Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Addict Biol ; 22(3): 766-778, 2017 May.
Article in English | MEDLINE | ID: mdl-26786379

ABSTRACT

The basolateral nucleus of the amygdala (BLA) is critical to the pathophysiology of anxiety-driven alcohol drinking and relapse. The endogenous cannabinoid/type 1 cannabinoid receptor (eCB/CB1 ) system curbs BLA-driven anxiety and stress responses via a retrograde negative feedback system that inhibits neurotransmitter release, and BLA CB1 activation reduces GABA release and drives anxiogenesis. Additionally, decreased amygdala CB1 is observed in abstinent alcoholic patients and ethanol withdrawn rats. Here, we investigated the potential disruption of eCB/CB1 signaling on GABAergic transmission in BLA pyramidal neurons of rats exposed to 2-3 weeks intermittent ethanol. In the naïve rat BLA, the CB1 agonist WIN 55,212-2 (WIN) decreased GABA release, and this effect was prevented by the CB1 antagonist AM251. AM251 alone increased GABA release via a mechanism requiring postsynaptic calcium-dependent activity. This retrograde tonic eCB/CB1 signaling was diminished in chronic ethanol exposed rats, suggesting a functional impairment of the eCB/CB1 system. In contrast, acute ethanol increased GABAergic transmission similarly in naïve and chronic ethanol exposed rats, via both presynaptic and postsynaptic mechanisms. Notably, CB1 activation impaired ethanol's facilitation of GABAergic transmission across both groups, but the AM251-induced and ethanol-induced facilitation of GABA release was additive, suggesting independent presynaptic sites of action. Collectively, the present findings highlight a critical CB1 influence on BLA GABAergic transmission that is dysregulated by chronic ethanol exposure and, thus, may contribute to the alcohol-dependent state.


Subject(s)
Alcoholism/physiopathology , Basolateral Nuclear Complex/drug effects , Ethanol/toxicity , Receptor, Cannabinoid, CB1/drug effects , gamma-Aminobutyric Acid/drug effects , Animals , Basolateral Nuclear Complex/physiopathology , Central Nervous System Depressants/toxicity , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
2.
Addict Biol ; 21(4): 788-801, 2016 07.
Article in English | MEDLINE | ID: mdl-25940135

ABSTRACT

The endogenous cannabinoids (eCBs) influence the acute response to ethanol and the development of tolerance, dependence and relapse. Chronic alcohol exposure alters eCB levels and Type 1 cannabinoid receptor (CB1 ) expression and function in brain regions associated with addiction. CB1 inhibits GABA release, and GABAergic dysregulation in the central nucleus of the amygdala (CeA) is critical in the transition to alcohol dependence. We investigated possible disruptions in CB1 signaling of rat CeA GABAergic transmission following intermittent ethanol exposure. In the CeA of alcohol-naive rats, CB1 agonist WIN 55,212-2 (WIN) decreased the frequency of spontaneous and miniature GABAA receptor-mediated inhibitory postsynaptic currents (s/mIPSCs). This effect was prevented by CB1 antagonism, but not Type 2 cannabinoid receptor (CB2 ) antagonism. After 2-3 weeks of intermittent ethanol exposure, these WIN inhibitory effects were attenuated, suggesting ethanol-induced impairments in CB1 function. The CB1 antagonist AM251 revealed a tonic eCB/CB1 control of GABAergic transmission in the alcohol-naive CeA that was occluded by calcium chelation in the postsynaptic cell. Chronic ethanol exposure abolished this tonic CB1 influence on mIPSC, but not sIPSC, frequency. Finally, acute ethanol increased CeA GABA release in both naive and ethanol-exposed rats. Although CB1 activation prevented this effect, the AM251- and ethanol-induced GABA release were additive, ruling out a direct participation of CB1 signaling in the ethanol effect. Collectively, these observations demonstrate an important CB1 influence on CeA GABAergic transmission and indicate that the CeA is particularly sensitive to alcohol-induced disruptions of CB1 signaling.


Subject(s)
Alcoholism/physiopathology , Central Amygdaloid Nucleus/drug effects , Ethanol/pharmacology , GABAergic Neurons/drug effects , Receptor, Cannabinoid, CB1/drug effects , Receptors, GABA-A/drug effects , Animals , Central Amygdaloid Nucleus/physiopathology , Central Nervous System Depressants/pharmacology , Chronic Disease , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Synapses/drug effects
3.
Front Pharmacol ; 6: 49, 2015.
Article in English | MEDLINE | ID: mdl-25852553

ABSTRACT

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.

4.
Article in English | MEDLINE | ID: mdl-24926240

ABSTRACT

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.

5.
Brain Behav Immun ; 40: 191-202, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24675033

ABSTRACT

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.


Subject(s)
Central Amygdaloid Nucleus/drug effects , Ethanol/pharmacology , Immunity, Innate , Lipopolysaccharide Receptors/metabolism , Neurons/drug effects , Receptors, GABA-A/metabolism , Toll-Like Receptor 4/metabolism , Animals , Central Amygdaloid Nucleus/immunology , Central Amygdaloid Nucleus/physiology , Immunologic Factors/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/pharmacology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/immunology , Neurons/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/immunology , Toll-Like Receptor 4/antagonists & inhibitors
6.
Addict Biol ; 17(4): 694-705, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21955024

ABSTRACT

The neuropeptide galanin and its three receptor subtypes (GalR1-3) are expressed in the central amygdala (CeA), a brain region involved in stress- and anxiety-related behaviors, as well as alcohol dependence. Galanin also has been suggested to play a role in alcohol intake and alcohol dependence. We examined the effects of galanin in CeA slices from wild-type and knockout (KO) mice deficient of GalR2 and both GalR1 and GalR2 receptors. Galanin had dual effects on gamma-aminobutyric acid (GABA)-ergic transmission, decreasing the amplitudes of pharmacologically isolated GABAergic inhibitory postsynaptic potentials (IPSPs) in over half of CeA neurons but augmenting IPSPs in the others. The increase in IPSP size was absent after superfusion of the GalR3 antagonist SNAP 37889, whereas the IPSP depression was absent in CeA neurons of GalR1 × GalR2 double KO and GalR2 KO mice. Paired-pulse facilitation studies showed weak or infrequent effects of galanin on GABA release. Thus, galanin may act postsynaptically through GalR3 to augment GABAergic transmission in some CeA neurons, whereas GalR2 receptors likely are involved in the depression of IPSPs. Co-superfusion of ethanol, which augments IPSPs presynaptically, together with galanin caused summated effects of ethanol and galanin in those CeA neurons showing galanin-augmented IPSPs, suggesting the two agents act via different mechanisms in this population. However, in neurons showing IPSP-diminishing galanin effects, galanin blunted the ethanol effects, suggesting a preemptive effect of galanin. These findings may increase understanding of the complex cellular mechanisms that underlie the anxiety-related behavioral effects of galanin and ethanol in CeA.


Subject(s)
Amygdala/drug effects , Galanin/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Receptors, Corticotropin-Releasing Hormone/drug effects , Animals , Anxiety/etiology , Central Nervous System Depressants/pharmacology , Drug Interactions , Ethanol/pharmacology , Evoked Potentials/drug effects , GABAergic Neurons/drug effects , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/etiology , gamma-Aminobutyric Acid/drug effects
7.
Addict Biol ; 16(4): 551-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21182569

ABSTRACT

We investigated possible alterations of pharmacologically-isolated, evoked GABA(A) inhibitory postsynaptic potentials (eIPSPs) and miniature GABA(A) inhibitory postsynaptic currents (mIPSCs) in the rat central amygdala (CeA) elicited by acute application of µ-opioid receptor (MOR) agonists (DAMGO and morphine; 1 µM) and by chronic morphine treatment with morphine pellets. The acute activation of MORs decreased the amplitudes of eIPSPs, increased paired-pulse facilitation (PPF) of eIPSPs and decreased the frequency (but not the amplitude) of mIPSCs in a majority of CeA neurons, suggesting that acute MOR-dependent modulation of this GABAergic transmission is mediated predominantly via presynaptic inhibition of GABA release. We observed no significant changes in the membrane properties, eIPSPs, PPF or mIPSCs of CeA neurons during chronic morphine treatment compared to CeA of naïve or sham rats. Superfusion of the MOR antagonist CTOP (1 µM) increased the mean amplitude of eIPSPs in a majority of CeA neurons to the same degree in both naïve/sham and morphine-treated rats, suggesting a tonic activation of MORs in both conditions. Superfusion of DAMGO decreased eIPSP amplitudes and the frequency of mIPSCs equally in both naïve/sham and morphine-treated rats but decreased the amplitude of mIPSCs only in morphine treated rats, an apparent postsynaptic action. Our combined findings suggest the development of tolerance of the CeA GABAergic system to inhibitory effects of acute activation of MORs on presynaptic GABA release and possible alteration of MOR-dependent postsynaptic mechanisms that may represent important neuroadaptations of the GABAergic and MOR systems during chronic morphine treatment.


Subject(s)
Amygdala/drug effects , Amygdala/physiology , Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Morphine/pharmacology , Narcotics/pharmacology , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Receptors, Opioid, mu/agonists , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Animals , Drug Tolerance , Male , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/physiology , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
8.
Biol Psychiatry ; 67(9): 831-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20060104

ABSTRACT

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.


Subject(s)
Alcoholism/metabolism , Amygdala/drug effects , Amygdala/metabolism , Corticotropin-Releasing Hormone/pharmacology , gamma-Aminobutyric Acid/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Alcoholism/blood , Alcoholism/pathology , Amygdala/pathology , Animals , Body Weight/drug effects , Central Nervous System Depressants/administration & dosage , Corticotropin-Releasing Hormone/genetics , Cyclophilins/pharmacology , Ethanol/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , Hormone Antagonists/pharmacology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Male , Microdialysis/methods , Neurons/drug effects , Neurons/physiology , Patch-Clamp Techniques , Phosphinic Acids/pharmacology , Propanolamines/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Time Factors , Valine/analogs & derivatives , Valine/pharmacology
9.
ScientificWorldJournal ; 9: 68-85, 2009 Jan 18.
Article in English | MEDLINE | ID: mdl-19151899

ABSTRACT

Corticotropin-releasing factor (CRF) is a 41-amino-acid neuropeptide involved in stress responses initiated from several brain areas, including the amygdala formation. Research shows a strong relationship between stress, brain CRF, and excessive alcohol consumption. Behavioral studies suggest that the central amygdala (CeA) is significantly involved in alcohol reward and dependence. We recently reported that the ethanol augmentation of GABAergic synaptic transmission in rat CeA involves CRF1 receptors, because both CRF and ethanol significantly enhanced the amplitude of evoked GABAergic inhibitory postsynaptic currents (IPSCs) in CeA neurons from wild-type (WT) and CRF2 knockout (KO) mice, but not in neurons of CRF1 KO mice. The present study extends these findings using selective CRF receptor ligands, gene KO models, and miniature IPSC (mIPSC) analysis to assess further a presynaptic role for the CRF receptors in mediating ethanol effects in the CeA. In whole-cell patch recordings of pharmacologically isolated GABAAergic IPSCs from slices of mouse CeA, both CRF and ethanol augmented evoked IPSCs in a concentration-dependent manner, with low EC50s. A CRF1 (but not CRF2) KO construct and the CRF1-selective nonpeptide antagonist NIH-3 (LWH-63) blocked the augmenting effect of both CRF and ethanol on evoked IPSCs. Furthermore, the new selective CRF1 agonist stressin1, but not the CRF2 agonist urocortin 3, also increased evoked IPSC amplitudes. Both CRF and ethanol decreased paired-pulse facilitation (PPF) of evoked IPSCs and significantly enhanced the frequency, but not the amplitude, of spontaneous miniature GABAergic mIPSCs in CeA neurons of WT mice, suggesting a presynaptic site of action. The PPF effect of ethanol was abolished in CeA neurons of CRF1 KO mice. The CRF1 antagonist NIH-3 blocked the CRF- and ethanol-induced enhancement of mIPSC frequency in CeA neurons. These data indicate that presynaptic CRF1 receptors play a critical role in permitting or mediating ethanol enhancement of GABAergic synaptic transmission in CeA, via increased vesicular GABA release, and thus may be a rational target for the treatment of alcohol abuse and alcoholism.


Subject(s)
Amygdala/drug effects , Amygdala/metabolism , Ethanol/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/biosynthesis , Animals , Electrophysiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/deficiency , Receptors, Corticotropin-Releasing Hormone/genetics
10.
J Pharmacol Exp Ther ; 328(1): 284-93, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18854491

ABSTRACT

Endogenous opioid systems are implicated in the actions of ethanol. For example, mu-opioid receptor (MOR) knockout (KO) mice self-administer less alcohol than the genetically intact counterpart wild-type (WT) mice (Roberts et al., 2000). MOR KO mice also exhibit less anxiety-like behavior than WT mice (Filliol et al., 2000). To investigate the neurobiological mechanisms underlying these behaviors, we examined the effect of ethanol in brain slices from MOR KO and WT mice using sharp-electrode and whole-cell patch recording techniques. We focused our study in the central nucleus of the amygdala (CeA) because it is implicated in alcohol drinking behavior and stress behavior. We found that the amplitudes of evoked inhibitory postsynaptic currents (IPSCs) or inhibitory postsynaptic potentials (IPSPs) were significantly greater in MOR KO mice than WT mice. In addition, the baseline frequencies of spontaneous and miniature GABA(A) receptor-mediated inhibitory postsynaptic currents were significantly greater in CeA neurons from MOR KO than WT mice. However, ethanol enhancements of evoked IPSP and IPSC amplitudes and the frequency of miniature IPSCs were comparable between WT and MOR KO mice. Baseline spontaneous and miniature excitatory postsynaptic currents (EPSCs) and ethanol effects on EPSCs were not significantly different between MOR KO and WT mice. Based on knowledge of CeA circuitry and projections, we hypothesize that the role of MOR- and GABA receptor-mediated mechanisms in CeA underlying reinforcing effects of ethanol operate independently, possibly through pathway-specific responses within CeA.


Subject(s)
Amygdala/physiology , Ethanol/pharmacology , Receptors, Opioid, mu/physiology , Synapses/physiology , Synaptic Transmission/physiology , Amygdala/drug effects , Animals , Crosses, Genetic , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Patch-Clamp Techniques , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/drug effects , Synapses/drug effects
11.
Neurochem Res ; 33(2): 285-91, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17973188

ABSTRACT

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.


Subject(s)
Galanin/pharmacology , Raphe Nuclei/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism , Animals , In Vitro Techniques , Male , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Rats , Rats, Sprague-Dawley
12.
J Neurosci Res ; 83(4): 532-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16453311

ABSTRACT

The nucleus accumbens (NAcc) and central amygdala (CeA) are parts of the extended amygdala, a complex that plays a key role in drug abuse and dependence. Our previous studies showed that opiates and ethanol alter glutamatergic transmission in these regions. N-methyl-D-aspartate (NMDA) receptors are key components of glutamatergic transmission likely involved in the development of opiate tolerance and dependence. In this study we examined the effects of chronic morphine administration on gene and protein expression of three major NMDA receptors subunits (NR1, NR2A, and NR2B) in NAcc and CeA. Real-time PCR showed no differences in mRNA levels of any of the subunits in the whole NAcc between naïve and morphine-dependent rats. However, at the protein level, immunoblotting revealed that chronic morphine significantly increased levels of NR1 and NR2B subunits. In contrast to the case for NAcc, in CeA we found an increased mRNA level for the NR1 subunit only but unchanged protein levels of all three subunits in morphine-dependent rats. The altered expressions of NMDA receptor subunits, especially in NAcc, of morphine-dependent rats may represent a neuroadaptation to chronic morphine and suggest a mechanism for the changes of glutamatergic transmission found in the extended amygdala in dependent rats. In addition, our results indicate a region-specific response of NMDA receptor subunits to chronic morphine administration at the gene and protein levels.


Subject(s)
Amygdala/metabolism , Morphine/pharmacology , Narcotics/pharmacology , Receptors, N-Methyl-D-Aspartate/biosynthesis , Actins/biosynthesis , Amygdala/drug effects , Animals , Blotting, Western , Gene Expression/drug effects , Male , Morphine Dependence/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Neuropsychopharmacology ; 31(5): 988-96, 2006 May.
Article in English | MEDLINE | ID: mdl-16052244

ABSTRACT

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.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Amygdala/drug effects , Ethanol/adverse effects , Receptors, N-Methyl-D-Aspartate/drug effects , Substance Withdrawal Syndrome/metabolism , Synaptic Transmission/drug effects , Alcohol-Induced Disorders, Nervous System/physiopathology , Amygdala/metabolism , Animals , Central Nervous System Depressants/adverse effects , Disease Models, Animal , Drug Administration Schedule , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Male , Organ Culture Techniques , Presynaptic Terminals/metabolism , Protein Subunits/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Substance Withdrawal Syndrome/physiopathology , Synaptic Transmission/physiology
14.
J Neurosci ; 24(45): 10159-66, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537886

ABSTRACT

The central nucleus of amygdala (CeA) is important in regulating alcohol consumption and plays a major role in the anxiogenic response to ethanol withdrawal. We showed previously that acute ethanol augments GABA(A) receptor-mediated IPSPs and IPSCs, possibly by a presynaptic mechanism. Here, we have examined the interaction of acute ethanol with the GABAergic system in chronic ethanol-treated (CET) rats using an in vitro CeA slice preparation and in vivo brain microdialysis. We found that in CeA slices from CET rats, the baseline evoked IPSP and IPSC amplitudes were increased, and paired-pulse facilitation ratios were lower than in naive rats, suggesting an increased GABAergic transmission after chronic ethanol treatment. Interestingly, acute ethanol (5-66 mm) significantly enhanced IPSPs and IPSCs equally in CET and naive rats, indicating a lack of tolerance for this effect of acute ethanol. Analysis of miniature IPSC frequency suggests that the increased GABAergic transmission by both acute and chronic ethanol arises from a presynaptic mechanism involving enhanced vesicular release of GABA. These data are supported by microdialysis studies showing that CET rats presented a fourfold increase in baseline GABA dialysate content compared with naive rats. In vivo administration of ethanol (0.1, 0.3, and 1.0 m) produced a dose-dependent increase in GABA release in the CeA dialysate in both CET and naive rats. These combined findings suggest that acute and chronic ethanol increases GABA release in CeA and support previous reports that the behavioral actions of ethanol are mediated, in part, by increased GABAergic transmission in the CeA.


Subject(s)
Alcoholism/physiopathology , Amygdala/metabolism , Ethanol/toxicity , Substance-Related Disorders/physiopathology , gamma-Aminobutyric Acid/metabolism , Amygdala/drug effects , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Electric Stimulation , Ethanol/administration & dosage , Ethanol/pharmacology , Male , Microdialysis , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
15.
J Pharmacol Exp Ther ; 311(1): 172-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15152029

ABSTRACT

gamma-Hydroxybutyrate (GHB) is used for the treatment of alcoholism and to induce absence seizures in animals, but it has also recently emerged as a drug of abuse. In hippocampal neurons, GHB may activate its own putative receptor as well as GABA(B) receptors to affect synaptic transmission. We used voltage-clamp recordings of rat CA1 pyramidal neurons to characterize the postsynaptic conductances affected by GHB and to further clarify the site of GHB action. Low concentrations of GHB (0.1-1 mM) did not affect postsynaptic properties, but 10 mM GHB elicited an outward current at resting potential by augmenting an inwardly rectifying potassium current and concomitantly decreased the hyperpolarization-activated H-current (I(h)). Like GHB, the selective GABA(B)-receptor agonist baclofen (20 microM) increased a potassium current and decreased I(h). In the presence of 10 mM GHB, the baclofen effects were largely occluded. The selective GABA(B) receptor antagonist CGP 55845 [3-N[1-(S)-(3,4-dichlorophenyl)ethyl]amino-2-(S)-hydroxypropyl-p-benzyl-phosphinic acid] blocked the effects of both GHB and baclofen, whereas the putative GHB receptor antagonist NCS-382 [(2E)-(5-hydroxy-5,7,8,9-tetrahydro-6H-benzo[a][7]annulen-6-ylidene ethanoic acid] was ineffective. The GHB and baclofen effects were prevented in the presence of 200 microM barium, indicating that GHB augments a K(+) conductance, probably a G protein-coupled inwardly rectifying K(+) (GIRK) current. The decrease of I(h) by GHB and baclofen was also prevented by barium, suggesting that the diminution of I(h) is secondary to GIRK augmentation. Our results indicate that high GHB levels, which can be reached during abuse or intoxication, activate only GABA(B) receptors and not GHB receptors at the postsynaptic level to augment an inwardly rectifying K(+) current and decrease I(h).


Subject(s)
Potassium/physiology , Receptors, GABA-B/physiology , Sodium Oxybate/pharmacology , Animals , Baclofen/pharmacology , Barium/pharmacology , GABA Agonists/pharmacology , Protons , Rats , Rats, Sprague-Dawley , Receptors, GABA-B/drug effects , Synaptic Transmission/drug effects
16.
Science ; 303(5663): 1512-4, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-15001778

ABSTRACT

The central amygdala (CeA) plays a role in the relationship among stress, corticotropin-releasing factor (CRF), and alcohol abuse. In whole-cell recordings, both CRF and ethanol enhanced gamma-aminobutyric acid-mediated (GABAergic) neurotransmission in CeA neurons from wild-type and CRF2 receptor knockout mice, but not CRF1 receptor knockout mice. CRF1 (but not CRF2) receptor antagonists blocked both CRF and ethanol effects in wild-type mice. These data indicate that CRF1 receptors mediate ethanol enhancement of GABAergic synaptic transmission in the CeA, and they suggest a cellular mechanism underlying involvement of CRF in ethanol's behavioral and motivational effects.


Subject(s)
Amygdala/physiology , Ethanol/pharmacology , Neurons/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism , Alcohol Drinking , Amygdala/drug effects , Animals , Corticotropin-Releasing Hormone/pharmacology , Dose-Response Relationship, Drug , Evoked Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Patch-Clamp Techniques , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, GABA-A/metabolism , Stress, Psychological/physiopathology
17.
J Neurosci ; 24(7): 1594-603, 2004 Feb 18.
Article in English | MEDLINE | ID: mdl-14973247

ABSTRACT

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.


Subject(s)
Amygdala/physiology , Ethanol/pharmacology , Glutamic Acid/metabolism , Synaptic Transmission/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Male , Microdialysis , Neural Inhibition/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology , Time Factors
18.
J Neurosci ; 23(34): 10884-91, 2003 Nov 26.
Article in English | MEDLINE | ID: mdl-14645483

ABSTRACT

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.


Subject(s)
Hippocampus/drug effects , Ion Channels/drug effects , Neurons/drug effects , Neuropeptides/pharmacology , Animals , Hippocampus/cytology , Hippocampus/physiology , In Vitro Techniques , Ion Channels/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/physiology , Patch-Clamp Techniques , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Sleep/drug effects , Sleep/physiology , Somatostatin/pharmacology
19.
Proc Natl Acad Sci U S A ; 100(4): 2053-8, 2003 Feb 18.
Article in English | MEDLINE | ID: mdl-12566570

ABSTRACT

We examined the interaction of ethanol with the gamma-aminobutyric acid (GABA)ergic system in neurons of slices of the rat central amygdala nucleus (CeA), a brain region thought to be critical for the reinforcing effects of ethanol. Brief superfusion of 11-66 mM ethanol significantly increased GABA type A (GABA(A)) receptor-mediated inhibitory postsynaptic potentials (IPSPs) and currents (IPSCs) in most CeA neurons, with a low apparent EC(50) of 20 mM. Acute superfusion of 44 mM ethanol increased the amplitude of evoked GABA(A) IPSPs and IPSCs in 70% of CeA neurons. The ethanol enhancement of IPSPs and IPSCs occurred to a similar extent in the presence of the GABA type B (GABA(B)) receptor antagonist CGP 55845A, suggesting that this receptor is not involved in the ethanol effect on CeA neurons. Ethanol superfusion also decreased paired-pulse facilitation of evoked GABA(A) IPSPs and IPSCs and always increased the frequency and sometimes the amplitude of spontaneous miniature GABA(A) IPSCs as well as responses to local GABA application, indicating both presynaptic and postsynaptic sites of action for ethanol. Thus, the CeA is the first brain region to reveal, without conditional treatments such as GABA(B) antagonists, consistent, low-dose ethanol enhancement of GABAergic transmission at both pre- and postsynaptic sites. These findings add further support to the contention that the ethanol-GABA interaction in CeA plays an important role in the reinforcing effects of ethanol.


Subject(s)
Amygdala/drug effects , Ethanol/pharmacology , Neurons/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/physiology , Amygdala/cytology , Amygdala/physiology , Animals , Evoked Potentials , In Vitro Techniques , Neurons/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...