Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Epilepsy Curr ; 19(3): 177-181, 2019.
Article in English | MEDLINE | ID: mdl-31037960

ABSTRACT

A large body of evidence that has accumulated over the past decade strongly supports the role of both blood-brain barrier (BBB) dysfunction and perivascular inflammation in the pathophysiology of epilepsy. Recent preclinical studies indicate that prolonged seizure- or brain injury-induced BBB dysfunction and subsequent perivascular inflammation may play an important role in post-traumatic epileptogenesis. In turn, perivascular inflammation can further sustain BBB dysfunction. In genetic epilepsies, such as tuberous sclerosis complex and other related epileptogenic developmental pathologies, there is an association between the underlying gene mutation, BBB dysfunction, and perivascular inflammation, but evidence for a causal link to epilepsy is lacking. Future neuroimaging studies might shed light on the role of BBB function in different epilepsies and address the potential for disease modification by targeting both the BBB and perivascular inflammation in acquired and genetic epilepsies.

2.
J Neuroinflammation ; 15(1): 212, 2018 Jul 23.
Article in English | MEDLINE | ID: mdl-30037344

ABSTRACT

BACKGROUND: Previous studies in various rodent epilepsy models have suggested that mammalian target of rapamycin (mTOR) inhibition with rapamycin has anti-epileptogenic potential. Since treatment with rapamycin produces unwanted side effects, there is growing interest to study alternatives to rapamycin as anti-epileptogenic drugs. Therefore, we investigated curcumin, the main component of the natural spice turmeric. Curcumin is known to have anti-inflammatory and anti-oxidant effects and has been reported to inhibit the mTOR pathway. These properties make it a potential anti-epileptogenic compound and an alternative for rapamycin. METHODS: To study the anti-epileptogenic potential of curcumin compared to rapamycin, we first studied the effects of both compounds on mTOR activation, inflammation, and oxidative stress in vitro, using cell cultures of human fetal astrocytes and the neuronal cell line SH-SY5Y. Next, we investigated the effects of rapamycin and intracerebrally applied curcumin on status epilepticus (SE)-induced inflammation and oxidative stress in hippocampal tissue, during early stages of epileptogenesis in the post-electrical SE rat model for temporal lobe epilepsy (TLE). RESULTS: Rapamycin, but not curcumin, suppressed mTOR activation in cultured astrocytes. Instead, curcumin suppressed the mitogen-activated protein kinase (MAPK) pathway. Quantitative real-time PCR analysis revealed that curcumin, but not rapamycin, reduced the levels of inflammatory markers IL-6 and COX-2 in cultured astrocytes that were challenged with IL-1ß. In SH-SY5Y cells, curcumin reduced reactive oxygen species (ROS) levels, suggesting anti-oxidant effects. In the post-SE rat model, however, treatment with rapamycin or curcumin did not suppress the expression of inflammatory and oxidative stress markers 1 week after SE. CONCLUSIONS: These results indicate anti-inflammatory and anti-oxidant properties of curcumin, but not rapamycin, in vitro. Intracerebrally applied curcumin modified the MAPK pathway in vivo at 1 week after SE but failed to produce anti-inflammatory or anti-oxidant effects. Future studies should be directed to increasing the bioavailability of curcumin (or related compounds) in the brain to assess its anti-epileptogenic potential in vivo.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Curcumin/therapeutic use , Oxidative Stress/drug effects , Sirolimus/therapeutic use , Status Epilepticus , Animals , Astrocytes/drug effects , Brain/cytology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fetus/cytology , Gene Expression Regulation/drug effects , Humans , Inflammation , Male , Neuroblastoma/pathology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Status Epilepticus/complications , Status Epilepticus/drug therapy , Status Epilepticus/physiopathology
3.
Sci Rep ; 7(1): 11592, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912503

ABSTRACT

Temporal lobe epilepsy (TLE) is a common chronic neurological disease in humans. A number of studies have demonstrated differential expression of miRNAs in the hippocampus of humans with TLE and in animal models of experimental epilepsy. However, the dissimilarities in experimental design have led to largely discordant results across these studies. Thus, a comprehensive comparison is required in order to better characterize miRNA profiles obtained in various post-status epilepticus (SE) models. We therefore created a database and performed a meta-analysis of differentially expressed miRNAs across 3 post-SE models of epileptogenesis (electrical stimulation, pilocarpine and kainic acid) and human TLE with hippocampal sclerosis (TLE-HS). The database includes data from 11 animal post-SE studies and 3 human TLE-HS studies. A total of 378 differentially expressed miRNAs were collected (274 up-regulated and 198 down-regulated) and analyzed with respect to the post-SE model, time point and animal species. We applied the novel robust rank aggregation method to identify consistently differentially expressed miRNAs across the profiles. It highlighted common and unique miRNAs at different stages of epileptogenesis. The pathway analysis revealed involvement of these miRNAs in key pathogenic pathways underlying epileptogenesis, including inflammation, gliosis and deregulation of the extracellular matrix.


Subject(s)
Epilepsy, Temporal Lobe/genetics , Gene Expression Regulation , Genetic Association Studies , MicroRNAs/genetics , Animals , Biomarkers , Computational Biology/methods , Disease Models, Animal , Epilepsy, Temporal Lobe/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , Oligonucleotide Array Sequence Analysis , Signal Transduction , Species Specificity
4.
Semin Cell Dev Biol ; 38: 26-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25444846

ABSTRACT

The blood-brain barrier (BBB) is a dynamic and complex system which separates the brain from the blood. It helps to maintain the homeostasis of the brain, which is essential for normal neuronal functioning. BBB function is impaired in several neurological diseases, including epilepsy in which it may lead to abnormal and excessive neuronal firing. In this review we will discuss how BBB dysfunction can affect neuronal function and how this can lead to seizures and epilepsy. We will also summarize new therapies that aim to preserve or restore BBB function in order to prevent or reduce epileptogenesis.


Subject(s)
Blood-Brain Barrier/physiopathology , Epilepsy/physiopathology , Seizures/physiopathology , Animals , Blood-Brain Barrier/physiology , Epilepsy/drug therapy , Humans , Seizures/drug therapy
5.
Neuroscience ; 277: 455-73, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25080160

ABSTRACT

Temporal lobe epilepsy (TLE) is the most common form of focal epilepsies in adults. It is often initiated by an insult or brain injury which triggers a series of alterations which ultimately lead to seizures (epilepsy). In 50-70% of people with TLE the condition cannot be adequately treated by the present antiepileptic drugs. During the last decade the blood-brain barrier (BBB) has received renewed interest as a potential target to treat TLE or its progression. BBB changes have been observed in brain tissue of people with epilepsy as well as in experimental models at the structural, cellular and molecular level that could explain its role in the development and progression of epilepsy (epileptogenesis) as well as the development of drug resistance. Here, we will discuss the role of the BBB in TLE and drug resistance and summarize potential new therapies that may restore normal BBB function in order to put a brake on epileptogenesis and/or to improve drug treatment.


Subject(s)
Blood-Brain Barrier/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain/physiopathology , Drug Resistance , Epilepsy, Temporal Lobe/drug therapy , Humans , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology
6.
Neurobiol Dis ; 63: 74-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24321435

ABSTRACT

The blood-brain barrier (BBB) plays an important role in the homeostasis of the brain. BBB dysfunction has been implicated in the pathophysiology of various neurological disorders, including epilepsy in which it may contribute to disease progression. Precise understanding of BBB dynamics during epileptogenesis may be of importance for the assessment of future therapies, including BBB leakage blocking-agents. Longitudinal changes in BBB integrity can be studied with in vivo magnetic resonance imaging (MRI) in combination with paramagnetic contrast agents. Although this approach has shown to be suitable to detect major BBB leakage during the acute phase in experimental epilepsy models, so far no studies have provided information on dynamics of the extent of BBB leakage towards later phases. Therefore a sensitive and quantitative approach was used in the present study, involving fast T1 mapping (dynamic approach) during a steady-state infusion of gadobutrol, as well as pre- and post-contrast T1-weighted MRI (post-pre approach). This was applied in an experimental epilepsy model in which previous MRI studies failed to detect BBB leakage during epileptogenesis. Adult male Sprague-Dawley rats were injected with kainic acid to induce status epilepticus (SE). MRI experiments were performed before SE (control) and during the acute (1 day) and chronic epileptic phases (6 weeks after SE). BBB leakage was quantified by fast T1 mapping (Look-Locker gradient echo MRI) with a time resolution of 48 s from 5 min before up to 45 min after 20 min step-down infusion of 0.2M gadobutrol. In addition, T1-weighted MRI was acquired before and 45 min after infusion. MRI data were compared to post-mortem microscopic analysis using the BBB tracer fluorescein. Our MRI data showed BBB leakage, which was evident at 1 day and 6 weeks after SE in the hippocampus, entorhinal cortex, amygdala and piriform cortex. These findings were confirmed by microscopic analysis of fluorescein leakage. Furthermore, our MRI data revealed non-uniform BBB leakage throughout epileptogenesis. This study demonstrates BBB leakage in specific brain regions during epileptogenesis, which can be quantified using MRI. Therefore, MRI may be a valuable tool for experimental or clinical studies to elucidate the role of the BBB in epileptogenesis.


Subject(s)
Blood-Brain Barrier/physiopathology , Capillary Permeability/physiology , Status Epilepticus/complications , Status Epilepticus/pathology , Animals , Blood-Brain Barrier/pathology , Brain/pathology , Brain/physiopathology , Contrast Media/pharmacokinetics , Disease Models, Animal , Image Processing, Computer-Assisted , Longitudinal Studies , Magnetic Resonance Imaging , Male , Organometallic Compounds/pharmacokinetics , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors
7.
Neuroscience ; 188: 109-16, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21575686

ABSTRACT

Since the membrane attack complex (MAC), an end product of the activated complement cascade, has been shown to play a role in neurodegeneration, we investigated to which extent MAC contributes to structural reorganization, neuronal cell death, and seizure development in two rat models for temporal lobe epilepsy. We used the electrically-induced status epilepticus (SE) model and the kindling model in C6-deficient rats (that are unable to form MAC) and wild-type (WT) PVG/c rats. Structural reorganization was investigated using hilar cell counts and mossy fiber sprouting. Seizure development was monitored using electroencephalographic (EEG) recordings. 4 weeks after electrically stimulated SE, hilar cell counts in C6-deficient and WT post-SE rats were significantly decreased compared to an unstimulated control group, but not different between C6-deficient and WT post-SE. Since seizure development was unexpectedly absent in most post-SE rats we assessed epileptogenesis using the kindling rate as main parameter. Kindling development was slightly delayed in C6-deficient rats compared to WT rats. The lack of effect of C6 deficiency on hilar cell death and mossy fiber sprouting after electrically-induced SE or kindling argues against a role of the terminal complement complex in neuronal cell death induced by SE or seizures. A small but significant delay of kindling epileptogenesis suggests a subtle role of MAC in seizure spread. Whether complement components upstream of MAC play a crucial role in neuronal death and/or epileptogenesis needs to be further investigated.


Subject(s)
Brain/physiopathology , Complement C6/deficiency , Mossy Fibers, Hippocampal/pathology , Seizures/physiopathology , Animals , Brain/pathology , Cell Death , Complement Membrane Attack Complex/metabolism , Disease Models, Animal , Electric Stimulation , Electroencephalography , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/physiopathology , Female , Kindling, Neurologic , Mossy Fibers, Hippocampal/physiopathology , Rats , Seizures/pathology
8.
Neuroscience ; 170(1): 28-41, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20621164

ABSTRACT

Recent data support the involvement of the endocannabinoid signaling in early brain development, as well as a key role of cannabinoid receptors (CBR) in pathological conditions associated with unbalanced neuronal excitability and inflammation. Using immunocytochemistry, we explored the expression and cellular pattern of CBR 1 and 2 (CB1 and CB2) during prenatal human cortical development, as well as in focal malformations of cortical development associated with intractable epilepsy (focal cortical dysplasia; cortical tubers in patients with the tuberous sclerosis complex and glioneuronal tumors). Strong CB1 immunoreactivity was detected in the cortical plate in developing human brain from the earliest stages tested (gestational week 9) and it persisted throughout prenatal development. Both cannabinoid receptors were not detected in neural progenitor cells located in the ventricular zone. Only CB1 was expressed in the subventricular zone and in Cajal-Retzius cells in the molecular zone of the developing neocortex. CB2 was detected in cells of the microglia/macrophage lineage during development. In malformations of cortical development, prominent CB1 expression was demonstrated in dysplastic neurons. Both CBR were detected in balloon/giant cells, but CB2 appeared to be more frequently expressed than CB1 in these cell types. Reactive astrocytes were mainly stained with CB1, whereas cells of the microglia/macrophage lineage were stained with CB2. These findings confirm the early expression pattern of cannabinoid receptors in the developing human brain, suggesting a function for CB1 in the early stages of corticogenesis. The expression patterns in malformations of cortical development highlight the role of cannabinoid receptors as mediators of the endocannabinoid signaling and as potential pharmacological targets to modulate neuronal and glial cell function in epileptogenic developmental pathologies.


Subject(s)
Cerebral Cortex/growth & development , Epilepsy/metabolism , Epilepsy/pathology , Gene Expression Regulation, Developmental , Receptor, Cannabinoid, CB1/biosynthesis , Receptor, Cannabinoid, CB2/biosynthesis , Adult , Cell Lineage/genetics , Cerebral Cortex/embryology , Cerebral Cortex/pathology , Child , Child, Preschool , Epilepsy/genetics , Female , Humans , Infant , Infant, Newborn , Macrophages/cytology , Macrophages/metabolism , Male , Microglia/cytology , Microglia/metabolism , Middle Aged , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Young Adult
9.
Neuroscience ; 167(3): 929-45, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20219643

ABSTRACT

A growing body of evidence demonstrates the involvement of plasminogen activators (PAs) in a number of physiologic and pathologic events in the CNS. Induction of both tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA) has been observed in different experimental models of epilepsy and tPA has been implicated in the mechanisms underlying seizure activity. We investigated the expression and the cellular distribution of tPA and uPA in several epileptogenic pathologies, including hippocampal sclerosis (HS; n=6), and developmental glioneuronal lesions, such as focal cortical dysplasia (FCD, n=6), cortical tubers in patients with the tuberous sclerosis complex (TSC; n=6) and in gangliogliomas (GG; n=6), using immuno-cytochemical, western blot and real-time quantitative PCR analysis. TPA and uPA immunostaining showed increased expression within the epileptogenic lesions compared to control specimens in both glial and neuronal cells (hippocampal neurons in HS and dysplastic neurons in FCD, TSC and GG specimens). Confocal laser scanning microscopy confirmed expression of both proteins in astrocytes and microglia, as well as in microvascular endothelium. Immunoblot demonstrated also up-regulation of the uPA receptor (uPAR; P<0.05). Increased expression of tPA, uPA, uPAR and tissue PA inhibitor type mRNA levels was also detected by PCR analysis in different epileptogenic pathologies (P<0.05). Our data support the role of PA system components in different human focal epileptogenic pathologies, which may critically influence neuronal activity, inflammatory response, as well as contributing to the complex remodeling of the neuronal networks occurring in epileptogenic lesions.


Subject(s)
Brain Neoplasms/metabolism , Brain/metabolism , Epilepsy/metabolism , Nervous System Malformations/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Adolescent , Adult , Astrocytes/metabolism , Biomarkers/metabolism , Blotting, Western , Brain/abnormalities , Brain/pathology , Brain Neoplasms/complications , Brain Neoplasms/physiopathology , Child , Epilepsy/etiology , Epilepsy/physiopathology , Female , Ganglioglioma/complications , Ganglioglioma/metabolism , Ganglioglioma/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Immunohistochemistry , Male , Malformations of Cortical Development/complications , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/physiopathology , Microglia/metabolism , Middle Aged , Nervous System Malformations/complications , Nervous System Malformations/physiopathology , RNA, Messenger/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Plasminogen Activator/analysis , Tissue Plasminogen Activator/genetics , Tuberous Sclerosis/complications , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/physiopathology , Urokinase-Type Plasminogen Activator/analysis , Urokinase-Type Plasminogen Activator/genetics , Young Adult
10.
Eur J Neurosci ; 31(6): 1100-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20214679

ABSTRACT

Increasing evidence supports the involvement of inflammatory and immune processes in temporal lobe epilepsy (TLE). MicroRNAs (miRNA) represent small regulatory RNA molecules that have been shown to act as negative regulators of gene expression controlling different biological processes, including immune-system homeostasis and function. We investigated the expression and cellular distribution of miRNA-146a (miR-146a) in a rat model of TLE as well as in human TLE. miR-146a analysis in rat hippocampus was performed by polymerase chain reaction and immunocytochemistry at 1 week and 3-4 months after induction of status epilepticus (SE). Prominent upregulation of miR-146a activation was evident at 1 week after SE and persisted in the chronic phase. The miR-146a expression was confirmed to be present in reactive astrocytes. In human TLE with hippocampal sclerosis, increased astroglial expression of miR-146a was observed mainly in regions where neuronal cell loss and reactive gliosis occurred. The increased and persistent expression of miR-146a in reactive astrocytes supports the possible involvement of miRNAs in the modulation of the astroglial inflammatory response occurring in TLE and provides a target for future studies aimed at developing strategies against pro-epileptogenic inflammatory signalling.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Adult , Animals , Disease Models, Animal , Electric Stimulation/adverse effects , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/pathology , Female , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , MicroRNAs/genetics , Middle Aged , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Time Factors , Young Adult
11.
Neurobiol Dis ; 36(1): 81-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19596445

ABSTRACT

An increasing number of observations suggest an important role for voltage-gated potassium (Kv) channels in epilepsy. We studied the cell-specific distribution of Kv4.2, phosphorylated (p) Kv4.2 and the Kv4.2 interacting protein NCS-1 using immunocytochemistry in different epilepsy-associated focal lesions. In hippocampal sclerosis (HS), Kv4.2 and pKv4.2 immunoreactivity (IR) was reduced in the neuropil in regions with prominent neuronal cell loss. In both HS and malformations of cortical development (MCD), intense labeling was found in neuronal somata, but not in dendrites. Strong NCS-1 IR was observed in neurons in all lesion types. Western blot analysis demonstrated an increase of total Kv4.2 in all lesions and activation of the ERK pathway in HS and ganglioglioma. These findings indicate that Kv4.2 is expressed in both neuronal and glial cells and its regulation may involve potassium channel interacting proteins, alterations in the subcellular localization of the channel, as well as phosphorylation-mediated posttranslational modifications.


Subject(s)
Epilepsy/pathology , Hippocampus/metabolism , Hippocampus/pathology , Malformations of Cortical Development/metabolism , Shal Potassium Channels/metabolism , Adolescent , Adult , Animals , Child , Child, Preschool , Epilepsy/complications , Female , Humans , Male , Malformations of Cortical Development/complications , Malformations of Cortical Development/pathology , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Neurons/metabolism , Postmortem Changes , Rats , Sclerosis/complications , Sclerosis/pathology , Young Adult
12.
Epilepsy Res ; 84(1): 56-66, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19186029

ABSTRACT

Inflammation is an important biological process that is activated after status epilepticus and could be implicated in the development of epilepsy. Here we tested whether an anti-inflammatory treatment with a selective cox-2 inhibitor (SC58236) could prevent the development of epilepsy or modify seizure activity during the chronic epileptic phase. SC58236 was orally administered (10mg/kg) during the latent period for 7 days, starting 4h after electrically induced SE. Seizures were monitored using EEG/video monitoring until 35 days after SE. Cell death and inflammation were investigated using immunocytochemistry (NeuN and Ox-42). Sprouting was studied using Timm's staining after 1 week and after 4-5 months when rats were chronic epileptic. SC58236 was also administered during 5 days in chronic epileptic rats. Hippocampal EEG seizures were continuously monitored before, during and after treatment. SC58236 effectively reduced PGE(2) production but did not modify seizure development or the extent of cell death or microglia activation in the hippocampus. SC58236 treatment in chronic epileptic rats did not show any significant change in seizure duration or frequency of daily seizures. The fact that cox-2 inhibition, which effectively reduced prostaglandin levels, did not modify epileptogenesis or chronic seizure activity suggests that this type of treatment (starting after SE) will not provide an effective anti-epileptogenic or anti-epileptic therapy.


Subject(s)
Cyclooxygenase 2 Inhibitors/administration & dosage , Epilepsy, Temporal Lobe/prevention & control , Pyrazoles/administration & dosage , Seizures/drug therapy , Sulfonamides/administration & dosage , Adolescent , Adult , Animals , Brain/metabolism , CD11b Antigen/metabolism , Cell Death/drug effects , Cell Death/physiology , Cyclooxygenase 2/metabolism , Dinoprostone , Disease Models, Animal , Electroencephalography , Electroshock/adverse effects , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/physiopathology , Female , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/physiopathology , Humans , Inflammation/chemically induced , Inflammation/prevention & control , Male , Middle Aged , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Seizures/etiology , Seizures/pathology , Time Factors , Young Adult
13.
Neuroscience ; 156(1): 203-15, 2008 Sep 22.
Article in English | MEDLINE | ID: mdl-18706978

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disorder associated with cortical malformations (cortical tubers) and the development of glial tumors (subependymal giant-cell tumors, SGCTs). Expression of metabotropic glutamate receptor (mGluR) subtypes is developmentally regulated and several studies suggest an involvement of mGluR-mediated glutamate signaling in the regulation of proliferation and survival of neural stem-progenitor cells, as well as in the control of tumor growth. In the present study, we have investigated the expression and cell-specific distribution of group I (mGluR1, mGluR5), group II (mGluR2/3) and group III (mGluR4 and mGluR8) mGluR subtypes in human TSC specimens of both cortical tubers and SGCTs, using immunocytochemistry. Strong group I mGluR immunoreactivity (IR) was observed in the large majority of TSC specimens in dysplastic neurons and in giant cells within cortical tubers, as well as in tumor cells within SGCTs. In particular mGluR5 appeared to be most frequently expressed, whereas mGluR1alpha was detected in a subpopulation of neurons and giant cells. Cells expressing mGluR1alpha and mGluR5, demonstrate IR for phospho-S6 ribosomal protein (PS6), which is a marker of the mammalian target of rapamycin (mTOR) pathway activation. Group II and particularly group III mGluR IR was less frequently observed than group I mGluRs in dysplastic neurons and giant cells of tubers and tumor cells of SGCTs. Reactive astrocytes were mainly stained with mGluR5 and mGluR2/3. These findings expand our knowledge concerning the cellular phenotype in cortical tubers and in SGCTs and highlight the role of group I mGluRs as important mediators of glutamate signaling in TSC brain lesions. Individual mGluR subtypes may represent potential pharmacological targets for the treatment of the neurological manifestations associated with TSC brain lesions.


Subject(s)
Brain Neoplasms/metabolism , Cerebral Cortex/metabolism , Giant Cells/metabolism , Glioma, Subependymal/metabolism , Receptors, Metabotropic Glutamate/metabolism , Tuberous Sclerosis/metabolism , Adolescent , Adult , Astrocytes/metabolism , Astrocytes/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cerebral Cortex/pathology , Child , Child, Preschool , Female , Giant Cells/pathology , Glioma, Subependymal/pathology , Glutamic Acid/metabolism , Humans , Immunohistochemistry , Infant , Male , Neurons/metabolism , Neurons/pathology , Protein Kinases/metabolism , Receptor, Metabotropic Glutamate 5 , TOR Serine-Threonine Kinases , Tuberous Sclerosis/pathology , Young Adult
14.
Neuroscience ; 151(1): 272-92, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18093740

ABSTRACT

Gangliogliomas (GG) constitute the most frequent tumor entity in young patients undergoing surgery for intractable epilepsy. The histological composition of GG, with the presence of dysplastic neurons, corroborates their maldevelopmental origin. However, their histogenesis, the pathogenetic relationship with other developmental lesions, and the molecular alterations underlying the epileptogenicity of these tumors remain largely unknown. We performed gene expression analysis using the Affymetrix Gene Chip System (U133 plus 2.0 array). We used GENMAPP and the Gene Ontology database to identify global trends in gene expression data. Our analysis has identified various interesting genes and processes that are differentially expressed in GG when compared with normal tissue. The immune and inflammatory responses were the most prominent processes expressed in GG. Several genes involved in the complement pathway displayed a high level of expression compared with control expression levels. Higher expression was also observed for genes involved in cell adhesion, extracellular matrix and proliferation processes. We observed differential expression of genes as cyclin D1 and cyclin-dependent kinases, essential for neuronal cell cycle regulation and differentiation. Synaptic transmission, including GABA receptor signaling was an under-expressed process compared with control tissue. These data provide some suggestions for the molecular pathogenesis of GG. Furthermore, they indicate possible targets that may be investigated in order to dissect the mechanisms of epileptogenesis and possibly counteract the epileptogenic process in these developmental lesions.


Subject(s)
Brain Neoplasms/complications , Brain Neoplasms/genetics , Epilepsy/complications , Epilepsy/genetics , Ganglioglioma/complications , Ganglioglioma/genetics , Gene Expression Profiling , Adult , Cell Adhesion/drug effects , Complement System Proteins/biosynthesis , Complement System Proteins/genetics , DNA Primers , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Inflammation/pathology , Male , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Oligonucleotide Array Sequence Analysis , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Synaptic Transmission/physiology , Tissue Fixation , Wnt Proteins/biosynthesis , gamma-Aminobutyric Acid/physiology
15.
Neurobiol Dis ; 26(3): 497-511, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17412602

ABSTRACT

We investigated the involvement of the complement cascade during epileptogenesis in a rat model of temporal lobe epilepsy (TLE), and in the chronic epileptic phase in both experimental as well as human TLE. Previous rat gene expression analysis using microarrays indicated prominent activation of the classical complement pathway which peaked at 1 week after SE in CA3 and entorhinal cortex. Increased expression of C1q, C3 and C4 was confirmed in CA3 tissue using quantitative PCR at 1 day, 1 week and 3-4 months after status epilepticus (SE). Upregulation of C1q and C3d protein expression was confirmed mainly to be present in microglia and in a few hippocampal neurons. In human TLE with hippocampal sclerosis, astroglial, microglial and neuronal (5/8 cases) expression of C1q, C3c and C3d was observed particularly within regions where neuronal cell loss occurs. The membrane attack protein complex (C5b-C9) was predominantly detected in activated microglial cells. The persistence of complement activation could contribute to a sustained inflammatory response and could destabilize neuronal networks involved.


Subject(s)
Complement System Proteins/immunology , Encephalitis/immunology , Epilepsy, Temporal Lobe/immunology , Gliosis/immunology , Hippocampus/immunology , Up-Regulation/immunology , Adolescent , Adult , Aged , Animals , Astrocytes/immunology , Astrocytes/metabolism , Complement C1q/genetics , Complement C1q/immunology , Complement C1q/metabolism , Complement C3c/genetics , Complement C3c/immunology , Complement C3c/metabolism , Complement C3d/genetics , Complement C3d/immunology , Complement C3d/metabolism , Complement C5b/genetics , Complement C5b/immunology , Complement C5b/metabolism , Complement System Proteins/genetics , Complement System Proteins/metabolism , Disease Models, Animal , Encephalitis/genetics , Encephalitis/physiopathology , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/physiopathology , Female , Gliosis/genetics , Gliosis/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Male , Microglia/immunology , Microglia/metabolism , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Status Epilepticus/genetics , Status Epilepticus/immunology , Status Epilepticus/physiopathology , Up-Regulation/genetics
16.
J Pharmacol Exp Ther ; 322(1): 141-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17392402

ABSTRACT

Recent studies have suggested that overexpression of the multidrug transporter P-glycoprotein (P-gp) in the hippocampal region leads to decreased levels of antiepileptic drugs and contributes to pharmacoresistance that occurs in a subset of epileptic patients. Whether P-gp expression and function is affected in other brain regions and in organs that are involved in drug metabolism is less studied. Therefore, we investigated P-gp expression in different brain regions and liver of chronic epileptic rats, several months after electrically induced status epilepticus (SE), using Western blot analysis. P-gp function was determined by measuring phenytoin (PHT) levels in these brain regions using high-performance liquid chromatography, in the absence and presence of a P-gp-specific inhibitor, tariquidar (TQD). In addition, the pharmacokinetic profile of PHT was determined. PHT concentration was reduced by 20 to 30% in brain regions that had P-gp overexpression (temporal hippocampus and parahippocampal cortex) and not in brain regions in which P-gp expression was not changed after SE. Inhibition of P-gp by TQD significantly increased the PHT concentration, specifically in regions that showed P-gp overexpression. Despite increased P-gp expression in the liver of epileptic rats, pharmacokinetic analysis showed no significant change of PHT clearance in control versus epileptic rats. These findings show that overexpression of P-gp at the blood-brain barrier of specific limbic brain regions causes a decrease of local PHT levels in the rat brain.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Anticonvulsants/pharmacokinetics , Blood-Brain Barrier , Brain/metabolism , Epilepsy/drug therapy , Phenytoin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Epilepsy/metabolism , Male , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley
17.
Epilepsy Res ; 74(1): 33-44, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17267178

ABSTRACT

Developmental glioneuronal lesions, such as gangliogliomas (GG) are increasingly recognized causes of chronic pharmaco-resistant epilepsy. It has been postulated that chronic epilepsy in patients with malformations of cortical development is associated with dysfunction of the inhibitory GABA-ergic system. We aimed to identify the subtypes of interneurons present within GG specimens and the expression and cellular distribution patterns of GABA receptors (GABAR) and GABA transporter 1 (GAT1). The expression of the various components of the GABA-ergic system were also analyzed in the perilesional cortex. We investigated the expression of parvalbumin, calbindin, calretinin, GABA(A)R (a1 subunit)(,) GABA(B) (R1 and R2) and GAT-1 using immunocytochemistry in 30 specimens of GG obtained during epilepsy surgery, including 10 cases with sufficient amount of perilesional cortex. Immunocytochemistry for calbindin (CB), calretinin (CR) and parvalbumin (PV) demonstrate the presence of inhibitory neurons of different subtypes within the GG specimens. Calcium-binding protein-positive interneurons represent a small fraction of the total neuronal population. Both GABA(A)R and GABA(B)R (R1 and R2) subtypes were detected within the neuronal component of GG specimens. In addition, GABA(B)R2 immunoreactivity (IR) was observed in glial cells. GG specimens displayed also expression of GAT-1 IR. Compared to normal cortex, the density of PV- and CB-immunoreactive interneurons was reduced in the perilesional cortex of GG patients, whereas CR-labeling was similar to that observed in normal cortex. GAT-1 IR was also significantly reduced in the perilesional specimens. The cellular distribution of components of the GABA-ergic system in GG, together with the perilesional changes suggest that alterations of the GABA-ergic system may contribute to the complex abnormal functional network of these highly epileptogenic developmental lesions.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Ganglioglioma/metabolism , Interneurons/metabolism , Proteins/metabolism , Adolescent , Adult , Calbindin 2 , Calbindins , Cerebral Cortex/pathology , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/pathology , Female , GABA Plasma Membrane Transport Proteins/metabolism , Ganglioglioma/complications , Humans , Immunohistochemistry , Male , Middle Aged , Parvalbumins/metabolism , Receptors, GABA-A/metabolism , Receptors, GABA-B/metabolism , S100 Calcium Binding Protein G/metabolism
18.
Neuroscience ; 145(1): 185-96, 2007 Mar 02.
Article in English | MEDLINE | ID: mdl-17207578

ABSTRACT

Malformations of cortical development are recognized causes of chronic medically intractable epilepsy. An increasing number of observations suggests an important role for cation-chloride co-transporters (CCTs) in controlling neuronal function. Deregulation of their expression may contribute to the mechanisms of hyperexcitability that lead to seizures. In the present study the expression and cell-specific distribution of Na+-K+-2Cl--cotransporter (NKCC1) and K+-Cl--cotransporter (KCC2) were studied immunocytochemically in different developmental lesions, including focal cortical dysplasia (FCD) type IIB (n=9), hemimegalencephaly (HMEG, n=6) and ganglioglioma (GG, n=9) from patients with medically intractable epilepsy and in age-matched controls. In normal control adult cortex, NKCC1 displayed low neuronal and glial expression levels. In contrast KCC2 showed strong and diffuse neuropil staining. Notable glial immunoreactivity (IR) was not found for KCC2. NKCC1 was highly expressed in the majority of FCD, HMEG and GG specimens. NKCC1 IR was observed in neurons of different size, including large dysplastic neurons, in balloon cells (in FCD and HMEG cases) and in glial cells with astrocytic morphology. The immunoreactivity pattern of KCC2 in FCD, HMEG and GG specimens was characterized by less neuropil staining and more intrasomatic IR compared with control. KCC2 IR was observed in neurons of different size, including large dysplastic neurons, but not in balloon cells or in glial cells with astrocytic morphology. Double-labeling experiments confirmed the differential cellular distribution of the two CCTs and their expression in GABA(A) receptor (alpha1 subunit)-positive dysplastic neurons. The cellular distribution of CCTs, with high expression of NKCC1 in dysplastic neurons and altered subcellular distribution of KCC2 resembles that of immature cortex and suggests a possible contribution of CCTs to the high epileptogenicity of malformations of cortical development.


Subject(s)
Cerebral Cortex , Epilepsy/pathology , Gene Expression Regulation, Developmental/physiology , Sodium-Potassium-Chloride Symporters/metabolism , Symporters/metabolism , Adolescent , Adult , Cerebral Cortex/abnormalities , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/pathology , Child , Child, Preschool , Epilepsy/etiology , Female , Ganglioglioma/complications , Ganglioglioma/pathology , Humans , Immunohistochemistry/methods , Infant , Male , Nerve Tissue Proteins/metabolism , Postmortem Changes , Solute Carrier Family 12, Member 2 , K Cl- Cotransporters
19.
Brain ; 130(Pt 2): 521-34, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17124188

ABSTRACT

Leakage of the blood-brain barrier (BBB) is associated with various neurological disorders, including temporal lobe epilepsy (TLE). However, it is not known whether alterations of the BBB occur during epileptogenesis and whether this can affect progression of epilepsy. We used both human and rat epileptic brain tissue and determined BBB permeability using various tracers and albumin immunocytochemistry. In addition, we studied the possible consequences of BBB opening in the rat for the subsequent progression of TLE. Albumin extravasation in human was prominent after status epilepticus (SE) in astrocytes and neurons, and also in hippocampus of TLE patients. Similarly, albumin and tracers were found in microglia, astrocytes and neurons of the rat. The BBB was permeable in rat limbic brain regions shortly after SE, but also in the latent and chronic epileptic phase. BBB permeability was positively correlated to seizure frequency in chronic epileptic rats. Artificial opening of the BBB by mannitol in the chronic epileptic phase induced a persistent increase in the number of seizures in the majority of rats. These findings indicate that BBB leakage occurs during epileptogenesis and the chronic epileptic phase and suggest that this can contribute to the progression of epilepsy.


Subject(s)
Blood-Brain Barrier/physiopathology , Epilepsy, Temporal Lobe/physiopathology , Acute Disease , Adolescent , Adult , Albumins , Animals , Brain/metabolism , Chronic Disease , Coloring Agents , Disease Progression , Electroencephalography/methods , Epilepsy, Temporal Lobe/metabolism , Evans Blue , Fluoresceins , Humans , Male , Middle Aged , Organic Chemicals , Permeability , Rats , Rats, Sprague-Dawley , Status Epilepticus/physiopathology
20.
Neuroscience ; 138(2): 457-74, 2006.
Article in English | MEDLINE | ID: mdl-16413123

ABSTRACT

Hippocampal sclerosis constitutes the most frequent neuropathological finding in patients with medically intractable mesial temporal lobe epilepsy. Serial analysis of gene expression was used to get a global view of the gene profile in human hippocampus in control condition and in epileptic condition associated with hippocampal sclerosis. Libraries were generated from control hippocampus, obtained by rapid autopsy, and from hippocampal surgical specimens of patients with mesial temporal lobe epilepsy and the classical pattern of hippocampal sclerosis. More than 50,000 tags were analyzed (28,282, control hippocampus; 25,953, hippocampal sclerosis) resulting in 9206 (control hippocampus) and 9599 (hippocampal sclerosis) unique tags (genes), each representing a specific mRNA transcript. Comparison of the two libraries resulted in the identification of 143 transcripts that were differentially expressed. These genes belong to a variety of functional classes, including basic metabolism, transcription regulation, protein synthesis and degradation, signal transduction, structural proteins, regeneration and synaptic plasticity and genes of unknown identity of function. The database generated by this study provides an extensive inventory of genes expressed in human control hippocampus, identifies new high-abundant genes associated with altered hippocampal morphology in patients with mesial temporal lobe epilepsy and serves as a reference for future studies aimed at detecting hippocampal transcriptional responses under various pathological conditions.


Subject(s)
Epilepsy, Temporal Lobe/genetics , Gene Expression Regulation , Hippocampus/physiopathology , Base Sequence , DNA Primers , Enzymes/genetics , Expressed Sequence Tags , Hippocampus/pathology , Humans , Nerve Regeneration/genetics , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , RNA/genetics , RNA/isolation & purification , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...