 3 and 6 days afterwards. The earlier responses, at day 3 after injection, included hyperphosphorylation of intermediate filament (IF) proteins from astrocyte (glial fibrillary acidic protein - GFAP - and vimentin) and neuron (low-, medium- and high molecular weight neurofilament subunits: NF-L, NF-M and NF-H); increased mitogen-activated protein kinase (MAPK) (Erk and p38MAPK) and cAMP-dependent protein kinase (PKA) activities. Also, reactive astrogliosis takes part of the early responses to the insult with (PhTe)(2), evidenced by upregulated GFAP in Western blot, PCR and immunofluorescence analysis. Six days after (PhTe)(2) injection we found persistent astrogliosis, increased propidium iodide (PI) positive cells in NeuN positive population evidenced by flow cytometry and reduced immunofluorescence for NeuN, suggesting that the in vivo exposure to (PhTe)(2) progressed to neuronal death. Moreover, activated caspase 3 suggested apoptotic neuronal death. Neurodegeneration was related with decreased [(3)H]glutamate uptake and decreased Akt immunoreactivity, however phospho-GSK-3-ß (Ser9) was not altered in (PhTe)(2) injected rat. Therefore, the present results show that the earlier cerebellar responses to (PhTe)(2) include disruption of cytoskeletal homeostasis that could be related with MAPK and PKA activation and reactive astrogliosis. Akt inhibition observed at this time could also play a role in the neuronal death evidenced afterwards. The later events of the neurodegenerative process are characterized by persistent astrogliosis and activation of apoptotic neuronal death through caspase 3 mediated mechanisms, which could be related with glutamate excitotoxicity. The progression of these responses are therefore likely to be critical for the outcome of the neurodegeneration provoked by (PhTe)(2) in rat cerebellum.


Apoptosis/drug effects , Astrocytes/drug effects , Benzene Derivatives/toxicity , Cerebellum/drug effects , Cytoskeleton/drug effects , Nerve Degeneration , Organometallic Compounds/toxicity , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/pathology , Benzene Derivatives/administration & dosage , Blotting, Western , Caspase 3/metabolism , Cerebellum/metabolism , Cerebellum/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Cytoskeleton/pathology , Female , Flow Cytometry , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Homeostasis , Injections, Subcutaneous , Male , Mitogen-Activated Protein Kinases/metabolism , Neurofilament Proteins/metabolism , Organometallic Compounds/administration & dosage , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors , Vimentin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Neurotoxicology ; 33(5): 1106-16, 2012 Oct.
Article En | MEDLINE | ID: mdl-22705628

In the present report we examined the effect of maternal exposure to diphenyl ditelluride (PhTe)(2) (0.01 mg/kg body weight) during the first 14 days of lactational period on the activity of some protein kinases targeting the cytoskeleton of striatum and cerebellum of their offspring. We analyzed the phosphorylating system associated with glial fibrillary acidic protein (GFAP), and neurofilament of low, medium and high molecular weight (NF-L, NF-M and NF-H, respectively) of pups on PND 15, 21, 30 and 45. We found that (PhTe)(2) induced hyperphosphorylation of all the proteins studied on PND 15 and 21, recovering control values on PND 30 and 45. The immunocontent of GFAP, NF-L, NF-M and NF-H in the cerebellum of 15-day-old pups was increased. Western blot assays showed activation/phosphorylation of Erk1/2 on PND 21 and activation/phosphorylation of JNK on PND 15. Otherwise, p38MAPK was not activated in the striatum of (PhTe)(2) exposed pups. On the other hand, the cerebellum of pups exposed to (PhTe)(2) presented activated/phosphorylated Erk1/2 on PND 15 and 21 as well as activated/phosphorylated p38MAPK on PND 21, while JNK was not activated. Western blot assays showed that both in the striatum and in the cerebellum of (PhTe)(2) exposed pups, the immunocontent of the catalytic subunit of PKA (PKAcα) was increased on PND 15. Western blot showed that the phosphorylation level of NF-L Ser55 and NF-M/NF-H KSP repeats was increased in the striatum and cerebellum of both 15- and 21-day-old pups exposed to (PhTe)(2). Diphenyl diselenide (PhSe)(2), the selenium analog of (PhTe)(2), prevented (PhTe)(2)-induced hyperphosphorylation of striatal intermediate filament (IF) proteins but it failed to prevent the action of (PhTe)(2) in cerebellum. Western blot assay showed that the (PhSe)(2) prevented activation/phosphorylation of Erk1/2, JNK and PKAcα but did not prevent the stimulatory effect of (PhTe)(2) on p38MAPK in cerebellum at PND 21. In conclusion, this study demonstrated that dam exposure to low doses of (PhTe)(2) can alter cellular signaling targeting the cytoskeleton of striatum and cerebellum in the offspring in a spatiotemporal manner, which can be related to the neurotoxic effects of (PhTe)(2).


Benzene Derivatives/toxicity , Cerebellum/cytology , Corpus Striatum/cytology , Cytoskeleton/metabolism , Homeostasis/drug effects , Neurons/drug effects , Organometallic Compounds/toxicity , Prenatal Exposure Delayed Effects/pathology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Body Weight/drug effects , Female , Gene Expression Regulation, Developmental/drug effects , Glial Fibrillary Acidic Protein/metabolism , In Vitro Techniques , Neurofilament Proteins/metabolism , Neurons/metabolism , Phosphates/pharmacokinetics , Phosphorus Isotopes/pharmacokinetics , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar , Signal Transduction/drug effects , Time Factors
6.
J Neuroimmunol ; 249(1-2): 8-15, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22560157

In this study we investigated the anti-inflammatory effects of chronic ethanol (EtOH) treatment on lipopolysaccharide (LPS)-stimulated C6 glioma cells. The cells were chronically treated with 200mM EtOH; coincubation with LPS and EtOH was obtained upon addition of 2µg/ml LPS to the incubation medium in the last 24h of EtOH exposure. We found that EtOH prevented the LPS-induced production of tumor necrosis factor α (TNFα) without decreasing cell viability. Either LPS treated or EtOH plus LPS treated cells presented upregulated glial fibrillary acidic protein (GFAP) and downregulated vimentin levels characterizing a program of reactive astrogliosis. Also, EtOH plus LPS stimulation greatly increased the oxidative stress generation evaluated by DCF-DA measurement, while either EtOH alone or LPS alone was unable to induce oxidative stress. Western blot analysis indicated that either EtOH, LPS or EtOH plus LPS treatments are unable to affect Akt/GSK3ß signaling pathway. However, LPS alone and EtOH plus LPS co-treatment inhibited Erk phosphorylation. A dramatic loss of stress fibers was found in EtOH exposed cells, evaluated by cytochemistry using phalloidin-fluorescein. However, LPS alone was not able to disrupt actin organization. Furthermore, cells co-incubated with LPS and EtOH presented reversion of the disrupted stress fibers provoked by EtOH. Supporting this action, RhoA and vinculin immunocontent were upregulated in response to EtOH plus LPS. Interestingly, EtOH suppresses the inflammatory cascade (TNFα production) in response to LPS. Concomitantly it sustains Erk inhibition, increases oxidative stress generation and induces reactive astrogliosis in the presence of LPS, conditions associated with neurotoxicity. The effects observed were not supported by actin reorganization. Altogether, these findings suggest that Erk signaling inhibition could play a role in both suppressing TNFα production and inducing oxidative stress generation and astrogliosis, therefore modulating a dual action of EtOH plus LPS in glial cells.


Anti-Inflammatory Agents/pharmacology , Ethanol/pharmacology , MAP Kinase Signaling System/drug effects , Neuroglia/drug effects , Oxidative Stress/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival , Electrophoresis, Polyacrylamide Gel , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/pathology , Immunohistochemistry , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Neuroglia/metabolism , Neuroglia/pathology , Rats , Stress Fibers/drug effects , Stress Fibers/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Exp Neurol ; 233(1): 391-9, 2012 Jan.
Article En | MEDLINE | ID: mdl-22116044

The studies of signaling mechanisms involved in the disruption of the cytoskeleton homeostasis were performed in a model of quinolinic acid (QUIN) neurotoxicity in vitro. This investigation focused on the phosphorylation level of intermediate filament (IF) subunits of astrocytes (glial fibrillary acidic protein - GFAP) and neurons (low, medium and high molecular weight neurofilament subunits - NFL, NFM and NFH, respectively). The activity of the phosphorylating system associated with the IFs was investigated in striatal slices of rat exposed to QUIN or treated simultaneously with QUIN plus glutamate receptor antagonists, calcium channel blockers or kinase inhibitors. Results showed that in astrocytes, the action of 100 µM QUIN was mainly due to increased Ca(2+) influx through NMDA and L-type voltage-dependent Ca(2+) channels (L-VDCC). In neuronal cells QUIN acted through metabotropic glutamate receptor (mGluR) activation and influx of Ca(2+) through NMDA receptors and L-VDCC, as well as Ca(2+) release from intracellular stores. These mechanisms then set off a cascade of events including activation of PKA, PKCaMII and PKC, which phosphorylate head domain sites on GFAP and NFL. Also, Cdk5 was activated downstream of mGluR5, phosphorylating the KSP repeats on NFM and NFH. mGluR1 was upstream of phospholipase C (PLC) which, in turn, produced diacylglycerol (DAG) and inositol 3,4,5 triphosphate (IP3). DAG is important to activate PKC and phosphorylate NFL, while IP(3) contributed to Ca(2+) release from internal stores promoting hyperphosphorylation of KSP repeats on the tail domain of NFM and NFH. The present study supports the concept of glutamate and Ca(2+) contribution in excitotoxic neuronal damage provoked by QUIN associated to dysfunction of the cytoskeleton homeostasis and highlights the differential signaling mechanisms elicited in striatal astrocytes and neurons.


Astrocytes/cytology , Corpus Striatum/cytology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Neurons/cytology , Quinolinic Acid/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Animals , Astrocytes/drug effects , Cell Survival/drug effects , Corpus Striatum/drug effects , Drug Interactions , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Rats , Rats, Wistar
8.
Arch Toxicol ; 86(2): 217-30, 2012 Feb.
Article En | MEDLINE | ID: mdl-21863293

We studied the effect of different concentrations of diphenyl ditelluride (PhTe)(2) on the in vitro phosphorylation of glial fibrillary acidic protein (GFAP) and neurofilament (NF) subunits from cerebral cortex and hippocampus of rats during development. (PhTe)(2)-induced hypophosphorylation of GFAP and NF subunits only in cerebral cortex of 9- and 15-day-old animals but not in hippocampus. Hypophosphorylation was dependent on ionotropic glutamate receptors, as demonstrated by the specific inhibitors 10 µM DL-AP5 and 50 µM MK801, 100 µM CNQX and 100 µM DNQX. Also, 10 µM verapamil and 10 µM nifedipine, two L-voltage-dependent Ca(2+) channels (L-VDCC) blockers; 50 µM dantrolene, a ryanodine channel blocker, and the intracellular Ca(2+) chelator Bapta-AM (50 µM) totally prevented this effect. Results obtained with 0.2 µM calyculin A (PP1 and PP2A inhibitor), 1 µM Fostriecin a potent protein phosphatase 2A (PP2A) inhibitor, 100 µM FK-506 or 100 µM cyclosporine A, specific protein phosphatase 2B inhibitors, pointed to PP1 as the protein phosphatase directly involved in the hypophosphorylating effect of (PhTe)(2). Finally, we examined the activity of DARPP-32, an important endogenous Ca(2+)-mediated inhibitor of PP1 activity. Western blot assay using anti-DARPP-32, anti-pThr34DARPP-32, and anti-pThr75DARPP-32 antibodies showed a decreased phosphorylation level of the inhibitor at Thr34, compatible with inactivation of protein kinase A (PKA) by pThr75 DARPP-32. Decreased cAMP and catalytic subunit of PKA support that (PhTe)(2) acted on neuron and astrocyte cytoskeletal proteins through PKA-mediated inactivation of DARPP-32, promoting PP1 release and hypophosphorylation of IF proteins of those neural cells. Moreover, in the presence of Bapta, the level of the PKA catalytic subunit was not decreased by (PhTe)(2), suggesting that intracellular Ca(2+) levels could be upstream the signaling pathway elicited by this neurotoxicant and targeting the cytoskeleton.


Benzene Derivatives/pharmacology , Cerebral Cortex/drug effects , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Intermediate Filaments/drug effects , Organometallic Compounds/pharmacology , Animals , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intermediate Filaments/metabolism , Phosphorylation/drug effects , Protein Phosphatase 1/metabolism , Rats , Signal Transduction
9.
Chem Res Toxicol ; 24(10): 1754-64, 2011 Oct 17.
Article En | MEDLINE | ID: mdl-21879721

In the present report, we showed that diphenyl ditelluride (PhTe)(2) induced in vitro hyperphosphorylation of glial fibrillary acidic protein (GFAP), vimentin and neurofilament (NF) subunits in hippocampus of 21 day-old rats. Hyperphosphorylation was dependent on L-voltage dependent Ca(2+) channels (L-VDCC), N-methyl-d-aspartate (NMDA) and metabotropic glutamate receptors, as demonstrated by the specific inhibitors verapamil, DL-AP5 and MCPG, respectively. Also, dantrolene, a ryanodine channel blocker, EGTA and Bapta-AM, extra and intracellular Ca(2+) chelators respectively, totally prevented this effect. Activation of metabotropic glutamate receptors by (PhTe)(2) upregulates phospholipase C (PLC), producing inositol 1, 4, 5-trisphosphate (IP(3)) and diacylglycerol (DAG). Therefore, high Ca(2+) levels and DAG directly activate Ca(2+)/calmodulin-dependent protein kinase (PKCaMII) and protein kinase C (PCK), resulting in the hyperphosphorylation of Ser-57 in the carboxyl-terminal tail domain of the low molecular weight NF subunit (NF-L). Also, the activation of Erk1/2, and p38MAPK resulted in hyperphosphorylation of KSP repeats of the medium molecular weight NF subunit (NF-M). It is noteworthy that PKCaMII and PKC inhibitors prevented (PhTe)(2)-induced Erk1/2MAPK and p38MAPK activation as well as hyperphosphorylation of KSP repeats on NF-M, suggesting that PKCaMII and PKC could be upstream of this activation. Taken together, our results highlight the role of Ca(2+) as a mediator of the (PhTe)(2)-elicited signaling targeting specific phosphorylation sites on IF proteins of neural cells of rat hippocampus. Interestingly, this action shows a significant cross-talk among signaling pathways elicited by (PhTe)(2), connecting glutamate metabotropic cascade with activation of Ca(2+) channels. The extensively phosphorylated amino- and carboxyl- terminal sites could explain, at least in part, the neural dysfunction associated with (PhTe)(2) exposure.


Benzene Derivatives/toxicity , Calcium/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , MAP Kinase Signaling System/drug effects , Neurofilament Proteins/metabolism , Organometallic Compounds/toxicity , Vimentin/metabolism , Animals , Benzylamines/pharmacology , Blotting, Western , Calcium Channels, L-Type/metabolism , Cerebral Cortex/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Electrophoresis, Polyacrylamide Gel , Hippocampus/metabolism , In Vitro Techniques , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Wistar , Staurosporine/pharmacology , Sulfonamides/pharmacology
...