Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Neuropharmacology ; 39(1): 110-22, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10665824

RESUMEN

Quantification of receptor binding sites and their encoding mRNAs, and electrophysiological recordings, were used to assess central serotonin (5-HT) neurotransmission in rats 24 h after a 2-3 week treatment with the selective 5-HT reuptake inhibitor fluoxetine (8 mg/kg i.p., daily). Binding studies showed that this treatment affected neither 5-HT1A nor 5-HT1B binding sites in all brain areas examined. However, a significant decrease (-38%) in 5-HT1A mRNA levels in the anterior raphe area (but not forebrain regions) and increases in 5-HT1B mRNA levels in the striatum (+127%) and the cerebral cortex (+34%) were noted in fluoxetine-treated rats. Electrophysiological recordings in brain slices showed that chronic fluoxetine treatment reduced the potency of the 5-HT1A agonist 8-hydroxy-2-(di-n-propylamino)tetralin to inhibit neuronal activity in the dorsal raphe nucleus, but did not affect 5-HT1A-evoked responses of CA1 pyramidal cells in the hippocampus. These data further demonstrate that fluoxetine-induced adaptive changes in 5-HT neurotransmission exhibit marked regional differences. The decrease in 5-HT1A mRNA levels in the anterior raphe suggests that fluoxetine-induced desensitization of 5-HT1A autoreceptors involves changes at the transcription level.


Asunto(s)
Encéfalo/fisiología , Proteínas Portadoras/metabolismo , Fluoxetina/farmacología , Glicoproteínas de Membrana , Proteínas de Transporte de Membrana , Proteínas del Tejido Nervioso , Neuronas/fisiología , Receptores de Serotonina/metabolismo , Animales , Sitios de Unión , Encéfalo/efectos de los fármacos , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Corteza Cerebral/fisiología , Cuerpo Estriado/fisiología , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Estimulación Eléctrica , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Masculino , Neuronas/efectos de los fármacos , Prosencéfalo/fisiología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Núcleos del Rafe/fisiología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1B , Receptores de Serotonina/efectos de los fármacos , Receptores de Serotonina/genética , Receptores de Serotonina 5-HT1 , Transcripción Genética/efectos de los fármacos
2.
Neuroscience ; 91(3): 947-58, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10391473

RESUMEN

Extracellular and intracellular recording techniques were used to assess possible changes in the functional properties of 5-hydroxytryptamine-1A receptors in brain slices prepared from rats subjected to different stress paradigms. Whereas a 30-min restraint stress did not alter the inhibitory influence of ipsapirone on the firing of serotoninergic neurons in the dorsal raphe nucleus, the same session followed by a 24-h isolation produced a significant decrease in the potency of the 5-hydroxytryptamine-1A agonist to inhibit the electrical activity of these cells. Similarly, exposure of the animals to novel uncontrolled environmental conditions for 16 h significantly reduced the potency of ipsapirone to decrease the firing rate of serotoninergic neurons in brain stem slices. The effects of the latter two stressful paradigms were observed in slices from intact rats, but not in those from adrenalectomized animals. Intracellular recording showed that exposure of the animals to novel uncontrolled environmental conditions markedly reduced the potency of 5-carboxamidotryptamine to hyperpolarize serotoninergic neurons in the dorsal raphe nucleus and to decrease the input resistance of their plasma membrane. In contrast, the same stressful paradigm exerted no significant influence on the membrane effects of this 5-hydroxytryptamine-1A agonist on pyramidal cells in the CA1 hippocampal area. These data show that, like the direct application of corticosterone on to brain slices [Laaris N. et al. (1995) Neuropharmacology 34, 1201-1210], the stress-induced in vivo elevation of serum levels of endogenous corticosterone is associated with desensitization of somatodendritic 5-hydroxytryptamine-1A receptors in the dorsal raphe nucleus. The differential changes in 5-hydroxytryptamine-1A receptor sensitivity due to stress in the latter area versus the hippocampus further support the idea that somatodendritic and postsynaptic 5-hydroxytryptamine-1A receptors are regulated differently in the rat brain.


Asunto(s)
Encéfalo/metabolismo , Terminales Presinápticos/metabolismo , Receptores de Serotonina/metabolismo , Estrés Fisiológico/metabolismo , Sinapsis/metabolismo , Animales , Autorradiografía , Encéfalo/citología , Encéfalo/fisiopatología , Corticosterona/sangre , Electrofisiología , Técnicas In Vitro , Masculino , Neuronas/fisiología , Terminales Presinápticos/fisiología , Ratas , Ratas Sprague-Dawley , Restricción Física , Aislamiento Social , Estrés Fisiológico/etiología , Estrés Fisiológico/fisiopatología , Sinapsis/fisiología
3.
Br J Pharmacol ; 123(3): 449-62, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9504386

RESUMEN

1. The aim of the present work was to characterize the 5-hydroxytryptamine1A (5-HT1A) antagonistic actions of (-)-pindolol and WAY 100635 (N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl) cyclohexane carboxamide). Studies were performed on 5-HT1A receptors located on 5-hydroxytryptaminergic neurones in the dorsal raphe nucleus (DRN) and on pyramidal cells in the CA1 and CA3 regions of the hippocampus in rat brain slices. 2. Intracellular electrophysiological recording of CA1 pyramidal cells and 5-hydroxytryptaminergic DRN neurones showed that the 5-HT1A receptor agonist 5-carboxamidotryptamine (5-CT) evoked in both cell types a concentration-dependent cell membrane hyperpolarization and a decrease in cell input resistance. On its own, (-)-pindolol did not modify the cell membrane potential and resistance at concentrations up to 10 microM, but it antagonized the 5-CT effects in a concentration-dependent manner. Similar antagonism of 5-CT effects was observed in the CA3 hippocampal region. (-)-Pindolol also prevented the 5-HT1A receptor-mediated hyperpolarization of CA1 pyramidal cells due to 5-HT (15 microM). In contrast, the 5-HT-induced depolarization mediated by presumed 5-HT4 receptors persisted in the presence of 3 microM (-)-pindolol. 3. In the hippocampus, (-)-pindolol completely prevented the hyperpolarization of CA1 pyramidal cells by 100 nM 5-CT (IC50=92 nM; apparent KB=20.1 nM), and of CA3 neurones by 300 nM 5-CT (IC50=522 nM; apparent KB= 115.1 nM). The block by (-)-pindolol was surmounted by increasing the concentration of 5-CT, indicating a reversible and competitive antagonistic action. 4. Extracellular recording of the firing rate of 5-hydroxytryptaminergic neurones in the DRN showed that (-)-pindolol blocked, in a concentration-dependent manner, the decrease in firing elicited by 100 nM 5-CT (IC50=598 nM; apparent KB= 131.7 nM) or 100 nM ipsapirone (IC50= 132.5 nM; apparent KB= 124.9 nM). The effect of (-)-pindolol was surmountable by increasing the concentration of the agonist. Intracellular recording experiments showed that 10 microM (-)-pindolol were required to antagonize completely the hyperpolarizing effect of 100 nM 5-CT. 5. In vivo labelling of brain 5-HT1A receptors by i.v. administration of [3H]-WAY 100635 ([O-methyl-3H]-N-(2-(4-(2-methoxyphenyl)-1 -piperazinyl)ethyl-N-(2-pyridyl)cyclo-hexane-carboxamide) was used to assess their occupancy following in vivo treatment with (-)-pindolol. (-)-Pindolol (15 mg kg[-1]) injected i.p. either subchronically (2 day-treatment before i.v. injection of [3H]-WAY 100635) or acutely (20 min before i.v. injection of [3H]-WAY 100635) markedly reduced [3H]-WAY 100635 accumulation in all 5-HT1A receptor-containing brain areas. In particular, no differences were observed in the capacity of (-)-pindolol to prevent [3H]-WAY 100635 accumulation in the DRN and the CAI and CA3 hippocampal areas. 6. Intracellular electrophysiological recording of 5-hydroxytryptaminergic DRN neurones showed that WAY 100635 prevented the hyperpolarizing effect of 100 nM 5-CT in a concentration-dependent manner (IC50=4.9 nM, apparent KB=0.25 nM). In CA1 pyramidal cells, hyperpolarization induced by 50 nM 5-CT was also antagonized by WAY 100635 (IC50 = 0.80 nM, apparent KB= 0.28 nM).


Asunto(s)
Hipocampo/efectos de los fármacos , Pindolol/farmacología , Piperazinas/farmacología , Piridinas/farmacología , Núcleos del Rafe/efectos de los fármacos , Receptores de Serotonina/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Sinapsis/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Hipocampo/metabolismo , Técnicas In Vitro , Masculino , Núcleos del Rafe/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina/metabolismo , Receptores de Serotonina 5-HT1 , Sinapsis/metabolismo
4.
Br J Pharmacol ; 133(1): 138-44, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11325803

RESUMEN

1. Neuropeptides FF (NPFF) and AF (NPAF) are involved in pain modulation and opioid tolerance. These peptides were known to act through uncharacterized G protein-coupled receptors (GPCR). We describe here, using an aequorin-based assay as screening tool, that an orphan GPCR, previously designated HLWAR77, is a functional high affinity receptor for NPFF and related peptides. This receptor is further designated as NPFFR. 2. Binding experiments were performed with a new radioiodinated probe, [(125)I]-EYF, derived from the EFW-NPSF sequence of the rat NPFF precursor. Chinese hamster ovary (CHO) cell membranes expressing NPFFR bound [(125)I]-EYF with a K(d) of 0.06 nM. Various NPFF analogues and related peptides inhibited [(125)I]-EYF specific binding with the following rank order (K(i)): human NPAF (0.22 nM), SQA-NPFF (0.29 nM), NPFF (0.30 nM), 1DMe (0.31 nM), EYW-NPSF (0.32 nM), QFW-NPSF (0.35 nM), 3D (1.12 nM), Met-enk-RF-NH(2) (3.25 nM), FMRF-NH(2) (10.5 nM) and NPSF (12.1 nM). 3. The stimulatory activity of the same set of peptides was measured by a functional assay based on the co-expression of NPFFR, G(alpha 16) and apoaequorin. The rank order of potency was consistent with the results of the binding assay. 4. Membranes from NPFFR expressing CHO cells bound GTP gamma[(35)S] in the presence of SQA-NPFF. This functional response was prevented by pertussis toxin treatment, demonstrating the involvement of G(i) family members. 5. SQA-NPFF inhibited forskolin induced cyclic AMP accumulation in recombinant CHO cells in a dose dependent manner. This response was abolished as well by pertussis toxin pre-treatment. 6. RT -- PCR analysis of human tissues mRNA revealed that expression of NPFFR was mainly detected in placenta, thymus and at lower levels in pituitary gland, spleen and testis.


Asunto(s)
Oligopéptidos/metabolismo , Receptores de Neuropéptido/metabolismo , Aequorina , Animales , Unión Competitiva , Células CHO , Calcio/metabolismo , Clonación Molecular , Colforsina/antagonistas & inhibidores , Colforsina/farmacología , Cricetinae , AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Perfilación de la Expresión Génica , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Toxina del Pertussis , ARN Mensajero/análisis , ARN Mensajero/genética , Receptores de Neuropéptido/agonistas , Receptores de Neuropéptido/genética , Especificidad por Sustrato , Termodinámica , Factores de Virulencia de Bordetella/farmacología
5.
Eur J Pharmacol ; 365(2-3): 165-73, 1999 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-9988099

RESUMEN

The effects of long-term (7, 14 or 21 days) administration of the 5-HT1A receptor agonist alnespirone [5 mg/(kg day), i.p.] on the binding characteristics of 5-HT1A, 5-HT2A and 5-HT3 receptors, and the functional status of 5-HT1A autoreceptors were assessed using biochemical and electrophysiological approaches in rats. Whatever the treatment duration, the specific binding of [3H]8 hydroxy-2-(di-n-propylamino)tetralin ([3H]8-OH-DPAT), [3H]trans,4-[(3Z)3-(2-dimethylaminoethyl) oxyimino-3(2-fluorophenyl) propen-1-yl] phenol hemifumarate ([3H]SR 46349B), and [3H]S-zacopride to 5-HT1A, 5-HT2A and 5-HT3 receptors, respectively, were unaltered in all the brain areas examined. In contrast, in vitro electrophysiological recordings performed 24 h after the last injection of alnespirone showed that the potency of the 5-HT1A receptor agonist, 8-OH-DPAT, to depress the firing of serotoninergic neurons in the dorsal raphe nucleus, was significantly reduced after a 21-day treatment with alnespirone. However, no changes were noted after a 7-day or 14-day treatment. These data indicate that desensitization of somatodendritic 5-HT1A autoreceptors is a selective but slowly developing adaptive phenomenon in response to their chronic stimulation in rats.


Asunto(s)
Núcleos del Rafe/efectos de los fármacos , Receptores de Serotonina/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Compuestos de Espiro/farmacología , Animales , Autorradiografía , Conducta Animal/efectos de los fármacos , Sitios de Unión , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Masculino , Núcleos del Rafe/fisiología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A , Receptores de Serotonina 5-HT1 , Receptores de Serotonina 5-HT3 , Factores de Tiempo
6.
Naunyn Schmiedebergs Arch Pharmacol ; 352(2): 141-8, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7477436

RESUMEN

Electrophysiological and autoradiographic approaches were used to assess possible changes in 5-hydroxytryptamine (serotonin) 5-HT1A receptors in the rat dorsal raphe nucleus after a subchronic treatment with fluoxetine or paroxetine, two specific serotonin reuptake inhibitors with antidepressant properties. Fluoxetine or paroxetine were injected daily (5 mg/kg, i.p.) for various time periods up to 21 days. Electrophysiological recordings performed 24 h after the last injection showed that the potency of the 5-HT1A receptor agonist, 8-OH-DPAT, to depress the firing of serotoninergic neurons in the dorsal raphe nucleus within brain stem slices was significantly reduced as early as after a 3-day treatment with either drug. The proportion of recorded neurons showing desensitization of somatodendritic 5-HT1A autoreceptors increased along the treatment from approximately 40% on the 3rd day to 60-80% on the 21st day. At no time during the treatment, was the specific binding of [3H]8-OH-DPAT (agonist radioligand) or [3H]WAY-100 635 (antagonist radioligand) to 5-HT1A receptors modified in the dorsal raphe nucleus or in other brain areas, suggesting that neither the density nor the coupling of these receptors to G-proteins were probably altered in rats injected with fluoxetine or paroxetine for up to 21 days. These results show that adaptive desensitization of somatodendritic 5-HT1A autoreceptors within the dorsal raphe nucleus can already be detected after a 3-day treatment with selective serotonin reuptake inhibitors. Rather than the desensitization per se, it may be the progressive increase in the number of serotoninergic neurons with desensitized 5-HT1A autoreceptors which plays a critical role in the (slowly developing) antidepressant action of these drugs.


Asunto(s)
Fluoxetina/farmacología , Paroxetina/farmacología , Receptores de Serotonina/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Autorradiografía , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiología , Electrofisiología , Técnicas In Vitro , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Piperazinas/farmacología , Piridinas/farmacología , Núcleos del Rafe/citología , Núcleos del Rafe/efectos de los fármacos , Núcleos del Rafe/fisiología , Ratas , Ratas Sprague-Dawley , Agonistas de Receptores de Serotonina/farmacología
7.
Fundam Clin Pharmacol ; 11(3): 206-14, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9243251

RESUMEN

Somatodendritic 5-HT1A autoreceptors play a key role in the control of the electrical and metabolic activity of serotoninergic neurons in the dorsal raphe nucleus. These neurons also possess intracellular glucocorticoid receptors which may be involved in the well established modulation of serotonin (5-hydroxytryptamine, 5-HT) metabolism by corticosterone in stressed animals. The possible mediation by somatodendritic 5-HT1A autoreceptors of such corticosterone-dependent changes in serotoninergic neuron activity was investigated using an in vitro electrophysiological approach. 5-HT1A autoreceptor-mediated inhibition of the firing of serotoninergic neurons was examined in brain stem slices from rats whose serum corticosterone concentrations had been markedly increased (+100-200%) by two different stressful conditions. Immobilization for 30 or 90 min (restraint stress) did not modify the concentration-dependent inhibition of the firing of serotoninergic neurons by the 5-HT1A receptor agonist ipsapirone. In contrast, placing the rats in novel uncontrolled environmental conditions for 16 h significantly reduced the cell response to ipsapirone, indicating a decreased sensitivity of somatodendritic 5-HT1A autoreceptors. Such a change was not observed in adrenalectomized rats subjected to the same stressful conditions. These data show that some forms of stress can reduce the 5-HT1A autoreceptor-dependent inhibitory control of the electrophysiological activity of serotoninergic neurons in the dorsal raphe nucleus. Both the nature and duration of stress seem to be critical factors for triggering the (corticosterone-dependent) mechanism(s) responsible for the functional desensitization of 5-HT1A autoreceptors in stressed rats.


Asunto(s)
Autorreceptores/fisiología , Núcleos del Rafe/fisiología , Receptores de Serotonina/fisiología , Estrés Fisiológico/fisiopatología , Adrenalectomía , Animales , Autorreceptores/efectos de los fármacos , Corticosterona/sangre , Electrofisiología , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Pirimidinas/farmacología , Núcleos del Rafe/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina/efectos de los fármacos , Receptores de Serotonina 5-HT1 , Restricción Física , Agonistas de Receptores de Serotonina/farmacología , Estrés Fisiológico/etiología
8.
Encephale ; 21(2): 123-32, 1995.
Artículo en Francés | MEDLINE | ID: mdl-7781583

RESUMEN

The hypothesis that a dysfunction of serotonergic neurotransmission is implicated in depression is supported by the clinical efficiency of selective serotonin (5-hydroxytryptamine, 5-HT) reuptake inhibitors (SSRIs) in the treatment of depressive disorders. These drugs, such as fluoxetine and paroxetine, exert their antidepressant activity by increasing 5-HT concentration in the synaptic cleft and thus enhancing serotonergic neurotransmission. However, two to three weeks of treatment are necessary to see the first signs of clinical efficiency. Several hypothetical mechanisms have been put forward to account for this delay, taking into account pharmacokinetic considerations, neurotransmitter metabolism, and/or adaptive regulation of pre and/or post-synaptic receptors. The aim of this study was to look for such adaptive changes in the course of a 3-week treatment with fluoxetine (5 mg/kg/day, i.p.) or paroxetine (5 mg/kg/day, i.p.) in adult rats. In vitro binding and quantitative autoradiographic studies showed that neither 5-HT1A, 5-HT1B, 5-HT2A, nor 5-HT3 receptor binding sites in various brain areas were affected by these treatments. Furthermore, comparison of the specific binding of [3H]8-OH-DPAT to 5-HT1A receptors functionally coupled to G proteins with that of [3H]WAY 100635 to all 5-HT1A receptor binding sites (i.e. coupled and uncoupled with regard to G proteins) revealed no significant change in rats treated with either SSRI. Accordingly, the proportion of functional 5-HT1A receptors (i.e. those physically coupled to G proteins) appeared to remain unaltered all along a 3-week treatment with either fluoxetine or paroxetine. Nevertheless, in vitro electrophysiological recordings of serotonergic neurons in the dorsal raphe nucleus allowed the demonstration of a clearcut functional desensitization of somatodendritic 5-HT1A autoreceptors. Thus, the potency of the 5-HT1A autoreceptor agonist, 8-OH-DPAT, to depress the firing of serotonergic neurons in brain stem slices was significantly reduced as soon as after a 3-day treatment with either SSRI. The proportion of recorded neurons showing desensitization of somatodendritic 5-HT1A autoreceptors then increased along the treatment, and was generally larger with fluoxetine than with paroxetine. As 5-HT1A autoreceptor desensitization can contribute to facilitate serotoninergic neurotransmission, the remarkable efficiency of fluoxetine to trigger this adaptive regulatory mechanism might account, at least partly, for its potent antidepressant activity.


Asunto(s)
Encéfalo/efectos de los fármacos , Fluoxetina/farmacología , Paroxetina/farmacología , Receptores de Serotonina/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Autorradiografía , Autorreceptores/efectos de los fármacos , Tronco Encefálico/efectos de los fármacos , Técnicas de Cultivo , Inyecciones Intraperitoneales , Cuidados a Largo Plazo , Masculino , Ratas , Ratas Sprague-Dawley
9.
Synapse ; 27(4): 303-12, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9372553

RESUMEN

Previous in vitro studies showed that glucocorticoid receptor activation (notably by corticosterone) could induce a functional desensitization of somatodendritic 5-HT1A autoreceptors in the dorsal raphe nucleus [Laaris et al. (1995) Neuropharmacology 34:1201-1210], similar to that due to in vivo subchronic treatment with a 5-HT reuptake inhibitor, such as fluoxetine, in rats. In the present study, we investigated whether a link might exist between these effects, i.e., whether glucocorticoid receptor activation could be responsible for the fluoxetine-induced desensitization of 5-HT1A autoreceptors. In vitro recording in the dorsal raphe nucleus of brain-stem slices showed that subchronic treatment with fluoxetine (5 mg/kg intraperitoneally (i.p.), daily for 3-7 days) significantly reduced the potency of the 5-HT1A receptor agonist ipsapirone to inhibit the firing rate of serotoninergic neurons. Parallel experiments in adrenalectomized and sham-operated rats indicated that subchronic fluoxetine treatment produced a similar shift to the right of the ipsapirone inhibition curve in both groups of animals. Furthermore, the subchronic blockade of glucocorticoid receptors by RU 38486 (25 mg/kg subcutaneously (s.c.), daily) in intact rats treated with fluoxetine (5 mg/kg i.p., daily for 3 days) did not affect the ability of the latter treatment to reduce the potency of ipsapirone to inhibit the firing of serotoninergic neurons. These data suggest that glucocorticoid receptors (and their possible activation by corticosterone) are not involved in the functional desensitization of somatodendritic 5-HT1A autoreceptors, which occurs during long-term treatment with a serotonin reuptake inhibitor such as fluoxetine.


Asunto(s)
Antidepresivos de Segunda Generación/uso terapéutico , Dendritas/efectos de los fármacos , Fluoxetina/uso terapéutico , Glucocorticoides/fisiología , Receptores de Serotonina/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Glándulas Suprarrenales/fisiología , Adrenalectomía , Animales , Autorreceptores/efectos de los fármacos , Corticosterona/sangre , Corticosterona/fisiología , Técnicas In Vitro , Masculino , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Agonistas de Receptores de Serotonina/farmacología
10.
Acta Physiol Hung ; 84(4): 407-9, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-9328615

RESUMEN

The novel 5-HT1A receptor antagonist WAY 100635 [(N-(2-(-4(2-metoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl)cyc lohexane carboxamide)] has been tested on 5-HT1A receptor-mediated inhibition of firing and intracellularly recorded hyperpolarisation of serotoninergic cells of the dorsal raphe nucleus (DRN) and on hyperpolarisation of hippocampal CA1 pyramidal cells. WAY 100635 selectively blocked 5-HT1A receptor-mediated responses of 5-HT, 8-OH-DPAT, lesopitron and 5-CT. The antagonism of the hyperpolarisation elicited by 5-CT was competitive in the DRN and non competitive in CA1, probably because of the existence of a 5-HT1A receptor reserve in serotoninergic cells of DRN.


Asunto(s)
Neuronas/metabolismo , Piperazinas/farmacología , Células Piramidales/metabolismo , Piridinas/farmacología , Núcleos del Rafe/citología , Antagonistas de la Serotonina/farmacología , Animales , Electrofisiología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Técnicas In Vitro , Potenciales de la Membrana/efectos de los fármacos , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Núcleos del Rafe/efectos de los fármacos , Ratas
11.
J Sleep Res ; 5(4): 238-45, 1996 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9065875

RESUMEN

Sleep deprivation (SD) for one night induces mood improvement in depressed patients. However, relapse often occurs on the day after deprivation subsequently to a sleep episode. In light of the possible involvement of central serotonin (5-hydroxytryptamine, 5-HT) neurotransmission in both depression and sleep mechanisms, we presently investigated, in the rat, the effects of SD and recovery sleep on the electrophysiological response of 5-HT neurons in the nucleus raphe dorsalis (NRD) to an acute challenge with the 5-HT reuptake blocker citalopram. In all rats, citalopram induced a dose-dependent inhibition of the firing of NRD neurons recorded under chloral hydrate anaesthesia. After SD, achieved by placing rats in a slowly rotating cylinder for 24 h, the inhibitory action of citalopram was significantly reduced (with a concomitant 53% increase in its ED50 value). After a recovery period of 4 h, a normal susceptibility of the firing to citalopram was restored. The decreased sensitivity of 5-HT neuronal firing to the inhibitory effect of citalopram after SD probably results in an enhancement of 5-HT neurotransmission. Such an adaptive phenomenon (similar to that reported after chronic antidepressant treatment), and its normalization after recovery sleep, parallel the mood improvement effect of SD and the subsequent relapse observed in depressed patients. These data suggest that the associated changes in 5-HT autocontrol of the firing of NRD serotoninergic neurons are relevant to the antidepressant action of SD.


Asunto(s)
Citalopram/farmacología , Núcleos del Rafe/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/metabolismo , Privación de Sueño , Animales , Corticosterona/sangre , Masculino , Ratas , Ratas Wistar , Sueño REM , Vigilia/efectos de los fármacos
12.
J Pharmacol Exp Ther ; 278(2): 679-88, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8768719

RESUMEN

The aim of the present study was to examine the effects of N-(2-(4-2-methoxphenyl)-1-piperazinyl)ethyl)-N-(2-pyridnyl) cyclohexane carboxamide (WAY 100635) on 5-HT1A receptor-mediated responses in the dorsal raphe nucleus (DRN) and the CA1 hippocampal region. In DRN slices superfused with WAY 100635 (10 nM), the majority of putative 5-HT neurons increased their firing rate (13 +/- 2% of baseline rate). In addition, WAY 100635 completely prevented the decrease in firing rate produced by 5-HT (3-15 microM), 8-OH-DPAT (10 nM), 5-carboxamidotryptamine (20 nM) and lesopitron (100 nM). The antagonism exerted by WAY 100635 (IC50 = 0.95 +/- 0.12 nM against 15 microM 5-HT) was fully surmounted by increasing the concentration of 5-HT to 300 microM. In hippocampal slices, WAY 100635 (0.5-10 nM) did not alter the resting membrane potential or the membrane input resistance of intracellularly recorded CA1 pyramidal cells. However, WAY 100635 completely prevented (IC50 = 0.9-1.7 nM) the hyperpolarization and the decrease in membrane input resistance produced by 5-HT (15-30 microM) and by 5-carboxamidotryptamine (50-300 nM). In contrast, WAY 100635 affected neither the block of action potential frequency adaptation and slow afterhyperpolarization produced by 5-HT (15 microM) nor the hyperpolarization and decrease in membrane input resistance evoked by bath application of GABA(B) receptor agonist baclofen (10 microM). The cumulative concentration-hyperpolarization curve for 5-carboxamidotryptamine (3 nM-10 microM) was shifted to the right by WAY 100635 (apparent Kb = 0.23 +/- 0.07 nM), and the latter drug also reduced the maximal response to the agonist. These data show the WAY 100635 is a potent antagonist at 5-HT1A receptors, both in the DRN and in the CA1 region of the hippocampus. The antagonism is apparently competitive in the DRN and partly noncompetitive in the hippocampus. Kinetic characteristics of the antagonist-receptor interactions might account for these regional differences.


Asunto(s)
Hipocampo/efectos de los fármacos , Piperazinas/farmacología , Piridinas/farmacología , Núcleos del Rafe/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Animales , Recuento de Células/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
13.
J Biol Chem ; 276(26): 23795-804, 2001 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-11323418

RESUMEN

CCR5 is a CC chemokine receptor expressed on memory lymphocytes, macrophages, and dendritic cells and also constitutes the main coreceptor for macrophage-tropic (or R5) strains of human immunodeficiency viruses. In the present study, we investigated whether CCR5 was palmitoylated in its carboxyl-terminal domain by generating alanine substitution mutants for the three cysteine residues present in this region, individually or in combination. We found that wild-type CCR5 was palmitoylated, but a mutant lacking all three Cys residues was not. Through the use of green fluorescent fusion proteins and immunofluorescence studies, we found that the absence of receptor palmitoylation resulted in sequestration of CCR5 in intracellular biosynthetic compartments. By using the fluorescence recovery after photobleaching technique, we showed that the non-palmitoylated mutant had impaired diffusion properties within the endoplasmic reticulum. We next studied the ability of the mutants to bind and signal in response to chemokines. Chemokines binding and activation of G(i)-mediated signaling pathways, such as calcium mobilization and inhibition of adenylate cyclase, were not affected. However, the duration of the functional response, as measured by a microphysiometer, and the ability to increase [(35)S]guanosine 5'-3-O-(thio)triphosphate binding to membranes were severely affected for the non-palmitoylated mutant. The ability of RANTES (regulated on activation normal T cell expressed and secreted) and aminooxypentane-RANTES to promote CCR5 endocytosis was not altered by cysteine replacements. Finally, we found that the absence of receptor palmitoylation reduced the human immunodeficiency viruses coreceptor function of CCR5, but this effect was secondary to the reduction in surface expression. In conclusion, we found that palmitoylated cysteines play an important role in the intracellular trafficking of CCR5 and are likely necessary for efficient coupling of the receptor to part of its repertoire of signaling cascades.


Asunto(s)
Palmitatos/metabolismo , Receptores CCR5/metabolismo , Transducción de Señal , Acilación , Secuencia de Aminoácidos , Animales , Células CHO , Compartimento Celular , Membrana Celular/metabolismo , Quimiocina CCL5/farmacología , Cricetinae , Cisteína/fisiología , Citoplasma/metabolismo , Endocitosis , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , VIH/metabolismo , Humanos , Datos de Secuencia Molecular , Transporte de Proteínas , Receptores CCR5/genética , Receptores CCR5/fisiología , Alineación de Secuencia
14.
J Biol Chem ; 276(37): 34631-6, 2001 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-11457843

RESUMEN

Natural peptides displaying agonist activity on the orphan G protein-coupled receptor GPR54 were isolated from human placenta. These 54-, 14,- and 13-amino acid peptides, with a common RF-amide C terminus, derive from the product of KiSS-1, a metastasis suppressor gene for melanoma cells, and were therefore designated kisspeptins. They bound with low nanomolar affinities to rat and human GPR54 expressed in Chinese hamster ovary K1 cells and stimulated PIP(2) hydrolysis, Ca(2+) mobilization, arachidonic acid release, ERK1/2 and p38 MAP kinase phosphorylation, and stress fiber formation but inhibited cell proliferation. Human GPR54 was highly expressed in placenta, pituitary, pancreas, and spinal cord, suggesting a role in the regulation of endocrine function. Stimulation of oxytocin secretion after kisspeptin administration to rats confirmed this hypothesis.


Asunto(s)
Genes Supresores de Tumor , Metástasis de la Neoplasia/genética , Proteínas/genética , Receptores de Neuropéptido/análisis , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Proteínas de Unión al GTP/fisiología , Humanos , Kisspeptinas , Ligandos , Datos de Secuencia Molecular , Metástasis de la Neoplasia/prevención & control , Proteínas/aislamiento & purificación , Proteínas/metabolismo , Ratas , Receptores Acoplados a Proteínas G , Receptores de Kisspeptina-1 , Receptores de Neuropéptido/metabolismo , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA