Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 14(9): e0222855, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31557210

RESUMO

Trazodone is an antidepressant drug with considerable affinity for 5-HT1A receptors and α1-adrenoceptors for which the drug is competitive agonist and antagonist, respectively. In this study, we used cell-attached or whole-cell patch-clamp recordings to characterize the effects of trazodone at somatodendritic 5-HT1A receptors (5-HT1AARs) and α1-adrenoceptors of serotonergic neurons in rodent dorsal raphe slices. To reveal the effects of trazodone at α1-adrenoceptors, the baseline firing of 5-HT neurons was facilitated by applying the selective α1-adrenoceptor agonist phenylephrine at various concentrations. In the absence of phenylephrine, trazodone (1-10 µM) concentration-dependently silenced neurons through activation of 5-HT1AARs. The effect was fully antagonized by the selective 5-HT1A receptor antagonist Way-100635. With 5-HT1A receptors blocked by Way-100635, trazodone (1-10 µM) concentration-dependently inhibited neuron firing facilitated by 1 µM phenylephrine. Parallel rightward shift of dose-response curves for trazodone recorded in higher phenylephrine concentrations (10-100 µM) indicated competitive antagonism at α1-adrenoceptors. Both effects of trazodone were also observed in slices from Tph2-/- mice that lack synthesis of brain serotonin, showing that the activation of 5-HT1AARs was not mediated by endogenous serotonin. In whole-cell recordings, trazodone activated 5-HT1AAR-coupled G protein-activated inwardly-rectifying (GIRK) channel conductance with weak partial agonist efficacy (~35%) compared to that of the full agonist 5-CT. Collectively our data show that trazodone, at concentrations relevant to its clinical effects, exerts weak partial agonism at 5-HT1AARs and disfacilitation of firing through α1-adrenoceptor antagonism. These two actions converge in inhibiting dorsal raphe serotonergic neuron activity, albeit with varying contribution depending on the intensity of α1-adrenoceptor stimulation.


Assuntos
Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Antidepressivos/farmacologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Neurônios Serotoninérgicos/efeitos dos fármacos , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Trazodona/farmacologia , Animais , Núcleo Dorsal da Rafe/citologia , Núcleo Dorsal da Rafe/metabolismo , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Fenilefrina/farmacologia , Piperazinas/farmacologia , Piridinas/farmacologia , Ratos , Receptor 5-HT1A de Serotonina/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Neurônios Serotoninérgicos/metabolismo , Triptofano Hidroxilase/genética
2.
Front Neurosci ; 13: 245, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31068767

RESUMO

Brain serotonin (5-hydroxytryptamine, 5-HT) system dysfunction is implicated in exaggerated fear responses triggering various anxiety-, stress-, and trauma-related disorders. However, the underlying mechanisms are not well understood. Here, we investigated the impact of constitutively inactivated 5-HT synthesis on context-dependent fear learning and extinction using tryptophan hydroxylase 2 (Tph2) knockout mice. Fear conditioning and context-dependent fear memory extinction paradigms were combined with c-Fos imaging and electrophysiological recordings in the dorsal hippocampus (dHip). Tph2 mutant mice, completely devoid of 5-HT synthesis in brain, displayed accelerated fear memory formation and increased locomotor responses to foot shock. Furthermore, recall of context-dependent fear memory was increased. The behavioral responses were associated with increased c-Fos expression in the dHip and resistance to foot shock-induced impairment of hippocampal long-term potentiation (LTP). In conclusion, increased context-dependent fear memory resulting from brain 5-HT deficiency involves dysfunction of the hippocampal circuitry controlling contextual representation of fear-related behavioral responses.

3.
Hippocampus ; 28(3): 217-225, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29266595

RESUMO

The hippocampus is functionally differentiated along its longitudinal axis, with the dorsal and ventral segments preferentially involved in cognitive and emotional processing, respectively. Serotonergic modulation of hippocampal function has been extensively studied, but its relation to the dorsoventral axis has remained largely unknown. To examine the modulation of hippocampal output along the dorsoventral axis by endogenous serotonin (5-HT) we compared the effect of the 5-HT/noradrenaline (NA)-releaser, 3,4-methylenedioxymethamphetamine (MDMA), in transversal slices encompassing the entire rat hippocampus. Co-release of 5-HT and NA by MDMA resulted in a gradient of effects on evoked population spikes in the CA1 area along the dorsoventral axis of the hippocampus. Selective 5-HT release decreased population spike amplitude in slices from dorsal hippocampus, whereas an increase was produced in the ventral hippocampus, indicating differential modulation of CA1 impulse flow along the dorsoventral axis by endogenous 5-HT. Selective NA release increased population spike amplitude with no gradient indicating facilitatory effect of endogenous NA along the entire dorsoventral axis. Blockade of 5-HT1A receptors prevented the inhibitory component of MDMA action and the emergence of the gradient, indicating that activation of 5-HT1A receptors is required for differential modulation of CA1 impulse flow by endogenous 5-HT. These findings suggest that a dorsoventral shift in CA1 output level may represent an integral component of 5-HT action on hippocampal information processing. Given the preferential role of ventral hippocampus in emotional and anxiety-related behavior, it can be proposed that serotonin tone encodes the emotional salience of the signal processed by hippocampus.


Assuntos
Hipocampo/metabolismo , Serotonina/metabolismo , Inibidores da Captação Adrenérgica/farmacologia , Animais , Hipocampo/efeitos dos fármacos , Masculino , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Norepinefrina/metabolismo , Piperazinas/farmacologia , Piridinas/farmacologia , Ratos Wistar , Receptor 5-HT1A de Serotonina/metabolismo , Serotoninérgicos/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Técnicas de Cultura de Tecidos
4.
Eur Neuropsychopharmacol ; 27(12): 1258-1267, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29126768

RESUMO

Firing activity of serotonergic neurons is under regulatory control by somatodendritic 5-HT1A autoreceptors (5-HT1AARs). Enhanced 5-HT1AAR functioning may cause decreased serotonergic signaling in brain and has thereby been implicated in the etiology of mood and anxiety disorders. Tryptophan hydroxylase-2 knockout (Tph2-/-) mice exhibit sensitization of 5-HT1A agonist-induced inhibition of serotonergic neuron firing and thus represents a unique animal model of enhanced 5-HT1AAR functioning. To elucidate the mechanisms underlying 5-HT1AAR supersensitivity in Tph2-/- mice, we characterized the activation of G protein-coupled inwardly-rectifying potassium (GIRK) conductance by the 5-HT1A receptor agonist 5-carboxamidotryptamine using whole-cell recordings from serotonergic neurons in dorsal raphe nucleus. Tph2-/- mice exhibited a mean twofold leftward shift of the agonist concentration-response curve (p < 0.001) whereas the maximal response, proportional to the 5-HT1AAR number, was not different (p = 0.42) compared to Tph2+/- and Tph2+/+ littermates. No differences were found in the basal inwardly-rectifying potassium conductance, determined in the absence of agonist, (p = 0.80) nor in total GIRK conductance activated by intracellular application of GTP-γ-S (p = 0.69). These findings indicate increased functional coupling of 5-HT1AARs to GIRK channels in Tph2-/- mice without a concomitant increase in 5-HT1AARs and/or GIRK channel density. In addition, no changes were found in α1-adrenergic facilitation of firing (p = 0.72) indicating lack of adaptive changes Tph2-/- mice. 5-HT1AAR supersensitivity may represents a previously unrecognized cause of serotonergic system hypofunction and associated disorders and provides a possible explanation for conflicting results on the correlation between 5-HT1AAR density and depression in clinical imaging studies.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Regulação da Expressão Gênica/genética , Receptor 5-HT1A de Serotonina/metabolismo , Neurônios Serotoninérgicos/fisiologia , Triptofano Hidroxilase/deficiência , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Animais Recém-Nascidos , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Núcleo Dorsal da Rafe/citologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Feminino , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Ácidos Fosfínicos/farmacologia , Propanolaminas/farmacologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Serotonina/análogos & derivados , Serotonina/farmacologia , Serotoninérgicos/farmacologia , Fatores de Tempo , Triptofano Hidroxilase/genética
5.
Front Cell Neurosci ; 10: 195, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27536220

RESUMO

Tonic spiking of serotonergic neurons establishes serotonin levels in the brain. Since the first observations, slow regular spiking has been considered as a defining feature of serotonergic neurons. Recent studies, however, have revealed the heterogeneity of serotonergic neurons at multiple levels, comprising their electrophysiological properties, suggesting the existence of functionally distinct cellular subpopulations. In order to examine in an unbiased manner whether serotonergic neurons of the dorsal raphe nucleus (DRN) are heterogeneous, we used a non-invasive loose-seal cell-attached method to record α1 adrenergic receptor-stimulated spiking of a large sample of neurons in brain slices obtained from transgenic mice lines that express fluorescent marker proteins under the control of serotonergic system-specific Tph2 and Pet-1 promoters. We found wide homogeneous distribution of firing rates, well fitted by a single Gaussian function (r (2) = 0.93) and independent of anatomical location (P = 0.45), suggesting that in terms of intrinsic firing properties, serotonergic neurons in the DRN represent a single cellular population. Characterization of the population in terms of spiking regularity was hindered by its dependence on the firing rate. For instance, the coefficient of variation of the interspike intervals (ISI), a common measure of spiking irregularity, is of limited usefulness since it correlates negatively with the firing rate (r = -0.33, P < 0.0001). Nevertheless, the majority of neurons exhibited regular, pacemaker-like activity, with coefficient of variance of the ISI lower than 0.5 in ~97% of cases. Unexpectedly, a small percentage of neurons (~1%) exhibited a particular spiking pattern, characterized by low frequency (~0.02-0.1 Hz) oscillations in the firing rate. Transitions between regular and oscillatory firing were observed, suggesting that the oscillatory firing is an alternative firing pattern of serotonergic neurons.

6.
PLoS One ; 10(10): e0140369, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26460748

RESUMO

G protein-activated inwardly rectifying potassium (GIRK) channels in 5-HT neurons are assumed to be principal effectors of 5-hydroxytryptamine 1A (5-HT1A) autoreceptors, but their pharmacology, subunit composition and the role in regulation of 5-HT neuron activity have not been fully elucidated. We sought for a pharmacological tool for assessing the functional role of GIRK channels in 5-HT neurons by characterizing the effects of drugs known to block GIRK channels in the submicromolar range of concentrations. Whole-cell voltage-clamp recording in brainstem slices were used to determine concentration-response relationships for the selected GIRK channel blockers on 5-HT1A autoreceptor-activated inwardly rectifying K+ conductance in rat dorsal raphe 5-HT neurons. 5-HT1A autoreceptor-activated GIRK conductance was completely blocked by the nonselective inwardly rectifying potassium channels blocker Ba2+ (EC50 = 9.4 µM, full block with 100 µM) and by SCH23390 (EC50 = 1.95 µM, full block with 30 µM). GIRK-specific blocker tertiapin-Q blocked 5-HT1A autoreceptor-activated GIRK conductance with high potency (EC50 = 33.6 nM), but incompletely, i.e. ~16% of total conductance resulted to be tertiapin-Q-resistant. U73343 and SCH28080, reported to block GIRK channels with submicromolar EC50s, were essentially ineffective in 5-HT neurons. Our data show that inwardly rectifying K+ channels coupled to 5-HT1A autoreceptors display pharmacological properties generally expected for neuronal GIRK channels, but different from GIRK1-GIRK2 heteromers, the predominant form of brain GIRK channels. Distinct pharmacological properties of GIRK channels in 5-HT neurons should be explored for the development of new therapeutic agents for mood disorders.


Assuntos
Núcleo Dorsal da Rafe/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Neurônios Serotoninérgicos/metabolismo , Animais , Bário/farmacologia , Venenos de Abelha/farmacologia , Benzazepinas/farmacologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Condutividade Elétrica , Estrenos/farmacologia , Masculino , Maleatos/farmacologia , Pirrolidinonas/farmacologia , Ratos Wistar , Neurônios Serotoninérgicos/efeitos dos fármacos
7.
Eur Neuropsychopharmacol ; 25(11): 2022-35, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26409296

RESUMO

Considerable evidence links dysfunction of serotonin (5-hydroxytryptamine, 5-HT) transmission to neurodevelopmental and psychiatric disorders characterized by compromised "social" cognition and emotion regulation. It is well established that the brain 5-HT system is under autoregulatory control by its principal transmitter 5-HT via its effects on activity and expression of 5-HT system-related proteins. To examine whether 5-HT itself also has a crucial role in the acquisition and maintenance of characteristic rhythmic firing of 5-HT neurons, we compared their intrinsic electrophysiological properties in mice lacking brain 5-HT, i.e. tryptophan hydroxylase-2 null mice (Tph2(-/-)) and their littermates, Tph2(+/-) and Tph2(+/+), by using whole-cell patch-clamp recordings in a brainstem slice preparation and single unit recording in anesthetized animals. We report that the active properties of dorsal raphe nucleus (DRN) 5-HT neurons in vivo (firing rate magnitude and variability; the presence of spike doublets) and in vitro (firing in response to depolarizing current pulses; action potential shape) as well as the resting membrane potential remained essentially unchanged across Tph2 genotypes. However, there were subtle differences in subthreshold properties, most notably, an approximately 25% higher input conductance in Tph2(-/-) mice compared with Tph2(+/-) and Tph2(+/+) littermates (p<0.0001). This difference may at least in part be a consequence of slightly bigger size of the DRN 5-HT neurons in Tph2(-/-) mice (approximately 10%, p<0.0001). Taken together, these findings show that 5-HT neurons acquire and maintain their signature firing properties independently of the presence of their principal neurotransmitter 5-HT, displaying an unexpected functional resilience to complete brain 5-HT deficiency.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Dorsal da Rafe/fisiologia , Neurônios Serotoninérgicos/fisiologia , Triptofano Hidroxilase/deficiência , Potenciais de Ação/efeitos dos fármacos , Animais , Núcleo Dorsal da Rafe/citologia , Núcleo Dorsal da Rafe/efeitos dos fármacos , Capacitância Elétrica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos Knockout , Microscopia de Fluorescência , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Canais de Potássio/metabolismo , Neurônios Serotoninérgicos/citologia , Neurônios Serotoninérgicos/efeitos dos fármacos , Técnicas de Cultura de Tecidos , Triptofano Hidroxilase/genética
8.
J Gen Physiol ; 145(3): 225-51, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25712017

RESUMO

The firing activity of serotonergic neurons in raphe nuclei is regulated by negative feedback exerted by extracellular serotonin (5-HT)o acting through somatodendritic 5-HT1A autoreceptors. The steady-state [5-HT]o, sensed by 5-HT1A autoreceptors, is determined by the balance between the rates of 5-HT release and reuptake. Although it is well established that reuptake of 5-HTo is mediated by 5-HT transporters (SERT), the release mechanism has remained unclear. It is also unclear how selective 5-HT reuptake inhibitor (SSRI) antidepressants increase the [5-HT]o in raphe nuclei and suppress serotonergic neuron activity, thereby potentially diminishing their own therapeutic effect. Using an electrophysiological approach in a slice preparation, we show that, in the dorsal raphe nucleus (DRN), continuous nonexocytotic 5-HT release is responsible for suppression of phenylephrine-facilitated serotonergic neuron firing under basal conditions as well as for autoinhibition induced by SSRI application. By using 5-HT1A autoreceptor-activated G protein-gated inwardly rectifying potassium channels of patched serotonergic neurons as 5-HTo sensors, we show substantial nonexocytotic 5-HT release under conditions of abolished firing activity, Ca(2+) influx, vesicular monoamine transporter 2-mediated vesicular accumulation of 5-HT, and SERT-mediated 5-HT transport. Our results reveal a cytosolic origin of 5-HTo in the DRN and suggest that 5-HTo may be supplied by simple diffusion across the plasma membrane, primarily from the dense network of neurites of serotonergic neurons surrounding the cell bodies. These findings indicate that the serotonergic system does not function as a sum of independently acting neurons but as a highly interdependent neuronal network, characterized by a shared neurotransmitter pool and the regulation of firing activity by an interneuronal, yet activity-independent, nonexocytotic mechanism.


Assuntos
Potenciais de Ação , Exocitose , Neurônios Serotoninérgicos/metabolismo , Serotonina/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Masculino , Ratos , Ratos Wistar , Neurônios Serotoninérgicos/fisiologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
9.
J Neural Transm (Vienna) ; 122(2): 177-85, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24872079

RESUMO

Encoding of episodic memory requires long-term potentiation (LTP) of neurotransmission at excitatory synapses of the hippocampal circuitry. Previous data obtained with the application of exogenous 5-hydroxytryptamine (5-HT) in hippocampal slices indicate that 5-HT blocks LTP, which contrasts with the facilitatory effect of selective serotonin reuptake inhibitors (SSRIs) on learning and memory observed in vivo. Here, we investigated the effects of endogenous 5-HT, released from terminals by the monoamine releaser 3,4-methylenedioxymethamphetamine (MDMA), on LTP of field EPSPs induced by theta-burst stimulation and recorded at CA3/CA1 synapses of rat hippocampal slices. LTP was greater in the presence of MDMA (10 µM; 45.76 ± 15.75%; n = 28) than in controls (31.26 ± 11.03; n = 21; p < 0.01). This facilitatory effect on LTP persisted when the entry of MDMA in noradrenergic terminals was prevented by the selective noradrenaline reuptake inhibitor nisoxetine (44.90 ± 14.07%; n = 27 vs. 34.49 ± 12.94%; n = 20 in controls; p < 0.05). In both conditions, the facilitation of LTP was abolished by the SSRI citalopram that prevented the entry of MDMA in 5-HT terminals and the subsequent 5-HT release. These data show that, unlike exogenous 5-HT application, release of endogenous 5-HT does not impair cellular mechanisms responsible for induction of LTP, indicating that 5-HT is not detrimental to learning and memory. Moreover, facilitation of LTP by endogenous 5-HT may underlie the in vivo positive effects of augmented 5-HT tone on cognitive performance.


Assuntos
Região CA1 Hipocampal/citologia , Região CA3 Hipocampal/citologia , Potenciação de Longa Duração/fisiologia , Células Piramidais/fisiologia , Serotonina/metabolismo , Sinapses/fisiologia , Análise de Variância , Animais , Biofísica , Estimulação Elétrica , Fluoxetina/análogos & derivados , Fluoxetina/farmacologia , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Serotoninérgicos/farmacologia , Sinapses/efeitos dos fármacos
10.
Front Pharmacol ; 4: 97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935583

RESUMO

Firing activity of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) is controlled by inhibitory somatodendritic 5-HT1A autoreceptors. This autoinhibitory mechanism is implicated in the etiology of disorders of emotion regulation, such as anxiety disorders and depression, as well as in the mechanism of antidepressant action. Here, we investigated how persistent alterations in brain 5-HT availability affect autoinhibition in two genetically modified mouse models lacking critical mediators of serotonergic transmission: 5-HT transporter knockout (Sert-/-) and tryptophan hydroxylase-2 knockout (Tph2-/-) mice. The degree of autoinhibition was assessed by loose-seal cell-attached recording in DRN slices. First, application of the 5-HT1A-selective agonist R(+)-8-hydroxy-2-(di-n-propylamino)tetralin showed mild sensitization and marked desensitization of 5-HT1A receptors in Tph2-/- mice and Sert-/- mice, respectively. While 5-HT neurons from Tph2-/- mice did not display autoinhibition in response to L-tryptophan, autoinhibition of these neurons was unaltered in Sert-/- mice despite marked desensitization of their 5-HT1A autoreceptors. When the Tph2-dependent 5-HT synthesis step was bypassed by application of 5-hydroxy-L-tryptophan (5-HTP), neurons from both Tph2-/- and Sert-/- mice decreased their firing rates at significantly lower concentrations of 5-HTP compared to wildtype controls. Our findings demonstrate that, as opposed to the prevalent view, sensitivity of somatodendritic 5-HT1A receptors does not predict the magnitude of 5-HT neuron autoinhibition. Changes in 5-HT1A receptor sensitivity may rather be seen as an adaptive mechanism to keep autoinhibition functioning in response to extremely altered levels of extracellular 5-HT resulting from targeted inactivation of mediators of serotonergic signaling.

11.
J Neurosci ; 33(20): 8678-88, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23678112

RESUMO

Numerous studies link decreased serotonin metabolites with increased impulsive and aggressive traits. However, although pharmacological depletion of serotonin is associated with increased aggression, interventions aimed at directly decreasing serotonin neuron activity have supported the opposite association. Furthermore, it is not clear if altered serotonin activity during development may contribute to some of the observed associations. Here, we used two pharmacogenetic approaches in transgenic mice to selectively and reversibly reduce the firing of serotonin neurons in behaving animals. Conditional overexpression of the serotonin 1A receptor (Htr1a) in serotonin neurons showed that a chronic reduction in serotonin neuron firing was associated with heightened aggression. Overexpression of Htr1a in adulthood, but not during development, was sufficient to increase aggression. Rapid suppression of serotonin neuron firing by agonist treatment of mice expressing Htr1a exclusively in serotonin neurons also led to increased aggression. These data confirm a role of serotonin activity in setting thresholds for aggressive behavior and support a direct association between low levels of serotonin homeostasis and increased aggression.


Assuntos
Potenciais de Ação/fisiologia , Agressão/fisiologia , Inibição Neural/fisiologia , Núcleos da Rafe/citologia , Neurônios Serotoninérgicos/fisiologia , Serotonina/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralina/toxicidade , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Ansiedade/tratamento farmacológico , Ansiedade/etiologia , Ansiedade/genética , Ansiedade/patologia , Autorradiografia , Distribuição de Qui-Quadrado , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Hipotermia/induzido quimicamente , Isótopos de Iodo/farmacocinética , Locomoção/efeitos dos fármacos , Locomoção/genética , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Técnicas de Patch-Clamp , Piperazinas/farmacocinética , Piridinas/farmacocinética , Receptor 5-HT1A de Serotonina/genética , Receptor 5-HT1A de Serotonina/metabolismo , Serotoninérgicos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Agonistas do Receptor de Serotonina/toxicidade , Fatores de Tempo , Triptofano Hidroxilase/genética
12.
PLoS One ; 7(9): e45072, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028768

RESUMO

BACKGROUND: Serotonergic system participates in a wide range of physiological processes and behaviors, but its role is generally considered as modulatory and noncrucial, especially concerning life-sustaining functions. We recently created a transgenic mouse line in which a functional deficit in serotonin homeostasis due to excessive serotonin autoinhibition was produced by inducing serotonin 1A receptor (Htr1a) overexpression selectively in serotonergic neurons (Htr1a raphe-overexpressing or Htr1a(RO) mice). Htr1a(RO) mice exhibit episodes of autonomic dysregulation, cardiovascular crises and death, resembling those of sudden infant death syndrome (SIDS) and revealing a life-supporting role of serotonergic system in autonomic control. Since midbrain serotonergic neurons are chemosensitive and are implicated in arousal we hypothesized that their chemosensitivity might be impaired in Htr1a(RO) mice. PRINCIPAL FINDINGS: Loose-seal cell-attached recordings in brainstem slices revealed that serotonergic neurons in dorsal raphe nucleus of Htr1a(RO) mice have dramatically reduced responses to hypercapnic challenge as compared with control littermates. In control mice, application of 9% CO(2) produced an increase in firing rate of serotonergic neurons (0.260 ± 0.041 Hz, n=20, p=0.0001) and application of 3% CO(2) decreased their firing rate (-0.142 ± 0.025 Hz, n=17, p=0.0008). In contrast, in Htr1a(RO) mice, firing rate of serotonergic neurons was not significantly changed by 9% CO(2) (0.021 ± 0.034 Hz, n=16, p=0.49) and by 3% CO(2) (0.012 ± 0.046 Hz, n=12, p=0.97). CONCLUSIONS: Our findings support the hypothesis that chemosensitivity of midbrain serotonergic neurons provides a physiological mechanism for arousal responses to life-threatening episodes of hypercapnia and that functional impairment, such as excessive autoinhibition, of midbrain serotonergic neuron responses to hypercapnia may contribute to sudden death.


Assuntos
Células Quimiorreceptoras/metabolismo , Núcleos da Rafe/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Neurônios Serotoninérgicos/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Dióxido de Carbono/farmacologia , Células Quimiorreceptoras/efeitos dos fármacos , Feminino , Hipercapnia/metabolismo , Hipercapnia/fisiopatologia , Masculino , Camundongos , Norepinefrina/metabolismo , Núcleos da Rafe/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Neurônios Serotoninérgicos/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
13.
PLoS One ; 7(8): e43157, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912815

RESUMO

Brain serotonin (5-HT) is implicated in a wide range of functions from basic physiological mechanisms to complex behaviors, including neuropsychiatric conditions, as well as in developmental processes. Increasing evidence links 5-HT signaling alterations during development to emotional dysregulation and psychopathology in adult age. To further analyze the importance of brain 5-HT in somatic and brain development and function, and more specifically differentiation and specification of the serotonergic system itself, we generated a mouse model with brain-specific 5-HT deficiency resulting from a genetically driven constitutive inactivation of neuronal tryptophan hydroxylase-2 (Tph2). Tph2 inactivation (Tph2-/-) resulted in brain 5-HT deficiency leading to growth retardation and persistent leanness, whereas a sex- and age-dependent increase in body weight was observed in Tph2+/- mice. The conserved expression pattern of the 5-HT neuron-specific markers (except Tph2 and 5-HT) demonstrates that brain 5-HT synthesis is not a prerequisite for the proliferation, differentiation and survival of raphe neurons subjected to the developmental program of serotonergic specification. Furthermore, although these neurons are unable to synthesize 5-HT from the precursor tryptophan, they still display electrophysiological properties characteristic of 5-HT neurons. Moreover, 5-HT deficiency induces an up-regulation of 5-HT(1A) and 5-HT(1B) receptors across brain regions as well as a reduction of norepinephrine concentrations accompanied by a reduced number of noradrenergic neurons. Together, our results characterize developmental, neurochemical, neurobiological and electrophysiological consequences of brain-specific 5-HT deficiency, reveal a dual dose-dependent role of 5-HT in body weight regulation and show that differentiation of serotonergic neuron phenotype is independent from endogenous 5-HT synthesis.


Assuntos
Encéfalo/metabolismo , Inativação Gênica/fisiologia , Crescimento e Desenvolvimento/fisiologia , Núcleos da Rafe/metabolismo , Serotonina/deficiência , Triptofano Hidroxilase/genética , Fatores Etários , Animais , Autorradiografia , Peso Corporal , Crescimento e Desenvolvimento/genética , Técnicas Histológicas , Ácido Hidroxi-Indolacético/metabolismo , Camundongos , Norepinefrina/metabolismo , Receptores de Serotonina/metabolismo , Fatores Sexuais
14.
Eur Neuropsychopharmacol ; 21(1): 80-91, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20727723

RESUMO

In rats and primates, (±)3,4-Methylenedioxymethamphetamine (MDMA, ecstasy) produces both long-lasting damage to serotonergic axons and memory impairment. Our objective was to determine effects of neurotoxic dose of MDMA on long-term potentiation (LTP) in hippocampal area CA1 in Dark-Agouti (DA) rats. One week after neurotoxic MDMA treatment in vivo (12.5mg/kg i.p., once a week, per three weeks), serotonergic deficit was evident in hippocampal slices as 56.3% reduction in 5-HT content (p=0.04) and as 68.4% reduction in the effect of endogenous 5-HT release on synaptic neurotransmission (p<0.01). In hippocampal slices from the same animals, LTP was on average 46% greater than that observed in sham-treated controls (42.9 ± 3.5%; n=12 vs. 29.2 ± 3.2%; n=12; p<0.01). Non-neurotoxic dose of MDMA (12.5 mg/kg, i.p., one time) did not change LTP one week after the treatment, suggesting correlation between serotonergic deficit and enhanced synaptic plasticity. We conclude that MDMA-induced impairment of learning and memory is not a consequence of hippocampal LTP inhibition.


Assuntos
Alucinógenos/administração & dosagem , Hipocampo/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/administração & dosagem , Animais , Potenciais Evocados/efeitos dos fármacos , Alucinógenos/farmacologia , Hipocampo/fisiologia , Aprendizagem/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Norepinefrina/análise , Ratos , Serotonina/análise , Fatores de Tempo
15.
Science ; 321(5885): 130-3, 2008 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-18599790

RESUMO

Sudden infant death syndrome is the leading cause of death in the postneonatal period in developed countries. Postmortem studies show alterations in serotonin neurons in the brainstem of such infants. However, the mechanism by which altered serotonin homeostasis might cause sudden death is unknown. We investigated the consequences of altering the autoinhibitory capacity of serotonin neurons with the reversible overexpression of serotonin 1A autoreceptors in transgenic mice. Overexpressing mice exhibited sporadic bradycardia and hypothermia that occurred during a limited developmental period and frequently progressed to death. Moreover, overexpressing mice failed to activate autonomic target organs in response to environmental challenges. These findings show that excessive serotonin autoinhibition is a risk factor for catastrophic autonomic dysregulation and provide a mechanism for a role of altered serotonin homeostasis in sudden infant death syndrome.


Assuntos
Sistema Nervoso Autônomo/fisiologia , Inibição Neural , Neurônios/fisiologia , Serotonina/metabolismo , Morte Súbita do Lactente/etiologia , Animais , Autorreceptores/metabolismo , Temperatura Corporal , Doxiciclina/farmacologia , Eletrocardiografia , Retroalimentação Fisiológica , Frequência Cardíaca , Homeostase , Humanos , Lactente , Camundongos , Camundongos Transgênicos , Atividade Motora , Neurônios/metabolismo , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Piridinas/administração & dosagem , Piridinas/farmacologia , Núcleos da Rafe/citologia , Núcleos da Rafe/metabolismo , Receptor 5-HT1A de Serotonina/genética , Receptor 5-HT1A de Serotonina/metabolismo , Antagonistas da Serotonina/administração & dosagem , Antagonistas da Serotonina/farmacologia , Sistema Nervoso Simpático/fisiologia , Transmissão Sináptica , Triptofano/metabolismo , Triptofano/farmacologia
16.
Neuropsychopharmacology ; 33(6): 1464-75, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17653110

RESUMO

It is well documented that N-methyl-3,4-methylenedioxyamphetamine (MDMA, ecstasy) releases brain serotonin (5-HT; 5-hydroxytryptamine), noradrenaline (NE; norepinephrine), and dopamine, but the consequent effect on brain functioning remains elusive. In this study, we characterized the effects of MDMA on electrically evoked responses in the ventral CA1 region of a rat hippocampal slice preparation. Superfusion with MDMA (10 microM, 30 min) increased the population spike amplitude (PSA) by 48.9+/-31.2% and decreased population spike latency (PSL) by 103+/-139 mus (both: mean+/-SD, n=123; p<0.0001, Wilcoxon test), without affecting field excitatory postsynaptic potential (fEPSP). This effect persisted for at least 1 h after MDMA washout; we have called this EPSP-spike potentiation (ESP) by MDMA, ESP MDMA. Antagonism of GABAergic transmission did not prevent ESP MDMA, suggesting that an increase in excitability of pyramidal cells underlies this MDMA action. Block of serotonin transporter (SERT) with citalopram or 5-HT depletion with (+/-)-p-chlorophenylalanine pretreatment partially inhibited the ESP MDMA. Block of both SERT and NE transporter prevented ESP MDMA, indicating its dependence on release of both 5-HT and NE. ESP MDMA is produced by simultaneous activation of 5-HT4 and beta1 receptors, with a predominant role of 5-HT4 receptors. Block of both 5-HT4 and beta1 receptors revealed an inhibitory component of the MDMA action mediated by 5-HT1A receptor. The concentration range of MDMA which produced ESP MDMA (1-30 microM) corresponds to that commonly reached in human plasma following the ingestion of psychoactive MDMA doses, suggesting that release of both 5-HT and NE, and consequent ESP MDMA may underlie some of the psychoactive effects of MDMA in humans.


Assuntos
Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Norepinefrina/metabolismo , Serotoninérgicos/farmacologia , Serotonina/metabolismo , Adrenérgicos/farmacologia , Animais , Relação Dose-Resposta a Droga , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos da radiação , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Receptores Adrenérgicos beta/fisiologia , Receptores 5-HT4 de Serotonina/fisiologia , Estatísticas não Paramétricas
17.
Eur J Neurosci ; 24(3): 719-31, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16930402

RESUMO

Recent studies implicated involvement of the 5-hydroxytryptamine4 (5-HT4) receptor in cognitive and emotional processes. The highest 5-HT4 receptor densities in the brain are found in the limbic system including the hippocampus. Here we used the selective 5-HT4 receptor full agonist, N-pentyl-N'-aminoguanidine carbazimidamide (SDZ-216454) to characterize effects of 5-HT4 receptor activation in whole-cell and field recordings in the area CA1 in hippocampal slices prepared from 3 to 4- and 6 to 9-week-old rats, respectively. Extracellular recordings showed that transient 5-HT4 receptor activation by 10-20 min application of SDZ-216454 induces field excitatory postsynaptic potential (fEPSP)-population spike potentiation (ESP(5-HT4)), which persisted for as long as we held the recordings (> 2 h). ESP(5-HT4) displayed characteristics different from EPSP-spike potentiation that accompanies long-term potentiation; it developed without an associated increase in synaptic transmission, was independent on afferent input, activity of postsynaptic neurons and N-methyl-d-aspartate receptor activation; and was expressed in the presence of GABA receptor antagonists. ESP(5-HT4) was also induced by transient application of the natural neurotransmitter, 5-HT. The increase in the evoked population spike (PS) induced by SDZ-216454 was not prevented by blockers of hyperpolarization-activated cation current (Ih), Cs+ and ZD-7288, but was mimicked and occluded by 150 microm Ba2+. Whole-cell voltage-clamp recordings from pyramidal neurons demonstrated that SDZ-216454 application increases membrane resistance with a concomitant decrease in a Ba2+-sensitive inwardly rectifying K+ current and the Ba2+-insensitive K+ current underlying slow afterhyperpolarization (I(sAHP)). We conclude that 5-HT4 receptor activation may cause a long-lasting excitability increase in CA1 pyramidal neurons by inhibition of a Ba2+-sensitive inwardly rectifying K+ current.


Assuntos
Potenciais de Ação/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Células Piramidais/metabolismo , Receptores 5-HT4 de Serotonina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Bário/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Serotonina/metabolismo , Serotonina/farmacologia , Agonistas do Receptor 5-HT4 de Serotonina , Agonistas do Receptor de Serotonina/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
18.
Neuroreport ; 16(12): 1351-5, 2005 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-16056138

RESUMO

5-Hydroxytryptamine neurons in the dorsal raphe nucleus are under autoinhibitory control by endogenous 5-hydroxytryptamine. Tonic activation of 5-hydroxytryptamine 1A autoreceptors was demonstrated in awake animals, but was inconsistently observed in anaesthetized animals and slice preparations, leading to questioning of its physiological significance. We re-evaluated autoinhibition in single-unit recordings from deeply seated 5-hydroxytryptamine neurons in slices in which endogenous 5-hydroxytryptamine bioavailability was restored by supplementing its precursor L-tryptophan. In these conditions, the application of the neutral 5-hydroxytryptamine 1A receptor antagonist WAY-100635 markedly increased 5-hydroxytryptamine neuron firing. Responses to WAY-100635 in single experiments ranged from a lack of effect to a several-fold increase in firing rate, suggesting that 5-hydroxytryptamine neurons in the dorsal raphe nucleus represent a heterogeneous population regarding their susceptibility to autoinhibition by endogenous 5-hydroxytryptamine.


Assuntos
Potenciais de Ação/fisiologia , Inibição Neural/fisiologia , Neurônios/fisiologia , Núcleos da Rafe/citologia , Serotonina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Agonistas alfa-Adrenérgicos/farmacologia , Análise de Variância , Animais , Relação Dose-Resposta a Droga , Ácido Hidroxi-Indolacético/metabolismo , Técnicas In Vitro , Masculino , Inibição Neural/efeitos dos fármacos , Fenilefrina/farmacologia , Piperazinas/farmacologia , Piridinas/farmacologia , Ratos , Ratos Wistar , Antagonistas da Serotonina/farmacologia , Fatores de Tempo , Triptofano/farmacologia
19.
J Pharmacol Exp Ther ; 315(1): 109-17, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15951403

RESUMO

The pharmacological properties of cyclohexanecarboxylic acid, {2-[4-(2-bromo-5-methoxybenzyl)piperazin-1-yl]ethyl}-(2-trifluoromethoxyphenyl)amide (Rec 27/0224), and cyclohexanecarboxylic acid, (2-methoxy-phenyl)-{2-[4-(2-methoxyphenyl)-piperazin-1-yl]ethyl}amide (Rec 27/0074), were characterized using radioligand displacement and guanosine 5'-O-(3-[35S]thiotriphosphate) ([35S]GTPgammaS) binding assays, as well as electrophysiological experiments, in rat hippocampal and dorsal raphe nucleus (DRN) slices. Both compounds showed a high affinity (Ki, approximately 1 nM) and selectivity (>70-fold) at human 5-hydroxytryptamine (5-HT)1A receptors versus other 5-HT receptors. In [35S]GTPgammaS binding assays on HeLa cells stably expressing human 5-HT1A receptors, Rec 27/0224 and Rec 27/0074 inhibited basal [35S]GTPgammaS binding by 44.8 +/- 1.7% (pEC50 = 8.58) and 25 +/- 2.5% (pEC50 = 8.86), respectively. In intracellularly recorded CA1 pyramidal cells, 5-HT1A (hetero)receptor-mediated hyperpolarization, elicited by 100 nM 5-carboxamidoytryptamine (5-CT), was partially antagonized by Rec 27/0224 (approximately 50%; IC50 = 18.0 nM) and Rec 27/0074 (74%; IC50 = 0.8 nM). In extracellularly recorded DRN serotonergic neurons, Rec 27/0224 and Rec 27/0074 fully antagonized the inhibition of firing caused by the activation of 5-HT1A (auto)receptors by 30 nM 5-CT with an IC50 of 34.9 nM and 16.5 nM, respectively. The antagonism had a slow time course, reaching a steady state within 60 min. Both compounds also antagonized the citalopram-elicited, endogenous 5-HT-mediated inhibition of cell firing. In conclusion, Rec 27/0224 and Rec 27/0074 exhibited inverse agonism in [35S]GTPgammaS binding assays and differential antagonistic properties on 5-HT1A receptor-mediated responses in the hippocampus but not in the DRN. Whether this differential effect is causally related to inverse agonist activity is unclear. The qualitatively different nature of the antagonism in the hippocampus versus the DRN clearly distinguishes the compounds from neutral antagonists, such as N-{2-[4-(2-methoxyphenyl)-1-piperazinyl]ethyl}-N-2-pyridinylcyclo-hexanecarboxamide (WAY-100635).


Assuntos
Ácidos Cicloexanocarboxílicos/farmacologia , Hipocampo/efeitos dos fármacos , Piperazinas/farmacologia , Núcleos da Rafe/efeitos dos fármacos , Agonistas do Receptor 5-HT1 de Serotonina , Agonistas do Receptor de Serotonina/farmacologia , Animais , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HeLa , Hipocampo/fisiologia , Humanos , Masculino , Piridinas/farmacologia , Ensaio Radioligante , Núcleos da Rafe/fisiologia , Ratos , Ratos Wistar , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Receptor 5-HT1A de Serotonina/fisiologia
20.
Eur J Neurosci ; 18(6): 1559-71, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14511335

RESUMO

A multitude of different serotonin (5-HT) receptor types are expressed in the hippocampus, but the identity of receptors actually mediating the physiological response to endogenous 5-HT has not been determined. We combined pharmacologically induced release of 5-HT with patch-clamp recordings on disinhibited rat CA1 minislices to determine effects of endogenous 5-HT on the excitability of pyramidal neurons and synaptic transmission among them. We found that application of 5-HT releasers, 3,4-methylenedioxy-methamphetamine (MDMA) or p-methylthioamphetamine, at concentrations ranging from 2 to 50 microm, reduced the excitatory synaptic transmission between CA1 pyramidal neurons without altering their basal electrical properties. This effect of MDMA was blocked by the selective 5-HT1B antagonist GR 55562, was dependent on endogenous 5-HT content and was mediated by presynaptically located, pertussis-toxin sensitive mechanisms. We found no other MDMA effects in our preparation, which indicates that the release of endogenous 5-HT preferentially stimulates 5-HT1B receptors on CA1 pyramidal neurons. Therefore, 5-HT1B receptor activation may represent a predominant component of the physiological response to endogenous 5-HT in the CA1. The high sensitivity of the 5-HT1B receptor-mediated reduction of polysynaptic excitatory responses to the extracellular 5-HT level enabled us to study mechanisms of the 5-HT releasing action of MDMA. Block of the serotonin transporter (SERT) with citalopram slowed the time course and reduced overall 5-HT release by MDMA. Depletion of vesicular 5-HT, by inhibition of vesicular monoamine transporter type 2 with tetrabenazine prevented the release. Thus although the SERT reversal contributes, a direct vesicle-depleting action is essential for MDMA release of 5-HT.


Assuntos
Proteínas de Transporte/fisiologia , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana Transportadoras , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Proteínas do Tecido Nervoso , Células Piramidais/efeitos dos fármacos , Receptor 5-HT1B de Serotonina/metabolismo , Vesículas Secretórias/efeitos dos fármacos , Serotoninérgicos/farmacologia , Serotonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Antagonistas GABAérgicos/farmacologia , Hipocampo/citologia , Hipocampo/fisiologia , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Condução Nervosa/efeitos dos fármacos , Toxina Pertussis/farmacologia , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Vesículas Secretórias/metabolismo , Antagonistas da Serotonina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Serotonina , Transmissão Sináptica/fisiologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...