Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Georgian Med News ; (243): 84-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26087739

ABSTRACT

Ischemic stroke is a major cause of adult disability. Stroke-induced brain damage is accompanied by inflammation - activation of resident microglia and infiltration of blood-circulating monocytes. The effect of these cells on neuro-plasticity and recovery after stroke could be detrimental as well as beneficial. The future challenge is to understand the mechanisms of action of immune cells on cellular plasticity occurring in post-stroke brain and divert them towards support of functional recovery.


Subject(s)
Brain Injuries/pathology , Brain Ischemia/therapy , Inflammation/therapy , Microglia/immunology , Stroke/therapy , Adult , Animals , Brain Injuries/therapy , Brain Ischemia/diagnosis , Brain Ischemia/immunology , Cytokines/immunology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , Microglia/pathology , Stroke/diagnosis , Stroke/immunology
2.
Eur J Neurol ; 20(3): 473-479, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23057628

ABSTRACT

BACKGROUND AND PURPOSE: Clinical stroke trials with stem cell-based approaches aiming for trophic actions, modulation of inflammation and neuroprotection are ongoing. However, experimental studies also suggest that neuronal replacement by grafted neural stem cells (NSCs) and possibly by endogenous NSCs from the subventricular zone (SVZ) may restore function in the stroke-damaged striatum. To evaluate the potential clinical impact of these findings, we analyzed the spatial relationship of infarcts to the SVZ and the proportion of individuals with striatal lesions in a consecutive series of ischaemic stroke patients. METHODS: Patients aged 20-75 years with first-ever ischaemic stroke underwent DW-MRI of the brain within 4 days after stroke onset. We analyzed location, size, number of acute focal ischaemic abnormalities and their spatial relationship to the SVZ. Stroke severity was assessed using NIH Stroke Scale (NIHSS). RESULTS: Of 108 included patients, the distance from the nearest margin of the infarct(s) to the SVZ was ≤2 mm in 51/102 patients with visible ischaemic lesions on DW-MRI. Twenty-four patients had involvement of striatum. Eight of these had predominantly striatal lesions, that is >50% of the total ischaemic lesion volume was located in caudate nucleus and/or putamen. These 8 patients had a median NIHSS of 3. CONCLUSIONS: Many stroke patients have infarcts located close to the SVZ, providing some supportive evidence that optimized endogenous neurogenesis may have therapeutic potential. However, predominantly striatal infarcts are rare and tend to give mild neurological deficits, indicating that striatum should not be the primary target for neuronal replacement efforts in humans.


Subject(s)
Brain Infarction/pathology , Corpus Striatum/pathology , Adult , Aged , Diffusion Magnetic Resonance Imaging , Female , Humans , Male , Middle Aged , Neurogenesis/physiology , Stroke/pathology , Young Adult
3.
Neuroscience ; 214: 159-70, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22531371

ABSTRACT

Stroke-induced neurogenesis originates from a neural stem cell (NSC) niche in subventricular zone (SVZ). In mice, NSCs are concentrated in a so-called "neurogenic spot" in the lateral angle area of SVZ. We aimed to identify the "neurogenic spot" in the rat SVZ and to characterize the cellular changes in the ependymal cell compartment in this area at different time points after middle cerebral artery occlusion. The majority of ependymal cells outlining the ventricular wall did not proliferate, and their numbers in the "neurogenic spot" declined at 6 and 16weeks after stroke. Cells with the ultrastructural properties of ependymal cells were detected in the adjacent striatum. The number of these ectopic ependymal cells (EE cells) correlated positively with the magnitude of lateral ventricular enlargement and negatively with the ependymal cell number in the "neurogenic spot". EE cells were found along blood vessels, accumulated in the pericyst regions, and participated in scar formation but did not incorporate BrdU. We provide the first evidence for the occurrence of EE cells in the ischemic striatum following stroke.


Subject(s)
Choristoma/pathology , Corpus Striatum/cytology , Disease Models, Animal , Ependyma/cytology , Neurogenesis , Stroke/pathology , Animals , Corpus Striatum/physiology , Corpus Striatum/ultrastructure , Ependyma/physiology , Ependyma/ultrastructure , Male , Neurogenesis/physiology , Rats , Rats, Wistar
4.
Neuroscience ; 158(3): 1021-9, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-18662748

ABSTRACT

In the adult mammalian brain, neurogenesis from neural stem/progenitor cells continues in two regions: the subgranular zone in the dentate gyrus and the subventricular zone lining the lateral ventricles. The generated neuroblasts migrate to their appropriate location and differentiate to mature granule cells and olfactory bulb interneurons, respectively. Following injury such as stroke, neuroblasts generated in the subventricular zone migrate also into areas which are not normally neurogenic, e.g. striatum and cerebral cortex. In the initial studies in rodents, brain inflammation and microglia activation were found to be detrimental for the survival of the new hippocampal neurons early after they had been born. The role of inflammation for adult neurogenesis has, however, turned out to be much more complex. Recent experimental evidence indicates that microglia under certain circumstances can be beneficial and support the different steps in neurogenesis, progenitor proliferation, survival, migration, and differentiation. Here we summarize the current knowledge on the role of inflammation and in particular of microglia in adult neurogenesis in the intact and injured mammalian brain. We conclude that microglia activation, as an indicator of inflammation, is not pro- or antineurogenic per se but the net outcome is dependent on the balance between secreted molecules with pro- and antiinflammatory action.


Subject(s)
Encephalitis/immunology , Microglia/immunology , Nerve Regeneration/immunology , Neurogenesis/immunology , Neuronal Plasticity/immunology , Stroke/immunology , Animals , Cell Communication/immunology , Cytokines/metabolism , Encephalitis/physiopathology , Gliosis/immunology , Gliosis/physiopathology , Humans , Stroke/physiopathology
5.
Exp Cell Res ; 313(3): 588-601, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17156776

ABSTRACT

Isolation and expansion of neural stem cells (NSCs) of human origin are crucial for successful development of cell therapy approaches in neurodegenerative diseases. Different epigenetic and genetic immortalization strategies have been established for long-term maintenance and expansion of these cells in vitro. Here we report the generation of a new, clonal NSC (hc-NSC) line, derived from human fetal cortical tissue, based on v-myc immortalization. Using immunocytochemistry, we show that these cells retain the characteristics of NSCs after more than 50 passages. Under proliferation conditions, when supplemented with epidermal and basic fibroblast growth factors, the hc-NSCs expressed neural stem/progenitor cell markers like nestin, vimentin and Sox2. When growth factors were withdrawn, proliferation and expression of v-myc and telomerase were dramatically reduced, and the hc-NSCs differentiated into glia and neurons (mostly glutamatergic and GABAergic, as well as tyrosine hydroxylase-positive, presumably dopaminergic neurons). RT-PCR analysis showed that the hc-NSCs retained expression of Pax6, Emx2 and Neurogenin2, which are genes associated with regionalization and cell commitment in cortical precursors during brain development. Our data indicate that this hc-NSC line could be useful for exploring the potential of human NSCs to replace dead or damaged cortical cells in animal models of acute and chronic neurodegenerative diseases. Taking advantage of its clonality and homogeneity, this cell line will also be a valuable experimental tool to study the regulatory role of intrinsic and extrinsic factors in human NSC biology.


Subject(s)
Cell Transformation, Viral , Cerebral Cortex/cytology , Fetal Stem Cells/physiology , Neurons/physiology , Oncogene Protein p55(v-myc)/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Clone Cells/physiology , Down-Regulation , Fetal Stem Cells/enzymology , Gene Expression Regulation, Developmental , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Oncogene Protein p55(v-myc)/genetics , Phenotype , Telomerase , Tubulin/metabolism
6.
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
7.
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
8.
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
9.
Neuroscience ; 106(1): 27-41, 2001.
Article in English | MEDLINE | ID: mdl-11564414

ABSTRACT

Gene expression for glial cell line-derived neurotrophic factor (GDNF) family ligands and receptors was analyzed with in situ hybridization after two focal ischemic insults of different severities. Focal ischemia was induced in rats by either 30 min or 2 h of middle cerebral artery occlusion (MCAO), causing damage to the striatum only, or involving also the parietal cortex, respectively. We found modest, transient elevation of GDNF mRNA in the dentate granule cell layer. In addition, the number of GDNF mRNA-expressing cells increased in the cortex and striatum after 2 h or 30 min of MCAO, respectively. No changes of neurturin or persephin mRNA expression were detected. Both c-Ret and GFRalpha1 mRNA levels were markedly increased in the ipsilateral cortex outside the ischemic lesion at 6-24 h after the 2-h insult, whereas GFRalpha2 expression was decreased in cortical areas both within and outside the lesion. Similar increases of c-Ret and GFRalpha1 mRNA levels were detected in the striatum, and to a lesser extent, in the cortex following 30 min of MCAO. The 2-h insult also gave rise to transient increases of c-Ret and GFRalpha1 mRNA in hippocampal subregions. Thirty minutes and 2 h of MCAO lead to elevated c-Ret, and GFRalpha1 or GFRalpha2 mRNA expression, respectively, in the ipsilateral ventroposterolateral thalamic nucleus. Both insults induced increased levels of GFRalpha1 mRNA in the subventricular zone of the lateral ventricle. Our data indicate major changes of GDNF family signaling in the forebrain, regulated mainly through altered receptor levels, in the post-ischemic phase. These changes could enhance neuroprotective and neuroregenerative responses both to endogenous and exogenous GDNF ligands.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Drosophila Proteins , Gene Expression Regulation/physiology , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Stroke/metabolism , Animals , Brain/pathology , Brain/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/physiopathology , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Immunohistochemistry , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Ligands , Male , Neostriatum/metabolism , Neostriatum/pathology , Neostriatum/physiopathology , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurturin , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Rats , Rats, Wistar , Receptor Protein-Tyrosine Kinases/metabolism , Stroke/pathology , Stroke/physiopathology , Time Factors
10.
Brain Res ; 913(2): 117-32, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11549375

ABSTRACT

Detailed quantitative analysis of the vulnerability of different hippocampal and striatal neurons to global forebrain ischemia has not previously been performed. Here we have studied the survival of immunocytochemically identified neurons using an unbiased stereological method in rats subjected to global forebrain ischemia for 30 min and sacrificed 48 h, 1 week or 4 weeks thereafter. Within the hippocampal formation, there was extensive, progressive loss of CA1 pyramidal neurons and dentate hilar neuropeptide Y (NPY)-positive interneurons. In contrast, no reduction of the number of CA3 and CA4 pyramidal neurons or hilar parvalbumin-positive interneurons was detected. In the dorsolateral striatum, the insult caused a major loss of projection neurons immunoreactive to dopamine- and adenosine 3':5'-monophosphate-regulated phosphoprotein with a molecular weight of 32 kilodalton (DARPP-32). The number of parvalbumin-positive striatal interneurons was significantly reduced, while NPY-positive interneurons were resistant. All striatal cholinergic interneurons survived the ischemic insult. At 48 h following the ischemia, the cholinergic interneurons within the lesioned striatum transiently expressed the p75 neurotrophin receptor (p75(NTR)), as shown by double-label immunocytochemistry. Furthermore, there was a significant increase in the number of choline acetyltransferase (ChAT)- and TrkA-immunoreactive interneurons at 4 weeks after the insult. Injections with the cell mitotic division marker BrdU provided no evidence that the elevated cholinergic cell number was due to neurogenesis. Probably, the higher number of ChAT- and TrkA-positive interneurons reflected increased intracellular levels of the corresponding proteins leading to more cells detectable with immunocytochemistry. This study gives the first quantitative description of the vulnerability of defined hippocampal and striatal neurons after global forebrain ischemia. The ischemia-induced increases of p75(NTR), TrkA and ChAT in cholinergic striatal interneurons at various time points after the insult suggest that neurotrophin signaling might be important for the survival and function of these cells in the post-ischemic phase.


Subject(s)
Brain Ischemia/metabolism , Cell Survival/physiology , Hippocampus/metabolism , Neostriatum/metabolism , Nerve Degeneration/metabolism , Nerve Tissue Proteins , Neurons/metabolism , Animals , Antigens, Surface/metabolism , Body Weight/physiology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Count , Choline O-Acetyltransferase/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32 , Fluoresceins , Fluorescent Dyes/pharmacokinetics , Hippocampus/pathology , Hippocampus/physiopathology , Immunohistochemistry , Interneurons/metabolism , Interneurons/pathology , Male , Neostriatum/pathology , Neostriatum/physiopathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Neurons/pathology , Neuropeptide Y/metabolism , Organic Chemicals , Parvalbumins/metabolism , Phosphoproteins/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Rats , Rats, Wistar , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism
11.
Eur J Neurosci ; 14(1): 10-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11488944

ABSTRACT

Neurogenesis in the adult rat dentate gyrus was studied following focal ischemic insults produced by middle cerebral artery occlusion (MCAO). Animals were subjected to either 30 min of MCAO, which causes damage confined to the striatum, or 2 h of MCAO, which leads to both striatal and cortical infarction. When compared to sham-operated rats, MCAO-rats showed a marked increase of the number of cells double-labelled for 5-bromo-2'-deoxyuridine-5'-monophosphate (BrdU; injected during 4-6 days postischemia) and neuronal-specific antigen (NeuN; a marker of postmitotic neurons) in the ipsilateral dentate granule cell layer and subgranular zone at 5 weeks following the 2 h insult. Only a modest and variable increase of BrdU-labelled cells was found after 30 min of MCAO. The enhanced neurogenesis was not dependent on cell death in the hippocampus, and its magnitude was not correlated to the degree of cortical damage. Systemic administration of the N-methyl-D-aspartate (NMDA) receptor blocker dizocilpine maleate (MK-801) completely suppressed the elevated neurogenesis following 2 h of MCAO. Our findings indicate that stroke leads to increased neurogenesis in the adult rat dentate gyrus through glutamatergic mechanisms acting on NMDA receptors. This modulatory effect may be mediated through changes in the levels of several growth factors, which occur after stroke, and could influence various regulatory steps of neurogenesis.


Subject(s)
Cell Differentiation/physiology , Cell Division/physiology , Dentate Gyrus/growth & development , Infarction, Middle Cerebral Artery/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Antimetabolites/pharmacokinetics , Biomarkers/analysis , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Bromodeoxyuridine/pharmacokinetics , Cell Differentiation/drug effects , Cell Division/drug effects , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Neuronal Plasticity/drug effects , Neurons/cytology , Neurons/drug effects , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Recovery of Function/drug effects , Recovery of Function/physiology
12.
Neurosci Lett ; 305(3): 169-72, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11403932

ABSTRACT

Tissue levels of brain-derived neurotrophic factor (BDNF) protein were studied using enzyme immunoassay in different forebrain regions in the ipsi- and contralateral hemispheres of rats housed under enriched or standard conditions after the middle cerebral artery ligation. BDNF levels in the ipsilateral to ligation side was significantly higher only in the frontal cortex of standard as compared to enriched rats. However, BDNF overall was more abundant in standard than in enriched group. In addition, BDNF levels detected in the hippocampus and frontal cortex on the ischemic side of standard rats was higher as compared to contralateral side. The present study shows that housing conditions after permanent middle cerebral artery ligation leads to differential regulation of BDNF protein levels in forebrain regions which might have important implication for post-ischemic recovery.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Environment , Prosencephalon/metabolism , Stroke/metabolism , Animals , Brain Ischemia/metabolism , Frontal Lobe/metabolism , Hippocampus/metabolism , Housing, Animal , Immunoenzyme Techniques , Rats , Tissue Distribution
13.
Exp Neurol ; 169(2): 351-63, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11358448

ABSTRACT

The survival of different neuron types and the expression of the p75 neurotrophin receptor (p75(NTR)) after focal cerebral ischemia were studied in the mouse striatum using immunocytochemical and histochemical techniques and stereological procedures. As assessed at 1 week after 30 min of middle cerebral artery occlusion, the order of vulnerability was projection neurons > parvalbumin-expressing interneurons > nitric oxide synthase-containing interneurons > cholinergic interneurons. Within the ischemic lesion, projection neurons were almost completely lost whereas cholinergic interneurons were spared. Calretinin-immunoreactive interneurons also seemed resistant to the insult. Expression of p75(NTR) was induced in cholinergic interneurons within the lesioned area, raising the possibility of a protective action. However, the number of cholinergic interneurons was unaffected in p75(NTR) knockout mice subjected to the same ischemic insult. These quantitative data demonstrate that striatal neurons in the mouse are differentially susceptible to ischemic damage and argue against a significant role of p75(NTR) for the high resistance of cholinergic interneurons.


Subject(s)
Corpus Striatum/metabolism , Ischemic Attack, Transient/genetics , Neurons/metabolism , Receptors, Nerve Growth Factor/genetics , Stroke/genetics , Up-Regulation , Animals , Calbindin 2 , Corpus Striatum/pathology , Functional Laterality , Gene Expression Regulation , Humans , Interneurons/metabolism , Interneurons/pathology , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Cerebral Artery , Neurons/pathology , Nitric Oxide Synthase/metabolism , Parvalbumins/analysis , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/physiology , S100 Calcium Binding Protein G/analysis , Stroke/metabolism , Stroke/pathology
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.
Curr Protoc Toxicol ; Chapter 2: Unit 2.7, 2001 May.
Article in English | MEDLINE | ID: mdl-23045047

ABSTRACT

This unit describes two methods of in situ hybridization: one uses an ³5S-labeled oligonucleotide probe and the other uses a digoxigenin-labeled oligonucleotide probe on frozen, cryostat-sectioned samples. These methods allow detection of the physical distribution and expression levels of target mRNA. Protocols are also included for labeling the probes and preparing the sample material.


Subject(s)
Cells/metabolism , Histocytochemistry/methods , RNA, Messenger/metabolism , Animals , Biological Transport/drug effects , Brain/cytology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells/drug effects , Cells/pathology , Humans , In Situ Hybridization , Molecular Probe Techniques , Organ Specificity , Xenobiotics/toxicity
16.
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
17.
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
18.
Gene Ther ; 6(11): 1851-66, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10602381

ABSTRACT

Neural stem cell lines represent a homogeneous source of cells for genetic, developmental, and gene transfer and repair studies in the nervous system. Since both gene transfer of neurotrophic factors and cell replacement strategies are of immediate interest for therapeutical purposes, we have generated BDNF-secreting neural stem cell lines and investigated to what extent different endogenous levels of BDNF expression affect in vitro survival, proliferation and differentiation of these cells. Also, we have investigated the in vivo effects of such BDNF gene transfer procedure in the rat neostriatum. Hippocampus- and cerebellum-derived cell lines reacted differently to manipulations aimed at varying their levels of BDNF production. Over-expression of BDNF enhanced survival of both cell types, in a serum-deprivation assay. Conversely, and ruling out unspecific effects, expression of an antisense version of BDNF resulted in compromised survival of cerebellum-derived cells, and in a lethal phenotype in hippocampal progenitors. These data indicate that endogenous BDNF level strongly influences the in vitro survival of these cells. These effects are more pronounced for hippocampus- than for cerebellum-derived progenitors. Hippocampus-derived BDNF overproducers showed no major change in their capacity to differentiate towards a neuronal phenotype in vitro. In contrast, cerebellar progenitors overproducing BDNF did not differentiate into neurons, whereas cells expressing the antisense BDNF construct generated cells with morphological features of neurons and expressing immunological neuronal markers. Taken together, these results provide evidence that BDNF controls both the in vitro survival and differentiation of neural stem cells. After in vivo transplantation of BDNF-overproducing cells to the rat neostriatum, these survived better than the control ones, and induced the expected neurotrophic effects on cholinergic neurons. However, long-term (3 months) administration of BDNF resulted in detrimental effects, at this location. These findings may be of importance for the understanding of brain development, for the design of therapeutic neuro-regenerative strategies, and for cell replacement and gene therapy studies.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cerebellum/metabolism , Gene Transfer Techniques , Hippocampus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebellum/cytology , Female , Hippocampus/cytology , Immunohistochemistry , Neurons/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...