Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Epilepsia ; 62(7): 1665-1676, 2021 07.
Article in English | MEDLINE | ID: mdl-34002394

ABSTRACT

OBJECTIVE: Dravet syndrome (DS) is a rare but catastrophic genetic epilepsy, with 80% of patients carrying a mutation in the SCN1A gene. Currently, no antiseizure drug (ASD) exists that adequately controls seizures. In the clinic, individuals with DS often present first with a febrile seizure and, subsequently, generalized tonic-clonic seizures that can continue throughout life. To facilitate the development of ASDs for DS, the contract site of the National Institute of Neurological Disorders and Stroke (NINDS) Epilepsy Therapy Screening Program (ETSP) has evaluated a mouse model of DS using the conditional knock-in Scn1aA1783V/WT mouse. METHODS: Survival rates and temperature thresholds for Scn1aA1783V/WT were determined. Prototype ASDs were administered via intraperitoneal injections at the time-to-peak effect, which was previously determined, prior to the induction of hyperthermia-induced seizures. ASDs were considered effective if they significantly increased the temperature at which Scn1aA1783V/WT mice had seizures. RESULTS: Approximately 50% of Scn1aA1783V/WT survive to adulthood and all have hyperthermia-induced seizures. The results suggest that hyperthermia-induced seizures in this model of DS are highly refractory to a battery of ASDs. Exceptions were clobazam, tiagabine, levetiracetam, and the combination of clobazam and valproic acid with add-on stiripentol, which elevated seizure thresholds. SIGNIFICANCE: Overall, the data demonstrate that the proposed model for DS is suitable for screening novel compounds for the ability to block hyperthermia-induced seizures and that heterozygous mice can be evaluated repeatedly over the course of several weeks, allowing for higher throughput screening.


Subject(s)
Anticonvulsants/therapeutic use , Epilepsies, Myoclonic/drug therapy , Seizures/drug therapy , Seizures/etiology , Animals , Body Temperature , Dioxolanes/therapeutic use , Drug Resistant Epilepsy/genetics , Drug Therapy, Combination , Epilepsies, Myoclonic/genetics , Female , Gene Knock-In Techniques , High-Throughput Screening Assays , Hyperthermia/complications , Injections, Intraperitoneal , Male , Mice , NAV1.1 Voltage-Gated Sodium Channel/genetics , National Institute of Neurological Disorders and Stroke (U.S.) , United States
2.
Exp Neurol ; 318: 50-60, 2019 08.
Article in English | MEDLINE | ID: mdl-31022385

ABSTRACT

The contribution of glial transporters to glutamate movement across the membrane has been identified as a potential target for anti-seizure therapies. Two such glutamate transporters, GLT-1 and system xc-, are expressed on glial cells, and modulation of their expression and function have been identified as a means by which seizures, neuronal injury, and gliosis can be reduced in models of brain injury. While GLT-1 is responsible for the majority of glutamate uptake in the brain, system xc- releases glutamate in the extracellular cleft in exchange for cystine and represents as such the major source of hippocampal extracellular glutamate. Using the Theiler's Murine Encephalomyelitis Virus (TMEV) model of viral-induced epilepsy, we have taken two well-studied approaches, one pharmacological, one genetic, to investigate the potential role(s) of GLT-1 and system xc- in TMEV-induced pathology. Our findings suggest that the methods we utilized to modulate these glial transporters, while effective in other models, are not sufficient to reduce the number or severity of behavioral seizures in TMEV-infected mice. However, genetic knockout of xCT, the specific subunit of system xc-, may have cellular effects, as we observed a slight decrease in neuronal injury caused by TMEV and an increase in astrogliosis in the CA1 region of the hippocampus. Furthermore, xCT knockout caused an increase in GLT-1 expression selectively in the cortex. These findings have significant implications for both the characterization of the TMEV model as well as for future efforts to discover novel and effective anti-seizure drugs.


Subject(s)
Amino Acid Transport System y+/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Seizures/metabolism , Animals , Brain/pathology , Cardiovirus Infections/complications , Cardiovirus Infections/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Seizures/pathology , Seizures/virology , Theilovirus
3.
eNeuro ; 4(2)2017.
Article in English | MEDLINE | ID: mdl-28497109

ABSTRACT

Central nervous system infection can induce epilepsy that is often refractory to established antiseizure drugs. Previous studies in the Theiler's murine encephalomyelitis virus (TMEV)-induced mouse model of limbic epilepsy have demonstrated the importance of inflammation, especially that mediated by tumor necrosis factor-α (TNFα), in the development of acute seizures. TNFα modulates glutamate receptor trafficking via TNF receptor 1 (TNFR1) to cause increased excitatory synaptic transmission. Therefore, we hypothesized that an increase in TNFα signaling after TMEV infection might contribute to acute seizures. We found a significant increase in both mRNA and protein levels of TNFα and the protein expression ratio of TNF receptors (TNFR1:TNFR2) in the hippocampus, a brain region most likely involved in seizure initiation, after TMEV infection, which suggests that TNFα signaling, predominantly through TNFR1, may contribute to limbic hyperexcitability. An increase in hippocampal cell-surface glutamate receptor expression was also observed during acute seizures. Although pharmacological inhibition of TNFR1-mediated signaling had no effect on acute seizures, several lines of genetically modified animals deficient in either TNFα or TNFRs had robust changes in seizure incidence and severity after TMEV infection. TNFR2-/- mice were highly susceptible to developing acute seizures, suggesting that TNFR2-mediated signaling may provide beneficial effects during the acute seizure period. Taken together, the present results suggest that inflammation in the hippocampus, caused predominantly by TNFα signaling, contributes to hyperexcitability and acute seizures after TMEV infection. Pharmacotherapies designed to suppress TNFR1-mediated or augment TNFR2-mediated effects of TNFα may provide antiseizure and disease-modifying effects after central nervous system infection.


Subject(s)
Hippocampus/metabolism , Seizures/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Epilepsy, Temporal Lobe/virology , Hippocampus/virology , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor, Type II/metabolism , Seizures/pathology , Seizures/virology , Signal Transduction , Temporal Lobe/pathology , Theilovirus
4.
Epilepsia ; 57(12): 1958-1967, 2016 12.
Article in English | MEDLINE | ID: mdl-27739576

ABSTRACT

OBJECTIVE: Infection with Theiler's murine encephalomyelitis virus (TMEV) in C57Bl/6J mice induces acute seizures and development of spontaneous recurrent seizures and behavioral comorbidities weeks later. The present studies sought to determine whether acute therapeutic intervention with an anti-inflammatory-based approach could prevent or modify development of TMEV-induced long-term behavioral comorbidities. Valproic acid (VPA), in addition to its prototypical anticonvulsant properties, inhibits histone deacetylase (HDAC) activity, which may alter expression of the inflammasome. Minocycline (MIN) has previously demonstrated an antiseizure effect in the TMEV model via direct anti-inflammatory mechanisms, but the long-term effect of MIN treatment on the development of chronic behavioral comorbidities is unknown. METHODS: Mice infected with TMEV were acutely administered MIN (50 mg/kg, b.i.d. and q.d.) or VPA (100 mg/kg, q.d.) during the 7-day viral infection period. Animals were evaluated for acute seizure severity and subsequent development of chronic behavioral comorbidities and seizure threshold. RESULTS: Administration of VPA reduced the proportion of mice with seizures, delayed onset of symptomatic seizures, and reduced seizure burden during the acute infection. This was in contrast to the effects of administration of once-daily MIN, which did not affect the proportion of mice with seizures or delay onset of acute symptomatic seizures. However, VPA-treated mice were no different from vehicle (VEH)-treated mice in long-term behavioral outcomes, including open field activity and seizure threshold. Once-daily MIN treatment, despite no effect on the maximum observed Racine stage seizure severity, was associated with improved long-term behavioral outcomes and normalized seizure threshold. SIGNIFICANCE: Acute seizure control alone is insufficient to modify chronic disease comorbidities in the TMEV model. This work further supports the role of an inflammatory response in the development of chronic behavioral comorbidities and further highlights the utility of this platform for the development of mechanistically novel pharmacotherapies for epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Behavior, Animal/drug effects , Epilepsy, Temporal Lobe , Minocycline/therapeutic use , Theilovirus/pathogenicity , Valproic Acid/therapeutic use , Animals , Anxiety Disorders/drug therapy , Anxiety Disorders/etiology , Body Weight/drug effects , Chi-Square Distribution , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/virology , Exploratory Behavior/drug effects , Mice , Motor Activity/drug effects , Psychomotor Performance/drug effects , Rotarod Performance Test
5.
J Neuropathol Exp Neurol ; 75(4): 366-78, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26945036

ABSTRACT

It is estimated that 30%-40% of epilepsy patients are refractory to therapy and animal models are useful for the identification of more efficacious therapeutic agents. Various well-characterized syndrome-specific models are needed to assess their relevance to human seizure disorders and their validity for testing potential therapies. The corneal kindled mouse model of temporal lobe epilepsy (TLE) allows for the rapid screening of investigational compounds, but there is a lack of information as to the specific inflammatory pathology in this model. Similarly, the Theiler murine encephalomyelitis virus (TMEV) model of TLE may prove to be useful for screening, but quantitative assessment of hippocampal pathology is also lacking. We used immunohistochemistry to characterize and quantitate acute neuronal injury and inflammatory features in dorsal CA1 and dentate gyrus regions and in the directly overlying posterior parietal cortex at 2 time points in each of these TLE models. Corneal kindled mice were observed to have astrogliosis, but not microgliosis or neuron cell death. In contrast, TMEV-injected mice had astrogliosis, microgliosis, neuron death, and astrocyte and microglial proliferation. Our results suggest that these 2 animal models might be appropriate for evaluation of distinct therapies for TLE.


Subject(s)
Cell Proliferation/physiology , Disease Models, Animal , Epilepsy, Temporal Lobe/complications , Gliosis/etiology , Neuroglia/pathology , Neurons/pathology , Animals , Calcium-Binding Proteins/metabolism , Cardiovirus Infections/complications , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/virology , Fluoresceins/metabolism , Ki-67 Antigen/metabolism , Kindling, Neurologic/physiology , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Microscopy, Confocal , Nerve Tissue Proteins/metabolism , Theilovirus/pathogenicity
6.
J Pharmacol Exp Ther ; 353(2): 318-29, 2015 May.
Article in English | MEDLINE | ID: mdl-25755209

ABSTRACT

Central nervous system infections can underlie the development of epilepsy, and Theiler's murine encephalomyelitis virus (TMEV) infection in C57BL/6J mice provides a novel model of infection-induced epilepsy. Approximately 50-65% of infected mice develop acute, handling-induced seizures during the infection. Brains display acute neuropathology, and a high number of mice develop spontaneous, recurrent seizures and behavioral comorbidities weeks later. This study characterized the utility of this model for drug testing by assessing whether antiseizure drug treatment during the acute infection period attenuates handling-induced seizures, and whether such treatment modifies associated comorbidities. Male C57BL/6J mice infected with TMEV received twice-daily valproic acid (VPA; 200 mg/kg), carbamazepine (CBZ; 20 mg/kg), or vehicle during the infection (days 0-7). Mice were assessed twice daily during the infection period for handling-induced seizures. Relative to vehicle-treated mice, more CBZ-treated mice presented with acute seizures; VPA conferred no change. In mice displaying seizures, VPA, but not CBZ, reduced seizure burden. Animals were then randomly assigned to acute and long-term follow-up. VPA was associated with significant elevations in acute (day 8) glial fibrillary acidic protein (astrocytes) immunoreactivity, but did not affect NeuN (neurons) immunoreactivity. Additionally, VPA-treated mice showed improved motor performance 15 days postinfection (DPI). At 36 DPI, CBZ-treated mice traveled significantly less distance through the center of an open field, indicative of anxiety-like behavior. CBZ-treated mice also presented with significant astrogliosis 36 DPI. Neither CBZ nor VPA prevented long-term reductions in NeuN immunoreactivity. The TMEV model thus provides an etiologically relevant platform to evaluate potential treatments for acute seizures and disease modification.


Subject(s)
Anticonvulsants/pharmacology , Behavior, Animal/drug effects , Carbamazepine/pharmacology , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/etiology , Theilovirus/physiology , Valproic Acid/pharmacology , Animals , Anticonvulsants/adverse effects , Anticonvulsants/therapeutic use , Anxiety/chemically induced , Carbamazepine/adverse effects , Carbamazepine/therapeutic use , Cardiovirus Infections/complications , Comorbidity , DNA-Binding Proteins , Disease Models, Animal , Epilepsy, Temporal Lobe/complications , Epilepsy, Temporal Lobe/virology , Glial Fibrillary Acidic Protein , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Rotarod Performance Test , Theilovirus/drug effects , Time Factors , Valproic Acid/adverse effects , Valproic Acid/therapeutic use
7.
Neurobiol Dis ; 64: 98-106, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24412221

ABSTRACT

Viral infection of the CNS can result in encephalitis and acute seizures, increasing the risk for later-life epilepsy. We have previously characterized a novel animal model of temporal lobe epilepsy that recapitulates key sequela in the development of epilepsy following viral infection. C57BL/6J mice inoculated with the Daniel's strain of Theiler's Murine Encephalomyelitis Virus (TMEV; 3×10(5) PFU, i.c.) display acute limbic seizures that secondarily generalize. A majority of acutely seized animals develop spontaneous seizures weeks to months later. As part of our investigation, we sought to assess behavioral comorbidity following TMEV inoculation. Anxiety, depression, cognitive impairment, and certain psychoses are diagnosed in persons with epilepsy at rates far more frequent than in the general population. We used a battery of behavioral tests to assess anxiety, depression, cognitive impairment, and general health in acutely seized animals inoculated with TMEV and compared behavioral outcomes against age-matched controls receiving a sham injection. We determined that TMEV-seized animals are less likely to move through the exposed center of an open field and are less likely to enter into the lighted half of a light/dark box; both behaviors may be indicative of anxiety-like behavior. TMEV-seized animals also display early and persistent reductions in novel object exploration during novel object place tasks and do not improve in their ability to find a hidden escape platform in Morris water maze testing, indicative of impairment in episodic and spatial memory, respectively. Cresyl violet staining at 35 and 250 days after injection reveals bilateral reductions in hippocampal area, with extensive sclerosis of CA1 evident bilaterally along the rostral-caudal axis. Early and persistent behavioral changes in the TMEV model provide surrogate markers for assessing disease progression as well as endpoints in screening for the efficacy of novel compounds to manage both seizure burden and comorbid conditions.


Subject(s)
Anxiety Disorders/etiology , Cognition Disorders/etiology , Epilepsy, Temporal Lobe/complications , Epilepsy, Temporal Lobe/psychology , Hippocampus/pathology , Seizures/etiology , Acute Disease , Animals , Anxiety Disorders/pathology , Behavior, Animal , CA1 Region, Hippocampal/pathology , Comorbidity , Disease Models, Animal , Disease Progression , Epilepsy, Temporal Lobe/pathology , Male , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Neuropsychological Tests , Sclerosis/pathology , Seizures/pathology , Theilovirus , Time Factors
8.
PLoS Genet ; 5(9): e1000649, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19763161

ABSTRACT

A follow-up study of a large Utah family with significant linkage to chromosome 2q24 led us to identify a new febrile seizure (FS) gene, SCN9A encoding Na(v)1.7. In 21 affected members, we uncovered a potential mutation in a highly conserved amino acid, p.N641Y, in the large cytoplasmic loop between transmembrane domains I and II that was absent from 586 ethnically matched population control chromosomes. To establish a functional role for this mutation in seizure susceptibility, we introduced the orthologous mutation into the murine Scn9a ortholog using targeted homologous recombination. Compared to wild-type mice, homozygous Scn9a(N641Y/N641Y) knockin mice exhibit significantly reduced thresholds to electrically induced clonic and tonic-clonic seizures, and increased corneal kindling acquisition rates. Together, these data strongly support the SCN9A p.N641Y mutation as disease-causing in this family. To confirm the role of SCN9A in FS, we analyzed a collection of 92 unrelated FS patients and identified additional highly conserved Na(v)1.7 missense variants in 5% of the patients. After one of these children with FS later developed Dravet syndrome (severe myoclonic epilepsy of infancy), we sequenced the SCN1A gene, a gene known to be associated with Dravet syndrome, and identified a heterozygous frameshift mutation. Subsequent analysis of 109 Dravet syndrome patients yielded nine Na(v)1.7 missense variants (8% of the patients), all in highly conserved amino acids. Six of these Dravet syndrome patients with SCN9A missense variants also harbored either missense or splice site SCN1A mutations and three had no SCN1A mutations. This study provides evidence for a role of SCN9A in human epilepsies, both as a cause of FS and as a partner with SCN1A mutations.


Subject(s)
Abnormalities, Multiple/genetics , Epilepsy/complications , Epilepsy/genetics , Seizures, Febrile/etiology , Seizures, Febrile/genetics , Sodium Channels/genetics , Abnormalities, Multiple/physiopathology , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Base Sequence , DNA Mutational Analysis , Electroshock , Epilepsy/physiopathology , Female , Gene Knock-In Techniques , Humans , Kindling, Neurologic/physiology , Male , Mice , Molecular Sequence Data , Mutation/genetics , NAV1.1 Voltage-Gated Sodium Channel , NAV1.7 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Pedigree , Protein Subunits/genetics , Seizures, Febrile/physiopathology , Sequence Alignment , Sodium Channels/chemistry , Syndrome
9.
Epilepsia ; 50(7): 1752-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19453707

ABSTRACT

PURPOSE: Benign familial neonatal convulsions (BFNC) is caused by mutations in the KCNQ2 and KCNQ3 genes, which encode subunits of the M-type potassium channel. The purpose of this study was to examine the effects of orthologous BFNC-causing mutations on seizure thresholds and the acquisition of corneal kindling in mice with heterozygous expression of the mutations. METHODS: The effects of the Kcnq2 gene A306T mutation and the Kcnq3 gene G311V mutation were determined for minimal clonic, minimal tonic hindlimb extension, and partial psychomotor seizures. The rate of corneal kindling acquisition was also determined for Kcnq2 A306T and Kcnq3 G311V mice. RESULTS: Seizure thresholds were significantly altered relative to wild-type animals in the minimal clonic, minimal tonic hindlimb extension, and partial psychomotor seizure models. Differences in seizure threshold were found to be dependent on the mutation expressed, the seizure testing paradigm, the genetic background strain, and the gender of the animal. Mutations in Kcnq2 and Kcnq3 were associated with an increased rate of corneal kindling. In the Kcnq2 A306T mice, an increased incidence of death occurred during and immediately following the conclusion of the kindling acquisition period. CONCLUSIONS: These results suggest that genetic alterations in the subunits that underlie the M-current and cause BFNC alter seizure susceptibility in a sex-, mouse strain-, and seizure-test dependent manner. Although the heterozygous mice do not appear to have spontaneous seizures, the increased seizure susceptibility and incidence of death during and after kindling suggests that these mutations lead to altered excitability in these animals.


Subject(s)
Epilepsy, Benign Neonatal/genetics , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/genetics , Kindling, Neurologic/physiology , Mutation/genetics , Seizures/genetics , Action Potentials/physiology , Animals , Disease Models, Animal , Electric Stimulation , Electrocardiography , Epilepsy, Benign Neonatal/physiopathology , Female , Gene Knock-In Techniques/methods , Genetic Predisposition to Disease , Heterozygote , Humans , KCNQ2 Potassium Channel/physiology , KCNQ3 Potassium Channel/physiology , Kindling, Neurologic/genetics , Male , Mice , Mutation, Missense/genetics , Nerve Tissue Proteins/genetics , Seizures/physiopathology , Sex Factors
10.
J Physiol ; 586(14): 3405-23, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18483067

ABSTRACT

The childhood epilepsy syndrome of benign familial neonatal convulsions (BFNC) exhibits the remarkable feature of clinical remission within a few weeks of onset and a favourable prognosis, sparing cognitive abilities despite persistent expression of the mutant KCNQ2 or KCNQ3 potassium channels throughout adulthood. To better understand such dynamic neuroprotective plasticity within the developing brain, we introduced missense mutations that underlie human BFNC into the orthologous murine Kcnq2 (Kv7.2) and Kcnq3 (Kv7.3) genes. Mutant mice were examined for altered thresholds to induced seizures, spontaneous seizure characteristics, hippocampal histology, and M-current properties of CA1 hippocampal pyramidal neurons. Adult Kcnq2(A306T/+) and Kcnq3(G311V/+) heterozygous knock-in mice exhibited reduced thresholds to electrically induced seizures compared to wild-type littermate mice. Both Kcnq2(A306T/A306T) and Kcnq3(G311V/G311V) homozygous mutant mice exhibited early onset spontaneous generalized tonic-clonic seizures concurrent with a significant reduction in amplitude and increased deactivation kinetics of the neuronal M-current. Mice had recurrent seizures into adulthood that triggered molecular plasticity including ectopic neuropeptide Y (NPY) expression in granule cells, but without hippocampal mossy fibre sprouting or neuronal loss. These novel knocking mice recapitulate proconvulsant features of the human disorder yet show that inherited M-current defects spare granule cells from reactive changes in adult hippocampal networks. The absence of seizure-induced pathology found in these epileptic mouse models parallels the benign neurodevelopmental cognitive profile exhibited by the majority of BFNC patients.


Subject(s)
Epilepsy, Benign Neonatal/genetics , KCNQ2 Potassium Channel/genetics , KCNQ3 Potassium Channel/genetics , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Synapses/physiology , Action Potentials/physiology , Animals , Disease Models, Animal , Electrocardiography , Gene Expression Regulation , Humans , KCNQ2 Potassium Channel/metabolism , KCNQ3 Potassium Channel/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Neurons/physiology , Seizures/genetics , Seizures/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...