Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Exp Med Biol ; 978: 255-275, 2017.
Article in English | MEDLINE | ID: mdl-28523551

ABSTRACT

Despite being clinically described 150 years ago, the mechanisms underlying amyotrophic lateral sclerosis (ALS) pathogenesis have not yet been fully understood. Studies in both animal models of ALS and human patients reveal a plethora of alterations such as increased glutamate-mediated excitotoxicity, redox stress, increased apoptosis, defective axonal transport, protein-misfolding events, mitochondrial impairment and sustained unregulated immune responses. Regardless of being sporadic or familiar ALS, the final outcome at the cellular level is the death of upper and lower motor neurons, and once diagnosed, ALS is typically lethal within the next 5 years. There are neither clear biomarkers nor therapeutic or disease-modifying treatments for ALS.Accumulating evidence supports the concept that epigenetic-driven modifications, including altered chromatin remodelling events, RNA editing and non-coding RNA molecules, might shed light into the pathogenic mechanisms underlying sporadic/familiar ALS onset and/or severity to facilitate the identification of effective therapies, early diagnosis and potentially early-stage therapeutic interventions to increase the survival outcome of ALS patients.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , Acetylation , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA Methylation/genetics , Disease Models, Animal , Environmental Exposure , Gene-Environment Interaction , Histone Code/genetics , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/physiology , Humans , MicroRNAs/genetics , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation , Oxidative Stress , Protein Processing, Post-Translational , Riluzole/therapeutic use , Superoxide Dismutase-1/deficiency , Superoxide Dismutase-1/genetics
2.
J Neurosci ; 36(22): 5920-32, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27251615

ABSTRACT

UNLABELLED: Neuroinflammation is thought to contribute to the pathogenesis and maintenance of temporal lobe epilepsy, but the underlying cell and molecular mechanisms are not fully understood. The P2X7 receptor is an ionotropic receptor predominantly expressed on the surface of microglia, although neuronal expression has also been reported. The receptor is activated by the release of ATP from intracellular sources that occurs during neurodegeneration, leading to microglial activation and inflammasome-mediated interleukin 1ß release that contributes to neuroinflammation. Using a reporter mouse in which green fluorescent protein is induced in response to the transcription of P2rx7, we show that expression of the receptor is selectively increased in CA1 pyramidal and dentate granule neurons, as well as in microglia in mice that developed epilepsy after intra-amygdala kainic acid-induced status epilepticus. P2X7 receptor levels were increased in hippocampal subfields in the mice and in resected hippocampus from patients with pharmacoresistant temporal lobe epilepsy. Cells transcribing P2rx7 in hippocampal slices from epileptic mice displayed enhanced agonist-evoked P2X7 receptor currents, and synaptosomes from these animals showed increased P2X7 receptor levels and altered calcium responses. A 5 d treatment of epileptic mice with systemic injections of the centrally available, potent, and specific P2X7 receptor antagonist JNJ-47965567 (30 mg/kg) significantly reduced spontaneous seizures during continuous video-EEG monitoring that persisted beyond the time of drug presence in the brain. Hippocampal sections from JNJ-47965567-treated animals obtained >5 d after treatment ceased displayed strongly reduced microgliosis and astrogliosis. The present study suggests that targeting the P2X7 receptor has anticonvulsant and possibly disease-modifying effects in experimental epilepsy. SIGNIFICANCE STATEMENT: Temporal lobe epilepsy is the most common and drug-resistant form of epilepsy in adults. Neuroinflammation is implicated as a pathomechanism, but the upstream mechanisms driving gliosis and how important this is for seizures remain unclear. In our study, we show that the ATP-gated P2X7 receptor is upregulated in experimental epilepsy and resected hippocampus from epilepsy patients. Targeting the receptor with a new centrally available antagonist, JNJ-47965567, suppressed epileptic seizures well beyond the time of treatment and reduced underlying gliosis in the hippocampus. The findings suggest a potential disease-modifying treatment for epilepsy based on targeting the P2X7 receptor.


Subject(s)
Epilepsy, Temporal Lobe/complications , Epilepsy, Temporal Lobe/drug therapy , Gliosis/drug therapy , Gliosis/etiology , Purinergic P2X Receptor Antagonists/therapeutic use , Seizures/drug therapy , Seizures/etiology , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Adolescent , Adult , Animals , Brain/metabolism , Brain/ultrastructure , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Epilepsy, Temporal Lobe/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Middle Aged , Nerve Tissue Proteins/metabolism , Niacinamide/analogs & derivatives , Niacinamide/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Piperazines/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , Young Adult
3.
Sci Rep ; 5: 17486, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26631939

ABSTRACT

The ATP-gated ionotropic P2X7 receptor (P2X7R) modulates glial activation, cytokine production and neurotransmitter release following brain injury. Levels of the P2X7R are increased in experimental and human epilepsy but the mechanisms controlling P2X7R expression remain poorly understood. Here we investigated P2X7R responses after focal-onset status epilepticus in mice, comparing changes in the damaged, ipsilateral hippocampus to the spared, contralateral hippocampus. P2X7R-gated inward currents were suppressed in the contralateral hippocampus and P2rx7 mRNA was selectively uploaded into the RNA-induced silencing complex (RISC), suggesting microRNA targeting. Analysis of RISC-loaded microRNAs using a high-throughput platform, as well as functional assays, suggested the P2X7R is a target of microRNA-22. Inhibition of microRNA-22 increased P2X7R expression and cytokine levels in the contralateral hippocampus after status epilepticus and resulted in more frequent spontaneous seizures in mice. The major pro-inflammatory and hyperexcitability effects of microRNA-22 silencing were prevented in P2rx7(-/-) mice or by treatment with a specific P2X7R antagonist. Finally, in vivo injection of microRNA-22 mimics transiently suppressed spontaneous seizures in mice. The present study supports a role for post-transcriptional regulation of the P2X7R and suggests therapeutic targeting of microRNA-22 may prevent inflammation and development of a secondary epileptogenic focus in the brain.


Subject(s)
Hippocampus/physiology , MicroRNAs/genetics , Receptors, Purinergic P2X7/genetics , Status Epilepticus/genetics , Animals , Astrocytes/pathology , Electroencephalography , Gene Expression Regulation , Hippocampus/physiopathology , Inflammation/genetics , Inflammation/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , RNA Interference , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/metabolism , Receptors, Purinergic P2X7/metabolism , Status Epilepticus/metabolism , Status Epilepticus/physiopathology
4.
CNS Neurosci Ther ; 20(6): 556-64, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24750893

ABSTRACT

AIMS: Early-life seizures, particularly when prolonged, may be harmful to the brain. Current pharmacotherapy is often ineffective; therefore, novel neuro- and/or glio-transmitter systems should be explored for targeting. The P2X7 receptor is a cation-permeable channel with trophic and excitability effects on neurons and glia which is activated by high amounts of ATP that may be released in the setting of injury after severe seizures. Here, we tested the effects of A-438079, a potent and selective P2X7 receptor antagonist in a lesional model of early-life status epilepticus. METHODS: Seizures were induced by intra-amygdala kainic acid in 10-day-old rat pups. Electrographic seizure severity, changes to P2X7 receptor expression, inflammatory responses and histological effects were evaluated. RESULTS: Seizures induced by intra-amygdala kainic acid increased levels of P2X7 receptor protein and interleukin-1ß and caused significant cell death within the ipsilateral hippocampus. A-438079 rapidly reached the brain following systemic injection in P10 rats. Intraperitoneal injection of A-438079 (5 and 15 mg/kg) 60 min after triggering seizures reduced seizure severity and neuronal death within the hippocampus. A-438079 had superior neuroprotective effects compared with an equally seizure-suppressive dose of phenobarbital (25 mg/kg). CONCLUSIONS: These results suggest P2X7 receptor antagonists may be suitable as frontline or adjunctive treatments of pediatric status epilepticus or other early-life seizures, particularly when associated with brain damage.


Subject(s)
Hippocampus/drug effects , Purinergic P2X Receptor Antagonists/therapeutic use , Pyridines/therapeutic use , Status Epilepticus/drug therapy , Status Epilepticus/pathology , Tetrazoles/therapeutic use , Amygdala/injuries , Amygdala/physiology , Animals , Animals, Newborn , Bumetanide/pharmacology , Bumetanide/therapeutic use , Cell Death/drug effects , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/toxicity , Female , Hippocampus/metabolism , Kainic Acid/toxicity , Male , Pyridines/metabolism , Quinazolines , Rats , Rats, Sprague-Dawley , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Status Epilepticus/chemically induced , Tetrazoles/metabolism
5.
Epilepsia ; 54(9): 1551-61, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23808395

ABSTRACT

PURPOSE: ATP is an essential transmitter/cotransmitter in neuron function and pathophysiology and has recently emerged as a potential contributor to prolonged seizures (status epilepticus) through the activation of the purinergic ionotropic P2X7 receptor (P2X7R). Increased P2X7R expression has been reported in the hippocampus, and P2X7R antagonists reduced seizure-induced damage to this brain region. However, status epilepticus also produces damage to the neocortex. The present study was designed to characterize P2X7R in the neocortex and assess effects of P2X7R antagonists on cortical injury after status epilepticus. METHODS: Status epilepticus was induced in mice by intraamygdala microinjection of kainic acid. Specific P2X7R inhibitors were administered into the ventricle before seizure induction, and cortical electroencephalography and behavior was recorded to assess seizure severity. P2X7R expression was examined in neocortex up to 24 h after status epilepticus, in epileptic mice, and in resected neocortex from patients with pharmacoresistent temporal lobe epilepsy (TLE). In addition, the induction of P2X7R after status epilepticus was investigated using transgenic P2X7R reporter mice, which express enhanced green fluorescent protein under the control of the p2x7r promoter. KEY FINDINGS: Status epilepticus resulted in increased P2X7R protein levels in the neocortex of mice. Neocortical P2X7 receptor levels were also elevated in mice that developed epilepsy after status epilepticus and in resected neocortex from patients with pharmacoresistent TLE. Immunohistochemistry determined that neurons were the major cell population transcribing the P2X7R in the neocortex within the first 8 h after status epilepticus, whereas in epileptic mice, P2X7R up-regulation occurred in microglia as well as in neurons. Pretreatment of mice with the specific P2X7R inhibitor A-438079 reduced electrographic and clinical seizure severity during status epilepticus and reduced seizure-induced neuronal death in the neocortex. SIGNIFICANCE: Our findings identify neurons in the neocortex as an important site of P2X7R up-regulation after status epilepticus and in epilepsy, and provide support for the possible use of P2X7R antagonists for the treatment of status epilepticus and prevention of seizure-induced brain damage.


Subject(s)
Anticonvulsants/pharmacology , Pyridines/pharmacology , Receptors, Purinergic P2X7/metabolism , Seizures/drug therapy , Status Epilepticus/metabolism , Tetrazoles/pharmacology , Animals , Cell Death/physiology , Hippocampus/cytology , Hippocampus/drug effects , Mice , Mice, Transgenic , Neocortex/drug effects , Neuroprotective Agents/pharmacology , Seizures/chemically induced , Status Epilepticus/chemically induced , Status Epilepticus/pathology , Up-Regulation/drug effects
6.
Brain ; 136(Pt 2): 577-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23361066

ABSTRACT

Hippocampal sclerosis is a frequent pathological finding in patients with temporal lobe epilepsy and can be caused by prolonged single or repeated brief seizures. Both DNA damage and endoplasmic reticulum stress have been implicated as underlying molecular mechanisms in seizure-induced brain injury. The CCAAT/enhancer-binding protein homologous protein (CHOP) is a transcriptional regulator induced downstream of DNA damage and endoplasmic reticulum stress, which can promote or inhibit apoptosis according to context. Recent work has proposed inhibition of CHOP as a suitable neuroprotective strategy. Here, we show that transcript and protein levels of CHOP increase in surviving subfields of the hippocampus after prolonged seizures (status epilepticus) in mouse models. CHOP was also elevated in the hippocampus from epileptic mice and patients with pharmacoresistant epilepsy. The hippocampus of CHOP-deficient mice was much more vulnerable to damage in mouse models of status epilepticus. Moreover, compared with wild-type animals, CHOP-deficient mice subject to status epilepticus developed more spontaneous seizures, displayed protracted hippocampal neurodegeneration and a deficit in a hippocampus-dependent object-place recognition task. The absence of CHOP was associated with a supra-maximal induction of p53 after status epilepticus, and inhibition of p53 abolished the cell death-promoting consequences of CHOP deficiency. The protective effect of CHOP could be partly explained by activating transcription of murine double minute 2 that targets p53 for degradation. These data demonstrate that CHOP is required for neuronal survival after seizures and caution against inhibition of CHOP as a neuroprotective strategy where excitotoxicity is an underlying pathomechanism.


Subject(s)
Neurons/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Seizures/metabolism , Transcription Factor CHOP/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Survival/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/pathology , Proto-Oncogene Proteins c-mdm2/physiology , Seizures/genetics , Seizures/pathology , Tumor Suppressor Protein p53/physiology
7.
Article in English | MEDLINE | ID: mdl-23320131

ABSTRACT

The P2X7 receptor is an ATP-gated non-selective cation-permeable ionotropic receptor selectively expressed in neurons and glia in the brain. Activation of the P2X7 receptor has been found to modulate neuronal excitability in the hippocampus and it has also been linked to microglia activation and neuroinflammatory responses. Accordingly, interest developed on the P2X7 receptor in disorders of the nervous system, including epilepsy. Studies show that expression of the P2X7 receptor is elevated in damaged regions of the brain after prolonged seizures (status epilepticus) in both neurons and glia. P2X7 receptor expression is also increased in the hippocampus in experimental epilepsy. Recent data show that mice lacking the P2X7 receptor display altered susceptibility to status epilepticus and that drugs targeting the P2X7 receptor have potent anticonvulsant effects. Together, this suggests that P2X7 receptor ligands may be useful adjunctive treatments for refractory status epilepticus or perhaps pharmacoresistant epilepsy. This review summarizes the evidence of P2X7 receptor involvement in the pathophysiology of epilepsy and the potential of drugs targeting this receptor for seizure control.

SELECTION OF CITATIONS
SEARCH DETAIL