Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 349: 113954, 2022 03.
Article in English | MEDLINE | ID: mdl-34922908

ABSTRACT

The discovery and development of novel antiseizure drugs (ASDs) that are effective in controlling pharmacoresistant spontaneous recurrent seizures (SRSs) continues to represent a significant unmet clinical need. The Epilepsy Therapy Screening Program (ETSP) has undertaken efforts to address this need by adopting animal models that represent the salient features of human pharmacoresistant epilepsy and employing these models for preclinical testing of investigational ASDs. One such model that has garnered increased interest in recent years is the mouse variant of the Intra-Amygdala Kainate (IAK) microinjection model of mesial temporal lobe epilepsy (MTLE). In establishing a version of this model, several methodological variables were evaluated for their effect(s) on pertinent quantitative endpoints. Although administration of a benzodiazepine 40 min after kainate (KA) induced status epilepticus (SE) is commonly used to improve survival, data presented here demonstrates similar outcomes (mortality, hippocampal damage, latency periods, and 90-day SRS natural history) between mice given midazolam and those that were not. Using a version of this model that did not interrupt SE with a benzodiazepine, a 90-day natural history study was performed and survival, latency periods, SRS frequencies and durations, and SRS clustering data were quantified. Finally, an important step towards model adoption is to assess the sensitivities or resistances of SRSs to a panel of approved and clinically used ASDs. Accordingly, the following ASDs were evaluated for their effects on SRSs in these mice: phenytoin (20 mg/kg, b.i.d.), carbamazepine (30 mg/kg, t.i.d.), valproate (240 mg/kg, t.i.d.), diazepam (4 mg/kg, b.i.d.), and phenobarbital (25 and 50 mg/kg, b.i.d.). Valproate, diazepam, and phenobarbital significantly attenuated SRS frequency relative to vehicle controls at doses devoid of observable adverse behavioral effects. Only diazepam significantly increased seizure freedom. Neither phenytoin nor carbamazepine significantly altered SRS frequency or freedom under these experimental conditions. These data demonstrate that SRSs in this IAK model of MTLE are pharmacoresistant to two representative sodium channel-inhibiting ASDs (phenytoin and carbamazepine) and partially sensitive to GABA receptor modulating ASDs (diazepam and phenobarbital) or a mixed-mechanism ASD (valproate). Accordingly, this model is being incorporated into the NINDS-funded ETSP testing platform for treatment resistant epilepsy.


Subject(s)
Amygdala , Anticonvulsants/therapeutic use , Convulsants , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy , Kainic Acid , Seizures/chemically induced , Seizures/drug therapy , Animals , Behavior, Animal , Convulsants/administration & dosage , Diazepam/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Resistant Epilepsy/chemically induced , Drug Resistant Epilepsy/drug therapy , Epilepsy, Temporal Lobe/psychology , Kainic Acid/administration & dosage , Male , Mice , Mice, Inbred C57BL , Microinjections , Seizures/psychology , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy
2.
Epilepsia ; 59(11): 2035-2048, 2018 11.
Article in English | MEDLINE | ID: mdl-30328622

ABSTRACT

OBJECTIVE: Approximately 30% of patients with epilepsy are refractory to existing antiseizure drugs (ASDs). Given that the properties of the central nervous systems of these patients are likely to be altered due to their epilepsy, tissues from rodents that have undergone epileptogenesis might provide a therapeutically relevant disease substrate for identifying compounds capable of attenuating pharmacoresistant seizures. To facilitate the development of such a model, this study describes the effects of classical glutamate receptor antagonists and 20 ASDs on recurrent epileptiform discharges (REDs) in brain slices derived from the kainate-induced status epilepticus model of temporal lobe epilepsy (KA-rats). METHODS: Horizontal brain slices containing the medial entorhinal cortex (mEC) were prepared from KA-rats, and REDs were recorded from the superficial layers. 6-cyano-7-nitroquinoxaline-2,3-dione, (2R)-amino-5-phosphonovaleric acid, tetrodotoxin, or ASDs were bath applied for 20 minutes. Concentration-dependent effects and half maximal effective concentration values were determined for RED duration, frequency, and amplitude. RESULTS: ASDs targeting sodium and potassium channels (carbamazepine, eslicarbazepine, ezogabine, lamotrigine, lacosamide, phenytoin, and rufinamide) attenuated REDs at concentrations near their average therapeutic plasma concentrations. γ-aminobutyric acid (GABA)ergic synaptic transmission-modulating ASDs (clobazam, midazolam, phenobarbital, stiripentol, tiagabine, and vigabatrin) attenuated REDs only at higher concentrations and, in some cases, prolonged RED durations. ASDs with other/mixed mechanisms of action (bumetanide, ethosuximide, felbamate, gabapentin, levetiracetam, topiramate, and valproate) and glutamate receptor antagonists weakly or incompletely inhibited RED frequency, increased RED duration, or had no significant effects. SIGNIFICANCE: Taken together, these data suggest that epileptiform activity recorded from the superficial layers of the mEC in slices obtained from KA-rats is differentially sensitive to existing ASDs. The different sensitivities of REDs to these ASDs may reflect persistent molecular, cellular, and/or network-level changes resulting from disease. These data are expected to serve as a foundation upon which future therapeutics may be differentiated and assessed for potentially translatable efficacy in patients with refractory epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Entorhinal Cortex/drug effects , Epilepsy/chemically induced , Epilepsy/drug therapy , Excitatory Amino Acid Agonists/toxicity , Excitatory Postsynaptic Potentials/drug effects , Kainic Acid/toxicity , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Entorhinal Cortex/pathology , In Vitro Techniques , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
3.
Neurochem Res ; 42(7): 1904-1918, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28303498

ABSTRACT

The successful identification of promising investigational therapies for the treatment of epilepsy can be credited to the use of numerous animal models of seizure and epilepsy for over 80 years. In this time, the maximal electroshock test in mice and rats, the subcutaneous pentylenetetrazol test in mice and rats, and more recently the 6 Hz assay in mice, have been utilized as primary models of electrically or chemically-evoked seizures in neurologically intact rodents. In addition, rodent kindling models, in which chronic network hyperexcitability has developed, have been used to identify new agents. It is clear that this traditional screening approach has greatly expanded the number of marketed drugs available to manage the symptomatic seizures associated with epilepsy. In spite of the numerous antiseizure drugs (ASDs) on the market today, the fact remains that nearly 30% of patients are resistant to these currently available medications. To address this unmet medical need, the National Institute of Neurological Disorders and Stroke (NINDS) Epilepsy Therapy Screening Program (ETSP) revised its approach to the early evaluation of investigational agents for the treatment of epilepsy in 2015 to include a focus on preclinical approaches to model pharmacoresistant seizures. This present report highlights the in vivo and in vitro findings associated with the initial pharmacological validation of this testing approach using a number of mechanistically diverse, commercially available antiseizure drugs, as well as several probe compounds that are of potential mechanistic interest to the clinical management of epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Drug Evaluation, Preclinical/standards , Drug Resistant Epilepsy/drug therapy , Animals , Drug Evaluation, Preclinical/methods , Drug Resistant Epilepsy/chemically induced , Drug Resistant Epilepsy/etiology , Electroshock/adverse effects , Kainic Acid/toxicity , Kindling, Neurologic/drug effects , Kindling, Neurologic/physiology , Male , Mice , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
4.
Cephalalgia ; 36(10): 924-35, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26568161

ABSTRACT

BACKGROUND: Though migraine is disabling and affects 12%-15% of the population, there are few drugs that have been developed specifically for migraine prevention. Valproic acid (VPA) is a broad-spectrum antiepileptic drug (AED) that is also used for migraine prophylaxis, but its clinical use is limited by its side effect profile. sec-Butylpropylacetamide (SPD) is a novel VPA derivative, designed to be more potent and tolerable than VPA, that has shown efficacy in animal seizure and pain models. METHODS: We evaluated SPD's antimigraine potential in the cortical spreading depression (CSD) and nitroglycerin (NTG) models of migraine. To evaluate SPD's mechanism of action, we performed whole-cell recordings on cultured cortical neurons and neuroblastoma cells. RESULTS: In the CSD model, the SPD-treated group showed a significantly lower median number of CSDs compared to controls. In the NTG-induced mechanical allodynia model, SPD dose-dependently reduced mechanical sensitivity compared to controls. SPD showed both a significant potentiation of GABA-mediated currents and a smaller but significant decrease in NMDA currents in cultured cortical neurons. Kainic acid-evoked currents and voltage-dependent sodium channel currents were not changed by SPD. CONCLUSIONS: These results demonstrate SPD's potential as a promising novel antimigraine compound, and suggest a GABAergic mechanism of action.


Subject(s)
Amides/therapeutic use , Anticonvulsants/therapeutic use , Migraine Disorders/prevention & control , Migraine Disorders/physiopathology , Valproic Acid/analogs & derivatives , Amides/pharmacology , Animals , Anticonvulsants/pharmacology , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Male , Mice , Mice, Inbred C57BL , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
5.
Proc Natl Acad Sci U S A ; 111(35): E3631-40, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25136135

ABSTRACT

Defective mitochondrial distribution in neurons is proposed to cause ATP depletion and calcium-buffering deficiencies that compromise cell function. However, it is unclear whether aberrant mitochondrial motility and distribution alone are sufficient to cause neurological disease. Calcium-binding mitochondrial Rho (Miro) GTPases attach mitochondria to motor proteins for anterograde and retrograde transport in neurons. Using two new KO mouse models, we demonstrate that Miro1 is essential for development of cranial motor nuclei required for respiratory control and maintenance of upper motor neurons required for ambulation. Neuron-specific loss of Miro1 causes depletion of mitochondria from corticospinal tract axons and progressive neurological deficits mirroring human upper motor neuron disease. Although Miro1-deficient neurons exhibit defects in retrograde axonal mitochondrial transport, mitochondrial respiratory function continues. Moreover, Miro1 is not essential for calcium-mediated inhibition of mitochondrial movement or mitochondrial calcium buffering. Our findings indicate that defects in mitochondrial motility and distribution are sufficient to cause neurological disease.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Disease Models, Animal , Mice, Inbred C57BL , Mitochondria/physiology , Paraplegia/genetics , rho GTP-Binding Proteins/genetics , Adenosine Triphosphate/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axonal Transport/physiology , Calcium/metabolism , Cell Respiration/physiology , Female , Male , Mice , Mice, Knockout , Microtubules/metabolism , Motor Neurons/metabolism , Paraplegia/metabolism , Paraplegia/pathology , Phenotype , rho GTP-Binding Proteins/metabolism
6.
Epilepsia ; 55(2): 214-23, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24447124

ABSTRACT

OBJECTIVE: Cognitive comorbidities are increasingly recognized as an equal (or even more disabling) aspect of epilepsy. In addition, the actions of some antiseizure drugs (ASDs) can impact learning and memory. Accordingly, the National Institute of Neurological Disorders and Stroke (NINDS) epilepsy research benchmarks call for the implementation of standardized protocols for screening ASDs for their amelioration or exacerbation of cognitive comorbidities. Long-term potentiation (LTP) is a widely used model for investigating synaptic plasticity and its relationship to learning and memory. Although the effects of some ASDs on LTP have been examined, none of these studies employed physiologically relevant induction stimuli such as theta-burst stimulation (TBS). To systematically evaluate the effects of multiple ASDs in the same preparation using physiologically relevant stimulation protocols, we examined the effects of a broad panel of existing ASDs on TBS-induced LTP in area CA1 of in vitro brain slices, prepared in either normal or sucrose-based artificial cerebrospinal fluid (ACSF), from C57BL/6 mice. METHODS: Coronal brain slices containing the dorsal hippocampus were made using either standard or sucrose-based ACSF. Recordings were obtained from four slices at a time using the Scientifica Slicemaster high throughput recording system. Slices exposed to ASDs were paired with slices from the opposite hemisphere that served as controls. Field excitatory postsynaptic potentials (fEPSPs) were recorded, and all ASDs were applied to slices by bath perfusion for 20 min prior to the induction stimulus. LTP was induced by TBS or by high-frequency stimulation (HFS). The following ASDs were examined: 100 µM phenobarbital (PB), 80 µM phenytoin (PHT), 50 µM carbamazepine (CBZ), 600 µM valproate (VPA), 60 µM topiramate (TPM), 60 µM lamotrigine (LTG), 100 µM levetiracetam (LEV), 10 µM ezogabine (EZG), and 30 µM tiagabine (TGB). RESULTS: Among voltage-gated sodium channel inhibitors, CBZ significantly attenuated TBS-induced LTP, PHT attenuated both TBS-induced LTP and post-tetanic potentiation (PTP), and LTG failed to affect LTP but did attenuate PTP. ASDs that modulate γ-aminobutyric acid (GABA)ergic synaptic transmission, such as PB and TGB, significantly attenuated LTP in brain slices prepared in sucrose-based ACSF but not standard ACSF. Third generation ASDs, such as LEV and TPM, did not affect LTP in ACSF- or sucrose-prepared brain slices. Although EZG failed to affect LTP, it did significantly attenuate PTP under both slicing conditions. VPA failed to affect LTP in area CA1, both in C57BL/6 mice and Sprague-Dawley rats, using TBS or HFS. However, VPA did attenuate TBS-induced LTP in the dentate gyrus (DG). SIGNIFICANCE: The results of experiments describe herein provide a comprehensive summary of the effects of many commonly used ASDs on short- and long-term synaptic plasticity while, for the first time, using physiologically relevant LTP induction protocols and slice preparations from mice. Furthermore, methodologic variables, such as brain slice preparation protocols, were explored. These results provide comparative knowledge of ASD effects on synaptic plasticity in the mouse hippocampus and may ultimately contribute to an understanding of the differences in the cognitive side effect profiles of ASDs and the prediction of cognitive dysfunction associated with novel investigational ASDs.


Subject(s)
Anticonvulsants/pharmacology , Hippocampus/physiology , Long-Term Potentiation/physiology , Theta Rhythm/physiology , Animals , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , Models, Neurological , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Theta Rhythm/drug effects
7.
Orthopedics ; 34(9): e561-9, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21902158

ABSTRACT

With increasing usage of many types of total elbow replacements, there is a continuing need for clinical series that report survivorship, complications and revisions, and performance of single types of implants over extended time periods. The purpose of this study was to assess the long-term effectiveness of all implants of the Sorbie-QUESTOR (SQ) unlinked surface arthroplasty conducted by a single surgeon (C.S.) over 15 years at a single site, and to determine whether there were diagnostic group differences. Between 1995 and 2002, 51 S-Q prosthetic elbows were implanted into 44 patients. The patient groups were hemophilia, rheumatoid arthritis, and "other," which included osteoarthritis, traumatic arthritis, psoriatic arthritis, and reactive arthritis. Annual evaluations included scores of pain, range of motion, and function. The most recent annual evaluation was included in the data set. Details of complications and revisions were recorded. The hemophiliac group had the best survival outcomes at 87.5%. Eighteen prostheses required revision or removal with all but 3 retained or replaced. Postoperatively, 73% rated their pain as 'slight' or 'none'. The hemophilia and rheumatoid arthritis groups made very large total flexion/extension gains. The rheumatoid arthritis group made significant forearm motion gains. Average functional assessment gains were nearly 2 grades of 5 functional levels and were significant for all groups. The S-Q surface arthroplasty has demonstrated long-term effectiveness in patients with a variety of elbow joint pathologies showing reduction in pain, large gains in joint range and function, and good long-term survival.


Subject(s)
Arthroplasty, Replacement, Elbow/instrumentation , Elbow Joint/surgery , Joint Prosthesis , Activities of Daily Living , Adult , Aged , Arthritis/surgery , Arthroplasty, Replacement, Elbow/methods , Elbow Joint/physiopathology , Female , Hemophilia A/surgery , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prosthesis Failure , Range of Motion, Articular , Young Adult
8.
Neurotoxicology ; 32(4): 392-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21396956

ABSTRACT

Glutamate induced excitotoxic injury through over-activation of N-methyl-D-aspartate receptors (NMDARs) plays a critical role in the development of many neurodegenerative diseases. The present study was undertaken to evaluate the role of CGX-1007 (Conantokin G) as a neuroprotective agent against NMDA-induced excitotoxicity. Conantokin G, a cone snail peptide isolated from Conus geographus is reported to selectively inhibit NR2B containing NMDARs with high specificity and is shown to have potent anticonvulsant and antinociceptive effects. CGX-1007 significantly reduced the excitotoxic cell death induced by NMDA in organotypic hippocampal brain slice cultures in a concentration-dependent manner. In contrast, ifenprodil, another NR2B specific antagonist failed to offer neuroprotection against NMDA-induced excitotoxicity. We further determined that the neuroprotection observed is likely due to the action of CGX-1007 at multiple NMDA receptor subtypes. In a series of electrophysiology experiments, CGX-1007 inhibited NMDA-gated currents in human embryonic kidney (HEK) 293 cells expressing NMDA receptors containing either NR1a/NR2B or NR1a/NR2A subunit combinations. CGX-1007 produced a weak inhibition at NR1a/NR2C receptors, whereas it had no effect on NR1a/NR2D receptors. Further, the inhibition of NMDA receptors by CGX-1007 was voltage-dependent with greater inhibition seen at hyperpolarized membrane potentials. The voltage-dependence of CGX-1007 activity was also observed in recordings of NMDA-gated currents evoked in native receptors expressed in cortical neurons in culture. Based on our results, we conclude that CGX-1007 is a potent neuroprotective agent that acts as an antagonist at both NR2A and NR2B containing receptors.


Subject(s)
Conotoxins/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Neuroprotective Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Animals, Newborn , Cell Death/drug effects , Cytoprotection , Dose-Response Relationship, Drug , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Membrane Potentials , Mice , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors , Tissue Culture Techniques , Transfection
9.
Foot Ankle Int ; 29(3): 273-81, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18348822

ABSTRACT

BACKGROUND: Osteoarthritis of the first metatarsophalangeal (MTP) joint is a painful, disabling condition. It can interfere with the ability to run and even walk without pain. An implant of cobalt-chrome steel alloy to replace the base of the proximal phalanx is one solution. The purpose of this study is to review our results with one of these implants. MATERIALS AND METHODS: A series of 23 cases of hemiarthroplasty for the treatment of hallux rigidus from June 2000 to October 2001 has been evaluated using the AOFAS rating system, and the results are presented. RESULTS: The average preoperative AOFAS score was 57 (range, 39 to 80). The AOFAS score after hemiarthroplasty was 88 (range, 75 to 100) at last followup (34 to 72 months). There were no perioperative complications except for a small hematoma. Only one patient has required further surgery after 3 years for worsening of a preexisting tendency to hallux valgus. CONCLUSION: A hemiarthroplasty retained, in most cases, joint mobility, strength, and alignment while relieving pain. There was no evidence, at last followup, of component loosening or osteolysis.


Subject(s)
Arthroplasty, Replacement/instrumentation , Hallux Rigidus/surgery , Joint Prosthesis , Vitallium , Adult , Aged , Female , Follow-Up Studies , Hallux Rigidus/diagnostic imaging , Hallux Rigidus/physiopathology , Humans , Male , Middle Aged , Prosthesis Design , Radiography , Range of Motion, Articular , Treatment Outcome
10.
Epilepsy Res ; 79(1): 6-13, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18262393

ABSTRACT

Disruptions in GABAergic neurotransmission have been implicated in numerous CNS disorders, including epilepsy and neuropathic pain. Selective inhibition of neuronal and glial GABA transporter subtypes may offer unique therapeutic options for regaining balance between inhibitory and excitatory systems. The ability of two GABA transport inhibitors to modulate inhibitory tone via inhibition of mGAT1 (tiagabine) or mGAT2/BGT-1 (N-[4,4-bis(3-methyl-2-thienyl)-3-butenyl]-4-(methylamino-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol), also known as EF1502) was evaluated using an in vitro model of spontaneous interictal-like bursting (SB). SBs were recorded extracellularly in combined mEC-HC horizontal brain slices (400 microm; 31+/-1 degrees C) obtained from KA-treated rats. Slice recordings demonstrated that EF1502 exhibited a concentration-dependent reduction in SB frequency. EF1502 significantly reduced SB rate to 32% of control at the 30 microM concentration, while reducing the area and duration of SB activity to 60% and 46% of control, respectively, at the 10 microM concentration. In contrast, the GAT1 selective inhibitor tiagabine (3, 10, and 30 microM) was unable to significantly reduce the frequency of SB activity in the mEC, despite significantly reducing both the duration (51% of control) and area (58% of control) of the SB at concentrations as low as 3 microM. The ability of EF1502, but not tiagabine, to inhibit SBs in the mEC suggests that this in vitro model of pharmacoresistant SB activity is useful to differentiate between novel anticonvulsants with similar mechanisms of action and suggests a therapeutic potential for non-GAT1 transport inhibitors.


Subject(s)
Carrier Proteins/metabolism , Entorhinal Cortex/physiopathology , Evoked Potentials/physiology , Status Epilepticus/pathology , Animals , Carrier Proteins/antagonists & inhibitors , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Entorhinal Cortex/drug effects , Entorhinal Cortex/radiation effects , Evoked Potentials/drug effects , Evoked Potentials/radiation effects , GABA Agonists/pharmacology , GABA Plasma Membrane Transport Proteins , In Vitro Techniques , Isoxazoles/pharmacology , Kainic Acid , Male , Nipecotic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Status Epilepticus/chemically induced , Tiagabine
11.
Epilepsy Res ; 74(2-3): 97-106, 2007 May.
Article in English | MEDLINE | ID: mdl-17395429

ABSTRACT

Hyperexcitability in the medial entorhinal cortex-hippocampal (mEC-HC) circuit in the initial weeks after prolonged seizure activity may contribute to the epileptogenic process in animal models of temporal lobe epilepsy (TLE). The present study examined combined mEC-HC slices (400 microm) using field potential recordings 1-2 weeks following the multiple administration, low-dose kainic acid (KA) model of TLE [Hellier, J.L., Patrylo, P.R., Buckmaster, P.S., Dudek, F.E., 1998. Recurrent spontaneous motor seizures after repeated low-dose systemic treatment with kainate: assessment of a rat model of temporal lobe epilepsy. Epilepsy Res. 31, 73-84]. Field potential recordings in slices from KA-treated rats demonstrated hallmarks of hyperexcitability in the mEC and in the CA1 and CA3 cell body regions of the HC. Spontaneous burst (SB) activity was observed under baseline recording conditions in the mEC of several slices from KA-treated rats, but not in the slices from saline-treated control rats. Elevating ACSF [K(+)](o) (6mM) in the presence of picrotoxin (50 microM) increased SB rates in all slices tested. However, there was a significantly shorter latency to onset of bursting and prolonged evoked response durations in layer II of the mEC of slices from KA-treated rats versus those from controls. Neither carbamazepine (CBZ) nor phenytoin (PHT) abolished SB activity in slices from KA-treated rats; whereas, SB activity in slices from control rats was dose-dependently reduced at 100 microM CBZ. In contrast, the novel anticonvulsant retigabine (RGB) dramatically reduced SB frequency in both control and KA-treated groups. The hyperexcitability observed in combined mEC-HC brain slices from KA-treated rats suggests that the mEC, as well as the HC, may contribute to the epileptogenic process after KA-induced seizure activity. This model may provide an efficient, flexible in vitro paradigm for differentiating novel AEDs in a model of pharmacoresistant bursting.


Subject(s)
Anticonvulsants/therapeutic use , Carbamates/pharmacology , Carbamazepine/therapeutic use , Entorhinal Cortex/drug effects , Entorhinal Cortex/pathology , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/pathology , Phenylenediamines/pharmacology , Phenytoin/therapeutic use , Animals , Cell Count , Dose-Response Relationship, Drug , Drug Resistance , Electrophysiology , Epilepsy, Temporal Lobe/chemically induced , Evoked Potentials/drug effects , Excitatory Amino Acid Antagonists , Hippocampus/pathology , Ketamine , Male , Nerve Net/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...