Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
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.
Theranostics ; 10(18): 8415-8429, 2020.
Article in English | MEDLINE | ID: mdl-32724478

ABSTRACT

Rationale: Dysfunction or reduced levels of EAAT2 have been documented in epilepsy. We previously demonstrated the antiepileptic effects of Hsp90 inhibitor 17AAG in temporal lobe epilepsy by preventing EAAT2 degradation. Because of the potential toxicities of 17AAG, this study aimed to identify an alternative Hsp90 inhibitor with better performance on Hsp90 inhibition, improved blood-brain barrier penetration and minimal toxicity. Methods: We used cell-based screening and animal models of epilepsy, including mouse models of epilepsy and Alzheimer's disease, and a cynomolgus monkey model of epilepsy, to evaluate the antiepileptic effects of new Hsp90 inhibitors. Results: In both primary cultured astrocytes and normal mice, HSP990 enhanced EAAT2 levels at a lower dose than other Hsp90 inhibitors. In epileptic mice, administration of 0.1 mg/kg HSP990 led to upregulation of EAAT2 and inhibition of spontaneous seizures. Additionally, HSP990 inhibited seizures and improved cognitive functions in the APPswe/PS1dE9 transgenic model of Alzheimer's disease. In a cynomolgus monkey model of temporal lobe epilepsy, oral administration of low-dose HSP990 completely suppressed epileptiform discharges for up to 12 months, with no sign of hepatic and renal toxicity. Conclusions: These results support further preclinical studies of HSP990 treatment for temporal lobe epilepsy.


Subject(s)
Alzheimer Disease/drug therapy , Anticonvulsants/administration & dosage , Epilepsy, Temporal Lobe/drug therapy , Excitatory Amino Acid Transporter 2/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Administration, Oral , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Anticonvulsants/adverse effects , Astrocytes , Cells, Cultured , Cognition/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Female , HSP90 Heat-Shock Proteins/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Humans , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Macaca fascicularis , Male , Mice , Mice, Transgenic , Pentylenetetrazole/administration & dosage , Pentylenetetrazole/toxicity , Primary Cell Culture , Pyridones/adverse effects , Pyrimidines/adverse effects , Temporal Lobe/drug effects , Temporal Lobe/pathology , Up-Regulation/drug effects
3.
Neurosci Lett ; 622: 30-6, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27095588

ABSTRACT

In rodent models of epilepsy, EEG implantation surgery is an essential modality to evaluate electrographic seizures. The inflammatory consequences of EEG electrode-implantation and their resultant effects on seizure susceptibility are unclear. We evaluated electrode-implantation in a two-hit model of epileptogenesis in C57BL/6 mice that included brief, recurrent febrile seizures (FS) at P14 and kainic acid induced seizures (KA-SZ) at P28. During KA-SZ, latencies to first electrographic and behavioral seizures, seizure severity, and KA dose sensitivity were measured. Mice that received subdural screw electrode implants at P25 for EEG monitoring at P28 had significantly shorter latencies to seizures than sham mice, regardless of early life seizure experience. Electrode-implanted mice were sensitive to low dose KA as shown by high mortality rate at KA doses above 10mg/kg. We then directly compared electrode-implantation and KA-SZ in seizure naive CX3CR1(GFP/+) transgenic C57BL/6 mice, wherein microglia express green fluorescent protein (GFP), to determine if microglia activation related to surgery was associated with the increased seizure susceptibility in electrode-implanted mice from the two-hit model. Hippocampal microglia activation, as demonstrated by percent area GFP signal and GFP positive cell counts, prior to seizures was indistinguishable between electrode-implanted mice and controls, but was significantly greater in electrode-implanted mice following seizures. Electrode-implantation had a confounding priming effect on the inflammatory response to subsequent seizures.


Subject(s)
Epilepsy, Temporal Lobe/surgery , Animals , Dose-Response Relationship, Drug , Electrodes, Implanted/adverse effects , Electroencephalography , Epilepsy, Temporal Lobe/etiology , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/pathology , Hippocampus/surgery , Hyperthermia, Induced , Inflammation/etiology , Inflammation/pathology , Kainic Acid/administration & dosage , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology
4.
Mol Pain ; 122016.
Article in English | MEDLINE | ID: mdl-27030713

ABSTRACT

BACKGROUND: Stroke patients often suffer from a central neuropathic pain syndrome called central post-stroke pain. This syndrome is characterized by evoked pain hypersensitivity as well as spontaneous, on-going pain in the body area affected by the stroke. Clinical evidence strongly suggests a dysfunction in central pain pathways as an important pathophysiological factor in the development of central post-stroke pain, but the exact underlying mechanisms remain poorly understood. To elucidate the underlying pathophysiology of central post-stroke pain, we generated a mouse model that is based on a unilateral stereotactic lesion of the thalamic ventral posterolateral nucleus, which typically causes central post-stroke pain in humans. RESULTS: Behavioral analysis showed that the sensory changes in our model are comparable to the sensory abnormalities observed in patients suffering from central post-stroke pain. Surprisingly, pharmacological inhibition of spinal and peripheral key components of the pain system had no effect on the induction or maintenance of the evoked hypersensitivity observed in our model. In contrast, microinjection of lidocaine into the thalamic lesion completely reversed injury-induced hypersensitivity. CONCLUSIONS: These results suggest that the evoked hypersensitivity observed in central post-stroke pain is causally linked to on-going neuronal activity in the lateral thalamus.


Subject(s)
Pain/etiology , Pain/physiopathology , Stroke/complications , Stroke/physiopathology , Animals , Collagenases/administration & dosage , Disease Models, Animal , Hyperalgesia/complications , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Kainic Acid/administration & dosage , Lidocaine/administration & dosage , Mice, Inbred C57BL , Microinjections , Nerve Fibers, Unmyelinated/pathology , Sensation , Spinal Cord/pathology , Spinal Cord/physiopathology , TRPV Cation Channels/metabolism , Thalamus/pathology , Thalamus/physiopathology , Ventral Thalamic Nuclei/pathology , Ventral Thalamic Nuclei/physiopathology
5.
Mol Neurobiol ; 53(3): 1518-1532, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25650120

ABSTRACT

Abnormal hippocampal neurogenesis is a prominent feature of temporal lobe epilepsy (TLE) models, which is thought to contribute to abnormal brain activity. Stromal cell-derived factor-1 (SDF-1) and its specific receptor CXCR4 play important roles in adult neurogenesis. We investigated whether treatment with the CXCR4 antagonist AMD3100 suppressed aberrant hippocampal neurogenesis, as well as the long-term consequences in the intracerebroventricular kainic acid (ICVKA) model of epilepsy. Adult male rats were randomly assigned as control rats, rats subjected to status epilepticus (SE), and post-SE rats treated with AMD3100. Animals in each group were divided into two subgroups (acute stage and chronic stage). We used immunofluorescence staining of BrdU and DCX to analyze the hippocampal neurogenesis on post-SE days 10 or 74. Nissl staining and Timm staining were used to evaluate hippocampal damage and mossy fiber sprouting, respectively. On post-SE day 72, the frequency and mean duration of spontaneous seizures were measured by electroencephalography (EEG). Cognitive function was evaluated by Morris water maze testing on post-SE day 68. The ICVKA model of TLE resulted in aberrant neurogenesis such as altered proliferation, abnormal dendrite development of newborn neurons, as well as spontaneous seizures and spatial learning impairments. More importantly, AMD3100 treatment reversed the aberrant neurogenesis seen after TLE, which was accompanied by decreased long-term seizure activity, though improvement in spatial learning was not seen. AMD3100 could suppress long-term seizure activity and alter adult neurogenesis in the ICVKA model of TLE, which provided morphological evidences that AMD3100 might be beneficial for treating chronic epilepsy.


Subject(s)
Epilepsy/drug therapy , Heterocyclic Compounds/therapeutic use , Neurogenesis/drug effects , Receptors, CXCR4/antagonists & inhibitors , Animals , Animals, Newborn , Benzylamines , Cyclams , Dendrites/drug effects , Dendrites/ultrastructure , Doublecortin Protein , Drug Evaluation, Preclinical , Electroencephalography , Epilepsy/chemically induced , Epilepsy/pathology , Heterocyclic Compounds/pharmacology , Hippocampus/drug effects , Hippocampus/pathology , Infusions, Intraventricular , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Male , Maze Learning/drug effects , Mice , Mossy Fibers, Hippocampal/drug effects , Mossy Fibers, Hippocampal/pathology , Random Allocation , Rats, Wistar
6.
Neurosci Res ; 99: 62-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26003742

ABSTRACT

Arginine vasopressin (AVP) synthesis in the hypothalamo-neurohypophysial system (HNS) is up-regulated by kainic acid (KA)-induced seizure in rats. However, it remains unknown whether a subconvulsive dose of KA affects the HNS. Here we examined the effects of subcutaneous (s.c.) administration of a low dose of KA (4 mg/kg) on the gene expressions of the AVP, oxytocin (OXT) and neuronal nitric oxide synthase (nNOS) in the supraoptic (SON) and paraventricular nuclei (PVN) of the rat hypothalamus, using in situ hybridization histochemistry. The expression of the AVP gene in the SON and PVN was judged to be up-regulated in KA-treated rats in comparison with saline-treated rats as controls. Next, the expression of the OXT gene was significantly increased in the SON at 6-24h and in the PVN at 6 and 12h after s.c. administration of KA. Finally, the expression of the nNOS gene was significantly increased in the SON and PVN at 3 and 6h after s.c. administration of KA. These results suggest that up-regulation of the gene expressions of the AVP, OXT and nNOS in the rat hypothalamus may be differentially affected by peripheral administration of a subconvulsive dose of KA.


Subject(s)
Arginine Vasopressin/drug effects , Hypothalamus/drug effects , Kainic Acid/pharmacology , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Gene Expression/physiology , In Situ Hybridization/methods , Kainic Acid/administration & dosage , Male , Rats, Wistar , Up-Regulation
7.
Nutr Hosp ; 30(2): 338-42, 2014 Aug 01.
Article in Spanish | MEDLINE | ID: mdl-25208788

ABSTRACT

OBJECTIVE: To determine the effect of a polyphenolic extract from olive pit on the development of the nervous system as well as its effect on pain induced by the neurotoxin kainic acid, taking the zebrafish as the animal model. MATERIAL AND METHODS: We analyse the effect of the extract at the maximum tolerated dose (100 mg/ml of polyphenols) on the cholinergic activity in zebrafish larvae (72 hours post-fertilization). Only fecundated eggs with no abnormalities are used. 6 eggs/bowl are incubated in a 24 bowls microplate in 2 ml of water with DMSO (0.1%) at 26 ± 1º C: a) neurodevelopment: water (control) and 100 mg/ml of extract, as an essay; b) neuroprotection: water and kainic acid (100 µM) (control) and 100 mg/ml of extract (essay). All incubations are in triplicate. After 72 h, incubations are examined and checked for any abnormalities. Larvae are homogenized and acetyl cholinesterase activity and protein concentration in supernatants is quantified. RESULTS: The quantity of protein and the morphologic appreciation is similar in all the essays, showing a standard development. Acetyl cholinesterase in fish larvae, with the polyphenolic extract is 162.2% (SD 44.2) compared to controls (100% of activity) (p < 0.01). Fish larvae treated with kainic acid and polyphenolic acid show 140.1% (SD 22.0) of activity, compared to those only incubated with the neurotoxin (100%) (p < 0.05). CONCLUSION: Polyphenols extracted from olive pit produce an increase in the cholinergic activity during the larvae neurodevelopment in the zebrafish as well as protection against the neurotoxin kainic acid.


Objetivo: Determinar el efecto de un extracto polifenólico de hueso de oliva en el desarrollo del sistema nervioso y frente al daño inducido mediante la neurotoxina ácido kaínico, utilizando como modelo animal el pez cebra. Material y métodos: Se analiza el efecto del extracto a la máxima dosis tolerada (100 mg/ml de polifenoles) sobre la actividad colinérgica en larvas de pez cebra (72 horas post-fertilización). Se utilizan únicamente huevos fecundados sin anomalías. Se incuban 6 huevos/pocillo en microplaca de 24 pocillos en 2 ml de agua con DMSO (0,1%) a 26 ± 1º C: a) neurodesarrollo: agua (control) y con 100 mg/ml de extracto, como ensayo; b) neuroprotección: agua y ácido kaínico (100 M) (control) y con 100 mg/ml de extracto (ensayo). Todas las incubaciones por triplicado. A las 72 h se examinan y verifica ausencia de anomalías. Las larvas se homogeneizan y en los sobrenadantes se cuantifica actividad acetilnolinesterasa y concentración proteínas. Resultados: La cantidad de proteína y apreciación morfológica es análoga en todos los ensayos, indicando mismo desarrollo. La acetilcolinesterasa en las larvas de pez, con el extracto polifenólico es del 162,2%(SD 44,2) respecto a controles (100% de actividad) (p < 0,01). Las larvas de pez tratadas con ácido kaínico y extracto polifenólico presentan el 140,1% (SD 22,0) de actividad, respecto a las incubadas únicamente con la neurotoxina (100%) (p < 0,05). Conclusión: Los polifenoles extraídos de los huesos de aceituna producen incremento de actividad colinérgica durante el neurodesarrollo larvario en el pez cebra y protección frente a la neurotoxina ácido kaínico.


Subject(s)
Nervous System/drug effects , Nervous System/growth & development , Neuroprotective Agents/pharmacology , Olea , Plant Extracts/pharmacology , Seeds , Zebrafish/growth & development , Animals , Kainic Acid/administration & dosage , Pain/chemically induced , Pain/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Polyphenols
8.
J Appl Physiol (1985) ; 106(2): 620-30, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19074569

ABSTRACT

We have shown that electroacupuncture (EA) at P 5-6 (overlying median nerves) activates arcuate (ARC) neurons, which excite the ventrolateral periaqueductal gray (vlPAG) and inhibit cardiovascular sympathoexcitatory neurons in the rostral ventrolateral medulla (rVLM). To investigate whether the ARC inhibits rVLM activity directly or indirectly, we stimulated the splanchnic nerve to activate rVLM neurons. Micropipettes were inserted in the rVLM, vlPAG, and ARC for neural recording or injection. Microinjection of kainic acid (KA; 1 mM, 50 nl) in the ARC blocked EA inhibition of the splanchnic nerve stimulation-induced reflex increases in rVLM neuronal activity. Microinjection of d,l-homocysteic acid (4 nM, 50 nl) in the ARC, like EA, inhibited reflex increases in the rVLM neuronal discharge. The vlPAG neurons receive convergent input from the ARC, splanchnic nerve, P 5-6, and other acupoints. Microinjection of KA bilaterally into the rostral vlPAG partially reversed rVLM neuronal responses and cardiovascular inhibition during d,l-homocysteic acid stimulation of the ARC. On the other hand, injection of KA into the caudal vlPAG completely reversed these responses. We also observed that ARC neurons could be antidromically activated by stimulating the rVLM, and that ARC perikarya was labeled with retrograde tracer that had been microinjected into the rVLM. These neurons frequently contained beta-endorphin and c-Fos, activated by EA stimulation. Therefore, the vlPAG, particularly, the caudal vlPAG, is required for ARC inhibition of rVLM neuronal activation and subsequent EA-related cardiovascular activation. Direct projections from the ARC to the rVLM, which serve as an important source of beta-endorphin, appear also to exist.


Subject(s)
Blood Pressure , Brain/physiology , Cardiovascular System/innervation , Electroacupuncture , Neural Inhibition , Reflex , Sympathetic Nervous System/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Blood Pressure/drug effects , Brain/cytology , Brain/drug effects , Brain/metabolism , Cardiovascular System/drug effects , Cats , Excitatory Amino Acid Agonists/administration & dosage , Female , Homocysteine/administration & dosage , Homocysteine/analogs & derivatives , Kainic Acid/administration & dosage , Male , Medulla Oblongata/physiology , Microinjections , Neural Inhibition/drug effects , Neural Pathways/physiology , Periaqueductal Gray/physiology , Proto-Oncogene Proteins c-fos/metabolism , Reflex/drug effects , Splanchnic Nerves/physiology , Sympathetic Nervous System/drug effects , Time Factors , beta-Endorphin/metabolism
9.
Gynecol Endocrinol ; 22(2): 63-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16603429

ABSTRACT

Soy extracts are widely used as an alternative to hormone replacement therapy for the treatment of menopausal symptoms. Soy phytoestrogens, such as genistein, may act on the nervous system, affecting mood, cognitive function and behavior. In addition, several studies suggest that soy phytoestrogens are neuroprotective. The hypothesis of the present study was that soy extracts may exert neuroprotection and that this effect is mediated by phytoestrogens such as genistein. To test this hypothesis we assessed whether an acute administration of soy extract or genistein in vivo affects hippocampal neuronal loss induced by the systemic administration of kainic acid to adult Wistar female rats. One week after ovariectomy, animals received one intraperitoneal injection of soy extract (0.2, 1, 2 or 20 mg/kg), one injection of genistein (0.1, 1 or 10 mg/kg) or one injection of vehicle. Thirty minutes later, all animals received one intraperitoneal injection of kainic acid (7 mg/kg) or vehicle. One week after the injections, all animals were fixed by perfusion and the number of Nissl-stained neurons in the hilus of the dentate gyrus was estimated by the optical disector method. Administration of soy extract, even at high doses, did not induce neuronal loss and did not increase neuronal degeneration after kainic acid injury. On the contrary, soy extract at doses ranging from 1 to 20 mg/kg prevented neuronal loss induced by kainic acid. Genistein showed neuroprotective effects only at high dose (10 mg/kg), suggesting that other components in the soy extract are involved in the neuroprotective effect.


Subject(s)
Brain/drug effects , Glycine max/chemistry , Neuroprotective Agents/administration & dosage , Phytoestrogens/administration & dosage , Animals , Cell Count , Female , Genistein/administration & dosage , Hippocampus/cytology , Hippocampus/drug effects , Kainic Acid/administration & dosage , Neurons/drug effects , Ovariectomy , Plant Extracts/administration & dosage , Rats , Rats, Wistar
10.
Am J Physiol Heart Circ Physiol ; 290(6): H2543-53, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16428348

ABSTRACT

The periaqueductal gray (PAG) is an important integrative region in the regulation of autonomic outflow and cardiovascular function and may serve as a regulatory center as part of a long-loop pathway during somatic afferent stimulation with acupuncture. Because the ventrolateral PAG (vlPAG) provides input to the rostral ventrolateral medulla (rVLM), an important area for electroacupuncture (EA) regulation of sympathetic outflow, we hypothesized that the vlPAG plays a role in the EA-related modulation of rVLM premotor sympathetic neurons activated during visceral afferent stimulation and autonomic excitatory reflexes. Cats were anesthetized and ventilated, and heart rate and mean blood pressure were monitored. Stimulation of the splanchnic nerve by a pledget of filter paper soaked in bradykinin (BK, 10 mug/ml) every 10 min on the gallbladder induced consistent cardiovascular reflex responses. Bilateral stimulation with EA at acupoints over the pericardial meridian (P5-6) situated over the median nerve reduced the increases in blood pressure from 34 +/- 3 to 18 +/- 5 mmHg for a period of time that lasted for 60 min or more. Unilateral inactivation of neuronal activity in the vlPAG with 50-75 nl of kainic acid (KA, 1 mM) restored the blood pressure responses from 18 +/- 3 to 36 +/- 5 mmHg during BK-induced gallbladder stimulation, an effect that lasted for 30 min. In the absence of EA, unilateral microinjection of the excitatory amino acid dl-homocysteic acid (DLH, 4 nM) in the vlPAG mimicked the effect of EA and reduced the reflex blood pressure responses from 35 +/- 6 to 14 +/- 5 mmHg. Responses of 21 cardiovascular sympathoexcitatory rVLM neurons, including 12 that were identified as premotor neurons, paralleled the cardiovascular responses. Thus splanchnic nerve-evoked neuronal discharge of 32 +/- 4 spikes/30 stimuli in six neurons was reduced to 10 +/- 2 spikes/30 stimuli by EA, which was restored rapidly to 28 +/- 4 spikes/30 stimuli by unilateral injection of 50 nl KA into the vlPAG. Conversely, 50 nl of DLH in the vlPAG reduced the number of action potentials of 5 rVLM neurons from 30 +/- 4 to 18 +/- 4 spikes/30 stimuli. We conclude that the inhibitory influence of EA involves vlPAG stimulation, which, in turn, inhibits rVLM neurons in the EA-related attenuation of the cardiovascular excitatory response during visceral afferent stimulation.


Subject(s)
Cardiovascular Physiological Phenomena , Electroacupuncture , Medulla Oblongata/physiology , Mesencephalon/physiology , Periaqueductal Gray/physiology , Sympathetic Nervous System/physiology , Animals , Blood Pressure/physiology , Cats , Electric Stimulation , Electrophysiology , Excitatory Amino Acid Agonists/pharmacology , Female , Gallbladder/physiology , Homocysteine/administration & dosage , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Kainic Acid/administration & dosage , Kainic Acid/pharmacology , Male , Microinjections , Neural Conduction/drug effects , Neural Conduction/physiology , Reflex/physiology , Splanchnic Nerves/physiology
11.
Toxicol Appl Pharmacol ; 200(1): 48-53, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15451307

ABSTRACT

To investigate the role of tissue plasminogen activator (tPA) in retinal damage, tPA-deficient and wild-type mice were employed. Two different retinal neuron insult models were used in the present study. One is an excitotoxin-treated retinal model, created by direct intravitreal injection of glutamate analogs, NMDA or kainic acid (KA), and the other is an ischemia-reperfusion model induced by transient elevation of intraocular pressure. TdT-dUTP terminal nick-end labeling (TUNEL) method was used to examine the retinal cell nuclear damage. The number of TUNEL-positive cells in ganglion cell layer (GCL) and inner nuclear layer (INL) in tPA-deficient mice after low-, but not high-dose NMDA was significantly less compared to wild type. In contrast, neither intravitreal KA or transient ischemia produced significant difference in retinal damage in tPA vs. wild-type mice. These data show that tPA-deficient mice are resistant to retinal damage by intravitreal injection of NMDA, and indicate that tPA plays a role in the retinal cell damage induced by excitotoxins, especially NMDA.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , N-Methylaspartate/toxicity , Plasminogen Inactivators/physiology , Retinal Diseases/chemically induced , Animals , Cell Count , DNA Fragmentation , Excitatory Amino Acid Agonists/administration & dosage , Glutamates/administration & dosage , Glutamates/pharmacology , In Situ Nick-End Labeling , Injections , Kainic Acid/administration & dosage , Kainic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate/administration & dosage , Neurons/drug effects , Plasminogen Inactivators/deficiency , Recombinant Proteins/pharmacology , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Retina , Retinal Diseases/pathology , Retinal Ganglion Cells/drug effects
12.
Br J Pharmacol ; 142(4): 679-88, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15159282

ABSTRACT

1. Ingestion of a poisonous mushroom Clitocybe acromelalga is known to cause severe tactile pain (allodynia) in the extremities for a month and acromelic acid (ACRO), a kainate analogue isolated from the mushroom, produces selective damage of interneurons of the rat lower spinal cord when injected either systemically or intrathecally. Since ACRO has two isomers, ACRO-A and ACRO-B, here we examined their acute and late effects on induction of allodynia. 2. Intrathecal administration of ACRO-A and ACRO-B provoked marked allodynia by the first stimulus 5 min after injection, which lasted over the 50-min experimental period. Dose-dependency of the acute effect of ACRO-A on induction of allodynia showed a bell-shaped pattern from 50 ag x kg(-1) to 0.5 pg x kg(-1) and the maximum effect was observed at 50 fg x kg(-1). On the other hand, ACRO-B induced allodynia in a dose-dependent manner from 50 pg x kg(-1) to 50 ng x kg(-1). 3. N-methyl-d-aspartate (NMDA) receptor antagonists and Joro spider toxin, a Ca(2+)-permeable AMPA receptor antagonist, inhibited the allodynia induced by ACRO-A, but not by ACRO-B. However, other AMPA/kainate antagonists did not affect the allodynia induced by ACRO. 4. Whereas no neuronal damage was observed in the spinal cord in ACRO-A-treated mice, induction of allodynia by ACRO-A (50 fg x kg(-1)) and ACRO-B (50 ng x kg(-1)) was selectively lost 1 week after i.t. injection of a sublethal dose of ACRO-A (50 ng x kg(-1)) or ACRO-B (250 ng x kg(-1)). Higher doses of ACRO-A, however, could evoke allodynia dose-dependently from 50 pg x kg(-1) to 500 ng x kg(-1) in the ACRO-A-treated mice. The allodynia induced by ACRO-A (500 ng x kg(-1)) was not inhibited by Joro spider toxin or NMDA receptor antagonists. These properties of the late allodynia induced by ACRO-A were quite similar to those of the acute allodynia induced by ACRO-B. 5. ACRO-A could increase [Ca(2+)](i) in the deeper laminae, rather than in the superficial laminae, of the spinal cord. This increase was not blocked by the AMPA-preferring antagonist GYKI52466 and Joro spider toxin. 6. Taken together, these results demonstrate the stereospecificity of ACRO for the induction of allodynia and suggest the presence of a receptor specific to ACRO.


Subject(s)
Heterocyclic Compounds/adverse effects , Kainic Acid/analogs & derivatives , Kainic Acid/adverse effects , Pain/chemically induced , Structure-Activity Relationship , Animals , Basidiomycota/chemistry , Basidiomycota/isolation & purification , Benzodiazepines/pharmacology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Glutamates/pharmacology , Heterocyclic Compounds/administration & dosage , Indoles/pharmacology , Injections, Spinal , Japan , Kainic Acid/administration & dosage , Kainic Acid/antagonists & inhibitors , Kainic Acid/chemistry , Lumbosacral Region/injuries , Lumbosacral Region/pathology , Male , Mice , Mice, Inbred Strains , Mushroom Poisoning/complications , Oximes/pharmacology , Pain/complications , Pain/prevention & control , Quinoxalines/pharmacology , Receptors, AMPA/administration & dosage , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Spider Venoms/pharmacology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/ultrastructure , Stereoisomerism , Time Factors , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
13.
Brain Res ; 979(1-2): 153-64, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12850582

ABSTRACT

In four squirrel monkeys (Saimiri sciureus), the inferior colliculus, together with the neighboring superior colliculus, reticular formation, cuneiform nucleus and parabrachial area, were explored with microelectrodes, looking for neurons that might be involved in the discrimination between self-produced and external sounds. Vocalization was elicited by kainic acid injections into the periaqueductal gray of the midbrain. Acoustic tests were carried out with ascending and descending narrow-band noise sweeps spanning virtually the whole hearing range of the squirrel monkey. Altogether 577 neurons were analyzed. Neurons that both were audiosensitive and fired in advance of self-produced vocalization were found almost exclusively in the pericentral nuclei of the inferior colliculus and the adjacent reticular formation. Only the latter, however, contained, in addition, neurons that fired during external acoustic stimulation, but remained quiet during self-produced vocalization. These findings suggest that the reticular formation bordering the inferior colliculus is involved in the discrimination between self-produced and foreign vocalization on the basis of a vocalmotor feedforward mechanism.


Subject(s)
Brain Mapping , Neurons/physiology , Vocalization, Animal/physiology , Acoustic Stimulation , Animals , Brain/physiology , Electric Stimulation , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Agonists/pharmacology , Female , Inferior Colliculi/physiology , Injections, Intraventricular , Kainic Acid/administration & dosage , Kainic Acid/pharmacology , Male , Microelectrodes , Periaqueductal Gray/physiology , Saimiri
14.
Pol J Pharmacol ; 55(5): 727-33, 2003.
Article in English | MEDLINE | ID: mdl-14704468

ABSTRACT

Loreclezole (5 mg/kg) exerted a significant protective action in amygdala-kindled rats, reducing both seizure and afterdischarge durations. The combinations of loreclezole (2.5 mg/kg) with valproate, clonazepam, or carbamazepine (applied at their subprotective doses) also exhibited antiseizure effect in this test. However, only two first combinations occurred to be of pharmacodynamic nature. Among several chemoconvulsants, bicuculline, N-methyl-D-aspartic acid and BAY k-8644 (the opener of L-type calcium channels) reversed the protective activity of loreclezole alone and its combination with valproate. On the other hand, bicuculline, aminophylline and BAY k-8644 inhibited the anticonvulsive action of loreclezole combined with clonazepam. The results support the hypothesis that the protective activity of loreclezole and its combinations with other antiepileptics may involve potentiation of GABAergic neurotransmission and blockade of L-type of calcium channels.


Subject(s)
Amygdala/drug effects , Convulsants/adverse effects , Drug Combinations , Kindling, Neurologic/drug effects , Triazoles/antagonists & inhibitors , Triazoles/therapeutic use , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Aminophylline/administration & dosage , Aminophylline/adverse effects , Aminophylline/pharmacokinetics , Amygdala/physiopathology , Amygdala/surgery , Animals , Bicuculline/administration & dosage , Bicuculline/adverse effects , Bicuculline/pharmacokinetics , Carbamazepine/pharmacology , Clonazepam/antagonists & inhibitors , Clonazepam/pharmacology , Convulsants/administration & dosage , Convulsants/antagonists & inhibitors , Disease Models, Animal , Drug Interactions , Electrodes, Implanted , Injections, Intraperitoneal , Kainic Acid/administration & dosage , Kainic Acid/pharmacokinetics , Male , N-Methylaspartate/administration & dosage , N-Methylaspartate/adverse effects , N-Methylaspartate/pharmacokinetics , Rats , Rats, Wistar , Seizures/chemically induced , Seizures/physiopathology , Seizures/prevention & control , Stereotaxic Techniques , Strychnine/administration & dosage , Strychnine/adverse effects , Strychnine/pharmacokinetics , Time Factors , Triazoles/administration & dosage , Valproic Acid/administration & dosage , Valproic Acid/antagonists & inhibitors , Valproic Acid/pharmacokinetics
15.
Brain Res ; 880(1-2): 1-10, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-11032984

ABSTRACT

Previous work showed that the dorsal periaqueductal gray is involved in the inhibition of fear-potentiated startle. The present study investigated the effects of blockade and stimulation of Kainate/AMPA and GABA(A) receptors within the dorsal periaqueductal gray on expression and conditioned inhibition of fear-potentiated startle. Blockade of the Kainate/AMPA receptors enhanced whereas stimulation of the Kainate/AMPA receptors decreased expression of fear-potentiated startle. These effects do not reflect conditioned inhibition since this modulation was not changed by injections of Kainate/AMPA receptor agonists or antagonists into the dorsal periaqueductal gray. Stimulation and blockade of GABA(A) receptors within the dorsal periaqueductal gray neither affected expression of fear-potentiated startle nor its conditioned inhibition. The present results together with findings from the literature indicate that glutamate in the dorsal periaqueductal gray is a critical substrate for the expression and modulation of fear-related behaviours.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Fear/physiology , Periaqueductal Gray/physiology , Receptors, AMPA/physiology , Receptors, GABA-A/physiology , Receptors, Kainic Acid/physiology , Reflex, Startle/physiology , Acoustic Stimulation , Animals , Conditioning, Classical , Electroshock , Excitatory Amino Acid Agonists/administration & dosage , Excitatory Amino Acid Antagonists/administration & dosage , Kainic Acid/administration & dosage , Kainic Acid/pharmacology , Light , Male , Microinjections , Periaqueductal Gray/drug effects , Picrotoxin/pharmacology , Piperidines/administration & dosage , Piperidines/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley
16.
J Neurochem ; 73(4): 1573-83, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10501203

ABSTRACT

We investigated the temporal and spatial profile of mRNA transcription for the growth arrest and DNA damage-inducible gene GADD45, DNA fragmentation, and neuronal death in rat brain following focally evoked limbic seizures. GADD45 mRNA was detected by in situ hybridization, whereas fragmented DNA was detected using in situ nick end-labeling by the large (Klenow) fragment of DNA polymerase I. Kainic acid (0.1 microg) was injected into the right amygdala of rats to induce seizures for 45 min, after which diazepam (30 mg/kg) was administered. GADD45 mRNA, DNA fragmentation, and cell death were quantified bilaterally within six limbic brain regions 0-96 h following seizure cessation. All animals underwent seizures of equivalent severity and duration as determined electrographically. In situ hybridization detected bilateral up-regulation of GADD45 mRNA throughout the CA1, CA3, and dentate gyrus of the hippocampus, the piriform and retrosplenial cortices, and the thalamus within 1 h of seizure termination. GADD45 mRNA levels remained elevated for up to 6 h, declining to baseline within all structures by 16 h. Klenow-positive cells were only found within the CA3 pyramidal layer of the ipsilateral hippocampus and appeared 16-72 h following seizure cessation. Morphologic cell death was also restricted to the CA3 subfield. These data demonstrate that focally evoked limbic seizures trigger early bihemispheric GADD45 mRNA transcription within connected limbic structures, whereas subsequent DNA fragmentation and cell death are restricted to selectively vulnerable brain regions.


Subject(s)
Brain/metabolism , DNA Damage , Epilepsies, Partial/metabolism , Neurons/metabolism , Proteins/genetics , Seizures/metabolism , Amygdala/drug effects , Amygdala/pathology , Amygdala/physiology , Animals , Brain/drug effects , Brain/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , DNA Fragmentation , Electroencephalography/drug effects , Epilepsies, Partial/chemically induced , Epilepsies, Partial/pathology , Functional Laterality , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Intracellular Signaling Peptides and Proteins , Kainic Acid/administration & dosage , Kainic Acid/toxicity , Limbic System , Male , Microinjections , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/pathology , Thalamus/drug effects , Thalamus/metabolism , Thalamus/pathology , GADD45 Proteins
17.
Neuroscience ; 82(3): 701-8, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9483529

ABSTRACT

Previous studies have suggested that intracerebroventricular kainic acid injections alter brain anatomy and neurochemistry in a manner similar to what is observed in schizophrenic patients. Disturbances in sensory information processing are one of the major symptoms of schizophrenia. Thus, the present experiments were designed to evaluate the hypothesis that hippocampal damage, induced by administration of kainic acid, would alter the processing of auditory stimuli in a paired-click paradigm. Adult male Sprague-Dawley rats were implanted for surface recording of auditory evoked potentials. At the time of electrode implantation, the rats also received bilateral injections of either kainic acid or the vehicle solution. In vehicle-treated rats, the midlatency N40 component of the auditory evoked potential was diminished in amplitude by approximately 60% in response to the second of a pair of clicks delivered 0.5 s apart. By contrast, no reduction of the N40 wave evoked by the second click was observed in kainate-treated rats. Further, administration of haloperidol, a prototypical neuroleptic agent, did not improve this auditory processing dysfunction in kainate-treated animals. Loss of auditory filtering in the paired-click paradigm and a lack of response to haloperidol in this test are typically observed in schizophrenic humans. Thus, the present results demonstrate that kainate-lesioned rats possess a functional schizophrenia-like abnormality, further reinforcing the utility of this model system for studying the basic neurobiology of schizophrenia-induced sensory processing deficits.


Subject(s)
Auditory Perception/physiology , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Schizophrenia/chemically induced , Schizophrenic Psychology , Acoustic Stimulation , Animals , Antipsychotic Agents/pharmacology , Disease Models, Animal , Electrodes, Implanted , Electrophysiology , Evoked Potentials/drug effects , Excitatory Amino Acid Agonists/administration & dosage , Haloperidol/pharmacology , Hippocampus/anatomy & histology , Hippocampus/drug effects , Hippocampus/physiology , Injections, Intraventricular , Kainic Acid/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Reflex, Startle/physiology
18.
Neurochem Int ; 31(5): 723-30, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9364458

ABSTRACT

The present study was conducted to investigate the sensitivity of the cholinergic elements of ventral and dorsal striatal regions of the rat brain to the neurotoxin kainic acid (KA). Cholinergic activity was assessed by determining choline-acetyltransferase activity (CAT) and by measurements of acetylcholine (Ach) release from slices prelabeled with [3H]-choline. Direct stereotaxic injections of high-dose KA (4 micrograms/2 microliters) into specific brain regions, reduced CAT in caudate putamen (CP) by 91 +/- 1%, in nucleus accumbens (Nac) by 71 +/- 6%, but CAT in the olfactory tubercle (OT) was not affected by KA. The effects of KA on CP CAT were dose- and volume-dependent. In the OT, KA failed to affect CAT at low, moderate or high doses. Slices obtained from CP injected with KA (3 days prior) showed a 90% reduction in the electrically evoked release of [3H]-transmitter release; however, KA had no effect on transmitter release from OT. These results indicate that KA spares the cholinergic elements of the OT, and reveal the existence of marked differences in excitotoxic action of KA between ventral and dorsal striatal regions and among regions of the ventral striatum. Kainic acid preferentially damages neuronal cell bodies, dendrites and terminals intrinsic within the structures injected, with little or no effect on afferent axons and terminal boutons. Therefore, we propose that most of the Ach present in the OT may be within afferent axons and axon terminals. In the CP and NAc, KA lesions reflect loss of intrinsic cholinergic neurons. In addition, variable levels of excitatory inputs and of excitatory receptors, of the mechanisms available to reduce elevated intracellular calcium concentrations and of the levels of free-radical scavenging resources, also could account for the differences in KA neurotoxicity between OT and CP.


Subject(s)
Cholinergic Fibers/drug effects , Corpus Striatum/drug effects , Kainic Acid/pharmacology , Acetylcholine/metabolism , Animals , Choline O-Acetyltransferase/drug effects , Choline O-Acetyltransferase/metabolism , Corpus Striatum/cytology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Injections, Intraventricular , Kainic Acid/administration & dosage , Olfactory Pathways/drug effects , Olfactory Pathways/enzymology , Olfactory Pathways/metabolism , Putamen/drug effects , Putamen/enzymology , Putamen/metabolism , Rats , Rats, Sprague-Dawley , Stereotaxic Techniques
19.
Magnes Res ; 9(4): 281-91, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9247876

ABSTRACT

We developed three models of reversible magnesium depletion in rats resulting from the combined effects of kainic (KA) acid with magnesium deficiency, in order to compare the effects of various common magnesium salts (pidolate, aspartate, lactate, gluconate and chloride) and of magnesium acetyl taurinate (MgATa), administered daily (14 mg Mg2+, PO) for ten days. First, the immediate effects (wet dog shakes, clonic convulsions and death 1 h after injection) and late effects (fall from hole board between the second and tenth days post injection) of kainic acid at three different doses (3.6 and 11 mg/kg) were studied in magnesium deficient rats (50 ppm for 40 days) and in non-deficient rats (1700 ppm). The results showed that the effects of kainic acid were enhanced in magnesium deficient rats. Secondly, after ten days of physiological then pharmacological doses of magnesium, used as chronical supplementation, we showed that kainic acid administration combined with magnesium deficiency led to magnesium depletion of increasing severity depending on the dose of kainic acid. The observed magnesium depletions were weak at a dose of 3 mg/kg KA, moderate at a dose of 6 mg/kg and severe at a dose of 11 mg/kg. These depletions were more or less reversible, and this enabled the classification of the therapeutic effects of these salts on Mg depletion. Among common salts, magnesium pidolate presented the greatest efficacy but none of them fully prevented depletion. In contrast, MgATa was efficient on all the aspects of depletion, when administered preventively both chronically or acutely or as a single curative injection. Consequently the results we obtained in the present study, on a new model of magnesium depletion, showed the greatest efficacy of magnesium acetyl taurinate we demonstrated yet on other models of reversible magnesium depletion.


Subject(s)
Kainic Acid/administration & dosage , Magnesium Deficiency/metabolism , Magnesium Deficiency/prevention & control , Administration, Oral , Animals , Disease Models, Animal , Food, Fortified , Injections, Intraperitoneal , Kainic Acid/adverse effects , Magnesium/administration & dosage , Magnesium/pharmacology , Male , Rats , Rats, Sprague-Dawley , Taurine/analogs & derivatives , Taurine/pharmacology
20.
Hear Res ; 93(1-2): 28-51, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8735067

ABSTRACT

Brainstem regions involved in generating the brainstem auditory evoked potential (BAEP) were identified by examining the effects of lesions on the click-evoked BAEP in cats. An excitotoxin, kainic acid, was injected into various parts of the cochlear nucleus (CN) or into the superior olivary complex (SOC). The locations of the resulting lesions were correlated with the changes produced in the various extrema of the BAEP waveforms. The results indicate that: (1) the earliest BAEP extrema (P1, N1 (recorded between vertex and the earbar ipsilateral to the stimulus) and P1a, P1b, (vertex to contralateral earbar)) are generated by cells with somata peripheral to the CN; (2) P2 is primarily generated by posterior anteroventral CN (AVCNp) and anterior posteroventral CN (PVCNa) cells; (3) SOC, anterior anteroventral CN (AVCNa), AVCNp, and PVCNa cells are involved in generating P3; (4) AVCNa cells are the main CN cells involved in P4, N4, and P5 generation; (5) both ipsilateral and contralateral SOC cells have a role in generating monaurally evoked P4 and P5; and (6) P5 is generated by cells with characteristic frequencies below 10 kHz. From (2) and (4), it is clear that P2 and P4-P5 are generated by cells in distinct, parallel pathways.


Subject(s)
Cochlear Nucleus/drug effects , Evoked Potentials, Auditory, Brain Stem/drug effects , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Olivary Nucleus/drug effects , Acoustic Stimulation , Animals , Brain Stem/drug effects , Brain Stem/pathology , Cats , Cochlear Nucleus/cytology , Cochlear Nucleus/pathology , Evoked Potentials, Auditory, Brain Stem/physiology , Excitatory Amino Acid Agonists/administration & dosage , Kainic Acid/administration & dosage , Neurons/cytology , Neurons/drug effects , Neurons/pathology , Olivary Nucleus/cytology , Olivary Nucleus/pathology , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL