Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 355(2): 206-11, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26350161

RESUMEN

The corticotropin-releasing factor (CRF) and kappa-opioid receptor (KOR) systems are both implicated in stress-related behaviors and drug dependence. Although previous studies suggest that antagonism of each system blocks aspects of experimental models of drug dependence, the possible interaction between these systems at the neuronal level has not been completely examined. We used an in vitro brain slice preparation to investigate the interaction of these two peptide systems on inhibitory neurotransmission in the central nucleus of the amygdala (CeA). Application of exogenous CRF increased the mean frequency of GABAergic miniature inhibitory postsynaptic currents (mIPSC) by 20.2%, suggesting an increase in presynaptic GABA release. Although the pharmacological blockade of KORs by norBNI alone did not significantly affect mIPSC frequency, it significantly enhanced the effect of CRF (by 43.9%, P = 0.02). Similarly, the CRF effects in slices from KOR knockout (KO) mice (84.0% increase) were significantly greater than in wild-type (WT) mice (24.6%, P = 0.01), although there was no significant difference in baseline mIPSC frequency between slices from KOR KO and WT mice. The increase in CRF action in the presence of norBNI was abolished by a CRF-1 receptor antagonist but was unaffected by a CRF-2 receptor antagonist. We hypothesize that CRF facilitates the release of an endogenous ligand for KORs and that subsequent activation of KOR receptors modulates presynaptic effects of CRF in CeA. These results suggest that potential pharmacotherapies aimed at neurobehavioral and addictive disorders may need to involve both the KOR/dynorphin and the CRF systems in CeA.


Asunto(s)
Núcleo Amigdalino Central/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Receptores Opioides kappa/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Potenciales Postsinápticos Inhibidores , Ratones Endogámicos C57BL , Ratones Noqueados , Potenciales Postsinápticos Miniatura , Naltrexona/análogos & derivados , Naltrexona/farmacología , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/genética , Transmisión Sináptica
2.
Front Pharmacol ; 6: 49, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25852553

RESUMEN

Neuroinflammation is hypothesized to enhance alcohol consumption and contribute to the development of alcoholism. GABAergic transmission in the central amygdala (CeA) plays an important role in the transition to alcohol dependence. Therefore, we studied the effects of interleukin-1ß (IL-1ß), a proinflammatory cytokine mediating ethanol-induced neuroinflammation, and its interaction with ethanol on CeA GABAegic transmission in B6129SF2/J mice. We also assessed ethanol intake in B6129SF2/J mice. Intake with unlimited (24 h) ethanol access was 9.2-12.7 g/kg (3-15% ethanol), while limited (2 h) access produced an intake of 4.1 ± 0.5 g/kg (15% ethanol). In our electrophysiology experiments, we found that recombinant IL-1ß (50 and 100 ng/ml) significantly decreased the amplitude of evoked inhibitory postsynaptic potentials (eIPSPs), with no significant effects on paired-pulse facilitation (PPF). IL-1ß (50 ng/ml) had dual effects on spontaneous miniature inhibitory postsynaptic currents (mIPSCs): increasing mIPSC frequencies in most CeA neurons, but decreasing both mIPSC frequencies and amplitudes in a few cells. The IL-1ß receptor antagonist (IL-1ra; 100 ng/ml) also had dual effects on mIPSCs and prevented the actions of IL-1ß on mIPSC frequencies. These results suggest that IL-1ß can alter CeA GABAergic transmission at pre- and postsynaptic sites. Ethanol (44 mM) significantly increased eIPSP amplitudes, decreased PPFs, and increased mIPSC frequencies. IL-1ß did not alter ethanol's enhancement of the eIPSP amplitude, but, in IL-1ß-responsive neurons, the ethanol effects on mIPSC frequencies were lost. Overall, our data suggest that the IL-1 system is involved in basal GABAergic transmission and that IL-1ß interacts with the ethanol-induced facilitation of CeA GABAergic transmission.

3.
Artículo en Inglés | MEDLINE | ID: mdl-24926240

RESUMEN

The central amygdala (CeA) plays an important role in opioid addiction. Therefore, we examined the effects of naloxone-precipitated morphine withdrawal (WD) on GABAergic transmission in rat CeA neurons using whole-cell recordings with naloxone in the bath. The basal frequency of miniature inhibitory postsynaptic currents (mIPSCs) increased in CeA neurons from WD compared to placebo rats. Acute morphine (10 µ M) had mixed effects (≥20% change from baseline) on mIPSCs in placebo and WD rats. In most CeA neurons (64%) from placebo rats, morphine significantly decreased mIPSC frequency and amplitude. In 32% of placebo neurons, morphine significantly increased mIPSC amplitudes but had no effect on mIPSC frequency. In WD rats, acute morphine significantly increased mIPSC frequency but had no effect on mIPSC amplitude in 41% of CeA neurons. In 45% of cells, acute morphine significantly decreased mIPSC frequency and amplitude. Pre-treatment with the cyclic AMP inhibitor (R)-adenosine, cyclic 3',5'-(hydrogenphosphorothioate) triethylammonium (RP), prevented acute morphine-induced potentiation of mIPSCs. Pre-treatment of slices with the Gi/o G-protein subunit inhibitor pertussis toxin (PTX) did not prevent the acute morphine-induced enhancement or inhibition of mIPSCs. PTX and RP decreased basal mIPSC frequencies and amplitudes only in WD rats. The results suggest that inhibition of GABAergic transmission in the CeA by acute morphine is mediated by PTX-insensitive mechanisms, although PTX-sensitive mechanisms cannot be ruled out for non-morphine responsive cells; by contrast, potentiation of GABAergic transmission is mediated by activated cAMP signaling that also mediates the increased basal GABAergic transmission in WD rats. Our data indicate that during the acute phase of WD, the CeA opioid and GABAergic systems undergo neuroadaptative changes conditioned by a previous chronic morphine exposure and dependence.

4.
Brain Behav Immun ; 40: 191-202, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24675033

RESUMEN

Excessive ethanol drinking in rodent models may involve activation of the innate immune system, especially toll-like receptor 4 (TLR4) signaling pathways. We used intracellular recording of evoked GABAergic inhibitory postsynaptic potentials (eIPSPs) in central amygdala (CeA) neurons to examine the role of TLR4 activation by lipopolysaccharide (LPS) and deletion of its adapter protein CD14 in acute ethanol effects on the GABAergic system. Ethanol (44, 66 or 100mM) and LPS (25 and 50µg/ml) both augmented eIPSPs in CeA of wild type (WT) mice. Ethanol (44mM) decreased paired-pulse facilitation (PPF), suggesting a presynaptic mechanism of action. Acute LPS (25µg/ml) had no effect on PPF and significantly increased the mean miniature IPSC amplitude, indicating a postsynaptic mechanism of action. Acute LPS pre-treatment potentiated ethanol (44mM) effects on eIPSPs in WT mice and restored ethanol's augmenting effects on the eIPSP amplitude in CD14 knockout (CD14 KO) mice. Both the LPS and ethanol (44-66mM) augmentation of eIPSPs was diminished significantly in most CeA neurons of CD14 KO mice; however, ethanol at the highest concentration tested (100mM) still increased eIPSP amplitudes. By contrast, ethanol pre-treatment occluded LPS augmentation of eIPSPs in WT mice and had no significant effect in CD14 KO mice. Furthermore, (+)-naloxone, a TLR4-MD-2 complex inhibitor, blocked LPS effects on eIPSPs in WT mice and delayed the ethanol-induced potentiation of GABAergic transmission. In CeA neurons of CD14 KO mice, (+)-naloxone alone diminished eIPSPs, and subsequent co-application of 100mM ethanol restored the eIPSPs to baseline levels. In summary, our results indicate that TLR4 and CD14 signaling play an important role in the acute ethanol effects on GABAergic transmission in the CeA and support the idea that CD14 and TLR4 may be therapeutic targets for treatment of alcohol abuse.


Asunto(s)
Núcleo Amigdalino Central/efectos de los fármacos , Etanol/farmacología , Inmunidad Innata , Receptores de Lipopolisacáridos/metabolismo , Neuronas/efectos de los fármacos , Receptores de GABA-A/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Núcleo Amigdalino Central/inmunología , Núcleo Amigdalino Central/fisiología , Factores Inmunológicos/metabolismo , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/farmacología , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/inmunología , Neuronas/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/inmunología , Receptor Toll-Like 4/antagonistas & inhibidores
5.
J Pharmacol Exp Ther ; 346(1): 130-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23587526

RESUMEN

Human and animal studies indicate that κ-opioid receptors (KORs) are involved in ethanol drinking and dependence (Xuei et al., 2006; Walker and Koob, 2008; Walker et al., 2011). Using in vitro single-cell recording techniques in mouse brain slices, we examined the physiologic effects of KOR activation in the central amygdala (CeA) on GABAergic neurotransmission and its interaction with acute ethanol. A selective KOR agonist (U69593, 1 µM) diminished evoked GABAergic inhibitory postsynaptic currents (IPSCs) by 18% (n = 10), whereas blockade of KORs with a selective antagonist (nor-binaltorphimine, 1 µM) augmented the baseline evoked GABAergic IPSCs by 14% (P < 0.01; n = 34), suggesting that the KOR system contributes to tonic inhibition of GABAergic neurotransmission in the CeA. In addition, the enhancement by acute ethanol of GABAergic IPSC amplitudes was further augmented by pharmacologic blockade of KORs, from 14% (n = 36) to 27% (n = 26; P < 0.01), or by genetic deletion of KORs, from 14% in wild-type mice (n = 19) to 34% in KOR knockout mice (n = 13; P < 0.01). Subsequent experiments using tetrodotoxin to block activity-dependent neurotransmission suggest that KORs regulate GABA release at presynaptic sites. Our data support the idea that KORs modulate GABAergic synaptic responses and ethanol effects as one of multiple opioid system-dependent actions of ethanol in the CeA, possibly in a circuit-specific manner.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Terminales Presinápticos/efectos de los fármacos , Receptores Opioides kappa/metabolismo , Transmisión Sináptica/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Analgésicos/farmacología , Animales , Ansiolíticos/farmacología , Potenciales Evocados/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Masculino , Ratones , Ratones Noqueados , Antagonistas de Narcóticos/farmacología , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Inhibición Neural/efectos de los fármacos , Técnicas de Placa-Clamp , Terminales Presinápticos/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inhibidores , Receptores Opioides kappa/genética , Análisis de la Célula Individual , Bloqueadores de los Canales de Sodio/farmacología
6.
Cold Spring Harb Perspect Med ; 2(12): a012195, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23085848

RESUMEN

Alcohol dependence is a chronically relapsing disorder characterized by compulsive drug seeking and drug taking, loss of control in limiting intake, and the emergence of a withdrawal syndrome in the absence of the drug. Accumulating evidence suggests an important role for synaptic transmission in the central amygdala (CeA) in mediating alcohol-related behaviors and neuroadaptative mechanisms associated with alcohol dependence. Acute alcohol facilitates γ-aminobutyric acid-ergic (GABAergic) transmission in CeA via both pre- and postsynaptic mechanisms, and chronic alcohol increases baseline GABAergic transmission. Acute alcohol inhibits glutamatergic transmission via effects at N-methyl-d-aspartate (NMDA) and AMPA receptors in CeA, whereas chronic alcohol up-regulates N-methyl-d-aspartate receptor (NMDAR)-mediated transmission. Pro- (e.g., corticotropin-releasing factor [CRF]) and anti-stress (e.g., NPY, nociceptin) neuropeptides affect alcohol- and anxiety-related behaviors, and also alter the alcohol-induced effects on CeA neurotransmission. Alcohol dependence produces plasticity in these neuropeptide systems, reflecting a recruitment of those systems during the transition to alcohol dependence.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Ácido Glutámico/efectos de los fármacos , Neuropéptidos/efectos de los fármacos , Ácido gamma-Aminobutírico/efectos de los fármacos , Alcoholismo/fisiopatología , Hormona Liberadora de Corticotropina/fisiología , Humanos , Neuropéptido Y/fisiología , Péptidos Opioides/fisiología , Transmisión Sináptica/efectos de los fármacos , Nociceptina
7.
Front Neurosci ; 4: 207, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21258618

RESUMEN

The GABAergic system in the central amygdala (CeA) plays a major role in ethanol dependence and in the anxiogenic response to ethanol withdrawal. Previously, we found that both ethanol and corticotropin releasing factor (CRF) increase GABAergic transmission in mouse and rat CeA neurons, in part by enhancing the release of GABA via activation of presynaptic CRF1 receptors. CRF1 receptors are coupled to the enzyme adenylyl cyclase (AC), which produces the second messenger cyclic AMP. There are nine isoforms of AC, but we recently found that CRF1 receptors in the pituitary were coupled to the Type 7 AC (AC7). Therefore, using an in vitro electrophysiological approach in brain slices, here we have investigated a possible role of the AC7 signaling pathway in ethanol and CRF effects on CeA GABAergic synapses of genetically modified mice with diminished brain Adcy7 activity (HET) compared to their littermate male wild-type (WT) mice. We found no significant differences in basal membrane properties, mean baseline amplitude of evoked GABA(A) receptor-mediated inhibitory postsynaptic potentials (IPSPs), or paired-pulse facilitation (PPF) of GABA(A)-IPSPs between HET and WT mice. In CeA neurons of WT mice, ethanol superfusion significantly augmented (by 39%) GABAA-IPSPs and decreased PPF (by 25%), suggesting increased presynaptic GABA release. However, these effects were absent in HET mice. CRF superfusion also significantly augmented IPSPs (by 38%) and decreased PPF (by 23%) in WT CeA neurons, and still elicited a significant but smaller (by 13%) increase of IPSP amplitude, but no effect on PPF, in HET mice. These electrophysiological data suggest that AC7 plays an important role in ethanol and CRF modulation of presynaptic GABA release in CeA and thus may underlie ethanol-related behaviors such as anxiety and dependence.

8.
Neuropsychopharmacology ; 35(9): 1962-72, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20463657

RESUMEN

The central amygdala (CeA) has a major role in alcohol dependence and reinforcement, and behavioral and neurochemical evidence suggests a role for the endocannabinoid (eCB) system in ethanol binging and dependence. We used a slice preparation to investigate the physiological role of cannabinoids and their interaction with ethanol on inhibitory synaptic transmission in CeA. Superfusion of the cannabinoid receptor (CB1) agonist WIN55212-2 (WIN2) onto CeA neurons decreased evoked GABA(A) receptor-mediated inhibitory postsynaptic potentials (IPSPs) in a concentration-dependent manner, an effect prevented by the CB1 antagonists Rimonabant (SR141716, SR1) and AM251. SR1 or AM251 applied alone augmented IPSPs, revealing a tonic eCB activity that decreased inhibitory transmission in CeA. Paired-pulse analysis suggested a presynaptic CB1 mechanism. Intracellular BAPTA abolished the ability of AM251 to augment IPSPs, demonstrating the eCB-driven nature and postsynaptic origin of the tonic CB1-dependent control of GABA release. Superfusion of ethanol increased IPSPs and addition of WIN2 reversed the ethanol effect. Similarly, previous superfusion of WIN2 prevented subsequent ethanol effects on GABAergic transmission. The ethanol-induced augmentation of IPSPs was additive to CB1 blockade, ruling out a participation of CB1 in the action of acute ethanol. Our study points to an important role of CB1 in CeA in which the eCBs tonically regulate neuronal activity, and suggests a potent mechanism for modulating CeA tone during challenge with ethanol.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Moduladores de Receptores de Cannabinoides/fisiología , Depresores del Sistema Nervioso Central/farmacología , Endocannabinoides , Etanol/farmacología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Amígdala del Cerebelo/citología , Analgésicos/farmacología , Animales , Benzoxazinas/farmacología , Moduladores de Receptores de Cannabinoides/agonistas , Moduladores de Receptores de Cannabinoides/antagonistas & inhibidores , Moduladores de Receptores de Cannabinoides/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Técnicas In Vitro , Masculino , Morfolinas/farmacología , Naftalenos/farmacología , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Piperidinas/farmacología , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley , Rimonabant
9.
Biol Psychiatry ; 67(9): 831-9, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20060104

RESUMEN

BACKGROUND: Corticotropin-releasing factor (CRF) and gamma-aminobutyric acid (GABA)ergic systems in the central amygdala (CeA) are implicated in the high-anxiety, high-drinking profile associated with ethanol dependence. Ethanol augments CeA GABA release in ethanol-naive rats and mice. METHODS: Using naive and ethanol-dependent rats, we compared electrophysiologic effects and interactions of CRF and ethanol on CeA GABAergic transmission, and we measured GABA dialyzate in CeA after injection of CRF(1) antagonists and ethanol. We also compared mRNA expression in CeA for CRF and CRF(1) using real-time polymerase chain reaction. We assessed effects of chronic treatment with a CRF(1) antagonist on withdrawal-induced increases in alcohol consumption in dependent rats. RESULTS: CRF and ethanol augmented CeA GABAergic transmission in naive rats via increased GABA release. Three CRF1 receptor (CRF(1)) antagonists decreased basal GABAergic responses and abolished ethanol effects. Ethanol-dependent rats exhibited heightened sensitivity to CRF and CRF(1) antagonists on CeA GABA release. Intra-CeA CRF(1) antagonist administration reversed dependence-related elevations in GABA dialysate and blocked ethanol-induced increases in GABA dialyzate in both dependent and naive rats. Polymerase chain reaction studies indicate increased expression of CRF and CRF(1) in CeA of dependent rats. Chronic CRF(1) antagonist treatment blocked withdrawal-induced increases in alcohol drinking by dependent rats and tempered moderate increases in alcohol consumption by nondependent rats in intermittent testing. CONCLUSIONS: These combined findings suggest a key role for specific presynaptic CRF-GABA interactions in CeA in the development and maintenance of ethanol dependence.


Asunto(s)
Alcoholismo/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Hormona Liberadora de Corticotropina/farmacología , Ácido gamma-Aminobutírico/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Alcoholismo/sangre , Alcoholismo/patología , Amígdala del Cerebelo/patología , Animales , Peso Corporal/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Hormona Liberadora de Corticotropina/genética , Ciclofilinas/farmacología , Etanol/administración & dosificación , Antagonistas de Aminoácidos Excitadores/farmacología , Antagonistas del GABA/farmacología , Antagonistas de Hormonas/farmacología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Microdiálisis/métodos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp , Ácidos Fosfínicos/farmacología , Propanolaminas/farmacología , Pirimidinas/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Factores de Tiempo , Valina/análogos & derivados , Valina/farmacología
10.
Alcohol ; 43(7): 509-19, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19913194

RESUMEN

This article summarizes the proceedings of a symposium that was presented at a conference entitled "Alcoholism and Stress: A Framework for Future Treatment Strategies." The conference was held in Volterra, Italy on May 6-9, 2008 and this symposium was chaired by Jeff L. Weiner. The overall goal of this session was to review recent findings that may shed new light on the neurobiological mechanisms that underlie the complex relationships between stress, anxiety, and alcoholism. Dr. Danny Winder described a novel interaction between D1 receptor activation and the corticotrophin-releasing factor (CRF) system that leads to an increase in glutamatergic synaptic transmission in the bed nucleus of the stria terminalis. Dr. Marisa Roberto presented recent data describing how protein kinase C epsilon, ethanol, and CRF interact to alter GABAergic inhibition in the central nucleus of the amygdala. Dr. Jeff Weiner presented recent advances in our understanding of inhibitory circuitry within the basolateral amygdala (BLA) and how acute ethanol exposure enhances GABAergic inhibition in these pathways. Finally, Dr. Brian McCool discussed recent findings on complementary glutamatergic and GABAergic adaptations to chronic ethanol exposure and withdrawal in the BLA. Collectively, these investigators have identified novel mechanisms through which neurotransmitter and neuropeptide systems interact to modulate synaptic activity in stress and anxiety circuits. Their studies have also begun to describe how acute and chronic ethanol exposure influence excitatory and inhibitory synaptic communication in these pathways. These findings point toward a number of novel neurobiological targets that may prove useful for the development of more effective treatment strategies for alcohol use disorders.


Asunto(s)
Alcoholismo/etiología , Ansiedad/complicaciones , Estrés Psicológico/complicaciones , Alcoholismo/tratamiento farmacológico , Alcoholismo/fisiopatología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Ansiedad/fisiopatología , Hormona Liberadora de Corticotropina/fisiología , Etanol/farmacología , Humanos , Proteína Quinasa C-epsilon/fisiología , Receptores de GABA-B/fisiología , Estrés Psicológico/fisiopatología , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
11.
Proc Natl Acad Sci U S A ; 105(24): 8410-5, 2008 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-18541912

RESUMEN

In the central amygdala (CeA), ethanol acts via corticotrophin-releasing factor (CRF) type 1 receptors to enhance GABA release. Amygdala CRF mediates anxiety associated with stress and drug dependence, and it regulates ethanol intake. Because mutant mice that lack PKCepsilon exhibit reduced anxiety-like behavior and alcohol consumption, we investigated whether PKCepsilon lies downstream of CRF(1) receptors in the CeA. Compared with PKCepsilon(+/+) CeA neurons, PKCepsilon(-/-) neurons showed increased GABAergic tone due to enhanced GABA release. CRF and ethanol stimulated GABA release in the PKCepsilon(+/+) CeA, but not in the PKCepsilon(-/-) CeA. A PKCepsilon-specific inhibitor blocked both CRF- and ethanol-induced GABA release in the PKCepsilon(+/+) CeA, confirming findings in the PKCepsilon(-/-) CeA. These results identify a PKCepsilon signaling pathway in the CeA that is activated by CRF(1) receptor stimulation, mediates GABA release at nerve terminals, and regulates anxiety and alcohol consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Ansiedad/enzimología , Hormona Liberadora de Corticotropina/metabolismo , Etanol/farmacología , Proteína Quinasa C-epsilon/fisiología , Ácido gamma-Aminobutírico/metabolismo , Consumo de Bebidas Alcohólicas/genética , Amígdala del Cerebelo/enzimología , Animales , Ansiedad/genética , Hormona Liberadora de Corticotropina/farmacología , Masculino , Ratones , Ratones Mutantes , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Proteína Quinasa C-epsilon/genética , Receptores de Hormona Liberadora de Corticotropina/agonistas , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Transmisión Sináptica/efectos de los fármacos
12.
J Neurosci ; 28(22): 5762-71, 2008 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-18509038

RESUMEN

Gabapentin is a structural analog of GABA that has anticonvulsant properties. Despite the therapeutic efficacy of gabapentin, its molecular and cellular mechanisms of action are unclear. The GABAergic system in the central nucleus of the amygdala (CeA) plays an important role in regulating voluntary ethanol intake. Here, we investigated the effect of gabapentin on GABAergic transmission in CeA slices, on ethanol intake, and on an anxiety measure using animal models of ethanol dependence. Gabapentin increased the amplitudes of evoked GABA receptor-mediated IPSCs (GABA-IPSCs) in CeA neurons from nondependent rats, but decreased their amplitudes in CeA of ethanol-dependent rats. Gabapentin effects were blocked in the presence of a specific GABA(B) receptor antagonist. The sensitivity of the GABA-IPSCs to a GABA(B) receptor antagonist and an agonist was decreased after chronic ethanol, suggesting that ethanol-induced neuroadaptations of GABA(B) receptors associated with ethanol dependence may account for the differential effects of gabapentin after chronic ethanol. Systemic gabapentin reduced ethanol intake in dependent, but not in nondependent, rats and reversed the anxiogenic-like effects of ethanol abstinence using an acute dependence model. Gabapentin infused directly into the CeA also blocked dependence-induced elevation in operant ethanol responding. Collectively, these findings show that gabapentin reverses behavioral measures of ethanol dependence and, in turn, dependence reverses the effects of gabapentin on CeA neurons, and suggest that gabapentin represents a potential medication for treatment of alcoholism.


Asunto(s)
Alcoholismo , Aminas/farmacología , Amígdala del Cerebelo/patología , Conducta Animal/efectos de los fármacos , Ácidos Ciclohexanocarboxílicos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Neuronas/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología , Alcoholismo/tratamiento farmacológico , Alcoholismo/patología , Alcoholismo/fisiopatología , Animales , Depresores del Sistema Nervioso Central/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Etanol/administración & dosificación , Etanol/metabolismo , Antagonistas del GABA/farmacología , Gabapentina , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Técnicas de Placa-Clamp , Ácidos Fosfínicos/farmacología , Propanolaminas/farmacología , Ratas , Ratas Wistar , Autoadministración , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
13.
Neurochem Res ; 33(2): 285-91, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17973188

RESUMEN

The neuropeptide galanin and its three receptor subtypes (Gal R1-3) are highly expressed in the dorsal raphe nucleus (DRN), a region of the brain that contains a large population of serotonergic neurons. Galanin is co-expressed with serotonin in approximately 40% of the DRN neurons, and galanin and GALR2 expression are elevated by antidepressants like the SSRI fluoxetine, suggesting an interaction between serotonin and galanin. The present study examines the effect of galanin (Gal 1-29), a pan ligand for GalR (1-3) and the GalR2/GalR3-selective ligand, Gal 2-11, on the electrophysiological properties of DRN serotonergic neurons in a slice preparation. We recorded from cells in the DRN with electrophysiological characteristics consistent with those of serotonergic neurons that exhibit high input resistance, large after-hyperpolarizations and long spike duration as defined by Aghajanian and Vandermaelen. Both Gal 1-29 and Gal 2-11 decreased the amplitudes pharmacologically-isolated GABAergic inhibitory postsynaptic potentials (IPSPs) in these putative serotonergic neurons. Furthermore, based on paired pulse facilitation studies, we show that Gal 1-29 likely decreases GABA release through a presynaptic mechanism, whereas Gal 2-11 may act postsynaptically. These findings may enhance understanding of the cellular mechanisms underlying the effects of antidepressant treatments on galanin and galanin receptors in DRN.


Asunto(s)
Galanina/farmacología , Núcleos del Rafe/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Técnicas In Vitro , Masculino , Núcleos del Rafe/metabolismo , Núcleos del Rafe/fisiología , Ratas , Ratas Sprague-Dawley
14.
Proc Natl Acad Sci U S A ; 103(25): 9715-20, 2006 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-16788074

RESUMEN

Behavioral studies show that the GABAergic system in the central amygdala (CeA) nucleus has a complex role in the reinforcing effects effects of ethanol and the anxiogenic response to ethanol withdrawal. Opioid peptides and nociceptin/orphanin FQ (nociceptin) within the CeA are implicated also in regulating voluntary ethanol consumption and ethanol relapse. Recently, we reported that basal GABAergic transmission was increased in ethanol-dependent rats, and that acute ethanol increases GABA(A) receptor-mediated inhibitory postsynaptic currents (IPSCs) in CeA neurons from both naïve and ethanol-dependent rats to the same extent, suggesting lack of tolerance for the acute effect of ethanol. Here, we investigated the effect of nociceptin on IPSCs in CeA neurons and its interaction with ethanol effects on these GABA synapses. We found that nociceptin moderately decreased IPSC amplitudes, acting mostly presynaptically as it increased paired-pulse facilitation ratio of IPSCs and decreased miniature IPSC frequencies (but not amplitudes). Nociceptin also prevented the ethanol-induced augmentation of IPSCs in CeA of naïve rats. Interestingly, in CeA of ethanol-dependent rats, the nociceptin-induced inhibition of IPSCs was increased, indicating an enhanced sensitivity to nociceptin. Nociceptin also blocked the ethanol-induced augmentation of IPSCs in ethanol-dependent rats. Our data suggest that nociceptin has a role in regulating the GABAergic system and opposing the effect elicited by ethanol. Thus, nociceptin may represent a therapeutic target for alleviating alcohol dependence.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Etanol/farmacología , Péptidos Opioides/farmacología , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Sinapsis/metabolismo , Nociceptina
15.
Neuropsychopharmacology ; 31(5): 988-96, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16052244

RESUMEN

We recently reported that chronic ethanol treatment (CET) and early withdrawal (2-8 h) altered glutamatergic transmission at both pre- and postsynaptic sites in central nucleus of the amygdala (CeA). Acute ethanol (44 mM) inhibited the NMDA receptor (NMDAR)-mediated EPSCs (NMDA-EPSCs) more in CeA neurons from CET rats than from naïve rats and also decreased paired-pulse facilitation (PPF) of NMDA-EPSCs only in CET rats. To determine whether these CET effects persisted after prolonged withdrawal, we recorded intracellularly in rat CeA slices and measured mRNA and protein expression of CeA NMDAR subunits from CET rats and those withdrawn from ethanol for 1 or 2 weeks. At 1 week withdrawal, acute ethanol decreased evoked NMDA-EPSC amplitudes and NMDA currents induced by exogenous NMDA ( approximately 20%) equally to that in naïve rats, indicating that CET effects on postsynaptic mechanisms reversed 1 week after CET cessation. However, acute ethanol still decreased PPF of NMDA-EPSCs, indicating that the acute ethanol-induced increase in glutamate release in CeA seen in CET rats was still present at this time. CET also significantly increased mRNA levels of NR1 and NR2B NMDAR subunits compared to control rats. At 1 week withdrawal, mRNA levels for NR1 and NR2B subunits were significantly decreased. These changes reversed at 2 weeks withdrawal. In Western blots, a significant increase in protein for all three subunits occurred in CeA from CET rats, but not after 1 and 2 weeks of withdrawal. These data indicate that CET induces reversible neuroadaptations in synaptic function, gene expression, and protein composition of NMDAR at CeA synapses.


Asunto(s)
Trastornos del Sistema Nervioso Inducidos por Alcohol/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Etanol/efectos adversos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/metabolismo , Transmisión Sináptica/efectos de los fármacos , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Amígdala del Cerebelo/metabolismo , Animales , Depresores del Sistema Nervioso Central/efectos adversos , Modelos Animales de Enfermedad , Esquema de Medicación , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ácido Glutámico/metabolismo , Masculino , Técnicas de Cultivo de Órganos , Terminales Presinápticos/metabolismo , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Síndrome de Abstinencia a Sustancias/fisiopatología , Transmisión Sináptica/fisiología
16.
Mol Cell Neurosci ; 30(3): 465-75, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16182561

RESUMEN

Cortistatin-14 (CST) is a neuropeptide expressed in cortical and hippocampal interneurons that shares 11 of 14 residues with somatostatin. In contrast to somatostatin, infusion of CST decreases locomotor activity and selectively enhances slow wave sleep. Here, we show that transgenic mice that overexpress cortistatin under the control of neuron-specific enolase promoter do not express long-term potentiation in the dentate gyrus. This blockade of dentate LTP correlates with profound impairment of hippocampal-dependent spatial learning. Exogenously applied CST to slices of wild-type mice also blocked induction of LTP in the dentate gyrus. Our findings implicate cortistatin in the modulation of synaptic plasticity and cognitive function. Thus, increases in hippocampal cortistatin expression during aging could have an impact on age-related cognitive deficits.


Asunto(s)
Hipocampo/metabolismo , Discapacidades para el Aprendizaje/genética , Aprendizaje/fisiología , Potenciación a Largo Plazo/genética , Péptidos/metabolismo , Transmisión Sináptica/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Giro Dentado/metabolismo , Giro Dentado/fisiopatología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Péptidos y Proteínas de Señalización Intercelular , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Transgénicos , Péptidos/genética , Regiones Promotoras Genéticas/genética
17.
J Pharmacol Exp Ther ; 311(1): 265-73, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15263066

RESUMEN

Although there is now evidence of a role for N-methyl-D-aspartate (NMDA) receptors in nucleus accumbens (NAcc) neurons in the effects of chronic opiate treatment, the cellular and molecular mechanisms underlying this phenomenon are still unclear. Therefore, we studied the effects of chronic morphine on the pharmacological and biophysical properties of NMDA receptors in freshly isolated medium spiny neurons from NAcc. We found that chronic morphine treatment did not alter the affinity for NMDA receptor agonists such as glutamate, homoquinolinic acid, and NMDA, but decreased the affinity of glycine, the allosteric NMDA receptor coagonist, from 2.24 +/- 0.15 microM to 5.1 +/- 1.45 microM. Chronic morphine treatment also altered the affinity of two noncompetitive NMDA receptor antagonists, 7-chloro-kynurenic acid and ifenprodil. However, morphine had no effect on a third antagonist, D-(-)-2-amino-5-phosphonopentanoic acid. Single-exponential fits of desensitized NMDA current tails gave tau values ranging from 0.5 to 4 s in neurons from both control and morphine-treated rats. However, a shift to the left of the distribution of tau values after morphine treatment revealed that NMDA current desensitization rate was accelerated in a majority of NAcc neurons. Taken together with our recent molecular studies, our data are consistent with a shift away from NMDA receptor subunit (NR) NR2B and 2C function toward increased NR2A subunit expression or function after chronic morphine, a process that could alter excitability and integrative properties and may represent a neuroadaptation of NAcc medium spiny neurons underlying morphine dependence.


Asunto(s)
Morfina/farmacología , Neuronas/efectos de los fármacos , Núcleo Accumbens/citología , Receptores de N-Metil-D-Aspartato/metabolismo , Analgésicos Opioides/farmacología , Animales , Antagonistas de Aminoácidos Excitadores/farmacología , Glicina/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/efectos de los fármacos
18.
J Neuroimmunol ; 150(1-2): 37-47, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15081247

RESUMEN

Brain levels of CXC chemokine ligand 10 (CXCL10) are elevated in a number of neuropathological conditions. To determine its impact on neuronal function, we measured synaptic transmission and plasticity in hippocampal slices prepared from transgenic (TG) mice with chronic astroglial production of CXCL10. We also tested the acute effect of recombinant CXCL10 applied to slices from normal C57Bl/6J mice, CXCL10 TG mice and CXCR3 knock out (KO) mice. Chronic production of CXCL10 did not alter synaptic plasticity. By contrast, exogenous CXCL10 (10 ng/ml) significantly inhibited long-term potentiation (LTP) in slices from normal C57Bl/6J mice and CXCL10 TG. The effect was probably receptor-mediated because CXCL10-induced inhibition of LTP was not observed in CXCR3 KO mice. Our findings suggest that acute exposure to CXCL10 alters synaptic plasticity via CXCR3 in mouse hippocampus.


Asunto(s)
Astrocitos/inmunología , Astrocitos/metabolismo , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/farmacología , Plasticidad Neuronal/inmunología , Sinapsis/inmunología , Animales , Astrocitos/fisiología , Quimiocina CXCL10 , Quimiocinas CXC/genética , Quimiocinas CXC/fisiología , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/inmunología , Técnicas In Vitro , Potenciación a Largo Plazo/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Inhibición Neural/genética , Inhibición Neural/inmunología , Plasticidad Neuronal/genética , Receptores CXCR3 , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores de Quimiocina/fisiología , Proteínas Recombinantes/farmacología , Sinapsis/genética , Factores de Tiempo
19.
J Neurosci ; 24(7): 1594-603, 2004 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-14973247

RESUMEN

The modulation of glutamatergic transmission by ethanol may contribute to ethanol intoxication, reinforcement, tolerance, and dependence. Therefore, we used in vitro electrophysiological and in vivo microdialysis techniques to investigate the effects of acute and chronic ethanol on glutamatergic transmission in the central nucleus of amygdala (CeA). Superfusion of 5-66 mM ethanol decreased compound glutamatergic EPSPs and EPSCs in CeA neurons, with half-maximal inhibition elicited by 14 mM ethanol. Ethanol (44 mM) decreased both non-NMDAR- and NMDAR-mediated EPSPs and EPSCs by 21%. Both the ethanol- and ifenprodil-induced depression of NMDAR-mediated EPSPs and EPSCs was enhanced in rats that received chronic ethanol treatment (CET). Ifenprodil also occluded the ethanol effect, suggesting that NR2B subunit-containing receptors may be involved. With local applications of NMDA, acute ethanol elicited a greater inhibition of NMDA currents in slices taken from CET (47%) compared with naive (30%) animals, suggesting that CET sensitizes NMDA receptors to ethanol. Acute ethanol also reduced paired pulse facilitation of EPSPs and EPSCs only in CET animals, suggesting acute ethanol-induced increase of glutamate release. This finding was supported by in vivo experiments showing that infusion of ethanol (0.1-1 M) via reverse microdialysis significantly increased glutamate release into the CeA dialysate but only after CET. Moreover, baseline CeA glutamate content was significantly higher in CET compared with naive animals. These combined findings suggest that CET and withdrawal lead to neuroadaptations of glutamatergic transmission at both presynaptic and postsynaptic sites in CeA, and glutamatergic synapses in CeA may play an important role in ethanol dependence.


Asunto(s)
Amígdala del Cerebelo/fisiología , Etanol/farmacología , Ácido Glutámico/metabolismo , Transmisión Sináptica/efectos de los fármacos , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Electrofisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Masculino , Microdiálisis , Inhibición Neural/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/fisiología , Factores de Tiempo
20.
J Neurosci ; 23(34): 10884-91, 2003 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-14645483

RESUMEN

Cortistatin (CST) is a sleep-modulating peptide found exclusively in the brain. Although CST is closely related to somatostatin (SST) and binds to SST receptors, CST has effects on sleep and neuronal activity in cortex and hippocampus that differ from SST. To uncover the cellular mechanisms affected by CST, we studied the electrophysiological postsynaptic effects of CST and assessed its interaction with SST on hippocampal CA1 pyramidal neurons. CST altered intrinsic membrane properties and occluded SST effects, indicating that both peptides similarly augment the sustained K+ M- and leak-currents (IM and IK(L)). In the presence of SST, however, CST elicited an additional inwardly rectifying component in the hyperpolarized range. This effect was unaffected by barium, used to block K+ currents, but was completely prevented by the selective h-current (Ih) blocker ZD7288. CST, but not SST, selectively increased Ih in a concentration-dependent manner by augmenting its maximum conductance. CST did not shift the Ih activation curve, and the peptide effect was unaffected by a membrane-permeable analog of cAMP. We conclude that CST and SST similarly increase K+ conductances in hippocampal neurons, most likely by activating SST receptors. However, CST additionally augments Ih, a voltage-dependent current that plays a key role in the modulation of synaptic integration and regulates oscillatory activity. Our results indicate that CST targets a specific conductance unaffected by SST to modulate cellular mechanisms implicated in sleep regulation.


Asunto(s)
Hipocampo/efectos de los fármacos , Canales Iónicos/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuropéptidos/farmacología , Animales , Hipocampo/citología , Hipocampo/fisiología , Técnicas In Vitro , Canales Iónicos/fisiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Neuronas/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Células Piramidales/efectos de los fármacos , Células Piramidales/fisiología , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Sueño/efectos de los fármacos , Sueño/fisiología , Somatostatina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA