Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Front Neurol ; 6: 46, 2015.
Article in English | MEDLINE | ID: mdl-25806020

ABSTRACT

Exposure of the brain to brief, non-harmful seizures can activate protective mechanisms that temporarily generate a damage-refractory state. This process, termed epileptic tolerance, is associated with large-scale down-regulation of gene expression. Polycomb group (PcG) proteins are master controllers of gene silencing during development that are re-activated by injury to the brain. Here, we explored the transcriptional response of genes associated with polycomb repressive complex (PRC) 1 (Ring1A, Ring1B, and Bmi1) and PRC2 (Ezh1, Ezh2, and Suz12), as well as additional transcriptional regulators Sirt1, Yy1, and Yy2, in a mouse model of status epilepticus (SE). Findings were contrasted to changes after SE in mice previously given brief seizures to evoke tolerance. Real-time quantitative PCR showed SE prompted an early (1 h) increase in expression of several genes in PRC1 and PRC2 in the hippocampus, followed by down-regulation of many of the same genes at later times points (4, 8, and 24 h). Spatio-temporal differences were found among PRC2 genes in epileptic tolerance, including increased expression of Ezh2, Suz12, and Yy2 relative to the normal injury response to SE. In contrast, PRC1 complex genes including Ring 1B and Bmi1 displayed differential down-regulation in epileptic tolerance. The present study characterizes PcG gene expression following SE and shows prior seizure exposure produces select changes to PRC1 and PRC2 composition that may influence differential gene expression in epileptic tolerance.

2.
Brain ; 138(Pt 3): 616-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552301

ABSTRACT

Temporal lobe epilepsy is associated with large-scale, wide-ranging changes in gene expression in the hippocampus. Epigenetic changes to DNA are attractive mechanisms to explain the sustained hyperexcitability of chronic epilepsy. Here, through methylation analysis of all annotated C-phosphate-G islands and promoter regions in the human genome, we report a pilot study of the methylation profiles of temporal lobe epilepsy with or without hippocampal sclerosis. Furthermore, by comparative analysis of expression and promoter methylation, we identify methylation sensitive non-coding RNA in human temporal lobe epilepsy. A total of 146 protein-coding genes exhibited altered DNA methylation in temporal lobe epilepsy hippocampus (n = 9) when compared to control (n = 5), with 81.5% of the promoters of these genes displaying hypermethylation. Unique methylation profiles were evident in temporal lobe epilepsy with or without hippocampal sclerosis, in addition to a common methylation profile regardless of pathology grade. Gene ontology terms associated with development, neuron remodelling and neuron maturation were over-represented in the methylation profile of Watson Grade 1 samples (mild hippocampal sclerosis). In addition to genes associated with neuronal, neurotransmitter/synaptic transmission and cell death functions, differential hypermethylation of genes associated with transcriptional regulation was evident in temporal lobe epilepsy, but overall few genes previously associated with epilepsy were among the differentially methylated. Finally, a panel of 13, methylation-sensitive microRNA were identified in temporal lobe epilepsy including MIR27A, miR-193a-5p (MIR193A) and miR-876-3p (MIR876), and the differential methylation of long non-coding RNA documented for the first time. The present study therefore reports select, genome-wide DNA methylation changes in human temporal lobe epilepsy that may contribute to the molecular architecture of the epileptic brain.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic , Epilepsy, Temporal Lobe/pathology , Hippocampus/pathology , Adolescent , Adult , Computational Biology , CpG Islands/physiology , Epilepsy, Temporal Lobe/genetics , Female , Gene Expression Regulation , Hippocampus/metabolism , Humans , Immunoprecipitation , Male , MicroRNAs/metabolism , Microdissection , Middle Aged , Pilot Projects , Promoter Regions, Genetic , Sclerosis , Young Adult
3.
Methods Mol Biol ; 1067: 87-101, 2013.
Article in English | MEDLINE | ID: mdl-23975788

ABSTRACT

Epigenetic modification of DNA by methylation of the cytosine present in CG dinucleotides constitutes a key regulatory mechanism in the control of gene expression in neurological diseases. In this chapter, we describe an in-depth methodology of methylated DNA immunoprecipitation used in combination with tiling microarrays (MeDIP-chip) in order to analyze genome-wide gene promoter methylation in the hippocampus of mice following status epilepticus (prolonged seizure). While a specific mouse model and array format are described, the method can be applied to DNA from many tissues to analyze the methylation status of promoter regions across whole genomes, using a wide range of available array formats (both custom designed and commercially catalogued). We conclude the chapter with the description of bisulfite sequencing validation of MeDIP-chip results.


Subject(s)
DNA Methylation , DNA/genetics , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Status Epilepticus/genetics , Animals , DNA/analysis , Disease Models, Animal , Immunoprecipitation/methods , Mice , Sulfites/chemistry
4.
Epilepsy Res ; 103(2-3): 167-79, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23182415

ABSTRACT

Mesial temporal lobe epilepsy (MTLE) is the most common, intractable seizure disorder in adults. Blood-brain barrier (BBB) disruption, including interruption of endothelial tight cell junctions and serum protein and immunoglobulin G (IgG) extravasation into brain parenchyma, has been reported in experimental and human MTLE and implicated in disease pathogenesis. Triggering status epilepticus in mice by intra-amygdala microinjection of kainic acid produces damage mainly within the CA3 subfield of the ipsilateral hippocampus, and recurrent spontaneous seizures emerge during the following week. To investigate whether BBB impairment is a feature of this model, we characterized endothelial tight cell junction proteins and IgG and albumin in the hippocampus up to three weeks after status epilepticus. Hippocampal microvessels displayed a reduction in continuous staining for zonula occludens 1 (ZO-1), the main tight junction protein, after status epilepticus and in epileptic mice, although western blotting found ZO-1 protein levels in the hippocampal subfields were not different from controls at any time. Increased IgG and albumin were detected in damaged and non-damaged ipsilateral hippocampal subfields, mainly 4-24h after status epilepticus, although increased serum protein extravasation was also found in the CA3 subfield in epileptic mice. Thus, BBB opening or damage occurs mainly in the period shortly after status epilepticus but may also persist within the CA3 subfield as a feature of the pathophysiology of chronic epilepsy in this model.


Subject(s)
Amygdala/physiology , Blood-Brain Barrier/pathology , Kainic Acid/toxicity , Status Epilepticus/pathology , Amygdala/drug effects , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Injections, Intraventricular , Kainic Acid/administration & dosage , Male , Mice , Mice, Inbred C57BL , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Time Factors
5.
J Neurosci ; 32(5): 1577-88, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22302800

ABSTRACT

Prolonged seizures (status epilepticus) produce pathophysiological changes in the hippocampus that are associated with large-scale, wide-ranging changes in gene expression. Epileptic tolerance is an endogenous program of cell protection that can be activated in the brain by previous exposure to a non-harmful seizure episode before status epilepticus. A major transcriptional feature of tolerance is gene downregulation. Here, through methylation analysis of 34,143 discrete loci representing all annotated CpG islands and promoter regions in the mouse genome, we report the genome-wide DNA methylation changes in the hippocampus after status epilepticus and epileptic tolerance in adult mice. A total of 321 genes showed altered DNA methylation after status epilepticus alone or status epilepticus that followed seizure preconditioning, with >90% of the promoters of these genes undergoing hypomethylation. These profiles included genes not previously associated with epilepsy, such as the polycomb gene Phc2. Differential methylation events generally occurred throughout the genome without bias for a particular chromosomal region, with the exception of a small region of chromosome 4, which was significantly overrepresented with genes hypomethylated after status epilepticus. Surprisingly, only few genes displayed differential hypermethylation in epileptic tolerance. Nevertheless, gene ontology analysis emphasized the majority of differential methylation events between the groups occurred in genes associated with nuclear functions, such as DNA binding and transcriptional regulation. The present study reports select, genome-wide DNA methylation changes after status epilepticus and in epileptic tolerance, which may contribute to regulating the gene expression environment of the seizure-damaged hippocampus.


Subject(s)
CA3 Region, Hippocampal/metabolism , DNA Methylation/genetics , Status Epilepticus/genetics , Status Epilepticus/metabolism , Animals , Down-Regulation/genetics , Genome-Wide Association Study/methods , Male , Mice , Mice, Inbred C57BL , Status Epilepticus/prevention & control
6.
PLoS One ; 6(1): e14565, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21283688

ABSTRACT

The development of different brain regions involves the coordinated control of proliferation and cell fate specification along and across the neuraxis. Here, we identify Plxdc2 as a novel regulator of these processes, using in ovo electroporation and in vitro cultures of mammalian cells. Plxdc2 is a type I transmembrane protein with some homology to nidogen and to plexins. It is expressed in a highly discrete and dynamic pattern in the developing nervous system, with prominent expression in various patterning centres. In the chick neural tube, where Plxdc2 expression parallels that seen in the mouse, misexpression of Plxdc2 increases proliferation and alters patterns of neurogenesis, resulting in neural tube thickening at early stages. Expression of the Plxdc2 extracellular domain alone, which can be cleaved and shed in vivo, is sufficient for this activity, demonstrating a cell non-autonomous function. Induction of proliferation is also observed in cultured embryonic neuroepithelial cells (ENCs) derived from E9.5 mouse neural tube, which express a Plxdc2-binding activity. These experiments uncover a direct molecular activity of Plxdc2 in the control of proliferation, of relevance in understanding the role of this protein in various cancers, where its expression has been shown to be altered. They also implicate Plxdc2 as a novel component of the network of signalling molecules known to coordinate proliferation and differentiation in the developing nervous system.


Subject(s)
Avian Proteins/physiology , Mitogens/physiology , Neural Stem Cells/cytology , Neurogenesis , Receptors, Cell Surface/physiology , Animals , Cell Differentiation , Cell Proliferation , Chick Embryo , Neural Tube
SELECTION OF CITATIONS
SEARCH DETAIL
...