Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 376: 114759, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38519010

ABSTRACT

Malformations of cortical development (MCDs) are caused by abnormal neuronal migration processes during the fetal period and are a major cause of intractable epilepsy in infancy. However, the timing of hyperexcitability or epileptogenesis in MCDs remains unclear. To identify the early developmental changes in the brain of the MCD rat model, which exhibits increased seizure susceptibility during infancy (P12-15), we analyzed the pathological changes in the brains of MCD model rats during the neonatal period and tested NMDA-induced seizure susceptibility. Pregnant rats were injected with two doses of methylazoxymethanol acetate (MAM, 15 mg/kg, i.p.) to induce MCD, while controls were administered normal saline. The cortical development of the offspring was measured by performing magnetic resonance imaging (MRI) on postnatal days (P) 1, 5, and 8. At P8, some rats were sacrificed for immunofluorescence, Golgi staining, and Western analysis. In another set of rats, the number and latency to onset of spasms were monitored for 90 min after the NMDA (5 mg/kg i.p.) injection at P8. In MCD rats, in vivo MR imaging showed smaller brain volume and thinner cortex from day 1 after birth (p < 0.001). Golgi staining and immunofluorescence revealed abnormal neuronal migration, with a reduced number of neuronal cell populations and less dendritic arborization at P8. Furthermore, MCD rats exhibited a significant reduction in the expression of NMDA receptors and AMPAR4, along with an increase in AMPAR3 expression (p < 0.05). Although there was no difference in the latency to seizure onset between MCD rats and controls, the MCD rats survived significantly longer than the controls. These results provide insights into the early developmental changes in the cortex of a MCD rat model and suggest that delayed and abnormal neuronal development in the immature brain is associated with a blunted response to NMDA-induced excitotoxic injury. These developmental changes may be involved in the sudden onset of epilepsy in patients with MCD or prenatal brain injury.


Subject(s)
Cell Movement , Disease Models, Animal , Malformations of Cortical Development , N-Methylaspartate , Neurons , Rats, Sprague-Dawley , Animals , Rats , N-Methylaspartate/toxicity , Female , Pregnancy , Cell Movement/drug effects , Neurons/pathology , Neurons/drug effects , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/pathology , Animals, Newborn , Methylazoxymethanol Acetate/toxicity , Methylazoxymethanol Acetate/analogs & derivatives , Cerebral Cortex/pathology , Cerebral Cortex/drug effects , Male , Magnetic Resonance Imaging
2.
Cells ; 10(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201497

ABSTRACT

Cortical dysplasias are alterations in the organization of the layers of the brain cortex due to problems in neuronal migration during development. The neuronal component has been widely studied in experimental models of cortical dysplasias. In contrast, little is known about how glia are affected. In the cerebellum, Bergmann glia (BG) are essential for neuronal migration during development, and in adult they mediate the control of fine movements through glutamatergic transmission. The aim of this study was to characterize the morphology and intracellular calcium dynamics of BG and astrocytes from mouse cerebellum and their modifications in a model of cortical dysplasia induced by carmustine (BCNU). Carmustine-treated mice were affected in their motor coordination and balance. Cerebellar dysplasias and heterotopias were more frequently found in lobule X. Morphology of BG cells and astrocytes was affected, as were their spontaneous [Ca2+]i transients in slice preparation and in vitro.


Subject(s)
Calcium Signaling , Cerebellum/pathology , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Neuroglia/metabolism , Neuroglia/pathology , Animals , Astrocytes/pathology , Carmustine , Cells, Cultured , Malformations of Cortical Development/chemically induced , Mice, Transgenic , Motor Activity
3.
Int J Dev Neurosci ; 68: 1-9, 2018 08.
Article in English | MEDLINE | ID: mdl-29605566

ABSTRACT

BACKGROUND: Melatonin, which is an antioxidant and neuroprotective agent, can be an effective treatment for neurological disorders. We assessed the effect of melatonin administration on histological changes, antioxidant enzyme levels, and behavioral changes in a neonate mouse model of cortical malformation. MATERIALS AND METHODS: Cortical malformation was induced by two injections of 15 mg/kg methylazoxymethanol (MAM) on gestational day 15 (E15). Pregnant Balb/c mice were randomly divided into the following six groups: Control (CO), Melatonin (MEL), Luzindole (LUZ), MAM, MEL + MAM1 (co-treatment), and MEL + MAM2 (pretreatment). Melatonin was intraperitoneally injected at a dose of 10 mg/kg daily (from E15 until delivery of from E6 for 20 days after delivery). On postnatal day 31, the activity and anxiety of mice were assessed by open field and elevated plus maze tests, respectively. Histopathological changes in the neonate cortex were studied using hematoxylin and eosin staining and neurofilament immunohistochemistry. Enzyme-linked immunosorbent assays were used to measure the activity of nitric oxide (NO), malondialdehyde (MDA), and antioxidant enzymes, including catalase (CAT), super oxide dismutase (SOD), and glutathione peroxidase (GPX). RESULTS: In the behavioral assessment of neonate mice, a significant increase in the crossing activity and decrease in anxiety were recorded in groups treated with MAM plus melatonin. In histological examination, heterotopic, dysmorphic, and ectopic cells, as well as dyslamination, were seen in the MAM and LUZ groups. However, these defects were attenuated in the MAM plus melatonin groups. Significant reductions were recorded in the SOD and GPX levels in the MAM and LUZ groups compared to the control, while the NO level was increased in these groups. Groups that received MAM plus melatonin showed significant increases in the levels of SOD and GPX and a significant decrease in the level of NO, compared to the MAM group. CONCLUSION: Melatonin increased the crossing activity and decreased the anxiety in the treated mice of the neonate mouse model of cortical malformation. Histologically, the administration of exogenous melatonin in pregnant mice and their neonates had a protective effect on the cerebral cortex of neonates. Also, this effect is elicited by decreasing NO and increasing antioxidative enzymes.


Subject(s)
Antioxidants/therapeutic use , Exploratory Behavior/drug effects , Gene Expression Regulation, Developmental/drug effects , Malformations of Cortical Development/complications , Malformations of Cortical Development/drug therapy , Melatonin/therapeutic use , Animals , Animals, Newborn , Carcinogens/toxicity , Catalase/metabolism , Disease Models, Animal , Female , Glutathione Peroxidase/metabolism , Intermediate Filaments/metabolism , Malformations of Cortical Development/chemically induced , Malondialdehyde/metabolism , Maze Learning/drug effects , Methylazoxymethanol Acetate/analogs & derivatives , Methylazoxymethanol Acetate/toxicity , Mice , Mice, Inbred BALB C , Nitroprusside/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Superoxide Dismutase/metabolism , Tryptamines/toxicity
4.
Neurochem Res ; 43(4): 878-885, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29468458

ABSTRACT

Cortical dysplasia is the most common etiology of intractable epilepsy. Both excitability changes in cortical neurons and neural network reconstitution play a role in cortical dysplasia epileptogenesis. Recent research shows that the axon initial segment, a subcompartment of the neuron important to the shaping of action potentials, adjusts its position in response to changes in input, which contributes to neuronal excitability and local circuit balance. It is unknown whether axon initial segment plasticity occurs in neurons involved in seizure susceptibility in cortical dysplasia. Here, we developed a "Carmustine"- "pilocarpine" rat model of cortical dysplasia and show that it exhibits a lower seizure threshold, as indicated by behavior studies and electroencephalogram monitoring. Using immunofluorescence, we measured the axon initial segment positions of deep L5 somatosensory neurons and show that it is positioned closer to the soma after acute seizure, and that this displacement is sustained in the chronic phase. We then show that Nifedipine has a dose-dependent protective effect against axon initial segment displacement and increased seizure susceptibility. These findings further our understanding of the pathophysiology of seizures in cortical dysplasia and suggests Nifedipine as a potential therapeutic agent.


Subject(s)
Axon Initial Segment/physiology , Disease Models, Animal , Malformations of Cortical Development/physiopathology , Neuronal Plasticity/physiology , Seizures/physiopathology , Animals , Axons/drug effects , Axons/physiology , Disease Susceptibility/chemically induced , Disease Susceptibility/diet therapy , Disease Susceptibility/physiopathology , Electroencephalography/drug effects , Electroencephalography/methods , Female , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/drug therapy , Nifedipine/pharmacology , Nifedipine/therapeutic use , Pilocarpine/toxicity , Pregnancy , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/drug therapy
5.
Toxicol Sci ; 160(1): 57-73, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28973696

ABSTRACT

Adequate levels of thyroid hormone (TH) are needed for proper brain development, deficiencies may lead to adverse neurologic outcomes in humans and animal models. Environmental chemicals have been linked to TH disruption, yet the relationship between developmental exposures and decline in serum TH resulting in neurodevelopmental impairment is poorly understood. The present study developed a quantitative adverse outcome pathway where serum thyroxin (T4) reduction following inhibition of thyroperoxidase in the thyroid gland are described and related to deficits in fetal brain TH and the development of a brain malformation, cortical heterotopia. Pregnant rats were exposed to 6-propylthiouracil (PTU 0, 0.1, 0.5, 1, 2, or 3 parts per million [ppm]) from gestational days 6-20, sequentially increasing PTU concentrations in maternal thyroid gland and serum as well as in fetal serum. Dams exposed to 0.5 ppm PTU and higher exhibited dose-dependent decreases in thyroidal T4. Serum T4 levels in the dam were significantly decreased with exposure to 2 and 3 ppm PTU. In the fetus, T4 decrements were first observed at a lower dose of 0.5 ppm PTU. Based on these data, fetal brain T4 levels were estimated from published literature sources, and quantitatively linked to increases in the size of the heterotopia present in the brains of offspring. These data show the potential of in vivo assessments and computational descriptions of biologic responses to predict the development of this structural brain malformation and use of quantitative adverse outcome pathway approach to evaluate brain deficits that may result from exposure to other TH disruptors.


Subject(s)
Adverse Outcome Pathways , Brain/drug effects , Endocrine Disruptors/toxicity , Enzyme Inhibitors/toxicity , Iodide Peroxidase/antagonists & inhibitors , Malformations of Cortical Development/chemically induced , Prenatal Exposure Delayed Effects , Propylthiouracil/toxicity , Thyroid Gland/drug effects , Thyroxine/biosynthesis , Animals , Biomarkers/blood , Brain/abnormalities , Brain/metabolism , Computer Simulation , Dose-Response Relationship, Drug , Female , Gestational Age , Iodide Peroxidase/metabolism , Malformations of Cortical Development/enzymology , Maternal Exposure/adverse effects , Pregnancy , Rats, Long-Evans , Thyroid Gland/enzymology , Thyroxine/blood , Time Factors
6.
J Neurosci Methods ; 260: 62-72, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-25850077

ABSTRACT

Pharmaco-resistant epilepsies, and also some neuropsychiatric disorders, are often associated with malformations in hippocampal and neocortical structures. The mechanisms leading to these cortical malformations causing an imbalance between the excitatory and inhibitory system are largely unknown. Animal models using chemical or physical manipulations reproduce different human pathologies by interfering with cell generation and neuronal migration. The model of in utero injection of methylazoxymethanol (MAM) acetate mimics periventricular nodular heterotopia. The freeze lesion model reproduces (poly)microgyria, focal heterotopia and schizencephaly. The in utero irradiation model causes microgyria and heterotopia. Intraperitoneal injections of carmustine 1-3-bis-chloroethyl-nitrosurea (BCNU) to pregnant rats produces laminar disorganization, heterotopias and cytomegalic neurons. The ibotenic acid model induces focal cortical malformations, which resemble human microgyria and ulegyria. Cortical dysplasia can be also observed following prenatal exposure to ethanol, cocaine or antiepileptic drugs. All these models of cortical malformations are characterized by a pronounced hyperexcitability, few of them also produce spontaneous epileptic seizures. This dysfunction results from an impairment in GABAergic inhibition and/or an increase in glutamatergic synaptic transmission. The cortical region initiating or contributing to this hyperexcitability may not necessarily correspond to the site of the focal malformation. In some models wide-spread molecular and functional changes can be observed in remote regions of the brain, where they cause pathophysiological activities. This paper gives an overview on different animal models of cortical malformations, which are mostly used in rodents and which mimic the pathology and to some extent the pathophysiology of neuronal migration disorders associated with epilepsy in humans.


Subject(s)
Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/physiopathology , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/physiopathology , Teratogens , Animals , Cerebral Cortex/abnormalities , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Freezing , Rodentia
7.
PLoS One ; 9(2): e89898, 2014.
Article in English | MEDLINE | ID: mdl-24587109

ABSTRACT

Whether severe epilepsy could be a progressive disorder remains as yet unresolved. We previously demonstrated in a rat model of acquired focal cortical dysplasia, the methylazoxymethanol/pilocarpine - MAM/pilocarpine - rats, that the occurrence of status epilepticus (SE) and subsequent seizures fostered a pathologic process capable of modifying the morphology of cortical pyramidal neurons and NMDA receptor expression/localization. We have here extended our analysis by evaluating neocortical and hippocampal changes in MAM/pilocarpine rats at different epilepsy stages, from few days after onset up to six months of chronic epilepsy. Our findings indicate that the process triggered by SE and subsequent seizures in the malformed brain i) is steadily progressive, deeply altering neocortical and hippocampal morphology, with atrophy of neocortex and CA regions and progressive increase of granule cell layer dispersion; ii) changes dramatically the fine morphology of neurons in neocortex and hippocampus, by increasing cell size and decreasing both dendrite arborization and spine density; iii) induces reorganization of glutamatergic and GABAergic networks in both neocortex and hippocampus, favoring excitatory vs inhibitory input; iv) activates NMDA regulatory subunits. Taken together, our data indicate that, at least in experimental models of brain malformations, severe seizure activity, i.e., SE plus recurrent seizures, may lead to a widespread, steadily progressive architectural, neuronal and synaptic reorganization in the brain. They also suggest the mechanistic relevance of glutamate/NMDA hyper-activation in the seizure-related brain pathologic plasticity.


Subject(s)
Epilepsies, Partial/etiology , Epilepsies, Partial/pathology , Malformations of Cortical Development/complications , N-Methylaspartate/metabolism , Synapses/pathology , Animals , Atrophy , Cerebral Cortex/pathology , Disease Models, Animal , Female , Hippocampus/pathology , Malformations of Cortical Development/chemically induced , Neocortex/pathology , Pregnancy , Pyramidal Cells/pathology , Rats , Receptors, N-Methyl-D-Aspartate/metabolism
8.
J Mol Neurosci ; 52(4): 586-97, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24057922

ABSTRACT

Microgyria is associated with epilepsy and due to developmental disruption of neuronal migration. However, the role of endogenous subventricular zone-derived neural progenitors (SDNPs) in formation and hyperexcitability has not been fully elucidated. Here, we establish a neonatal cortex freeze-lesion (FL) model, which was considered as a model for focal microgyria, and simultaneously label SDNPs by CM-DiI. Morphological investigation showed that SDNPs migrated into FL and differentiated into neuronal and glia cell types, suggesting the involvement of endogenous SDNPs in the formation of FL-induced microgyria. Patch-clamp recordings in CM-DiI positive (CM-DiI(+)) pyramidal neurons within FL indicated an increase in frequency of spontaneous action potentials, while the resting membrane potential did not differ from the controls. We also found that spontaneous excitatory postsynaptic currents (EPSCs) increased in frequency but not in amplitude compared with controls. The evoked EPSCs showed a significant increase of 10-90% in rise time and decay time in the CM-DiI(+) neurons. Moreover, paired-pulse facilitation was dramatically larger in CM-DiI(+) pyramidal neurons. Western blotting data showed that AMPA and NMDA receptors were increased to some extent in the FL cortex compared with controls, and the NMDA/AMPA ratio of eEPSCs at CM-DiI(+) pyramidal neurons was significantly increased. Taken together, our findings provide novel evidence for the contribution of endogenous SDNPs in the formation and epileptogenicity of FL-induced focal microgyria.


Subject(s)
Malformations of Cortical Development/pathology , Malformations of Cortical Development/physiopathology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stem Cell Niche/physiology , Action Potentials/physiology , Animals , Animals, Newborn , Carbocyanines/toxicity , Cell Movement/physiology , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Injections, Intraventricular , Malformations of Cortical Development/chemically induced , Organ Culture Techniques , Patch-Clamp Techniques , Pyramidal Cells/pathology , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/physiology
9.
Cereb Cortex ; 24(4): 1088-101, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23246779

ABSTRACT

Although focal cortical malformations are considered neuronal migration disorders, their formation mechanisms remain unknown. We addressed how the γ-aminobutyric acid (GABA)ergic system affects the GABAergic and glutamatergic neuronal migration underlying such malformations. A focal freeze-lesion (FFL) of the postnatal day zero (P0) glutamic acid decarboxylase-green fluorescent protein knock-in mouse neocortex produced a 3- or 4-layered microgyrus at P7. GABAergic interneurons accumulated around the necrosis including the superficial region during microgyrus formation at P4, whereas E17.5-born, Cux1-positive pyramidal neurons outlined the GABAergic neurons and were absent from the superficial layer, forming cell-dense areas in layer 2 of the P7 microgyrus. GABA imaging showed that an extracellular GABA level temporally increased in the GABAergic neuron-positive area, including the necrotic center, at P4. The expression of the Cl(-) transporter KCC2 was downregulated in the microgyrus-forming GABAergic and E17.5-born glutamatergic neurons at P4; these cells may need a high intracellular Cl(-) concentration to induce depolarizing GABA effects. Bicuculline decreased the frequency of spontaneous Ca(2+) oscillations in these microgyrus-forming cells. Thus, neonatal FFL causes specific neuronal accumulation, preceded by an increase in ambient GABA during microgyrus formation. This GABA increase induces GABAA receptor-mediated Ca(2+) oscillation in KCC2-downregulated microgyrus-forming cells, as seen in migrating cells during early neocortical development.


Subject(s)
Down-Regulation/physiology , GABAergic Neurons/pathology , Malformations of Cortical Development/metabolism , Malformations of Cortical Development/pathology , Symporters/metabolism , gamma-Aminobutyric Acid/metabolism , Age Factors , Animals , Animals, Newborn , Bicuculline/pharmacology , Cell Count , Cerebral Cortex/pathology , Disease Models, Animal , Embryo, Mammalian , Female , GABA-A Receptor Antagonists/pharmacology , Glutamate Decarboxylase/genetics , Male , Malformations of Cortical Development/chemically induced , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitrogen/toxicity , Symporters/genetics , K Cl- Cotransporters
10.
J Neurophysiol ; 111(2): 323-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155012

ABSTRACT

Cortical dysplasia (CD) associates with clinical pathologies, including epilepsy and mental retardation. CD results from impaired migration of immature neurons to their cortical targets, leading to clustering of neural cells and changes in cortical properties. We developed a CD model by administering methylazoxymethanol (MAM), an anti-mitotic, to pregnant ferrets on embryonic day 33; this leads to reduction in cortical thickness in addition to redistribution and increased expression of GABAA receptors (GABAAR). We evaluated the impact of MAM treatment on GABAAR-mediated synaptic transmission in postnatal day 0-1 neurons, leaving the ganglionic eminence (GE) and in layer 2/3 pyramidal cells of postnatal day 28-38 ferrets. Embryonic day 33 MAM treatment significantly increases the amplitude and frequency of spontaneous GABAAR-mediated inhibitory postsynaptic currents (IPSCs) in the cells leaving the GE. In older MAM-treated animals, the amplitude and frequency of GABAAR-mediated spontaneous IPSCs in layer 2/3 pyramidal cells is increased, as are the amplitude and frequency of miniature IPSCs. The kinetics of GABAAR opening also altered following treatment with MAM. Western blot analysis shows that the expression of the GABAAα3R and GABAAγ2R subunits amplified in our model animals. We did not observe any significant change in the passive properties of either the layer 2/3 pyramidal cells or cells leaving the GE after MAM treatment. These observations reinforce the idea that synaptic neurotransmission through GABAAR enhances following treatment with MAM and coincides with our finding of increased GABAAαR expression within the upper cortical layers. Overall, we demonstrate that small amounts of toxins delivered during corticogenesis can result in long-lasting changes in ambient expression of GABAAR that influence intrinsic neuronal properties.


Subject(s)
Inhibitory Postsynaptic Potentials , Malformations of Cortical Development/physiopathology , Miniature Postsynaptic Potentials , Neocortex/physiopathology , Receptors, GABA-A/metabolism , Animals , Disease Models, Animal , Female , Ferrets , Malformations of Cortical Development/chemically induced , Methylazoxymethanol Acetate/analogs & derivatives , Methylazoxymethanol Acetate/toxicity , Neocortex/cytology , Neocortex/embryology , Neocortex/metabolism , Pregnancy , Protein Subunits/genetics , Protein Subunits/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Receptors, GABA-A/genetics , Teratogens/toxicity
11.
PLoS One ; 8(12): e84492, 2013.
Article in English | MEDLINE | ID: mdl-24358362

ABSTRACT

Children with malformations of cortical development (MCD) frequently have associated cognitive impairments which reduce quality of life. We hypothesized that cognitive deficits associated with MCD can be improved with environmental manipulation or additional training. The E17 methylazoxymethanol acetate (MAM) exposure model bears many anatomical hallmarks seen in human MCDs as well as similar behavioral and cognitive deficits. We divided control and MAM exposed Sprague-Dawley rats into enriched and non-enriched groups and tested performance in the Morris water maze. Another group similarly divided underwent sociability testing and also underwent Magnetic Resonance Imaging (MRI) scans pre and post enrichment. A third group of control and MAM rats without enrichment were trained until they reached criterion on the place avoidance task. MAM rats had impaired performance on spatial tasks and enrichment improved performance of both control and MAM animals. Although MAM rats did not have a deficit in sociability they showed similar improvement with enrichment as controls. MRI revealed a whole brain volume decrease with MAM exposure, and an increase in both MAM and control enriched volumes in comparison to non-enriched animals. In the place avoidance task, MAM rats required approximately 3 times as long to reach criterion as control animals, but with additional training were able to reach control performance. Environmental manipulation and additional training can improve cognition in a rodent MCD model. We therefore suggest that patients with MCD may benefit from appropriate alterations in educational strategies, social interaction and environment. These factors should be considered in therapeutic strategies.


Subject(s)
Cognition , Conditioning, Psychological , Malformations of Cortical Development/psychology , Animals , Avoidance Learning , Behavior, Animal , Choice Behavior , Female , Magnetic Resonance Imaging , Male , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/diagnosis , Maze Learning , Methylazoxymethanol Acetate/administration & dosage , Methylazoxymethanol Acetate/adverse effects , Pregnancy , Prenatal Exposure Delayed Effects , Rats
12.
Eur J Neurosci ; 37(1): 150-62, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23095101

ABSTRACT

Cortical dysplasias (CDs) include a spectrum of cerebral lesions resulting from cortical development abnormalities during embryogenesis that lead to cognitive disabilities and epilepsy. The experimental model of CD obtained by means of in utero administration of BCNU (1-3-bis-chloroethyl-nitrosurea) to pregnant rats on embryonic day 15 mimics the histopathological abnormalities observed in many patients. The aim of this study was to investigate the behavioural, electrophysiological and anatomical profile of BCNU-treated rats in order to determine whether cortical and hippocampal lesions can directly lead to cognitive dysfunction. The BCNU-treated rats showed impaired short-term working memory but intact long-term aversive memory, whereas their spontaneous motor activity and anxiety-like response were normal. The histopathological and immunohistochemical analyses, made after behavioural tests, revealed the disrupted integrity of neuronal populations and connecting fibres in hippocampus and prefrontal and entorhinal cortices, which are involved in memory processes. An electrophysiological evaluation of the CA1 region of in vitro hippocampal slices indicated a decrease in the efficiency of excitatory synaptic transmission and impaired paired pulse facilitation, but enhanced long-term potentiation (LTP) associated with hyperexcitability in BCNU-treated rats compared with controls. The enhanced LTP, associated with hyperexcitability, may indicate a pathological distortion of long-term plasticity. These findings suggest that prenatal developmental insults at the time of peak cortical neurogenesis can induce anatomical abnormalities associated with severe impairment of spatial working memory in adult BCNU-treated rats and may help to clarify the pathophysiological mechanisms of cognitive dysfunction that is often associated with epilepsy in patients with CD.


Subject(s)
Entorhinal Cortex/pathology , Frontal Lobe/pathology , Hippocampus/pathology , Malformations of Cortical Development/physiopathology , Animals , Carmustine , Cognition/drug effects , Disease Models, Animal , Entorhinal Cortex/drug effects , Entorhinal Cortex/embryology , Excitatory Postsynaptic Potentials/drug effects , Female , Frontal Lobe/drug effects , Frontal Lobe/embryology , Hippocampus/drug effects , Hippocampus/embryology , Long-Term Potentiation/drug effects , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/pathology , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Motor Activity/drug effects , Nerve Fibers/pathology , Neurogenesis/drug effects , Neurons/pathology , Pregnancy , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
13.
Epilepsy Res ; 102(1-2): 113-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22647842

ABSTRACT

There is a high incidence of epilepsy in patients with polymicrogyria; however, the epileptogenic mechanisms are largely unknown. The density of parvalbumin-immunoreactive interneurons was evaluated in an experimental model of polymicrogyria, in order to assess the potential changes in the development of one population of inhibitory interneurons. Newborn hamsters received an intracerebral injection of ibotenate, and all injected animals showed abnormal cortical layers characterized by one or two microgyrus in the fronto-parietal cortex. A quantitative analysis revealed that the ratios of parvalbumin-immunoreactive neurons in total neurons were significantly reduced in the medial paramicrogyral area, and in the medial and central parts of microgyrus in comparison to that in the lateral part of microgyrus (P<0.01). The lateral paramicrogyral area had the greatest number of parvalbumin-immunoreactive neurons, which was increased significantly in comparison to that in the control cortex (P<0.01). We suggest that the callosal, thalamic and intracortical afferents to the microgyrus and paramicrogyral area may induce a remarkable imbalance between the excitatory and inhibitory activities of the cortical structures, associated with the epileptogenic mechanism in polymicrogyria.


Subject(s)
Epilepsy/pathology , Interneurons/pathology , Malformations of Cortical Development/pathology , Neural Inhibition , Animals , Animals, Newborn , Cerebral Cortex/pathology , Corpus Callosum/pathology , Cricetinae , Disease Models, Animal , Epilepsy/chemically induced , Epilepsy/epidemiology , Excitatory Amino Acid Agonists/toxicity , Female , Ibotenic Acid/toxicity , Incidence , Interneurons/metabolism , Male , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/epidemiology , Neurons, Afferent/metabolism , Neurons, Afferent/pathology , Parvalbumins/metabolism , Thalamus/pathology
14.
Brain ; 134(Pt 10): 2828-43, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21482549

ABSTRACT

We have generated an experimental 'double-hit' model of chronic epilepsy to recapitulate the co-existence of abnormal cortical structure and frequently recurrent seizures as observed in human focal cortical dysplasia. We induced cortical malformations by exposing rats prenatally to methylazoxymethanol acetate and triggered status epilepticus and recurrent seizures in adult methylazoxymethanol acetate rats with pilocarpine. We studied the course of epilepsy and the long-term morphologic and molecular changes induced by the occurrence of status epilepticus and subsequent chronic epilepsy in the malformed methylazoxymethanol acetate exposed brain. Behavioural and electroencephalographic analyses showed that methylazoxymethanol acetate pilocarpine rats develop more severe epilepsy than naïve rats. Morphologic and molecular analyses demonstrated that status epilepticus and subsequent seizures, but not pilocarpine treatment per se, was capable of affecting both cortical architectural and N-methyl-D-aspartate receptor abnormalities induced by methylazoxymethanol acetate. In particular, cortical thickness was further decreased and N-methyl-D-aspartate regulatory subunits were recruited at the postsynaptic membrane. In addition, methylazoxymethanol acetate pilocarpine rats showed abnormally large cortical pyramidal neurons with neurofilament over-expression. These neurons bear similarities to the hypertrophic/dysmorphic pyramidal neurons observed in acquired human focal cortical dysplasia. These data show that status epilepticus sets in motion a pathological process capable of significantly changing the cellular and molecular features of pre-existing experimental cortical malformations. They suggest that seizure recurrence in human focal cortical dysplasia might be an additional factor in establishing a pathological circuitry that favours chronic neuronal hyperexcitability.


Subject(s)
Cerebral Cortex/pathology , Malformations of Cortical Development/pathology , Neurons/pathology , Status Epilepticus/pathology , Animals , Cerebral Cortex/physiopathology , Disease Models, Animal , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/physiopathology , Methylazoxymethanol Acetate , Neurons/physiology , Pilocarpine , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology
15.
Neuroscience ; 175: 380-93, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21130845

ABSTRACT

Cortical dysplasia (CD) comprises a wide range of cerebral cortex alterations ranging from severe brain malformations to local disruption of the cortical structure. Most hypotheses focused on the role of embryonic/perinatal development insults as the main cause for the majority of CD. Rats with prenatal exposure to BCNU (1-3-bis-chloroethyl-nitrosurea) represent an injury-based model and reproduce many anatomical features seen in human patients with CD, such as altered cortical layering and the presence of heterotopia and dysmorphic/heterotopic neurons. With the aim to investigate the formation and evolution of CD during development, we analysed the expression of a panel of layer-specific genes (Nurr1, Er81, Ror-ß and Cux2, markers of layers VI, V, IV and superficial layers, respectively) in BCNU-treated cortices from E17 to postnatal day 14. By means of appropriate immunohistochemical markers, we also analysed the structural organization of embryonic ventricular zone and of glial and axonal fibres, substrates supporting radial and tangential migration, respectively. The main results of the present study are: (i) the ventricular zone appeared disorganized and the neuroependyma was partially disrupted; (ii) radial glia scaffold and tangential fibres were deeply disarranged, thus explaining the neuronal migration defects; (iii) cortical heterotopia were detectable by E19, whereas periventricular heterotopia were detectable after birth; (iv) both cortical and periventricular heterotopia showed a pseudo-laminar structure, with cells of the upper cortical layers in the core of the nodules and cells of layer IV and V at their border; (v) the distribution of GABAergic cells was altered since the embryonic stages, as a consequence of the derangement of tangential fibres. Our analysis sheds light on how a malformed cortex develops after a temporally discrete environmental insult and adds additional knowledge on specific aspects of the etiopathogenesis of CD.


Subject(s)
Carmustine/toxicity , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/genetics , Nervous System Malformations/chemically induced , Nervous System Malformations/genetics , Teratogens/toxicity , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Movement/drug effects , Cell Movement/genetics , Disease Models, Animal , Epilepsy/etiology , Epilepsy/genetics , Epilepsy/pathology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Humans , Malformations of Cortical Development/pathology , Nervous System Malformations/pathology , Rats , Rats, Sprague-Dawley
16.
Epilepsia ; 51(9): 1809-19, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20738387

ABSTRACT

PURPOSE: Hippocampal sclerosis, characterized by prominent neuronal loss and reactive gliosis, is the most common pathology in human temporal lobe epilepsy (TLE). Although prolonged febrile convulsion (FC) is a risk factor of TLE, it is not clear whether FC provokes hippocampal sclerosis and subsequent TLE. Given that underlying brain lesions, such as cortical dysplasia (CD), in the immature brain predispose patients to FC, CD may link FC and TLE. However, the role of CD in epileptogenesis after FC is also unclear. Here, we investigated whether inborn CD increases the risk of later epilepsy induced by prolonged FC using a rat model. METHODS: Experimental CD was induced by in utero exposure of methylazoxymethanol (MAM). Rat pups from MAM-treated or control rats were then subjected to prolonged FC. We examined morphologic changes in the hippocampi with respect to neuronal loss, reactive gliosis, and synaptogenesis, and evaluated spontaneous recurrent seizures (SRS) by long-term video-EEG (electroencephalography). RESULTS: The MAM+FC group had a significantly lower hippocampal neuronal density in the CA1 and dentate hilus than other control groups. A robust increase in glial cells and synaptic reorganization was also detected in the MAM+FC groups. Furthermore, later SRS occurred in all rats in the MAM+FC group and in 50% and 25% of the rats in the FC-only and MAM-only group, respectively. The frequency and total duration of SRS was highest in the MAM+FC group. DISCUSSION: Our results suggest that preexisting CD in the immature brain augments the proepileptogenic effects of prolonged FC, leading to TLE.


Subject(s)
Epilepsy, Temporal Lobe/etiology , Malformations of Cortical Development/physiopathology , Seizures, Febrile/congenital , Seizures, Febrile/physiopathology , Animals , Disease Models, Animal , Electroencephalography/methods , Electroencephalography/statistics & numerical data , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Malformations of Cortical Development/chemically induced , Methylazoxymethanol Acetate/analogs & derivatives , Mossy Fibers, Hippocampal/pathology , Rats , Rats, Sprague-Dawley , Sclerosis/pathology , Sclerosis/physiopathology , Seizures, Febrile/chemically induced , Synapses/pathology
17.
Childs Nerv Syst ; 26(11): 1575-81, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20461523

ABSTRACT

OBJECTIVES: Cortical dysplasia is a cortical malformation resulting from any developmental defects during different periods of development. This study aims to investigate the hippocampal histopathological alterations in the neonates with cortical dysplasia due to the prenatal exposure to carmustine (1,3-bis (2-chloroethyl)-1-nitrosourea; BCNU) and the possible effects of prophylaxis with melatonin, a neuroprotective agent. METHODS: Wistar albino female rats were randomly divided into four experimental groups; control, melatonin-treated, BCNU-exposed and BCNU-exposed+melatonin-treated. Light microscopy and immunohistochemistry were carried out on the newborn hippocampus. RESULTS: Histopathology of hippocampus from the control and melatonin-treated groups showed continuity of migration and maturation as pathognomonic signs of the normal newborn hippocampus. Hippocampal cortex from the newborns exposed in utero to BCNU showed the histology of early embryonic hippocampal formation with immunohistochemical increase in the number of nestin positive cells and decreases in the immunoreactivity of glial fibrillary acidic protein (GFAP) and synaptophysin. These findings indicate a significant delay in hippocampal maturation, migration, and synaptogenesis. Intrauterine treatment of BCNU-exposed rats with melatonin resulted in histopathological features almost similar to control group. CONCLUSION: It has been concluded that cortical dysplasia induced by intrauterine BCNU administration results in delayed hippocampal maturation, which is successfully restored by intrauterine melatonin treatment.


Subject(s)
Hippocampus/drug effects , Malformations of Cortical Development/pathology , Melatonin/pharmacology , Animals , Animals, Newborn , Antineoplastic Agents, Alkylating/toxicity , Carmustine/toxicity , Female , Glial Fibrillary Acidic Protein/analysis , Humans , Immunoenzyme Techniques , Intermediate Filament Proteins/analysis , Malformations of Cortical Development/chemically induced , Nerve Tissue Proteins/analysis , Nestin , Pregnancy , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Rats , Rats, Wistar , Synaptophysin/analysis
18.
Eur Radiol ; 20(9): 2204-12, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20393713

ABSTRACT

PURPOSE: To assess the feasibility of magnetic resonance imaging (MRI) enhanced with ultrasmall superparamagnetic particles of iron oxide (USPIO) for assessing excitotoxic brain lesions in an experimental model of neonatal periventricular white matter (PWM) lesions. MATERIALS AND METHODS: Brain lesions were induced by intracerebral injection of ibotenate in 14 newborn rats. Pre- and post-USPIO T2-weighted MRI was performed in seven of them (group A) and in five control newborns (group C). In seven newborns with induced cerebral lesions, USPIO-enhanced MRI was not performed (group B). We compared the signal intensity of the lesion to the contralateral unaffected brain (lesion-to-brain contrast, LBC) and the lesion signal-to-noise ratio (SNR) before and after USPIO injection. MR imaging was correlated with histology. RESULTS: USPIO injection significantly (P<0.05) decreased LBC and SNR of brain lesion but induced no changes in normal controls. The densities of macrophages and iron-laden cells were higher on the lesion side than on the contralateral side (P<0.05). Neither lesion size nor the surrounding macrophage infiltrate was significantly different between groups A and B. CONCLUSION: Post-USPIO T2-weighted MRI demonstrated negative enhancement of neonatal excitotoxic brain lesion. USPIO injection does not appear to exacerbate brain lesions.


Subject(s)
Dextrans , Disease Models, Animal , Ibotenic Acid , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/pathology , Nerve Fibers, Myelinated/pathology , Animals , Animals, Newborn , Contrast Media , Humans , Image Enhancement/methods , Neurotoxins , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
19.
Birth Defects Res A Clin Mol Teratol ; 88(2): 128-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19764076

ABSTRACT

BACKGROUND: Intrauterine exposure to alcohol may result in a distinct pattern of craniofacial abnormalities and central nervous system dysfunction, designated fetal alcohol syndrome (FAS). The spectrum of malformations of the brain associated with maternal alcohol abuse during pregnancy is much broader than the relatively uniform clinical phenotype of FAS. Among these malformations the most striking abnormalities involve the impairment of neuronal cell migration. However, polymicrogyria (PMG) has so far been reported only once in a human autopsy study of a child with FAS. CASE: A 16-year-old girl with confirmed maternal alcohol consumption during pregnancy and full phenotype of FAS presented after two generalized epileptic seizures for neurologic assessment. Cranial magnetic resonance imaging revealed bilateral PMG in the superior frontal gyrus with asymmetric distribution. History, clinical features, and genetic investigations provided no evidence for any of the known genetic or acquired causes of PMG. Therefore, we propose that prenatal alcohol exposure is the cause of PMG in this patient rather than a mere coincidence. CONCLUSION: Our observation represents only the second patient of PMG in FAS and confirms the phenotypic variability of cerebral malformations associated with maternal alcohol abuse during pregnancy. In patients with clinical features of FAS and neurologic deficits or seizures neuroimaging is recommended. Furthermore, FAS should be considered as a differential diagnosis for PMG.


Subject(s)
Fetal Alcohol Spectrum Disorders/diagnosis , Malformations of Cortical Development/diagnosis , Prenatal Exposure Delayed Effects/diagnosis , Adolescent , Diagnosis, Differential , Epilepsy/chemically induced , Epilepsy/congenital , Female , Fetal Alcohol Spectrum Disorders/pathology , Functional Laterality/drug effects , Germany , Humans , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/pathology , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Russia
20.
Ann Neurol ; 66(1): 19-27, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19670444

ABSTRACT

OBJECTIVE: Brain arteriovenous malformations (AVMs) are an important cause of neurological morbidity in young adults. The pathophysiology of these lesions is poorly understood. A soluble form of endoglin (sEng) has been shown to cause endothelial dysfunction and induce preeclampsia. We tested if sEng would be elevated in brain AVM tissues relative to epilepsy brain tissues, and also investigated whether sEng overexpression via gene transfer in the mouse brain would induce vascular dysplasia and associated changes in downstream signaling pathways. METHODS: Expression levels of sEng in surgical specimens were determined by Western blot assay and enzyme-linked immunosorbent assay. Vascular dysplasia, levels of matrix metalloproteinase (MMP), and oxidative stress were determined by immunohistochemistry and gelatin zymography. RESULTS: Brain AVMs (n = 33) had higher mean sEng levels (245 +/- 175 vs 100 +/- 60, % of control, p = 0.04) compared with controls (n = 8), as determined by Western blot. In contrast, membrane-bound Eng was not significantly different (108 +/- 79 vs 100 +/- 63, % of control, p = 0.95). sEng gene transduction in the mouse brain induced abnormal vascular structures. It also increased MMP activity by 490 +/- 30% (MMP-9) and 220 +/- 30% (MMP-2), and oxidants by 260 +/- 20% (4-hydroxy-2-nonenal) at 2 weeks after injection, suggesting that MMPs and oxidative radicals may mediate sEng-induced pathological vascular remodeling. INTERPRETATION: The results suggest that elevated sEng may play a role in the generation of sporadic brain AVMs. Our findings may provide new targets for therapeutic intervention for patients with brain AVMs. Ann Neurol 2009;66:19-27.


Subject(s)
Antigens, CD/metabolism , Central Nervous System Vascular Malformations/metabolism , Gene Expression Regulation, Enzymologic/physiology , Receptors, Cell Surface/metabolism , Adult , Aldehydes/pharmacology , Animals , Central Nervous System Vascular Malformations/pathology , Cysteine Proteinase Inhibitors/pharmacology , Disease Progression , Endoglin , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression Regulation, Enzymologic/drug effects , Humans , Male , Malformations of Cortical Development/chemically induced , Matrix Metalloproteinases/classification , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Statistics, Nonparametric , Superoxides/metabolism , Vascular Endothelial Growth Factor A/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...