Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 4: e606, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23618904

ABSTRACT

Prolonged seizures (status epilepticus, SE) can cause neuronal death within brain regions such as the hippocampus. This may contribute to impairments in cognitive functioning and trigger or exacerbate epilepsy. Seizure-induced neuronal death is mediated, at least in part, by apoptosis-associated signaling pathways. Indeed, mice lacking certain members of the potently proapoptotic BH3-only subfamily of Bcl-2 proteins are protected against hippocampal damage caused by status epilepticus. The recently identified BH3-only protein Bcl-2-modifying factor (Bmf) normally interacts with the cytoskeleton, but upon certain cellular stresses, such as loss of extracellular matrix adhesion or energy crisis, Bmf relocalizes to mitochondria, where it can promote Bax activation and mitochondrial dysfunction. Although Bmf has been widely reported in the hematopoietic system to exert a proapoptotic effect, no studies have been undertaken in models of neurological disorders. To examine whether Bmf is important for seizure-induced neuronal death, we studied Bmf induction after prolonged seizures induced by intra-amygdala kainic acid (KA) in mice, and examined the effect of Bmf-deficiency on seizures and damage caused by SE. Seizures triggered an early (1-8 h) transcriptional activation and accumulation of Bax in the cell death-susceptible hippocampal CA3 subfield. Bmf mRNA was biphasically upregulated beginning at 1 h after SE and returning to normal by 8 h, while again being found elevated in the hippocampus of epileptic mice. Bmf upregulation was prevented by Compound C, an inhibitor of adenosine monophosphate-activated protein kinase, indicating Bmf expression may be induced in response to bioenergetic stress. Bmf-deficient mice showed normal sensitivity to the convulsant effects of KA, but, surprisingly, displayed significantly more neuronal death in the hippocampal CA1 and CA3 subfields after SE. These are the first studies investigating Bmf in a model of neurologic injury, and suggest that Bmf may protect neurons against seizure-induced neuronal death in vivo.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Hippocampus/physiopathology , Status Epilepticus/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Hippocampus/metabolism , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Time Factors , Up-Regulation/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
2.
Neuroscience ; 238: 218-29, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23485811

ABSTRACT

MicroRNA (miRNA) is a class of small non-coding RNA which regulates post-transcriptional gene expression by repressing and thereby fine-tuning protein production, mainly via sequence-specific binding within the 3'untranslated region of mRNA transcripts. Although in humans there are only ∼1600 miRNAs, bioinformatics, systems studies and advanced quantitative proteomics reveal miRNA regulation of over half of all protein-coding genes and that each miRNA can regulate multiple proteins. Epilepsy is a common, serious neurologic disorder characterized by recurring unprovoked seizures that result from abnormal firing of populations of neurons in the brain. The brain expresses several unique miRNAs which control dendritic morphology as well as ion channel levels, neuronal migration and glial function. There is an emerging view that the patho-mechanisms underlying the process of epileptogenesis, as well as maintenance and progression of the epileptic state, involve miRNAs that control multiple genes and proteins on a systems level. Expression profiling studies reveal select changes to brain miRNA levels following prolonged seizures (status epilepticus) in animal models. Inflammation, stress signaling and neuronal excitation are among the pathways most impacted. Analysis of miRNA expression in human epilepsy has also been performed, where again neuroinflammatory processes were prominent. These studies suggest that miRNAs may regulate certain key processes but are not necessarily broadly altering all patho-mechanisms in epilepsy. Functional studies employing antagomirs have identified contributions from miR-34a and miR-132 to seizure-induced neuronal death whereas silencing miR-134 potently reduced status epilepticus, seizure-damage and the later occurrence of spontaneous seizures. Efforts to identify the in vivo target(s) of epilepsy-regulated miRNAs, is now a priority. Last, miRNAs are stable, information-carrying (paracrine) signals. Profiling miRNA in biofluids may represent a novel source of disease biomarkers in epilepsy. In summary, miRNA is emerging as a critical new layer of gene expression control with implications for the cause and treatment of epilepsy.


Subject(s)
Epilepsy/genetics , Hippocampus/metabolism , MicroRNAs/genetics , Neurons/metabolism , Animals , Disease Models, Animal , Epilepsy/metabolism , Epilepsy/pathology , Hippocampus/pathology , Humans , MicroRNAs/metabolism , Neurons/pathology , Sclerosis/genetics , Sclerosis/metabolism , Sclerosis/pathology
3.
Cell Death Dis ; 3: e287, 2012 Mar 22.
Article in English | MEDLINE | ID: mdl-22436728

ABSTRACT

MicroRNAs (miRNAs) are short, noncoding RNAs that function as posttranscriptional regulators of gene expression by controlling translation of mRNAs. A subset of miRNAs may be critical for the control of cell death, including the p53-regulated miRNA, miR-34a. Because seizures activate p53, and p53-deficient mice are reportedly resistant to damage caused by prolonged seizures, we investigated the role of miR-34a in seizure-induced neuronal death in vivo. Status epilepticus was induced by intra-amygdala microinjection of kainic acid in mice. This led to an early (2 h) multifold upregulation of miR-34a in the CA3 and CA1 hippocampal subfields and lower protein levels of mitogen-activated kinase kinase kinase 9, a validated miR-34a target. Immunoprecipitation of the RNA-induced silencing complex component, Argonaute-2, eluted significantly higher levels of miR-34a after seizures. Injection of mice with pifithrin-α, a putative p53 inhibitor, prevented miR-34a upregulation after seizures. Intracerebroventricular injection of antagomirs targeting miR-34a reduced hippocampal miR-34a levels and had a small modulatory effect on apoptosis-associated signaling, but did not prevent hippocampal neuronal death in models of either severe or moderate severity status epilepticus. Thus, prolonged seizures cause subfield-specific, temporally restricted upregulation of miR-34a, which may be p53 dependent, but miR-34a is probably not important for seizure-induced neuronal death in this model.


Subject(s)
Apoptosis/drug effects , Hippocampus/metabolism , MicroRNAs/metabolism , Seizures/metabolism , Up-Regulation , Animals , Argonaute Proteins/metabolism , Benzothiazoles/pharmacology , Hippocampus/drug effects , Immunoprecipitation , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Protein Binding , Seizures/pathology , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Neuroscience ; 171(2): 556-65, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20837105

ABSTRACT

Brief, non-harmful seizures can activate endogenous protective programmes which render the brain resistant to damage caused by prolonged seizure episodes. Whether protection in epileptic tolerance is long-lasting or influences the subsequent development of epilepsy is uncertain. Presently, we investigated the relationship between hippocampal pathology, neuropeptide Y rearrangement and spontaneous seizures in sham- and seizure-preconditioned mice after status epilepticus induced by intra-amygdala kainate. Seizure-induced neuronal death at 24 h was significantly reduced in the ipsilateral hippocampal CA3 and hilus of tolerance mice compared to sham-preconditioned animals subject to status epilepticus. Damage to the CA3-hilus remained reduced in tolerance mice 21 days post-status. In sham-preconditioned mice subject to status epilepticus correlative statistics showed there was a strong inverse relationship between CA3, but not hilar, neuron counts and the number of spontaneous seizures. A strong positive association was also found between neuropeptide Y score and spontaneous seizure count in these mice. In contrast, there was no significant association between spontaneous seizure count and CA3 neuron loss or neuropeptide Y rearrangement in the tolerance mice. These data show that tolerance-conferred neuroprotection is long-lasting and that tolerance disrupts the normal association between CA3 damage, synaptic rearrangement and occurrence of spontaneous seizures in this model.


Subject(s)
CA3 Region, Hippocampal/pathology , Kainic Acid , Neuropeptide Y/metabolism , Seizures/prevention & control , Status Epilepticus/prevention & control , Amygdala , Animals , Cell Count , Cell Death , Cytoprotection , Male , Mice , Mice, Inbred C57BL , Mossy Fibers, Hippocampal/pathology , Neurons/pathology , Seizures/pathology , Seizures/physiopathology , Status Epilepticus/chemically induced , Status Epilepticus/physiopathology , Synapses/pathology , Time Factors
5.
Cell Death Dis ; 1: e79, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-21368852

ABSTRACT

The p53 tumor suppressor is a multifunctional protein, which regulates cell cycle, differentiation, DNA repair and apoptosis. Experimental seizures up-regulate p53 in the brain, and acute seizure-induced neuronal death can be reduced by genetic deletion or pharmacologic inhibition of p53. However, few long-term functional consequences of p53 deficiency have been explored. Here, we investigated the development of epilepsy triggered by status epilepticus in wild-type and p53-deficient mice. Analysis of electroencephalogram (EEG) recordings during status epilepticus induced by intra-amygdala kainic acid (KA) showed that seizures lasted significantly longer in p53-deficient mice compared with wild-type animals. Nevertheless, neuronal death in the hippocampal CA3 subfield and the neocortex was significantly reduced at 72 h in p53-deficient mice. Long-term continuous EEG telemetry recordings after status epilepticus determined that the sum duration of spontaneous seizures was significantly longer in p53-deficient compared with wild-type mice. Hippocampal damage and neuropeptide Y distribution at the end of chronic recordings was found to be similar between p53-deficient and wild-type mice. The present study identifies protracted KA-induced electrographic status as a novel outcome of p53 deficiency and shows that the absence of p53 leads to an exacerbated epileptic phenotype. Accordingly, targeting p53 to protect against status epilepticus or related neurologic insults may be offset by deleterious consequences of reduced p53 function during epileptogenesis or in chronic epilepsy.


Subject(s)
Seizures/physiopathology , Status Epilepticus/physiopathology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , CA3 Region, Hippocampal/metabolism , Electroencephalography/drug effects , Kainic Acid/toxicity , Mice , Mice, Knockout , Neurons/metabolism , Neuropeptide Y/metabolism , Phenotype , Seizures/chemically induced , Status Epilepticus/chemically induced , Tumor Suppressor Protein p53/genetics
6.
Cell Death Differ ; 17(3): 459-68, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19779495

ABSTRACT

Prolonged seizures (status epilepticus) are associated with brain region-specific regulation of apoptosis-associated signaling pathways. Bcl-2 homology domain 3-only (BH3) members of the Bcl-2 gene family are of interest as possible initiators of mitochondrial dysfunction and release of apoptogenic molecules after seizures. Previously, we showed that expression of the BH3-only protein, Bcl-2 interacting mediator of cell death (Bim), increased in the rat hippocampus but not in the neocortex after focal-onset status epilepticus. In this study, we examined Bim expression in mice and compared seizure damage between wild-type and Bim-deficient animals. Status epilepticus induced by intra-amygdala kainic acid (KA) caused extensive neuronal death within the ipsilateral hippocampal CA3 region. Hippocampal activation of factors associated with transcriptional and posttranslational activation of Bim, such as CHOP and c-Jun NH(2)-terminal kinases, was significant within 1 h. Upregulation of bim mRNA was evident after 2 h and Bim protein increased between 4 and 24 h. Hippocampal CA3 neurodegeneration was reduced in Bim-deficient mice compared with wild-type animals after seizures in vivo, and short interfering RNA molecules targeting bim reduced cell death after KA treatment of hippocampal organotypic cultures. In contrast, neocortical Bim expression declined after status epilepticus, and neocortex damage in Bim-deficient mice was comparable with that in wild-type animals. These results show region-specific differential contributions of Bim to seizure-induced neuronal death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Neocortex/metabolism , Neuroprotective Agents/metabolism , Proto-Oncogene Proteins/metabolism , Status Epilepticus/metabolism , Animals , Anthracenes/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Hippocampus/cytology , Hippocampus/pathology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Kainic Acid/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neocortex/cytology , Proto-Oncogene Proteins/genetics , Rats , Status Epilepticus/chemically induced , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
7.
Neuroscience ; 150(2): 467-77, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17935890

ABSTRACT

A neuroprotected state can be acquired by preconditioning brain with a stimulus that is subthreshold for damage (tolerance). Acquisition of tolerance involves coordinate, bi-directional changes to gene expression levels and the re-programmed phenotype is determined by the preconditioning stimulus. While best studied in ischemic brain there is evidence brief seizures can confer tolerance against prolonged seizures (status epilepticus). Presently, we developed a model of epileptic preconditioning in mice and used microarrays to gain insight into the transcriptional phenotype within the target hippocampus at the time tolerance had been acquired. Epileptic tolerance was induced by an episode of non-damaging seizures in adult C57Bl/6 mice using a systemic injection of kainic acid. Neuron and DNA damage-positive cell counts 24 h after status epilepticus induced by intraamygdala microinjection of kainic acid revealed preconditioning given 24 h prior reduced CA3 neuronal death by approximately 45% compared with non-tolerant seizure mice. Microarray analysis of over 39,000 transcripts (Affymetrix 430 2.0 chip) from microdissected CA3 subfields was undertaken at the point at which tolerance was acquired. Results revealed a unique profile of small numbers of equivalently up- and down-regulated genes with biological functions that included transport and localization, ubiquitin metabolism, apoptosis and cell cycle control. Select microarray findings were validated post hoc by real-time polymerase chain reaction and Western blotting. The present study defines a paradigm for inducing epileptic preconditioning in mice and first insight into the global transcriptome of the seizure-damage refractory brain.


Subject(s)
Brain Damage, Chronic/physiopathology , Brain Damage, Chronic/therapy , Epilepsy/physiopathology , Gene Expression/physiology , Hippocampus/physiopathology , Animals , Brain Damage, Chronic/etiology , Convulsants/therapeutic use , Disease Models, Animal , Down-Regulation/genetics , Epilepsy/complications , Excitatory Amino Acid Agonists/therapeutic use , Gene Expression Profiling/methods , Hippocampus/metabolism , Ischemic Preconditioning/methods , Kainic Acid/therapeutic use , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Nerve Degeneration/therapy , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , RNA, Messenger/metabolism , Status Epilepticus/physiopathology , Status Epilepticus/prevention & control , Status Epilepticus/therapy , Treatment Outcome , Up-Regulation/genetics
8.
J Neurosci Res ; 67(6): 713-9, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11891784

ABSTRACT

Several lines of evidence have indicated that changes in the structure of neuronal cytoskeleton provide the support for the dramatic morphological changes that occur during neuronal differentiation. It has been proposed that microtubule-associated proteins can contribute to the development of this phenomenon by controlling the dynamic properties of microtubules. In this report we have characterized the effect of the combined suppression of MAP1B and tau, and MAP1B and MAP2 on neuronal polarization in cultured hippocampal cells grown on a laminin-containing substrate. We have taken advantage of the use of a mouse line deficient in MAP1B expression obtained by the gene trapping approach. In addition to this engineered mice line we used the antisense oligonucleotide approach to induce the suppression of tau or MAP2, in wild type and MAP1B-deficient neurons. Together these results show a synergistic role for MAP1B/MAP2 and MAP1B/TAU.


Subject(s)
Cell Polarity/physiology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cells, Cultured , Female , Fluorescent Antibody Technique , Hippocampus/cytology , Male , Mice , Mice, Knockout , Oligonucleotides, Antisense/pharmacology , Polylysine , Pregnancy , tau Proteins/genetics , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...