Open access peer-reviewed chapter

Contribution of Noradrenaline, Serotonin, and the Basolateral Amygdala to Alcohol Addiction: Implications for Novel Pharmacotherapies for AUDs

Written By

Omkar L. Patkar, Arnauld Belmer and Selena E. Bartlett

Submitted: 25 October 2015 Reviewed: 04 March 2016 Published: 14 July 2016

DOI: 10.5772/62843

From the Edited Volume

Recent Advances in Drug Addiction Research and Clinical Applications

Edited by William M. Meil and Christina L. Ruby

Chapter metrics overview

2,082 Chapter Downloads

View Full Metrics

Abstract

Alcohol use disorders (AUDs) constitute one of the 10 leading causes of preventable deaths worldwide. To date, there are only a few Food and Drug Administration (FDA)‐approved medications for AUDs, all of which are only moderately effective. The development of improved and effective strategies for the management of AUDs is greatly needed. This review focuses on understanding the neurobiological basis of alcohol addiction with a special emphasis on the role of serotonin (5‐hydroxytryptamine, 5‐HT) and noradrenaline (NE) in AUDs and sheds light on their complex interplay in the basolateral amygdala (BLA)––a brain region widely implicated in addiction. There is a significant evidence to support the role of the amygdala in stress‐induced negative emotional states resulting from withdrawal from alcohol; in fact, it has been hypothesized that this leads to craving and relapse. Dysregulation of 5‐HT and NE signaling in the BLA have been proposed to alter affective behavior, memory consolidation, and most importantly increase the propensity for addiction to alcohol and other common drugs of abuse. Improving deficits in 5‐HT and NE receptor signaling may provide ideal targets for the treatment of AUDs.

Keywords

  • Addiction
  • alcohol use disorders
  • noradrenaline
  • serotonin
  • basolateral amygdala

1. Introduction

1.1 Alcohol addiction: one drink too many

Alcohol dependence or alcohol abuse, now collectively known as alcohol use disorders (AUDs), causes significant loss of productivity, health concerns, emotional instability, career‐oriented failures, and socioeconomic problems [1]. It is estimated that AUDs amount to 3.8% of global deaths and 4.6% of disability‐adjusted life years [2]. The Diagnostic and Statistical Manual of Mental Disorders, 4th edition (DSM‐IV‐TR), defines AUDs on the persistence of dependence symptoms like tolerance, withdrawal, increased amounts of alcohol consumed over time, ineffective efforts to reduce use, interference with personal or professional life, significant amount of time spent obtaining, using, and recovering from alcohol or continued use of alcohol despite harmful consequences [3]. The U.S. National Institute of Alcohol Abuse and Alcoholism (NIAAA) defined men who consume more than 14 drinks per week and women having more than 11 drinks per week belong to the “At Risk” category of alcohol consumers.

1.2 Neurobiology of alcohol addiction: a vicious cycle

Alcohol addiction like any other drug addiction is a chronic relapsing disorder characterized by compulsive alcohol use and alcohol‐seeking behavior [4, 5]. The neurobiology of alcohol addiction is increasingly complex; however, for the purpose of simplicity, it can be delineated in three stages. The first phase of this cycle is the Binge and intoxication stage [5]. During this phase, reward areas of the brain involving the mesocorticolimbic system like the dorsal striatum and nucleus accumbens (NAc) are activated, which results in pleasurable and rewarding feelings [5, 6]. Dopamine is a key neurotransmitter involved in this stage [79]. The positive reinforcement is triggered by the pleasurable effects of alcohol where the user wants “more” to experience the hedonic effects. This is then followed by the Withdrawal stage [5]. During this phase, brain regions that are associated with negative feelings and emotions are activated, such as the amygdala and bed nucleus of stria terminalis (BNST) [4, 5]. Chronic withdrawal‐induced stress blunts the activity of the stress–response system and sensitizes extrahypothalamic structures of the extended amygdala [6, 10]. This stage marks a critical phase in the addiction cycle where alcohol use is primarily motivated by the desire to avoid negative feelings of stress, dysphoria, and negative emotional states of alcohol withdrawal. The third phase is the Preoccupation and anticipation stage [5]. During this phase, brain regions like the frontal cortex and hippocampus [11] that respond to previously paired alcohol cues and contexts are activated, intensifying alcohol‐seeking behavior [12, 13]. Since the frontal cortex is involved in decision‐making and higher executive functions, alcohol‐induced neuroadaptations of the frontal cortex [14] impair higher cognitive and decision‐making processes, increasing the rate of relapse in alcoholics.

Over time, as this cycle is repeated, alcohol‐induced neuroadaptations in the reward circuitry, stress–response pathway, and brain regions involved in higher cognitive functions facilitate the transition from nondependent to dependent alcohol consumption. These maladaptive neuromodulations contribute to sensitization, tolerance, craving, and relapse to alcohol‐seeking [4]. For instance, alcohol‐induced plasticity in glutamatergic signaling in the NAc may contribute to behavioral sensitization to the effects of alcohol [15], while changes in the synaptic properties of NAc‐medium spiny neurons contribute to relapse during withdrawal [16]. Furthermore, chronic alcohol modulates presynaptic and postsynaptic functions on glutamate neurons in the basolateral amygdala (BLA) [17]. Finally, alcohol impairs communication between the amygdala and prefrontal cortex to disrupt cognitive and emotional responses that lead to altered affective states that further contribute to the development of alcohol dependence [18, 19].

1.3. Pharmacotherapy: available treatment options for AUDs

Bill Wilson and Bob Smith took early steps toward alcohol remediation in 1935 with the introduction of Alcoholic Anonymous (AA) [20, 21]. This 12‐step approach toward rehabilitation was built on the premise of acceptance of individual helplessness during addiction to alcohol and other drugs of abuse [22]. This method was adopted by the “Minnesota model of addiction treatment” in a 28‐day rehabilitation setting [23]. Parallel efforts to treat alcoholism by understanding the nature and cause of alcohol dependence were gaining momentum, which led to the foundation of the National Institute of Alcohol Abuse and Alcoholism (NIAAA) in 1970 [24].

Since then, several approaches to understand and treat alcoholism were designed that took into consideration individual differences and susceptibility to AUDs. Cognitive behavioral therapy or motivational therapy was adopted as the first line of treatment to match the needs of the addict to help recuperate in a 12‐week therapy session called “Project MATCH” [25]. This project was successful in rehabilitation of patients that did not have any psychiatric conditions. The next step was to combine behavioral and pharmacotherapy in the treatment of alcoholism called “Project COMBINE” [26]. This study evaluated the efficacy of available pharmacotherapies, namely acamprosate and naltrexone, in conjunction with or without medical assistance and with or without cognitive–behavioral therapy [27].

Acamprosate (CampralTM), the calcium salt of N‐acetyl homotaurine, suppresses alcohol consumption and relapse [28, 29]. Early reports delineating the mechanism of action of acamprosate were unclear [30]; however, recent studies have shown that acamprosate works through the calcium ion in its molecular structure [31]. This was supported with improved results in patients that showed an increase in plasma calcium levels following acamprosate treatment [31]. Acamprosate has been shown to have a good safety and a tolerability profile and is highly effective in maintenance of abstinence in patients who are abstinent at treatment initiation [32].

In addition to acamprosate, the mu‐opioid receptor antagonist, naltrexone (Re ViaTM), was found effective as a treatment for alcohol consumption and relapse [33]. However, studies have shown that naltrexone is ineffective in achieving abstinence in alcoholic subjects; instead it is more effective to reduce consumption [34, 35]. Also, recent research demonstrated that it acts more specifically for a cohort with single nucleotide polymorphism (SNP) in exon 1 of the mu‐opioid receptor gene (OPRM1) [36] limiting broader efficacy. Nevertheless, naltrexone reduces alcohol consumption through a dopaminergic/opioidergic reinforcement system, causing increased sedation and less arousal in patients consuming alcohol [35]. Both these drugs were successful in reducing drinking in combination with behavioral therapy, as highlighted by the COMBINE project [37].

In addition to acamprosate and naltrexone, disulfiram (Antabuse®) was approved as a therapeutic treatment for alcoholism. The anti‐alcohol addiction properties of disulfiram were serendipitously discovered, when a Danish physician Jacobsen accidentally ingested alcohol over disulfiram and experienced its unpleasant and nauseous effects [38, 39]. Disulfiram inhibits the enzyme aldehyde dehydrogenase (ALDH), which results in the accumulation of acetaldehyde on alcohol ingestion [40]. This toxic metabolite produces aversive symptoms, such as flushing, nausea, and vomiting, and a desire to avoid this reaction encourages abstinence [41]. Disulfiram also inhibits dopamine‐β‐hydroxylase (DBH), the enzyme required to synthesize noradrenaline (NE). It reduces NE concentrations and elevates dopamine (DA) concentrations to facilitate normal DA functioning [40, 41], a pharmacotherapeutic feature of the drug that makes it an excellent treatment option even for cocaine addicts.

In addition to this, our lab has investigated the role of neuronal nicotinic acetylcholine receptors in alcohol addiction and came up with varenicline (ChampixTM) as a treatment option for AUDs [42, 43]. Varenicline was found to be more efficacious in heavy‐drinking smokers because of the comorbid nature of both the types of addiction involving the recruitment of nicotinic acetylcholine receptors. Varenicline is now in its third stage of clinical trial as a treatment option for AUDs [44, 45].

1.4. Shortcomings of available treatment options for AUDs: need for better pharmaceutical alternatives

Acamprosate, naltrexone, and disulfiram are the only available medications for alcoholism approved by the Food and Drug Administration (FDA), while nalmefene (SelincroTM), an opioid receptor antagonist having a similar mechanism of action to naltrexone [46], is approved as a medication for alcohol abstinence in Europe [47]. Most of these drugs treat one aspect of alcoholism at best without significantly altering other parameters of alcohol addiction.

Drugs like acamprosate reduce consumption and are effective in motivating abstinence for a certain period of time. However, acamprosate does not significantly affect abstinence‐induced rebound consumption of alcohol [48]. Also, despite achieving an aversion for alcohol, the likelihood of the addict returning to drinking with increased tolerance cannot be assured. A case study also indicated the development of Parkinson's‐like syndrome with acamprosate use [49].

Although naltrexone was shown to be very effective with and without cognitive behavioral therapy, noncompliance with maintenance of drug regimen was shown to limit efficacy [50]. About 37% patients were reported to discontinue naltrexone therapy by 12 weeks and 80% by 6 months [50]. It is possible that some of the severe complications involved with naltrexone use, that is, renal failure and hepatitis, may have contributed to its early discontinuation [51]. Furthermore, the efficacy of naltrexone appears to be related to alcohol abusers having the mu‐opioid SNP [36].

All the above drugs work best when combined with an individual's motivation to quit drinking. Disulfiram works on this principle as it deters the positive reinforcing effects of alcohol and masks them with aversive and negative feelings stimulated by the action of the drug post‐alcohol consumption [52]. As a result, this drug is effective for alcoholics with a goal to achieve complete abstinence, but has limited efficacy for alcoholics without these goals. Noncompliance is one of the biggest challenges in the use of disulfiram, illustrated by the 20% compliance measure in the largest controlled trial to date [53]. Also, disulfiram is contraindicated in patients with cardiac disease and on rare occasions may cause severe liver damage [54].

Despite the availability of these pharmacotherapies and behavioral therapy, AUDs are widely prevalent. As illustrated by COMBINE, no single medication or treatment strategy is effective in every case or in every person [37]. A detailed investigation of other neurobiological factors that play a role in alcohol dependence is needed as are further strategies to treat alcoholism.

The remainder of this chapter highlights the role of serotonin (5‐hydroxytryptamine, 5‐HT), NE, and BLA in alcohol addiction with a view to improve current treatment strategies for AUDs.

Advertisement

2. NE and serotonin: role in alcohol dependence

Prolonged alcohol exposure causes maladaptive changes in regions of the extended amygdala that cause sensitization to negative emotional states and reinforcement of addictive behaviors during withdrawal. These neuroadaptations alter the activity of important neurotransmitters particularly involved in stress. Such changes are well documented for increasing the activity of the stress neurotransmitter corticotrophin‐releasing factor (CRF) in rodent models of alcohol dependence [4]. Additionally, changes in the function and signaling of other neurotransmitters including 5‐HT [5557] and NE [5561] have also been implicated in the development of alcohol addiction.

NE and 5‐HT play a crucial role in regulating mood, emotions, and importantly, behavioral adaptations to stress that include addictive phenotypes [57, 60]. As these neurochemicals widely innervate the reward system [6266] and extrahypothalamic regions involving the amygdala [6771], these are prime candidates to influence alcohol and even other drug‐seeking behaviors.

Dysregulation of the 5‐HT pathway is implicated in AUDs and other affective states like depression and anxiety disorders [57, 72, 73]. Recent studies have demonstrated an increase in the immunoreactivity of tryptophan hydroxylase (TRH)––the rate‐limiting step in 5‐HT synthesis, in the dorsal raphe nuclei (DRN) of alcohol‐dependent victims of depression and suicide compared to normal psychiatric controls [74]. Such disruptions in brain serotonin levels in these individuals have widespread implications in the role of 5‐HT to regulate emotional and behavioral vulnerability to alcohol and other drugs of abuse. Alcohol increases 5‐HT levels in the ventral tegmental area (VTA), NAc, and amygdala [75]. These brain regions play a pivotal role in processing of information from emotional and rewarding stimuli. Chronic alcohol abuse alters the activity of these brain areas, resulting in changes in motivational and goal‐directed behaviors, which further drive alcohol‐seeking behavior [76, 77]. For instance, studies have shown that behavioral sensitization to alcohol is mediated by accumbal 5‐HT2C receptors [76], and blockade of 5‐HT3 receptors especially in the VTA attenuates alcohol consumption [77]. The 5‐HT receptors, 5‐HT1A, 5‐HT1B, 5‐HT2A, and 5‐HT2C, [7880] have been widely implicated in alcohol consumption in animal models with new evidence also implicating 5‐HT3 and 5‐HT6 receptors in alcohol addiction [81, 82].

NE has been shown to play a significant role in negative emotional states which contribute to alcohol consumption [60, 83, 84]. Acute alcohol decreases [85], while chronic alcohol and withdrawal increases the activity of neurons in the locus coeruleus (LC), a region that provides the majority of NE in the brain [86]. Activation of the α2‐adrenergic autoreceptors has been shown to attenuate the overall negative effects of withdrawal [87], and blocking α1‐adrenergic receptors (ARs) using prazosin reduced alcohol consumption in dependent rats [88] and human alcoholics [89]. Likewise, treatment with the β‐AR antagonist, propranolol, reduced drinking in dependent rats [60]. Evidence also suggests that β‐ARs may also contribute in mediating the anxiolytic effects of alcohol [58].

Furthermore, CRF is a regulating factor in the activation of the hypothalamus–pituitary–adrenal (HPA) axis to stress [9094]. Chronic alcohol consumption affects CRF signaling in the central nucleus of amygdala (CeA) and BNST, as evidenced by alterations in CRF transmission during withdrawal [95]. Interestingly, NE and 5‐HT have been shown to interact with the neurotransmitter CRF in neuroanatomical sites like the LC, DRN, CeA, and BNST [96100] to influence addictive behaviors. For instance, yohimbine, a pharmacological agent used to promote stress in rats, has effects on NE, 5‐HT, and CRF signaling to potentiate alcohol drinking and reinstatement [101, 102], suggesting possible mutual regulatory roles of these neurotransmitters in alcohol dependence and relapse. This was further evidenced by CRF antagonism in the DRN to attenuate yohimbine‐induced alcohol‐seeking behavior in rats [100]. Also, CRF and NE antagonism has been shown to be effective in reducing stress‐induced reinstatement in human alcoholics [88, 103].

Advertisement

3. The BLA: role in alcohol addiction

The amygdaloid complex is made up 13 distinct nuclei which are divided in three groups: the deep or basolateral group, the superficial or cortical‐like group, and the centromedial group [104]. These nuclei have been proposed to be located in such a way to maximize the amygdala's connections with other limbic, cortical, and subcortical regions of the brain to help facilitate its function in emotional processing, learning, and fear memory [105107]. The basolateral amygdalar complex, comprising of lateral amygdala (LA), basal and basomedial nuclei [108, 109] controls behavioral expressions like emotional arousal, fear, and stress that are linked to traumatic incidents, stressful environmental stimuli, or pharmacological stressors, and consolidates them as memories [70]. The BLA communicates through excitatory efferents to the prefrontal cortex and structures of the limbic system involving the hippocampus, NAc, dorsomedial striatum (DMS), and BNST [110114], while it receives feedback from these structures through glutamatergic afferents [115, 116], majority of which converge with the cortical inputs [114] running toward the BLA.

The role of the BLA in fear, memory consolidation, and emotional learning along with its contribution in associative learning for appetitive conditioning is well documented [70, 105, 117, 118]. Since the BLA can impart incentive salience to a previously neutral stimulus in response to a motivational or a goal‐directed task [119], recent efforts have now focused on the role of the BLA in drug‐seeking, including cocaine [120], morphine [121], and alcohol [122124].

Alcohol has been shown to increase neuronal activity and glucose utilization in the BLA [125]. Additionally, long‐term alcohol exposure alters glutamate transmission in the BLA [126] and NAc [127], which is implicated in increased alcohol self‐administration in rodents [128]. Furthermore, alcohol‐induced withdrawal stress increases presynaptic glutamatergic function in thalamic afferents to the BLA that may explain the increased emotional dysregulation during withdrawal [129]. It has also been shown that altered neuropeptide S function in the BLA following long‐term alcohol exposure may contribute to relapse [130]. Furthermore, a recent study has shown that IL‐1 receptor signaling in the BLA contributes to binge‐like alcohol consumption in mice [131].

There is growing body of evidence that supports the role of the BLA in conditioned–cued relapse [132] and context‐induced reinstatement [133] for alcohol and a variety of other drugs [134137]. It was shown that the BLA may play a significant role in cue‐induced alcohol reinstatement [138], following exposure to previously alcohol‐paired environmental cues [123]. Research has also shown that BLA–glutamatergic signaling attributes salience to conditioned cues that are related to alcohol‐seeking [132], while the opioidergic system of the BLA may play a role in context‐induced alcohol‐seeking [140]. Indeed, since the BLA extensively communicates with the NAc, alcohol withdrawal‐induced changes in glutamatergic function in the BLA get perpetuated in structures of the reward system that may contribute to craving and relapse [141].

It is well documented that repeated and chronic stress leads to adverse behavioral outcomes, and many studies support the reinforcing effects of chronic stress in drug addiction in animal models [86, 142144]. Stress alters the morphology of BLA principal cells and impairs fear extinction memory [145] that may have implications in the development of affective disorders like PTSD and depression. It has been shown that the BLA modulates chronic stress‐induced learning and memory deficits in the hippocampus, suggesting that dysregulation of BLA–hippocampal signaling may affect memory storage, retrieval, and extinction of fear memory that may contribute to emotional disorders and drug dependence [146]. Furthermore, early life stress causes increased excitability of pyramidal cells in the BLA [147], while chronic restraint stress in adolescent and adult rats increases BLA activity [148]. Increased BLA excitability has been positively correlated with increased anxiety and increased alcohol‐seeking behavior [141, 147, 149, 150].

Long‐term exposure to alcohol simulates chronic stress‐like conditions [130] that have a profound effect on fear memory consolidation [151]. Alcohol withdrawal‐induced stress has been shown to increase conditioned fear [152] and impair extinction of fear memory [153]. A recent study also showed that repeated alcohol exposures enhance retrieval of previously consolidated fear memories and augments activity in BLA and other brain regions involved in fear memory retrieval [154].

Advertisement

4. Role of 5‐HT and NE in the BLA in alcohol addiction

There is significant evidence that supports the role of NE and 5‐HT in drug dependence and alcohol addiction [87, 155157]. Moreover, the BLA which is highly implicated in dependence to alcohol‐seeking [17, 123, 131, 132] is densely innervated by these neurotransmitters [58, 71, 158, 159]. Since chronic alcohol exposure causes neuroadaptations that affect the signaling and receptor subtypes of these neurotransmitters, dysregulation of NE and 5‐HT transmission in the BLA may lead to a constellation of aversive outcomes including altered consolidation of alcohol‐related memories, anxiety disorders, and eventually higher rates of relapse [132, 138].

NE plays a vital role in facilitating the function of the BLA in fear memory consolidation [70]. It has been shown that intra‐BLA infusions of β‐AR agonists enhance retention of inhibitory avoidance [160], while β‐AR antagonists block fear memory enhancement [69]. Also, α1‐AR activation in the BLA enhances fear memory consolidation through an interaction with β‐ARs [161]. This evidence suggests that noradrenergic receptors strongly contribute to BLA function. It is possible that alteration in NE activity in the BLA may lead to altered memory consolidation and stress‐coping mechanism that may enhance alcohol‐seeking and relapse [162]. Indeed, antagonism of α1‐ARs reduced dependence‐induced increase in alcohol consumption in rats [88]. Furthermore, recent evidence supports the role of β‐ARs in alcohol‐induced enhancement of GABA synapses in the BLA, suggesting a possible noradrenergic mechanism mediating the anxiolytic effects of alcohol [58] (Figure 1). This was further evidenced by intra‐BLA infusions of a β3‐AR agonist that enhanced inhibitory GABA signaling on BLA pyramidal cells to reduce anxiety‐like and alcohol‐seeking behavior [163]. Furthermore, the neuroadaptive changes associated with chronic alcohol consumption including desensitization of β‐ARs in the BLA have been shown to modulate its activity [164] (Figure 1).

Figure 1.

Changes in NE signaling and BLA output following acute and chronic alcohol exposure or withdrawal. Acute alcohol decreases NE signaling in the BLA, which is regulated by a feedback loop through presynaptic α2‐adrenergic autoreceptors expressed on NE fibers in the LC. Decreased BLA‐NE levels decrease the excitation of BLA principal cells through postsynaptic α1‐ARs. Acute alcohol further enhances the inhibition of BLA principal cells by NE‐mediated enhancement of GABA synapses through β‐ARs expressed on GABAergic LPSCs. The net result of this inhibition is decreased BLA principal neuron excitability and BLA activity which has been suggested to reduce anxiety and may explain the anxiolytic effect of acute alcohol. Chronic alcohol/withdrawal increases NE levels that enhance α1‐AR mediated excitation of BLA principal cells. Chronic ethanol has been shown to desensitize β‐ARs in the brain which leads to a reduction in NE's effects on GABA‐LPSCs causing a decrease in the inhibitory tone over BLA principal cells, increasing excitability. This increases the net excitability of BLA principal cells and increases BLA activity causing anxiety during withdrawal and may contribute to alcohol‐seeking and relapse.

In contrast to excitatory dopaminergic/glutamatergic signaling in the BLA that increases its activity, serotonergic transmission in the BLA is inhibitory [165]. The serotonergic innervations on principal glutamate cells in the BLA decrease the overall excitatory activity of these cells [166] through 5‐HT1A receptors [167] and modulate BLA output (Figure 2). This is supported by a recent study where depletion of serotonin in the BLA increased glutamate receptor density and fear‐potentiated startle in mice, indicating that serotonergic inhibition regulates excitatory signaling in the BLA to modulate affective behaviors like anxiety [68]. Chronic alcohol‐induced neuroadaptations change 5‐HT receptor expression and function in the brain [168] that alters the regulatory control of serotonin over BLA principal cells. Loss of inhibition on BLA principal neurons increases BLA output, increasing anxiety [169, 170] and other symptoms of withdrawal. In support of this, chronic alcohol or withdrawal stress increases the expression of 5‐HT1A autoreceptors in the raphe nucleus [168] which causes a reduction in 5‐HT levels in the BLA. This increases BLA activity, which contributes to anxiety‐like behaviors following withdrawal from chronic alcohol (Figure 2). Furthermore, 5‐HT2A/2C receptors have been suggested to potentiate inhibitory GABAergic tone on principal BLA glutamatergic cells to decrease excitability [67]. Chronic alcohol causes adaptive changes that lower the expression levels of these receptors, reducing inhibition over BLA principal neurons [67]. This augments BLA output and increases the possibility of anxiety‐induced relapse following a period of chronic alcohol exposure [141] (Figure 2). In addition to this, chronic alcohol‐induced neuroadaptations in other receptor subtypes like the GABA‐A receptors facilitate the anxiolytic effects of alcohol [171]. Increasing the activity of 5‐HT on GABA‐A receptors on BLA principal cells may contribute in reducing withdrawal‐induced anxiety and alcohol‐seeking.

Figure 2.

Changes in 5‐HT signaling and BLA output following acute and chronic alcohol exposure or withdrawal. Acute alcohol increases 5‐HT release in the BLA which is regulated by a feedback loop through 5‐HT1A autoreceptors expressed on 5‐HT neurons in the DRN. Increased BLA‐5‐HT levels enhance the inhibition of BLA activity through postsynaptic 5‐HT1A receptors expressed on principal neurons. Increased 5‐HT signaling also activates 5‐HT2A/2C receptors expressed on GABAergic interneurons in the BLA that further increase the inhibition on BLA principal cells through increased GABAergic tone. The net result of this inhibition is decreased BLA principal neuron excitability and BLA activity, which has been shown to reduce anxiety and may explain the anxiolytic effect of acute alcohol. Chronic alcohol/withdrawal increases the expression of 5‐HT1A autoreceptors in the DRN which decreases 5‐HT levels in the BLA. This reduces 5‐HT1A‐mediated inhibition on BLA principal cells. Chronic alcohol‐induced withdrawal downregulates the expression of 5‐HT2A/2C receptors on GABAergic interneurons to further decrease the inhibitory GABA tone on BLA principal cells, increasing excitability. Chronic alcohol also upregulates GABA receptors on principal cells. This results in a net increase in BLA activity causing anxiety that may contribute to alcohol‐seeking and relapse.

Furthermore, cross‐modulation of synaptic transmission in the BLA by 5‐HT1A/1B receptors and β‐ARs dictates BLA output [159] that may affect behavioral outcomes like stress, anxiety, and drug dependence. In support of this, we have shown that pindolol, a drug having dual pharmacological activity on 5‐HT1A/1B receptors and β1/β2 ARs, decreases alcohol consumption in mice following long‐term alcohol exposure. Our electrophysiological experiments also indicate that the BLA may mediate the effects of pindolol on alcohol consumption [172].

Advertisement

5. Conclusion

Research in the past few decades has significantly increased our understanding of the neurobiological basis of alcohol dependence. Recent research has targeted pathways that mediate more than just the reinforcing properties of alcohol. However, despite these concerted efforts, effective pharmacological interventions for the management of AUDs remain elusive.

Chronic alcohol consumption causes maladaptive changes in brain regions like the extended amygdala that cause sensitization to negative emotional states of withdrawal. These changes disrupt the signaling of many neurotransmitters including those involved in stress. Dysregulation of NE and 5‐HT signaling has been widely implicated in the development of affective disorders and alcohol addiction. Specifically, NE and/or 5‐HT impairments in the BLA, a region involved in stress, emotional processing, and reward‐seeking have been suggested to play a major role in the development of alcohol dependence (Figures 1 and 2).

In addition to the growing evidence in animal models of alcohol addiction, pharmacological compounds that target NE and 5‐HT receptors have also shown promise as potential treatment strategies for AUDs in human patients [173, 174]. Noradrenergic compounds like propranolol [175, 176] and atenolol [174] have been shown to attenuate alcohol‐seeking behavior and reduce craving in human alcoholics. Similarly, serotonergic compounds like buspirone show efficacy to reduce anxiety‐induced consumption in alcoholics [177, 178]. Moreover, our research indicates that pindolol, the FDA‐approved antihypertensive drug having activity on both 5‐HT and NE receptors, may have a similar mechanism of action to more effectively reduce alcohol consumption following chronic intake [172].

Since the BLA plays a vital role in affective disorders and stress‐induced maladaptive behavioral conditioning, drugs that selectively modulate NE and 5‐HT signaling in the BLA offer great promise in the treatment of AUDs. With the increasing need for improved pharmacotherapeutic strategies for the management of AUDs combined with the modest efficacy of current treatments, putative compounds that target 5‐HT and NE receptors may prove useful for the development of more effective treatment strategies for alcohol dependence.

Advertisement

Acknowledgments

This work was supported by funding from grants from the Australian Research Council and National Health and Medical Research Council to S.E.B. We thank Dr. Paul Klenowski and Ms Joan Holgate in providing their valuable suggestions to this manuscript.

Financial disclosures

All the authors who have participated in this study report no biomedical financial interests or potential conflicts of interest.

Author's contributions

All authors have been involved in the preparation and have approved the submitted manuscript. Omkar Patkar was the lead author and responsible for conducting the literature review and writing the manuscript. Arnauld Belmer assisted in writing and editing the manuscript. As the senior author, Selena Bartlett supervised Omkar Patkar's work, reviewed, and edited the manuscript.

References

  1. 1. Gnanavel S, Robert RS. Diagnostic and Statistical Manual of Mental Disorders, fifth edition, and the impact of events scale—revised. Chest. 2013;144(6):1974.
  2. 2. Rehm J, Mathers C, Popova S, Thavorncharoensap M, Teerawattananon Y, Patra J. Global burden of disease and injury and economic cost attributable to alcohol use and alcohol‐use disorders. The Lancet. 2009;373(9682):2223–33.
  3. 3. Trull TJ, Verges A, Wood PK, Jahng S, Sher KJ. The structure of Diagnostic and Statistical Manual of Mental Disorders (4th edition, text revision) personality disorder symptoms in a large national sample. Personal Disorders. 2012;3(4):355–69.
  4. 4. Gilpin NW, Koob GF. Neurobiology of alcohol dependence: focus on motivational mechanisms. Alcohol Research & Health: The Journal of the National Institute on Alcohol Abuse and Alcoholism. 2008;31(3):185–95.
  5. 5. Koob GF, Volkow ND. Neurocircuitry of addiction. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2010;35(1):217–38.
  6. 6. Koob GF. A role for brain stress systems in addiction. Neuron. 2008;59(1):11–34.
  7. 7. Harris RA, Brodie MS, Dunwiddie TV. Possible substrates of ethanol reinforcement: GABA and dopamine. Annals of the New York Academy of Sciences. 1992;654:61–9.
  8. 8. Yorgason JT, Ferris MJ, Steffensen SC, Jones SR. Frequency‐dependent effects of ethanol on dopamine release in the nucleus accumbens. Alcoholism: Clinical and Experimental Research. 2014;38(2):438–47.
  9. 9. Young KA, Gobrogge KL, Wang Z. The role of mesocorticolimbic dopamine in regulating interactions between drugs of abuse and social behavior. Neuroscience & Biobehavioral Reviews. 2011;35(3):498–515.
  10. 10. Zorrilla, E.P., M.L. Logrip, and G.F. Koob, Corticotropin releasing factor: a key role in the neurobiology of addiction. Front Neuroendocrinol, 2014. 35(2): p. 234-44.
  11. 11. Kalivas PW, O'Brien C. Drug addiction as a pathology of staged neuroplasticity. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2008;33(1):166–80.
  12. 12. Baimel C, Bartlett SE, Chiou L‐C, Lawrence AJ, Muschamp JW, Patkar O, Tung L‐W, Borgland SL. Orexin/hypocretin role in reward: implications for opioid and other addictions. British Journal of Pharmacology. 2015;172(2):334–48.
  13. 13. Abernathy K, chandler lJ, Woodward JJ. Alcohol and the prefrontal cortex. International Review of Neurobiology. 2010;91:289–320.
  14. 14. Bechara A, Dolan S, Denburg N, Hindes A, Anderson SW, Nathan PE. Decision‐making deficits, linked to a dysfunctional ventromedial prefrontal cortex, revealed in alcohol and stimulant abusers. Neuropsychologia. 2001;39(4):376–89.
  15. 15. Abrahao KP, Ariwodola OJ, Butler TR, Rau AR, Skelly MJ, Carter E, Alexander NP, McCool BA, Souza‐Formigoni MLO, Weiner JL. Locomotor sensitization to ethanol impairs NMDA receptor‐dependent synaptic plasticity in the nucleus accumbens and increases ethanol self‐administration. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2013;33(11):4834–42.
  16. 16. Marty VN, Spigelman I. Effects of alcohol on the membrane excitability and synaptic transmission of medium spiny neurons in the nucleus accumbens. Alcohol (Fayetteville, NY). 2012;46(4):317–27.
  17. 17. Lack AK, Diaz MR, Chappell A, DuBois DW, McCool BA. Chronic ethanol and withdrawal differentially modulate pre‐ and postsynaptic function at glutamatergic synapses in rat basolateral amygdala. Journal of Neurophysiology. 2007;98(6):3185–96.
  18. 18. Stephens DN, Duka T. Cognitive and emotional consequences of binge drinking: role of amygdala and prefrontal cortex. Philosophical Transactions of the Royal Society B: Biological Sciences. 2008;363(1507):3169–79.
  19. 19. Pleil KE, Lowery‐Gionta EG, Crowley NA, Li C, Marcinkiewcz CA, Rose JH, McCall NM, Maldonado‐Devincci AM, Morrow AL, Jones SR, Kash TL. Effects of chronic ethanol exposure on neuronal function in the prefrontal cortex and extended amygdala. Neuropharmacology. 2015;99:735–49.
  20. 20. Krentzman AR, Farkas KJ, Townsend AL. Spirituality, religiousness, and alcoholism treatment outcomes: a comparison between black and white participants. Alcoholism Treatment Quarterly. 2010;28(2):128–50.
  21. 21. Tonigan JS, Rynes KN, McCrady BS. Spirituality as a change mechanism in 12‐step programs: a replication, extension, and refinement. Substance Use & Misuse. 2013;48(12):1161–73.
  22. 22. Bogenschutz MP, Rice SL, Tonigan JS, Vogel HS, Nowinski J, Hume D, Arenella PB. 12‐Step facilitation for the dually diagnosed: a randomized clinical trial. Journal of Substance Abuse Treatment. 2014;46(4):403–11.
  23. 23. Anderson DJ, McGovern JP, DuPont RL. The origins of the Minnesota model of addiction treatment – a first person account. Journal of Addictive Diseases. 1999;18(1):107–14.
  24. 24. Huebner RB, Kantor LW. Advances in alcoholism treatment. Alcohol Research & Health: The Journal of the National Institute on Alcohol Abuse and Alcoholism. 2011;33(4):295–9.
  25. 25. Project MATCH secondary a priori hypotheses. Project MATCH Research Group. Addiction. 1997;92(12):1671–98.
  26. 26. Gueorguieva R, Wu R, Donovan D, Rounsaville BJ, Couper D, Krystal JH, O'Malley SS. Naltrexone and combined behavioral intervention effects on trajectories of drinking in the COMBINE study. Drug and Alcohol Dependence. 2010;107(2–3):221–9.
  27. 27. Kranzler HR, Kirk J. Efficacy of naltrexone and acamprosate for alcoholism treatment: a meta-analysis. Alcoholism: Clinical and Experimental Research. 2001;25(9):1335–41.
  28. 28. Acamprosate. Drugs in R&D. 2002;3(1):13–8.
  29. 29. Acamprosate campral for alcoholism. Medical Letter on Drugs and Therapeutics. 2005;47(1199):1–3.
  30. 30. De Witte P, Littleton J, Parot P, Koob G. Neuroprotective and abstinence‐promoting effects of acamprosate: elucidating the mechanism of action. CNS Drugs. 2005;19(6):517–37.
  31. 31. Spanagel R, Vengeliene V, Jandeleit B, Fischer W‐N, Grindstaff K, Zhang X, Gallop MA, Krstew EV, Lawrence AJ, Kiefer F. Acamprosate produces its anti‐relapse effects via calcium. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2014;39(4):783–91.
  32. 32. Yahn SL, Watterson LR, Olive MF. Safety and efficacy of acamprosate for the treatment of alcohol dependence. Substance Abuse: Research and Treatment. 2013;7:1–12.
  33. 33. Hoque MM, Hossain KJ, Kamal MM, Akhtaruzzaman M. Naltrexone in drug addiction: significance in the prevention of relapse. Mymensingh Medical Journal. 2009;18(1 Suppl):S56–65.
  34. 34. Del Re AC, Maisel N, Blodgett J, Finney J. The declining efficacy of naltrexone pharmacotherapy for alcohol use disorders over time: a multivariate meta‐analysis. Alcoholism: Clinical and Experimental Research. 2013;37(6):1064–8.
  35. 35. Chen A, Morgenstern J, Davis CM, Kuerbis AN, Covault J, Kranzler HR. Variation in Mu‐Opioid Receptor Gene (OPRM1) as a moderator of naltrexone treatment to reduce heavy drinking in a high functioning cohort. Journal of Drug & Alcohol Dependence. 2013;1(1):101.
  36. 36. Oslin DW, Berrettini W, Kranzler HR, Pettinati H, Gelernter J, Volpicelli JR, O'Brien CP. A functional polymorphism of the mu‐opioid receptor gene is associated with naltrexone response in alcohol‐dependent patients. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2003;28(8):1546–52.
  37. 37. Anton RF, O'Malley SS, Ciraulo DA, Cisler RA, Couper D, Donovan DM, Gastfriend DR, Hosking JD, Johnson BA, LoCastro JS, Longabaugh R, Mason BJ, Mattson ME, Miller WR, Pettinati HM, Randall CL, Swift R, Weiss RD, Williams LD, Zweben A, Group CSR. Combined pharmacotherapies and behavioral interventions for alcohol dependence: the COMBINE study: a randomized controlled trial. JAMA. 2006;295(17):2003–17.
  38. 38. Hald J, Jacobsen E. A drug sensitizing the organism to ethyl alcohol. The Lancet. 1948;2(6539):1001–4.
  39. 39. Krampe H, Ehrenreich H. Supervised disulfiram as adjunct to psychotherapy in alcoholism treatment. Current Pharmaceutical Design. 2010;16(19):2076–90.
  40. 40. Kalra G, De Sousa A, Shrivastava A. Disulfiram in the management of alcohol dependence: a comprehensive clinical review. Open Journal of Psychiatry. 2014;4:43.
  41. 41. Schroeder JP, Cooper DA, Schank JR, Lyle MA, Gaval‐Cruz M, Ogbonmwan YE, Pozdeyev N, Freeman KG, Iuvone PM, Edwards GL, Holmes PV, Weinshenker D. Disulfiram attenuates drug‐primed reinstatement of cocaine seeking via inhibition of dopamine beta‐hydroxylase. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2010;35(12):2440–9.
  42. 42. Bito‐Onon JJ, Simms JA, Chatterjee S, Holgate J, Bartlett SE. Varenicline, a partial agonist at neuronal nicotinic acetylcholine receptors, reduces nicotine‐induced increases in 20% ethanol operant self‐administration in Sprague–Dawley rats. Addiction Biology. 2011;16(3):440–9.
  43. 43. Steensland P, Simms JA, Holgate J, Richards JK, Bartlett SE. Varenicline, an alpha4beta2 nicotinic acetylcholine receptor partial agonist, selectively decreases ethanol consumption and seeking. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(30):12518–23.
  44. 44. Mitchell JM, Teague CH, Kayser AS, Bartlett SE, Fields HL. Varenicline decreases alcohol consumption in heavy‐drinking smokers. Psychopharmacology. 2012;223(3):299–306.
  45. 45. Litten RZ, Ryan ML, Fertig JB, Falk DE, Johnson B, Dunn KE, Green AI, Pettinati HM, Ciraulo DA, Sarid‐Segal O, Kampman K, Brunette MF, Strain EC, Tiouririne NA, Ransom J, Scott C, Stout R. A double‐blind, placebo‐controlled trial assessing the efficacy of varenicline tartrate for alcohol dependence. Journal of Addiction Medicine. 2013;7(4):277–86.
  46. 46. Osborn MD, Lowery JJ, Skorput AGJ, Giuvelis D, Bilsky EJ. In vivo characterization of the opioid antagonist nalmefene in mice. Life Sciences. 2010;86(15–16):624–30.
  47. 47. Robin C. Alcohol use disorder: pathophysiology, effects, and pharmacologic options for treatment. 2014;5:1–12.
  48. 48. Mann K, Lemenager T, Hoffmann S, Reinhard I, Hermann D, Batra A, Berner M, Wodarz N, Heinz A, Smolka MN, Zimmermann US, Wellek S, Kiefer F, Anton RF, Team PS. Results of a double‐blind, placebo‐controlled pharmacotherapy trial in alcoholism conducted in Germany and comparison with the US COMBINE study. Addiction Biology. 2013;18(6):937–46.
  49. 49. Sidana AK, Mangla D. Unusual side effects with acamprosate. Indian Journal of Psychiatry. 2007;49(2):143.
  50. 50. Hulse GK. Improving clinical outcomes for naltrexone as a management of problem alcohol use. British Journal of Clinical Pharmacology. 2013;76(5):632–41.
  51. 51. Waal H, Christophersen AS, Frogopsahl G, Olsen LH, Morland J. [Naltrexone implants – a pilot project]. Tidsskr Nor Laegeforen. 2003;123(12):1660–1.
  52. 52. Weiss RD. Pharmacotherapy of alcohol dependence: how and when to use disulfiram and naltrexone. Current Psychiatry. 2002;1(1):51–60.
  53. 53. Fuller RK, Branchey L, Brightwell DR, Derman RM, Emrick CD, Iber FL, James KE, Lacoursiere RB, Lee KK, Lowenstam I, et al. Disulfiram treatment of alcoholism. A Veterans Administration cooperative study. JAMA. 1986;256(11):1449–55.
  54. 54. Fuller RK, Gordis E. Does disulfiram have a role in alcoholism treatment today? Addiction. 2004;99(1):21–4.
  55. 55. Gorelick DA. Serotonin uptake blockers and the treatment of alcoholism. Recent Developments in Alcoholism: An Official Publication of the American Medical Society on Alcoholism, the Research Society on Alcoholism, and the National Council on Alcoholism. 1989;7:267–81.
  56. 56. George FR. Pharmacotherapy in alcoholism treatment: integrating and understanding the use of serotonin reuptake inhibitors. Alcohol and Alcoholism (Oxford, Oxfordshire) Supplement. 1994;2:537–43.
  57. 57. Gallant D. The serotonin system and alcoholism: basic research and clinical problems. Alcoholism: Clinical and Experimental Research. 1993;17(6):1345.
  58. 58. Silberman Y, Ariwodola OJ, Weiner JL. beta1‐adrenoceptor activation is required for ethanol enhancement of lateral paracapsular GABAergic synapses in the rat basolateral amygdala. The Journal of Pharmacology and Experimental Therapeutics. 2012;343(2):451–9.
  59. 59. Petrakis IL, Ralevski E, Desai N, Trevisan L, Gueorguieva R, Rounsaville B, Krystal JH. Noradrenergic vs serotonergic antidepressant with or without naltrexone for veterans with PTSD and comorbid alcohol dependence. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2012;37(4):996–1004.
  60. 60. Gilpin NW, Koob GF. Effects of beta‐adrenoceptor antagonists on alcohol drinking by alcohol‐dependent rats. Psychopharmacology. 2010;212(3):431–9.
  61. 61. Fahlke C, Berggren U, Berglund KJ, Zetterberg H, Blennow K, Engel JA, Balldin J. Neuroendocrine assessment of serotonergic, dopaminergic, and noradrenergic functions in alcohol‐dependent individuals. Alcoholism: Clinical and Experimental Research. 2012;36(1):97–103.
  62. 62. Ossewaarde L, Verkes RJ, Hermans EJ, Kooijman SC, Urner M, Tendolkar I, van Wingen GA, Fernandez G. Two‐week administration of the combined serotonin‐noradrenaline reuptake inhibitor duloxetine augments functioning of mesolimbic incentive processing circuits. Biological Psychiatry. 2011;70(6):568–74.
  63. 63. Orejarena MJ, Lanfumey L, Maldonado R, Robledo P. Involvement of 5‐HT2A receptors in MDMA reinforcement and cue‐induced reinstatement of MDMA‐seeking behaviour. International Journal of Neuropsychopharmacology. 2011;14(7):927–40.
  64. 64. Gomez‐Milanes I, Almela P, Garcia‐Carmona JA, Garcia‐Gutierrez MS, Aracil‐Fernandez A, Manzanares J, Milanes Maquilon MV, Laorden ML. Accumbal dopamine, noradrenaline and serotonin activity after naloxone‐conditioned place aversion in morphine‐dependent mice. Neurochemistry International. 2012;61(3):433–40.
  65. 65. Fallon S, Shearman E, Sershen H, Lajtha A. Food reward‐induced neurotransmitter changes in cognitive brain regions. Neurochemical Research. 2007;32(10):1772–82.
  66. 66. Dzirasa K, Phillips HW, Sotnikova TD, Salahpour A, Kumar S, Gainetdinov RR, Caron MG, Nicolelis MA. Noradrenergic control of cortico‐striato‐thalamic and mesolimbic cross‐structural synchrony. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2010;30(18):6387–97.
  67. 67. McCool BA, Christian DT, Fetzer JA, Chappell AM. Lateral/basolateral amygdala serotonin type‐2 receptors modulate operant self‐administration of a sweetened ethanol solution via inhibition of principal neuron activity. Frontiers in Integrative Neuroscience. 2014;8:5.
  68. 68. Tran L, Lasher BK, Young KA, Keele NB. Depletion of serotonin in the basolateral amygdala elevates glutamate receptors and facilitates fear‐potentiated startle. Translational Psychiatry. 2013;3:e298.
  69. 69. Roozendaal B, Nguyen BT, Power AE, McGaugh JL. Basolateral amygdala noradrenergic influence enables enhancement of memory consolidation induced by hippocampal glucocorticoid receptor activation. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(20):11642–7.
  70. 70. Pare D. Role of the basolateral amygdala in memory consolidation. Progress in Neurobiology. 2003;70(5):409–20.
  71. 71. Zhang J, Muller JF, McDonald AJ. Noradrenergic innervation of pyramidal cells in the rat basolateral amygdala. Neuroscience. 2013;228:395–408.
  72. 72. Commons KG, Connolley KR, Valentino RJ. A neurochemically distinct dorsal raphe‐limbic circuit with a potential role in affective disorders. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2003;28(2):206–15.
  73. 73. Le AD, Harding S, Juzytsch W, Fletcher PJ, Shaham Y. The role of corticotropin‐releasing factor in the median raphe nucleus in relapse to alcohol. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2002;22(18):7844–9.
  74. 74. Bonkale WL, Turecki G, Austin MC. Increased tryptophan hydroxylase immunoreactivity in the dorsal raphe nucleus of alcohol‐dependent, depressed suicide subjects is restricted to the dorsal subnucleus. Synapse. 2006;60(1):81–5.
  75. 75. Ketcherside A, Mathews I, Filbey F. The serotonin link between alcohol use and 25 affective disorders. Journal of Addiction and Prevention. 2013;1(2).3p. n/a-n/a.
  76. 76. Andrade AL, Abrahao KP, Goeldner FO, Souza‐Formigoni ML. Administration of the 5‐HT2C receptor antagonist SB‐242084 into the nucleus accumbens blocks the expression of ethanol‐induced behavioral sensitization in Albino Swiss mice. Neuroscience. 2011;189:178–86.
  77. 77. Ding ZM, Oster SM, Hauser SR, Toalston JE, Bell RL, McBride WJ, Rodd ZA. Synergistic self‐administration of ethanol and cocaine directly into the posterior ventral tegmental area: involvement of serotonin‐3 receptors. The Journal of Pharmacology and Experimental Therapeutics. 2012;340(1):202–9.
  78. 78. Burnett EJ, Grant KA, Davenport AT, Hemby SE, Friedman DP. The effects of chronic ethanol self‐administration on hippocampal 5‐HT 1A receptors in monkeys. Drug and Alcohol Dependence. 2014;136:135–142.
  79. 79. Kasper JM. Characterization of the effects of novel 5‐HT2C receptor agonists on neurotransmission and voluntary alcohol consumption in rats. Thesis, University of Florida; 2013.
  80. 80. Sari Y. Role of 5‐hydroxytryptamine 1B (5‐HT1B) receptors in the regulation of ethanol intake in rodents. Journal of Psychopharmacology. 2013;27(1):3–12.
  81. 81. Yang, J. and M.D. Li, Association and Interaction Analyses of 5-HT3 Receptor and Serotonin Transporter Genes with Alcohol, Cocaine, and Nicotine Dependence Using the SAGE Data. Human genetics, 2014. 133(7): p. 905-918. .
  82. 82. De Bruin N, McCreary A, Van Loevezijn A, De Vries T, Venhorst J, Van Drimmelen M, Kruse C. A novel highly selective 5‐HT 6 receptor antagonist attenuates ethanol and nicotine seeking but does not affect inhibitory response control in Wistar rats. Behavioural Brain Research. 2013;236:157–65.
  83. 83. Smith RJ, Aston‐Jones G. Noradrenergic transmission in the extended amygdala: role in increased drug‐seeking and relapse during protracted drug abstinence. Brain Structure and Function. 2008;213(1–2):43–61.
  84. 84. Becker HC. Effects of alcohol dependence and withdrawal on stress responsiveness and alcohol consumption. Alcohol Research. 2012;34(4):448–58.
  85. 85. Howes LG, Reid JL. Changes in plasma free 3,4‐dihydroxyphenylethylene glycol and noradrenaline levels after acute alcohol administration. Clinical Science. 1985;69(4):423–8.
  86. 86. Higley AE, Koob GF, Mason BJ. Treatment of alcohol dependence with drug antagonists of the stress response. Alcohol Research. 2012;34(4):516–21.
  87. 87. Le AD, Harding S, Juzytsch W, Funk D, Shaham Y. Role of alpha‐2 adrenoceptors in stress‐induced reinstatement of alcohol seeking and alcohol self‐administration in rats. Psychopharmacology. 2005;179(2):366–73.
  88. 88. Walker BM, Rasmussen DD, Raskind MA, Koob GF. alpha1‐noradrenergic receptor antagonism blocks dependence‐induced increases in responding for ethanol. Alcohol. 2008;42(2):91–7.
  89. 89. Simpson TL, Saxon AJ, Meredith CW, Malte CA, McBride B, Ferguson LC, Gross CA, Hart KL, Raskind M. A pilot trial of the alpha‐1 adrenergic antagonist, prazosin, for alcohol dependence. Alcoholism: Clinical and Experimental Research. 2009;33(2):255–63.
  90. 90. Walsh JJ, Friedman AK, Sun H, Heller EA, Ku SM, Juarez B, Burnham VL, Mazei‐Robison MS, Ferguson D, Golden SA, Koo JW, Chaudhury D, Christoffel DJ, Pomeranz L, Friedman JM, Russo SJ, Nestler EJ, Han MH. Stress and CRF gate neural activation of BDNF in the mesolimbic reward pathway. Nature Neuroscience. 2014;17(1):27–9.
  91. 91. Philbert J, Pichat P, Palme R, Belzung C, Griebel G. The CRF(1) receptor antagonist SSR125543 attenuates long‐term cognitive deficit induced by acute inescapable stress in mice, independently from the hypothalamic pituitary adrenal axis. Pharmacology, Biochemistry, and Behavior. 2012;102(3):415–22.
  92. 92. Lemos JC, Wanat MJ, Smith JS, Reyes BA, Hollon NG, Van Bockstaele EJ, Chavkin C, Phillips PE. Severe stress switches CRF action in the nucleus accumbens from appetitive to aversive. Nature. 2012;490(7420):402–6.
  93. 93. Huang MM, Overstreet DH, Knapp DJ, Angel R, Wills TA, Navarro M, Rivier J, Vale W, Breese GR. 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. The Journal of Pharmacology and Experimental Therapeutics. 2010;332(1):298–307.
  94. 94. Hauger RL, Risbrough V, Oakley RH, Olivares‐Reyes JA, Dautzenberg FM. Role of CRF receptor signaling in stress vulnerability, anxiety, and depression. Annals of the New York Academy of Sciences. 2009;1179:120–43.
  95. 95. Koob G, Kreek MJ. Stress, dysregulation of drug reward pathways, and the transition to drug dependence. American Journal of Psychiatry. 2007;164(8):1149–59.
  96. 96. Hammack SE, Guo JD, Hazra R, Dabrowska J, Myers KM, Rainnie DG. The response of neurons in the bed nucleus of the stria terminalis to serotonin: implications for anxiety. Progress in Neuro‐Psychopharmacology & Biological Psychiatry. 2009;33(8):1309–20.
  97. 97. Wenzel JM, Cotten SW, Dominguez HM, Lane JE, Shelton K, Su ZI, Ettenberg A. Noradrenergic beta‐receptor antagonism within the central nucleus of the amygdala or bed nucleus of the stria terminalis attenuates the negative/anxiogenic effects of cocaine. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2014;34(10):3467–74.
  98. 98. Flavin SA, Winder DG. Noradrenergic control of the bed nucleus of the stria terminalis in stress and reward. Neuropharmacology. 2013;70:324–30.
  99. 99. Leri F, Flores J, Rodaros D, Stewart J. Blockade of stress‐induced but not cocaine‐induced reinstatement by infusion of noradrenergic antagonists into the bed nucleus of the stria terminalis or the central nucleus of the amygdala. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2002;22(13):5713–8.
  100. 100. Le AD, Funk D, Coen K, Li Z, Shaham Y. Role of corticotropin‐releasing factor in the median raphe nucleus in yohimbine‐induced reinstatement of alcohol seeking in rats. Addiction Biology. 2013;18(3):448–51.
  101. 101. Dzung Le A, Funk D, Harding S, Juzytsch W, Fletcher PJ. The role of noradrenaline and 5‐hydroxytryptamine in yohimbine‐induced increases in alcohol‐seeking in rats. Psychopharmacology. 2009;204(3):477–88.
  102. 102. Marinelli PW, Funk D, Juzytsch W, Harding S, Rice KC, Shaham Y, Le A. The CRF1 receptor antagonist antalarmin attenuates yohimbine‐induced increases in operant alcohol self‐administration and reinstatement of alcohol seeking in rats. Psychopharmacology. 2007;195(3):345–55.
  103. 103. Funk CK, Koob GF. A CRF(2) agonist administered into the central nucleus of the amygdala decreases ethanol self‐administration in ethanol‐dependent rats. Brain Research. 2007;1155:172–8.
  104. 104. Krettek JE, Price JL. A description of the amygdaloid complex in the rat and cat with observations on intra‐amygdaloid axonal connections. The Journal of Comparative Neurology. 1978;178(2):255–79.
  105. 105. Davis M, Rainnie D, Cassell M. Neurotransmission in the rat amygdala related to fear and anxiety. Trends in Neurosciences. 1994;17(5):208–14.
  106. 106. Cardinal RN, Parkinson JA, Hall J, Everitt BJ. Emotion and motivation: the role of the amygdala, ventral striatum, and prefrontal cortex. Neuroscience & Biobehavioral Reviews. 2002;26(3):321–52.
  107. 107. Cahill L, Weinberger NM, Roozendaal B, McGaugh JL. Is the amygdala a locus of “conditioned fear”? Some questions and caveats. Neuron. 1999;23(2):227–8.
  108. 108. Johnston JB. Further contributions to the study of the evolution of the forebrain. The Journal of Comparative Neurology. 1923;35(5):337–481.
  109. 109. Roozendaal B, McEwen BS, Chattarji S. Stress, memory and the amygdala. Nature Reviews Neuroscience. 2009;10(6):423–33.
  110. 110. Paré D, Smith Y, Paré JF. Intra‐amygdaloid projections of the basolateral and basomedial nuclei in the cat: phaseolus vulgaris‐leucoagglutinin anterograde tracing at the light and electron microscopic level. Neuroscience. 1995;69(2):567–83.
  111. 111. Dong H‐W, Petrovich GD, Swanson LW. Topography of projections from amygdala to bed nuclei of the stria terminalis. Brain Research Reviews. 2001;38(1–2):192–246.
  112. 112. Petrovich GD, Canteras NS, Swanson LW. Combinatorial amygdalar inputs to hippocampal domains and hypothalamic behavior systems. Brain Research Reviews. 2001;38(1–2):247–89.
  113. 113. McDonald AJ. Topographical organization of amygdaloid projections to the caudatoputamen, nucleus accumbens, and related striatal‐like areas of the rat brain. Neuroscience. 1991;44(1):15–33.
  114. 114. Ray JP, Price JL. The organization of the thalamocortical connections of the mediodorsal thalamic nucleus in the rat, related to the ventral forebrain–prefrontal cortex topography. The Journal of Comparative Neurology. 1992;323(2):167–97.
  115. 115. Ottersen OP, Fischer BO, Rinvik E, Storm‐Mathisen J. Putative amino acid transmitters in the amygdala. Advances in Experimental Medicine and Biology. 1986;203:53–66.
  116. 116. Amaral D, Insausti R. Retrograde transport of D‐[3H]‐aspartate injected into the monkey amygdaloid complex. Experimental Brain Research. 1992;88(2):375–88.
  117. 117. Killcross S, Robbins TW, Everitt BJ. Different types of fear‐conditioned behaviour mediated by separate nuclei within amygdala. Nature. 1997;388(6640):377–80.
  118. 118. Nomura M, Izaki Y, Takita M, Tanaka J, Hori K. Extracellular level of basolateral amygdalar dopamine responding to reversal of appetitive‐conditioned discrimination in young and old rats. Brain Research. 2004;1018(2):241–6.
  119. 119. Ramirez DR, Savage LM. Differential involvement of the basolateral amygdala, orbitofrontal cortex, and nucleus accumbens core in the acquisition and use of reward expectancies. Behavioral Neuroscience. 2007;121(5):896–906.
  120. 120. Meil WM, See RE. Lesions of the basolateral amygdala abolish the ability of drug associated cues to reinstate responding during withdrawal from self‐administered cocaine. Behavioural Brain Research. 1997;87(2):139–48.
  121. 121. Valizadegan F, Oryan S, Nasehi M, Zarrindast MR. Interaction between morphine and noradrenergic system of basolateral amygdala on anxiety and memory in the elevated plus‐maze test based on a test‐retest paradigm. Archives of Iranian Medicine. 2013;16(5):281–7.
  122. 122. Varodayan, F.P., et al., Chronic alcohol exposure disrupts CB1 regulation of GABAergic transmission in the rat basolateral amygdala. Addiction Biology, 2016: p. n/a-n/a.In press.
  123. 123. Sciascia JM, Reese RM, Janak PH, Chaudhri N. Alcohol‐seeking triggered by discrete pavlovian cues is invigorated by alcohol contexts and mediated by glutamate signaling in the basolateral amygdala. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2015;40(12):2801–12.
  124. 124. Gass JT, Sinclair CM, Cleva RM, Widholm JJ, Olive MF. Alcohol‐seeking behavior is associated with increased glutamate transmission in basolateral amygdala and nucleus accumbens as measured by glutamate‐oxidase‐coated biosensors. Addiction Biology. 2011;16(2):215–28.
  125. 125. Porrino LJ, Williams‐Hemby L, Whitlow C, Bowen C, Samson HH. Metabolic mapping of the effects of oral alcohol self‐administration in rats. Alcoholism: Clinical and Experimental Research. 1998;22(1):176–82.
  126. 126. Läck AK, Diaz MR, Chappell A, DuBois DW, McCool BA. Chronic ethanol and withdrawal differentially modulate pre‐ and post‐synaptic function at glutamatergic synapses in rat basolateral amygdala. Journal of Neurophysiology. 2007;98(6):3185–96.
  127. 127. Rassnick S, Pulvirenti L, Koob GF. Oral ethanol self‐administration in rats is reduced by the administration of dopamine and glutamate receptor antagonists into the nucleus accumbens. Psychopharmacology. 1992;109(1):92–8.
  128. 128. Hyytia P, Kiianmaa K. Suppression of ethanol responding by centrally administered CTOP and naltrindole in AA and Wistar rats. Alcoholism: Clinical and Experimental Research. 2001;25(1):25–33.
  129. 129. Christian DT, Alexander NJ, Diaz MR, McCool BA. Thalamic glutamatergic afferents into the rat basolateral amygdala exhibit increased presynaptic glutamate function following withdrawal from chronic intermittent ethanol. Neuropharmacology. 2013;65:134–42.
  130. 130. Enquist J, Ferwerda M, Madhavan A, Hok D, Whistler JL. Chronic ethanol potentiates the effect of neuropeptide s in the basolateral amygdala and shows increased anxiolytic and anti‐depressive effects. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2012;37(11):2436–45.
  131. 131. Marshall SA, Casachahua JD, Rinker JA, Blose AK, Lysle DT, Thiele TE. IL‐1 receptor signaling in the basolateral amygdala modulates binge‐like ethanol consumption in male C57BL/6J mice. Brain, Behavior, and Immunity. 2016;51:258–67.
  132. 132. Sinclair CM, Cleva RM, Hood LE, Olive MF, Gass JT. mGluR5 receptors in the basolateral amygdala and nucleus accumbens regulate cue‐induced reinstatement of ethanol‐seeking behavior. Pharmacology, Biochemistry, and Behavior. 2012;101(3):329–35.
  133. 133. Chaudhri N, Woods CA, Sahuque LL, Gill TM, Janak PH. Unilateral inactivation of the basolateral amygdala attenuates context‐induced renewal of pavlovian‐conditioned alcohol‐seeking. The European Journal of Neuroscience. 2013;38(5):2751–61.
  134. 134. McLaughlin J, See RE. Selective inactivation of the dorsomedial prefrontal cortex and the basolateral amygdala attenuates conditioned‐cued reinstatement of extinguished cocaine‐seeking behavior in rats. Psychopharmacology. 2003;168(1–2):57–65.
  135. 135. Fuchs RA, Feltenstein MW, See RE. The role of the basolateral amygdala in stimulus‐reward memory and extinction memory consolidation and in subsequent conditioned cued reinstatement of cocaine seeking. The European Journal of Neuroscience. 2006;23(10):2809–13.
  136. 136. Di Ciano P, Everitt BJ. Direct interactions between the basolateral amygdala and nucleus accumbens core underlie cocaine‐seeking behavior by rats. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2004;24(32):7167–73.
  137. 137. Alderson HL, Robbins TW, Everitt BJ. The effects of excitotoxic lesions of the basolateral amygdala on the acquisition of heroin‐seeking behaviour in rats. Psychopharmacology. 2000;153(1):111–9.
  138. 138. Radwanska K, Wrobel E, Korkosz A, Rogowski A, Kostowski W, Bienkowski P, Kaczmarek L. Alcohol relapse induced by discrete cues activates components of AP‐1 transcription factor and ERK pathway in the rat basolateral and central amygdala. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2008;33(8):1835–46.
  139. 139. Wilden J, Hauser S, Deehanjr G, Ding Z‐M, Truitt W, Mcbride W, Rodd Z. The basolateral amygdala is a critical structure for environmental cue augmentation of EtOH‐seeking. 2012.
  140. 140. Marinelli PW, Funk D, Juzytsch W, Le AD. Opioid receptors in the basolateral amygdala but not dorsal hippocampus mediate context‐induced alcohol seeking. Behavioural Brain Research. 2010;211(1):58–63.
  141. 141. Gass JT, Sinclair CM, Cleva RM, Widholm JJ, Olive MF. Alcohol‐seeking behavior is associated with increased glutamate transmission in basolateral amygdala and nucleus accumbens as measured by glutamate‐oxidase‐coated biosensors. Addiction Biology. 2011;16(2):215–28.
  142. 142. Silberman Y, Matthews RT, Winder DG. A corticotropin releasing factor pathway for ethanol regulation of the ventral tegmental area in the bed nucleus of the stria terminalis. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2013;33(3):950–60.
  143. 143. Simms JA, Haass‐Koffler CL, Bito‐Onon J, Li R, Bartlett SE. Mifepristone in the central nucleus of the amygdala reduces yohimbine stress‐induced reinstatement of ethanol‐seeking. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology. 2012;37(4):906–18.
  144. 144. Valdez GR, Sabino V, Koob GF. Increased anxiety‐like behavior and ethanol self‐administration in dependent rats: reversal via corticotropin‐releasing factor‐2 receptor activation. Alcoholism: Clinical and Experimental Research. 2004;28(6):865–72.
  145. 145. Maroun M, Ioannides PJ, Bergman KL, Kavushansky A, Holmes A, Wellman CL. Fear extinction deficits following acute stress associate with increased spine density and dendritic retraction in basolateral amygdala neurons. The European Journal of Neuroscience. 2013;38(4):2611–20.
  146. 146. Rei D, Mason X, Seo J, Gräff J, Rudenko A, Wang J, Rueda R, Siegert S, Cho S, Canter RG, Mungenast AE, Deisseroth K, Tsai L‐H. Basolateral amygdala bidirectionally modulates stress‐induced hippocampal learning and memory deficits through a p25/Cdk5‐dependent pathway. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(23):7291–6.
  147. 147. Rau AR, Chappell AM, Butler TR, Ariwodola OJ, Weiner JL. Increased basolateral amygdala pyramidal cell excitability may contribute to the anxiogenic phenotype induced by chronic early‐life stress. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2015;35(26):9730–40.
  148. 148. Zhang W, Rosenkranz JA. Repeated restraint stress increases basolateral amygdala neuronal activity in an age‐dependent manner. Neuroscience. 2012;226:459–74.
  149. 149. Silberman Y, Bajo M, Chappell AM, Christian DT, Cruz M, Diaz MR, Kash T, Lack AK, Messing RO, Siggins GR, Winder D, Roberto M, McCool BA, Weiner JL. Neurobiological mechanisms contributing to alcohol–stress–anxiety interactions. Alcohol. 2009;43(7):509–19.
  150. 150. Baculis BC, Diaz MR, Valenzuela CF. Third trimester‐equivalent ethanol exposure increases anxiety‐like behavior and glutamatergic transmission in the basolateral amygdala. Pharmacology, Biochemistry, and Behavior. 2015;137:78–85.
  151. 151. Weitemier AZ, Ryabinin AE. Alcohol‐induced memory impairment in trace fear conditioning: a hippocampus‐specific effect. Hippocampus. 2003;13(3):305–15.
  152. 152. Bertotto ME, Bustos SG, Molina VA, Martijena ID. Influence of ethanol withdrawal on fear memory: effect of d‐cycloserine. Neuroscience. 2006;142(4):979–90.
  153. 153. Holmes A, Fitzgerald PJ, MacPherson KP, DeBrouse L, Colacicco G, Flynn SM, Masneuf S, Pleil KE, Li C, Marcinkiewcz CA, Kash TL, Gunduz‐Cinar O, Camp M. Chronic alcohol remodels prefrontal neurons and disrupts NMDAR‐mediated fear extinction encoding. Nature neuroscience. 2012;15(10):1359–61.
  154. 154. Quinones‐Laracuente K, Hernandez‐Rodriguez MY, Bravo‐Rivera C, Melendez RI, Quirk GJ. The effect of repeated exposure to ethanol on pre‐existing fear memories in rats. Psychopharmacology. 2015;232(19):3615–22.
  155. 155. Le AD, Funk D, Juzytsch W, Coen K, Navarre BM, Cifani C, Shaham Y. Effect of prazosin and guanfacine on stress‐induced reinstatement of alcohol and food seeking in rats. Psychopharmacology. 2011;218(1):89–99.
  156. 156. Marcinkiewcz, C.A., Serotonergic Systems In The Pathophysiology Of Ethanol Dependence: Relevance To Clinical Alcoholism. ACS Chem Neurosci, 2015. 6(7): p. 1026-39.
  157. 157. McBride WJ, Guan XM, Chernet E, Lumeng L, Li TK. Regional serotonin1A receptors in the CNS of alcohol‐preferring and ‐nonpreferring rats. Pharmacology, Biochemistry, and Behavior. 1994;49(1):7–12.
  158. 158. Muller JF, Mascagni F, McDonald AJ. Serotonin‐immunoreactive axon terminals innervate pyramidal cells and interneurons in the rat basolateral amygdala. The Journal of Comparative Neurology. 2007;505(3):314–35.
  159. 159. Wang SJ, Cheng LL, Gean PW. Cross‐modulation of synaptic plasticity by beta‐adrenergic and 5‐HT1A receptors in the rat basolateral amygdala. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 1999;19(2):570–7.
  160. 160. Barbara Ferry and Gina L. Quirarte (2012). Role of Norepinephrine in Modulating Inhibitory Avoidance Memory Storage: Critical Involvement of the Basolateral Amygdala, The Amygdala - A Discrete Multitasking Manager, Dr. Barbara Ferry (Ed.), InTech, DOI: 10.5772/53246. Available from: http://www.intechopen.com/books/the-amygdala-a-discrete-multitasking-manager/role-of-norepinephrine-in-modulating-inhibitory-avoidance-memory-storage-critical-involvement-of-the.
  161. 161. Ferry B, Roozendaal B, McGaugh JL. Basolateral amygdala noradrenergic influences on memory storage are mediated by an interaction between β‐ and α1‐adrenoceptors. Journal of Neuroscience. 1999;19(12):5119–23.
  162. 162. Barak S, Liu F, Hamida SB, Yowell QV, Neasta J, Kharazia V, Janak PH, Ron D. Disruption of alcohol‐related memories by mTORC1 inhibition prevents relapse. Nature Neuroscience. 2013;16(8):1111–7.
  163. 163. Butler TR, Chappell AM, Weiner JL. Effect of beta3 adrenoceptor activation in the basolateral amygdala on ethanol seeking behaviors. Psychopharmacology. 2014;231(1):293–303.
  164. 164. Hoffman PL, Valverius P, Kwast M, Tabakoff B. Comparison of the effects of ethanol on beta‐adrenergic receptors in heart and brain. Alcohol and Alcoholism (Oxford, Oxfordshire) Supplement. 1987;1:749–54.
  165. 165. Rainnie DG. Serotonergic modulation of neurotransmission in the rat basolateral amygdala. Journal of Neurophysiology. 1999;82(1):69–85.
  166. 166. Cheng LL, Wang SJ, Gean PW. Serotonin depresses excitatory synaptic transmission and depolarization‐evoked Ca2+ influx in rat basolateral amygdala via 5‐HT1A receptors. The European Journal of Neuroscience. 1998;10(6):2163–72.
  167. 167. de Andrade Strauss CV, Vicente MA, Zangrossi Jr H. Activation of 5‐HT1A receptors in the rat basolateral amygdala induces both anxiolytic and antipanic‐like effects. Behavioural Brain Research. 2013;246:103–10.
  168. 168. Nevo I, Langlois X, Laporte AM, Kleven M, Koek W, Lima L, Maudhuit C, Martres MP, Hamon M. Chronic alcoholization alters the expression of 5‐HT1A and 5‐HT1B receptor subtypes in rat brain. The European Journal of Pharmacology. 1995;281(3):229–39.
  169. 169. Christianson JP, Ragole T, Amat J, Greenwood BN, Strong PV, Paul ED, Fleshner M, Watkins LR, Maier SF. 5‐Hydroxytryptamine 2C receptors in the basolateral amygdala are involved in the expression of anxiety after uncontrollable traumatic stress. Biological Psychiatry. 2010;67(4):339–45.
  170. 170. Gonzalez LE, Andrews N, File SE. 5‐HT1A and benzodiazepine receptors in the basolateral amygdala modulate anxiety in the social interaction test, but not in the elevated plus‐maze. Brain Research. 1996;732(1–2):145–53.
  171. 171. McCool BA, Frye GD, Pulido MD, Botting SK. Effects of chronic ethanol consumption on rat GABA(A) and strychnine‐sensitive glycine receptors expressed by lateral/basolateral amygdala neurons. Brain Research. 2003;963(1–2):165–77.
  172. 172. Patkar, O.L., et al., The antihypertensive drug pindolol attenuates long-term but not short-term binge-like ethanol consumption in mice. Addiction Biology, 2016: p. n/a-n/a. In press. 2016.
  173. 173. Kraus ML, Gottlieb LD, Horwitz RI, Anscher M. Randomized clinical trial of atenolol in patients with alcohol withdrawal. The New England Journal of Medicine. 1985;313(15):905–9.
  174. 174. Horwitz RI, Gottlieb LD, Kraus ML. The efficacy of atenolol in the outpatient management of the alcohol withdrawal syndrome. Results of a randomized clinical trial. Archives of Internal Medicine. 1989;149(5):1089–93.
  175. 175. Johnsson G, Regardh CG. Clinical pharmacokinetics of beta‐adrenoreceptor blocking drugs. Clinical Pharmacokinetics. 1976;1(4):233–63.
  176. 176. Carlsson C, Johansson T. The psychological effects of propranolol in the abstinence phase of chronic alcoholics. The British Journal of Psychiatry: The Journal of Mental Science. 1971;119(553):605–6.
  177. 177. Malcolm R, Anton RF, Randall CL, Johnston A, Brady K, Thevos A. A placebo‐controlled trial of buspirone in anxious inpatient alcoholics. Alcoholism: Clinical and Experimental Research. 1992;16(6):1007–13.
  178. 178. Kranzler HR, Burleson JA, Del Boca FK, Babor TF, Korner P, Brown J, Bohn MJ. Buspirone treatment of anxious alcoholics. A placebo‐controlled trial. Archives of General Psychiatry. 1994;51(9):720–31.

Written By

Omkar L. Patkar, Arnauld Belmer and Selena E. Bartlett

Submitted: 25 October 2015 Reviewed: 04 March 2016 Published: 14 July 2016