Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Neuroscience ; 306: 18-27, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26283024

ABSTRACT

INTRODUCTION: Neuro-vascular rearrangement occurs in brain disorders, including epilepsy. Platelet-derived growth factor receptor beta (PDGFRß) is used as a marker of perivascular pericytes. Whether PDGFRß(+) cell reorganization occurs in regions of neuro-vascular dysplasia associated with seizures is unknown. METHODS: We used brain specimens derived from epileptic subjects affected by intractable seizures associated with focal cortical dysplasia (FCD) or temporal lobe epilepsy with hippocampal sclerosis (TLE-HS). Tissues from cryptogenic epilepsy, non-sclerotic hippocampi or peritumoral were used for comparison. An in vivo rat model of neuro-vascular dysplasia was obtained by pre-natal exposure to methyl-axozy methanoic acid (MAM). Status epilepticus (SE) was induced in adult MAM rats by intraperitoneal pilocarpine. MAM tissues were also used to establish organotypic hippocampal cultures (OHC) to further assess pericytes positioning at the dysplastic microvasculature. PDGFRß and its colocalization with RECA-1 or CD34 were used to segregate perivascular pericytes. PDGFRß and NG2 or IBA1 colocalization were performed. Rat cortices and hippocampi were used for PDGFRß western blot analysis. RESULTS: Human FCD displayed the highest perivascular PDGFRß immunoreactivity, indicating pericytes, and presence of ramified PDGFRß(+) cells in the parenchyma and proximal to microvessels. Tissues deriving from human cryptogenic epilepsy displayed a similar pattern of immunoreactivity, although to a lesser extent compared to FCD. In TLE-HS, CD34 vascular proliferation was paralleled by increased perivascular PDGFRß(+) pericytes, as compared to non-HS. Parenchymal PDGFRß immunoreactivity co-localized with NG2 but was distinct from IBA1(+) microglia. In MAM rats, we found pericyte-vascular changes in regions characterized by neuronal heterotopias. PDGFRß immunoreactivity was differentially distributed in the heterotopic and adjacent normal CA1 region. The use of MAM OHC revealed microvascular-pericyte dysplasia at the capillary tree lining the dentate gyrus (DG) molecular layer as compared to control OHC. Severe SE induced PDGFRß(+) immunoreactivity mostly in the CA1 region of MAM rats. CONCLUSION: Our descriptive study points to microvascular-pericyte changes in the epileptic pathology. The possible link between PDGFRß(+) cells, neuro-vascular dysplasia and remodeling during seizures is discussed.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Epilepsy, Temporal Lobe/pathology , Malformations of Cortical Development/pathology , Pericytes/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Adolescent , Adult , Animals , Calcium-Binding Proteins , Cerebral Cortex/abnormalities , Cerebral Cortex/metabolism , Child , Child, Preschool , DNA-Binding Proteins/metabolism , Disease Models, Animal , Epilepsy, Temporal Lobe/complications , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Infant , Malformations of Cortical Development/complications , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/physiopathology , Microfilament Proteins , Pericytes/metabolism , Rats , Rats, Sprague-Dawley , Seizures/complications , Young Adult
2.
Neuroscience ; 281: 124-34, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25280786

ABSTRACT

P450 metabolic enzymes are expressed in the human and rodent brain. Recent data support their involvement in the pathophysiology of epilepsy. However, the determinants of metabolic enzyme expression in the epileptic brain are unclear. We tested the hypothesis that status epilepticus (SE) or exposure to phenytoin or phenobarbital affects brain expression of the metabolic enzyme CYP2E1. SE was induced in C57BL/6J mice by systemic kainic acid. Brain CYP2E1 expression was evaluated 18-24h after severe SE by immunohistochemistry. Co-localization with neuronal nuclei (NEUN), glial fibrillary acidic protein (GFAP) and CD31 was determined by confocal microscopy. The effect of phenytoin, carbamazepine and phenobarbital on CYP2E1 expression was evaluated in vivo or by using organotypic hippocampal cultures in vitro. CYP2E1 expression was investigated in brain resections from a cohort of drug-resistant epileptic brain resections and human endothelial cultures (EPI-EC). Immunohistochemistry showed an increase of CYP2E1 expression limited to hippocampal CA2/3 and hilar neurons after severe SE in mice. CYP2E1 expression was also observed at the astrocyte-vascular interface. Analysis of human brain specimens revealed CYP2E1 expression in neurons and vascular endothelial cells (EC). CYP2E1 was expressed in cultured human EC and over-expressed by EPI-EC. When analyzing the effect of drug exposure on CYP2E1 expression we found that, in vivo or in vitro, ethanol increased CYP2E1 levels in the brain and liver. Treatment with phenytoin induced localized CYP2E1 expression in the brain whereas no significant effects were exerted by carbamazepine or phenobarbital. Our data indicate that the effect of acute SE on brain CYP2E1 expression is localized and cell specific. Exposure to selected anti-epileptic drugs could play a role in determining CYP2E1 brain expression. Additional investigation is required to fully reproduce the culprits of P450 enzyme expression as observed in the human epileptic brain.


Subject(s)
Anticonvulsants/pharmacology , Brain/metabolism , Central Nervous System Depressants/pharmacology , Cytochrome P-450 CYP2E1/metabolism , Endothelial Cells/metabolism , Ethanol/pharmacology , Neurons/metabolism , Phenytoin/pharmacology , Status Epilepticus/metabolism , Adolescent , Adult , Animals , Brain/drug effects , Carbamazepine/pharmacology , Cells, Cultured , Child, Preschool , Cytochrome P-450 CYP2E1/drug effects , Disease Models, Animal , Female , Humans , Infant , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenobarbital/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...