Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
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
5.
J Pharmacol Exp Ther ; 320(2): 917-25, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17114568

ABSTRACT

Endogenous opioid systems are implicated in the reinforcing effects of ethanol consumption. For example, delta opioid receptor (DOR) knockout (KO) mice show greater ethanol consumption than wild-type (WT) mice (Roberts et al., 2001). To explore the neurobiological correlates underlying these behaviors, we examined effects of acute ethanol application in brain slices from DOR KO mice using whole-cell patch recording techniques. We examined the central nucleus of amygdala (CeA) because the CeA is implicated in alcohol reinforcement (Koob et al., 1998). We found that the acute ethanol effects on GABA(A) receptor-mediated inhibitory postsynaptic currents (IPSCs) were greater in DOR KO mice than in WT mice. Ethanol increased the frequency of miniature IPSCs (mIPSCs) significantly more in DOR KO mice than in WT mice. In CeA of WT mice, application of ICI 174864 [[allyl]2-Tyr-alpha-amino-isobutyric acid (Aib)-Aib-Phe-Leu-OH], a DOR inverse agonist, augmented ethanol actions on mIPSC frequency comparable with ethanol effects seen in DOR KO mice. Superfusion of the selective DOR agonist D-Pen(2),D-Pen(5)-enkephalin decreased the mean frequency of mIPSCs; this effect was reversed by the DOR antagonist naltrindole. These findings suggest that endogenous opioids may reduce ethanol actions on IPSCs of CeA neurons in WT mice through DOR-mediated inhibition of GABA release and that the increased ethanol effect on IPSCs in CeA of DOR KO mice could be, at least in part, due to absence of DOR-mediated inhibition of GABA release. This result supports the hypothesis that endogenous opioid peptides modulate the ethanol-induced augmentation of GABA(A) receptor-dependent circuitry in CeA (Roberto et al., 2003).


Subject(s)
Amygdala/drug effects , Ethanol/pharmacology , Receptors, Opioid, delta/physiology , Amygdala/physiology , Animals , Enkephalin, D-Penicillamine (2,5)-/pharmacology , Enkephalin, Leucine/analogs & derivatives , Enkephalin, Leucine/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Male , Mice , Mice, Inbred C57BL , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/metabolism
6.
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
7.
Alcohol Clin Exp Res ; 30(2): 222-32, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16441271

ABSTRACT

This article presents the proceedings of a symposium entitled "The Tipsy Terminal: Presynaptic Effects of Ethanol" (held at the annual meeting of the Research Society on Alcoholism, in Santa Barbara, CA, June 27, 2005). The objective of this symposium was to focus on a cellular site of ethanol action underrepresented in the alcohol literature, but quickly becoming a "hot" topic. The chairs of the session were Marisa Roberto and George Robert Siggins. Our speakers were chosen on the basis of the diverse electrophysiological and other methods used to discern the effects of acute and chronic ethanol on presynaptic terminals and on the basis of significant insights that their data provide for understanding ethanol actions on neurons in general, as mechanisms underlying problematic behavioral effects of alcohol. The 5 presenters drew from their recent studies examining the effects of acute and chronic ethanol using a range of sophisticated methods from electrophysiological analysis of paired-pulse facilitation and spontaneous and miniature synaptic currents (Drs. Weiner, Valenzuela, Zhu, and Morrisett), to direct recording of ion channel activity and peptide release from acutely isolated synaptic terminals (Dr. Treistman), to direct microscopic observation of vesicular release (Dr. Morrisett). They showed that ethanol administration could both increase and decrease the probability of release of different transmitters from synaptic terminals. The effects of ethanol on synaptic terminals could often be correlated with important behavioral or developmental actions of alcohol. These and other novel findings suggest that future analyses of synaptic effects of ethanol should attempt to ascertain, in multiple brain regions, the role of presynaptic terminals, relevant presynaptic receptors and signal transduction linkages, exocytotic mechanisms, and their involvement in alcohol's behavioral actions. Such studies could lead to new treatment strategies for alcohol intoxication, alcohol abuse, and alcoholism.


Subject(s)
Alcoholic Intoxication/physiopathology , Alcoholism/physiopathology , Brain/drug effects , Receptors, Presynaptic/drug effects , Animals , Brain/physiopathology , Glutamic Acid/metabolism , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neuropeptides/metabolism , Receptors, Presynaptic/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/physiology , gamma-Aminobutyric Acid/metabolism
8.
Pharmacol Ther ; 107(1): 80-98, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15963352

ABSTRACT

Considerable evidence suggests that the synapse is the most sensitive CNS element for ethanol effects. Although most alcohol research has focussed on the postsynaptic sites of ethanol action, especially regarding interactions with the glutamatergic and GABAergic receptors, few such studies have directly addressed the possible presynaptic loci of ethanol action, and even fewer describe effects on synaptic terminals. Nonetheless, there is burgeoning evidence that presynaptic terminals play a major role in ethanol effects. The methods used to verify such ethanol actions range from electrophysiological analysis of paired-pulse facilitation (PPF) and spontaneous and miniature synaptic potentials to direct recording of ion channel activity and transmitter/messenger release from acutely isolated synaptic terminals, and microscopic observation of vesicular release, with a focus predominantly on GABAergic, glutamatergic, and peptidergic synapses. The combined data suggest that acute ethanol administration can both increase and decrease the release of these transmitters from synaptic terminals, and more recent results suggest that prolonged or chronic ethanol treatment (CET) can also alter the function of presynaptic terminals. These new findings suggest that future analyses of synaptic effects of ethanol should attempt to ascertain the role of presynaptic terminals and their involvement in alcohol's behavioral actions. Other future directions should include an assessment of ethanol's effects on presynaptic signal transduction linkages and on the molecular machinery of transmitter release and exocytosis in general. Such studies could lead to the formulation of new treatment strategies for alcohol intoxication, alcohol abuse, and alcoholism.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Glutamates , Presynaptic Terminals/drug effects , Signal Transduction/drug effects , gamma-Aminobutyric Acid , Animals , Glutamates/drug effects , Glutamates/metabolism , Glutamates/physiology , Humans , Signal Transduction/physiology , gamma-Aminobutyric Acid/drug effects , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/physiology
9.
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
10.
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
11.
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
12.
Ann N Y Acad Sci ; 1003: 196-211, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14684447

ABSTRACT

Both the nucleus accumbens (NAcc) and central amygdala (CeA) are thought to play roles in tolerance to, and dependence on, abused drugs. Although our past studies in rat brain slices suggested a role for NMDA receptors (NMDARs) in NAcc neurons in the effects of acute and chronic opiate treatment, the cellular and molecular mechanisms remained unclear. Therefore, we examined the effects of morphine dependence on electrophysiological properties of NMDARs in freshly isolated NAcc neurons and on expression of mRNA coding for NR2A-C subunits using single-cell RT-PCR. Chronic morphine did not alter the affinity for NMDAR agonists glutamate, homoquinolinate, or NMDA, but decreased the affinity of the coagonist glycine. Chronic morphine altered the NMDAR inhibition by two NMDAR antagonists, 7-Cl-kynurenate and ifenprodil, but not that by d-APV or Mg2+. Chronic morphine accelerated the NMDA current desensitization rate in NAcc neurons. In single-cell RT-PCR, chronic morphine predominantly reduced the number of neurons expressing multiple NR2 subunits. Ethanol also alters NMDARs. We found that low ethanol concentrations (IC50 = 13 mM) inhibited NMDA currents and NMDA-EPSPs in most NAcc neurons in a slice preparation. NAcc neurons from ethanol-dependent rats showed enhanced NMDA sensitivity. In CeA neurons, acute ethanol decreased (by 10-25%) non-NMDA- and NMDA-EPSPs in most neurons. In CeA neurons from ethanol-dependent rats, acute ethanol decreased the non-NMDA-EPSPs to the same extent as in naïve rats, but inhibited (by 30-40%) NMDA-EPSPs significantly more than in controls, suggesting sensitization to ethanol. Preliminary studies with microdialysis and real-time PCR analysis support this idea: local ethanol administration in vivo had no effect on glutamate release, but chronic ethanol nearly tripled the expression of NR2B subunits (the most ethanol sensitive) in CeA. These combined findings suggest that changes in glutamatergic transmission in NAcc and CeA may underlie the neuroadaptions that lead to opiate and ethanol dependence.


Subject(s)
Alcoholism/physiopathology , Glutamates/physiology , Opioid-Related Disorders/physiopathology , Synaptic Transmission/physiology , Amygdala/physiology , Animals , Ethanol/pharmacology , Glycine/metabolism , Humans , Morphine Dependence/physiopathology , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/physiology , Rats , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/drug effects , Synaptic Transmission/drug effects
13.
J Pharmacol Exp Ther ; 302(3): 1135-45, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183673

ABSTRACT

In the course of studying N-methyl-D-aspartate (NMDA) receptors of the nucleus accumbens (NAcc), we found that 20% of freshly isolated medium spiny neurons, as well as all interneurons, responded in an unexpected way to long (5-s) coapplication of NMDA and glycine, the coagonist of NMDA receptors. Whereas the reversal potential of the peak NMDA current of this subset of neurons was still around 0 mV, the desensitizing current became outward at hyperpolarized potentials around -30 mV. A Cl(-)-free solution shifted the equilibrium potentials of the desensitized currents to around 0 mV. This outward current was not blocked by a Ca(2+)-free, Ba(2+)-containing solution, suggesting that the anionic conductance was not activated by Ca(2+) influx through NMDA receptor channels. Interestingly, glycine alone also evoked a current with a similar hyperpolarized reversal potential in this subset of neurons. The glycine current reversed around -50 mV, rectified outwardly, and inactivated strongly. Its desensitization was best fitted with a double exponential. Only the slow desensitization showed clear voltage dependence. The glycine current was not blocked by 200 microM picrotoxin and 10 microM zinc, was weakly antagonized by 1 microM strychnine, and was not enhanced by 1 microM zinc. In addition, 1 mM taurine, but not GABA, inactivated glycine currents, and 1 mM glycine occluded 10 mM taurine-mediated currents. These data indicate that a subset of nucleus accumbens neurons expresses glycine receptors and that either glycine or taurine could be an endogenous agonist for these receptors.


Subject(s)
Neurons/metabolism , Nucleus Accumbens/metabolism , Receptors, Glycine/biosynthesis , Animals , Electrophysiology , Excitatory Amino Acid Agonists/pharmacology , GABA Agonists/pharmacology , Glycine/pharmacology , Glycine Agents/pharmacology , Male , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Neurons/drug effects , Nucleus Accumbens/cytology , Nucleus Accumbens/drug effects , Patch-Clamp Techniques , Picrotoxin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glycine/agonists , Receptors, Glycine/antagonists & inhibitors , Strychnine/pharmacology , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...