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








Base de dados
Intervalo de ano de publicação
1.
Neuropsychopharmacology ; 49(6): 1014-1023, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38368493

RESUMO

In the central nervous system, noradrenaline transmission controls the degree to which we are awake, alert, and attentive. Aberrant noradrenaline transmission is associated with pathological forms of hyper- and hypo-arousal that present in numerous neuropsychiatric disorders often associated with dysfunction in serotonin transmission. In vivo, noradrenaline regulates the release of serotonin because noradrenergic input drives the serotonin neurons to fire action potentials via activation of excitatory α1-adrenergic receptors (α1-AR). Despite the critical influence of noradrenaline on the activity of dorsal raphe serotonin neurons, the source of noradrenergic afferents has not been resolved and the presynaptic mechanisms that regulate noradrenaline-dependent synaptic transmission have not been described. Using an acute brain slice preparation from male and female mice and electrophysiological recordings from dorsal raphe serotonin neurons, we found that selective optogenetic activation of locus coeruleus terminals in the dorsal raphe was sufficient to produce an α1-AR-mediated excitatory postsynaptic current (α1-AR-EPSC). Activation of inhibitory α2-adrenergic receptors (α2-AR) with UK-14,304 eliminated the α1-AR-EPSC via presynaptic inhibition of noradrenaline release, likely via inhibition of voltage-gated calcium channels. In a subset of serotonin neurons, activation of postsynaptic α2-AR produced an outward current through activation of GIRK potassium conductance. Further, in vivo activation of α2-AR by systemic administration of clonidine reduced the expression of c-fos in the dorsal raphe serotonin neurons, indicating reduced neural activity. Thus, α2-AR are critical regulators of serotonin neuron excitability.


Assuntos
Núcleo Dorsal da Rafe , Locus Cerúleo , Receptores Adrenérgicos alfa 2 , Neurônios Serotoninérgicos , Transmissão Sináptica , Animais , Núcleo Dorsal da Rafe/efeitos dos fármacos , Núcleo Dorsal da Rafe/fisiologia , Núcleo Dorsal da Rafe/metabolismo , Masculino , Receptores Adrenérgicos alfa 2/metabolismo , Receptores Adrenérgicos alfa 2/fisiologia , Receptores Adrenérgicos alfa 2/efeitos dos fármacos , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/fisiologia , Feminino , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Camundongos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Optogenética , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Camundongos Endogâmicos C57BL , Norepinefrina/metabolismo , Camundongos Transgênicos
2.
Neuropsychopharmacology ; 49(5): 864-875, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37848733

RESUMO

Psychiatric and obstetric diseases are growing threats to public health and share high rates of co-morbidity. G protein-coupled receptor signaling (e.g., vasopressin, serotonin) may be a convergent psycho-obstetric risk mechanism. Regulator of G Protein Signaling 2 (RGS2) mutations increase risk for both the gestational disease preeclampsia and for depression. We previously found preeclampsia-like, anti-angiogenic obstetric phenotypes with reduced placental Rgs2 expression in mice. Here, we extend this to test whether conserved cerebrovascular and serotonergic mechanisms are also associated with risk for neurobiological phenotypes in the Rgs2 KO mouse. Rgs2 KO exhibited anxiety-, depression-, and hedonic-like behaviors. Cortical vascular density and vessel length decreased in Rgs2 KO; cortical and white matter thickness and cell densities were unchanged. In Rgs2 KO, serotonergic gene expression was sex-specifically changed (e.g., cortical Htr2a, Maoa increased in females but all serotonin targets unchanged or decreased in males); redox-related expression increased in paraventricular nucleus and aorta; and angiogenic gene expression was changed in male but not female cortex. Whole-cell recordings from dorsal raphe serotonin neurons revealed altered 5-HT1A receptor-dependent inhibitory postsynaptic currents (5-HT1A-IPSCs) in female but not male KO neurons. Additionally, serotonin transporter blockade by the SSRI sertraline increased the amplitude and time-to-peak of 5-HT1A-IPSCs in KO neurons to a greater extent than in WT neurons in females only. These results demonstrate behavioral, cerebrovascular, and sertraline hypersensitivity phenotypes in Rgs2 KOs, some of which are sex-specific. Disruptions may be driven by vascular and cell stress mechanisms linking the shared pathogenesis of psychiatric and obstetric disease to reveal future targets.


Assuntos
Pré-Eclâmpsia , Serotonina , Humanos , Feminino , Masculino , Camundongos , Gravidez , Animais , Serotonina/metabolismo , Sertralina , Pré-Eclâmpsia/metabolismo , Placenta/metabolismo , Núcleo Dorsal da Rafe/metabolismo , Camundongos Knockout , Receptor 5-HT1A de Serotonina/metabolismo
3.
EMBO Rep ; 24(7): e56801, 2023 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-37154294

RESUMO

Ion channel function of native delta glutamate receptors (GluDR ) is incompletely understood. Previously, we and others have shown that activation of Gαq protein-coupled receptors (GqPCR) produces a slow inward current carried by GluD1R . GluD1R also carries a tonic cation current of unknown cause. Here, using voltage-clamp electrophysiological recordings from adult mouse brain slices containing the dorsal raphe nucleus, we find no role of ongoing G-protein-coupled receptor activity in generating or sustaining tonic GluD1R currents. Neither augmentation nor disruption of G protein activity affects tonic GluD1R currents, suggesting that ongoing G-protein-coupled receptor activity does not give rise to tonic GluD1R currents. Further, the tonic GluD1R current is unaffected by the addition of external glycine or D-serine, which influences GluD2R current at millimolar concentrations. Instead, GqPCR-stimulated and tonic GluD1R currents are regulated by physiological levels of external calcium. In current-clamp recordings, block of GluD1R channels hyperpolarizes the membrane by ~7 mV at subthreshold potentials, reducing excitability. Thus, GluD1R carries a G-protein-independent tonic current that contributes to subthreshold neuronal excitation in the dorsal raphe nucleus.


Assuntos
Canais Iônicos , Neurônios , Camundongos , Animais , Potenciais da Membrana/fisiologia , Neurônios/fisiologia , Encéfalo , Receptores Acoplados a Proteínas G , Glutamato Desidrogenase
4.
Biophys J ; 122(12): 2383-2395, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37177782

RESUMO

In this article, we review contemporary evidence that GluD receptors are functional ion channels whose depolarizing currents contribute to their biological functions, akin to all other members of the ionotropic glutamate receptor (iGluR) family.


Assuntos
Receptores Ionotrópicos de Glutamato
6.
J Neurosci ; 42(6): 968-979, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34921047

RESUMO

Activity of dorsal raphe neurons is controlled by noradrenaline afferents. In this brain region, noradrenaline activates Gαq-coupled α1-adrenergic receptors (α1-AR), causing action potential (AP) firing and serotonin release. In vitro, electrical stimulation elicits vesicular noradrenaline release and subsequent activation of α1-AR to produce an EPSC (α1-AR-EPSC). The duration of the α1-AR-EPSC (∼27 s) is much longer than that of most other synaptic currents, but the factors that govern the spatiotemporal dynamics of α1-AR are poorly understood. Using an acute brain slice preparation from adult male and female mice and electrophysiological recordings from dorsal raphe neurons, we found that the time course of the α1-AR-EPSC was slow, but highly consistent within individual serotonin neurons. The amount of noradrenaline released influenced the amplitude of the α1-AR-EPSC without altering the time constant of decay suggesting that once released, extracellular noradrenaline was cleared efficiently. Reuptake of noradrenaline via noradrenaline transporters was a primary means of terminating the α1-AR-EPSC, with little evidence for extrasynaptic diffusion of noradrenaline unless transporter-dependent reuptake was impaired. Taken together, the results demonstrate that despite slow intrinsic signaling kinetics, noradrenaline-dependent synaptic transmission in the dorsal raphe is temporally and spatially controlled and noradrenaline transporters are critical regulators of serotonin neuron excitability. Given the functionally distinct types of neurons intermingled in the dorsal raphe nucleus and the unique roles of these neural circuits in physiological responses, transporters may preserve independence of each synapse to encode a long-lasting but discrete signal.SIGNIFICANCE STATEMENT The dorsal raphe nucleus is the predominant source of serotonin in the brain and is controlled by another monoamine, noradrenaline. In this brain region, noradrenaline activates G-protein-coupled α1-adrenergic receptors (α1-AR) causing action potential (AP) firing and serotonin release. Despite high interest in pharmacotherapies to enhance serotonin signaling, the factors that govern noradrenaline α1-AR signaling have received little attention. Here, we show using mouse brain slices that the time course of α1-AR signaling is slow, persisting for tens of seconds. Despite slow intrinsic signaling kinetics, noradrenaline-dependent synaptic transmission in the dorsal raphe is controlled temporally and spatially by efficient noradrenaline transporter-dependent clearance of extracellular noradrenaline. Thus, noradrenaline transporters are critical regulators of serotonin neuron excitability.


Assuntos
Núcleo Dorsal da Rafe/fisiologia , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Neurônios Serotoninérgicos/fisiologia , Transmissão Sináptica/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34260402

RESUMO

Sinoatrial node myocytes (SAMs) act as cardiac pacemaker cells by firing spontaneous action potentials (APs) that initiate each heartbeat. The funny current (If) is critical for the generation of these spontaneous APs; however, its precise role during the pacemaking cycle remains unresolved. Here, we used the AP-clamp technique to quantify If during the cardiac cycle in mouse SAMs. We found that If is persistently active throughout the sinoatrial AP, with surprisingly little voltage-dependent gating. As a consequence, it carries both inward and outward current around its reversal potential of -30 mV. Despite operating at only 2 to 5% of its maximal conductance, If carries a substantial fraction of both depolarizing and repolarizing net charge movement during the firing cycle. We also show that ß-adrenergic receptor stimulation increases the percentage of net depolarizing charge moved by If, consistent with a contribution of If to the fight-or-flight increase in heart rate. These properties were confirmed by heterologously expressed HCN4 channels and by mathematical models of If Modeling further suggested that the slow rates of activation and deactivation of the HCN4 isoform underlie the persistent activity of If during the sinoatrial AP. These results establish a new conceptual framework for the role of If in pacemaking, in which it operates at a very small fraction of maximal activation but nevertheless drives membrane potential oscillations in SAMs by providing substantial driving force in both inward and outward directions.


Assuntos
Relógios Biológicos/fisiologia , Fenômenos Eletrofisiológicos , Miócitos Cardíacos/fisiologia , Nó Sinoatrial/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Relógios Biológicos/efeitos dos fármacos , Simulação por Computador , Diástole/efeitos dos fármacos , Diástole/fisiologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Células HEK293 , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Ivabradina/farmacologia , Moduladores de Transporte de Membrana/farmacologia , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Nó Sinoatrial/efeitos dos fármacos
8.
Elife ; 102021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33822716

RESUMO

Ultrapotent chemogenetics, including the chloride-permeable inhibitory PSAM4-GlyR receptor, were recently proposed as a powerful strategy to selectively control neuronal activity in awake, behaving animals. We aimed to validate the inhibitory function of PSAM4-GlyR in dopamine D1 receptor-expressing medium spiny neurons (D1-MSNs) in the ventral striatum. Activation of PSAM4-GlyR with the uPSEM792 ligand enhanced rather than suppressed the activity of D1-MSNs in vivo as indicated by increased c-fos expression in D1-MSNs and in vitro as indicated by cell-attached recordings from D1-MSNs in mouse brain slices. Whole-cell recordings showed that activation of PSAM4-GlyR depolarized D1-MSNs, attenuated GABAergic inhibition, and shifted the reversal potential of PSAM4-GlyR current to more depolarized potentials, perpetuating the depolarizing effect of receptor activation. These data show that 'inhibitory' PSAM4-GlyR chemogenetics may activate certain cell types and highlight the pitfalls of utilizing chloride conductances to inhibit neurons.


Assuntos
Cloretos/metabolismo , Neurônios/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Glicina/metabolismo , Estriado Ventral/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Feminino , Masculino , Camundongos , Técnicas de Patch-Clamp , Receptores de GABA-A/metabolismo
9.
Elife ; 92020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32234214

RESUMO

The dorsal raphe nucleus is the predominant source of central serotonin, where neuronal activity regulates complex emotional behaviors. Action potential firing of serotonin dorsal raphe neurons is driven via α1-adrenergic receptors (α1-AR) activation. Despite this crucial role, the ion channels responsible for α1-AR-mediated depolarization are unknown. Here, we show in mouse brain slices that α1-AR-mediated excitatory synaptic transmission is mediated by the ionotropic glutamate receptor homolog cation channel, delta glutamate receptor 1 (GluD1). GluD1R-channels are constitutively active under basal conditions carrying tonic inward current and synaptic activation of α1-ARs augments tonic GluD1R-channel current. Further, loss of dorsal raphe GluD1R-channels produces an anxiogenic phenotype. Thus, GluD1R-channels are responsible for α1-AR-dependent induction of persistent pacemaker-type firing of dorsal raphe neurons and regulate dorsal raphe-related behavior. Given the widespread distribution of these channels, ion channel function of GluD1R as a regulator of neuronal excitability is proposed to be widespread in the nervous system.


Serotonin is a chemical that allows cells to communicate in the nervous system of many animals. It is also particularly important in the treatment of mental health disorders: a large number of antidepressants work by preventing nerve cells from clearing away serotonin, therefore increasing the overall level of the molecule in the brain. Yet, exactly how serotonin is released remains unclear. When a serotonin-producing cell is activated, a series of biochemical processes lead to the creation of an electric current that, ultimately, is required for the cell to release serotonin. This mechanism starts when the α1-adrenergic receptor, a protein at the surface of the cell, detects noradrenaline molecules. However, on its own, the α1-adrenergic receptor is unable to create an electric current: this requires ion channels, another type of protein which can let charged particles in and out of the cell. Here, Gantz et al. set out to determine the identity of the ion channel that allows noradrenaline signals to generate electrical activity in cells which can release serotonin. Electrical and chemical manipulation of mouse brain slices revealed that an ion channel called delta-glutamate 1 was active in serotonin-producing cells exposed to noradrenaline. In fact, applying toxins that specifically blocked the activity of this channel also prevented the cells from responding electrically to noradrenaline. Further experiments used mice whose serotonin-producing cells were genetically modified to turn off delta-glutamate 1. In turn, these animals showed anxiety-like behaviors, which could be consistent with a drop in serotonin levels. This is in line with previous human studies showing that patients with depression and other mental health conditions have mutations in the gene for delta-glutamate 1. Taken together, these results give an insight into the electrical activity of serotonin-producing cells. Further work is now required to examine how changes in the gene that codes for delta-glutamate 1 ultimately affect the release of serotonin. This could potentially help to understand if certain individuals may not be able to properly produce this chemical. As many antidepressants work by retaining serotonin that is already present in the brain, this knowledge could ultimately help patients who do not currently respond to treatment.


Assuntos
Potenciais de Ação/fisiologia , Núcleo Dorsal da Rafe/fisiologia , Neurônios/fisiologia , Receptores de Glutamato/metabolismo , Serotonina/metabolismo , Animais , Camundongos , Receptores Adrenérgicos alfa 1/metabolismo
10.
Proc Natl Acad Sci U S A ; 117(15): 8611-8615, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32229573

RESUMO

Electrical or optogenetic stimulation of lateral hypothalamic (LH) GABA neurons induces rapid vigorous eating in sated animals. The dopamine system has been implicated in the regulation of feeding. Previous work has suggested that a subset of LH GABA neurons projects to the ventral tegmental area (VTA) and targets GABA neurons, inhibiting them and thereby disinhibiting dopaminergic activity and release. Furthermore, stimulation-induced eating is attenuated by dopamine lesions or receptor antagonists. Here we explored the involvement of dopamine in LH stimulation-induced eating. LH stimulation caused sated mice to pick up pellets of standard chow with latencies that varied based on stimulation intensity; once food was picked up, animals ate for the remainder of the 60-s stimulation period. However, lesion of VTA GABA neurons failed to disrupt this effect. Moreover, direct stimulation of VTA or substantia nigra dopamine cell bodies failed to induce food approach or eating. Looking further, we found that some LH GABA fibers pass through the VTA to more caudal sites, where they synapse onto neurons near the locus coeruleus (LC). Similar eating was induced by stimulation of LH GABA terminals or GABA cell bodies in this peri-LC region. Lesion of peri-LC GABA neurons blocked LH stimulation-induced eating, establishing them as a critical downstream circuit element for LH neurons. Surprisingly, lesions did not alter body weight, suggesting that this system is not involved in the hunger or satiety mechanisms that govern normal feeding. Thus, we present a characterization of brain circuitry that may promote overeating and contribute to obesity.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Neurônios GABAérgicos/metabolismo , Região Hipotalâmica Lateral/fisiologia , Área Tegmentar Ventral/fisiologia , Animais , Comportamento Animal , Dopamina/metabolismo , Neurônios Dopaminérgicos/citologia , Feminino , Neurônios GABAérgicos/citologia , Região Hipotalâmica Lateral/citologia , Masculino , Camundongos , Vias Neurais , Receptores de GABA-A/metabolismo , Recompensa , Área Tegmentar Ventral/citologia , Ácido gama-Aminobutírico/metabolismo
11.
Curr Opin Neurobiol ; 54: 66-72, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30237117

RESUMO

Cocaine exposure induces persistent changes in synaptic transmission and intrinsic properties of ventral tegmental area (VTA) dopamine neurons. Despite significant progress in understanding cocaine-induced plasticity, an effective treatment of cocaine addiction is lacking. Chronic cocaine potentiates excitatory and alters inhibitory transmission to dopamine neurons, induces dopamine neuron hyperexcitability, and reduces dopamine release in projection areas. Understanding how intrinsic and synaptic plasticity interact to control dopamine neuron firing and dopamine release could prove useful in the development of new therapeutics. In this review, we examine recent literature discussing cocaine-induced plasticity in the VTA and highlight potential therapeutic interventions.


Assuntos
Cocaína/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Área Tegmentar Ventral/citologia , Animais , Transtornos Relacionados ao Uso de Cocaína/patologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Humanos , Neurotransmissores/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
12.
Annu Rev Physiol ; 80: 219-241, 2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28938084

RESUMO

In recent years, the population of neurons in the ventral tegmental area (VTA) and substantia nigra (SN) has been examined at multiple levels. The results indicate that the projections, neurochemistry, and receptor and ion channel expression in this cell population vary widely. This review centers on the intrinsic properties and synaptic regulation that control the activity of dopamine neurons. Although all dopamine neurons fire action potentials in a pacemaker pattern in the absence of synaptic input, the intrinsic properties that underlie this activity differ considerably. Likewise, the transition into a burst/pause pattern results from combinations of intrinsic ion conductances, inhibitory and excitatory synaptic inputs that differ among this cell population. Finally, synaptic plasticity is a key regulator of the rate and pattern of activity in different groups of dopamine neurons. Through these fundamental properties, the activity of dopamine neurons is regulated and underlies the wide-ranging functions that have been attributed to dopamine.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Substância Negra/metabolismo , Área Tegmentar Ventral/metabolismo , Potenciais de Ação/fisiologia , Animais , Humanos , Plasticidade Neuronal/fisiologia , Substância Negra/citologia , Área Tegmentar Ventral/citologia
13.
Neuron ; 93(6): 1375-1387.e2, 2017 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-28262417

RESUMO

The major endocannabinoid in the mammalian brain is the bioactive lipid 2-arachidonoylglycerol (2-AG). The best-known effects of 2-AG are mediated by G-protein-coupled cannabinoid receptors. In principle, 2-AG could modify neuronal excitability by acting directly on ion channels, but such mechanisms are poorly understood. Using a preparation of dissociated mouse midbrain dopamine neurons to isolate effects on intrinsic excitability, we found that 100 nM 2-AG accelerated pacemaking and steepened the frequency-current relationship for burst-like firing. In voltage-clamp experiments, 2-AG reduced A-type potassium current (IA) through a cannabinoid receptor-independent mechanism mimicked by arachidonic acid, which has no activity on cannabinoid receptors. Activation of orexin, neurotensin, and metabotropic glutamate Gq/11-linked receptors mimicked the effects of exogenous 2-AG and their actions were prevented by inhibiting the 2-AG-synthesizing enzyme diacylglycerol lipase α. The results show that 2-AG and related lipid signaling molecules can directly tune neuronal excitability in a cell-autonomous manner by modulating IA.


Assuntos
Potenciais de Ação/fisiologia , Ácidos Araquidônicos/fisiologia , Neurônios Dopaminérgicos/fisiologia , Endocanabinoides/fisiologia , Glicerídeos/fisiologia , Potenciais da Membrana/fisiologia , Mesencéfalo/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Ácido Araquidônico/farmacologia , Ácidos Araquidônicos/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Endocanabinoides/farmacologia , Feminino , Glicerídeos/farmacologia , Lipase Lipoproteica/antagonistas & inibidores , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Receptores de Orexina/agonistas , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Neurotensina/agonistas
14.
Elife ; 42015 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-26308580

RESUMO

D2 autoreceptors regulate dopamine release throughout the brain. Two isoforms of the D2 receptor, D2S and D2L, are expressed in midbrain dopamine neurons. Differential roles of these isoforms as autoreceptors are poorly understood. By virally expressing the isoforms in dopamine neurons of D2 receptor knockout mice, this study assessed the calcium-dependence and drug-induced plasticity of D2S and D2L receptor-dependent G protein-coupled inwardly rectifying potassium (GIRK) currents. The results reveal that D2S, but not D2L receptors, exhibited calcium-dependent desensitization similar to that exhibited by endogenous autoreceptors. Two pathways of calcium signaling that regulated D2 autoreceptor-dependent GIRK signaling were identified, which distinctly affected desensitization and the magnitude of D2S and D2L receptor-dependent GIRK currents. Previous in vivo cocaine exposure removed calcium-dependent D2 autoreceptor desensitization in wild type, but not D2S-only mice. Thus, expression of D2S as the exclusive autoreceptor was insufficient for cocaine-induced plasticity, implying a functional role for the co-expression of D2S and D2L autoreceptors.


Assuntos
Autorreceptores/metabolismo , Cálcio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Receptores de Dopamina D2/metabolismo , Transdução de Sinais , Animais , Cocaína/metabolismo , Camundongos Knockout , Isoformas de Proteínas/metabolismo
15.
Cell Rep ; 12(6): 944-54, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26235617

RESUMO

Imbalance between the dopamine and serotonin (5-HT) neurotransmitter systems has been implicated in the comorbidity of Parkinson's disease (PD) and psychiatric disorders. L-DOPA, the leading treatment of PD, facilitates the production and release of dopamine. This study assessed the action of L-DOPA on monoamine synaptic transmission in mouse brain slices. Application of L-DOPA augmented the D2-receptor-mediated inhibitory postsynaptic current (IPSC) in dopamine neurons of the substantia nigra. This augmentation was largely due to dopamine release from 5-HT terminals. Selective optogenetic stimulation of 5-HT terminals evoked dopamine release, producing D2-receptor-mediated IPSCs following treatment with L-DOPA. In the dorsal raphe, L-DOPA produced a long-lasting depression of the 5-HT1A-receptor-mediated IPSC in 5-HT neurons. When D2 receptors were expressed in the dorsal raphe, application of L-DOPA resulted in a D2-receptor-mediated IPSC. Thus, treatment with L-DOPA caused ectopic dopamine release from 5-HT terminals and a loss of 5-HT-mediated synaptic transmission.


Assuntos
Levodopa/farmacologia , Serotonina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , Eletrofisiologia , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
Proc Natl Acad Sci U S A ; 111(44): E4779-88, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25331903

RESUMO

Despite the critical role of the presynaptic dopamine (DA) transporter (DAT, SLC6A3) in DA clearance and psychostimulant responses, evidence that DAT dysfunction supports risk for mental illness is indirect. Recently, we identified a rare, nonsynonymous Slc6a3 variant that produces the DAT substitution Ala559Val in two male siblings who share a diagnosis of attention-deficit hyperactivity disorder (ADHD), with other studies identifying the variant in subjects with bipolar disorder (BPD) and autism spectrum disorder (ASD). Previously, using transfected cell studies, we observed that although DAT Val559 displays normal total and surface DAT protein levels, and normal DA recognition and uptake, the variant transporter exhibits anomalous DA efflux (ADE) and lacks capacity for amphetamine (AMPH)-stimulated DA release. To pursue the significance of these findings in vivo, we engineered DAT Val559 knock-in mice, and here we demonstrate in this model the presence of elevated extracellular DA levels, altered somatodendritic and presynaptic D2 DA receptor (D2R) function, a blunted ability of DA terminals to support depolarization and AMPH-evoked DA release, and disruptions in basal and psychostimulant-evoked locomotor behavior. Together, our studies demonstrate an in vivo functional impact of the DAT Val559 variant, providing support for the ability of DAT dysfunction to impact risk for mental illness.


Assuntos
Anfetamina/farmacologia , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Transtornos Mentais/metabolismo , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Animais , Dopamina/genética , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Neurônios Dopaminérgicos/patologia , Feminino , Humanos , Masculino , Transtornos Mentais/genética , Transtornos Mentais/patologia , Camundongos , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo
17.
Neuron ; 78(5): 807-12, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23764286

RESUMO

G protein-coupled receptors (GPCRs) affect many physiological processes by modulating both intrinsic membrane conductances and synaptic transmission. This study describes spontaneous miniature inhibitory postsynaptic currents mediated by vesicular dopamine release acting locally on metabotropic D2 receptors leading to the activation of a G protein-coupled inwardly rectifying potassium conductance. Thus, individual exocytotic events result in spontaneous GPCR-mediated transmission, similar to synaptic activation of classical ligand-gated ion channels.


Assuntos
Potenciais Pós-Sinápticos Inibidores/fisiologia , Neurônios/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Análise de Variância , Animais , Quelantes/farmacologia , Colforsina/farmacologia , Dopamina/metabolismo , Dopaminérgicos/farmacologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Feminino , Antagonistas GABAérgicos/farmacologia , Humanos , Técnicas In Vitro , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/genética , Levodopa/farmacologia , Masculino , Mesencéfalo/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Compostos Organofosforados/farmacologia , Técnicas de Patch-Clamp , Receptores de Dopamina D2/genética , Serotoninérgicos/farmacologia , Tirosina 3-Mono-Oxigenase/genética
18.
J Neurosci ; 31(35): 12629-37, 2011 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-21880923

RESUMO

Mutations in the methyl-CpG-binding protein 2 (MeCP2) result in Rett syndrome (RTT), an X-linked disorder that disrupts neurodevelopment. Girls with RTT exhibit motor deficits similar to those in Parkinson's disease, suggesting defects in the nigrostriatal pathway. This study examined age-dependent changes in dopamine neurons of the substantia nigra (SN) from wild-type, presymptomatic, and symptomatic Mecp2(+/-) mice. Mecp2(+) neurons in the SN in Mecp2(+/-) mice were indistinguishable in morphology, resting conductance, and dopamine current density from neurons in wild-type mice. However, the capacitance, total dendritic length, and resting conductance of Mecp2(-) neurons were less than those of Mecp2(+) neurons as early as 4 weeks after birth, before overt symptoms. These differences were maintained throughout life. In symptomatic Mecp2(+/-) mice, the current induced by activation of D(2) dopamine autoreceptors was significantly less in Mecp2(-) neurons than in Mecp2(+) neurons, although D(2) receptor density was unaltered in Mecp2(+/-) mice. Electrochemical measurements revealed that significantly less dopamine was released after stimulation of striatum in adult Mecp2(+/-) mice compared to wild type. The decrease in size and function of Mecp2(-) neurons observed in adult Mecp2(+/-) mice was recapitulated in dopamine neurons from symptomatic Mecp2(-/y) males. These results show that mutation in Mecp2 results in cell-autonomous defects in the SN early in life and throughout adulthood. Ultimately, dysfunction in terminal dopamine release and D(2) autoreceptor-dependent currents in dopamine neurons from symptomatic females support the idea that decreased dopamine transmission due to heterogeneous Mecp2 expression contributes to the parkinsonian features of RTT in Mecp2(+/-) mice.


Assuntos
Corpo Estriado/fisiologia , Dopamina/metabolismo , Proteína 2 de Ligação a Metil-CpG/deficiência , Vias Neurais/fisiologia , Neurônios/fisiologia , Substância Negra/citologia , Fatores Etários , Análise de Variância , Animais , Benzamidas/farmacocinética , Biofísica , Antagonistas de Dopamina/farmacocinética , Estimulação Elétrica/métodos , Técnicas Eletroquímicas/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/genética , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp/métodos , Ligação Proteica/efeitos dos fármacos , Ensaio Radioligante , Fatores Sexuais , Trítio/farmacocinética
19.
J Neurosci ; 30(20): 6975-83, 2010 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-20484639

RESUMO

Midbrain dopamine neurons release dopamine from both axons and dendrites. The mechanism underlying release at these different sites has been proposed to differ. This study used electrochemical and electrophysiological methods to compare the time course and calcium dependence of somatodendritic dopamine release in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) to that of axonal dopamine release in the dorsal striatum. The amount of dopamine released in the striatum was approximately 20-fold greater than in cell body regions of the VTA or SNc. However, the calcium dependence and time to peak of the dopamine transients were similar. These results illustrate an unexpected overall similarity in the mechanisms of dopamine release in the striatum and cell body regions. To examine how diffusion regulates the time course of dopamine following release, dextran was added to the extracellular solution to slow diffusion. In the VTA, dextran slowed the rate of rise and fall of the extracellular dopamine transient as measured by fast-scan cyclic voltammetry yet did not alter the kinetics of the dopamine-dependent IPSC. Dextran failed to significantly alter the time course of the rise and fall of the dopamine transient in the striatum, suggesting a more influential role for reuptake in the striatum. The conclusion is that the time course of dopamine within the extracellular space of the VTA is dependent on both diffusion and reuptake, whereas the activation of D(2) receptors on dopamine neurons is primarily limited by reuptake.


Assuntos
Axônios/metabolismo , Dendritos/metabolismo , Dopamina/metabolismo , Neurônios/citologia , Análise de Variância , Animais , Biofísica/métodos , Cálcio/metabolismo , Dextranos/metabolismo , Dopamina/farmacologia , Estimulação Elétrica/métodos , Eletroquímica/métodos , Feminino , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Indóis/farmacologia , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Iontoforese/métodos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Técnicas de Patch-Clamp/métodos , Ácidos Fosfínicos/farmacologia , Propanolaminas/farmacologia , Serotonina/farmacologia , Substância Negra/citologia , Fatores de Tempo , Área Tegmentar Ventral/citologia
20.
Neuropsychopharmacology ; 34(8): 1926-35, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19279570

RESUMO

Dopamine neurons in the ventral midbrain contribute to learning and memory of natural and drug-related rewards. Corticotropin-releasing factor (CRF), a stress-related peptide, is thought to be involved in aspects of relapse following drug withdrawal, but the cellular actions are poorly understood. This study investigates the action of CRF on G-protein-linked inhibitory postsynaptic currents (IPSCs) mediated by GIRK (Kir3) channels in dopamine neurons. CRF enhanced the amplitude and slowed the kinetics of IPSCs following activation of D2-dopamine and GABA(B) receptors. This action was postsynaptic and dependent on the CRF(1) receptor. The enhancement induced by CRF was attenuated by repeated in vivo exposures to psychostimulants or restraint stress. The results indicate that CRF influences dopamine- and GABA-mediated inhibition in the midbrain, suggesting implications for the chronic actions of psychostimulants and stress on dopamine-mediated behaviors.


Assuntos
Hormônio Liberador da Corticotropina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Reforço Psicológico , Estresse Psicológico/fisiopatologia , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Estimulantes do Sistema Nervoso Central/farmacologia , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Potenciais Pós-Sinápticos Inibidores/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Neurônios/metabolismo , Receptores de Hormônio Liberador da Corticotropina/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/metabolismo , Receptores de GABA-B/efeitos dos fármacos , Receptores de GABA-B/metabolismo , Recidiva , Restrição Física/fisiologia , Restrição Física/psicologia , Estresse Psicológico/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Área Tegmentar Ventral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA