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.
Brain ; 144(5): 1576-1589, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33769452

ABSTRACT

Seizures can emerge from multiple or large foci in temporal lobe epilepsy, complicating focally targeted strategies such as surgical resection or the modulation of the activity of specific hippocampal neuronal populations through genetic or optogenetic techniques. Here, we evaluate a strategy in which optogenetic activation of medial septal GABAergic neurons, which provide extensive projections throughout the hippocampus, is used to control seizures. We utilized the chronic intrahippocampal kainate mouse model of temporal lobe epilepsy, which results in spontaneous seizures and as is often the case in human patients, presents with hippocampal sclerosis. Medial septal GABAergic neuron populations were immunohistochemically labelled and were not reduced in epileptic conditions. Genetic labelling with mRuby of medial septal GABAergic neuron synaptic puncta and imaging across the rostral to caudal extent of the hippocampus, also indicated an unchanged number of putative synapses in epilepsy. Furthermore, optogenetic stimulation of medial septal GABAergic neurons consistently modulated oscillations across multiple hippocampal locations in control and epileptic conditions. Finally, wireless optogenetic stimulation of medial septal GABAergic neurons, upon electrographic detection of spontaneous hippocampal seizures, resulted in reduced seizure durations. We propose medial septal GABAergic neurons as a novel target for optogenetic control of seizures in temporal lobe epilepsy.


Subject(s)
GABAergic Neurons/physiology , Hippocampus/physiopathology , Optogenetics , Seizures/physiopathology , Septal Nuclei/physiopathology , Animals , Epilepsy, Temporal Lobe/physiopathology , Female , Male , Mice
2.
J Neurosci ; 39(16): 3159-3169, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30755487

ABSTRACT

Refractory focal epilepsy is a devastating disease for which there is frequently no effective treatment. Gene therapy represents a promising alternative, but treating epilepsy in this way involves irreversible changes to brain tissue, so vector design must be carefully optimized to guarantee safety without compromising efficacy. We set out to develop an epilepsy gene therapy vector optimized for clinical translation. The gene encoding the voltage-gated potassium channel Kv1.1, KCNA1, was codon optimized for human expression and mutated to accelerate the recovery of the channels from inactivation. For improved safety, this engineered potassium channel (EKC) gene was packaged into a nonintegrating lentiviral vector under the control of a cell type-specific CAMK2A promoter. In a blinded, randomized, placebo-controlled preclinical trial, the EKC lentivector robustly reduced seizure frequency in a male rat model of focal neocortical epilepsy characterized by discrete spontaneous seizures. When packaged into an adeno-associated viral vector (AAV2/9), the EKC gene was also effective at suppressing seizures in a male rat model of temporal lobe epilepsy. This demonstration of efficacy in a clinically relevant setting, combined with the improved safety conferred by cell type-specific expression and integration-deficient delivery, identify EKC gene therapy as being ready for clinical translation in the treatment of refractory focal epilepsy.SIGNIFICANCE STATEMENT Pharmacoresistant epilepsy affects up to 0.3% of the population. Although epilepsy surgery can be effective, it is limited by risks to normal brain function. We have developed a gene therapy that builds on a mechanistic understanding of altered neuronal and circuit excitability in cortical epilepsy. The potassium channel gene KCNA1 was mutated to bypass post-transcriptional editing and was packaged in a nonintegrating lentivector to reduce the risk of insertional mutagenesis. A randomized, blinded preclinical study demonstrated therapeutic effectiveness in a rodent model of focal neocortical epilepsy. Adeno-associated viral delivery of the channel to both hippocampi was also effective in a model of temporal lobe epilepsy. These results support clinical translation to address a major unmet need.


Subject(s)
Brain/metabolism , Epilepsy/therapy , Genetic Therapy , Kv1.1 Potassium Channel/genetics , Seizures/therapy , Animals , Disease Models, Animal , Epilepsy/genetics , Genetic Vectors , Kv1.1 Potassium Channel/metabolism , Male , Rats , Seizures/genetics
3.
eNeuro ; 5(2)2018.
Article in English | MEDLINE | ID: mdl-29780880

ABSTRACT

Network hyperexcitability is a feature of Alzheimer' disease (AD) as well as numerous transgenic mouse models of AD. While hyperexcitability in AD patients and AD animal models share certain features, the mechanistic overlap remains to be established. We aimed to identify features of network hyperexcitability in AD models that can be related to epileptiform activity signatures in AD patients. We studied network hyperexcitability in mice expressing amyloid precursor protein (APP) with mutations that cause familial AD, and compared a transgenic model that overexpresses human APP (hAPP) (J20), to a knock-in model expressing APP at physiological levels (APPNL/F). We recorded continuous long-term electrocorticogram (ECoG) activity from mice, and studied modulation by circadian cycle, behavioral, and brain state. We report that while J20s exhibit frequent interictal spikes (IISs), APPNL/F mice do not. In J20 mice, IISs were most prevalent during daylight hours and the circadian modulation was associated with sleep. Further analysis of brain state revealed that IIS in J20s are associated with features of rapid eye movement (REM) sleep. We found no evidence of cholinergic changes that may contribute to IIS-circadian coupling in J20s. In contrast to J20s, intracranial recordings capturing IIS in AD patients demonstrated frequent IIS in non-REM (NREM) sleep. The salient differences in sleep-stage coupling of IIS in APP overexpressing mice and AD patients suggests that different mechanisms may underlie network hyperexcitability in mice and humans. We posit that sleep-stage coupling of IIS should be an important consideration in identifying mouse AD models that most closely recapitulate network hyperexcitability in human AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Circadian Rhythm/physiology , Cortical Excitability/physiology , Disease Models, Animal , Epilepsy/physiopathology , Nerve Net/physiopathology , Sleep Stages/physiology , Amyloid beta-Peptides/genetics , Animals , Electrocorticography , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Brain ; 138(Pt 11): 3159-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26373601

ABSTRACT

Most patients with N-methyl D-aspartate-receptor antibody encephalitis develop seizures but the epileptogenicity of the antibodies has not been investigated in vivo. Wireless electroencephalogram transmitters were implanted into 23 C57BL/6 mice before left lateral ventricle injection of antibody-positive (test) or healthy (control) immunoglobulin G. Mice were challenged 48 h later with a subthreshold dose (40 mg/kg) of the chemo-convulsant pentylenetetrazol and events recorded over 1 h. Seizures were assessed by video observation of each animal and the electroencephalogram by an automated seizure detection programme. No spontaneous seizures were seen with the antibody injections. However, after the pro-convulsant, the test mice (n = 9) had increased numbers of observed convulsive seizures (P = 0.004), a higher total seizure score (P = 0.003), and a higher number of epileptic 'spike' events (P = 0.023) than the control mice (n = 6). At post-mortem, surprisingly, the total number of N-methyl D-aspartate receptors did not differ between test and control mice, but in test mice the levels of immunoglobulin G bound to the left hippocampus were higher (P < 0.0001) and the level of bound immunoglobulin G correlated with the seizure scores (R(2) = 0.8, P = 0.04, n = 5). Our findings demonstrate the epileptogenicity of N-methyl D-aspartate receptor antibodies in vivo, and suggest that binding of immunoglobulin G either reduced synaptic localization of N-methyl D-aspartate receptors, or had a direct effect on receptor function, which could be responsible for seizure susceptibility in this acute short-term model.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/immunology , Autoantibodies/immunology , Hippocampus/immunology , Receptors, N-Methyl-D-Aspartate/immunology , Seizures/immunology , Animals , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/complications , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/physiopathology , Brain/immunology , Brain/metabolism , Convulsants/toxicity , Disease Models, Animal , Electroencephalography , Female , Hippocampus/metabolism , Humans , Immunization, Passive , Immunoglobulin G , Mice , Mice, Inbred C57BL , Pentylenetetrazole/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/etiology , Seizures/physiopathology
5.
Sci Transl Med ; 4(161): 161ra152, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23147003

ABSTRACT

Neocortical epilepsy is frequently drug-resistant. Surgery to remove the epileptogenic zone is only feasible in a minority of cases, leaving many patients without an effective treatment. We report the potential efficacy of gene therapy in focal neocortical epilepsy using a rodent model in which epilepsy is induced by tetanus toxin injection in the motor cortex. By applying several complementary methods that use continuous wireless electroencephalographic monitoring to quantify epileptic activity, we observed increases in high frequency activity and in the occurrence of epileptiform events. Pyramidal neurons in the epileptic focus showed enhanced intrinsic excitability consistent with seizure generation. Optogenetic inhibition of a subset of principal neurons transduced with halorhodopsin targeted to the epileptic focus by lentiviral delivery was sufficient to attenuate electroencephalographic seizures. Local lentiviral overexpression of the potassium channel Kv1.1 reduced the intrinsic excitability of transduced pyramidal neurons. Coinjection of this Kv1.1 lentivirus with tetanus toxin fully prevented the occurrence of electroencephalographic seizures. Finally, administration of the Kv1.1 lentivirus to an established epileptic focus progressively suppressed epileptic activity over several weeks without detectable behavioral side effects. Thus, gene therapy in a rodent model can be used to suppress seizures acutely, prevent their occurrence after an epileptogenic stimulus, and successfully treat established focal epilepsy.


Subject(s)
Epilepsies, Partial/genetics , Epilepsies, Partial/therapy , Genetic Therapy , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/therapeutic use , Neocortex/pathology , Optogenetics , Animals , Disease Models, Animal , Electroencephalography , Epilepsies, Partial/pathology , Epilepsies, Partial/physiopathology , Lentivirus/genetics , Male , Neocortex/metabolism , Neocortex/physiopathology , Neurons/pathology , Pyramidal Cells/pathology , Rats , Rats, Sprague-Dawley , Tetanus Toxin/administration & dosage
6.
J Neurosci Methods ; 201(1): 106-15, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21820010

ABSTRACT

It has become increasingly evident that continuous EEG monitoring is necessary to observe the development of epilepsy in animals, and to determine the effect of drugs on spontaneous seizures. Telemetric recording systems have been increasingly used to monitor EEG in freely moving animals. One challenge faced by such systems is to monitor frequencies above 80Hz continuously for weeks. We present an implantable, 2.4-ml, telemetric sensor that can monitor EEG at 512 samples per second for eight weeks in a freely moving animal. With minor modifications, the same transmitter can operate at higher sample rates with a proportional decrease in operating life. Signal transmission is through bursts of 915-MHz radio power. The burst transmission and several other novel techniques reduce the transmitter's power consumption by two orders of magnitude while allowing 8 transmitters to share the same recording system. The use of radio-frequency transmission permits digitization within the sensor to sixteen-bit resolution, thus eliminating transmission-generated signal noise. The result is a signal with dynamic range 9mV, bandwidth 160Hz, input noise 12µV, and AC power interference less than 1µV. All circuit diagrams are open-source. Data acquisition takes place over the Internet using open-source software that works on multiple operating systems. The resulting system permits long-term, continuous, monitoring of EEG signals, therefore providing continuous and reliable data upon which to base studies of epilepsy in freely moving animals.


Subject(s)
Electroencephalography/trends , Telemetry/trends , Animals , Electrodes, Implanted/trends , Electroencephalography/instrumentation , Electroencephalography/methods , Male , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology , Telemetry/instrumentation , Telemetry/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...