Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 35(12): 1966-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21110090

ABSTRACT

Overactivation of glutamate receptors results in neurodegeneration in a variety of brain pathologies, including ischemia, epilepsy, traumatic brain injury and slow-progressing neurodegenerative disorders. In all these pathologies, it is well accepted that the calcium-dependent cysteine proteases calpains are key players in the mechanisms of neuronal cell death. Many research groups have been actively pursuing to establish a link between the deregulation of intracellular Ca(2+) homeostasis associated with excitotoxicity and calpain activity. It is well established that these two events are connected and interact synergistically to promote neurodegeneration, but whether calpain activity depends on or contributes to Ca(2+) deregulation is still under debate.


Subject(s)
Calcium/physiology , Calpain/physiology , Receptors, Glutamate/physiology , Animals , Calpain/metabolism , Enzyme Activation , Homeostasis , Humans
2.
Neurochem Res ; 33(8): 1501-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18368487

ABSTRACT

We investigated the contribution of L-, N- and P/Q-type Ca(2+) channels to the [Ca(2+)](i) changes, evoked by kainate, in the cell bodies of hippocampal neurons, using a pharmacological approach and Ca(2+) imaging. Selective Ca(2+) channel blockers, namely nitrendipine, omega-Conotoxin GVIA (omega-GVIA) and omega-Agatoxin IVA (omega-AgaIVA) were used. The [Ca(2+)](i) changes evoked by kainate presented a high variability, and were abolished by NBQX, a AMPA/kainate receptor antagonist, but the N-methyl-D-aspartate (NMDA) receptor antagonist, D-AP5, was without effect. Each Ca(2+) channel blocker caused differential inhibitory effects on [Ca(2+)](i) responses evoked by kainate. We grouped the neurons for each blocker in three subpopulations: (1) neurons with responses below 60% of the control; (2) neurons with responses between 60% and 90% of the control, and (3) neurons with responses above 90% of the control. The inhibition caused by nitrendipine was higher than the inhibition caused by omega-GVIA or omega-AgaIVA. Thus, in the presence of nitrendipine, the percentage of cells with responses below 60% of the control was 41%, whereas in the case of omega-GVIA or omega-AgaIVA the values were 9 or 17%, respectively. The results indicate that hippocampal neurons differ in what concerns their L-, N- and P/Q-type Ca(2+) channels activated by stimulation of the AMPA/kainate receptors.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Excitatory Amino Acid Agonists/pharmacology , Hippocampus , Kainic Acid/pharmacology , Neurons/drug effects , Animals , Calcium/metabolism , Calcium Channel Blockers/metabolism , Cells, Cultured , Excitatory Amino Acid Antagonists/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Neurons/cytology , Neurons/metabolism , Nitrendipine/metabolism , Quinoxalines/metabolism , Rats , Rats, Wistar , omega-Agatoxin IVA/metabolism , omega-Conotoxin GVIA/metabolism
3.
J Neurosci Res ; 83(5): 832-44, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16477614

ABSTRACT

Brain-derived neurotrophic factor (BDNF) modulates glutamatergic excitatory transmission in hippocampal primary cultures by acting at a presynaptic locus. Although it has been suggested that BDNF also modulates adult hippocampus glutamatergic transmission, this remains a matter of controversy. To clarify a putative role for this neurotrophin in the modulation of glutamate release we applied exogenous BDNF to isolated adult rat hippocampal nerve terminals. BDNF, at 100 ng/ml, potentiated by 25% the K(+)-evoked release of [(3)H]glutamate from hippocampal synaptosomes. The small effect of BDNF on [(3)H]glutamate release correlated with a modest increase in phospholipase Cgamma (PLCgamma) phosphorylation, and with the lack of effect of BDNF on extracellular-signal regulated kinase (ERK) and Akt phosphorylation. Immunocytochemistry studies demonstrated that only about one-third of glutamatergic synaptosomes were positive for TrkB immunoreactivity. Furthermore, biotinylation and subsynaptic fractionation studies showed that only one-fourth of total full-length TrkB was present at the plasma membrane, evenly distributed between the presynaptic active zone and the postsynaptic density. These results indicate that BDNF modulates synaptic transmission presynaptically in a small subset of hippocampal glutamatergic synapses that contain TrkB and that express the receptor on the plasma membrane.


Subject(s)
Glutamic Acid/metabolism , Hippocampus/metabolism , Presynaptic Terminals/metabolism , Receptor, trkB/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippocampus/drug effects , Immunoblotting , Immunohistochemistry , Male , Presynaptic Terminals/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Synaptosomes/drug effects , Synaptosomes/metabolism
4.
J Neurochem ; 91(6): 1322-31, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15584909

ABSTRACT

In this work, we investigated the involvement of calpains in the neurotoxicity induced by short-term exposure to kainate (KA) in non-desensitizing conditions of AMPA receptor activation (cyclothiazide present, CTZ), in cultured rat hippocampal neurons. The calpain inhibitor MDL28170 had a protective effect in cultures treated with KA plus CTZ (p < 0.01), preventing the decrease in MTT reduction caused by exposure to KA (p < 0.001). Caspase inhibition by ZVAD-fmk was not neuroprotective against the toxic effect of KA. At 1 h after treatment, we could already observe significantly increased calpain activity, which was prevented by MDL 28170 and NBQX. Western blot analysis of calpain substrates, GluR1, neuronal nitric oxide synthase (nNOS) and nonerythroid spectrin (fodrin), showed a time-dependent and MDL 28170-sensitive proteolysis of these proteins. This effect was due to calpains, but not caspases, since ZVAD-fmk was ineffective in preventing proteolytic events. Breakdown products of fodrin (BDPs) were detected as early as 15 min after exposure to KA. Overall, these results show early activation of calpains following activation of AMPA receptors as well as compromise of neuronal survival, likely due to proteolytic events that affect proteins involved in neuronal signaling.


Subject(s)
Calpain/metabolism , Hippocampus/physiology , Neurons/physiology , Receptors, AMPA/physiology , Animals , Calpain/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Survival/physiology , Cells, Cultured , Embryo, Mammalian , Hippocampus/cytology , Hippocampus/drug effects , Kainic Acid/poisoning , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurotoxins/poisoning , Rats , Rats, Wistar
5.
Cancer Lett ; 216(2): 183-9, 2004 Dec 28.
Article in English | MEDLINE | ID: mdl-15533594

ABSTRACT

Photodynamic therapy (PDT) of cancer is a promising technique based on the formation of singlet oxygen following irradiation of a sensitizer with visible light. In the present work we investigated the role of calpains in PDT, using the human lymphoblastoid CCRF-CEM cells and bisulfonated aluminum phthalocyanine (AlPcS2) as a sensitizer. Photosensitization induced apoptotic cell death and a time-dependent activation of calpains, as determined using the fluorogenic substrate succinyl-Leu-Leu-Val-Tyr-7-amido-4-methylcoumarin (SLLVY-AMC). However, inhibition of calpains with calpain inhibitor II or with PD 150606 did not affect the demise process. The results indicate that although calpains are activated in PDT, they do not play a major role in tumor cell death.


Subject(s)
Apoptosis , Calpain/metabolism , Photochemotherapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Indoles/pharmacology , Organometallic Compounds/pharmacology , Photosensitizing Agents/pharmacology , Time Factors
6.
Biochim Biophys Acta ; 1704(2): 59-86, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15363861

ABSTRACT

In photodynamic therapy (PDT) a sensitizer, light and oxygen are used to induce death of tumor cells and in the treatment of certain noncancerous conditions. Cell death in PDT may occur by apoptosis or by necrosis, depending on the sensitizer, on the PDT dose and on the cell genotype. Some sensitizers that have been used in PDT are accumulated in the mitochondria, and this may explain their efficiency in inducing apoptotic cell death, both in vitro and in vivo. In this review we will focus on the events that characterize apoptotic death in PDT and on the intracellular signaling events that are set in motion in photosensitized cells. Activation of phospholipases, changes in ceramide metabolism, a rise in the cytosolic free Ca2+ concentration, stimulation of nitric oxide synthase (NOS), changes in protein phosphorylation and alterations in the activity of transcription factors and on gene expression have all been observed in PDT-treated cells. Although many of these metabolic reactions contribute to the demise process, some of them may antagonize cell death. Understanding the signaling mechanisms in PDT may provide means to modulate the PDT effects at the molecular level and potentiate its antitumor effectiveness.


Subject(s)
Apoptosis , Calcium/metabolism , Photochemotherapy , Signal Transduction , Calcium/pharmacokinetics , Cyclic AMP/metabolism , Homeostasis , Humans , Mitogen-Activated Protein Kinase Kinases/pharmacology , Necrosis , Neoplasms/drug therapy , Neovascularization, Pathologic , Proteins/metabolism
7.
Exp Dermatol ; 13(1): 18-26, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15009112

ABSTRACT

Nuclear factor kappa B (NF-kB) and activating protein-1 (AP-1) transcription factors are ubiquitously expressed signaling molecules known to regulate the transcription of a large number of genes involved in immune responses, namely the inducible isoform of nitric oxide synthase (iNOS). In this study, we demonstrate that a fetal skin-derived dendritic cell line (FSDC) produces nitric oxide (NO) in response to the contact sensitizer nickel sulfate (NiSO(4)) and increases the expression of the iNOS protein, as determined by immunofluorescence and Western blot analysis. The sensitizer NiSO(4) increased cytoplasmic iNOS expression by 31.9 +/- 10.3% and nitrite production, as assayed by the Griess reaction, by 27.6 +/- 9.5%. Electrophoretic mobility shift assay (EMSA), showed that 30 min of FSDC exposure to NiSO(4) activates the transcription factor NF-kB by 58.2 +/- 7.0% and 2 h of FSDC exposure to NiSO(4) activates the transcription factor AP-1 by 26.0 +/- 1.4%. Together, these results indicate that NiSO(4) activates the NF-kB and AP-1 pathways and induces iNOS expression in skin dendritic cells.


Subject(s)
Dendritic Cells/metabolism , Irritants/pharmacology , NF-kappa B/metabolism , Nickel/pharmacology , Nitric Oxide Synthase/metabolism , Skin/metabolism , Transcription Factor AP-1/metabolism , Animals , Cell Line , Dendritic Cells/drug effects , Dendritic Cells/enzymology , Fetus , Fluorescent Antibody Technique , Immunohistochemistry , Mice , NF-kappa B/drug effects , Nitric Oxide Synthase/drug effects , Nitrites/metabolism , Skin/drug effects , Skin/enzymology , Transcription Factor AP-1/drug effects
8.
Neurochem Res ; 28(10): 1453-7, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14570390

ABSTRACT

The expression and localization of syntaxin isoforms 1A and 1B in adrenergic and noradrenergic chromaffin cells were examined by both immunoblot analysis and confocal immunofluorescence microscopy. Syntaxin 1A was found in higher levels in noradrenergic cells, whereas syntaxin 1B was similarly expressed in most noradrenergic and adrenergic cells. However, some heterogeneity was observed within each catecholaminergic phenotype. Although the majority of adrenergic cells appeared to express low levels of syntaxin 1A, about 7% was strongly stained for syntaxin 1A. A subpopulation of noradrenergic cells, about 17%, expressed greater levels of syntaxin 1B. Syntaxin 1B labeling showed a punctate appearance in the cytoplasm, whereas syntaxin 1A appeared predominantly localized to the plasma membrane. These data show differences in the exocytotic machinery of the two subtypes of chromaffin cells that may underlie some of the distinct characteristics of adrenaline and noradrenaline secretion.


Subject(s)
Antigens, Surface/metabolism , Chromaffin Cells/metabolism , Epinephrine/metabolism , Nerve Tissue Proteins/metabolism , Norepinephrine/metabolism , Adrenal Glands/cytology , Animals , Cattle , Cells, Cultured , Microscopy, Confocal , Microscopy, Fluorescence , Phenotype , Protein Isoforms/metabolism , Syntaxin 1
9.
J Neurochem ; 85(3): 791-800, 2003 May.
Article in English | MEDLINE | ID: mdl-12694405

ABSTRACT

In this work, we investigated the role of nitric oxide (NO) in neurotoxicity triggered by alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) receptor activation in cultured hippocampal neurons. In the presence of cyclothiazide (CTZ), short-term exposures to kainate (KA; 5 and 15 min, followed by 24-h recovery) decreased cell viability. Both NBQX and d-AP-5 decreased the neurotoxicity caused by KA plus CTZ. Long-term exposures to KA plus CTZ (24 h) resulted in increased toxicity. In short-, but not in long-term exposures, the presence of NO synthase (NOS) inhibitors (l-NAME and 7-NI) decreased the toxicity induced by KA plus CTZ. We also found that KA plus CTZ (15-min exposure) significantly increased cGMP levels. Furthermore, short-term exposures lead to decreased intracellular ATP levels, which was prevented by NBQX, d-AP-5 and NOS inhibitors. Immunoblot analysis revealed that KA induced neuronal NOS (nNOS) proteolysis, gradually lowering the levels of nNOS according to the time of exposure. Calpain, but not caspase-3 inhibitors, prevented this effect. Overall, these results show that NO is involved in the neurotoxicity caused by activation of non-desensitizing AMPA receptors, although to a limited extent, since AMPA receptor activation triggers mechanisms that lead to nNOS proteolysis by calpains, preventing a further contribution of NO to the neurotoxic process.


Subject(s)
Hippocampus/cytology , Kainic Acid/toxicity , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Adenosine Triphosphate/metabolism , Animals , Antibodies/pharmacology , Calpain/metabolism , Cells, Cultured , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Neurons/cytology , Neurons/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Peptide Hydrolases/metabolism , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
10.
FASEB J ; 17(9): 1118-20, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12692082

ABSTRACT

Glutamate and NPY have been implicated in hippocampal neuropathology in temporal lobe epilepsy. Thus, we investigated the involvement of NPY receptors in mediating neuroprotection against excitotoxic insults in organotypic cultures of rat hippocampal slices. Exposure of hippocampal slice cultures to 2 microM AMPA (alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionate) induced neuronal degeneration, monitored by propidium iodide uptake, of granule cells and CA1 pyramidal cells. For dentate granule cells, selective activation of Y1, Y2, or Y5 receptors with 1 microM [Leu31,Pro34]NPY, 300 nM NPY13-36 or 1 microM 500 nM NPY(19-23)-(Gly1,Ser3,Gln4,Thr6,Ala31,Aib32,Gln34)-PP, respectively, had a neuroprotective effect against AMPA, whereas only the activation of Y2 receptors was effective for CA1 pyramidal cells. When the slice cultures were exposed to 6 microM kainate, the CA3 pyramidal cells displayed significant degeneration, and in this case the activation of Y1, Y2, and Y5 receptors was neuroprotective. For the kainic acid-induced degeneration of CA1 pyramidal cells, it was again found that only the Y2 receptor activation was effective. Based on the present findings, it was concluded that Y1, Y2, and Y5 receptors effectively can modify glutamate receptor-mediated neurodegeneration in the hippocampus.


Subject(s)
Hippocampus/drug effects , Nerve Degeneration/prevention & control , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/physiology , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Hippocampus/cytology , Kainic Acid/toxicity , Nerve Degeneration/chemically induced , Neuroprotective Agents/metabolism , Organ Culture Techniques , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats , Receptors, Neuropeptide/physiology , Receptors, Neuropeptide Y/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity
11.
Neuropharmacology ; 44(2): 282-92, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12623227

ABSTRACT

We investigated the functional interaction between neuropeptide Y (NPY) receptors using nerve terminals and cultured rat hippocampal neurons, and we evaluated the involvement of voltage-gated Ca(2+) channels (VGCCs) in NPY receptors-induced inhibition of Ca(2+) influx and glutamate release. The KCl-evoked release of glutamate from hippocampal synaptosomes was inhibited by 1 microM NPY and this effect was insensitive to either BIBP3226 (Y1 receptor antagonist) or L-152,804 (Y5 receptor antagonist), but was sensitive to BIIE0246 (Y2 receptor antagonist). We could also pharmacologically dissect the NPY receptors activity by using Y1, Y2 and Y5 receptor agonists ([Leu(31),Pro(34)]NPY, NPY13-36, NPY (19-23)-(Gly(1),Ser(3),Gln(4),Thr(6),Ala(31),Aib(32),Gln(34))-pancreatic polypeptide (PP), respectively), and in all the cases we observed that these agonists could inhibited the KCl-induced release of glutamate. However, the selective and specific co-activation of both Y1 and Y2 or Y2 and Y5 receptors resulted in non-additive inhibition, and this effect was prevented in the presence of the Y2 antagonist, but was insensitive to the Y1 or Y5 receptor antagonist. Moreover, as we previously showed for Y1 receptors, we also observed that the activation of Y5 receptors inhibited the glutamate release in the dentate gyrus and CA3 subregion, without significant effect in the CA1 subregion of the hippocampus. The same qualitative results were obtained when we investigated the role of NPY Y1 and Y2 receptors in modulating the changes in [Ca(2+)](i) due to KCl depolarisation in cultured hippocampal neurons. The inhibitory effect of nitrendipine (L-type VGCC blocker) or omega-conotoxin GVIA (omega-CgTx; N-type VGCC blocker) was not potentiated by the simultaneous activation of Y1 or Y2 receptors. Moreover, the exocytotic release of glutamate was inhibited by omega-agatoxin IVA (omega-Aga; P-/Q-type VGCC blocker), and this VGCC blocker did not potentiate Y1, Y2 or Y5 receptor-mediated inhibition of glutamate release. Also, the effect of ionomycin in inducing the exocytotic release of glutamate from hippocampal synaptosomes was insensitive to the activation of NPY receptors. In the present paper, we identified a role for NPY Y1, Y2 and Y5 receptors in modulating the exocytotic release of glutamate and the [Ca(2+)](i) changes in the rat hippocampus. In conditions of co-activation, there appears to exist a physiological cross-talk between Y1 and Y2 and also between Y2 and Y5 receptors, in which Y2 receptors play a predominant role. Moreover, we also show that Y1 and Y2 receptors exert their inhibitory action by directly modulating L-, N-, and P-/Q-type VGCCs, whereas the inhibition of glutamate release mediated by the Y5 receptors seems to involve P-/Q-type VGCCs.


Subject(s)
Arginine/analogs & derivatives , Calcium Channels/physiology , Hippocampus/physiology , Neurons/physiology , Neuropeptide Y/pharmacology , Receptor Cross-Talk , Receptors, Neuropeptide Y/physiology , Animals , Arginine/pharmacology , Benzazepines/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Cells, Cultured , Cyclohexanes/pharmacology , Drug Interactions , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Ionomycin/pharmacology , Ionophores/pharmacology , Neurons/drug effects , Neuropeptide Y/agonists , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/classification , Peptide Fragments/classification , Peptide Fragments/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Wistar/embryology , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/classification , Receptors, Neuropeptide Y/drug effects , Synaptosomes/drug effects , Xanthenes/pharmacology , omega-Agatoxin IVA/pharmacology , omega-Conotoxin GVIA/pharmacology
12.
Int J Toxicol ; 22(1): 43-8, 2003.
Article in English | MEDLINE | ID: mdl-12573948

ABSTRACT

In this work, a dendritic cell line derived from mouse skin (FSDC) was used, as an in vitro experimental model, to evaluate the cytotoxic effect of two chemical sensitizers, a strong sensitizer (2,4-dinitrofluorobenzene, DNFB) and a weak sensitizer (2,4-dichloronitrobenzene, DCNB). The results indicated that DNFB reduces the cellular metabolism of FSDC, as evaluated by the reduction of the tetrazolium salt, 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT). All the DNFB concentrations tested, ranging from 5.2 micro M to 26 micro M, significantly inhibited the MTT reduction after 1 hour of cell exposure to the sensitizer. In contrast, incubation of FSDC with the weak sensitizer DCNB had no significant effect on the MTT reduction assay. When the cells were incubated with DNFB (13 micro M), for 3 and 6 hours, morphological changes characteristics of cell death by apoptosis were observed, as assessed by propidium iodide (PI) DNA staining and annexin-V externalization analysis. These results correlate well with an increase of caspase-3-like activity after FSDC exposure to DNFB (13 micro M) for 6 hours. Together, these results indicate that apoptotic death of skin dendritic cells occurs after exposure to the sensitizer DNFB, although necrotic cell death was also observed when the cells were incubated with high concentrations of DNFB (26 micro M), or after long periods of cell exposure to the chemical DNFB (13 micro M, for 6 hours).


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Dendritic Cells/drug effects , Dinitrofluorobenzene/toxicity , Animals , Caspase 3 , Cell Death/drug effects , Cell Line , Dendritic Cells/cytology , Dendritic Cells/enzymology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Mice , Microscopy, Fluorescence , Skin/cytology , Skin/drug effects , Skin/enzymology , Time Factors
13.
Neurochem Int ; 42(2): 179-88, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12421598

ABSTRACT

The role of protein tyrosine kinases on glutamate release was investigated by determining the effect of broad range inhibitors of tyrosine kinases on the release of glutamate from rat hippocampal synaptosomes. We found that lavendustin A and herbimycin A did not inhibit glutamate release stimulated by 15 mM KCl, but genistein, also a broad range inhibitor of tyrosine kinases did inhibit the intracellular Ca(2+) concentration response to KCl and, concomitantly, decreased glutamate release evoked by the same stimulus, in a dose-dependent manner. These effects were not observed with the inactive analogue genistin. Therefore, we investigated the mechanism whereby genistein modulates Ca(2+) influx and glutamate release. Studies with voltage-gated Ca(2+) channel inhibitors showed that omega-conotoxin GVIA did not further inhibit glutamate release or the Ca(2+) influx stimulated by KCl in the presence of genistein. This tyrosine kinase inhibitor and omega-agatoxin IVA had a partially additive effect on those events. Nitrendipine did not reduce significantly the KCl-induced responses. Genistein further reduced Ca(2+) influx in response to KCl in the presence of nitrendipine, omega-conotoxin GVIA and omega-agatoxin IVA, simultaneously. The effect of tyrosine phosphatase inhibitors was also tested on the influx of Ca(2+) and on glutamate release stimulated by KCl-depolarization. We found that the broad range inhibitors sodium orthovanadate and dephostatin did not significantly affect these KCl-evoked events. Our results suggest that genistein inhibits glutamate release and Ca(2+) influx in response to KCl independently of tyrosine kinase inhibition, and that tyrosine kinases and phosphatases are not key regulators of glutamate release in hippocampal nerve terminals.


Subject(s)
Calcium/metabolism , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Glutamic Acid/metabolism , Hippocampus/metabolism , Synaptosomes/metabolism , Animals , Calcium Channel Blockers/pharmacology , Hippocampus/drug effects , Immunoblotting , In Vitro Techniques , Male , Nerve Endings/drug effects , Nerve Endings/enzymology , Phosphorylation/drug effects , Potassium Chloride/pharmacology , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Rats, Wistar , Synaptosomes/drug effects , omega-Agatoxin IVA/pharmacology
14.
Photochem Photobiol ; 76(4): 423-30, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12405151

ABSTRACT

Photodynamic therapy (PDT) of cancer is a very promising technique based on the formation of singlet oxygen induced by a sensitizer after irradiation with visible light. The stimulation of tumor growth by nitric oxide (NO) was reported recently, and NO was shown to have a protective effect against PDT-induced tumor death. We investigated a putative direct effect of NO on tumor cell death induced by PDT, using the human lymphoblastoid CCRF-CEM cells and bisulfonated aluminum phthalocyanine (AlPcS2) as a sensitizer. Cells were incubated with AlPcS2 in the presence or absence of NO donors ((Z)-1-[(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate, hydroxylamine and S-nitroso-N-acetylpenicillamine) or L-arginine. Under these conditions, in the absence of NO donors or L-arginine the cells died rapidly by apoptosis upon photosensitization. In the presence of NO donors or L-arginine, apoptotic cell death after photosensitization was significantly decreased. Modulation of cell death by NO was not due to S-nitrosylation of caspases and occurred at the level or upstream of caspase-9 processing. The protective effect of NO was reversed by incubating the cells with 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, an inhibitor of guanylyl cyclase, or with KT5823, an inhibitor of protein kinase G (PKG). Incubation with 8-bromo-cyclic guanosine monophosphate, a membrane permeable cyclic guanosine monophosphate analog, also decreased cell death induced by PDT. Although the protective effect of NO against apoptotic cell death in several models has been attributed to an increase in the expression of heme oxygenase-1, heat shock protein 70 or Bcl-2, this was not the case under our experimental conditions. These results show that NO decreases the extent of apoptotic cell death after PDT treatment through a PKG-dependent mechanism, upstream or at the level of caspase activation.


Subject(s)
Apoptosis/drug effects , Cyclic GMP/physiology , Nitric Oxide/physiology , Photochemotherapy , Apoptosis/physiology , Humans , Nitric Oxide Donors/pharmacology , Tumor Cells, Cultured
15.
Neurochem Res ; 27(1-2): 121-30, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11926264

ABSTRACT

Carbamazepine (CBZ) has been extensively used in the treatment of epilepsy, as well as in the treatment of neuropathic pain and affective disorders. However, the mechanisms of action of this drug are not completely elucidated and are still a matter of debate. Since CBZ is not very effective in some epileptic patients and may cause several adverse effects, several antiepileptic drugs have been developed by structural variation of CBZ, such as oxcarbazepine (OXC), which is used in the treatment of epilepsy since 1990. (S)-(-)-10-acetoxy-10,11-dihydro-5H-dibenz [b,f]azepine-5-carboxamide (BIA 2-093) and 10,11-dihydro-10-hydroxyimino-5H-dibenz[b,f] azepine-5-carboxamide (BIA 2-024), which were recently developed by BIAL, are new putative antiepileptic drugs, with some improved properties. In this review, we will focus on the mechanisms of action of CBZ and its derivatives, OXC, BIA 2-093 and BIA 2-024. The available data indicate that the anticonvulsant efficacy of these AEDs is mainly due to the inhibition of sodium channel activity.


Subject(s)
Anticonvulsants/therapeutic use , Carbamazepine/analogs & derivatives , Carbamazepine/therapeutic use , Dibenzazepines/therapeutic use , Epilepsy/drug therapy , Animals , Anticonvulsants/chemistry , Carbamazepine/chemistry , Humans , Oxcarbazepine , Sodium Channel Blockers/therapeutic use
16.
Eur J Neurosci ; 15(3): 465-74, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11876774

ABSTRACT

We have previously reported that the activity of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptors is potentiated by protein kinase C (PKC) in cultured chick retina amacrine neurons, and that constitutive PKC activity is necessary for basal AMPA receptor activity (Carvalho et al., 1998). In this study, we evaluated the phosphorylation of the GluR4 subunit, which is very abundant in cultured amacrine neurons, to correlate it with the effects of PKC on AMPA receptor activity in these cells. 32P-labelling of GluR4 increased upon AMPA receptor stimulation or cell treatment with phorbol 12-myristate 13-acetate (PMA) before stimulating with kainate. By contrast, phosphorylation of GluR4 was not changed when PKC was inhibited by treating the cells with the selective PKC inhibitor GF 109203X before stimulation with kainate. We conclude that GluR4 is phosphorylated upon PKC activation and/or stimulation of AMPA receptors in cultured amacrine cells. Additionally, AMPA receptor activation with kainate in cultured chick amacrine cells leads to translocation of conventional and novel PKC isoforms to the cell membrane, suggesting that PKC could be activated upon AMPA receptor stimulation in these cells.


Subject(s)
Amacrine Cells/enzymology , Glutamic Acid/metabolism , Protein Kinase C/metabolism , Receptors, AMPA/metabolism , Synapses/enzymology , Synaptic Transmission/physiology , Amacrine Cells/cytology , Amacrine Cells/drug effects , Animals , Cells, Cultured , Chick Embryo , Enzyme Inhibitors/pharmacology , Kainic Acid/pharmacology , Phosphorylation , Protein Isoforms/metabolism , Protein Kinase C/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Receptors, AMPA/drug effects , Receptors, Kainic Acid/metabolism , Synapses/drug effects , Synaptic Membranes/drug effects , Synaptic Membranes/metabolism , Synaptic Transmission/drug effects
17.
Arch Dermatol Res ; 294(9): 419-25, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12522580

ABSTRACT

Dendritic cell (DC) maturation is essential for the initiation of T-dependent immune responses. Nuclear factor kappa B (NF-kappaB) transcription factors are ubiquitously expressed signalling molecules, known to regulate the transcription of a large number of genes involved in immune responses, including cytokines and cell surface molecules. In this work, we studied the time-dependent activation of five members of the NF-kappaB family, p50, p52, p65, RelB and cRel, in a mouse skin DC line in response to stimulation with the strong sensitizer, 2,4-dinitrofluorobenzene (DNFB). Western blot assay revealed that exposure of fetal skin DC (FSDC) to DNFB induced the degradation of the inhibitor of NF-kappaB (IkappaB). Three out of its five members, i.e. p50, p52, and RelB, were similarly activated upon DNFB stimulation, with subsequent translocation of these subunits from the cytosol to the nucleus, but with different kinetics. In contrast, p65 expression was diminished in both the nucleus and the cytosol. The electrophoretic mobility shift assay (EMSA) showed that exposure of FSDC to DNFB induced DNA binding to NF-kappaB. Together, these results show that DNFB differentially activates the various members of the NF-kappaB family in skin DC.


Subject(s)
Dendritic Cells/metabolism , Dinitrofluorobenzene/pharmacology , NF-kappa B/drug effects , NF-kappa B/physiology , Skin/metabolism , Animals , Biological Transport , Cell Line , Cell Nucleus/metabolism , Cytosol/metabolism , DNA/metabolism , Fetus , I-kappa B Proteins/metabolism , Mice , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , NF-kappa B p50 Subunit , Protein Isoforms/drug effects , Protein Isoforms/physiology , Proto-Oncogene Proteins/metabolism , Skin/cytology , Transcription Factor RelA , Transcription Factor RelB , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...