Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Heliyon ; 9(7): e17984, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37539249

RESUMEN

Leucine-rich glioma-inactivated 1 (LGI1) was identified as a causative gene of autosomal dominant lateral temporal lobe epilepsy. We previously reported that Lgi1-mutant rats carrying a missense mutation (L385R) showed audiogenic seizure-susceptibility. To explore the pathophysiological mechanisms underlying Lgi1-related epilepsy, we evaluated changes in glutamate and GABA release in Lgi1-mutant rats. Acoustic priming (AP) for audiogenic seizure-susceptibility was performed by applying intense sound stimulation (130 dB, 10 kHz, 5 min) on postnatal day 16. Extracellular glutamate and GABA levels in the hippocampus CA1 region were evaluated at 8 weeks of age, using in vivo microdialysis techniques. Under naïve conditions without AP, glutamate and GABA release evoked by high-K+ depolarization was more prominent in Lgi1-mutant than in wild-type (WT) rats. The AP treatment on day 16 significantly increased basal glutamate levels and depolarization-induced glutamate release both in Lgi1-mutant and WT rats, yielding greater depolarization-induced glutamate release in Lgi1-mutant rats. On the other hand, the AP treatment enhanced depolarization-induced GABA release only in WT rats, and not in Lgi1-mutant rats, illustrating reduced GABAergic neurotransmission in primed Lgi1-mutant rats. The present results suggest that enhanced glutamatergic and reduced GABAergic neurotransmission are involved in the audiogenic seizure-susceptibility associated with Lgi1-mutation.

2.
Front Pharmacol ; 13: 938175, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35784764

RESUMEN

Nicotine induces kinetic tremor, which resembles pharmacological features of essential tremors, via activating the inferior olive (IO) neurons. Since nicotine is known to enhance dopamine release by stimulating α4ß2 and/or α6 nACh receptors, we examined the effects of various dopamine receptor ligands on nicotine-induced tremor to clarify the role of the dopaminergic system in modulating nicotine tremor. A tremorgenic dose of nicotine increased the dopamine level in the pons and medulla oblongata (P/MO), and the levels of dopamine metabolites in the hippocampus, P/MO, and striatum. Treatment of animals with the D1/5 agonist SKF-38393 inhibited the induction of nicotine tremor, whereas the D3 agonist PD-128,907 facilitated nicotine-induced tremor. The D2 agonist sumanirole showed no effect. In addition, nicotine tremor was significantly enhanced by the D1/5 antagonist SCH-23390 and inhibited by the D3 antagonist U-99194. Neither the D2 (L-741,626) nor D4 (L-745,870) antagonist affected the generation of nicotine tremor. Furthermore, microinjection of U-99194 into the cerebellum significantly inhibited nicotine-induced tremor, whereas its injection into IO or the striatum did not affect tremor generation. Although intrastriatal injection of SCH-23390 showed no effects, its injection into IO tended to enhance nicotine-induced tremor. The present study suggests that dopamine D3 and D1/5 receptors regulate the induction of nicotine tremor in an opposite way, D3 receptors facilitately and D1/5 receptors inhibitorily. In addition, the cerebellar D3 receptors may play an important role in modulating the induction of nicotine tremor mediated by the olivo-cerebellar system.

3.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638578

RESUMEN

Inwardly rectifying Kir4.1 channels in astrocytes mediate spatial potassium (K+) buffering, a clearance mechanism for excessive extracellular K+, in tripartite synapses. In addition to K+ homeostasis, astrocytic Kir4.1 channels also play an essential role in regulating extracellular glutamate levels via coupling with glutamate transporters. Moreover, Kir4.1 channels act as novel modulators of the expression of brain-derived neurotrophic factor (BDNF) in astrocytes. Specifically, inhibition of astrocytic Kir4.1 channels elevates extracellular K+ and glutamate levels at synapses and facilitates BDNF expression in astrocytes. These changes elevate neural excitability, which may facilitate synaptic plasticity and connectivity. In this article, we summarize the functions and pharmacological features of Kir4.1 channels in astrocytes and highlight the importance of these channels in the treatment of brain diseases. Although further validation in animal models and human patients is required, astrocytic Kir4.1 channel could potentially serve as a novel therapeutic target for the treatment of depressive disorders and epilepsy.


Asunto(s)
Astrocitos/metabolismo , Encefalopatías/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Potasio/metabolismo , Animales , Ácido Glutámico/metabolismo , Humanos , Potenciales de la Membrana/fisiología , Sinapsis/metabolismo
4.
J Pharmacol Sci ; 145(1): 60-68, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33357781

RESUMEN

Emerging evidence suggests that dysfunctions in glutamatergic signaling are associated with the pathophysiology of depression. Several molecules that act on glutamate binding sites, so-called glutamatergic modulators, are rapid-acting antidepressants that stimulate synaptogenesis. Their antidepressant response involves the elevation of both extracellular glutamate and brain-derived neurotrophic factor (BDNF) levels, as well as the postsynaptic activation of the mammalian target of rapamycin complex 1. The mechanisms involved in the antidepressant outcomes of glutamatergic modulators, including ketamine, suggest that astrocytes must be considered a cellular target for developing rapid-acting antidepressants. It is well known that extracellular glutamate levels and glutamate intrasynaptic time-coursing are maintained by perisynaptic astrocytes, where inwardly rectifying potassium channels 4.1 (Kir4.1 channels) regulate both potassium and glutamate uptake. In addition, ketamine reduces membrane expression of Kir4.1 channels, which raises extracellular potassium and glutamate levels, increasing postsynaptic neural activities. Furthermore, inhibition of Kir4.1 channels stimulates BDNF expression in astrocytes, which may enhance synaptic connectivity. In this review, we discuss glutamatergic modulators' actions in regulating extracellular glutamate and BDNF levels, and reinforce the importance of perisynaptic astrocytes for the development of novel antidepressant drugs.


Asunto(s)
Antidepresivos/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Sinapsis , Fármacos actuantes sobre Aminoácidos Excitadores , Humanos , Ketamina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo
5.
Acta Neuropathol Commun ; 8(1): 206, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33256836

RESUMEN

The Ihara epileptic rat (IER) is a mutant model with limbic-like seizures whose pathology and causative gene remain elusive. In this report, via linkage analysis, we identified Down syndrome cell adhesion molecule-like 1(Dscaml1) as the responsible gene for IER. A single base mutation in Dscaml1 causes abnormal splicing, leading to lack of DSCAML1. IERs have enhanced seizure susceptibility and accelerated kindling establishment. Furthermore, GABAergic neurons are severely reduced in the entorhinal cortex (ECx) of these animals. Voltage-sensitive dye imaging that directly presents the excitation status of brain slices revealed abnormally persistent excitability in IER ECx. This suggests that reduced GABAergic neurons may cause weak sustained entorhinal cortex activations, leading to natural kindling via the perforant path that could cause dentate gyrus hypertrophy and epileptogenesis. Furthermore, we identified a single nucleotide substitution in a human epilepsy that would result in one amino acid change in DSCAML1 (A2105T mutation). The mutant DSCAML1A2105T protein is not presented on the cell surface, losing its homophilic cell adhesion ability. We generated knock-in mice (Dscaml1A2105T) carrying the corresponding mutation and observed reduced GABAergic neurons in the ECx as well as spike-and-wave electrocorticogram. We conclude that DSCAML1 is required for GABAergic neuron placement in the ECx and suppression of seizure susceptibility in rodents. Our findings suggest that mutations in DSCAML1 may affect seizure susceptibility in humans.


Asunto(s)
Moléculas de Adhesión Celular/genética , Corteza Entorrinal/patología , Neuronas GABAérgicas/patología , Convulsiones/genética , Animales , Electroencefalografía , Predisposición Genética a la Enfermedad , Excitación Neurológica/genética , Ratones , Ratas , Ratas Mutantes
6.
Exp Anim ; 69(4): 388-394, 2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-32507787

RESUMEN

We recently demonstrated that aspartoacylase (Aspa) and hyperpolarization-activated cyclic nucleotide-gated potassium channel 1 (Hcn1) genes were causative of essential tremor (ET) in rats. This finding was obtained using Aspaem34Kyo/Hcn1A354V double-mutant rats, but they were bred on a heterogeneous genetic background of two strains, F344 and WTC. Here, we developed an Aspaem34Kyo/Hcn1em1Kyo double-knockout rat strain with a homogenous F344 genetic background and studied the ability of glutamate receptor antagonists to suppress ET. The F344-Aspa/Hcn1 double-knockout rats exhibited spontaneous, intense body tremor equivalent to that in the double-mutant rats. N-acetyl-aspartate (NAA), a substrate of ASPA, showed accumulation in all brain regions and in the spinal cord. However, N-acetyl-aspartyl-glutamate (NAAG), which is derived from NAA and interacts with glutamatergic receptors, was decreased in the medulla oblongata of the double-knockout rats. The tremor was suppressed by 3-[(R)-2-carboxypiperazin-4-yl]-prop-2-enyl-1-phosphonic acid, an N-methyl-D-aspartate (NMDA) receptor antagonist, in F344-Aspa/Hcn1 double-knockout rats. The non-NMDA glutamate receptor antagonist NBQX weakly inhibited the tremor, while the metabotropic glutamate receptor antagonist LY341495 showed no effect. In addition, both NR2B subunit-specific (Ro 25-6981) and NR2C/NR2D subunit-specific (cis-piperidine dicarboxylic acid) NMDA receptor antagonists suppressed the tremor. These data indicated that the pathogenesis of tremor in Aspa/Hcn1 double-knockout rats involved ionotropic glutamate receptors, particularly NMDA receptors.


Asunto(s)
Amidohidrolasas/genética , Temblor Esencial/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales de Potasio/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/fisiología , Amidohidrolasas/metabolismo , Animales , Encéfalo/metabolismo , Temblor Esencial/tratamiento farmacológico , Técnicas de Inactivación de Genes , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Terapia Molecular Dirigida , Fenoles/farmacología , Fenoles/uso terapéutico , Piperidinas/farmacología , Piperidinas/uso terapéutico , Canales de Potasio/metabolismo , Quinoxalinas/farmacología , Quinoxalinas/uso terapéutico , Ratas Endogámicas F344 , Ratas Mutantes , Médula Espinal/metabolismo
7.
Front Neurol ; 11: 626658, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424762

RESUMEN

Astrocytes regulate potassium and glutamate homeostasis via inwardly rectifying potassium (Kir) 4.1 channels in synapses, maintaining normal neural excitability. Numerous studies have shown that dysfunction of astrocytic Kir4.1 channels is involved in epileptogenesis in humans and animal models of epilepsy. Specifically, Kir4.1 channel inhibition by KCNJ10 gene mutation or expressional down-regulation increases the extracellular levels of potassium ions and glutamate in synapses and causes hyperexcitation of neurons. Moreover, recent investigations demonstrated that inhibition of Kir4.1 channels facilitates the expression of brain-derived neurotrophic factor (BDNF), an important modulator of epileptogenesis, in astrocytes. In this review, we summarize the current understanding on the role of astrocytic Kir4.1 channels in epileptogenesis, with a focus on functional and expressional changes in Kir4.1 channels and their regulation of BDNF secretion. We also discuss the potential of Kir4.1 channels as a therapeutic target for the prevention of epilepsy.

8.
Front Pharmacol ; 10: 1045, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31607910

RESUMEN

Antipsychotic drugs are often used for the treatment of behavioral and psychological symptoms of dementia (BPSD), especially psychosis and behavioral disturbances (e.g., aggression and agitation). They are prescribed alone or in conjunction with anti-dementia (e.g., anti-Alzheimer's disease drugs) and other psychotropic drugs (e.g., antidepressants). However, antipsychotic drugs frequently produce serious extrapyramidal side effects (EPS) including Parkinsonian symptoms (e.g., bradykinesia, akinesia, tremor, and muscle rigidity). Therefore, appropriate drug choice and combination strategy are important in the treatment of BPSD. Among anti-Alzheimer's disease drugs, cholinesterase inhibitors (ChEIs, e.g., donepezil and galantamine) have a propensity to potentiate EPS associated with antipsychotic treatment in a synergistic manner. In contrast, the NMDA receptor antagonist memantine reduces antipsychotic-induced EPS. Antidepressant drugs, which inhibit 5-HT reuptake into the nerve terminals, also synergistically augment antipsychotic-induced EPS, while mirtazapine (α2, 5-HT2 and 5-HT3 antagonist) reduces the EPS induction. Importantly, previous studies showed that multiple 5-HT receptors play crucial roles in modulating EPS associated with antipsychotic treatment. Specifically, activation of 5-HT1A receptors or blockade of 5-HT2, 5-HT3 and 5-HT6 receptors can alleviate EPS induction both by antipsychotics alone and by combined antipsychotic treatments with ChEIs or 5-HT reuptake inhibitors. In this article, we review antipsychotic use in treating BPSD and discuss the favorable drug selection in terms of the management of antipsychotic-induced EPS.

9.
Behav Brain Res ; 369: 111922, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31039378

RESUMEN

Phf24 is known as Gαi-interacting protein (GINIP) and is associated with the GABAB receptor. To study the function of Phf24 protein in the central nervous system (CNS), we have newly developed Phf24-null rats and investigated their behavioral phenotypes, especially changes in seizure sensitivity, emotional responses and cognitive functions. Phf24-null rats did not exhibit any spontaneous seizures. However, they showed a higher sensitivity to pentylenetetrazol (PTZ)- or pilocarpine-induced convulsive seizures. Phf24-null rats also showed an elevated susceptibility to kindling development with repeated PTZ treatments, suggesting that Phf24 acts as an inhibitory modulator in epileptogenesis. Although young Phf24-null rats showed normal gross behaviors, elevated spontaneous locomotor activity, especially in terms of the circadian dark period, emotional hyper-reactivity, reduced anxiety behaviors in the elevated plus-maze (EPM) test, and cognitive deficits in the Morris water maze test were explicitly observed at older age (20-week-old). The present results suggest that Phf24 is essential for proper functioning of the CNS, especially in preventing epileptogenesis and controlling emotional and cognitive functions.


Asunto(s)
Disfunción Cognitiva/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Convulsiones/etiología , Animales , Sistema Nervioso Central/metabolismo , Cognición/fisiología , Trastornos del Conocimiento/genética , Disfunción Cognitiva/metabolismo , Emociones/fisiología , Técnicas de Inactivación de Genes/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Excitación Neurológica/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Dedos de Zinc PHD/genética , Ratas , Ratas Endogámicas F344 , Receptores de GABA-B/genética , Convulsiones/genética , Convulsiones/metabolismo
10.
Int J Mol Sci ; 20(5)2019 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-30813600

RESUMEN

The dysfunction of astrocytic inwardly rectifying potassium (Kir) 4.1 channels, which mediate the spatial potassium-buffering function of astrocytes, is known to be involved in the development of epilepsy. Here, we analyzed the Kir4.1 expressional changes in Leucine-Rich Glioma-Inactivated 1 (Lgi1) mutant rats, which is a model of autosomal dominant lateral temporal lobe epilepsy in humans, to clarify the role of astrocytic Kir4.1 channels in Lgi1-related epileptogenesis. Priming acoustic stimulation (at postnatal day 16) conferred seizure susceptibility on Lgi1 mutant rats, which evoked audiogenic seizures with test stimulation at eight weeks. In the seizure-susceptible Lgi1 mutant rats (before test stimulation), astrocytic Kir4.1 expression was down-regulated region-specifically in the cerebral cortex, hippocampus, and amygdala. In addition, prophylactic treatments of Lgi1 mutant rats with valproic acid (VPA, 30 mg/kg and 200 mg/kg) for two weeks prevented both the development of seizure susceptibility and the down-regulation of Kir4.1 expression in astrocytes. The present study demonstrated for the first time that the astrocytic Kir4.1 expression was reduced in the Lgi1-related seizure model, suggesting that the down-regulation of Kir4.1 channels in astrocytes is involved in audiogenic epileptogenesis caused by Lgi1 mutation. In addition, VPA seemed to have a prophylactic effect on Lgi1-related seizures.


Asunto(s)
Astrocitos/metabolismo , Regulación hacia Abajo , Epilepsia Refleja/genética , Mutación/genética , Canales de Potasio de Rectificación Interna/genética , Proteínas/genética , Acústica , Animales , Susceptibilidad a Enfermedades , Epilepsia Refleja/tratamiento farmacológico , Proteína Ácida Fibrilar de la Glía/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas/metabolismo , Ratas Endogámicas F344 , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico
11.
Neurotox Res ; 35(3): 575-583, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30729450

RESUMEN

Organophosphates (OPs) inhibit cholinesterase and hyperactivate the acetylcholinergic nervous system in the brain, causing motor disorders (e.g., tremor and seizures). Here, we performed behavioral and immunohistochemical studies in mice and rats to investigate the tremorgenic mechanism of paraoxon, an active metabolite of parathion. Treating animals with paraoxon (0.15-0.6 mg/kg, i.p.) elicited kinetic tremor in a dose-dependent manner. Expressional analysis of Fos protein, a biomarker of neural excitation, revealed that a tremorgenic dose of paraoxon (0.6 mg/kg) significantly and region-specifically elevated Fos expression in the cerebral cortex (e.g., sensory cortex), hippocampal CA1, globus pallidus, medial habenula, and inferior olive (IO) among 48 brain regions examined. A moderate increase in Fos expression was also observed in the dorsolateral striatum while the change was not statistically significant. Paraoxon-induced tremor was inhibited by the nicotinic acetylcholine (nACh) receptor antagonist mecamylamine (MEC), but not affected by the muscarinic acetylcholine receptor antagonist trihexyphenidyl (THP). In addition, paraoxon-induced Fos expression in the IO was also antagonized by MEC, but not by THP, and lesioning of the IO markedly suppressed tremorgenic action of paraoxon. The present results suggest that OPs elicit kinetic tremor at least partly by activating IO neurons via nACh receptors.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Discinesia Inducida por Medicamentos/metabolismo , Paraoxon/efectos adversos , Temblor/inducido químicamente , Temblor/metabolismo , Animales , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Discinesia Inducida por Medicamentos/tratamiento farmacológico , Discinesia Inducida por Medicamentos/patología , Expresión Génica/efectos de los fármacos , Masculino , Mecamilamina/farmacología , Ratones , Antagonistas Muscarínicos/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Antagonistas Nicotínicos/farmacología , Proteínas Oncogénicas v-fos/metabolismo , Ratas , Receptores Nicotínicos/metabolismo , Temblor/tratamiento farmacológico , Temblor/patología , Trihexifenidilo/farmacología
12.
Brain Res ; 1706: 209-217, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30408474

RESUMEN

Hyperpolarized-activated cyclic nucleotide-gated (HCN) channels underlie hyperpolarization-activated current (Ih) and are involved in controlling the excitability and electrical responsiveness of neurons. Absence epilepsy is clinically defined by a sudden, brief impairment of consciousness and behavioral arrest. Spike-and-wave discharges (SWDs) on electroencephalograms (EEG) are a diagnostic hallmark of absence epilepsy. In rat models of absence epilepsy, impaired function or expression of HCN1, a subtype of HCN channels, has been found. Here, to evaluate whether HCN1 deficiency causes absence epilepsy in rats, we developed Hcn1-knockout rats by transcription activator-like effector nuclease mutagenesis. The cortical and hippocampal pyramidal neurons of these rats displayed a significant reduction of Ih, a pronounced hyperpolarizing shift of the resting membrane potential, and increased input resistance, which indicated that the Hcn1-knockout rats were deficient in HCN1 function. The Hcn1-knockout rats were also more vulnerable to pentylenetetrazol-induced acute convulsions. More importantly, they exhibited spontaneous SWDs, which were accompanied by behavioral arrest, both of which were suppressed by ethosuximide. These results confirm the involvement of the HCN1 subunit in the regulation of input resistance and provide direct evidence that a deficiency of HCN1 caused absence epilepsy in rats.


Asunto(s)
Epilepsia Tipo Ausencia/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales de Potasio/metabolismo , Potenciales de Acción/fisiología , Animales , Corteza Cerebral/metabolismo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia Tipo Ausencia/etiología , Técnicas de Inactivación de Genes , Hipocampo/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Masculino , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/genética , Canales de Potasio/fisiología , Células Piramidales/fisiología , Ratas , Ratas Endogámicas F344 , Convulsiones/metabolismo
13.
Nihon Yakurigaku Zasshi ; 152(6): 275-280, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-30531097

RESUMEN

Epilepsy is a chronic neurologic disease characterized by recurrent seizures, affecting nearly 1% of the population. Synaptic vesicle protein 2A (SV2A) is a membrane protein specifically expressed in synaptic vesicles and is now implicated in the pathogenesis of epileptic disorders. This is because 1) Sv2a-knockout mice exhibit severe seizures, 2) SV2A serves as a specific binding site for certain antiepileptics (e.g., levetiracetam and its analogues) and 3) the SV2A expression changes under various epileptic conditions both in animals (e.g., kindling) and humans (e.g., intractable temporal lobe epilepsy and focal cortical dysplasia). Furthermore, it has been shown that a missense mutation in the SV2A gene caused intractable epilepsy, involuntary movements and developmental retardation, indicating a causative role of SV2A dysfunction in epilepsy. In order to explore the mechanism of SV2A in modulating development of epileptogenesis, we recently developed a novel rat model (Sv2aL174Q rat) carrying a missense mutation (Leu174Gln) in the Sv2a gene. These rats were highly susceptible to the kindling development associated with repeated pentylenetetrazole treatments or electrical stimulations of the amygdala. In addition, the Sv2aL174Q mutation specifically impaired depolarization-induced GABA, but not glutamate, release in the hippocampus and amygdala. All this evidence indicates that the SV2A-GABAergic system plays a crucial role in modulating epileptogenesis and encourages discovery research into the novel antiepileptic agents which enhance the function of the SV2A-GABA system.


Asunto(s)
Epilepsia , Excitación Neurológica , Animales , Humanos , Glicoproteínas de Membrana , Ratones , Proteínas del Tejido Nervioso , Ratas , Vesículas Sinápticas
14.
Int J Mol Sci ; 19(11)2018 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-30356026

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a key molecule essential for neural plasticity and development, and is implicated in the pathophysiology of various central nervous system (CNS) disorders. It is now documented that BDNF is synthesized not only in neurons, but also in astrocytes which actively regulate neuronal activities by forming tripartite synapses. Inwardly rectifying potassium (Kir) channel subunit Kir4.1, which is specifically expressed in astrocytes, constructs Kir4.1 and Kir4.1/5.1 channels, and mediates the spatial potassium (K⁺) buffering action of astrocytes. Recent evidence illustrates that Kir4.1 channels play important roles in bringing about the actions of antidepressant drugs and modulating BDNF expression in astrocytes. Although the precise mechanisms remain to be clarified, it seems likely that inhibition (down-regulation or blockade) of astrocytic Kir4.1 channels attenuates K⁺ buffering, increases neuronal excitability by elevating extracellular K⁺ and glutamate, and facilitates BDNF expression. Conversely, activation (up-regulation or opening) of Kir4.1 channels reduces neuronal excitability by lowering extracellular K⁺ and glutamate, and attenuates BDNF expression. Particularly, the former pathophysiological alterations seem to be important in epileptogenesis and pain sensitization, and the latter in the pathogenesis of depressive disorders. In this article, we review the functions of Kir4.1 channels, with a focus on their regulation of spatial K⁺ buffering and BDNF expression in astrocytes, and discuss the role of the astrocytic Kir4.1-BDNF system in modulating CNS disorders.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Trastorno Depresivo/metabolismo , Epilepsia/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Humanos , Neuronas/metabolismo , Neuronas/fisiología , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética
15.
Front Pharmacol ; 9: 845, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30127740

RESUMEN

Inwardly rectifying potassium (Kir) channel subunits Kir4.1 are specifically expressed in astrocytes and regulate neuronal excitability by mediating spatial potassium buffering. In addition, it is now known that astrocytic Kir4.1 channels are closely involved in the pathogenesis of epilepsy. Here, to explore the role of Kir4.1 channels in the treatment of epilepsy, we evaluated the effects of the antiepileptic drugs, valproate, phenytoin, phenobarbital and ethosuximide, on Kir4.1 expression in astrocytes using immunohistochemical techniques. Repeated treatment of rats with valproate (30-300 mg/kg, i.p., for 1-10 days) significantly elevated the Kir4.1 expression levels in the cerebral cortex, amygdala and hippocampus. Up-regulation of Kir4.1 expression by valproate occurred in a dose- and treatment period-related manner, and did not accompany an increase in the number of astrocytes probed by glial fibrillary acidic protein (GFAP). In addition, repeated treatment with phenytoin (30 mg/kg, i.p., for 10 days) or phenobarbital (30 mg/kg, i.p., for 10 days) also elevated Kir4.1 expression region-specifically in the amygdala. However, ethosuximide (100 mg/kg, i.p., for 10 days), which can alleviate absence but not convulsive seizures, showed no effects on the astrocytic Kir4.1 expression. The present results demonstrated for the first time that the antiepileptic drugs effective for convulsive seizures (valproate, phenytoin, and phenobarbital) commonly elevate the astrocytic Kir4.1 channel expression in the limbic regions, which may be related to their antiepileptic actions.

16.
J Pharmacol Sci ; 137(2): 162-169, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29945769

RESUMEN

We previously showed that nicotine evoked kinetic tremor by activating the inferior olive, which is implicated in the pathogenesis of essential tremor, via α7 nicotinic acetylcholine receptors. Here, we evaluated the effects of various anti-tremor and anti-epileptic agents on nicotine-induced tremor in mice to clarify the pharmacological characteristics of nicotine tremor. Drugs effective for essential tremor, propranolol, diazepam and phenobarbital, all significantly inhibited kinetic tremor induced by an intraperitoneal (i.p.) injection of nicotine (1 mg/kg). In contrast, none of the medications for Parkinson's disease, l-DOPA, bromocriptine or trihexyphenidyl, affected the nicotine tremor. Among the anti-epileptic agents examined, valproate, carbamazepine and ethosuximide, significantly inhibited nicotine-induced tremor. In addition, a selective T-type Ca2+ channel blocker, TTA-A2, also suppressed the nicotine tremor. However, neither gabapentin, topiramate, zonisamide nor levetiracetam significantly affected nicotine-induced tremor. The present results show that nicotine-induced tremor resembles essential tremor not only on the neural basis, but also in terms of the pharmacological responses to anti-tremor agents, implying that nicotine-induced tremor can serve as a model for essential tremor. In addition, it is suggested that anti-epileptic agents, which have stimulant actions on the GABAergic system or blocking actions on voltage-gated Na+ channels and T-type Ca2+ channels, can alleviate essential tremor.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Bencenoacetamidas/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Carbamazepina/uso terapéutico , Diazepam/uso terapéutico , Modelos Animales de Enfermedad , Temblor Esencial/inducido químicamente , Temblor Esencial/tratamiento farmacológico , Etosuximida/uso terapéutico , Nicotina/efectos adversos , Fenobarbital/uso terapéutico , Propranolol/uso terapéutico , Piridinas/uso terapéutico , Ácido Valproico/uso terapéutico , Animales , Antiparkinsonianos/uso terapéutico , Masculino , Ratones Endogámicos
18.
Behav Genet ; 47(6): 609-619, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28936718

RESUMEN

The Noda epileptic rat (NER) exhibits generalized tonic-clonic seizures (GTCS). A genetic linkage analysis identified two GTCS-associated loci, Ner1 on Chr 1 and Ner3 on Chr 5. The wild-type Ner1 and Ner3 alleles suppressed GTCS when combined in double-locus congenic lines, but not when present in single-locus congenic lines. Global expression analysis revealed that cholecystokinin B receptor (Cckbr) and suppressor of tumorigenicity 5 (St5), which map within Ner1, and PHD finger protein 24 (Phf24), which maps within Ner3, were significantly downregulated in NER. De novo BAC sequencing detected an insertion of an endogenous retrovirus sequence in intron 2 of the Phf24 gene in the NER genome, and PHF24 protein was almost absent in the NER brain. Phf24 encodes a Gαi-interacting protein involved in GABAB receptor signaling pathway. Based on these findings, we conclude that Cckbr, St5, and Phf24 are strong candidate genes for GTCS in NER.


Asunto(s)
Epilepsia Tónico-Clónica/genética , Receptor de Colecistoquinina B/genética , Proteínas Supresoras de Tumor/genética , Animales , Cromosomas de los Mamíferos/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Electroencefalografía/veterinaria , Epilepsia/genética , Ligamiento Genético/genética , Sitios Genéticos/genética , Dedos de Zinc PHD/genética , Ratas , Ratas Wistar/genética , Receptor de Colecistoquinina B/fisiología , Convulsiones/genética
19.
Brain Res ; 1676: 38-45, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28917524

RESUMEN

Tremor dominant Kyoto (Trdk) is an autosomal dominant mutation that appeared in F344/NSlc rats mutagenized with N-ethyl-N-nitrosourea (ENU). In this study, we characterized and genetically analyzed F344-Trdk/+ heterozygous rats. The rats exhibited a tremor that was especially evident around weaning but persisted throughout life. The tremors of F344-Trdk/+ rats were attenuated by drugs effective against essential tremor (ET) but not drugs used to treat Parkinson's disease-related tremor, indicating that the pharmacological phenotype of F344-Trdk/+ rats was similar to human ET. Using positional candidate approach, we identified the Trdk mutation as a missense substitution (c. 866T>A, p. I289N) in Kcnn2, which encodes the SK2 subunit of the small-conductance Ca2+-activated K+ channel. In vitro electrophysiological studies revealed that the I289N mutation diminished SK2 channel activity. These findings demonstrate that F344-Trdk/+ rats represent a novel model of ET, and strongly suggest that Kcnn2 is the causative gene for the tremor phenotype in F344-Trdk/+ rats.


Asunto(s)
Mutación Missense , Ratas Endogámicas F344 , Ratas Mutantes , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Temblor/genética , Animales , Antidiscinéticos/farmacología , Encéfalo/metabolismo , Encéfalo/patología , Mapeo Cromosómico , Modelos Animales de Enfermedad , Temblor Esencial/tratamiento farmacológico , Temblor Esencial/genética , Temblor Esencial/metabolismo , Temblor Esencial/patología , Células HEK293 , Humanos , Inmunohistoquímica , Hibridación in Situ , Técnicas de Placa-Clamp , Fenotipo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Transfección , Temblor/tratamiento farmacológico , Temblor/metabolismo , Temblor/patología
20.
Int J Mol Sci ; 18(7)2017 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-28671605

RESUMEN

Dysfunction of the N-methyl-d-aspartate (NMDA) receptor has been implicated in the pathogenesis of schizophrenia. Although agonists for the glycine-binding sites of NMDA receptors have potential as new medication for schizophrenia, their modulation of antipsychotic-induced extrapyramidal side effects (EPS) has not yet been clarified. We herein evaluated the effects of glycine-binding site stimulants of NMDA receptors on antipsychotic-induced EPS in mice and rats. d-cycloserine (DCS) and d-serine significantly improved haloperidol (HAL)-induced bradykinesia in mice, whereas glycine showed no effects. Sodium benzoate, a d-amino acid oxidase inhibitor, also attenuated HAL-induced bradykinesia. Improvements in HAL-induced bradykinesia by DCS were antagonized by the NMDA antagonist dizocilpine or nitric oxide synthase inhibitor L-NG-Nitro-l-arginine methyl ester. In addition, DCS significantly reduced HAL-induced Fos expression in the dorsolateral striatum without affecting that in the nucleus accumbens. Furthermore, a microinjection of DCS into the substantia nigra pars compacta significantly inhibited HAL-induced EPS concomitant with elevations in dopamine release in the striatum. The present results demonstrated for the first time that stimulating the glycine-binding sites of NMDA receptors alleviates antipsychotic-induced EPS by activating the nigrostriatal dopaminergic pathway, suggesting that glycine-binding site stimulants are beneficial not only for efficacy, but also for side-effect management.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Glicina/metabolismo , Trastornos Motores/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sustancia Negra/metabolismo , Animales , Sitios de Unión , Cuerpo Estriado/efectos de los fármacos , Cicloserina/farmacología , Maleato de Dizocilpina/farmacología , Haloperidol , Hipocinesia/inducido químicamente , Masculino , Ratones , Microdiálisis , Microinyecciones , NG-Nitroarginina Metil Éster/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/agonistas , Sustancia Negra/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...