Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Int J Mol Sci ; 22(8)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923533

RESUMEN

It has been established that the selective α2A adrenoceptor agonist guanfacine reduces hyperactivity and improves cognitive impairment in patients with attention-deficit/hyperactivity disorder (ADHD). The major mechanisms of guanfacine are considered to involve the activation of the postsynaptic α2A adrenoceptor of glutamatergic pyramidal neurons in the frontal cortex, but the effects of chronic guanfacine administration on catecholaminergic and glutamatergic transmissions associated with the orbitofrontal cortex (OFC) are yet to be clarified. The actions of guanfacine on catecholaminergic transmission, the effects of acutely local and systemically chronic (for 7 days) administrations of guanfacine on catecholamine release in pathways from the locus coeruleus (LC) to OFC, the ventral tegmental area (VTA) and reticular thalamic-nucleus (RTN), from VTA to OFC, from RTN to the mediodorsal thalamic-nucleus (MDTN), and from MDTN to OFC were determined using multi-probe microdialysis with ultra-high performance liquid chromatography. Additionally, the effects of chronic guanfacine administration on the expression of the α2A adrenoceptor in the plasma membrane fraction of OFC, VTA and LC were examined using a capillary immunoblotting system. The acute local administration of therapeutically relevant concentrations of guanfacine into the LC decreased norepinephrine release in the OFC, VTA and RTN without affecting dopamine release in the OFC. Systemically, chronic administration of therapeutically relevant doses of guanfacine for 14 days increased the basal release of norepinephrine in the OFC, VTA, RTN, and dopamine release in the OFC via the downregulation of the α2A adrenoceptor in the LC, OFC and VTA. Furthermore, systemically, chronic guanfacine administration did not affect intrathalamic GABAergic transmission, but it phasically enhanced thalamocortical glutamatergic transmission. The present study demonstrated the dual actions of guanfacine on catecholaminergic transmission-acute attenuation of noradrenergic transmission and chronic enhancement of noradrenergic transmission and thalamocortical glutamatergic transmission. These dual actions of guanfacine probably contribute to the clinical effects of guanfacine against ADHD.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Guanfacina/farmacología , Corteza Prefrontal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Tálamo/efectos de los fármacos , Agonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Dopamina/metabolismo , Ácido Glutámico/metabolismo , Guanfacina/administración & dosificación , Guanfacina/uso terapéutico , Masculino , Norepinefrina/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Tálamo/metabolismo , Tálamo/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
2.
Int J Mol Sci ; 22(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572981

RESUMEN

The functional suppression of serotonin (5-HT) type 7 receptor (5-HT7R) is forming a basis for scientific discussion in psychopharmacology due to its rapid-acting antidepressant-like action. A novel mood-stabilizing atypical antipsychotic agent, lurasidone, exhibits a unique receptor-binding profile, including a high affinity for 5-HT7R antagonism. A member of a novel class of antidepressants, vortioxetine, which is a serotonin partial agonist reuptake inhibitor (SPARI), also exhibits a higher affinity for serotonin transporter, serotonin receptors type 1A (5-HT1AR) and type 3 (5-HT3R), and 5-HT7R. However, the effects of chronic administration of lurasidone, vortioxetine, and the selective serotonin reuptake inhibitor (SSRI), escitalopram, on 5-HT7R function remained to be clarified. Thus, to explore the mechanisms underlying the clinical effects of vortioxetine, escitalopram, and lurasidone, the present study determined the effects of these agents on thalamocortical glutamatergic transmission, which contributes to emotional/mood perception, using multiprobe microdialysis and 5-HT7R expression using capillary immunoblotting. Acute local administration of a 5-HT7R agonist and antagonist into the mediodorsal thalamic nucleus (MDTN) enhanced and reduced thalamocortical glutamatergic transmission, induced by N-methyl-D-aspartate (NMDA)/glutamate receptor inhibition in the reticular thalamic nucleus (RTN). Acute local administration of a relevant therapeutic concentration of vortioxetine and lurasidone into the MDTN suppressed the thalamocortical glutamatergic transmission via 5-HT7R inhibition, whereas that of escitalopram activated 5-HT7R. Subchronic administration of effective doses of vortioxetine and lurasidone (for 7 days) reduced the thalamocortical glutamatergic transmission, but escitalopram did not affect it, whereas subchronic administration of these three agents attenuated the stimulatory effects of the 5-HT7R agonist on thalamocortical glutamatergic transmission. Subchronic administration of effective doses of vortioxetine, lurasidone, and escitalopram downregulated the 5-HT7R expression of the plasma membrane in the MDTN; the 5-HT7R downregulation induced by vortioxetine and lurasidone was observed at 3 days, but that induced by escitalopram required a longer duration of 7 days. These results indicate that chronic administration of vortioxetine, escitalopram, and lurasidone generate downregulation of 5-HT7R in the thalamus; however, the direct inhibition of 5-HT7R associated with vortioxetine and lurasidone generates more rapid downregulation than the indirect elevation of the extracellular serotonin level via serotonin transporter inhibition by escitalopram.


Asunto(s)
Antidepresivos/farmacología , Antipsicóticos/farmacología , Citalopram/farmacología , Clorhidrato de Lurasidona/farmacología , Receptores de Serotonina/metabolismo , Vortioxetina/farmacología , Animales , Antidepresivos/administración & dosificación , Antipsicóticos/administración & dosificación , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Citalopram/administración & dosificación , Ácido Glutámico/metabolismo , Clorhidrato de Lurasidona/administración & dosificación , Masculino , Ratas , Ratas Sprague-Dawley , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transmisión Sináptica/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/metabolismo , Vortioxetina/administración & dosificación
3.
Neurosci Res ; 170: 195-200, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32702384

RESUMEN

Change-N1 peaking 90-180 ms after changes in a sound feature of a continuous sound is clearly attenuated by a preceding change stimulus (called a "prepulse"). Here, we investigated the effects of a preceding decrease in sound pressure on the degree of inhibition of the subsequent Change-N1 amplitude. Using 100-Hz click train sounds, we obtained Change-N1s from 11 healthy volunteers. The two types of test stimuli were an abrupt 10-dB increase from the baseline (70 dB) and the insertion of a 0.45-ms inter-aural time difference in the middle of the sound. Three consecutive clicks at 30, 40, and 50 ms before the change onset that was used as a prepulse were weaker than the background by 5 or 10 dB. The Change-N1 elicited by the two test stimuli was attenuated more strongly by the weaker prepulse, which was not congruent with the theory that the inhibition of the subsequent sensory/sensory-motor processing depends on the sound pressure level of a prepulse. These results suggest that a change in any type of sound feature elicits a change-related response that is inhibited by any type of preceding change stimulus, which reflects auto-inhibition of the change-responding circuit.


Asunto(s)
Potenciales Evocados Auditivos , Reflejo de Sobresalto , Estimulación Acústica , Humanos , Inhibición Psicológica , Inhibición Prepulso
4.
Biomolecules ; 10(7)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32630356

RESUMEN

Noncompetitive N-methyl-D-aspartate/glutamate receptor (NMDAR) antagonists contribute to the pathophysiology of schizophrenia and mood disorders but improve monoaminergic antidepressant-resistant mood disorder and suicidal ideation. The mechanisms of the double-edged sword clinical action of NMDAR antagonists remained to be clarified. The present study determined the interaction between the NMDAR antagonist (MK801), α1 adrenoceptor antagonist (prazosin), and α2A adrenoceptor agonist (guanfacine) on mesocortical and mesothalamic catecholaminergic transmission, and thalamocortical glutamatergic transmission using multiprobe microdialysis. The inhibition of NMDAR in the locus coeruleus (LC) by local MK801 administration enhanced both the mesocortical noradrenergic and catecholaminergic coreleasing (norepinephrine and dopamine) transmissions. The mesothalamic noradrenergic transmission was also enhanced by local MK801 administration in the LC. These mesocortical and mesothalamic transmissions were activated by intra-LC disinhibition of transmission of γ-aminobutyric acid (GABA) via NMDAR inhibition. Contrastingly, activated mesothalamic noradrenergic transmission by MK801 enhanced intrathalamic GABAergic inhibition via the α1 adrenoceptor, resulting in the suppression of thalamocortical glutamatergic transmission. The thalamocortical glutamatergic terminal stimulated the presynaptically mesocortical catecholaminergic coreleasing terminal in the superficial cortical layers, but did not have contact with the mesocortical selective noradrenergic terminal (which projected terminals to deeper cortical layers). Furthermore, the α2A adrenoceptor suppressed the mesocortical and mesothalamic noradrenergic transmissions somatodendritically in the LC and presynaptically/somatodendritically in the reticular thalamic nucleus (RTN). These discrepancies between the noradrenergic and catecholaminergic transmissions in the mesocortical and mesothalamic pathways probably constitute the double-edged sword clinical action of noncompetitive NMDAR antagonists.


Asunto(s)
Maleato de Dizocilpina/administración & dosificación , Guanfacina/administración & dosificación , Locus Coeruleus/metabolismo , Prazosina/administración & dosificación , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Corteza Cerebral/metabolismo , Maleato de Dizocilpina/farmacología , Dopamina/metabolismo , Guanfacina/farmacología , Hipotálamo/metabolismo , Locus Coeruleus/efectos de los fármacos , Masculino , Microdiálisis/instrumentación , Norepinefrina/metabolismo , Prazosina/farmacología , Ratas , Transmisión Sináptica/efectos de los fármacos
5.
Br J Pharmacol ; 176(20): 4002-4018, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31347694

RESUMEN

BACKGROUND AND PURPOSE: Lurasidone is an atypical mood-stabilizing antipsychotic with a unique receptor-binding profile, including 5-HT7 receptor antagonism; however, the detailed effects of 5-HT7 receptor antagonism on various transmitter systems relevant to schizophrenia, particularly the thalamo-insular glutamatergic system and the underlying mechanisms, are yet to be clarified. EXPERIMENTAL APPROACH: We examined the mechanisms underlying the clinical effects of lurasidone by measuring the release of l-glutamate, GABA, dopamine, and noradrenaline in the reticular thalamic nucleus (RTN), mediodorsal thalamic nucleus (MDTN) and insula of freely moving rats in response to systemic injection or local infusion of lurasidone or MK-801 using multiprobe microdialysis with ultra-HPLC. KEY RESULTS: Systemic MK-801 (0.5 mg·kg-1 ) administration increased insular release of l-glutamate, dopamine, and noradrenaline but decreased GABA release. Systemic lurasidone (1 mg·kg-1 ) administration also increased insular release of l-glutamate, dopamine, and noradrenaline but without affecting GABA. Local lurasidone administration into the insula (3 µM) did not affect MK-801-induced insular release of l-glutamate or catecholamine, whereas local lurasidone administration into the MDTN (1 µM) inhibited MK-801-induced insular release of l-glutamate and catecholamine, similar to the 5-HT7 receptor antagonist SB269970. CONCLUSIONS AND IMPLICATIONS: The present results indicate that MK-801-induced insular l-glutamate release is generated by activation of thalamo-insular glutamatergic transmission via MDTN GABAergic disinhibition resulting from NMDA receptor inhibition in the MDTN and RTN. Lurasidone inhibited this MK-801-evoked insular l-glutamate release through inhibition of excitatory 5-HT7 receptor in the MDTN. These effects on thalamo-insular glutamatergic transmission may contribute to the antipsychotic and mood-stabilizing actions of lurasidone.


Asunto(s)
Antipsicóticos/farmacología , Maleato de Dizocilpina/antagonistas & inhibidores , Clorhidrato de Lurasidona/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Animales , Antipsicóticos/administración & dosificación , Maleato de Dizocilpina/farmacología , Ácido Glutámico/efectos de los fármacos , Ácido Glutámico/metabolismo , Clorhidrato de Lurasidona/administración & dosificación , Masculino , Núcleo Talámico Mediodorsal/efectos de los fármacos , Perfusión , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Antagonistas de la Serotonina/administración & dosificación , Transmisión Sináptica/efectos de los fármacos , Tálamo/efectos de los fármacos , Tálamo/metabolismo
6.
Pharmacol Res Perspect ; 7(1): e00457, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30784207

RESUMEN

Deficiencies in N-methyl-d-aspartate (NMDA)/glutamate receptor (NMDAR) signaling have been considered central to the cognitive impairments of schizophrenia; however, an NMDAR antagonist memantine (MEM) improves cognitive impairments of Alzheimer's disease and schizophrenia. These mechanisms of paradoxical clinical effects of NMDAR antagonists remain unclear. To explore the mechanisms by which MK801 and MEM affect thalamocortical transmission, we determined interactions between local administrations of MK801, MEM, system xc- (Sxc), and metabotropic glutamate receptors (mGluRs) on extracellular glutamate and GABA levels in the mediodorsal thalamic nucleus (MDTN) and medial prefrontal cortex (mPFC) using dual-probe microdialysis with ultra-high-pressure liquid chromatography. Effects of MK801 and MEM on Sxc activity were also determined using primary cultured astrocytes. Sxc activity was enhanced by MEM, but was unaffected by MK801. MK801 enhanced thalamocortical glutamatergic transmission by GABAergic disinhibition in the MDTN. In the MDTN and the mPFC, MEM weakly increased glutamate release by activating Sxc, whereas MEM inhibited thalamocortical glutamatergic transmission. Paradoxical effects of MEM were induced following secondary activation of inhibitory II-mGluR and III-mGluR by exporting glutamate from astroglial Sxc. The present results suggest that the effects of therapeutically relevant concentrations of MEM on thalamocortical glutamatergic transmission are predominantly caused by activation of Sxc rather than inhibition of NMDAR. These demonstrations suggest that the combination between reduced NMDAR and activated Sxc contribute to the neuroprotective effects of MEM. Furthermore, activation of Sxc may compensate for the cognitive impairments that are induced by hyperactivation of thalamocortical glutamatergic transmission following activation of Sxc/II-mGluR in the MDTN and Sxc/II-mGluR/III-mGluR in the mPFC.


Asunto(s)
Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Memantina/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Ácido Glutámico/metabolismo , Masculino , Núcleo Talámico Mediodorsal/metabolismo , Microdiálisis/métodos , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/efectos de los fármacos , Tálamo/metabolismo
7.
Int J Mol Sci ; 19(11)2018 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-30463253

RESUMEN

To explore pathophysiology of schizophrenia, this study analyzed the regulation mechanisms that are associated with cystine/glutamate antiporter (Sxc), group-II (II-mGluR), and group-III (III-mGluR) metabotropic glutamate-receptors in thalamo-cortical glutamatergic transmission of MK801-induced model using dual-probe microdialysis. L-glutamate release in medial pre-frontal cortex (mPFC) was increased by systemic- and local mediodorsal thalamic nucleus (MDTN) administrations of MK801, but was unaffected by local administration into mPFC. Perfusion into mPFC of activators of Sxc, II-mGluR, and III-mGluR, and into the MDTN of activators of Sxc, II-mGluR, and GABAA receptor inhibited MK801-evoked L-glutamate release in mPFC. Perfusion of aripiprazole (APZ) into MDTN and mPFC also inhibited systemic MK801-evoked L-glutamate release in mPFC. Inhibition of II-mGluR in mPFC and MDTN blocked inhibitory effects of Sxc-activator and APZ on MK801-evoked L-glutamate release; however, their inhibitory effects were blocked by the inhibition of III-mGluR in mPFC but not in MDTN. These results indicate that reduced activation of the glutamate/NMDA receptor (NMDAR) in MDTN enhanced L-glutamate release in mPFC possibly through GABAergic disinhibition in MDTN. Furthermore, MDTN-mPFC glutamatergic transmission receives inhibitory regulation of Sxc/II-mGluR/III-mGluR functional complex in mPFC and Sxc/II-mGluR complex in MDTN. Established antipsychotic, APZ inhibits MK801-evoked L-glutamate release through the activation of Sxc/mGluRs functional complexes in both MDTN and mPFC.


Asunto(s)
Antiportadores/metabolismo , Aripiprazol/farmacología , Maleato de Dizocilpina/farmacología , Ácido Glutámico/metabolismo , N-Metilaspartato/antagonistas & inhibidores , Corteza Prefrontal/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Tálamo/fisiopatología , Acetilcisteína/farmacología , Animales , Aripiprazol/administración & dosificación , Maleato de Dizocilpina/administración & dosificación , Masculino , Modelos Biológicos , Perfusión , Corteza Prefrontal/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de GABA-A/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Tálamo/efectos de los fármacos
8.
Dig Surg ; 35(1): 1-10, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28171868

RESUMEN

PURPOSES: To clarify the incidence and risk factors of postoperative delirium in patients following pancreatic surgery, and the impact of yokukansan (TJ-54) administered to reduce delirium. METHODS: Fifty-nine consecutive patients who underwent pancreatic surgery (2012.4-2013.5) were divided into 2 groups: TJ-54 group: patients who received TJ-54 (n = 21) due to insomnia and the No-TJ-54 group: patients who did not receive TJ-54 (n = 38), and the medical records including the delirium rating scale - Japanese version (DRS-J) were retrospectively reviewed. RESULTS: Postoperative delirium occurred in 2 patients (9.5%) in the TJ-54 group and in 4 (10.5%) patients in the No-TJ-54 group (p = 0.90). The DRS-J on 5 days after surgery was lower in the TJ-54 group than in the No-TJ-54 group (rough p = 0.006), however, without any statistically significant differences with the Bonferroni correction. As for the hospital cost, there was no difference between the TJ-54 and the No-TJ-54 groups (p = 0.78). History of delirium was identified as an independent risk factor of postoperative delirium. CONCLUSION: The patients with preoperative insomnia, who were treated with TJ-54, did not have a higher incidence of postoperative delirium, compared to those without preoperative insomnia. The patients who had a history of delirium have an increased risk of postoperative delirium and should be cared for and treated prophylactically to prevent it.


Asunto(s)
Fármacos del Sistema Nervioso Central/uso terapéutico , Delirio , Medicamentos Herbarios Chinos/uso terapéutico , Pancreatectomía , Pancreaticoduodenectomía , Complicaciones Posoperatorias , Adenocarcinoma/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Delirio/epidemiología , Delirio/etiología , Delirio/prevención & control , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/cirugía , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Complicaciones Posoperatorias/prevención & control , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento
9.
Clin EEG Neurosci ; 49(3): 152-158, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-28490194

RESUMEN

A weak preceding sound stimulus attenuates the startle response evoked by an intense sound stimulus. Like startle reflexes, change-related auditory responses are suppressed by a weak leading stimulus (ie, a prepulse). We aim to examine whether a prepulse inhibits cerebral responses to the sound offset and how the prepulse magnitude affects the degree of the prepulse inhibition (PPI). Using magnetoencephalography, we recorded the Off-P50m elicited by an offset of a train sound of 100-Hz clicks in 12 healthy subjects. A single click slightly louder (+1.5, +3, or +5 dB) than the background sound of 80 dB was inserted 50 ms before the sound offset as a prepulse. We performed a dipole source analysis of the Off-P50m, and we measured its latency and amplitude using the source strength waveforms. The origin of the Off-P50m was estimated to be the auditory cortex on both hemispheres. The Off-P50m was clearly attenuated by the prepulses, and the degree of PPI was greater with a louder prepulse. The Off-P50m is considered to be a simple change-related response, which does not overlap with a processing of incoming sounds. Thus, the Off-P50m and its PPI comprise a valuable tool for investigating the neural inhibitory system.


Asunto(s)
Corteza Auditiva/fisiología , Magnetoencefalografía , Inhibición Prepulso/fisiología , Reflejo de Sobresalto/fisiología , Estimulación Acústica/métodos , Adulto , Electroencefalografía/métodos , Potenciales Evocados Auditivos , Femenino , Humanos , Magnetoencefalografía/métodos , Masculino , Persona de Mediana Edad , Adulto Joven
10.
Br J Pharmacol ; 168(5): 1088-100, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22882023

RESUMEN

BACKGROUND AND PURPOSE: Anticonvulsants have been developed according to the traditional neurotransmission imbalance hypothesis. However, the anticonvulsive pharmacotherapy currently available remains unsatisfactory. To develop new antiepileptic drugs with novel antiepileptic mechanisms, we have tested the antiepileptic actions of ONO-2506, a glial modulating agent, and its effects on tripartite synaptic transmission. EXPERIMENTAL APPROACH: Dose-dependent effects of ONO-2506 on maximal-electroshock seizure (MES), pentylenetetrazol-induced seizure (PTZ) and epileptic discharge were determined in a genetic model of absence epilepsy in mice (Cacna1a(tm2Nobs/tm2Nobs) strain). Antiepileptic mechanisms of ONO-2506 were analysed by examining the interaction between ONO-2506 and transmission-modulating toxins (tetanus toxin, fluorocitrate, tetrodotoxin) on release of l-glutamate, d-serine, GABA and kynurenic acid in the medial-prefrontal cortex (mPFC) of freely moving rats using microdialysis and primary cultured rat astrocytes. KEY RESULTS: ONO-2506 inhibited spontaneous epileptic discharges in Cacna1a(tm2Nobs/tm2Nobs) mice without affecting MES or PTZ. Given systemically, ONO-2506 increased basal release of GABA and kynurenic acid in the mPFC through activation of both neuronal and glial exocytosis, but inhibited depolarization-induced releases of all transmitters. ONO-2506 increased basal glial release of kynurenic acid without affecting those of l-glutamate, d-serine or GABA. However, ONO-2506 inhibited AMPA-induced releases of l-glutamate, d-serine, GABA and kynurenic acid. CONCLUSIONS AND IMPLICATIONS: ONO-2506 did not affect traditional convulsive tests but markedly inhibited epileptic phenomena in the genetic epilepsy mouse model. ONO-2506 enhanced release of inhibitory neuro- and gliotransmitters during the resting stage and inhibited tripartite transmission during the hyperactive stage. The results suggest that ONO-2506 is a novel potential glial-targeting antiepileptic drug. LINKED ARTICLE: This article is commented on by Onat, pp. 1086-1087 of this issue. To view this commentary visit http://dx.doi.org/10.1111/bph.12050.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Caprilatos/uso terapéutico , Epilepsia/tratamiento farmacológico , Animales , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Caprilatos/farmacología , Células Cultivadas , Convulsivantes , Modelos Animales de Enfermedad , Electrochoque , Epilepsia/etiología , Epilepsia/metabolismo , Epilepsia/fisiopatología , Ácido Glutámico/metabolismo , Ácido Quinurénico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Pentilenotetrazol , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Serina/metabolismo , Transmisión Sináptica , Ácido gamma-Aminobutírico/metabolismo
11.
Neurosci Res ; 73(3): 248-51, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22525281

RESUMEN

Quickly detecting changes in the surrounding environment is one of the most important functions of sensory processing. Comparison of a new event with preceding sensory conditions is necessary for the change-detection process. A sudden change in a continuous sound elicits auditory evoked potentials that peak approximately 100 ms after the onset of the change (Change-N1). In the present study, we recorded Change-N1 under an oddball paradigm in 19 healthy subjects using an abruptly moving sound (SM-stimulus) as a deviant stimulus and investigated effects of the probability of the SM-stimulus to reveal whether Change-N1 is a memory-based response. We compared the amplitude and latency of Change-N1 elicited by the SM-stimulus among three probability conditions (33, 50 and 100%). As the probability of the SM-stimulus decreased, the amplitude of Change-N1 increased and its latency decreased. The present results indicate that the preceding sensory history affects Change-N1 elicited by the SM-stimulus.


Asunto(s)
Atención/fisiología , Percepción Auditiva/fisiología , Potenciales Evocados Auditivos/fisiología , Memoria/fisiología , Estimulación Acústica/métodos , Adulto , Electroencefalografía/métodos , Femenino , Humanos , Masculino , Modelos Estadísticos , Tiempo de Reacción/fisiología , Localización de Sonidos/fisiología , Adulto Joven
12.
Cell Biochem Funct ; 27(8): 526-34, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19862690

RESUMEN

The requirement of large amounts of the recombinant human bone morphogenetic protein-2 (BMP-2) produces a huge translational barrier for its routine clinical use due to high cost. This leads to an urgent need to develop alternative methods to lower costs and/or increase efficacies for using BMP-2. In this study, we describe the development and optimization of a cell-based assay that is sensitive, reproducible, and reliable in identifying reagents that potentiate the effects of BMP-2 in inducing transdifferentiation of C2C12 myoblasts into the osteoblastic phenotype. The assay is based on a BMP-responsive Smad1-driven luciferase reporter gene. LIM mineralization protein-1 (LMP-1) is a novel intracellular LIM domain protein that has been shown by our group to enhance cellular responsiveness to BMP-2. Our previous report elucidated that the binding of LMP-1 with the WW2 domain in Smad ubiquitin regulatory factor-1 (Smurf1) rescues the osteogenic Smads from degradation. Here, using the optimized cell-based assay, we first evaluated the activity of the recombinantly prepared proteins, LMP-1, and its mutant (LMP-1DeltaSmurf1) that lacks the Smurf1-WW2 domain-binding motif. Both the wild type and the mutant proteins were engineered to contain an 11-amino acid HIV-TAT protein derived membrane transduction domain to aid the cellular delivery of recombinant proteins. The cell-based reporter assay confirmed that LMP-1 potentiates the BMP-induced stimulation of C2C12 cells towards the osteoblastic phenotype. The potentiating effect of LMP-1 was significantly reduced when a specific-motif known to interact with Smurf1 was mutated. We validated the results obtained in the reporter assay by also monitoring the expression of mRNA for osteocalcin and alkaline phosphatase (ALP) which is widely accepted osteoblast differentiation marker genes. Finally, we provide further confirmation of our results by measuring the activity of alkaline phosphatase in support of the accuracy and reliability of our cell-based assay. Direct delivery of synthesized protein can be limited by high cost, instability or inadequate post-translational modifications. Thus, there would be a clear benefit for a low cost, cell penetrable chemical compound. We successfully used our gene expression-based assay to choose an active compound from a select group of compounds that were identified by computational screenings as the most likely candidates for mimicking the function of LMP-1. Among them, we selected SVAK-3, a compound that showed a dose-dependent potentiation of BMP-2 activity in inducing osteoblastic differentiation of C2C12 cells. We show that either the full length LMP-1 protein or its potential mimetic compound consistently exhibit similar potentiation of BMP-2 activity even when multiple markers of the osteoblastic phenotype were parallely monitored.


Asunto(s)
Bioensayo/métodos , Proteína Morfogenética Ósea 2/agonistas , Proteína Morfogenética Ósea 2/farmacología , Evaluación Preclínica de Medicamentos/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proteínas del Citoesqueleto , Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM , Ratones , Unión Proteica , Bibliotecas de Moléculas Pequeñas/síntesis química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
Neurosci Lett ; 454(1): 91-6, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19429061

RESUMEN

To clarify the antiepileptic mechanisms of zonisamide (ZNS), we determined the interaction between ZNS and inositol-1,4,5-triphosphate receptor (IP3R) on exocytosis of GABA and glutamate in rat frontal cortex using microdialysis. ZNS increased basal GABA release, but not glutamate, concentration-dependently, and reduced concentration-dependently K(+)-evoked GABA and glutamate releases. Inhibition and activation of IP3R reduced and enhanced basal and K(+)-evoked GABA releases, respectively. The K(+)-evoked glutamate release was reduced and enhanced by IP3R antagonist and agonist, respectively, whereas basal glutamate release was increased by IP3R agonist but not affected by IP3R antagonist. Under extracellular Ca(2+) depletion, IP3R agonist increased basal GABA and glutamate releases. The latter effects of IP3R agonist were weakly enhanced by ZNS, but such stimulatory action of ZNS was abolished by extracellular Ca(2+) depletion. In contrast, ZNS inhibited the stimulatory effect of IP3R agonist on K(+)-evoked release. The stimulatory effect of IP3R agonist on basal release was regulated by N-type voltage-sensitive Ca(2+) channel (VSCC) rather than P- and L-type VSCCs, whereas the stimulatory effect of IP3R agonist on K(+)-evoked release was regulated by P- and L-type VSCCs rather than N-type VSCC. These results suggest that ZNS-activated N-type VSCC enhances IP3R-associated neurotransmitter release during resting stage, whereas ZNS-induced suppression of P- and L-type VSCCs possibly attenuates IP3R-associated neurotransmitter release during neuronal hyperexcitability. Therefore, the combination of both of these two actions of ZNS on IP3R-associated neurotransmitter release mechanism seems to be involved, at least in part, in the mechanisms of antiepileptic and neuroprotective actions of ZNS.


Asunto(s)
Anticonvulsivantes/farmacología , Lóbulo Frontal/efectos de los fármacos , Ácido Glutámico/efectos de los fármacos , Receptores de Inositol 1,4,5-Trifosfato/efectos de los fármacos , Isoxazoles/farmacología , Ácido gamma-Aminobutírico/efectos de los fármacos , Animales , Canales de Calcio , Relación Dosis-Respuesta a Droga , Lóbulo Frontal/metabolismo , Ácido Glutámico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Microdiálisis , Ratas , Ratas Sprague-Dawley , Zonisamida , Ácido gamma-Aminobutírico/metabolismo
14.
J Neurosci ; 28(47): 12465-76, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020039

RESUMEN

Mutations of genes encoding alpha4, beta2, or alpha2 subunits (CHRNA4, CHRNB2, or CHRNA2, respectively) of nAChR [neuronal nicotinic ACh (acetylcholine) receptor] cause nocturnal frontal lobe epilepsy (NFLE) in human. NFLE-related seizures are seen exclusively during sleep and are characterized by three distinct seizure phenotypes: "paroxysmal arousals," "paroxysmal dystonia," and "episodic wandering." We generated transgenic rat strains that harbor a missense mutation S284L, which had been identified in CHRNA4 in NFLE. The transgenic rats were free of biological abnormalities, such as dysmorphology in the CNS, and behavioral abnormalities. The mRNA level of the transgene (mutant Chrna4) was similar to the wild type, and no distorted expression was detected in the brain. However, the transgenic rats showed epileptic seizure phenotypes during slow-wave sleep (SWS) similar to those in NFLE exhibiting three characteristic seizure phenotypes and thus fulfilled the diagnostic criteria of human NFLE. The therapeutic response of these rats to conventional antiepileptic drugs also resembled that of NFLE patients with the S284L mutation. The rats exhibited two major abnormalities in neurotransmission: (1) attenuation of synaptic and extrasynaptic GABAergic transmission and (2) abnormal glutamate release during SWS. The currently available genetically engineered animal models of epilepsy are limited to mice; thus, our transgenic rats offer another dimension to the epilepsy research field.


Asunto(s)
Epilepsia del Lóbulo Frontal/genética , Leucina/genética , Mutación/genética , Receptores Nicotínicos/genética , Serina/genética , Transmisión Sináptica/genética , Ácido gamma-Aminobutírico/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Conducta Animal/fisiología , Corteza Cerebral/citología , Corteza Cerebral/patología , Relación Dosis-Respuesta a Droga , Electroencefalografía , Epilepsia del Lóbulo Frontal/dietoterapia , Epilepsia del Lóbulo Frontal/fisiopatología , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Ácido Glutámico/metabolismo , Calor/efectos adversos , Técnicas In Vitro , Microscopía Inmunoelectrónica , Actividad Motora/genética , Destreza Motora/fisiología , Neurotransmisores/metabolismo , Nicotina/farmacología , Dimensión del Dolor/métodos , Pentilenotetrazol/farmacología , Fenotipo , Ratas , Ratas Transgénicas , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/ultraestructura , Transmisión Sináptica/efectos de los fármacos
15.
Epilepsia ; 43 Suppl 9: 21-5, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12383275

RESUMEN

Genetic defects have been recently identified in certain inherited epilepsy syndromes in which the phenotypes are similar to those of common idiopathic epilepsies. Mutations in the neuronal nicotinic acetylcholine receptor alpha4 and beta2 subunit genes have been detected in families with autosomal dominant nocturnal frontal lobe epilepsy. Both receptors are components of neuronal acetylcholine receptor, a ligand-gated ion channel in the brain. Furthermore, mutations of two K+ channel genes also were identified as the underlying genetic abnormalities of benign familial neonatal convulsions. Mutations in the voltage-gated Na+-channel alpha1 and beta1 subunit genes were found as the cause of generalized epilepsy with febrile seizures plus, a clinical subset of febrile convulsions. Mutation of a voltage-gated K+-channel gene can cause partial seizures associated with periodic ataxia type 1 and some forms of juvenile myoclonic epilepsy can result from mutations of a Ca2+ channel. This line of evidence suggests the involvement of channels expressed in the brain in the pathogenesis of certain types of epilepsy. Our working hypothesis is to view certain idiopathic epilepsies as disorders of ion channels (i.e., "channelopathies"). Such a hypothesis should provide a new insight into our understanding of the genetic background of epilepsy.


Asunto(s)
Epilepsia Benigna Neonatal , Epilepsia , Epilepsia/genética , Canales Iónicos/genética , Adulto , Canales de Calcio/genética , Aberraciones Cromosómicas , Epilepsias Parciales/genética , Epilepsia/etiología , Epilepsia Benigna Neonatal/genética , Epilepsia del Lóbulo Frontal/genética , Epilepsia Generalizada/genética , Genes Dominantes , Humanos , Recién Nacido , Biología Molecular , Mutación , Linaje , Fenotipo , Canales de Potasio/genética , Canales de Potasio con Entrada de Voltaje/genética , Convulsiones Febriles/genética , Sueño , Síndrome , Factores de Tiempo
16.
Epilepsy Res ; 49(1): 49-60, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11948007

RESUMEN

To clarify the mechanisms of action of antiepileptic drugs (AEDs), carbamazepine (CBZ) and zonisamide (ZNS), on exocytosis mechanisms, the present study determined the concentration-dependent action of CBZ and ZNS, as well as the interaction between these AEDs and voltage-sensitive Ca(2+) channel (VSCC) activity on basal, Ca(2+)- and K(+)-evoked acetylcholine (ACh) release in frontal cortex of freely moving rat using in vivo microdialysis. Perfusion with therapeutic-relevant concentrations of CBZ and ZNS increased basal ACh release, which was regulated by N-type VSCC predominantly and P-type VSCC weakly, whereas supratherapeutic-relevant concentrations of these AEDs reduced this release. The 3.4 mM Ca(2+)-evoked release, which was regulated by N-type VSCC selectively, but not by P-type VSCC, was increased by therapeutic-relevant concentrations of CBZ and ZNS, whereas this release was reduced by supratherapeutic-relevant concentrations of them. The 50 mM K(+)-evoked release, which was regulated by P-type VSCC predominantly and N-type VSCC weakly, was decreased by CBZ and ZNS, in a concentration-dependent manner. These findings indicate that the interplay between enhancement of basal ACh release and reduction of depolarization-related ACh release in the frontal cortex are at least partially involved in a common mechanism of antiepileptic action between CBZ and ZNS.


Asunto(s)
Acetilcolina/metabolismo , Canales de Calcio/fisiología , Carbamazepina/farmacología , Lóbulo Frontal/efectos de los fármacos , Isoxazoles/farmacología , Animales , Canales de Calcio/metabolismo , Difusión , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Lóbulo Frontal/metabolismo , Masculino , Microdiálisis , Ratas , Ratas Wistar , Zonisamida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA