Skip to main content
Cold Spring Harbor Perspectives in Medicine logoLink to Cold Spring Harbor Perspectives in Medicine
. 2012 Dec;2(12):a012195. doi: 10.1101/cshperspect.a012195

The Central Amygdala and Alcohol: Role of γ-Aminobutyric Acid, Glutamate, and Neuropeptides

Marisa Roberto 1, Nicholas W Gilpin 2, George R Siggins 3
PMCID: PMC3543070  PMID: 23085848

Abstract

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.


Alcohol has strong and persistent effects on synaptic transmission in the central amygdala. Most notably, it potentiates the GABAergic system.


Alcoholism (i.e., dependence on alcohol) is a complex and dynamic disease process. Alcohol dependence is a chronically relapsing disorder characterized by (1) compulsive drug seeking and drug taking, (2) loss of control in limiting intake (in terms of amount of drug per bout and number of drug-taking bouts), and (3) the emergence of a withdrawal syndrome in the absence of the drug that includes, but is not limited to, dysphoria, sleep disturbances, disruption of autonomic processes, and increases in anxiety and irritability (ICD-10 and DSM-IV). Many years of research have shaped the current view that excessive alcohol consumption is largely mediated by an organism’s past experience with alcohol (e.g., intake pattern and frequency), and is driven by the emotional disturbances, rather than the physical disturbances, associated with alcohol withdrawal and abstinence (Koob 2003; Heilig et al. 2010). Although the central amygdala (CeA) is known to play a role in such negative affective alcohol responses, the neuronal circuitry underlying these behavioral stages is still under considerable scrutiny. This article will focus on neurotransmission in the central amygdala and its role in driving the negative affect characteristic of the withdrawal phase of alcohol addiction. Throughout this article, acute alcohol exposure refers to in vitro application of alcohol onto the slice preparation, whereas chronic alcohol exposure refers to long-duration (at least several weeks) in vivo alcohol exposure.

CENTRAL AMYGDALA IS A HUB FOR NEGATIVE EMOTIONAL CIRCUITRY

Chronic consumption of large quantities of drugs, including alcohol, promotes a transition from casual drug use to drug dependence that is defined in part by down-regulation of dopamine signaling in the mesocorticolimbic reward system, hyperactivity of glutamate signaling, and dysregulation of brain stress systems (Koob and Volkow 2010). Chronic alcohol effects on brain stress systems can refer to either alcohol-induced changes in neuroendocrine function (i.e., hypothalamic-pituitary-adrenal [HPA] axis; Kiefer and Wiedemann 2004; Clarke et al. 2008) or the recruitment of extrahypothalamic brain stress systems such as the amygdala (Koob 2009). The present article details the effects of acute and chronic alcohol on synaptic transmission and plasticity in the CeA and neighboring regions, and the role of these regions in mediating alcohol-related behaviors. We also will review the literature on peptidergic modulation of inhibitory and excitatory transmission in the central (and extended) amygdala, because these peptides share a common cellular target and interact with each other and alcohol.

Many of the long-term emotional disturbances associated with alcohol abuse and dependence are attributed to neurotransmission within a conceptual macrostructure in the basal forebrain called the “extended amygdala” (Koob 2008). In the context of drug addiction, the major constituents of the extended amygdala are the CeA, the lateral portion of the bed nucleus of stria terminalis (BNST), and nucleus accumbens (NAc) shell (Heimer and Alheid 1991). These regions show similar morphology, a high degree of interconnectivity, and overlapping afferents from limbic cortices, hippocampus, and basolateral amygdala (BLA). The outputs of the extended amygdala project largely to effector regions, including lateral hypothalamus and various brain stem regions, that produce behaviors related to fear and anxiety (Davis et al. 2010).

The role of the extended amygdala in fear and anxiety has been previously described in detail (Ciocchi et al. 2010; Davis et al. 2010). The CeA and BNST are integral in mediating fear and anxiety responses. The BLA receives significant sensory input from thalamus and cortex, sends prominent glutamatergic projections to CeA and BNST, and is integral in both conditioning (Phelps and LeDoux 2005) and extinction (Quirk and Mueller 2008) processes. The CeA is composed mostly of γ-aminobutyric acid-ergic (GABAergic) projection neurons and interneurons (Sun and Cassell 1993; Veinante and Freund-Mercier 1998), and the BNST is a major target of CeA projection neurons (Krettek and Price 1978; Weller and Smith 1982; Sun and Cassell 1993; Veinante and Freund-Mercier 1998). Of major relevance for this article, connections between CeA and BNST often contain neuropeptide cotransmitters. For example, the CeA is a major source of corticotropin-releasing factor (CRF) in the BNST (Sakanaka et al. 1986). Therefore, the CeA is uniquely situated to convert sensory information into behavioral and physiological responses, and this is particularly true for stress- and alcohol-related stimuli.

Neuropeptides in the CeA are important for producing the negative affective state observed during withdrawal from drugs, including alcohol (Koob 2008). Here we will describe alcohol-induced neuroadaptations in select peptidergic systems (CRF, nociception, and NPY), largely in the context of the CeA. It is becoming increasingly evident that these peptides interact in complex ways in the CeA to modulate GABAergic inhibitory and glutamatergic excitatory transmission, and that dysregulation of these peptide systems by alcohol alters the way in which they modulate such neurotransmission. Conceptually, these neuropeptides have been divided into prostress peptides and antistress peptides that respectively, promote and rescue negative affective disturbances during drug abstinence following heavy drug use. Prostress peptides include CRF, dynorphin, orexin, and vasopressin, whereas antistress peptides include neuropeptide Y (NPY), enkephalin, and nociceptin; however, owing to space limitations we will discuss only a select few of these peptides.

AMYGDALAR GABAergic SYSTEM AND ALCOHOL

GABA, the major inhibitory transmitter in the brain, acts on two classes of GABA receptors: GABAA (which includes GABAA-rho subclass—formerly GABAC) and GABAB. GABAA receptors are ligand-gated ion channels, whereas GABAB receptors are G protein coupled. There is considerable evidence that GABAergic transmission mediates some aspects of alcohol-drinking behavior, but there is ambiguity in the literature with respect to the directions of these effects. Early studies showed that systemic administration of GABAAR agonists increased voluntary alcohol drinking, whereas GABAAR antagonists and benzodiazepine inverse agonists decreased alcohol drinking (Boyle et al. 1993; Rassnick et al. 1993). Infusion of both GABAAR agonists and antagonists into the nucleus accumbens (NAcc) suppressed alcohol drinking by nondependent rats (Hodge et al. 1995). Systemic administration of a GABABR agonist suppressed alcohol drinking by all rats, but alcohol-dependent rats were more sensitive to this effect, suggesting an up-regulation of GABABR function (Walker and Koob 2007). Other studies highlight a role for GABA circuitry in alcohol-drinking behavior, particularly in regions implicated in the negative reinforcing properties of the drug (i.e., extended amygdala). Hyytia and Koob (Hyytia and Koob 1995) found that injection of GABAAR antagonists in the three major regions of the extended amygdala suppressed alcohol drinking by nondependent rats, but this effect was most potent and selective for alcohol when infused into the CeA. Another study showed that antagonism of GABAARs in the BNST reversed decreases in alcohol drinking elicited by a D2 receptor antagonist infused into the ventral tegmental area (VTA) of alcohol-preferring (P) rats (Eiler and June 2007). Interestingly, infusion of a GABAAR agonist directly into the amygdala suppresses drinking by alcohol-dependent rats without affecting intake by nondependent controls (Roberts et al. 1996). Although there are considerable methodological differences between these studies, they suggest that (1) chronic alcohol produces neuroadaptations in GABAergic neurotransmission and changes sensitivity to GABAergic compounds, (2) GABAergic neurotransmission regulates alcohol drinking, and (3) in the case of excessive alcohol consumption by alcohol-dependent rats, the CeA is a strong candidate region for localization of these effects.

ACUTE ALCOHOL AUGMENTS GABAergic TRANSMISSION IN CeA

The acute effects of alcohol on GABAAR function has been extensively studied, mostly in in vitro brain slice preparations, an approach that allows multiple methods for detecting changes in presynaptic transmitter release (for a review, see Criswell and Breese 2005; Siggins et al. 2005; Weiner and Valenzuela 2006; Lovinger and Roberto 2011). Alcohol (1–100 mm) selectively enhances the function of GABAARs containing certain subunit compositions, but such findings have been inconsistent across laboratories testing heterologous systems (reviewed in Aguayo et al. 2002; Lovinger and Homanics 2007). Alcohol increases GABAergic synaptic transmission in the CeA (Roberto et al. 2003) and BLA (Zhu and Lovinger 2006) via increased presynaptic GABA release. Specifically, alcohol augments evoked inhibitory postsynaptic currents (IPSCs), decreases paired-pulse facilitation (PPF) of evoked IPSCs, and increases the frequency of miniature inhibitory postsynaptic currents (mIPSCs) (i.e., in Tetrodotoxin to eliminate action potential firing) in most CeA neurons, suggesting that alcohol increases vesicular GABA release.

Although the molecular mechanism(s) for alcohol effects have yet to be identified, we showed that activation of adenylyl cylase (AC) and/or protein kinase C (PKC) mediate GABAergic transmission in CeA synapses (Bajo et al. 2008; Cruz et al. 2011). In CeA slices of mice lacking PKCε or pretreated with a PKCε antagonist, the ability of acute alcohol to augment IPSCs is impaired (Bajo et al. 2008), suggesting that PKCε facilitates alcohol-elicited vesicular GABA release. Notably, basal GABA release is greater in CeA of PKCε knockout than wild-type (WT) mice, suggesting that under normal conditions in WT neurons, PKCε limits spontaneous GABA release. Therefore, PKCε serves at least two roles in the CeA: (i) limiting baseline GABA release, and (ii) facilitating alcohol-stimulated release of GABA.

The ability of alcohol to facilitate GABA neurotransmission may be limited by GABA feedback onto presynaptic GABABRs (Wan et al. 1996; Ariwodola and Weiner 2004). For example, acute alcohol facilitates GABAergic transmission in hippocampus (Wu and Saggau 1994; Wan et al. 1996; Ariwodola and Weiner 2004) and nucleus accumbens (Nie et al. 2000) only if GABAB receptors are blocked. However, in the CeA, GABAB receptor blockade is not required for the enhancement of IPSPs by acute alcohol nor does it potentiate this effect (Roberto et al. 2003). Thus, the involvement of GABAB receptors in alcohol-induced GABA release may depend on the presence of presynaptic GABAB receptors in certain brain regions (Ariwodola and Weiner 2004; Breese et al. 2005).

CHRONIC ALCOHOL EFFECTS ON GABAergic TRANSMISSION IN CeA

Our in vitro electrophysiological results show that chronic alcohol exposure augments GABA release in the CeA (Roberto et al. 2004a). Compared to alcohol-naïve controls, alcohol-dependent rats show larger baseline evoked GABAA-mediated IPSP/C amplitudes, smaller baseline PPF of evoked IPSCs, and higher baseline frequency of mIPSCs in CeA neurons. Interestingly, acute alcohol augments IPSCs, decreases PPF of IPSCs, and increases mIPSC frequency similarly in alcohol-dependent and alcohol-naïve rats, suggesting a lack of tolerance for these acute effects of alcohol (Roberto et al. 2004a). In vivo microdialysis studies indicate a fourfold increase of baseline dialysate GABA concentrations in the CeA of alcohol-dependent rats relative to naïve controls, as well as lack of tolerance for acute alcohol-induced increases in dialysate GABA levels (Roberto et al. 2004a), strongly suggesting that both acute and chronic alcohol alter presynaptic elements of GABAergic synapses in the CeA. Future studies need to determine the molecular mechanisms responsible for chronic alcohol-induced adaptations in CeA neurons and their behavioral implications in alcohol-dependent and/or alcohol-withdrawn organisms. These ongoing studies may elucidate the mechanism(s) underlying reductions in alcohol withdrawal hyperexcitability produced by GABAmimetic drugs (Ticku and Burch 1980; McCown et al. 1985; Breese et al. 2006; Roberto et al. 2008), and could impact treatment of pathological alcohol-drinking behaviors.

Chronic alcohol exposure produces tolerance to many behavioral effects of the drug, including the anxiolytic, sedative, ataxic, and positive reinforcing effects (Kumar et al. 2004, 2009). Chronic alcohol also produces physical and motivational dependence, and alcohol withdrawal is associated with increased neuronal excitability in several brain regions (but not CeA) (Kliethermes 2005; Weiner and Valenzuela 2006). Chronic alcohol effects may reflect, in part, compensatory adaptations to the facilitatory effects of alcohol on GABAergic synapses (Siggins et al. 2005; Weiner and Valenzuela 2006). We showed that evoked IPSCs in CeA slices from alcohol-dependent rats are significantly larger than those from naive controls (Roberto et al. 2004a). Some CeA neurons from alcohol-dependent rats also show increased mIPSC amplitudes relative to naïve rats, suggesting a postsynaptic effect of chronic alcohol (Roberto et al. 2004a). Substantial evidence suggests that alcohol-induced behavioral and neural adaptations are attributable to changes in GABAAR subunit composition rather than changes in the number of GABAARs (Morrow et al. 1992; Eckardt et al. 1998; Grobin et al. 1998; Papadeas et al. 2001; Kumar et al. 2004, 2009).

In alcohol-naïve rats, a GABABR antagonist increased the amplitude of evoked IPSCs and decreased PPF of IPSCs in CeA, suggesting tonic activation of presynaptic GABABRs (Roberto et al. 2008). Conversely, a GABAB agonist markedly depressed evoked IPSC amplitudes and increased PPF of IPSCs in the CeA of alcohol-naïve rats, indicating decreased presynaptic GABA release. These effects of GABABR agonists and antagonists were absent or greatly attenuated in the CeA of alcohol-dependent rats, suggesting chronic alcohol-induced down-regulation of the GABABR system that may explain the increased GABAergic tone observed in the CeA of dependent rats (Roberto et al. 2008). These alcohol dependence-induced neuroadaptations of the GABABR system also may account for chronic alcohol-induced changes in gabapentin effects on inhibitory transmission in CeA. Gabapentin, a structural analog of GABA (Sills 2006), increases the amplitudes of evoked IPSCs in CeA neurons from nondependent rats (an effect blocked by a GABABR antagonist), but decreases IPSC amplitudes in CeA of alcohol-dependent rats. Notably, gabapentin infused into the CeA reverses dependence-induced increases in operant alcohol responding, but tends to increase alcohol drinking by nondependent rats (Roberto et al. 2008).

ALCOHOL AND GLUTAMATERGIC TRANSMISSION IN AMYGDALA

Glutamate, the major excitatory neurotransmitter has long been implicated in the reinforcing actions of alcohol. Glutamate receptors include three major classes of ionotropic receptors (iGluRs), with varying ratios of selectivity for Na+, K+, and Ca2+. The iGluRs include AMPA (AMPARs), NMDA (NMDARs), and kainate receptors (KARs). Additionally, there are various subclasses of metabotropic glutamate receptors (mGluRs) that are G-protein-coupled receptors (GPCRs).

In contrast to its potentiating effects on GABA systems, alcohol generally inhibits glutamate neurotransmission in the brain (Lovinger and Roberto 2011). Dysregulation of glutamate systems may contribute to hyperexcitability and craving associated with alcohol withdrawal (Pulvirenti and Diana 2001). In the CeA of alcohol-preferring (P) rats, long-term consumption of high quantities of alcohol increases expression of mGluRs, NMDA receptor subunits, and a scaffolding protein regulating expression of these receptors in the cell membrane (Obara et al. 2009). Group II mGluRs also may block stress- and cue-induced reinstatement of alcohol-seeking behavior via neuronal activation in CeA or BNST (Zhao et al. 2006). Excitatory transmission in the CeA may also mediate some of the aversive aspects of withdrawal from abused drugs [e.g., morphine (Watanabe et al. 2002)].

Acamprosate, approved for treatment of alcoholic patients, modulates glutamate transmission via actions at NMDARs and/or mGluRs (Berton et al. 1998; Blednov and Harris 2008; Mann et al. 2008). Interestingly, acamprosate dampens the increased glutamate levels in abstinent alcoholics measured by magnetic resonance spectroscopy (Umhau et al. 2010) and reduces excessive alcohol drinking in alcoholics, presumably by reducing craving and negative affect (for a review, see Littleton 2007). AMPARs may be important in regulating relapse-like behaviors without playing a central role in alcohol consumption per se (Sanchis-Segura et al. 2006).

ACUTE ALCOHOL EFFECTS ON GLUTAMATERGIC TRANSMISSION IN CeA

We showed that acute alcohol (5–66 mm) decreases excitatory postsynaptic potentials (EPSPs) and currents (EPSCs) in the CeA, and that these effects are mediated by both NMDAR and non-NMDAR mechanisms (Roberto et al. 2004b). In contrast to alcohol effects on GABA release, the majority of studies indicate that acute alcohol either has no effect on or inhibits glutamate release (for a review, see Siggins et al. 2005) and inhibits NMDAR, AMPAR, and KAR function in some neuron types (for a review, see Lovinger and Roberto 2011).

CHRONIC ALCOHOL EFFECTS ON GLUTAMATERGIC TRANSMISSION IN CeA

Chronic alcohol exposure produces neuroadaptation in glutamatergic synaptic transmission. For example, acute alcohol decreases NMDAR-mediated EPSPs and EPSCs in the CeA of alcohol-dependent rats more than in alcohol-naïve controls. With local NMDA application, acute alcohol inhibits NMDA currents more in slices from alcohol-dependent rats, suggesting that alcohol dependence sensitizes NMDARs to alcohol (Roberto et al. 2004b, 2006). NMDARs containing the NR2B subunit are most sensitive to chronic alcohol exposure (Floyd et al. 2003; Carpenter-Hyland et al. 2004; Roberto et al. 2004b; Kash et al. 2009). Chronic alcohol increases NR2B mRNA and/or protein levels (Roberto et al. 2006; Kash et al. 2009) in the CeA and BNST, but not in other brain regions (Cebers et al. 1999; Floyd et al. 2003; Lack et al. 2005). It is not yet clear if increased NR2B subunit expression is the major driving force behind alcohol-induced increases in NMDAR function, or what molecular mechanisms underlie these subunit changes.

Microdialysis experiments in the amygdala show increases in glutamate release following chronic alcohol exposure (at 2–8 h withdrawal in CeA: Roberto et al. 2004b; at 24 h withdrawal in BLA: Lack et al. 2007). Our laboratory found that chronic alcohol exposure unmasks the ability of acute alcohol to increase presynaptic glutamate release in CeA (Roberto et al. 2004b), and that this effect persists 2 weeks into abstinence (Roberto et al. 2006). Collectively, these data suggest that multiple factors contribute to increased extracellular glutamate levels and increased glutamatergic transmission following chronic alcohol exposure and withdrawal.

CENTRAL AMYGDALA NEUROPEPTIDES AND ALCOHOL DEPENDENCE

Neuropeptides play prominent roles in regulating anxiety-like and alcohol-drinking behaviors in subjects that are either alcohol dependent, genetically vulnerable to developing excessive drinking (e.g., via selective breeding), repeatedly cycled through periods of alcohol withdrawal, and/or innately anxious. The CeA contains high concentrations of prostress (e.g., CRF) and antistress (e.g., NPY and nociceptin) neuropeptides, and many effects of these neuropeptides on anxiety- and alcohol-related behaviors have been localized to the CeA. Here we review the actions of a few neuropeptides on inhibitory transmission in the CeA, with focus on the CRF system that is heavily recruited during the transition from casual alcohol use to dependence (Koob 2010).

The CeA is the major output center of the amygdala, and about 95% of CeA neurons are medium spiny GABAergic neurons (McDonald 1982). The CeA is not a homogeneous neuroanatomical structure, and can be subdivided into lateral (CeL) and medial (CeM) aspects that differ in neuropeptide content, origin of afferents, and target sites of efferent projections (for a review, see Pitkanen et al. 2000). The CeL contains a much higher density of neuropeptides (e.g., dynorphin, CRF [Veening et al. 1984; Cassell et al. 1986; Shimada et al. 1989]) than the CeM, receives input from cortex and thalamus, and projects to the substantia innominata. In contrast, the CeM receives prominent inputs from other amygdaloid nuclei (especially glutamatergic afferents from BLA), and projects to effector regions such as hypothalamus and brain stem nuclei (Krettek and Price 1978). Both the CeL and CeM project to the BNST (Sun et al. 1991).

CRF AND ALCOHOL-RELATED BEHAVIOR

CRF plays a central role in arousal and hormonal, sympathetic, and behavioral responses to stress. The CeA, BNST, and BLA contain abundant CRF neurons and receptors (De Souza et al. 1984; Sakanaka et al. 1986). Hyperfunction of CRF systems in the CeA, BLA, and BNST produce increases in anxiety-like behavior (Sajdyk et al. 1999; Rainnie et al. 2004; Lee et al. 2008). Extracellular CRF levels in CeA are elevated following exposure to stress and development of alcohol dependence (Merlo Pich et al. 1995; Zorrilla et al. 2001), and alcohol withdrawal increases CRF synthesis and release in CeA (Funk et al. 2006; Sommer et al. 2008; Roberto et al. 2010b). Likewise, alcohol withdrawal increases extracellular CRF in BNST (Olive et al. 2002), and these increases are normalized by alcohol consumption. CRFR antagonists suppress dependence-induced increases in alcohol drinking during acute withdrawal and protracted abstinence (Valdez et al. 2002) and reverse increases in stress-induced anxiety during protracted abstinence (Valdez et al. 2003). CRF repeatedly administered into the CeA, BLA, or dorsal BNST exaggerates alcohol withdrawal-induced increases in anxiety-like behavior via CRF1Rs (Huang et al. 2010). Conversely, antagonism of CRFRs in the CeA blunts the increases in anxiety-like behavior in rats during withdrawal from chronic high-dose alcohol exposure (Rassnick et al. 1993).

CRF1R antagonists block the anxiogenic effects of many stressors (Arborelius et al. 2000; Zorrilla and Koob 2004). CRF1R antagonists also block increases in alcohol self-administration elicited by stressors and withdrawal (Hansson et al. 2006; Funk et al. 2007; Gehlert et al. 2007; Marinelli et al. 2007; Lowery et al. 2008). Chronic treatment with a CRF1R antagonist abolishes dependence-induced escalation of drinking in rats chronically exposed to high doses of alcohol (Roberto et al. 2010b). Likewise, (1) stressors and alcohol withdrawal increase CRF1R expression in limbic brain regions (Aguilar-Valles et al. 2005; Sommer et al. 2008); (2) rats bred for high alcohol preference show increased anxiety-like behavior and CRF1R levels (Ciccocioppo et al. 2006); and (3) CRF1R knockout mice show decreased anxiety-like behavior (Muller et al. 2003) and decreased drinking following withdrawal from chronic high-dose alcohol (Chu et al. 2007).

CRF EFFECTS ON SYNAPTIC TRANSMISSION IN CeA

We showed that CRF robustly increases GABAergic transmission in CeA of rats (Roberto et al. 2010b) and mice (Nie et al. 2004, 2009). CRF increases, and CRF1R antagonists decrease, presynaptic GABA release, suggesting tonic facilitation of GABA release by endogenous CRF. CRF1R antagonists and CRF1R knockouts also block the alcohol-induced augmentation of GABAergic transmission in CeA. Both CRF- and alcohol-induced facilitation of GABAergic transmission in CeA require the PKCε signaling pathway (Bajo et al. 2008). Alcohol-dependent rats show increased sensitivity to the effects of CRF and CRF1R antagonists on GABA release in CeA, suggesting up-regulation of the CRF-CRF1R system. These findings are further supported by increased CRF and CRF1R messenger RNA (mRNA) levels seen in the CeA of alcohol-dependent rats, and by reversal of dependence-induced elevations in amygdalar GABA dialysate by a CRF1R antagonist (Roberto et al. 2010b). CRF also increases GABAergic transmission in the BNST, likely via actions at postsynaptic CRF1Rs (Kash and Winder 2006; Francesconi et al. 2009).

NOCICEPTIN/ORPHANIN FQ AND ALCOHOL-RELATED BEHAVIOR

Nociceptin is an opioid-like peptide (Meunier et al. 1995; Reinscheid et al. 1995; Meunier 1997) that acts at opioid-like (NOP) receptors, although it does not bind to opioid receptors and opioids do not bind to NOP receptors (NOPRs). Nociceptin is abundantly expressed in the CeA and BNST (Neal et al. 1999) and is described as a functional CRF antagonist (Ciccocioppo et al. 2003). Nociceptin and other NOPR agonists have an anxiolytic-like profile in animal studies (Jenck et al. 1997, 2000). Nociceptin knockout mice show increased anxiety-like behavior (Koster et al. 1999) and are more sensitive to social stress (Ouagazzal et al. 2003).

Nociceptin suppresses alcohol drinking and prevents relapse-like behavior in rats (Ciccocioppo et al. 2004; Kuzmin et al. 2007). Nociceptin blocks reinstatement of alcohol-seeking behavior by cues predictive of alcohol availability and footshock stress (Martin-Fardon et al. 2000; Ciccocioppo et al. 2004), a behavior mediated by brain CRF systems (Ciccocioppo et al. 2004). Generally, rats bred for high alcohol intake show increased sensitivity to the suppressive effects of nociceptin on drinking and related behaviors (Economidou et al. 2008, 2011). Human alcoholics express decreased levels of mRNA for NOPR1 in the CeA (Kuzmin et al. 2007), suggesting that the CeA is a critical site for a role of nociceptin in alcoholism.

NOCICEPTIN AND SYNAPTIC TRANSMISSION IN CeA

We found that nociceptin dose-dependently and reversibly reduced GABAAR-mediated IPSCs in CeA (Roberto and Siggins 2006). Nociceptin increased PPF of evoked IPSCs and decreased the frequency of mIPSCs in CeA, suggesting decreased presynaptic GABA release. Notably, nociceptin both prevented (when applied before alcohol) and totally reversed (applied during alcohol) acute alcohol-induced increases in evoked IPSC amplitudes and mIPSC frequencies and decreases in PPF, thus preempting the usual alcohol-induced increase in GABA release in CeA. Further, the ability of nociceptin to decrease GABAergic transmission in CeA is augmented following alcohol dependence, suggesting that the nociceptin system in the CeA adapts during chronic alcohol exposure (Roberto and Siggins 2006).

NEUROPEPTIDE Y (NPY) AND ALCOHOL-RELATED BEHAVIOR

The amygdala contains abundant NPY fibers and receptors (Allen et al. 1984; de Quidt and Emson 1986; Dumont et al. 1993; Gustafson et al. 1997). NPY anxiolytic effects may involve both the CeA (Heilig et al. 1993) and BLA (Sajdyk et al. 1999). Rats selectively bred for high alcohol preference express low levels of NPY and NPY mRNA in CeA that are restored by voluntary alcohol intake (Pandey et al. 2005). Alcohol-withdrawn rats show increases in anxiety-like behavior and decreased amygdalar NPY, possibly owing to changes in histone acetylation (Roy and Pandey 2002; Zhao et al. 2007; Pandey et al. 2008).

NPY microinjection into the CeA suppresses alcohol consumption in alcohol-dependent and abstinent P rats (Gilpin et al. 2008a,b), and also in rats showing innately high anxiety-like behavior (Primeaux et al. 2006). Both postsynaptic Y1 and presynaptic Y2 receptors (Y1Rs and Y2Rs) are implicated in the effects of NPY on anxiety-like behavior and alcohol consumption. Early studies suggested roles for amygdalar Y1Rs (Heilig et al. 1993) and Y2Rs (Sajdyk et al. 2002) in anxiety-like behavior. Mouse studies indicate that Y1Rs mediate the suppressive effects of NPY on alcohol drinking (Thiele et al. 2002; Sparta et al. 2004; Eva et al. 2006). Likewise, acute stress and alcohol withdrawal increase amygdalar Y1R expression in rodents (Aguilar-Valles et al. 2005; Eva et al. 2006; Sommer et al. 2008).

NPY AND SYNAPTIC TRANSMISSION IN CeA

Our lab found that NPY in CeA prevents acute alcohol-induced increases in evoked IPSPs and mIPSC frequency (Gilpin et al. 2011) and PPF decreases, suggesting that NPY effects arise from decreased presynaptic GABA release. Tests with antagonists confirm the presynaptic site of action and suggest that NPY blocks alcohol effects via actions at presynaptic Y2Rs. NPY also normalizes alcohol dependence-induced increases in GABA release in CeA, suggesting that chronic exposure causes neuroadaptations in NPY systems that affect inhibitory transmission here. These results concur with findings that NPY modulates GABA release in BNST via activation of presynaptic Y2Rs (Kash and Winder 2006) and suggest that Y2Rs function as autoreceptors regulating NPY release (Chen et al. 1997), and also as heteroceptors regulating release of other neurotransmitters (Greber et al. 1994). NPY actions at postsynaptic Y1Rs appear to function as a “brake” on presynaptic Y2R-mediated reductions in GABA release, a result that may explain previous findings that intra-amygdala infusion of a Y1R antagonist decreases alcohol drinking in rats (Schroeder et al. 2003). These combined results support the hypothesis that NPY blocks stress-induced reinstatement of alcohol-seeking behavior via activation of inhibitory neurons in CeA (Cippitelli et al. 2010).

DISINHIBITION MODEL OF CeA OUTPUT

As noted above, most neurons in the CeA are GABAergic inhibitory projection neurons or interneurons that cotransmit GABA and one or more neuropeptides. Peptides that promote anxiety-like behavior and alcohol self-administration (e.g., CRF) generally increase, whereas peptides that reduce anxiety-like behavior and alcohol self-administration (e.g., nociceptin, NPY) decrease GABAergic transmission in CeA. In our slice preparation, we stimulate and record locally in the medial portion of the CeA, and recordings of GABAergic transmission reflect the activity of inhibitory interneurons or projection neurons (via collaterals) within CeA. Therefore, increases in GABAergic transmission within CeA following application of acute alcohol or CRF will inhibit the activity of GABAergic neurons projecting out of CeA. Conversely, decreases in GABAergic transmission in CeA neurons (e.g., following nociceptin or NPY application) will reduce inhibition of GABAergic neurons projecting out of CeA, thereby facilitating release of GABA in downstream targets (e.g., BNST, periaqueductal gray). Thus, increases or decreases in inhibitory output from the CeA to downstream effector regions may decrease or increase anxiety-like behavior, respectively (Pare et al. 2004; Davis et al. 2010; Tye et al. 2011). Furthermore, alcohol markedly affects excitatory transmission in CeA, particularly via NMDARs (Roberto et al. 2004b, 2006), lending at least partial buffering of alcohol effects on inhibitory transmission. Finally, alcohol may alter the release of local opioids (Lam et al. 2008), endocannabinoids (Roberto et al. 2010a), and/or galanin (Bajo et al. 2011) in CeA that in turn may increase GABA-mediated inhibition of downstream target areas. Other neuropeptides (e.g., substance P, vasopressin) are also likely to regulate the synaptic transmission within CeA.

CONCLUSIONS

The data summarized here support the idea that the CeA is a critical locus of neuroadaptation during the transition to alcohol dependence. Alcohol has strong and persistent effects, particularly on inhibitory transmission, in the CeA of alcohol-dependent animals. Neuropeptides present at high levels in the CeA profoundly alter inhibitory transmission, and potentially also excitatory transmission. The ability of these neuropeptides to affect neurotransmission in the CeA, either alone or in combination with alcohol, is often dysregulated in alcohol dependence. Although manipulation of many of these peptides affects alcohol drinking in alcohol-dependent but not nondependent animals, it is unsurprising that these neuropeptides affect basal neurotransmission in CeA of alcohol-naïve animals, especially because all these peptides when microinjected into CeA modulate anxiety-related behavior independent of alcohol exposure history. This point also enhances our understanding of why these neuropeptide systems are recruited and/or up-regulated during the transition to alcohol dependence, a dynamic disease defined largely by a negative emotional state in the absence of the drug. Finally, our electrophysiological data suggest that synaptic transmission and the special neuronal circuitry in the CeA may be an important point of convergence for the neuroadaptations that occur during the transition to alcohol dependence. Our understanding of this pivotal system as a “bellwether” target for therapeutic testing for anxiety and alcohol use disorders may be reliably predicted by drug effects on synaptic transmission in the CeA. Thus, we predict that most drugs that decrease GABAergic transmission in CeA neurons will be logical candidates for treatment of anxiety and/or alcohol use disorders.

ACKNOWLEDGMENTS

This work is supported by the National Institute of Alcohol Abuse and Alcoholism grants AA015566, AA06420, AA016985, AA017447, AA018400, and AA13498. This is manuscript number 21515 from The Scripps Research Institute.

Footnotes

Editors: R. Christopher Pierce and Paul J. Kenny

Additional Perspectives on Addiction available at www.perspectivesinmedicine.org

REFERENCES

  1. Aguayo LG, Peoples RW, Yeh HH, Yevenes GE 2002. GABA(A) receptors as molecular sites of ethanol action. Direct or indirect actions? Curr Top Med Chem 2: 869–885 [DOI] [PubMed] [Google Scholar]
  2. Aguilar-Valles A, Sanchez E, de Gortari P, Balderas I, Ramirez-Amaya V, Bermudez-Rattoni F, Joseph-Bravo P 2005. Analysis of the stress response in rats trained in the water-maze: Differential expression of corticotropin-releasing hormone, CRH-R1, glucocorticoid receptors and brain-derived neurotrophic factor in limbic regions. Neuroendocrinology 82: 306–319 [DOI] [PubMed] [Google Scholar]
  3. Allen YS, Roberts GW, Bloom SR, Crow TJ, Polak JM 1984. Neuropeptide Y in the stria terminalis: Evidence for an amygdalofugal projection. Brain Res 321: 357–362 [DOI] [PubMed] [Google Scholar]
  4. Arborelius L, Skelton KH, Thrivikraman KV, Plotsky PM, Schulz DW, Owens MJ 2000. Chronic administration of the selective corticotropin-releasing factor 1 receptor antagonist CP-154,526: Behavioral, endocrine and neurochemical effects in the rat. J Pharmacol Exp Ther 294: 588–597 [PubMed] [Google Scholar]
  5. Ariwodola OJ, Weiner JL 2004. Ethanol potentiation of GABAergic synaptic transmission may be self-limiting: Role of presynaptic GABA(B) receptors. J Neurosci 24: 10679–10686 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bajo M, Cruz MT, Siggins GR, Messing R, Roberto M 2008. Protein kinase C epsilon mediation of CRF- and ethanol-induced GABA release in central amygdala. Proc Natl Acad Sci 105: 8410–8415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bajo M, Madamba SG, Lu X, Sharkey LM, Bartfai T, Siggins GR 2011. Receptor subtype-dependent galanin actions on γ-aminobutyric acidergic neurotransmission and ethanol responses in the central amygdala. Addict Biol 10.1111/j.1369-1600.2011.00360.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Berton F, Francesconi WG, Madamba SG, Zieglgansberger W, Siggins GR 1998. Acamprosate enhances N-methyl-D-apartate receptor-mediated neurotransmission but inhibits presynaptic GABA(B) receptors in nucleus accumbens neurons. Alcohol Clin Exp Res 22: 183–191 [PubMed] [Google Scholar]
  9. Blednov YA, Harris RA 2008. Metabotropic glutamate receptor 5 (mGluR5) regulation of ethanol sedation, dependence and consumption: Relationship to acamprosate actions. Int J Neuropsychopharmacol 11: 775–793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Boyle AE, Segal R, Smith BR, Amit Z 1993. Bidirectional effects of GABAergic agonists and antagonists on maintenance of voluntary ethanol intake in rats. Pharmacol Biochem Behav 46: 179–182 [DOI] [PubMed] [Google Scholar]
  11. Breese GR, Overstreet DH, Knapp DJ 2005. Conceptual framework for the etiology of alcoholism: A “kindling”/stress hypothesis. Psychopharmacology (Berl) 178: 367–380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Breese GR, Criswell HE, Carta M, Dodson PD, Hanchar HJ, Khisti RT, Mameli M, Ming Z, Morrow AL, Olsen RW, et al. 2006. Basis of the gabamimetic profile of ethanol. Alcohol Clin Exp Res 30: 731–744 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Carpenter-Hyland EP, Woodward JJ, Chandler LJ 2004. Chronic ethanol induces synaptic but not extrasynaptic targeting of NMDA receptors. J Neurosci 24: 7859–7868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cassell MD, Gray TS, Kiss JZ 1986. Neuronal architecture in the rat central nucleus of the amygdala: A cytological, hodological, and immunocytochemical study. J Comp Neurol 246: 478–499 [DOI] [PubMed] [Google Scholar]
  15. Cebers G, Cebere A, Wagner A, Liljequist S 1999. Prolonged inhibition of glutamate reuptake down-regulates NMDA receptor functions in cultured cerebellar granule cells. J Neurochem 72: 2181–2190 [DOI] [PubMed] [Google Scholar]
  16. Chen X, DiMaggio DA, Han SP, Westfall TC 1997. Autoreceptor-induced inhibition of neuropeptide Y release from PC-12 cells is mediated by Y2 receptors. Am J Physiol 273: H1737–1744 [DOI] [PubMed] [Google Scholar]
  17. Chu K, Koob GF, Cole M, Zorrilla EP, Roberts AJ 2007. Dependence-induced increases in ethanol self-administration in mice are blocked by the CRF1 receptor antagonist antalarmin and by CRF1 receptor knockout. Pharmacol Biochem Behav 86: 813–821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Ciccocioppo R, Economidou D, Fedeli A, Massi M 2003. The nociceptin/orphanin FQ/NOP receptor system as a target for treatment of alcohol abuse: A review of recent work in alcohol-preferring rats. Physiol Behav 79: 121–128 [DOI] [PubMed] [Google Scholar]
  19. Ciccocioppo R, Economidou D, Fedeli A, Angeletti S, Weiss F, Heilig M, Massi M 2004. Attenuation of ethanol self-administration and of conditioned reinstatement of alcohol-seeking behaviour by the antiopioid peptide nociceptin/orphanin FQ in alcohol-preferring rats. Psychopharmacology (Berl) 172: 170–178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Ciccocioppo R, Economidou D, Cippitelli A, Cucculelli M, Ubaldi M, Soverchia L, Lourdusamy A, Massi M 2006. Genetically selected Marchigian Sardinian alcohol-preferring (msP) rats: An animal model to study the neurobiology of alcoholism. Addict Biol 11: 339–355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Ciocchi S, Herry C, Grenier F, Wolff SB, Letzkus JJ, Vlachos I, Ehrlich I, Sprengel R, Deisseroth K, Stadler MB, et al. 2010. Encoding of conditioned fear in central amygdala inhibitory circuits. Nature 468: 277–282 [DOI] [PubMed] [Google Scholar]
  22. Cippitelli A, Damadzic R, Hansson AC, Singley E, Sommer WH, Eskay R, Thorsell A, Heilig M 2010. Neuropeptide Y (NPY) suppresses yohimbine-induced reinstatement of alcohol seeking. Psychopharmacology (Berl) 208: 417–426 [DOI] [PubMed] [Google Scholar]
  23. Clarke TK, Treutlein J, Zimmermann US, Kiefer F, Skowronek MH, Rietschel M, Mann K, Schumann G 2008. HPA-axis activity in alcoholism: Examples for a gene-environment interaction. Addict Biol 13: 1–14 [DOI] [PubMed] [Google Scholar]
  24. Criswell HE, Breese GR 2005. A conceptualization of integrated actions of ethanol contributing to its GABAmimetic profile: A commentary. Neuropsychopharmacology 30: 1407–1425 [DOI] [PubMed] [Google Scholar]
  25. Cruz MT, Bajo M, Maragnoli ME, Tabakoff B, Siggins GR, Roberto M 2011. Type 7 adenylyl cyclase is involved in the ethanol and CRF sensitivity of GABAergic synapses in mouse central amygdala. Front Neurosci 4: 207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Davis M, Walker DL, Miles L, Grillon C 2010. Phasic vs sustained fear in rats and humans: Role of the extended amygdala in fear vs anxiety. Neuropsychopharmacology 35: 105–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. de Quidt ME, Emson PC 1986. Distribution of neuropeptide Y-like immunoreactivity in the rat central nervous system—II. Immunohistochemical analysis. Neuroscience 18: 545–618 [DOI] [PubMed] [Google Scholar]
  28. De Souza EB, Perrin MH, Insel TR, Rivier J, Vale WW, Kuhar MJ 1984. Corticotropin-releasing factor receptors in rat forebrain: Autoradiographic identification. Science 224: 1449–1451 [DOI] [PubMed] [Google Scholar]
  29. Dumont Y, Fournier A, St-Pierre S, Quirion R 1993. Comparative characterization and autoradiographic distribution of neuropeptide Y receptor subtypes in the rat brain. J Neurosci 13: 73–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Eckardt MJ, File SE, Gessa GL, Grant KA, Guerri C, Hoffman PL, Kalant H, Koob GF, Li TK, Tabakoff B 1998. Effects of moderate alcohol consumption on the central nervous system. Alcohol Clin Exp Res 22: 998–1040 [DOI] [PubMed] [Google Scholar]
  31. Economidou D, Hansson AC, Weiss F, Terasmaa A, Sommer WH, Cippitelli A, Fedeli A, Martin-Fardon R, Massi M, Ciccocioppo R, et al. 2008. Dysregulation of nociceptin/orphanin FQ activity in the amygdala is linked to excessive alcohol drinking in the rat. Biol Psychiatry 64: 211–218 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Economidou D, Cippitelli A, Stopponi S, Braconi S, Clementi S, Ubaldi M, Martin-Fardon R, Weiss F, Massi M, Ciccocioppo R 2011. Activation of brain NOP receptors attenuates acute and protracted alcohol withdrawal symptoms in the rat. Alcohol Clin Exp Res 35: 747–755 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Eiler WJ II, June HL 2007. Blockade of GABA(A) receptors within the extended amygdala attenuates D(2) regulation of alcohol-motivated behaviors in the ventral tegmental area of alcohol-preferring (P) rats. Neuropharmacology 52: 1570–1579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Eva C, Serra M, Mele P, Panzica G, Oberto A 2006. Physiology and gene regulation of the brain NPY Y1 receptor. Front Neuroendocrinol 27: 308–339 [DOI] [PubMed] [Google Scholar]
  35. Floyd DW, Jung KY, McCool BA 2003. Chronic ethanol ingestion facilitates N-methyl-D-aspartate receptor function and expression in rat lateral/basolateral amygdala neurons. J Pharmacol Exp Ther 307: 1020–1029 [DOI] [PubMed] [Google Scholar]
  36. Francesconi W, Berton F, Koob GF, Sanna PP 2009. Intrinsic neuronal plasticity in the juxtacapsular nucleus of the bed nuclei of the stria terminalis (jcBNST). Prog Neuropsychopharmacol Biol Psychiatry 33: 1347–1355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Funk CK, O’Dell LE, Crawford EF, Koob GF 2006. Corticotropin-releasing factor within the central nucleus of the amygdala mediates enhanced ethanol self-administration in withdrawn, ethanol-dependent rats. J Neurosci 26: 11324–11332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Funk CK, Zorrilla EP, Lee MJ, Rice KC, Koob GF 2007. Corticotropin-releasing factor 1 antagonists selectively reduce ethanol self-administration in ethanol-dependent rats. Biol Psychiatry 61: 78–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Gehlert DR, Cippitelli A, Thorsell A, Le AD, Hipskind PA, Hamdouchi C, Lu J, Hembre EJ, Cramer J, Song M, et al. 2007. 3-(4-Chloro-2-morpholin-4-yl-thiazol-5-yl)-8-(1-ethylpropyl)-2,6-dimethyl-imidazo [1,2-b]pyridazine: A novel brain-penetrant, orally available corticotropin-releasing factor receptor 1 antagonist with efficacy in animal models of alcoholism. J Neurosci 27: 2718–2726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Gilpin NW, Misra K, Koob GF 2008a. Neuropeptide Y in the central nucleus of the amygdala suppresses dependence-induced increases in alcohol drinking. Pharmacol Biochem Behav 90: 475–480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Gilpin NW, Stewart RB, Badia-Elder NE 2008b. Neuropeptide Y administration into the amygdala suppresses ethanol drinking in alcohol-preferring (P) rats following multiple deprivations. Pharmacol Biochem Behav 90: 470–474 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Gilpin NW, Misra K, Herman MA, Cruz MT, Koob GF, Roberto M 2011. Neuropeptide Y opposes alcohol effects on γ-aminobutyric acid release in amygdala and blocks the transition to alcohol dependence. Biol Psychiatry 69: 1091–1099 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Greber S, Schwarzer C, Sperk G 1994. Neuropeptide Y inhibits potassium-stimulated glutamate release through Y2 receptors in rat hippocampal slices in vitro. Br J Pharmacol 113: 737–740 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Grobin AC, Matthews DB, Devaud LL, Morrow AL 1998. The role of GABAA receptors in the acute and chronic effects of ethanol. Psychopharmacology (Berl) 139: 2–19 [DOI] [PubMed] [Google Scholar]
  45. Gustafson EL, Smith KE, Durkin MM, Walker MW, Gerald C, Weinshank R, Branchek TA 1997. Distribution of the neuropeptide Y Y2 receptor mRNA in rat central nervous system. Brain Res Mol Brain Res 46: 223–235 [DOI] [PubMed] [Google Scholar]
  46. Hansson AC, Cippitelli A, Sommer WH, Fedeli A, Bjork K, Soverchia L, Terasmaa A, Massi M, Heilig M, Ciccocioppo R 2006. Variation at the rat Crhr1 locus and sensitivity to relapse into alcohol seeking induced by environmental stress. Proc Natl Acad Sci 103: 15236–15241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Heilig M, McLeod S, Brot M, Heinrichs SC, Menzaghi F, Koob GF, Britton KT 1993. Anxiolytic-like action of neuropeptide Y: Mediation by Y1 receptors in amygdala, and dissociation from food intake effects. Neuropsychopharmacology 8: 357–363 [DOI] [PubMed] [Google Scholar]
  48. Heilig M, Egli M, Crabbe JC, Becker HC 2010. Acute withdrawal, protracted abstinence and negative affect in alcoholism: Are they linked? Addict Biol 15: 169–184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Heimer L, Alheid G 1991. Piecing together the puzzle of basal forebrain anatomy. In The basal forebrain: Anatomy to function (ed. Napier TC, Kalivas PW, Hanin I), Advances in Experimental Medicine and Biology, Vol. 295, pp. 1–42 Plenum, New York: [DOI] [PubMed] [Google Scholar]
  50. Hodge CW, Chappelle AM, Samson HH 1995. GABAergic transmission in the nucleus accumbens is involved in the termination of ethanol self-administration in rats. Alcohol Clin Exp Res 19: 1486–1493 [DOI] [PubMed] [Google Scholar]
  51. Huang MM, Overstreet DH, Knapp DJ, Angel R, Wills TA, Navarro M, Rivier J, Vale W, Breese GR 2010. Corticotropin-releasing factor (CRF) sensitization of ethanol withdrawal-induced anxiety-like behavior is brain site specific and mediated by CRF-1 receptors: Relation to stress-induced sensitization. J Pharmacol Exp Ther 332: 298–307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hyytia P, Koob GF 1995. GABAA receptor antagonism in the extended amygdala decreases ethanol self-administration in rats. Eur J Pharmacol 283: 151–159 [DOI] [PubMed] [Google Scholar]
  53. Jenck F, Moreau JL, Martin JR, Kilpatrick GJ, Reinscheid RK, Monsma FJ Jr, Nothacker HP, Civelli O 1997. Orphanin FQ acts as an anxiolytic to attenuate behavioral responses to stress. Proc Natl Acad Sci 94: 14854–14858 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Jenck F, Ouagazzal AM, Pauly-Evers M, Moreau JL 2000. OrphaninFQ: Role in behavioral fear responses and vulnerability to stress? Mol Psychiatry 5: 572–574 [DOI] [PubMed] [Google Scholar]
  55. Kash TL, Winder DG 2006. Neuropeptide Y and corticotropin-releasing factor bi-directionally modulate inhibitory synaptic transmission in the bed nucleus of the stria terminalis. Neuropharmacology 51: 1013–1022 [DOI] [PubMed] [Google Scholar]
  56. Kash TL, Baucum AJ 2nd, Conrad KL, Colbran RJ, Winder DG 2009. Alcohol exposure alters NMDAR function in the bed nucleus of the stria terminalis. Neuropsychopharmacology 34: 2420–2429 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kiefer F, Wiedemann K 2004. Neuroendocrine pathways of addictive behaviour. Addict Biol 9: 205–212 [DOI] [PubMed] [Google Scholar]
  58. Kliethermes CL 2005. Anxiety-like behaviors following chronic ethanol exposure. Neurosci Biobehav Rev 28: 837–850 [DOI] [PubMed] [Google Scholar]
  59. Koob GF 2003. Alcoholism: Allostasis and beyond. Alcohol Clin Exp Res 27: 232–243 [DOI] [PubMed] [Google Scholar]
  60. Koob GF 2008. A role for brain stress systems in addiction. Neuron 59: 11–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Koob GF 2009. Brain stress systems in the amygdala and addiction. Brain Res 1293: 61–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Koob GF 2010. The role of CRF and CRF-related peptides in the dark side of addiction. Brain Res 1314: 3–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Koob GF, Volkow ND 2010. Neurocircuitry of addiction. Neuropsychopharmacology 35: 217–238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Koster A, Montkowski A, Schulz S, Stube EM, Knaudt K, Jenck F, Moreau JL, Nothacker HP, Civelli O, Reinscheid RK 1999. Targeted disruption of the orphanin FQ/nociceptin gene increases stress susceptibility and impairs stress adaptation in mice. Proc Natl Acad Sci 96: 10444–10449 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Krettek JE, Price JL 1978. Amygdaloid projections to subcortical structures within the basal forebrain and brainstem in the rat and cat. J Comp Neurol 178: 225–254 [DOI] [PubMed] [Google Scholar]
  66. Kumar S, Fleming RL, Morrow AL 2004. Ethanol regulation of γ-aminobutyric acid A receptors: Genomic and nongenomic mechanisms. Pharmacol Ther 101: 211–226 [DOI] [PubMed] [Google Scholar]
  67. Kumar S, Porcu P, Werner DF, Matthews DB, Diaz-Granados JL, Helfand RS, Morrow AL 2009. The role of GABAA receptors in the acute and chronic effects of ethanol: A decade of progress. Psychopharmacology (Berl) 205: 529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Kuzmin A, Kreek MJ, Bakalkin G, Liljequist S 2007. The nociceptin/orphanin FQ receptor agonist Ro 64–6198 reduces alcohol self-administration and prevents relapse-like alcohol drinking. Neuropsychopharmacology 32: 902–910 [DOI] [PubMed] [Google Scholar]
  69. Lack AK, Floyd DW, McCool BA 2005. Chronic ethanol ingestion modulates proanxiety factors expressed in rat central amygdala. Alcohol 36: 83–90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Lack AK, Diaz MR, Chappell A, DuBois DW, McCool BA 2007. Chronic ethanol and withdrawal differentially modulate pre- and postsynaptic function at glutamatergic synapses in rat basolateral amygdala. J Neurophysiol 98: 3185–3196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Lam MP, Marinelli PW, Bai L, Gianoulakis C 2008. Effects of acute ethanol on opioid peptide release in the central amygdala: An in vivo microdialysis study. Psychopharmacology (Berl) 201: 261–271 [DOI] [PubMed] [Google Scholar]
  72. Lee Y, Fitz S, Johnson PL, Shekhar A 2008. Repeated stimulation of CRF receptors in the BNST of rats selectively induces social but not panic-like anxiety. Neuropsychopharmacology 33: 2586–2594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Littleton JM 2007. Acamprosate in alcohol dependence: Implications of a unique mechanism of action. J Addict Med 1: 115–125 [DOI] [PubMed] [Google Scholar]
  74. Lovinger DM, Homanics GE 2007. Tonic for what ails us? High-affinity GABAA receptors and alcohol. Alcohol 41: 139–143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Lovinger DM, Roberto M 2011. Synaptic effects induced by alcohol. Curr Top Behav Neurosci 10.1007/7854-2011-143 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Lowery EG, Sparrow AM, Breese GR, Knapp DJ, Thiele TE 2008. The CRF-1 receptor antagonist, CP-154,526, attenuates stress-induced increases in ethanol consumption by BALB/cJ mice. Alcohol Clin Exp Res 32: 240–248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Mann K, Kiefer F, Spanagel R, Littleton J 2008. Acamprosate: Recent findings and future research directions. Alcohol Clin Exp Res 32: 1105–1110 [DOI] [PubMed] [Google Scholar]
  78. Marinelli PW, Funk D, Juzytsch W, Harding S, Rice KC, Shaham Y, Le AD 2007. The CRF1 receptor antagonist antalarmin attenuates yohimbine-induced increases in operant alcohol self-administration and reinstatement of alcohol seeking in rats. Psychopharmacology (Berl) 195: 345–355 [DOI] [PubMed] [Google Scholar]
  79. Martin-Fardon R, Ciccocioppo R, Massi M, Weiss F 2000. Nociceptin prevents stress-induced ethanol- but not cocaine-seeking behavior in rats. Neuroreport 11: 1939–1943 [DOI] [PubMed] [Google Scholar]
  80. McCown TJ, Frye GD, Breese GR 1985. Evidence for site specific ethanol actions in the CNS. Alcohol Drug Res 6: 423–429 [PubMed] [Google Scholar]
  81. McDonald AJ 1982. Cytoarchitecture of the central amygdaloid nucleus of the rat. J Comp Neurol 208: 401–418 [DOI] [PubMed] [Google Scholar]
  82. Merlo Pich E, Lorang M, Yeganeh M, Rodriguez de Fonseca F, Raber J, Koob GF, Weiss F 1995. Increase of extracellular corticotropin-releasing factor-like immunoreactivity levels in the amygdala of awake rats during restraint stress and ethanol withdrawal as measured by microdialysis. J Neurosci 15: 5439–5447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Meunier JC 1997. Nociceptin/orphanin FQ and the opioid receptor-like ORL1 receptor. Eur J Pharmacol 340: 1–15 [DOI] [PubMed] [Google Scholar]
  84. Meunier JC, Mollereau C, Toll L, Suaudeau C, Moisand C, Alvinerie P, Butour JL, Guillemot JC, Ferrara P, Monsarrat B, et al. 1995. Isolation and structure of the endogenous agonist of opioid receptor-like ORL1 receptor. Nature 377: 532–535 [DOI] [PubMed] [Google Scholar]
  85. Morrow AL, Herbert JS, Montpied P 1992. Differential effects of chronic ethanol administration on GABAA receptor α1 and α6 subunit mRNA levels in rat cerebellum. Mol Cell Neurosci 3: 251–258 [DOI] [PubMed] [Google Scholar]
  86. Muller MB, Zimmermann S, Sillaber I, Hagemeyer TP, Deussing JM, Timpl P, Kormann MS, Droste SK, Kuhn R, Reul JM, et al. 2003. Limbic corticotropin-releasing hormone receptor 1 mediates anxiety-related behavior and hormonal adaptation to stress. Nat Neurosci 6: 1100–1107 [DOI] [PubMed] [Google Scholar]
  87. Neal CR Jr, Mansour A, Reinscheid R, Nothacker HP, Civelli O, Watson SJ Jr 1999. Localization of orphanin FQ (nociceptin) peptide and messenger RNA in the central nervous system of the rat. J Comp Neurol 406: 503–547 [PubMed] [Google Scholar]
  88. Nie Z, Madamba SG, Siggins GR 2000. Ethanol enhances γ-aminobutyric acid responses in a subpopulation of nucleus accumbens neurons: Role of metabotropic glutamate receptors. J Pharmacol Exp Ther 293: 654–661 [PubMed] [Google Scholar]
  89. Nie Z, Schweitzer P, Roberts AJ, Madamba SG, Moore SD, Siggins GR 2004. Ethanol augments GABAergic transmission in the central amygdala via CRF1 receptors. Science 303: 1512–1514 [DOI] [PubMed] [Google Scholar]
  90. Nie Z, Zorrilla EP, Madamba SG, Rice KC, Roberto M, Siggins GR 2009. Presynaptic CRF1 receptors mediate the ethanol enhancement of GABAergic transmission in the mouse central amygdala. Sci World J 9: 68–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Obara I, Bell RL, Goulding SP, Reyes CM, Larson LA, Ary AW, Truitt WA, Szumlinski KK 2009. Differential effects of chronic ethanol consumption and withdrawal on homer/glutamate receptor expression in subregions of the accumbens and amygdala of P rats. Alcohol Clin Exp Res 33: 1924–1934 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Olive MF, Koenig HN, Nannini MA, Hodge CW 2002. Elevated extracellular CRF levels in the bed nucleus of the stria terminalis during ethanol withdrawal and reduction by subsequent ethanol intake. Pharmacol Biochem Behav 72: 213–220 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Ouagazzal AM, Moreau JL, Pauly-Evers M, Jenck F 2003. Impact of environmental housing conditions on the emotional responses of mice deficient for nociceptin/orphanin FQ peptide precursor gene. Behav Brain Res 144: 111–117 [DOI] [PubMed] [Google Scholar]
  94. Pandey SC, Zhang H, Roy A, Xu T 2005. Deficits in amygdaloid cAMP-responsive element-binding protein signaling play a role in genetic predisposition to anxiety and alcoholism. J Clin Invest 115: 2762–2773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Pandey SC, Ugale R, Zhang H, Tang L, Prakash A 2008. Brain chromatin remodeling: A novel mechanism of alcoholism. J Neurosci 28: 3729–3737 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Papadeas S, Grobin AC, Morrow AL 2001. Chronic ethanol consumption differentially alters GABAA receptor α1 and α4 subunit peptide expression and GABAA receptor-mediated 36Cl uptake in mesocorticolimbic regions of rat brain. Alcohol Clin Exp Res 25: 1270–1275 [PubMed] [Google Scholar]
  97. Pare D, Quirk GJ, Ledoux JE 2004. New vistas on amygdala networks in conditioned fear. J Neurophysiol 92: 1–9 [DOI] [PubMed] [Google Scholar]
  98. Phelps EA, LeDoux JE 2005. Contributions of the amygdala to emotion processing: From animal models to human behavior. Neuron 48: 175–187 [DOI] [PubMed] [Google Scholar]
  99. Pitkanen A, Pikkarainen M, Nurminen N, Ylinen A 2000. Reciprocal connections between the amygdala and the hippocampal formation, perirhinal cortex, and postrhinal cortex in rat. A review. Ann NY Acad Sci 911: 369–391 [DOI] [PubMed] [Google Scholar]
  100. Primeaux SD, Wilson SP, Bray GA, York DA, Wilson MA 2006. Overexpression of neuropeptide Y in the central nucleus of the amygdala decreases ethanol self-administration in “anxious” rats. Alcohol Clin Exp Res 30: 791–801 [DOI] [PubMed] [Google Scholar]
  101. Pulvirenti L, Diana M 2001. Drug dependence as a disorder of neural plasticity: Focus on dopamine and glutamate. Rev Neurosci 12: 141–158 [DOI] [PubMed] [Google Scholar]
  102. Quirk GJ, Mueller D 2008. Neural mechanisms of extinction learning and retrieval. Neuropsychopharmacology 33: 56–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Rainnie DG, Bergeron R, Sajdyk TJ, Patil M, Gehlert DR, Shekhar A 2004. Corticotrophin releasing factor-induced synaptic plasticity in the amygdala translates stress into emotional disorders. J Neurosci 24: 3471–3479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Rassnick S, Heinrichs SC, Britton KT, Koob GF 1993. Microinjection of a corticotropin-releasing factor antagonist into the central nucleus of the amygdala reverses anxiogenic-like effects of ethanol withdrawal. Brain Res 605: 25–32 [DOI] [PubMed] [Google Scholar]
  105. Reinscheid RK, Nothacker HP, Bourson A, Ardati A, Henningsen RA, Bunzow JR, Grandy DK, Langen H, Monsma FJ Jr, Civelli O 1995. Orphanin FQ: A neuropeptide that activates an opioidlike G protein-coupled receptor. Science 270: 792–794 [DOI] [PubMed] [Google Scholar]
  106. Roberto M, Siggins GR 2006. Nociceptin/orphanin FQ presynaptically decreases GABAergic transmission and blocks the ethanol-induced increase of GABA release in central amygdala. Proc Natl Acad Sci 103: 9715–9720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Roberto M, Madamba SG, Moore SD, Tallent MK, Siggins GR 2003. Ethanol increases GABAergic transmission at both pre- and postsynaptic sites in rat central amygdala neurons. Proc Natl Acad Sci 100: 2053–2058 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Roberto M, Madamba SG, Stouffer DG, Parsons LH, Siggins GR 2004a. Increased GABA release in the central amygdala of ethanol-dependent rats. J Neurosci 24: 10159–10166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Roberto M, Schweitzer P, Madamba SG, Stouffer DG, Parsons LH, Siggins GR 2004b. Acute and chronic ethanol alter glutamatergic transmission in rat central amygdala: An in vitro and in vivo analysis. J Neurosci 24: 1594–1603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Roberto M, Bajo M, Crawford E, Madamba SG, Siggins GR 2006. Chronic ethanol exposure and protracted abstinence alter NMDA receptors in central amygdala. Neuropsychopharmacology 31: 988–996 [DOI] [PubMed] [Google Scholar]
  111. Roberto M, Gilpin NW, O’Dell LE, Cruz MT, Morse AC, Siggins GR, Koob GF 2008. Cellular and behavioral interactions of gabapentin with alcohol dependence. J Neurosci 28: 5762–5771 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Roberto M, Cruz M, Bajo M, Siggins GR, Parsons LH, Schweitzer P 2010a. The endocannabinoid system tonically regulates inhibitory transmission and depresses the effect of ethanol in central amygdala. Neuropsychopharmacology 35: 1962–1972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Roberto M, Cruz MT, Gilpin NW, Sabino V, Schweitzer P, Bajo M, Cottone P, Madamba SG, Stouffer DG, Zorrilla EP, et al. 2010b. Corticotropin releasing factor-induced amygdala γ-aminobutyric acid release plays a key role in alcohol dependence. Biol Psychiatry 67: 831–839 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Roberts AJ, Cole M, Koob GF 1996. Intra-amygdala muscimol decreases operant ethanol self-administration in dependent rats. Alcohol Clin Exp Res 20: 1289–1298 [DOI] [PubMed] [Google Scholar]
  115. Roy A, Pandey SC 2002. The decreased cellular expression of neuropeptide Y protein in rat brain structures during ethanol withdrawal after chronic ethanol exposure. Alcohol Clin Exp Res 26: 796–803 [PubMed] [Google Scholar]
  116. Sajdyk TJ, Vandergriff MG, Gehlert DR 1999. Amygdalar neuropeptide Y Y1 receptors mediate the anxiolytic-like actions of neuropeptide Y in the social interaction test. Eur J Pharmacol 368: 143–147 [DOI] [PubMed] [Google Scholar]
  117. Sajdyk TJ, Schober DA, Gehlert DR 2002. Neuropeptide Y receptor subtypes in the basolateral nucleus of the amygdala modulate anxiogenic responses in rats. Neuropharmacology 43: 1165–1172 [DOI] [PubMed] [Google Scholar]
  118. Sakanaka M, Shibasaki T, Lederis K 1986. Distribution and efferent projections of corticotropin-releasing factor-like immunoreactivity in the rat amygdaloid complex. Brain Res 382: 213–238 [DOI] [PubMed] [Google Scholar]
  119. Sanchis-Segura C, Borchardt T, Vengeliene V, Zghoul T, Bachteler D, Gass P, Sprengel R, Spanagel R 2006. Involvement of the AMPA receptor GluR-C subunit in alcohol-seeking behavior and relapse. J Neurosci 26: 1231–1238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Schroeder JP, Olive F, Koenig H, Hodge CW 2003. Intra-amygdala infusion of the NPY Y1 receptor antagonist BIBP 3226 attenuates operant ethanol self-administration. Alcohol Clin Exp Res 27: 1884–1891 [DOI] [PubMed] [Google Scholar]
  121. Shimada S, Inagaki S, Kubota Y, Ogawa N, Shibasaki T, Takagi H 1989. Coexistence of peptides (corticotropin releasing factor/neurotensin and substance P/somatostatin) in the bed nucleus of the stria terminalis and central amygdaloid nucleus of the rat. Neuroscience 30: 377–383 [DOI] [PubMed] [Google Scholar]
  122. Siggins GR, Roberto M, Nie Z 2005. The tipsy terminal: Presynaptic effects of ethanol. Pharmacol Ther 107: 80–98 [DOI] [PubMed] [Google Scholar]
  123. Sills GJ 2006. The mechanisms of action of gabapentin and pregabalin. Curr Opin Pharmacol 6: 108–113 [DOI] [PubMed] [Google Scholar]
  124. Sommer WH, Rimondini R, Hansson AC, Hipskind PA, Gehlert DR, Barr CS, Heilig MA 2008. Upregulation of voluntary alcohol intake, behavioral sensitivity to stress, and amygdala crhr1 expression following a history of dependence. Biol Psychiatry 63: 139–145 [DOI] [PubMed] [Google Scholar]
  125. Sparta DR, Fee JR, Hayes DM, Knapp DJ, MacNeil DJ, Thiele TE 2004. Peripheral and central administration of a selective neuropeptide Y Y1 receptor antagonist suppresses ethanol intake by C57BL/6J mice. Alcohol Clin Exp Res 28: 1324–1330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Sun N, Cassell MD 1993. Intrinsic GABAergic neurons in the rat central extended amygdala. J Comp Neurol 330: 381–404 [DOI] [PubMed] [Google Scholar]
  127. Sun N, Roberts L, Cassell MD 1991. Rat central amygdaloid nucleus projections to the bed nucleus of the stria terminalis. Brain Res Bull 27: 651–662 [DOI] [PubMed] [Google Scholar]
  128. Thiele TE, Koh MT, Pedrazzini T 2002. Voluntary alcohol consumption is controlled via the neuropeptide Y Y1 receptor. J Neurosci 22: RC208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Ticku MK, Burch T 1980. Alterations in γ-aminobutyric acid receptor sensitivity following acute and chronic ethanol treatments. J Neurochem 34: 417–423 [DOI] [PubMed] [Google Scholar]
  130. Tye KM, Prakash R, Kim SY, Fenno LE, Grosenick L, Zarabi H, Thompson KR, Gradinaru V, Ramakrishnan C, Deisseroth K 2011. Amygdala circuitry mediating reversible and bidirectional control of anxiety. Nature 471: 358–362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Umhau JC, Momenan R, Schwandt ML, Singley E, Lifshitz M, Doty L, Adams LJ, Vengeliene V, Spanagel R, Zhang Y, et al. 2010. Effect of acamprosate on magnetic resonance spectroscopy measures of central glutamate in detoxified alcohol-dependent individuals: A randomized controlled experimental medicine study. Arch Gen Psychiatry 67: 1069–1077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Valdez GR, Roberts AJ, Chan K, Davis H, Brennan M, Zorrilla EP, Koob GF 2002. Increased ethanol self-administration and anxiety-like behavior during acute ethanol withdrawal and protracted abstinence: Regulation by corticotropin-releasing factor. Alcohol Clin Exp Res 26: 1494–1501 [DOI] [PubMed] [Google Scholar]
  133. Valdez GR, Zorrilla EP, Roberts AJ, Koob GF 2003. Antagonism of corticotropin-releasing factor attenuates the enhanced responsiveness to stress observed during protracted ethanol abstinence. Alcohol 29: 55–60 [DOI] [PubMed] [Google Scholar]
  134. Veening JG, Swanson LW, Sawchenko PE 1984. The organization of projections from the central nucleus of the amygdala to brainstem sites involved in central autonomic regulation: A combined retrograde transport-immunohistochemical study. Brain Res 303: 337–357 [DOI] [PubMed] [Google Scholar]
  135. Veinante P, Freund-Mercier MJ 1998. Intrinsic and extrinsic connections of the rat central extended amygdala: An in vivo electrophysiological study of the central amygdaloid nucleus. Brain Res 794: 188–198 [DOI] [PubMed] [Google Scholar]
  136. Walker BM, Koob GF 2007. The γ-aminobutyric acid-B receptor agonist baclofen attenuates responding for ethanol in ethanol-dependent rats. Alcohol Clin Exp Res 31: 11–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Wan FJ, Berton F, Madamba SG, Francesconi W, Siggins GR 1996. Low ethanol concentrations enhance GABAergic inhibitory postsynaptic potentials in hippocampal pyramidal neurons only after block of GABAB receptors. Proc Natl Acad Sci 93: 5049–5054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Watanabe T, Nakagawa T, Yamamoto R, Maeda A, Minami M, Satoh M 2002. Involvement of glutamate receptors within the central nucleus of the amygdala in naloxone-precipitated morphine withdrawal-induced conditioned place aversion in rats. Jpn J Pharmacol 88: 399–406 [DOI] [PubMed] [Google Scholar]
  139. Weiner JL, Valenzuela CF 2006. Ethanol modulation of GABAergic transmission: The view from the slice. Pharmacol Ther 111: 533–554 [DOI] [PubMed] [Google Scholar]
  140. Weller KL, Smith DA 1982. Afferent connections to the bed nucleus of the stria terminalis. Brain Res 232: 255–270 [DOI] [PubMed] [Google Scholar]
  141. Wu LG, Saggau P 1994. Presynaptic calcium is increased during normal synaptic transmission and paired-pulse facilitation, but not in long-term potentiation in area CA1 of hippocampus. J Neurosci 14: 645–654 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Zhao Y, Dayas CV, Aujla H, Baptista MA, Martin-Fardon R, Weiss F 2006. Activation of group II metabotropic glutamate receptors attenuates both stress and cue-induced ethanol-seeking and modulates c-fos expression in the hippocampus and amygdala. J Neurosci 26: 9967–9974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Zhao Y, Weiss F, Zorrilla EP 2007. Remission and resurgence of anxiety-like behavior across protracted withdrawal stages in ethanol-dependent rats. Alcohol Clin Exp Res 31: 1505–1515 [DOI] [PubMed] [Google Scholar]
  144. Zhu PJ, Lovinger DM 2006. Ethanol potentiates GABAergic synaptic transmission in a postsynaptic neuron/synaptic bouton preparation from basolateral amygdala. J Neurophysiol 96: 433–441 [DOI] [PubMed] [Google Scholar]
  145. Zorrilla EP, Koob GF 2004. The therapeutic potential of CRF1 antagonists for anxiety. Expert Opin Investig Drugs 13: 799–828 [DOI] [PubMed] [Google Scholar]
  146. Zorrilla EP, Valdez GR, Weiss F 2001. Changes in levels of regional CRF-like-immunoreactivity and plasma corticosterone during protracted drug withdrawal in dependent rats. Psychopharmacology (Berl) 158: 374–381 [DOI] [PubMed] [Google Scholar]

Articles from Cold Spring Harbor Perspectives in Medicine are provided here courtesy of Cold Spring Harbor Laboratory Press

RESOURCES