Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Cereb Cortex ; 33(10): 6171-6183, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36611229

ABSTRACT

During epileptic seizures, neuronal network activity is hyper synchronized whereby GABAergic parvalbumin-interneurons may have a key role. Previous studies have mostly utilized 4-aminopyridine to induce epileptiform discharges in brain slices from healthy animals. However, it is not clear if the seizure-triggering ability of parvalbumin-interneurons also holds true without the use of external convulsive agents. Here, we investigate whether synchronized activation of parvalbumin-interneurons or principal cells can elicit epileptiform discharges in subiculum slices of epileptic mice. We found that selective synchronized activation of parvalbumin-interneurons or principal cells with optogenetics do not result in light-induced epileptiform discharges (LIEDs) neither in epileptic nor in normal brain slices. Adding 4-aminopyridine to slices, activation of parvalbumin-interneurons still failed to trigger LIEDs. In contrast, such activation of principal neurons readily generated LIEDs with features resembling afterdischarges. When GABAA receptor blocker was added to the perfusion medium, the LIEDs were abolished. These results demonstrate that in subiculum, selective synchronized activation of principal excitatory neurons can trigger epileptiform discharges by recruiting a large pool of downstream interneurons. This study also suggests region-specific role of principal neurons and interneurons in ictogenesis, opening towards differential targeting of specific brain areas for future treatment strategies tailored for individual patients with epilepsy.


Subject(s)
Epilepsy , Parvalbumins , Mice , Animals , Parvalbumins/metabolism , Limbic System , Seizures , Interneurons/physiology , Hippocampus/metabolism , 4-Aminopyridine/pharmacology
2.
Sci Rep ; 8(1): 4158, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29515159

ABSTRACT

Resected hippocampal tissue from patients with drug-resistant epilepsy presents a unique possibility to test novel treatment strategies directly in target tissue. The post-resection time for testing and analysis however is normally limited. Acute tissue slices allow for electrophysiological recordings typically up to 12 hours. To enable longer time to test novel treatment strategies such as, e.g., gene-therapy, we developed a method for keeping acute human brain slices viable over a longer period. Our protocol keeps neurons viable well up to 48 hours. Using a dual-flow chamber, which allows for microscopic visualisation of individual neurons with a submerged objective for whole-cell patch-clamp recordings, we report stable electrophysiological properties, such as action potential amplitude and threshold during this time. We also demonstrate that epileptiform activity, monitored by individual dentate granule whole-cell recordings, can be consistently induced in these slices, underlying the usefulness of this methodology for testing and/or validating novel treatment strategies for epilepsy.


Subject(s)
Epilepsy, Temporal Lobe/physiopathology , Hippocampus/physiopathology , Neurons , Adolescent , Adult , Child , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Middle Aged , Patch-Clamp Techniques
3.
Gene Ther ; 23(10): 760-766, 2016 10.
Article in English | MEDLINE | ID: mdl-27416078

ABSTRACT

Epilepsy is a neurological disorder with a prevalence of ≈1% of general population. Available antiepileptic drugs (AEDs) have multiple side effects and are ineffective in 30% of patients. Therefore, development of effective treatment strategies is highly needed, requiring drug-screening models that are relevant and reliable. We investigated novel chemogenetic approach, using DREADDs (designer receptors exclusively activated by designer drugs) as possible inhibitor of epileptiform activity in organotypic hippocampal slice cultures (OHSCs). The OHSCs are characterized by increased overall excitability and closely resemble features of human epileptic tissue. Studies suggest that chemically induced epileptiform activity in rat OHSCs is pharmacoresistant to most of AEDs. However, high-frequency electric stimulus train-induced bursting (STIB) in OHSCs is responsive to carbamazepine and phenytoin. We investigated whether inhibitory DREADD, hM4Di, would be effective in suppressing STIB in OHSC. hM4Di is a mutated muscarinic receptor selectively activated by otherwise inert clozapine-N-oxide, which leads to hyperpolarization in neurons. We demonstrated that this hyperpolarization effectively suppresses STIB in mouse OHSCs. As we also found that STIB in mouse OHSCs is resistant to common AED, valproic acid, collectively our findings suggest that DREADD-based strategy may be effective in suppressing epileptiform activity in a pharamcoresitant epileptic brain tissue.


Subject(s)
Action Potentials , Anticonvulsants/pharmacology , Clozapine/pharmacology , Drug Resistant Epilepsy/genetics , Muscarinic Agonists/pharmacology , Neurons/metabolism , Receptors, Muscarinic/metabolism , Animals , Drug Resistant Epilepsy/metabolism , Hippocampus/cytology , Mice , Mice, Inbred BALB C , Mutation , Neurons/drug effects , Neurons/physiology , Receptors, Muscarinic/genetics
4.
Neuropeptides ; 46(2): 71-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22342800

ABSTRACT

Neuropeptide Y (NPY) has been implicated in anxiolytic- and antidepressant-like behaviour as well as seizure-suppressant effects in rodents. Although these effects appear to be predominantly mediated via other NPY receptors (Y1 and/or Y2), several studies have also indicated a role for Y5 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY, Y1 or Y2 receptors in the hippocampus or amygdala has previously been shown to modulate emotional behaviour and seizures in rodents. The present study explored the potential effects of gene therapy with the Y5 receptor, by testing effects of recombinant adeno-associated viral vector (rAAV) encoding Y5 (rAAV-Y5) in anxiety- and depression-like behaviour as well as in kainate-induced seizures in adult mice. The rAAV-Y5 vector injected into the hippocampus and amygdala induced a pronounced and sustained increase in Y5 receptor mRNA expression and functional Y5 receptor binding, but no significant effects were found with regard to anxiety- and depression-like behaviours or seizure susceptibility. Instead, rAAV-mediated Y5 receptor transgene overexpression resulted in moderate hyperactivity in the open field test. These results do not support a potential role for single transgene overexpression of Y5 receptors for modulating anxiety-/depression-like behaviours or seizures in adult mice. Whether the induction of hyperactivity by rAAV-Y5 could be relevant for other conditions remains to be studied.


Subject(s)
Anxiety/physiopathology , Depression/physiopathology , Hyperkinesis/metabolism , Receptors, Neuropeptide Y/metabolism , Seizures/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/physiology , Genetic Therapy , Genetic Vectors/therapeutic use , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred BALB C , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Receptors, Neuropeptide Y/agonists , Seizures/chemically induced , Seizures/therapy
5.
J Neurosci Res ; 90(2): 498-507, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21971867

ABSTRACT

Neuropeptide Y (NPY) exerts anxiolytic- and antidepressant-like effects in rodents that appear to be mediated via Y1 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY in the hippocampus or amygdala has previously been shown to confer anxiolytic-like effect in rodents. The present study explored an alternative and more specific approach: overexpression of Y1 receptors. Using a recombinant adeno-associated viral vector (rAAV) encoding the Y1 gene (rAAV-Y1), we, for the first time, induced overexpression of functional transgene Y1 receptors in the hippocampus of adult mice and tested the animals in anxiety- and depression-like behavior. Hippocampal Y1 receptors have been suggested to mediate seizure-promoting effect, so the effects of rAAV-induced Y1 receptor overexpression were also tested in kainate-induced seizures. Y1 receptor transgene overexpression was found to be associated with modest anxiolytic-like effect in the open field and elevated plus maze tests, but no effect was seen on depression-like behavior using the tail suspension and forced swim tests. However, the rAAV-Y1 vector modestly aggravated kainate-induced seizures. These data indicate that rAAV-induced overexpression of Y1 receptors in the hippocampus could confer anxiolytic-like effect accompanied by a moderate proconvulsant adverse effect. Further studies are clearly needed to determine whether Y1 gene therapy might have a future role in the treatment of anxiety disorders.


Subject(s)
Anti-Anxiety Agents/administration & dosage , Convulsants/administration & dosage , Dependovirus/genetics , Gene Expression Regulation, Viral , Genetic Vectors/administration & dosage , Hippocampus/metabolism , Receptors, Neuropeptide Y/biosynthesis , Seizures/metabolism , Animals , Convulsants/toxicity , Genetic Vectors/toxicity , Male , Mice , Receptors, Neuropeptide Y/genetics , Recombinant Fusion Proteins/genetics , Seizures/genetics , Seizures/virology
6.
Drugs Today (Barc) ; 47(1): 53-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21373649

ABSTRACT

Controlling activity of defined populations of neurons without affecting other neurons in the brain is now possible by a new gene- and neuroengineering technology termed optogenetics. Derived from microbial organisms, opsin genes encoding light-activated ion channels and pumps (channelrhodopsin-2 [ChR2]; halorhodopsin [NpHR], respectively), engineered for expression in the mammalian brain, can be genetically targeted into specific neural populations using viral vectors. When exposed to light with appropriate wavelength, action potentials can be triggered in ChR2-expressing neurons, whereas inhibition of action potentials can be obtained in NpHR-expressing neurons, thus allowing for powerful control of neural activity. Optogenetics is now intensively used in laboratory animals, both in vitro and in vivo, for exploring functions of complex neural circuits and information processing in the normal brain and during various neurological conditions. The clinical perspectives of adopting optogenetics as a novel treatment strategy for human neurological disorders have generated considerable interest, largely because of the enormous potential demonstrated in recent rodent and nonhuman primate studies. Restoration of dopamine-related movement dysfunction in parkinsonian animals, amelioration of blindness and recovery of breathing after spinal cord injury are a few examples of such perspectives.


Subject(s)
Genetic Engineering , Light , Nervous System Diseases/therapy , Photic Stimulation , Action Potentials , Brain/physiology , Humans , Neurons/physiology
7.
Neurochem Int ; 55(7): 697-702, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19563850

ABSTRACT

Both brain-derived neurotrophic factor (BDNF) and the serotonin receptor 2A (5-HT(2A)) have been related to depression pathology. Specific 5-HT(2A) receptor changes seen in BDNF conditional mutant mice suggest that BDNF regulates the 5-HT(2A) receptor level. Here we show a direct effect of BDNF on 5-HT(2A) receptor protein levels in primary hippocampal neuronal and mature hippocampal organotypic cultures exposed to different BDNF concentrations for either 1, 3, 5 or 7 days. In vivo effects of BDNF on hippocampal 5-HT(2A) receptor levels were further corroborated in (BDNF +/-) mice with reduced BDNF levels. In primary neuronal cultures, 7 days exposure to 25 and 50ng/mL BDNF resulted in downregulation of 5-HT(2A), but not of 5-HT(1A), receptor protein levels. The BDNF-associated downregulation of 5-HT(2A) receptor levels was also observed in mature hippocampal organotypic cultures, excluding confounding effects of BDNF on immature tissue. BDNF +/- mice showed significant increased 5-HT(2A) receptor levels in hippocampus confirming the association between 5-HT(2A) receptor and BDNF levels in vivo. In conclusion, our results point to a regulatory role of BDNF on 5-HT2A receptor levels. This interaction may be an important mechanism in the role of BDNF in affective disorders emphasizing the need for further elucidating the specificity and the mechanism behind this regulation.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Hippocampus/metabolism , Receptor, Serotonin, 5-HT2A/biosynthesis , Animals , Apoptosis/drug effects , Blotting, Western , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/physiology , Cells, Cultured , Down-Regulation/drug effects , Female , Hippocampus/growth & development , Mice , Mice, Knockout , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/biosynthesis , Organ Culture Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/metabolism
8.
Neuroscience ; 150(4): 984-92, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-17988802

ABSTRACT

Gene therapy-based overexpression of endogenous seizure-suppressing molecules represents a promising treatment strategy for epilepsy. Viral vector-based overexpression of the neuropeptide galanin has been shown to effectively suppress generalized seizures in various animal models of epilepsy. However, it has not been explored whether such treatment can also prevent the epileptogenesis. Using a recombinant adeno-associated viral (rAAV) vector, we induced hippocampal galanin overexpression under the neuron specific enolase promoter in rats. Here we report that in animals with galanin overexpression, the duration of electrographic afterdischarges was shortened and initiation of convulsions was delayed at generalized seizure stages. However, the hippocampal kindling development was unchanged. Short-term plasticity of mossy fiber-cornu ammonis (CA) 3 synapses was unaltered, as assessed by paired-pulse and frequency facilitation of field excitatory postsynaptic potentials (fEPSPs) in hippocampal slices, suggesting that despite high transgene galanin expression, overall release probability of glutamate in these synapses was unaffected. These data indicate that hippocampal rAAV-based galanin overexpression is capable of mediating anticonvulsant effects by lowering the seizure susceptibility once generalized seizures are induced, but does not seem to affect kindling development or presynaptic short-term plasticity in mossy fibers.


Subject(s)
Galanin/physiology , Gene Transfer Techniques , Hippocampus/physiopathology , Neuronal Plasticity/physiology , Seizures/pathology , Seizures/therapy , Animals , Dependovirus/physiology , Disease Models, Animal , Dose-Response Relationship, Radiation , Electric Stimulation/adverse effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Galanin/biosynthesis , Genetic Vectors/physiology , Hippocampus/radiation effects , Kindling, Neurologic/physiology , Male , Rats , Rats, Sprague-Dawley , Reaction Time , Seizures/etiology , Time Factors
9.
Neuropeptides ; 38(4): 253-60, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15337377

ABSTRACT

The endogenous NPY system in the brain is centrally involved in seizure regulation. The present paper reviews the evidence that exogenously applied NPY receptor ligands can inhibit epileptic seizures in various rodent in vitro and in vivo models. Agonists at Y2 and/or Y5 receptors and antagonists at Y1 receptors appear to inhibit seizures, depending on the seizure model studied. Although progress has been made, further studies are needed using transgenic animals as well as novel selective agonists and antagonists to firmly identify the NPY receptors mediating antiepileptic effects. This may lead to the development of future antiepileptic drug treatments targeting the NPY system.


Subject(s)
Anticonvulsants/metabolism , Neuropeptide Y , Receptors, Neuropeptide Y/metabolism , Seizures/metabolism , Animals , Brain/anatomy & histology , Brain/metabolism , Electric Stimulation , Ligands , Neuropeptide Y/agonists , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/metabolism , Seizures/chemically induced
10.
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
11.
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
12.
Neurosci Lett ; 315(1-2): 85-8, 2001 Nov 23.
Article in English | MEDLINE | ID: mdl-11711221

ABSTRACT

GABA(B) receptors play an important role in the excitability of neuronal networks and can influence seizure activity. Here we demonstrate for the first time that kindling, an animal model for human temporal lobe epilepsy, leads to both early and delayed changes of GABA(B) receptor immunoreactivity in hippocampal and cortical areas. We propose that the altered GABA(B) receptor levels might be a compensatory mechanism to reduce excitability induced by recurrent kindled seizures, or alternatively, may promote the development of kindled epilepsy.


Subject(s)
Brain/metabolism , Epilepsy/metabolism , Receptors, GABA-B/metabolism , Animals , Cerebral Cortex/metabolism , Hippocampus/metabolism , Immunohistochemistry , Kindling, Neurologic , Male , Rats , Rats, Sprague-Dawley
13.
Mol Cell Neurosci ; 17(5): 811-20, 2001 May.
Article in English | MEDLINE | ID: mdl-11358480

ABSTRACT

The specific identity of neuronal precursors within the embryonic brain is, at present, not clear. Here we show that cultures with glial characteristics derived from the embryonic mouse or human lateral ganglionic eminence (LGE) can be expanded over many passages and maintain their glial identity. Interestingly, removal of serum and EGF from the culture medium results in the generation of large numbers of neurons. The neurons derived from these cultures display many characteristic features of striatal neurons, which normally derive from the LGE, even after extensive expansion in vitro. Furthermore, a portion of the neurons generated in these cultures were shown to arise from glial fibrillary acidic protein (GFAP)-expressing cells. These results demonstrate that at least a subpopulation of neurogenic LGE precursors exhibit glial characteristics.


Subject(s)
Cell Differentiation/physiology , Cells, Cultured/cytology , Nerve Tissue Proteins , Neuroglia/cytology , Neurons/cytology , Stem Cells/cytology , Telencephalon/embryology , Animals , Cell Culture Techniques , Cells, Cultured/metabolism , Culture Media, Serum-Free/pharmacology , Epidermal Growth Factor/pharmacology , Fetus , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Mice , Neostriatum/cytology , Neostriatum/embryology , Neostriatum/metabolism , Nestin , Neuroglia/metabolism , Neurons/metabolism , Stem Cells/metabolism , Telencephalon/cytology , Telencephalon/metabolism , Tubulin/metabolism , Vimentin/metabolism
14.
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
15.
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
16.
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
17.
Eur J Neurosci ; 12(2): 662-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10712646

ABSTRACT

Neurotrophins modulate synaptic transmission and plasticity in the adult brain. We here show a novel feature of this synaptic modulation, i.e. that two populations of excitatory synaptic connections to granule cells in the dentate gyrus, lateral perforant path (LPP) and medial perforant path (MPP), are differentially influenced by the neurotrophins BDNF and NT-3. Using field recordings and whole-cell patch-clamp recordings in hippocampal slices, we found that paired-pulse (PP) depression at MPP-granule cell synapses was impaired in BDNF knock-out (+/-) mice, but PP facilitation at LPP synapses to the same cells was not impaired. In accordance, scavenging of endogenous BDNF with TrkB-IgG fusion protein also impaired PP depression at MPP-granule cell synapses, but not PP facilitation at LPP-granule cell synapses. Conversely, in NT-3+/- mice, PP facilitation was impaired at LPP-granule cell synapses whilst PP depression at MPP-granule cell synapses was unaffected. These deficits could be reversed by application of exogenous neurotrophins in an afferent-specific manner. Our data suggest that BDNF and NT-3 differentially regulate the synaptic impact of different afferent inputs onto single target neurons in the CNS.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Dentate Gyrus/drug effects , Neuronal Plasticity/drug effects , Neurotrophin 3/physiology , Perforant Pathway/drug effects , Synaptic Transmission/drug effects , Afferent Pathways/physiology , Animals , Brain-Derived Neurotrophic Factor/deficiency , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/pharmacology , Dentate Gyrus/physiology , Genotype , Mice , Mice, Knockout , Mice, Neurologic Mutants , Neurotrophin 3/deficiency , Neurotrophin 3/genetics , Neurotrophin 3/pharmacology , Patch-Clamp Techniques , Receptor, trkB/genetics , Receptor, trkB/physiology , Recombinant Fusion Proteins/physiology
18.
Hippocampus ; 10(6): 684-92, 2000.
Article in English | MEDLINE | ID: mdl-11153714

ABSTRACT

Subicular neurons receive direct afferent connections from the vast majority of CA1 pyramidal cells and send their axons to the various brain areas. Because of this strategic position, subicular cells can modulate output of the hippocampus and, thus, play a significant part in memory, spatial processing, and seizure amplification and propagation from the hippocampus. Despite its important role as a hippocampal interface with different brain regions, present knowledge of the subiculum and the plastic properties of the synapses on the subicular neurons is rather limited. By using IR-DIC videomicroscopy and whole-cell patch-clamp recordings in mouse hippocampal slices, I demonstrated that long-term potentiation (LTP) in CA1-subicular cell synapses can be readily induced by high-frequency stimulation (HFS) of the afferents, but not by pairing of low-frequency stimulation with depolarization of postsynaptic cells. This tetanus-induced LTP is input specific, insensitive to the N-methyl-D-aspartate (NMDA) receptor antagonist 3-[(R)-2Carboxipiperazin-4-yl]-propyl-1-phosphonic acid (R-CPP), and reduces paired-pulse facilitation in potentiated synapses. Subsequent morphologic analysis of the recorded cells, which were filled either with Lucifer Yellow or Biocytin, revealed pyramidal-shaped neurons localized predominantly in the deep layer of the subiculum, close to the CA1 border. Axons of the majority of these neurons extended to the alveus and on toward the hippocampus, probably exiting it via the fornix. These data indicate that CA1-subicular cell synapses in mice exhibit LTP, which can be expressed presynaptically, and its induction does not require NMDA-receptor activation. The observed activity-dependent plasticity might play an important role in the integrative mechanisms of the subiculum and may influence transfer of information from the hippocampus to subcortical and cortical brain areas.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Long-Term Potentiation/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Fluorescent Dyes , Isoquinolines , Long-Term Potentiation/drug effects , Lysine/analogs & derivatives , Male , Mice , Mice, Inbred C57BL , Microscopy, Video , Organ Culture Techniques , Patch-Clamp Techniques , Periodicity , Piperazines/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology
19.
J Neurosci ; 18(21): 8730-9, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9786980

ABSTRACT

In the adult brain, neurotrophin-3 (NT-3) is mainly localized in dentate granule cells, and its expression is decreased by various stimuli, e.g., seizure activity. We have examined the role of endogenous NT-3 for excitatory synaptic transmission at lateral perforant path-dentate granule cell synapses using hippocampal slices from NT-3 knock-out (+/-) and wild-type (+/+) mice. Paired-pulse facilitation (PPF) and also short-term synaptic plasticity induced by a brief, high-frequency train of afferent stimulation were reduced, but the expression of long-term potentiation was not affected in the NT-3+/- mice. Incubation of the slices with recombinant NT-3 reversed the deficit in PPF through a mechanism requiring de novo protein synthesis, implying that the impaired short-term plasticity does not result from a developmental alteration. No changes of overall presynaptic release probability, measured by the progressive block of NMDA receptor-mediated synaptic currents by MK-801, or desensitization of AMPA receptors were detected. Because NT-3 expression is reduced after focal seizures, impaired short-term facilitation may represent a protective response that limits the propagation of epileptiform activity from the entorhinal cortex to the hippocampus.


Subject(s)
Dentate Gyrus/physiology , Nerve Growth Factors/physiology , Perforant Pathway/physiology , Synapses/physiology , Animals , Dentate Gyrus/drug effects , Excitatory Postsynaptic Potentials , In Vitro Techniques , Long-Term Potentiation , Membrane Potentials/drug effects , Mice , Mice, Knockout , Mossy Fibers, Hippocampal/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Neuronal Plasticity , Neurotrophin 3 , Patch-Clamp Techniques , Perforant Pathway/drug effects , Presynaptic Terminals/metabolism , Protein Biosynthesis , Pyrrolidinones/pharmacology , Receptors, AMPA/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Recombinant Proteins/pharmacology
20.
Eur J Neurosci ; 10(1): 213-20, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9753129

ABSTRACT

Selective immunolesioning of the basal forebrain cholinergic system by 192 IgG-saporin, which leads to a dramatic loss of the cholinergic innervation in cortical and hippocampal regions, facilitates the development of hippocampal kindling in rats. The aim of the present study was to explore whether grafted cholinergic neurones are able to reverse the lesion-induced increase of seizure susceptibility. Intraventricular 192 IgG-saporin was administered to rats which 3 weeks later were implanted with rat embryonic, acetylcholine-rich septal-diagonal band tissue ('cholinergic grafts') or cortical tissue/vehicle ('sham grafts') bilaterally into the hippocampal formation. After 3 months, the grafted animals as well as non-lesioned control rats were subjected to daily hippocampal kindling stimulations. In the animals with cholinergic grafts, which had reinnervated the hippocampus and dentate gyrus bilaterally, there was a marked suppression of the development of seizures as compared with the hyperexcitable, sham-grafted rats. This effect was significantly correlated to the density of the graft-derived cholinergic innervation of the host hippocampal formation. The kindling rate in the rats with cholinergic grafts was similar to that in non-lesioned controls. These results provide further evidence that the intrinsic basal forebrain cholinergic system dampens kindling epileptogenesis and demonstrate that this function can be exerted also by grafted cholinergic neurones.


Subject(s)
Brain Tissue Transplantation , Cholinergic Fibers/transplantation , Epilepsy/surgery , Hippocampus/surgery , Kindling, Neurologic/physiology , Acetylcholinesterase/analysis , Animals , Antibodies, Monoclonal/pharmacology , Cholinergic Agents/pharmacology , Hippocampus/cytology , Hippocampus/physiopathology , Immunotoxins/pharmacology , Male , N-Glycosyl Hydrolases , Nerve Regeneration/physiology , Neurons/drug effects , Neurons/enzymology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins
SELECTION OF CITATIONS
SEARCH DETAIL
...