Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Neuroscience ; 118(3): 845-52, 2003.
Article in English | MEDLINE | ID: mdl-12710991

ABSTRACT

Recent experimental evidence indicates that neurotrophic factors play a role in the pathophysiology of epilepsy. The objective of this study was to explore whether signaling through one of the glial cell line-derived neurotrophic factor family receptors, GFRalpha2, influences the severity of kindling-evoked, rapidly recurring seizures and the subsequent development of permanent hyperexcitability. We applied the rapid kindling model to adult mice, using 40 threshold stimulations delivered with 5-min interval in the ventral hippocampus. Generalized seizures were fewer and developed later in response to kindling stimulations in mice lacking GFRalpha2. However, GFRalpha2 gene deletion did not influence the acquisition of the permanent abnormal excitability as assessed 4 weeks later. In situ hybridization revealed marked and dynamic changes of GFRalpha2 mRNA levels in several forebrain areas following the stimulus-evoked seizures. Our findings provide evidence that signaling through the GFRalpha2 receptor contributes to seizure generalization in rapid kindling.


Subject(s)
Brain/metabolism , Epilepsy/genetics , Kindling, Neurologic/genetics , Nerve Growth Factors/metabolism , Proto-Oncogene Proteins/deficiency , Receptor Protein-Tyrosine Kinases/deficiency , Animals , Brain/physiopathology , Epilepsy/metabolism , Epilepsy/physiopathology , Gene Deletion , Gene Expression Regulation/genetics , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Kindling, Neurologic/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics
2.
Neurosci Lett ; 333(2): 127-30, 2002 Nov 22.
Article in English | MEDLINE | ID: mdl-12419497

ABSTRACT

Previous studies show that neuropeptide Y (NPY) inhibits in vitro seizures in rodent hippocampus. Here, we explored the effect of NPY application on epileptiform discharges induced by perfusion with magnesium-free solution in slices of entorhinal cortex from two different mouse strains. NPY significantly reduced the duration of epileptiform discharges with a peak effect of 36-50%. This is the first study showing anti-epileptiform effect of NPY in the entorhinal cortex and also the first evidence that NPY inhibits seizures in a cortical region in mice. The entorhinal cortex has a central role in transferring information between the hippocampus and the rest of the brain. Therefore our data further strengthen the concept of NPY and its receptors as widespread regulators of epileptiform activity and as a potential future target for antiepileptic therapy.


Subject(s)
Action Potentials/drug effects , Anticonvulsants/pharmacology , Entorhinal Cortex/physiology , Neuropeptide Y/pharmacology , Animals , Culture Media , Epilepsy/chemically induced , Magnesium Deficiency , Male , Mice , Organ Culture Techniques , Patch-Clamp Techniques
3.
Proc Natl Acad Sci U S A ; 98(24): 14006-11, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11698649

ABSTRACT

The neuropeptide galanin has been shown to suppress epileptic seizures. In cortical and hippocampal areas, galanin is normally mainly expressed in noradrenergic afferents. We have generated a mouse overexpressing galanin in neurons under the platelet-derived growth factor B promoter. RIA and HPLC analysis revealed up to 8-fold higher levels of galanin in transgenic as compared with wild-type mice. Ectopic galanin overexpression was detected especially in dentate granule cells and hippocampal and cortical pyramidal neurons. Galanin-overexpressing mice showed retardation of seizure generalization during hippocampal kindling, a model for human complex partial epilepsy. The high levels of galanin in mossy fibers found in the transgenic mice were further increased after seizures. Frequency facilitation of field excitatory postsynaptic potentials, a form of short-term synaptic plasticity assessed in hippocampal slices, was reduced in mossy fiber-CA3 cell synapses of galanin-overexpressing mice, indicating suppressed glutamate release. This effect was reversed by application of the putative galanin receptor antagonist M35. These data provide evidence that ectopically overexpressed galanin can be released and dampen the development of epilepsy by means of receptor-mediated action, at least partly by reducing glutamate release from mossy fibers.


Subject(s)
Epilepsy/metabolism , Galanin/biosynthesis , Kindling, Neurologic/metabolism , Animals , Cerebral Cortex/metabolism , Choristoma/metabolism , Epilepsy/prevention & control , Female , Galanin/genetics , Galanin/physiology , Gene Expression , Hippocampus/metabolism , Kindling, Neurologic/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , RNA, Messenger/metabolism
4.
Neuroscience ; 102(4): 819-32, 2001.
Article in English | MEDLINE | ID: mdl-11182245

ABSTRACT

Widespread lesions of forebrain cholinergic or noradrenergic projections by intraventricular administration of 192 IgG-saporin or 6-hydroxydopamine, respectively, accelerate kindling epileptogenesis. Here we demonstrate both quantitative and qualitative differences between the two lesions in their effects on hippocampal kindling in rats. Epileptogenesis was significantly faster after noradrenergic as compared to cholinergic denervation, and when both lesions were combined, kindling development resembled that in animals with 6-hydroxydopamine lesion alone. Furthermore, whereas the 192 IgG-saporin lesion promoted the development only of the early stages of kindling, administration of 6-hydroxydopamine or both neurotoxins accelerated the late stages also. To investigate the contribution of different subparts of the basal forebrain cholinergic system to its seizure-suppressant action in hippocampal kindling, 192 IgG-saporin was injected into medial septum/vertical limb of the diagonal band of Broca or nucleus basalis magnocellularis, leading to selective hippocampal or cortical cholinergic deafferentation, respectively. The denervation of the hippocampus facilitated kindling similar to the extensive lesion caused by intraventricular 192 IgG-saporin, whereas the cortical lesion had no effect. These results indicate that although both noradrenergic and cholinergic projections to the forebrain exert powerful inhibitory effects on hippocampal kindling epileptogenesis, the action of the cholinergic system is less pronounced and occurs specifically prior to seizure generalization. In contrast, noradrenergic neurons inhibit the development of both focal and generalized seizures. The septo-hippocampal neurons are responsible for the antiepileptogenic effect of the cholinergic system in hippocampal kindling, whereas the cortical projection is not significantly involved. Conversely, we have previously shown [Ferencz I. et al. (2000) Eur. J. Neurosci., 12, 2107-2116] that seizure-suppression in amygdala kindling is exerted through the cortical and not the hippocampal cholinergic projection. This shows that, depending on the location of the primary epileptic focus, i.e. the site of stimulation, basal forebrain cholinergic neurons operate through different subsystems to counteract seizure development in kindling.


Subject(s)
Basal Nucleus of Meynert/physiology , Cholinergic Fibers/enzymology , Diagonal Band of Broca/physiology , Hippocampus/physiology , Kindling, Neurologic/physiology , Norepinephrine/physiology , Acetylcholine/physiology , Acetylcholinesterase/analysis , Animals , Antibodies, Monoclonal/pharmacology , Basal Nucleus of Meynert/cytology , Choline O-Acetyltransferase/analysis , Cholinergic Agents/pharmacology , Diagonal Band of Broca/cytology , Epilepsy/chemically induced , Epilepsy/physiopathology , Hippocampus/cytology , Immunotoxins/pharmacology , Injections, Intraventricular , Kindling, Neurologic/drug effects , Male , N-Glycosyl Hydrolases , Neurons/physiology , Neurons/ultrastructure , Oxidopamine/pharmacology , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins , Sympatholytics/pharmacology
5.
Proc Natl Acad Sci U S A ; 97(22): 12312-7, 2000 Oct 24.
Article in English | MEDLINE | ID: mdl-11050250

ABSTRACT

Seizure activity regulates gene expression for glial cell line-derived neurotrophic factor (GDNF) and neurturin (NRTN), and their receptor components, the transmembrane c-Ret tyrosine kinase and the glycosylphosphatidylinositol-anchored GDNF family receptor (GFR) alpha 1 and alpha 2 in limbic structures. We demonstrate here that epileptogenesis, as assessed in the hippocampal kindling model, is markedly suppressed in mice lacking GFR alpha 2. Moreover, at 6 to 8 wk after having reached the epileptic state, the hyperexcitability is lower in GFR alpha 2 knock-out mice as compared with wild-type mice. These results provide evidence that signaling through GFR alpha 2 is involved in mechanisms regulating the development and persistence of kindling epilepsy. Our data suggest that GDNF and NRTN may modulate seizure susceptibility by altering the function of hilar neuropeptide Y-containing interneurons and entorhinal cortical afferents at dentate granule cell synapses.


Subject(s)
Drosophila Proteins , Epilepsy/genetics , Kindling, Neurologic/genetics , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Glial Cell Line-Derived Neurotrophic Factor Receptors , Hippocampus/physiopathology , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics
6.
Eur J Neurosci ; 12(6): 2107-16, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10886350

ABSTRACT

Intraventricular administration of the immunotoxin 192 IgG-saporin in rats has been shown to cause a selective loss of cholinergic afferents to the hippocampus and cortical areas, and to facilitate seizure development in hippocampal kindling. Here we demonstrate that this lesion also accelerates seizure progression when kindling is induced by electrical stimulations in the amygdala. However, whereas intraventricular 192 IgG-saporin facilitated the development of the initial stages of hippocampal kindling, the same lesion promoted the late stages of amygdala kindling. To explore the role of various parts of the basal forebrain cholinergic system in amygdala kindling, selective lesions of the cholinergic projections to either hippocampus or cortex were produced by intraparenchymal injections of 192 IgG-saporin into medial septum/vertical limb of the diagonal band or nucleus basalis, respectively. Cholinergic denervation of the cortical regions caused acceleration of amygdala kindling closely resembling that observed after the more widespread lesion induced by intraventricular 192 IgG-saporin. In contrast, removal of the cholinergic input to the hippocampus had no effect on the development of amygdala kindling. These data indicate that basal forebrain cholinergic neurons suppress kindling elicited from amygdala, and that this dampening effect is mediated via cortical but not hippocampal projections.


Subject(s)
Amygdala/cytology , Amygdala/physiology , Basal Nucleus of Meynert/cytology , Hippocampus/cytology , Kindling, Neurologic/physiology , Animals , Antibodies, Monoclonal/pharmacology , Cholinergic Agents/pharmacology , Cholinergic Fibers/physiology , Epilepsy/physiopathology , Immunotoxins/pharmacology , Injections, Intraventricular , Male , N-Glycosyl Hydrolases , Neural Pathways , Neurons/physiology , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins , Septal Nuclei/cytology
7.
Eur J Neurosci ; 12(5): 1687-95, 2000 May.
Article in English | MEDLINE | ID: mdl-10792446

ABSTRACT

Loss of function mutations in the gene encoding the cysteine protease inhibitor, cystatin B (CSTB), are responsible for the primary defect in human progressive myoclonus epilepsy (EPM1). CSTB inhibits the cathepsins B, H, L and S by tight reversible binding, but little is known regarding its localization and physiological function in the brain and the relation between the depletion of the CSTB protein and the clinical symptoms in EPM1. We have analysed the expression of mRNA and protein for CSTB in the adult rat brain using in situ hybridization and immunocytochemistry. In the control brains, the CSTB gene was differentially expressed with the highest levels in the hippocampal formation and reticular thalamic nucleus, and moderate levels in amygdala, thalamus, hypothalamus and cortical areas. Detectable levels of CSTB were found in virtually all forebrain neurons but not in glial cells. Following 40 rapidly recurring seizures evoked by hippocampal kindling stimulations, CSTB mRNA expression showed marked bilateral increases in the dentate granule cell layer, CA1 and CA4 pyramidal layers, amygdala, and piriform and parietal cortices. Maximum levels were detected at 6 or 24 h, and expression had reached control values at 1 week post-seizures. The changes of mRNA expression were accompanied by transient elevations (at 6-24 h) of CSTB protein in the same brain areas. These findings demonstrate that seizure activity leads to rapid and widespread increases of the synthesis of CSTB in forebrain neurons. We propose that the upregulation of CSTB following seizures may counteract apoptosis by binding cysteine proteases.


Subject(s)
Brain/metabolism , Cystatins/genetics , Epilepsies, Myoclonic/genetics , Gene Expression Regulation , Hippocampus/metabolism , Neurons/metabolism , Seizures/genetics , Transcription, Genetic , Animals , Cathepsins/antagonists & inhibitors , Cystatin B , Cystatins/analysis , Cystatins/metabolism , Cysteine Proteinase Inhibitors/genetics , Dentate Gyrus/metabolism , Functional Laterality , Humans , Kindling, Neurologic , Male , Organ Specificity , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Seizures/metabolism
8.
J Cereb Blood Flow Metab ; 19(11): 1220-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10566968

ABSTRACT

The levels of brain-derived neurotrophic factor (BDNF) vary between different forebrain areas and show region-specific changes after cerebral ischemia. The present study explores the possibility that the levels of endogenous BDNF determine the susceptibility to ischemic neuronal death. To block BDNF activity the authors used the TrkB-Fc fusion protein, which was infused intraventricularly in rats during 1 week before and 1 week after 5 or 30 minutes of global forebrain ischemia. Ischemic damage was quantified in the striatum and hippocampal formation after 1 week of reperfusion using immunocytochemistry and stereological procedures. After the 30-minute insult, there was a significantly lower number of surviving CA4 pyramidal neurons, neuropeptide Y-immunoreactive dentate hilar neurons, and choline acetyltransferase- and TrkA-positive, cholinergic striatal interneurons in the TrkB-Fc-infused rats as compared to controls. In contrast, the TrkB-Fc treatment did not influence survival of CA1 or CA3 pyramidal neurons or striatal projection neurons. Also, after the mild ischemic insult (5 minutes), neuronal death in the CA1 region was similar in the TrkB-Fc-treated and control groups. These results indicate that endogenous BDNF can protect certain neuronal populations against ischemic damage. It is conceivable, though, that efficient neuroprotection after brain insults is dependent not only on this factor but on the concerted action of a large number of neurotrophic molecules.


Subject(s)
Brain Ischemia/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Prosencephalon/blood supply , Animals , Brain Ischemia/pathology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Cell Death , Male , Rats , Rats, Wistar , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Recombinant Fusion Proteins/pharmacology
9.
Brain Res Mol Brain Res ; 72(1): 17-29, 1999 Sep 08.
Article in English | MEDLINE | ID: mdl-10521595

ABSTRACT

Expression of mRNAs for neuropeptide Y (NPY) and its receptor subtypes Y1 (Y1-R), Y2 (Y2-R) and Y5 (Y5-R) was studied in adult rat brain using in situ hybridization after 40 rapidly recurring seizures induced with 5-min interval by hippocampal kindling stimulations. At 2-4 h post-seizure, NPY mRNA levels were markedly elevated in dentate granule cells, CA1 and CA3 pyramidal layers, amygdala and piriform and entorhinal cortices. Gene expression had returned to control level in the dentate granule cell layer at 48 h but remained high in the other areas, reaching baseline at 1 week. Transient decreases of Y1-R mRNA levels were detected at 2-4 h in hippocampal subregions, amygdala, piriform, entorhinal and somatosensory cortices. The Y2-R mRNA levels were reduced at 2-4 h in the CA3 region and piriform cortex, but exhibited marked increases at 48 h and 1 week post-seizure in the dentate gyrus, amygdala and piriform and entorhinal cortices. At 3 weeks, Y2-R mRNA expression had virtually returned to baseline. Elevated Y5-R mRNA levels were only detected at 2-4 h and confined to dentate granule cell layer and piriform and entorhinal cortices. These results demonstrate a cell- and region-specific, differential regulation of mRNA expression for NPY and Y1-R, Y2-R, and Y5-R in the limbic system following recurring seizures. Because the gene changes were transient, it seems unlikely that the presumed alterations of the corresponding proteins are involved in the maintenance of the epileptic syndrome, which develops up to 4 weeks post-seizure in the present model and is stable thereafter. Our data provide further support for the hypothesis that the changes of NPY and its receptors act to dampen seizure susceptibility, and suggest that the cascade of gene changes is orchestrated to optimize this anticonvulsant effect.


Subject(s)
Gene Expression Regulation , Kindling, Neurologic/genetics , Limbic System/metabolism , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , RNA, Messenger/biosynthesis , Receptors, Neuropeptide Y/genetics , Amygdala/metabolism , Animals , Dentate Gyrus/metabolism , Electric Stimulation , Entorhinal Cortex/metabolism , Hippocampus/physiopathology , In Situ Hybridization , Male , Nerve Tissue Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/metabolism , Somatosensory Cortex/metabolism
10.
Eur J Neurosci ; 11(4): 1202-16, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10103116

ABSTRACT

Expression of mRNAs for glial cell line-derived neurotrophic factor (GDNF), neurturin (NTN) and their receptors was studied in adult rat brain using in situ hybridization after 40 kindling-evoked, rapidly recurring seizures or 10 min of global forebrain ischaemia. Following seizures, GDNF and NTN mRNAs were elevated in dentate granule cells, and c-Ret mRNA in hilar neurons and non-pyramidal cells in CA1 and CA3 regions. GFRalpha-1 mRNA levels showed more widespread increases in the dentate granule cell layer and hilus, CA1 and CA3 pyramidal layers, basolateral amygdala and parietal cortex. The expression of GFRalpha-2 mRNA increased in the piriform cortex and decreased in the CA1 region and basolateral amygdala. Forebrain ischaemia induced elevated expression of GDNF mRNA in dentate granule cells, GFRalpha-1 mRNA in the dentate granule cell layer, hilus and CA3 pyramidal layer, and GFRalpha-2 mRNA in the parietal cortex. The gene expression patterns observed here suggest that GDNF and NTN may act as target-derived factors, but also in an autocrine or paracrine manner. GFRalpha-1 can be coexpressed with GFRalpha-2 and c-Ret mRNAs in the same hippocampal or thalamic neurons, but other neurons contain GFRalpha-1 alone or together with c-Ret mRNA. The gene expression changes for the ligands, and the receptor components are region-, cell- and insult-specific, and occur independently of each other, mainly within 24 h after seizures or ischaemia. This dynamic regulation of GDNF and NTN circuits primarily at the receptor level may be important for the effectiveness of neuroprotective responses but could also trigger plastic changes, e.g. those underlying the development of epileptic syndromes.


Subject(s)
Drosophila Proteins , Ischemic Attack, Transient/physiopathology , Multigene Family , Nerve Growth Factors , Nerve Tissue Proteins/genetics , Animals , Gene Expression Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , In Situ Hybridization , Male , Prosencephalon/blood supply , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Seizures/physiopathology
11.
Proc Natl Acad Sci U S A ; 94(19): 10432-7, 1997 Sep 16.
Article in English | MEDLINE | ID: mdl-9294228

ABSTRACT

Neuronal apoptosis was observed in the rat dentate gyrus in two experimental models of human limbic epilepsy. Five hours after one hippocampal kindling stimulation, a marked increase of in situ terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling (TUNEL) of fragmented DNA was observed in nuclei located within and on the hilar border of the granule cell layer and in the polymorphic region. Forty kindling stimulations with 5-min interval produced higher numbers of labeled nuclei compared with one stimulation. The increase of TUNEL-positive nuclei was prevented by the protein synthesis inhibitor cycloheximide but not affected by the N-methyl-D-aspartate receptor antagonist MK-801. Kainic acid-induced seizures lead to a pattern of labeling in the hippocampal formation identical to that evoked by kindling. A large proportion of cells displaying TUNEL-positive nuclei was double-labeled by the neuron-specific antigen NeuN, demonstrating the neuronal identity of apoptotic cells. Either 1 or 40 kindling stimulations also gave rise to a marked increase of the number of cells double-labeled with the mitotic marker bromodeoxyuridine and NeuN in the subgranular zone and on the hilar border of the dentate granule cell layer. The present data show that single and intermittent, brief seizures induce both apoptotic death and proliferation of dentate gyrus neurons. We hypothesize that these processes, occurring early during epileptogenesis, are primary events in the development of hippocampal pathology in animals and possibly also in patients suffering from temporal lobe epilepsy.


Subject(s)
Dentate Gyrus/pathology , Epilepsy/pathology , Limbic System/pathology , Neurons/pathology , Animals , Apoptosis , Cell Division , Dentate Gyrus/physiopathology , Epilepsy/physiopathology , Limbic System/physiopathology , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...