Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Dec 1.
Published in final edited form as: Pharmacol Ther. 2022 Oct 30;240:108299. doi: 10.1016/j.pharmthera.2022.108299

Neurosteroids (allopregnanolone) and Alcohol Use disorder: From mechanisms to potential pharmacotherapy

Eleonora Gatta 1, Diletta Camussi 1, James Auta 1, Alessandro Guidotti 1, Subhash C Pandey 1,2
PMCID: PMC9810076  NIHMSID: NIHMS1849185  PMID: 36323379

Abstract

Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder that is commonly comorbid with psychiatric disorders, including anxiety. Alcohol exposure produces a plethora of effects on neurobiology. Currently, therapeutic strategies are limited, and only a few treatments — disulfiram, acamprosate, and naltrexone — are available. Given the complexity of this disorder, there is a great need for the identification of novel targets to develop new pharmacotherapy. The GABAergic system, the primary inhibitory system in the brain, is one of the well-known targets for alcohol and is responsible for the anxiolytic effects of alcohol. Interestingly, GABAergic neurotransmission is fine-tuned by neuroactive steroids that exert a regulatory role on several endocrine systems involved in neuropsychiatric disorders including AUD. Mounting evidence indicates that alcohol alters the biosynthesis of neurosteroids, whereas acute alcohol increases and chronic alcohol decreases allopregnanolone levels. Our recent work highlighted that chronic alcohol-induced changes in neurosteroid levels are mediated by epigenetic modifications, e.g., DNA methylation, affecting key enzymes involved in neurosteroid biosynthesis. These changes were associated with changes in GABAA receptor subunit expression, suggesting an imbalance between excitatory and inhibitory signaling in AUD. This review will recapitulate the role of neurosteroids in the regulation of the neuroendocrine system, highlight their role in the observed allostatic load in AUD, and develop a framework from mechanisms to potential pharmacotherapy.

Keywords: GABA receptors, neurosteroids, alcohol use disorder, stress, HPA axis

1. Introduction

Alcohol Use Disorder (AUD) is a multifaceted relapsing disorder, one of the most prevalent psychiatric disorders (Grant et al., 2004; Rehm et al., 2015), and a considerable economic burden (Rehm and Shield, 2019). According to a recent survey, 14.5 million people over the age of 12 had an AUD in the United states in 2019 (National Survey on Drug Use and Health, SAMHSA), a number that has significantly worsened during the COVID-19 pandemic (Pollard et al., 2020; Yazdi et al., 2020). Individuals suffering from AUD have an increased vulnerability to develop other psychiatric disorders, including depression and anxiety (Birrell et al., 2015; Lai et al., 2015; Swendsen et al., 1998) and alterations of executive functions (Fernández-Serrano et al., 2010). Conversely, negative emotional states, including stress and anxiety, lead to higher consumption of alcohol (Koob, 2015; Peltier et al., 2019; Pandey et al., 2017). The complex interaction of environmental and genetic factors can predispose an individual to the development of AUD (Schuebel et al., 2016; Starkman et al., 2012). While early evidence indicated an increased likelihood of developing the disease if AUD cases exist in the family (Cotton, 1979; Devor & Cloninger, 1989), specific loci mediating the risk to develop an AUD have been difficult to identify. Genome-wide association studies conducted in the past decades pointed to risk loci in genes involved in alcohol metabolism, including the genes for alcohol dehydrogenase (ADH) and the mitochondrial form of aldehyde dehydrogenase (ALDH2) (Carvalho et al., 2019; Sanchez-Roige et al., 2019). In addition, a growing body of evidence has implicated epigenetic mechanisms, e.g., DNA methylation, histone modifications and noncoding RNAs, in the development and maintenance of alcohol addiction (Bohnsack & Pandey, 2021; Gatta et al., 2021; Kyzar et al., 2022). Recent epigenome-wide association studies of alcohol consumption identified significant differentially methylated CpGs in the cystine/glutamate transporter SLC7A11 gene (Lohoff et al., 2021) and in glucocorticoid signaling genes (Lohoff et al., 2020) in individuals with AUD when compared to controls, suggesting that alterations in excitatory pathways along with environmental stimuli are key players in the disease. The dysregulations of the neurocircuitry underpinning AUD have been characterized as an ‘allostatic load,’ a term coined by Dr. Bruce S. McEwen to define an inadequate and pathological response to chronic stress with excessive release of stress hormones and the emergence of a pro-inflammatory state (McEwen, 1998; McEwen & Stellar, 1993). This concept has been extended to AUD (Koob, 2003). Indeed, chronic alcohol use is known to affect both the reward and stress systems (Gatta et al., 2019; Koob, 2015) and impair numerous physiological functions including immune response (Crews & Vetreno, 2014). While there is a growing understanding of the neurobiological mechanisms of AUD, only a few therapeutic strategies are currently available for the treatment of AUD, i.e., disulfiram, acamprosate and naltrexone (O’Malley & O’Connor, 2011). Given the complex pathophysiology of AUD (Koob & Lemoal,1997; Pandey et al., 2017), there is a great need for the identification of new targets to develop improved pharmacotherapy for AUD. This review has discussed the role of neurosteroids, specifically allopregnanolone, in the regulation of the neuroendocrine system, in particular the stress system, and highlighted their role in the allostatic load observed in AUD. We also discussed how the neurosteroid system is epigenetically regulated by ethanol and whether this could serve as a potential target for pharmacotherapy development to prevent or treat AUD.

2. Stress systems in alcohol addiction: a spiraling interplay

Koob and Lemoal (1997) characterized addiction as a three stage cycle, i.e., preoccupation/anticipation, binge/intoxication, and withdrawal/negative affect. These stages contribute to the development of a pathological state by aggravating each other. The addiction cycle involves key brain regions that encode for reward, motivation as well as stress (Koob & Volkow, 2016), including, among others, the prefrontal cortex, the hypothalamus, the nucleus accumbens and the extended amygdala. It is important to note that all drugs of abuse stimulate the mesolimbic dopamine system that mediate their rewarding properties (Di Chiara & Imperato, 1988; Koob & Volkow, 2010). The transition from controlled to compulsive drinking is mediated by neuroadaptive changes of the dopaminergic mesolimbic and the GABAergic hypothalamic pathways resulting in an allostatic overload of both reward and neuroendocrine circuits (Sinha, 2013). The existence of a frequent comorbidity between stress-related disorders (Conway et al., 2006; Grant et al., 2015), including anxiety, depression and AUD, suggests common underlying mechanisms in the development of these psychiatric disorders.

Epidemiological studies have reported a long standing association between stress and AUD (Ayer et al., 2021; Becker, 2017; San José et al., 2000), with women being four times more likely to have an AUD diagnosis after a stressful life event when compared to men (Verplaetse et al., 2018). Environmental stress and adverse life events may be a trigger for excessive alcohol consumption (Enoch, 2011; Koob, 2008, 2013; Uhart & Wand, 2009), and some individuals drink alcohol in an attempt to self-medicate and attenuate stress-related symptoms (Becker, 2017; Bolton et al., 2009; Kushner et al., 2000). But, the early view of alcohol as an anxiolytic and tension-reducing agent (Cappell & Herman, 1972; Greeley & Oei, 1999) has been contrasted by evidence that higher doses of alcohol act as a stressor. Alcohol exposure and withdrawal from chronic alcohol activate the hypothalamic-pituitary-adrenal (HPA) axis (Blaine & Sinha, 2017; Richardson et al., 2008; Rose et al., 2010), leading to increased release of corticotropin-releasing factor (CRF), adrenocorticotropic hormone (ACTH) from the paraventricular nucleus (PVN) within the hypothalamus, as well as glucocorticoids released from the adrenals, which are key mediators of the neuroadaptations underlying addictive behaviors (Richardson et al., 2008; Rivier, 2014; Stephens & Wand, 2012). This increase is particularly evident as a subject goes through the drinking cycle with repeated episodes of alcohol intoxication and withdrawal (Adinoff, 1990; Adinoff et al., 2003). Moreover, individuals suffering from AUD develop a high tolerance to the rapid effects of alcohol, including sedation and motor impairments, and a higher vulnerability to withdrawal symptoms, e.g., anhedonia and anxiety (Becker, 2008; Chen et al., 2019; Hatzigiakoumis et al., 2011; Martinotti et al., 2008; Pandey et al., 2017). Hence, in recent decades, AUD has been defined as a stress surfeit disorder (Koob, 2008, 2013) with high comorbidities with other psychopathological conditions, such as major depression disorder (MDD), posttraumatic stress disorder (PTSD), and anxiety disorder (Grant et al., 2015).

The relationship between alcohol and stress has been shown to be bidirectional. Intracerebral infusion of glucocorticoids increases voluntary alcohol intake (Fahlke et al., 1996), while suppression of glucocorticoids release by adrenalectomy induces a reduction in alcohol intake in alcohol preferring rats (Fahlke & Eriksson, 2000). In alcohol-dependent rats, acute withdrawal was associated with a reduction of glucocorticoid receptor (GR, a key component of the stress system) expression in stress/reward brain regions, such as the prefrontal cortex and the amygdala (Roy et al., 2002; Vendruscolo et al., 2012). The use of a GR antagonist, i.e. mifepristone, prevented alcohol intake and compulsive behavior in both rodents and human subjects (Vendruscolo et al., 2012, 2015), strengthening the important overlap existing between stress and alcohol.

The development of mood and anxiety disorders appears to be a predictor of alcohol consumption (Moscato et al., 1997). Individuals suffering from stress-related disorders (e.g., MDD and PTSD) also show impairments in the reward system (Grant et al., 2004; Petrakis & Simpson, 2017; Russo & Nestler, 2013). The comorbidity of AUD with other psychiatric disorders leads to a worse prognosis than either disorder alone (Conway et al., 2006; Grant et al., 2004; Hasin et al., 2007). Moreover, there is a reciprocal interaction between these pathologies, with depression increasing the risk of AUD and vice versa (McHugh & Weiss, 2019). The relationship between alcohol and other psychiatric disorders is also relevant for PTSD. Individuals suffering from PTSD are 1.2 times more likely to also meet the criteria for AUD when compared to healthy controls (Simpson et al., 2019). This prevalence is significantly higher in veterans (Chen et al., 2018; Seal et al., 2011). It should be noted that alterations in the HPA axis are a common neurobiological underpinning for these disorders. Specifically, central and peripheral glucocorticoids levels have been key determinants in the development of a pathological state in both depression (Stetler & Miller, 2011; Young et al., 2001) and PTSD (Daskalakis et al., 2013). Preclinical models of PTSD show that female mice exposed to chronic stressors increase their alcohol self-administration when compared to males (Cozzoli et al., 2014). Similarly, exposure to a traumatic stress paradigm increased ethanol consumption in alcohol-dependent adult male mice (Piggott et al., 2020). These observations suggest that therapeutic strategies targeting the stress system could be promising for the treatment of co-occurring stress-related disorders.

3. Key role of altered GABAergic neurotransmission on the stress system

Neurosteroids and glucocorticoids are involved in number of crucial central and peripheral functions, including the regulation of metabolism, immune and inflammatory mechanisms as well as mood and anxiety disorders (Morrow et al., 2020). The biological mechanisms modulating the function of the stress system have been investigated for decades (Jones et al., 1976; Makara & Stark, 1974). Regulation of the HPA axis activity occurs via a bottom-up control exerted by glucocorticoids, which have both genomic and non-genomic actions (de Kloet et al., 2008). Specifically, the GR regulates stress response by acting as a negative feedback loop on the HPA axis, in addition to its metabolic and immune suppressant actions (de Kloet and Derijk, 2004; Silverman and Sternberg, 2012). Furthermore, GABAergic projections originating from the posterior bed nucleus of the stria terminalis (BNST) and the peri-PVN regions mediate the inhibitory control of the HPA axis (Herman, 2012). The limbic and forebrain regions, including the hippocampus and the prefrontal cortex, also regulate the HPA axis by projecting to GABAergic nuclei (Radley et al., 2006; Ulrich-Lai & Herman, 2009). GABA released by these projections primarily binds to synaptic and extra-synaptic GABAA receptors (GABAARs), which are expressed throughout the stress neurocircuitry (Cullinan et al., 2008; Maguire, 2014). The pharmacological and biophysical properties of GABAARs are determined by the combination of specific subunits, 19 identified so far (α1-6, β1-3, γ1-3, δ, ε, θ π and ρ1-3), which form a pentameric structure (Olsen & Sieghart, 2008, 2009) belonging to the family of ionotropic receptors, ligand-gated chloride anion channels (Nayeem et al., 1994). The expression profile of these subunits varies depending on the brain region; most GABAARs isoforms include two α, two β and a single γ, δ or ε subunit (Sente et al., 2022).

Two main GABAARs isoforms have been characterized (Fig. 1): synaptic receptors, which mediate phasic GABAergic transmission and mostly contain α, β and γ subunits, and the extrasynaptic receptors, which contain α, β and δ subunits responsible for tonic (Farrant & Nusser, 2005) and “spill-over” inhibition (Herd et al., 2013, Figure 1). It has recently been demonstrated that the neuronal location of GABAAR isoforms is the result of a dynamic trafficking between synaptic and extrasynaptic locations (Davenport et al., 2021; Wu et al., 2021). In addition, slow synaptic GABAergic inhibition is mediated by metabotropic GABAB receptors (Chebib & Johnston, 1999; Li & Slesinger, 2022), which are expressed both pre-and post-synaptically throughout the CNS.

Figure 1. GABAergic neurotransmission and modulation.

Figure 1.

GABA acts on both synaptic and extrasynaptic GABAA receptors (GABAARs) as well as on metabotropic GABAB receptors (GABABRs). Synaptic GABAARs are responsible for phasic current, which results from the releasing of a high, short-lasting concentration of GABA into the synaptic cleft. It acts on a relatively small number of GABA receptors on the postsynaptic membrane. Extrasynaptic GABAARs are, instead, responsible for tonic inhibition; a low concentration of GABA, persisting in the extracellular ambient despite the activity of GABA transaminases, is responsible for the tonic current. GABA tonically activates extrasynaptic high-affinity GABAARs, which are located far from the synaptic cleft. GABA binds between α and β subunits of both synaptic and extrasynaptic receptors. Neurosteroids bind at a site between α and β subunits of both synaptic and extrasynaptic receptors and show a greater affinity for the extrasynaptic receptor, where they act at nM concentrations. Several drugs known to be used in the treatment of alcohol use disorder (AUD) target GABAergic neurotransmission. Benzodiazepines (BDZ) act between α and γ subunits of synaptic receptors. Gabapentin binds the β subunit of the extrasynaptic receptor, while Topiramate only binds β 2/3 subunits. On the other hand, Sodium oxybate and Baclofen bind GABABR. Interestingly, ethanol binds both synaptic and extrasynaptic GABAARs and GABABRs.

Pharmacological studies have further highlighted the role of GABAergic neurotransmission in the inhibition of glucocorticoids release. Microinfusion of bicuculine, a GABAARs antagonist, into the PVN enhanced stress-induced glucocorticoid plasmatic levels, while the GABAARs agonist muscimol blunted the stress response in adult rats exposed to restraint stress (Cullinan, 1998). The importance of GABAergic neurotransmission in the regulation of the HPA axis is also supported by the evidence that γ2-deficient mice have neuroendocrine abnormalities which have been linked to HPA axis hyperactivity (Shen et al., 2010). γ2-containing receptors are the most abundant GABAARs in the brain, mediating fast phasic inhibition in response to synaptic GABA release (Belelli et al., 2021). Given the importance of HPA axis dysregulations in stress-related disorders, including depression and anxiety, modulators of the GABAergic transmission have been considered as putative therapeutic options. Considering that AUD has been labeled a “stress surfeit disorder” (Koob, 2013), this therapeutic strategy could also be promising for the treatment of alcohol dependence and withdrawal.

The balance between pro and anti-stress neuropeptides (e.g., CRF and Neuropeptide Y (NPY), respectively), which are highly expressed in the amygdala (Koob, 2015; Palmisano & Pandey, 2017), has also been shown to modulate inhibitory GABAergic currents. The central nucleus of the amygdala (CeA) is an anatomical structure functioning at the interface between the stress-system and addictive processes. Most neurons in the CeA co-release GABA and neuropeptides, which appear to have a key regulatory role in anxiety-like and alcohol-related behaviors (Walker, 2021). Both alcohol withdrawal and stress induce an increase in CRF production and release within the CeA, leading to stress-related behaviors via a robust increase in GABAergic transmission. Of note, CRF receptor 1 (CRF1) antagonists reverse this phenotype (Funk et al., 2007; Knapp et al., 2004; Overstreet et al., 2004). Conversely, NPY has a strong anxiolytic effect resulting from its action on both NPY presynaptic (Y2R) and postsynaptic (Y1R) receptors in both the CeA and the basolateral amygdala (BLA; Gilpin, 2012). Ablation of Y2R results in increased depression and anxiety-like behaviors, a response that might be associated with a dysregulation of GABAergic neurotransmission (Tasan et al., 2010). Thus, an ethanol withdrawal-induced decrease in NPY levels in the CeA may play a significant role in the increase of GABAergic tone in alcohol-dependent rats (Gilpin et al., 2011). Despite their opposite behavioral effects, NPY and CRF have a remarkably similar anatomical pattern of distribution; this, along with electrophysiological data, suggests that NPY and CRF interact on the regulation of the GABAergic output in the CeA leading to the development of chronic alcohol intake and AUD (Gilpin, 2012).

4. Modulatory role of neurosteroids (allopregnanolone) on the stress response

Neurosteroids are endogenous GABAAR positive allosteric neuromodulators and include allopregnanolone (3α,5α-tetrahydroprogesterone) and its potent stereoisomer pregnanolone (Belelli & Lambert, 2005; Figure 2). Unlike glucocorticoids, neurosteroids (progesterone metabolites 5α-pregnan-3α-tetrahydroprogesterone [allopregnanolone], 5β-pregnan-3α-tetrahydroprogesterone and the deoxycorticosterone [DOC] metabolite, 5α,3α-tetrahydrodeoxycorticosterone [5α3α-THDOC]) are produced in the periphery (predominantly by the adrenal cortex and the ovaries (Paul & Purdy, 1992)), in the peripheral (Giatti et al., 2015; Mensah-Nyagan et al., 2008; Patte-Mensah et al., 2006) as well as the central nervous system (Baulieu et al., 1981; Cheney et al., 1995; Do Rego et al., 2009) where they mediate both paracrine and autocrine actions (Agís-Balboa et al., 2006; Belelli & Lambert, 2005). In the CNS, neurosteroid biosynthesis begins with the transport of cholesterol within glial cells from the outer to the inner mitochondrial membrane by the 18kDa translocator protein (TSPO) (Costa & Guidotti, 1991; Rupprecht et al., 2009), and the steroidogenic acute regulatory (StAR) protein (Clark et al., 1994; Lacapere et al., 2020) (Fig. 2). While the origin of brain TSPO has been identified in a specific subtype of glial cells, e.g. microglia cells, recent evidence suggests that TSPO is also expressed in other brain cells, such as astrocytes and neurons (Nutma et al., 2021). Interestingly it has been demonstrated that numerous brain cells, including neurons, oligodendrocytes, pinealocytes and glial cells, possess all the enzymatic machinery necessary for the local biosynthesis of neurosteroids (Agís-Balboa et al., 2006; Baulieu & Robel, 1998; Kimoto et al., 2001; King et al., 2002; Lloyd-Evans & Waller-Evans, 2020; Melcangi et al., 1993; Mellon & Deschepper, 1993; Mellon & Vaudry, 2001). Although TSPO is expressed in glial cells, the synthesis of neurosteroids most likely occurs in neurons (Porcu et al., 2016). Additionally, diazepam binding inhibitor (DBI) binds to TSPO to stimulate the transport of cholesterol to the mitochondria, thus increasing the concentration of this rate limiting component for neurosteroid synthesis (Costa et al., 1994; Costa & Guidotti, 1991; Midzak et al., 2015). The mitochondrial cholesterol side-chain cleavage enzyme (P450scc) then converts cholesterol into pregnenolone, which is the precursor for all neurosteroids. Pregnenolone diffuses into the cytosol, where it is further metabolized into progesterone by the 3β-hydroxysteroid dehydrogenase (3β-HSD). Type-1 5α-reductase (5α-R1) and 3α-HSD then convert progesterone into allopregnanolone (Do Rego et al., 2009; Locci & Pinna, 2017). 5α-1 and 3α-HSD also convert peripherally derived glucocorticoid metabolite, DOC into 5α3α-THDOC (Karavolas & Hodges, 1990).

Figure 2. Alcohol-induced epigenetic modifications of neurosteroidogenesis at the GABAergic synapse.

Figure 2.

GABA is released from GABAergic interneurons and acts on synaptic (phasic current) and extrasynaptic (tonic current) GABAA receptors (GABAARs). GABAARs are modulated by the neurosteroids, including allopregnanolone. The synthesis of neurosteroids occurs both in the periphery (where they are produced from circulating progesterone) and the central nervous system. In the brain, neurosteroidogenesis starts in neurons or glial cells, where cholesterol is transported from the outer to the inner mitochondrial membrane by the translocator protein (TSPO) and is converted into pregnanolone. Pregnanolone is further transported into the cytosol and converted into Progesterone, 5α-Dihydroprogesterone and finally into allopregnanolone, which is a positive allosteric modulator of GABAARs. Both alcohol and stress activate the hypothalamic-pituitary-adrenal (HPA)-axis. Allopregnanolone acting on corticotropin releasing factor (CRF)-secreting neurons via GABAergic current-mediated mechanisms dampens the hyperactivation of the stress system. Chronic alcohol consumption alters GABAergic transmission and neurosteroidogenesis via epigenetic mechanisms, leading to the loss of inhibition of HPA-axis GABA-mediated control, physiologically modulated by allopregnanolone. PE=pregnanolone, Prog=progesterone, 5α-DHP=5α-dehydroprogesterone, TET=ten-eleven translocation methylcytosine dioxygenases, DNMT=DNA methyltransferase.

Neurosteroids exert their biological function by binding with high affinity to allosteric modulatory sites located on GABAARs. Unlike benzodiazepines, which exert their actions through different GABAARs subtypes (e.g. α1 and α2 exert an anxiolytic and sedative effect, while β3 exerts an anesthetic action), in vitro experiments have demonstrated that neurosteroids act at physiologically nanomolar concentrations on a variety of GABAARs subtypes (Lambert et al., 2003). In addition, α subunit of GABAARs does not influence the positive allosteric modulatory action of allopregnanolone on GABAARs-mediated Cl-influx, when expressed with β and γ subunit, strengthening the hypothesis that the neurosteroid modulatory effect on GABA complex action may be independent of the GABAARs complex subunit composition. Therefore, the modulatory effects of neurosteroids on GABAergic neurotransmission are dependent on cell and neuronal types, brain regions, and distribution density of GABAARs, expressed throughout the CNS. The binding of allopregnanolone to the transmembrane domains of α64 and δ subunits containing extrasynaptic GABAARs results in a highly efficacious positive allosteric modulatory GABAergic inhibitory neurotransmission (Concas et al., 1998; Hosie et al., 2006; Brickley & Mody, 2012; Callachan et al., 1987; Lambert et al., 1995; Shu et al., 2004). Indeed, the δ subunit of the GABAARs complex plays a pivotal role in the sensitivity of the receptor to the modulatory effect of neurosteroids on GABAergic neurotransmission. Most importantly, this subunit is highly expressed in thalamus, granule cells of the cerebellum, and hippocampal dentate gyrus, where it acts as a major component of the tonic inhibitory current (Follesa et al., 2015). Of note, in δ subunit knock-out mice, the modulatory effects of neurosteroids are significantly attenuated (Mihalek et al., 1999; Spigelman et al., 2003; Stell et al., 2003), and the replacement of γ subunit with δ subunit enhances the sensitivity of the receptor to neurosteroid modulatory actions (Belelli et al., 2002; Brown et al., 2016; Wohlfarth et al., 2002). A similar reduction in tonic inhibitory current was also reported in mice lacking the α4 subunit, which is often co-assembled with the δ subunit (Chandra et al., 2006).

By modulating neuronal activity, neurosteroids are also key players in the regulation of the stress response as they participate in a feed-forward process of the HPA axis (Tonhajzerova & Mestanik, 2017). Exposure to stressful stimuli has been shown to increase allopregnanolone levels in the plasma and the central nervous system (Barbaccia et al., 1996, 1997; Purdy et al., 1991; Serra et al., 2002). In vitro and in vivo experiments have demonstrated that allopregnanolone dampens the endocrine response to stress by reducing CRF, ACTH and glucocorticoids release (Owens et al., 1992; Patchev et al., 1994, 1996), resulting in anxiolytic and anti-depressant effects (Bitran et al., 1991, 1999; Carboni et al., 1996; Crawley et al., 1986). In mice, systemic administration of trilostane, a 3β-HSD inhibitor, reduced progesterone levels in the brain and produced antidepressant-like effects (Espallergues et al., 2012). Conversely, the administration of finasteride, a 5α-R1 inhibitor that blocks neurosteroidogenesis, increases anxiety/depressive-like behaviors (Pallarès et al., 2015; Yoshizawa et al., 2017). In addition to these neuromodulatory roles, early evidence also highlighted the transcriptional effect of allopregnanolone on CRF gene expression in the hypothalamus (Patchev et al., 1994). The regulation of the stress system appears to be influenced by a sex-dependent mechanism; it has been showed that allopregnanolone administration decreased CRF synthesis and release in hippocampus and amygdala of male but not female rats. This evidence suggests a more complex mechanism in need of further investigation (Boero et al., 2021).

Thus, allopregnanolone has an important role in restoring the physiological response of the organism after stress exposure (Gunn et al., 2015) through regulating the HPA axis, both directly by impacting genomic mechanisms and indirectly by modulating GABAergic neurotransmission. Interestingly, Deo and colleagues (Deo et al., 2010) showed that allopregnanolone could also exert its anxiolytic-like effect by increasing the expression of NPY Y1 receptors in CeA in mice. The intra-CeA administration of either allopregnanolone, NPY or NPY Y1/5-receptor agonists resulted in anxiolytic effects in a dose-dependent manner, while an intra-CeA injection of NPY Y1 antagonists resulted in a significant increase in anxiety-like behavior. The anxiolytic effect of allopregnanolone was even more significant when administrated with effective doses of NPY, thus empowering the hypothesis that there is an interaction between neuropeptides and the neurosteroid system in selective nuclei of the amygdala.

As described above, impairment in HPA axis regulation is one of the underlying causes for the development of psychiatric disorders, including depression, PTSD and AUD. Mounting evidence indicate that allopregnanolone might serve as a biomarker for these diseases (for review, Aspesi and Pinna, 2018; Tomaselli and Vallée, 2019; Almeida et al., 2021). Individuals suffering from depression present with reductions in peripheral allopregnanolone levels in blood (Romeo et al., 1998; Ströhle et al., 1999) as well as in cerebrospinal fluid (CSF, Uzunova et al., 1998). Similarly, both men and women with PTSD have decreased allopregnanolone levels in the CSF (Rasmusson et al., 2006, 2019). These changes are associated with reduced expression and activity of GABAARs (Croarkin et al., 2011; Sanacora & Saricicek, 2007). Recent studies have shown that the intravenous infusion of brexanolone, a formulation of the endogenous allopregnanolone, reduces emotional disturbances in women with postpartum depression for more than a month (Dacarett-Galeano & Diao, 2019; Kanes et al., 2017; Meltzer-Brody et al., 2018). These findings have led to the FDA approval of this drug for the treatment of post-partum depression (Powell et al., 2020) and strengthen the hypothesis that targeting the GABAergic transmission is a potential therapeutic strategy for the treatment of stress-related psychiatric disorders.

5. GABAergic transmission impairments in AUD: current pharmacological approaches

The GABAergic system is a well-known target for alcohol (Liang & Olsen, 2014). Specifically, GABAARs have been identified as a central target for the action of ethanol (Becker et al., 1998; Boehm et al., 2004; Koob, 2004; Olsen & Spigelman, 2012), and single nucleotide polymorphisms of the α2 and γ1 subunit have been associated with alcohol dependence (Edenberg et al., 2004; Ittiwut et al., 2012; Soyka et al., 2008). Both low doses and intoxicating high doses of alcohol bind to synaptic and extrasynaptic GABAARs (Davies, 2003; Olsen, 2018), which mediate its pharmacological effects (Figure 2). To better understand the development of alcohol tolerance and dependence, an understanding of the distinctive roles of the synaptic and the extrasynaptic GABAARs in mediating the modulatory effects of alcohol on neuronal signaling appears to be crucial (Figure 2). By binding to these receptors, acute alcohol induces anxiolysis, sedation as well as impairments in motor coordination. Further, chronic alcohol consumption results in lethargy, confusion and loss of sensation which may eventually lead to death (Valenzuela, 1997).

A plethora of preclinical studies have investigated the effects of ethanol in the brain. Acute ethanol potentiates GABAergic neurotransmission (Harris & Allan, 1989; Suzdak et al., 1986), and in the basolateral amygdala, this has been related with the anxiolytic effects of this drug (Silberman et al., 2012). Interestingly, these changes have been associated with epigenetic mechanisms regulating the expression of neuropeptide Y in the amygdala (Berkel et al., 2019; Sakharkar et al., 2012). Some studies have demonstrated that a single alcohol injection could change the expression of GABAAR subunits. Specifically, in the hippocampus, acute ethanol reduces the surface expression of extrasynaptic receptors containing α4βδ within minutes. However, while this effect is also visible for synaptic receptors (α1βγ), it requires a longer time (Gonzalez et al., 2012; Liang et al., 2007). One or two days following ethanol exposure, an upregulation of synaptic receptors expression is observed (Liang et al., 2007). These changes highlight that the regulation of GABAAR subunits is complex and dynamic, thus representing an interesting target for understanding the development of ethanol dependence.

Several animal models are used to investigate the long-term consequences of chronic alcohol exposure and to investigate the association of GABAergic plasticity with the development of ethanol dependance. Rats exposed to chronic intermittent ethanol consumption have shown significant changes in the expression of synaptic GABAaR subunits, with a reduction of α1 subunit (Cagetti et al., 2003; Kumar et al., 2003) and an increase in the synaptic expression of α4 (Liang et al., 2006; Papadeas et al., 2001). Of note, these subunits have been identified as being involved in the sedation, anticonvulsant activity (Rudolph & Knoflach, 2011), and in changes of mood and anxiety (Liang & Olsen, 2014). Our recent work highlighted a reduced α2 (synaptic) and δ (extrasynaptic) mRNA expression in the cerebellum and prefrontal cortex of subjects suffering from AUD, suggesting a loss of tonic inhibition after chronic and excessive alcohol consumption. These changes were associated with altered DNA methylation at the promoter levels of these genes (Gatta et al., 2017, 2020). Mounting evidence has demonstrated the loss of GABAergic inhibition in postmortem brain of individuals with AUD, with alteration in both synaptic and extrasynaptic GABAAR subunits expression (Behar et al., 1999; Jin et al., 2012; Lingford-Hughes et al., 2005; Mitsuyama et al., 1998; Volkow et al., 1993). GABAARs downregulation after abstinence from chronic alcohol use contributes to many of the symptoms of alcohol withdrawal syndrome ([AWS], Liang and Olsen, 2014), which include disordered sleep, delirium tremens, and status epilepticus. Indeed, the loss of GABAergic inhibition is thought to contribute to sleep loss and increased wakefulness later in night (Koob & Colrain, 2020). During alcohol withdrawal, the loss of the modulatory effect of alcohol on GABAARs is likely to lead to an additional dysregulation of GABAergic neurotransmission and the simultaneous hyperactivation of glutamatergic transmission and HPA axis (Adinoff et al., 2005). Similarly, the development of status epilepticus is characterized by alterations in GABAAR subunit composition (e.g., internalization and desensitization of synaptic subunits, α and β), leading to a resistance to benzodiazepine (BDZ) treatment.

The use of modulators of the GABA receptors (Figure 1) to reverse the effects of alcohol dependence and withdrawal has been investigated (Burnette et al., 2022; Farokhnia et al., 2018, 2019; Liang & Olsen, 2014). GABAARs have a number of binding allosteric sites that allow their modulation by endogenous or non-endogenous ligands (Liang & Olsen, 2014). BDZ, one of the most well-known molecules for the modulation of GABAergic transmission, binds between the α and the γ subunit of GABAAR (Griffin et al., 2013) and thus enhances phasic inhibition but not tonic inhibition (Stell et al., 2003). In the clinic, BDZs are traditionally used for decreasing the risk of seizures, the emergence of panic attacks, and are indicated for generalized anxiety disorder (GAD), insomnia, and catatonia. They are also known to reduce AWS symptoms. However, because BDZ possess abuse potential, addictive sedative effects on the CNS when combined with alcohol, and dependance and cross-tolerance with alcohol (Devaud et al., 1996), they are unlikely to be used as a chronic treatment for AUD and withdrawal, as guidelines recommend their use for no longer than a few weeks (Liang & Olsen, 2014).

Topiramate, a fructopyranose derivative, potentiates GABAARs and inhibits AMPA/kainate glutamatergic transmission. It is currently FDA approved for the treatment of epilepsy and migraine. Topiramate is also used off-label for psychiatric disorders such as bipolar disorder, bulimia nervosa, PTSD, binge-eating disorder, and obesity (Marcotte, 1998; Nickel et al., 2005; Vieta et al., 2002). Recent evidence indicates that topiramate could serve as a putative treatment for substance related disorders, including alcohol dependance and withdrawal, nicotine, cocaine, benzodiazepine dependance and ecstasy abuse (Shinn & Greenfield, 2010). Clinical trials comparing the efficacy of Topiramate to FDA approved drugs for the treatment of AUD (e.g., disulfiram and naltrexone) show that Topiramate decreases alcohol consumption and intake (Baltieri et al., 2008; De Sousa et al., 2008). Although promising in terms of efficacy, Topiramate administration was associated with serious adverse events (including paresthesia, memory or concentration impairment) as well as a greater likelihood of relapse when compared to disulfiram (De Sousa et al., 2008; Shinn & Greenfield, 2010).

Gabapentin is a GABA analog with anticonvulsant properties. While its effect on alcohol intake has raised some controversies in preclinical models (Ozburn et al., 2020; Roberto et al., 2008), clinical trials showed a decrease in the percentage of heavy-drinking and craving, with an increase in abstinent days and a longer time-to-relapse in patients with AUD when compared to controls (Furieri & Nakamura-Palacios, 2007; Mason et al., 2014). Contraindicating its use, however, the administration of gabapentin is associated with dizziness, ataxia, somnolence, and gait disorders. Additionally, there is great potential for misuse and abuse of this drug, especially in patients with substance use disorder (Smith et al., 2016).

The GABAergic transmission can also be modulated by targeting metabotropic GABABRs. Sodium oxybate, the sodium salt of γ-hydroxybutyrate (GHB), is approved in Italy and Austria for the maintenance of abstinence in subjects suffering from AUD (Addolorato et al., 2020; Guiraud et al., 2021). Sodium oxybate exerts an alcohol mimetic action on the GABAergic system, binding with low affinity GABABRs and with high affinity GHB-specific receptors. Clinical trials showed the superiority of sodium oxybate in maintaining a sustained abstinence when compared to naltrexone (Caputo et al., 2003). Despite these encouraging results, sodium oxybate shows adverse effects which could limit its therapeutic indications, including dizziness and vertigo, the potential of abuse and misuse (especially in patients with psychiatric comorbidities as well as those suffering from cocaine or heroin dependance), and the risk of CNS depression and dependence/withdrawal (van den Brink et al., 2018).

One of the selective agonists for the metabotropic GABABRs is Baclofen, a derivative of GABA. Baclofen is FDA approved for the treatment of muscle spasticity. In France, it is also approved for the treatment of AUD, while it is used off-label in other European countries and in Australia (Addolorato & Leggio, 2010; Naudet & Braillon, 2018). The therapeutic effect of Baclofen in reducing alcohol intake has been demonstrated in some human studies, depending on doses and timing of administration (Farokhnia et al., 2018; Garbutt et al., 2021), while other studies have not observed an improvement in abstinence rates when compared to placebo-treated individuals (Beraha et al., 2016; Reynaud et al., 2017). To date, Baclofen is the only compound capable of safely reducing alcohol intake in patients with liver alcohol-associate diseases (Morley et al., 2018), but many adverse side effects have been observed, particularly in women, including sedation, headache, vertigo, and confusion. Furthermore, a certain level of tolerance to baclofen can develop if chronically administrated at high doses (Santos & Olmedo, 2017).

Although promising, the existing pharmacotherapy for AUD needs to be adapted to account for individual variability, considerable adverse effects, and the risk of relapse. As described above, GABAergic neurotransmission can also be fine-tuned by neuroactive steroids, like allopregnanolone, that exert a regulatory role on several endocrine systems underpinning the development of neuropsychiatric disorders, including substance use disorder and alcohol use disorder (Morrow et al., 2006).

6. Allopregnanolone alterations in neuropsychiatric pathologies: focus on alcohol use disorder

Since the pioneering work of Purdy and colleagues in the 90’s, a number of studies have investigated the effect of adverse events on neurosteroid levels along with the anxiolytic and anti-depressant properties of neurosteroids (for review, Tomaselli and Vallée, 2019). Mounting evidence indicates that, like stress, alcohol alters the biosynthesis of neurosteroids (Peltier et al., 2019, 2021). These effects are complex, however, with acute alcohol increasing allopregnanolone levels in the brain of rodents (VanDoren et al., 2000) as well as plasma of male and female subjects (Torres & Ortega, 2003, 2004) and chronic alcohol reducing these levels (Cagetti et al., 2004; Maldonado-Devincci et al., 2014). In addition, a decrease in allopregnanolone concentration has been observed during alcohol withdrawal (Romeo et al., 1996). The infusion of a low dose of alcohol reduces plasmatic allopregnanolone levels in both women suffering from PMDD and healthy controls (Nyberg et al., 2007). Alcohol consumption also reduced allopregnanolone in both men and women, and low allopregnanolone levels were associated with alcohol craving in men (Pierucci-Lagha et al., 2006). However, this reduction was not observed for a moderate dose of alcohol in healthy subjects (Holdstock et al., 2005). Our recent work highlighted that chronic alcohol induced changes in neurosteroid biosynthesis and affected the cerebellar levels of allopregnanolone and pregnanolone. These changes are associated with epigenetic modifications, DNA methylation in particular, affecting neurosteroidogenic enzymes, i.e., TSPO and 3α-hydroxysteroid dehydrogenase (Gatta et al., 2020). In addition, we observed a reduced expression (both mRNA and protein) in GABAAR α2 and δ subunits in association with increased DNA methylation at the gene promoter region in the cerebellum (Gatta et al., 2017, 2020). Preclinical studies also demonstrated that ethanol-induced epigenetic modifications mediated changes in GABAAR expression (Bohnsack et al., 2017, 2018). Altogether, these data strengthen the hypothesis of an imbalance between excitatory and inhibitory signaling in AUD.

Adding further complexity, neurosteroid levels are influenced by the phase of the menstrual cycle, as progesterone (the precursor of all neurosteroids) fluctuates throughout the cycle and significantly rises after ovulation (i.e., the luteal phase). High progesterone and increased allopregnanolone levels are associated with the emergence of negative moods (Bäckström et al., 1983; Rubinow & Schmidt, 2018; Sundström Poromaa & Gingnell, 2014), which are particularly evident during the luteal phase in premenstrual dysphoric disorder (PMDD) (Bäckström et al., 2015). This observation suggests that the neuromodulatory effect of neurosteroids on GABAergic transmission depends on their concentration. Likely, in these conditions, the binding capacity of the neurosteroids to the GABAARs is saturated, and this may prevent further activation of the GABAARs. Alternatively, the paradoxical symptoms induced by neurosteroids are also dependent on an individual’s vulnerability and their environment (Bäckström et al., 2015), with the existence of an inverted U-shaped relationship between the occurrence of negative symptoms and the levels of allopregnanolone (high and low concentrations being less determinant for the onset of dysphoria) (Andréen et al., 2006). This counterintuitive relationship exists for all GABAergic modulators, including benzodiazepines, barbiturates as well as alcohol (Gourley et al., 2005; Miczek et al., 2003).

The promise of neurosteroids for the treatment of substance use disorder is strengthened by the evidence that stimulation of neurosteroidogenesis reduces drug intake. Brain and plasma levels of allopregnanolone and 5α3α-THDOC are decreased in patients and in preclinical models of cocaine and alcohol-addiction (Milivojevic et al., 2019). Schmoutz and colleagues (2014) showed that an intraperitoneal injection of metyrapone decreased cocaine self-administration in rats. Metyrapone is an 11β-hydroxylase inhibitor that blocks the synthesis of pro-stress neurosteroids (i.e., glucocorticoids) while shifting the production toward anti-stress products (e.g., allopregnanolone and 5α3α-THDOC) (Rupprecht et al., 1998). Of note, both bicuculline (an antagonist of GABAA receptor) and finasteride (that blocks 5α-R1) partially reverse the effect of metyrapone (Schmoutz et al., 2014), demonstrating the importance of allopregnanolone and 5α3α-THDOC in restoring GABAergic inhibition and in modulating cocaine-related behaviors. Interestingly, allopregnanolone’s effect on cocaine-primed reinstatement and cue-induced reinstatement has been found in women more often than in men, suggesting a sex specific effect of neurosteroids on reducing cocaine-induced effects (Milivojevic et al., 2016).

Recently, ganaxolone, a synthetic derivative of allopregnanolone, has been filed for New Drug Application at the FDA for the treatment of seizures associated with CDKL5 deficiency disorder (CDD), a rare, genetic epilepsy (FDA, 2022). Kaminski and colleagues (2003) demonstrated that ganaxolone inhibited the expression and development of cocaine-kindled seizure in male mice. In addition, ganaxolone reverses behavioral symptoms in rodent models of kindling and bicuculline-treated animals (Beekman et al., 1998) suggesting a promising therapeutic effect in AWS.

7. Conclusion

Mounting evidence highlights the central role of an imbalance of GABAergic neurotransmission in the development of AUD. Excessive and protracted alcohol consumption leads to changes in synaptic and extrasynaptic subunit composition of GABAARs, resulting in loss of GABAergic inhibition and disruption of GABAergic control on the stress system. This imbalance is likely to be connected to an altered production of neurosteroids (e.g. pregnanolone and allopregnanolone) both in the brain and the periphery, which is influenced by alcohol consumption through epigenetic mechanisms, among others (Fig.2). Indeed, active neurosteroids are endogenous GABAARs positive allosteric neuromodulators, and recent studies suggest that their administration can reduce alcohol intake and craving by restoring the physiological tone of GABAergic neurotransmission and its control on the stress system and the HPA-axis, specifically. Interestingly, an alteration in neurosteroidogenesis has also been reported in several neuropsychiatric conditions that often are comorbid with AUD. The FDA approval of Brexanolone, an endogenous formulation of allopregnanolone, for the treatment of women suffering from PPD is an encouraging result because, unlike BDZ, Brexanolone exerts a long-lasting effect without induction of tolerance or dependence liability. This evidence suggests that neurosteroids represent a neuroregulatory system that should be further investigated as a novel treatment for people suffering from AUD, anxiety, epilepsy and depression.

Acknowledgements

SCP is supported by the National Institute on Alcohol Abuse and Alcoholism (NIAAA) grants, P50AA022538, U01AA019971 and by the Department of Veterans Affairs (Merit Grant, I01 BX004517 and Senior Research Career Scientist Award, IK6BX006030). EG is supported by K99 grant from NIAAAA K99AA028817. Figures are generated using BioRender.com. Authors thank Ms. Gabriela M. Wandling for editing the manuscript.

Abbreviations:

AUD

Alcohol Use Disorder

ADH

alcohol dehydrogenase

ALDH2

Aldehyde Dehydrogenase

HPA

Hypothalamic-Pituitary-Adrenal axis

CRF

Corticotropin-Releasing Factor

ACTH

Adrenocorticotropic Hormone

PVN

Paraventricular Nucleus

MDD

Major Depression Disorder

PTSD

Post-Traumatic Stress Disorder

GR

Glucocorticoid Receptor

BNST

Bed Nucleus of the Stria Terminalis

GABAARs

Extrasynaptic GABAA Receptors

CNS

Central Nervous System

NPY

Neuropeptide Y

CeA

Central Nucleus of the Amygdala

CRFR1

CRF receptor 1

Y2R

NPY receptor type 2

Y1R

NPY receptor type1

BLA

Basolateral Amygdala

DOC

Deoxycorticosterone

α3α-THDOC

5α,3α-tetrahydrodeoxycorticosterone

TSPO

Translocator Protein

StAR

Steroidogenic Acute Regulatory Protein

DBI

Diazepam Binding Inhibitor

3β-HSD

3β-hydroxysteroid dehydrogenase

5α-R1

Type-1 5α-reductase

CSF

Cerebrospinal Fluid

AWS

Alcohol Withdrawal Syndrome

BDX

Benzodiazepine

GAD

Generalized Anxiety Disorder

GHB

γ-hydroxybutyrate

PMDD

Premenstrual Dysphoric Disorder

PPD

Post-Partum Depression

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest

Authors have nothing to disclose.

References

  1. Addolorato G, & Leggio L (2010). Safety and Efficacy of Baclofen in the Treatment of Alcohol-Dependent Patients. Current Pharmaceutical Design, 16(19), 2113–2117. [DOI] [PubMed] [Google Scholar]
  2. Addolorato G, Lesch O-M, Maremmani I, Walter H, Nava F, Raffaillac Q, & Caputo F (2020). Post-marketing and clinical safety experience with sodium oxybate for the treatment of alcohol withdrawal syndrome and maintenance of abstinence in alcohol-dependent subjects. Expert Opinion on Drug Safety, 19(2), 159–166. [DOI] [PubMed] [Google Scholar]
  3. Adinoff B (1990). Hypothalamic-pituitary-adrenal axis functioning and cerebrospinal fluid corticotropin releasing hormone and corticotropin levels in alcoholics after recent and long-term abstinence. Archives of General Psychiatry, 47(4), 325. [DOI] [PubMed] [Google Scholar]
  4. Adinoff B, Junghanns K, Kiefer F, & Krishnan-Sarin S (2005). Suppression of the HPA axis stress-response: Implications for relapse. Alcoholism: Clinical and Experimental Research, 29(7), 1351–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Adinoff B, Ruether K, Krebaum S, Iranmanesh A, & Williams MJ (2003). Increased salivary cortisol concentrations during chronic alcohol intoxication in a naturalistic clinical sample of men. Alcoholism: Clinical and Experimental Research, 27(9), 1420–1427. [DOI] [PubMed] [Google Scholar]
  6. Agís-Balboa RC, Pinna G, Zhubi A, Maloku E, Veldic M, Costa E, & Guidotti A (2006). Characterization of brain neurons that express enzymes mediating neurosteroid biosynthesis. Proceedings of the National Academy of Sciences USA, 103(39), 14602–14607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Almeida FB, Pinna G, & Barros HMT (2021). The Role of HPA Axis and Allopregnanolone on the Neurobiology of Major Depressive Disorders and PTSD International Journal of Molecular Sciences, 22(11), 5495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Andréen L, Sundström-Poromaa I, Bixo M, Nyberg S, & Bäckström T (2006). Allopregnanolone concentration and mood—A bimodal association in postmenopausal women treated with oral progesterone. Psychopharmacology, 187(2), 209–221. [DOI] [PubMed] [Google Scholar]
  9. Aspesi D, & Pinna G (2018). Could a blood test for PTSD and depression be on the horizon? Expert Review of Proteomics, 15(12), 983–1006. [DOI] [PubMed] [Google Scholar]
  10. Ayer L, Ramchand R, Karimi G, & Wong EC (2022). Co-occurring alcohol and mental health problems in the military: Prevalence, disparities, and service utilization. Psychology of Addictive Behaviors, 36(4):419–427. [DOI] [PubMed] [Google Scholar]
  11. Bäckström T, Bixo M, & Strömberg J (2015). GABAA receptor-modulating steroids in relation to women’s behavioral health. Current Psychiatry Reports, 17(11), 92. [DOI] [PubMed] [Google Scholar]
  12. Bäckström T, Sanders D, Leask R, Davidson D, Warner P, & Bancroft J (1983). Mood, sexuality, hormones, and the menstrual cycle: II. Hormone levels and their relationship to the premenstrual syndrome. Psychosomatic Medicine, 45(6), 503–507. [DOI] [PubMed] [Google Scholar]
  13. Baltieri DA, Daró FR, Ribeiro PL, & de Andrade AG (2008). Comparing topiramate with naltrexone in the treatment of alcohol dependence. Addiction (Abingdon, England), 103(12), 2035–2044. [DOI] [PubMed] [Google Scholar]
  14. Barbaccia ML, Roscetti G, Trabucchi M, Mostallino MC, Concas A, Purdy RH, & Biggio G (1996). Time-dependent changes in rat brain neuroactive steroid concentrations and GABAA receptor function after acute stress. Neuroendocrinology, 63(2), 166–172. [DOI] [PubMed] [Google Scholar]
  15. Barbaccia ML, Roscetti G, Trabucchi M, Purdy RH, Mostallino MC, Concas A, & Biggio G (1997). The effects of inhibitors of GABAergic transmission and stress on brain and plasma allopregnanolone concentrations. British Journal of Pharmacology, 120(8), 1582–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Baulieu EE, & Robel P (1998). Dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEAS) as neuroactive neurosteroids. Proceedings of the National Academy of Sciences USA, 95(8), 4089–4091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Baulieu EE (1981). Steroid hormones in the brain: several mechanisms? In Fuxe K, Gustafsson J-Å, & Wetterberg L (Eds.), Steroid Hormone Regulation of the Brain (pp. 3–14). Pergamon. [Google Scholar]
  18. Becker HC (2008). Alcohol dependence, withdrawal, and relapse. Alcohol Research & Health, 31(4), 348–361. [PMC free article] [PubMed] [Google Scholar]
  19. Becker HC (2017). Influence of stress associated with chronic alcohol exposure on drinking. Neuropharmacology, 122, 115–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Becker HC, Veatch LM, & Diaz-Granados JL (1998). Repeated ethanol withdrawal experience selectively alters sensitivity to different chemoconvulsant drugs in mice. Psychopharmacology, 139(1–2), 145–153. [DOI] [PubMed] [Google Scholar]
  21. Beekman M, Ungard JT, Gasior M, Carter RB, Dijkstra D, Goldberg SR, & Witkin JM (1998). Reversal of behavioral effects of pentylenetetrazol by the neuroactive steroid ganaxolone. Journal of Pharmacology and Experimental Therapeutics, 284(3), 868–877. [PubMed] [Google Scholar]
  22. Behar KL, Rothman DL, Petersen KF, Hooten M, Delaney R, Petroff OAC, Shulman GI, Navarro V, Petrakis IL, Charney DS, & Krystal JH (1999). Preliminary evidence of low cortical GABA levels in localized 1H-MR spectra of alcohol-dependent and hepatic encephalopathy patients. American Journal of Psychiatry, 156(6), 952–954. [DOI] [PubMed] [Google Scholar]
  23. Belelli D, Casula A, Ling A, & Lambert JJ (2002). The influence of subunit composition on the interaction of neurosteroids with GABA(A) receptors. Neuropharmacology, 43(4), 651–661. [DOI] [PubMed] [Google Scholar]
  24. Belelli D, & Lambert JJ (2005). Neurosteroids: Endogenous regulators of the GABA(A) receptor. Nature Reviews. Neuroscience, 6(7), 565–575. [DOI] [PubMed] [Google Scholar]
  25. Belelli D, Phillips GD, Atack JR, & Lambert JJ (2021). Relating neurosteroid modulation of inhibitory neurotransmission to behaviour. Journal of Neuroendocrinology, e13045. [DOI] [PubMed] [Google Scholar]
  26. Beraha EM, Salemink E, Goudriaan AE, Bakker A, de Jong D, Smits N, Zwart JW, Geest D van, Bodewits P, Schiphof T, Defourny H, van Tricht M, van den Brink W, & Wiers RW (2016). Efficacy and safety of high-dose baclofen for the treatment of alcohol dependence: A multicentre, randomised, double-blind controlled trial. European Neuropsychopharmacology, 26(12), 1950–1959. [DOI] [PubMed] [Google Scholar]
  27. Berkel TDM, Zhang H, Teppen T, Sakharkar AJ, & Pandey SC (2019). Essential role of histone methyltransferase G9a in rapid tolerance to the anxiolytic effects of ethanol. The International Journal of Neuropsychopharmacology, 22(4), 292–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Birrell L, Newton NC, Teesson M, Tonks Z, & Slade T (2015). Anxiety disorders and first alcohol use in the general population. Findings from a nationally representative sample. Journal of Anxiety Disorders, 31, 108–113. [DOI] [PubMed] [Google Scholar]
  29. Bitran D, Dugan M, Renda P, Ellis R, & Foley M (1999). Anxiolytic effects of the neuroactive steroid pregnanolone (3α-OH-5β-pregnan-20-one) after microinjection in the dorsal hippocampus and lateral septum. Brain Research, 850(1), 217–224. [DOI] [PubMed] [Google Scholar]
  30. Bitran D, Hilvers RJ, & Kellogg CK (1991). Anxiolytic effects of 3 alpha-hydroxy-5 alpha[beta]-pregnan-20-one: Endogenous metabolites of progesterone that are active at the GABAA receptor. Brain Research, 561(1), 157–161. [DOI] [PubMed] [Google Scholar]
  31. Blaine SK, & Sinha R (2017). Alcohol, stress, and glucocorticoids: From risk to dependence and relapse in alcohol use disorders. Neuropharmacology, 122, 136–147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Boehm SL, Ponomarev I, Jennings AW, Whiting PJ, Rosahl TW, Garrett EM, Blednov YA, & Harris RA (2004). gamma-Aminobutyric acid A receptor subunit mutant mice: New perspectives on alcohol actions. Biochemical Pharmacology, 68(8), 1581–1602. [DOI] [PubMed] [Google Scholar]
  33. Boero G, Tyler RE, Todd CA, O’Buckley TK, Balan I, Besheer J, & Morrow AL (2021). (3α,5α)3-hydroxypregnan-20-one (3α,5α-THP) regulation of hypothalamic and extrahypothalamic corticotropin releasing factor (CRF): Sexual dimorphism and brain region specificity in Sprague Dawley rats. Neuropharmacology, 186, 108463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Bohnsack JP, Hughes BA, O’Buckley TK, Edokpolor K, Besheer J, & Morrow AL (2018). Histone deacetylases mediate GABAA receptor expression, physiology, and behavioral maladaptations in rat models of alcohol dependence. Neuropsychopharmacology, 43(7), 1518–1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Bohnsack JP, & Pandey SC (2021). Histone modifications, DNA methylation, and the epigenetic code of alcohol use disorder. International Review of Neurobiology, 156, 1–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Bohnsack JP, Patel VK, & Morrow AL (2017). Ethanol exposure regulates Gabra1 expression via histone deacetylation at the promoter in cultured cortical neurons. The Journal of Pharmacology and Experimental Therapeutics, 363(1), 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Bolton JM, Robinson J, & Sareen J (2009). Self-medication of mood disorders with alcohol and drugs in the National Epidemiologic Survey on Alcohol and Related Conditions. Journal of Affective Disorders, 115(3), 367–375. [DOI] [PubMed] [Google Scholar]
  38. Brickley SG, & Mody I (2012). Extrasynaptic GABA(A) receptors: Their function in the CNS and implications for disease. Neuron, 73(1), 23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Brown AR, Mitchell SJ, Peden DR, Herd MB, Seifi M, Swinny JD, Belelli D, & Lambert JJ (2016). During postnatal development endogenous neurosteroids influence GABA-ergic neurotransmission of mouse cortical neurons. Neuropharmacology, 103, 163–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Burnette EM, Nieto SJ, Grodin EN, Meredith LR, Hurley B, Miotto K, Gillis AJ, & Ray LA (2022). Novel agents for the pharmacological treatment of alcohol use disorder. Drugs, 82(3), 251–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Cagetti E, Liang J, Spigelman I, & Olsen RW (2003). Withdrawal from chronic intermittent ethanol treatment changes subunit composition, reduces synaptic function, and decreases behavioral responses to positive allosteric modulators of GABAA receptors. Molecular Pharmacology, 63(1), 53–64. [DOI] [PubMed] [Google Scholar]
  42. Cagetti E, Pinna G, Guidotti A, Baicy K, & Olsen RW (2004). Chronic intermittent ethanol (CIE) administration in rats decreases levels of neurosteroids in hippocampus, accompanied by altered behavioral responses to neurosteroids and memory function. Neuropharmacology, 46(4), 570–579. [DOI] [PubMed] [Google Scholar]
  43. Callachan H, Cottrell GA, Hather NY, Lambert JJ, Nooney JM, Peters JA, & Callan HG (1987). Modulation of the GABAA receptor by progesterone metabolites. Proceedings of the Royal Society of London. Series B. Biological Sciences, 231(1264), 359–369. [DOI] [PubMed] [Google Scholar]
  44. Cappell H, & Herman CP (1972). Alcohol and Tension Reduction; A Review. Quarterly Journal of Studies on Alcohol, 33(1), 33–64. [PubMed] [Google Scholar]
  45. Caputo F, Addolorato G, Lorenzini F, Domenicali M, Greco G, del RE A, Gasbarrini G, Stefanini GF, & Bernardi M (2003). Gamma-hydroxybutyric acid versus naltrexone in maintaining alcohol abstinence: An open randomized comparative study. Drug and Alcohol Dependence, 70(1), 85–91. [DOI] [PubMed] [Google Scholar]
  46. Carboni E, Gee KW, Wieland S, & Lan NC (1996). Anxiolytic properties of endogenously occurring pregnanediols in two rodent models of anxiety. Psychopharmacology, 126(2), 173–178. [DOI] [PubMed] [Google Scholar]
  47. Carvalho AF, Heilig M, Perez A, Probst C, & Rehm J (2019). Alcohol use disorders. The Lancet, 394(10200), 781–792. [DOI] [PubMed] [Google Scholar]
  48. Chandra D, Jia F, Liang J, Peng Z, Suryanarayanan A, Werner DF, Spigelman I, Houser CR, Olsen RW, Harrison NL, & Homanics GE (2006). GABAA receptor alpha 4 subunits mediate extrasynaptic inhibition in thalamus and dentate gyrus and the action of gaboxadol. Proceedings of the National Academy of Sciences of the United States of America, 103(41), 15230–15235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Chebib M, & Johnston GA (1999). The “ABC” of GABA receptors: A brief review. Clinical and Experimental Pharmacology & Physiology, 26(11), 937–940. [DOI] [PubMed] [Google Scholar]
  50. Chen JA, Owens MD, Browne KC, & Williams EC (2018). Alcohol-related and mental health care for patients with unhealthy alcohol use and posttraumatic stress disorder in a National Veterans Affairs cohort. Journal of Substance Abuse Treatment, 85, 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Chen W-Y, Zhang H, Gatta E, Glover EJ, Pandey SC, & Lasek AW (2019). The Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid (SAHA) Alleviates Depression-Like Behavior and Normalizes Epigenetic Changes in the Hippocampus During Ethanol Withdrawal. Alcohol (Fayetteville, N.Y.). [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Cheney DL, Uzunov D, Costa E, & Guidotti A (1995). Gas chromatographic-mass fragmentographic quantitation of 3 alpha- hydroxy-5 alpha-pregnan-20-one (allopregnanolone) and its precursors in blood and brain of adrenalectomized and castrated rats. Journal of Neuroscience, 15(6), 4641–4650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Clark BJ, Wells J, King SR, & Stocco DM (1994). The purification, cloning, and expression of a novel luteinizing hormone-induced mitochondrial protein in MA-10 mouse Leydig tumor cells. Characterization of the steroidogenic acute regulatory protein (StAR). Journal of Biological Chemistry, 269(45), 28314–28322. [PubMed] [Google Scholar]
  54. Concas A, Mostallino MC, Porcu P, Follesa P, Barbaccia ML, Trabucchi M, Purdy RH, Grisenti P, & Biggio G (1998). Role of brain allopregnanolone in the plasticity of gamma-aminobutyric acid type A receptor in rat brain during pregnancy and after delivery. Proceedings of the National Academy of Sciences of the United States of America, 95(22), 13284–13289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Conway KP, Compton W, Stinson FS, & Grant BF (2006). Lifetime Comorbidity of DSM-IV Mood and Anxiety Disorders and Specific Drug Use Disorders: Results From the National Epidemiologic Survey on Alcohol and Related Conditions. The Journal of Clinical Psychiatry, 67(02), 247–258. [DOI] [PubMed] [Google Scholar]
  56. Costa E, Cheney DL, Grayson DR, Korneyev A, Longone P, Pani L, Romeo E, Zivkovich E, & Guidotti A (1994). Pharmacology of Neurosteroid Biosynthesis. Role of the Mitochondrial DBI Receptor (MDR) Complex. Annals of the New York Academy of Sciences, 746(1), 223–242. [DOI] [PubMed] [Google Scholar]
  57. Costa E, & Guidotti A (1991). Diazepam binding inhibitor (DBI): A peptide with multiple biological actions. Life Sciences, 49(5), 325–344. [DOI] [PubMed] [Google Scholar]
  58. Cotton NS (1979). The familial incidence of alcoholism: A review. Journal of Studies on Alcohol, 40(1), 89–116. [DOI] [PubMed] [Google Scholar]
  59. Cozzoli DK, Tanchuck-Nipper MA, Kaufman MN, Horowitz CB, & Finn DA (2014). Environmental stressors influence limited-access ethanol consumption by C57BL/6J mice in a sex-dependent manner. Alcohol (Fayetteville, N.Y.), 48(8), 741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Crawley JN, Glowa JR, Majewska MD, & Paul SM (1986). Anxiolytic activity of an endogenous adrenal steroid. Brain Research, 398(2), 382–385. [DOI] [PubMed] [Google Scholar]
  61. Crews FT, & Vetreno RP (2014). Neuroimmune basis of alcoholic brain damage. International Review of Neurobiology, 118, 315–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Croarkin PE, Levinson AJ, & Daskalakis ZJ (2011). Evidence for GABAergic inhibitory deficits in major depressive disorder. Neuroscience & Biobehavioral Reviews, 35(3), 818–825. [DOI] [PubMed] [Google Scholar]
  63. Cullinan WE, Ziegler DR, & Herman JP (2008). Functional role of local GABAergic influences on the HPA axis. Brain Structure & Function, 213(1–2), 63–72. [DOI] [PubMed] [Google Scholar]
  64. Cullinan WE (1998). Evidence for a PVN site of action for gamma aminobutyric acid in the regulatory control of the rat stress axis. Physiologist, 41, 379. [Google Scholar]
  65. Dacarett-Galeano DJ, & Diao XY (2019). Brexanolone: A Novel Therapeutic in the Treatment of Postpartum Depression. American Journal of Psychiatry Residents’ Journal, 15(2), 2–4 [Google Scholar]
  66. Daskalakis NP, Lehrner A, & Yehuda R (2013). Endocrine aspects of posttraumatic stress disorder and implications for diagnosis and treatment. Endocrinology and Metabolism Clinics of North America, 42(3), 503–513. [DOI] [PubMed] [Google Scholar]
  67. Davenport CM, Rajappa R, Katchan L, Taylor CR, Tsai M-C, Smith CM, de Jong JW, Arnold DB, Lammel S, & Kramer RH (2021). Relocation of an Extrasynaptic GABAA Receptor to Inhibitory Synapses Freezes Excitatory Synaptic Strength and Preserves Memory. Neuron, 109(1), 123–134.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Davies M (2003). The role of GABAA receptors in mediating the effects of alcohol in the central nervous system. Journal of Psychiatry & Neuroscience: JPN, 28(4), 263–274. [PMC free article] [PubMed] [Google Scholar]
  69. de Kloet ER, & Derijk R (2004). Signaling Pathways in Brain Involved in Predisposition and Pathogenesis of Stress-Related Disease: Genetic and Kinetic Factors Affecting the MR/GR Balance. Annals of the New York Academy of Sciences, 1032(1), 14–34. [DOI] [PubMed] [Google Scholar]
  70. de Kloet ER, Karst H, & Joëls M (2008). Corticosteroid hormones in the central stress response: Quick-and-slow. Frontiers in Neuroendocrinology, 29(2), 268–272. [DOI] [PubMed] [Google Scholar]
  71. De Sousa AA, De Sousa J, & Kapoor H (2008). An open randomized trial comparing disulfiram and topiramate in the treatment of alcohol dependence. Journal of Substance Abuse Treatment, 34(4), 460–463. [DOI] [PubMed] [Google Scholar]
  72. Deo GS, Dandekar MP, Upadhya MA, Kokare DM, & Subhedar NK (2010). Neuropeptide Y Y1 receptors in the central nucleus of amygdala mediate the anxiolytic-like effect of allopregnanolone in mice: Behavioral and immunocytochemical evidences. Brain Research, 1318, 77–86. [DOI] [PubMed] [Google Scholar]
  73. Devaud LL, Purdy RH, Finn DA, & Morrow AL (1996). Sensitization of gamma-aminobutyric acidA receptors to neuroactive steroids in rats during ethanol withdrawal. Journal of Pharmacology and Experimental Therapeutics, 278(2), 510–517. [PubMed] [Google Scholar]
  74. Devor EJ, & Cloninger CR (1989). Genetics of Alcoholism. Annual Review of Genetics, 23(1), 19–36. [DOI] [PubMed] [Google Scholar]
  75. Di Chiara G, & Imperato A (1988). Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proceedings of the National Academy of Sciences of the United States of America, 85(14), 5274–5278. Scopus. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Do Rego JL, Seong JY, Burel D, Leprince J, Luu-The V, Tsutsui K, Tonon M-C, Pelletier G, & Vaudry H (2009). Neurosteroid biosynthesis: Enzymatic pathways and neuroendocrine regulation by neurotransmitters and neuropeptides. Frontiers in Neuroendocrinology, 30(3), 259–301. [DOI] [PubMed] [Google Scholar]
  77. Edenberg HJ, Dick DM, Xuei X, Tian H, Almasy L, Bauer LO, Crowe RR, Goate A, Hesselbrock V, Jones K, Kwon J, Li T-K, Nurnberger JI, O’Connor SJ, Reich T, Rice J, Schuckit MA, Porjesz B, Foroud T, & Begleiter H (2004). Variations in GABRA2, encoding the alpha 2 subunit of the GABA(A) receptor, are associated with alcohol dependence and with brain oscillations. American Journal of Human Genetics, 74(4), 705–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Enoch M-A (2011). The role of early life stress as a predictor for alcohol and drug dependence. Psychopharmacology, 214(1), 17–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Espallergues J, Mamiya T, Vallée M, Koseki T, Nabeshima T, Temsamani J, Laruelle C, & Maurice T (2012). The antidepressant-like effects of the 3β-hydroxysteroid dehydrogenase inhibitor trilostane in mice is related to changes in neuroactive steroid and monoamine levels. Neuropharmacology, 62(1), 492–502. [DOI] [PubMed] [Google Scholar]
  80. Fahlke C, & Eriksson CJP (2000). Effect of adrenalectomy and exposure to corticosterone on alcohol intake in alcohol-preferring and alcohol-avoiding rat lines. Alcohol and Alcoholism, 35(2), 139–144. [DOI] [PubMed] [Google Scholar]
  81. Fahlke C, Hård E, & Hansen S (1996). Facilitation of ethanol consumption by intracerebroventricular infusions of corticosterone. Psychopharmacology, 127(1), 133–139. [DOI] [PubMed] [Google Scholar]
  82. Farokhnia M, Browning BD, & Leggio L (2019). Prospects for pharmacotherapies to treat alcohol use disorder: An update on recent human studies. Current Opinion in Psychiatry, 32(4), 255–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Farokhnia M, Deschaine SL, Sadighi A, Farinelli LA, Lee MR, Akhlaghi F, & Leggio L (2018). A deeper insight into how GABA-B receptor agonism via baclofen may affect alcohol seeking and consumption: Lessons learned from a human laboratory investigation. Molecular Psychiatry 26(2), 545–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Farrant M, & Nusser Z (2005). Variations on an inhibitory theme: Phasic and tonic activation of GABA(A) receptors. Nature Reviews. Neuroscience, 6(3), 215–229. [DOI] [PubMed] [Google Scholar]
  85. Fernández-Serrano MJ, Pérez-García M, Schmidt Río-Valle J, & Verdejo-García A (2010). Neuropsychological consequences of alcohol and drug abuse on different components of executive functions. Journal of Psychopharmacology, 24(9), 1317–1332. Scopus. [DOI] [PubMed] [Google Scholar]
  86. Follesa P, Floris G, Asuni GP, Ibba A, Tocco MG, Zicca L, Mercante B, Deriu F, & Gorini G (2015). Chronic intermittent ethanol regulates hippocampal GABA(A) receptor delta subunit gene expression. Frontiers in Cellular Neuroscience, 9, 445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Funk CK, Zorrilla EP, Lee M-J, Rice KC, & Koob GF (2007). Corticotropin-releasing factor 1 antagonists selectively reduce ethanol self-administration in ethanol-dependent rats. Biological Psychiatry, 61(1), 78–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Furieri FA, & Nakamura-Palacios EM (2007). Gabapentin reduces alcohol consumption and craving: A randomized, double-blind, placebo-controlled trial. The Journal of Clinical Psychiatry, 68(11), 1691–1700. [DOI] [PubMed] [Google Scholar]
  89. Garbutt JC, Kampov-Polevoy AB, Pedersen C, Stansbury M, Jordan R, Willing L, & Gallop RJ (2021). Efficacy and tolerability of baclofen in a U.S. community population with alcohol use disorder: A dose-response, randomized, controlled trial. Neuropsychopharmacology, 46(13), 2250–2256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Gatta E, Auta J, Gavin DP, Bhaumik DK, Grayson DR, Pandey SC, & Guidotti A (2017). Emerging Role of One-Carbon Metabolism and DNA Methylation Enrichment on δ-Containing GABAA Receptor Expression in the Cerebellum of Subjects with Alcohol Use Disorders (AUD). The International Journal of Neuropsychopharmacology, 20(12), 1013–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Gatta E, Grayson DR, Auta J, Saudagar V, Dong E, Chen Y, Krishnan HR, Drnevich J, Pandey SC, & Guidotti A (2019). Genome-wide methylation in alcohol use disorder subjects: Implications for an epigenetic regulation of the cortico-limbic glucocorticoid receptors (NR3C1). Molecular Psychiatry, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Gatta E, Guidotti A, Saudagar V, Grayson DR, Aspesi D, Pandey SC, & Pinna G (2020). Epigenetic regulation of GABAergic neurotransmission and neurosteroid biosynthesis in alcohol use disorder. International Journal of Neuropsychopharmacology, 24(2), 130–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Gatta E, Saudagar V, & Guidotti A (2021). Chapter 25—Alcohol use disorder and associated alterations in brain epigenetic marks. In Peedicayil J, Grayson DR, & Avramopoulos D (Eds.), Epigenetics in Psychiatry (Second Edition) (pp. 599–617). Academic Press. [Google Scholar]
  94. Giatti S, Romano S, Pesaresi M, Cermenati G, Mitro N, Caruso D, Tetel MJ, Garcia-Segura LM, & Melcangi RC (2015). Neuroactive steroids and the peripheral nervous system: An update. Steroids, 103, 23–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Gilpin NW (2012). Corticotropin-releasing factor (CRF) and neuropeptide Y (NPY): Effects on inhibitory transmission in central amygdala, and anxiety- & alcohol-related behaviors. Alcohol (Fayetteville, N.Y.), 46(4), 329–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Gilpin NW, Misra K, Flerman MA, Cruz MT, Koob GF, & Roberto M (2011). Neuropeptide Y opposes alcohol effects on gamma-aminobutyric acid release in amygdala and blocks the transition to alcohol dependence. Biological Psychiatry, 69(11), 1091–1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Gonzalez C, Moss SJ, & Olsen RW (2012). Ethanol promotes clathrin adaptor-mediated endocytosis via the intracellular domain of δ-containing GABAA receptors. Journal of Neuroscience, 32(49), 17874–17881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Gourley SL, DeBold JF, Yin W, Cook J, & Miczek KA (2005). Benzodiazepines and heightened aggressive behavior in rats: Reduction by GABAA/α1 receptor antagonists. Psychopharmacology, 178(2), 232–240. [DOI] [PubMed] [Google Scholar]
  99. Grant BF, Goldstein RB, Saha TD, Chou SP, Jung J, Zhang H, Pickering RP, Ruan WJ, Smith SM, Huang B, & Hasin DS (2015). Epidemiology of DSM-5 alcohol use disorder: Results from the National Epidemiologic Survey on Alcohol and Related Conditions III. JAMA Psychiatry, 72(8), 757–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Grant BF, Stinson FS, Dawson DA, Chou SP, Dufour MC, Compton W, Pickering RP, & Kaplan K (2004). Prevalence and co-occurrence of substance use disorders and independent mood and anxiety disorders: Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Archives of General Psychiatry, 61(8), 807–816. [DOI] [PubMed] [Google Scholar]
  101. Greeley J, & Oei T (1999). Alcohol and tension reduction. In Psychological theories of drinking and alcoholism, 2nd ed (pp. 14–53). The Guilford Press. [Google Scholar]
  102. Griffin CE, Kaye AM, Bueno FR, & Kaye AD (2013). Benzodiazepine pharmacology and central nervous system-mediated effects. The Ochsner Journal, 13(2), 214–223. [PMC free article] [PubMed] [Google Scholar]
  103. Guiraud J, Addolorato G, Aubin H-J, Batel P, de Bejczy A, Caputo F, Goudriaan AE, Gual A, Lesch O, Maremmani I, Perney P, Poulnais R, Raffaillac Q, Soderpalm B, Spanagel R, Walter H, van den Brink W, & SMO032 study group. (2021). Treating alcohol dependence with an abuse and misuse deterrent formulation of sodium oxybate: Results of a randomised, double-blind, placebo-controlled study. European Neuropsychopharmacology, 52, 18–30. [DOI] [PubMed] [Google Scholar]
  104. Gunn BG, Cunningham L, Mitchell SG, Swinny JD, Lambert JJ, & Belelli D (2015). GABAA receptor-acting neurosteroids: A role in the development and regulation of the stress response. Frontiers in Neuroendocrinology, 36, 28–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Harris RA, & Allan AM (1989). Alcohol intoxication: Ion channels and genetics. FASEB Journal, 3(6), 1689–1695. [DOI] [PubMed] [Google Scholar]
  106. Hasin DS, Stinson FS, Ogburn E, & Grant BF (2007). Prevalence, correlates, disability, and comorbidity of DSM-IV alcohol abuse and dependence in the United States: Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Archives of General Psychiatry, 64(7), 830–842. [DOI] [PubMed] [Google Scholar]
  107. Hatzigiakoumis DS, Martinotti G, Di Giannantonio M, & Janiri L (2011). Anhedonia and Substance Dependence: Clinical Correlates and Treatment Options. Frontiers in Psychiatry, 2. https://www.frontiersin.org/article/10.3389/fpsyt.2011.00010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Herd MB, Brown AR, Lambert JJ, & Belelli D (2013). Extrasynaptic GABAA receptors couple presynaptic activity to postsynaptic inhibition in the somatosensory thalamus. Journal of Neuroscience, 33(37), 14850–14868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Herman JP (2012). Neural pathways of stress integration. Alcohol Research: Current Reviews, 34(4), 441–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Holdstock L, Penland SN, Morrow AL, & de Wit H (2005). Moderate doses of ethanol fail to increase plasma levels of neurosteroid 3α-hydroxy-5α-pregnan-20-one-like immunoreactivity in healthy men and women. Psychopharmacology, 186(3), 442. [DOI] [PubMed] [Google Scholar]
  111. Hosie AM, Wilkins ME, da Silva HMA, & Smart TG (2006). Endogenous neurosteroids regulate GABAA receptors through two discrete transmembrane sites. Nature, 444(7118), 486–489. [DOI] [PubMed] [Google Scholar]
  112. Ittiwut C, Yang B-Z, Kranzler HR, Anton RF, Hirunsatit R, Weiss RD, Covault J, Farrer LA, & Gelernter J (2012). GABRG1 and GABRA2 variation associated with alcohol dependence in African Americans. Alcoholism: Clinical and Experimental Research, 36(4), 588–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Jin Z, Bazov I, Kononenko O, Korpi ER, Bakalkin G, & Birnir B (2012). Selective Changes of GABAA Channel Subunit mRNAs in the Hippocampus and Orbitofrontal Cortex but not in Prefrontal Cortex of Human Alcoholics. Frontiers in Cellular Neuroscience, 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Jones MT, Hillhouse EW, & Burden J (1976). Effect on various putative neurotransmitters on the secretion of corticotrophin-releasing hormone from the rat hypothalamus in vitro-a model of the neurotransmitters involved. The Journal of Endocrinology, 69(1), 1–10. [DOI] [PubMed] [Google Scholar]
  115. Kaminski RM, Gasior M, Carter RB, & Witkin JM (2003). Protective efficacy of neuroactive steroids against cocaine kindled-seizures in mice. European Journal of Pharmacology, 474(2–3), 217–222. [DOI] [PubMed] [Google Scholar]
  116. Kanes S, Colquhoun H, Gunduz-Bruce H, Raines S, Arnold R, Schacterle A, Doherty J, Epperson CN, Deligiannidis KM, Riesenberg R, Hoffmann E, Rubinow D, Jonas J, Paul S, & Meltzer-Brody S (2017). Brexanolone (SAGE-547 injection) in post-partum depression: A randomised controlled trial. The Lancet, 390(10093), 480–489. [DOI] [PubMed] [Google Scholar]
  117. Karavolas HJ, & Hodges DR (1990). Neuroendocrine metabolism of progesterone and related progestins. Ciba Foundation Symposium, 153, 22–44; discussion 44-55. [DOI] [PubMed] [Google Scholar]
  118. Kimoto T, Tsurugizawa T, Ohta Y, Makino J, Tamura H, Hojo Y, Takata N, & Kawato S (2001). Neurosteroid Synthesis by Cytochrome P450-Containing Systems Localized in the Rat Brain Hippocampal Neurons: N-Methyl-d-Aspartate and Calcium-Dependent Synthesis. Endocrinology, 142(8), 3578–3589. [DOI] [PubMed] [Google Scholar]
  119. King SR, Manna PR, Ishii T, Syapin PJ, Ginsberg SD, Wilson K, Walsh LP, Parker KL, Stocco DM, Smith RG, & Lamb DJ (2002). An Essential Component in Steroid Synthesis, the Steroidogenic Acute Regulatory Protein, Is Expressed in Discrete Regions of the Brain. Journal of Neuroscience, 22(24), 10613–10620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Knapp DJ, Overstreet DH, Moy SS, & Breese GR (2004). SB242084, flumazenil, and CRA1000 block ethanol withdrawal–induced anxiety in rats. Alcohol, 32(2), 101–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Koob GF (2003). Alcoholism: Allostasis and Beyond. Alcoholism: Clinical and Experimental Research, 27(2), 232–243. [DOI] [PubMed] [Google Scholar]
  122. Koob GF (2004). A role for GABA mechanisms in the motivational effects of alcohol. Biochemical Pharmacology, 68(8), 1515–1525. [DOI] [PubMed] [Google Scholar]
  123. Koob GF (2008). A role for brain stress systems in addiction. Neuron, 59(1), 11–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Koob GF (2013). Addiction is a Reward Deficit and Stress Surfeit Disorder. Frontiers in Psychiatry, 4. 10.3389/fpsyt.2013.00072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Koob GF (2015). The dark side of emotion: The addiction perspective. European Journal of Pharmacology, 753, 73–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Koob GF, & Colrain IM (2020). Alcohol use disorder and sleep disturbances: A feed-forward allostatic framework. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology, 45(1), 141–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Koob GF, & Le Moal M (1997). Drug abuse: Hedonic homeostatic dysregulation. Science (New York, N.Y.), 278(5335), 52–58. [DOI] [PubMed] [Google Scholar]
  128. Koob GF, & Volkow ND (2010). Neurocircuitry of addiction. Neuropsychopharmacology: Official Publication of the American College of Neuropsychopharmacology, 35(1), 217–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Koob GF, &Volkow ND (2016). Neurobiology of addiction: A neurocircuitry analysis. The Lancet. Psychiatry, 3(8), 760–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Kumar S, Kralic JE, O’Buckley TK, Grobin AC, & Morrow AL (2003). Chronic ethanol consumption enhances internalization of alpha1 subunit-containing GABAA receptors in cerebral cortex. Journal of Neurochemistry, 86(3), 700–708. [DOI] [PubMed] [Google Scholar]
  131. Kushner MG, Abrams K, & Borchardt C (2000). The relationship between anxiety disorders and alcohol use disorders: A review of major perspectives and findings. Clinical Psychology Review, 20(2), 149–171. [DOI] [PubMed] [Google Scholar]
  132. Kyzar EJ, Bohnsack JP, & Pandey SC (2022). Current and Future Perspectives of Noncoding RNAs in Brain Function and Neuropsychiatric Disease. Biological Psychiatry, 91(2), 183–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Lacapere J-J, Duma L, Finet S, Kassiou M, & Papadopoulos V (2020). Insight into structural features of TSPO: Implications for drug development. Trends in Pharmacological Sciences, 41(2), 110–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Lai HMX, Cleary M, Sitharthan T, & Hunt GE (2015). Prevalence of comorbid substance use, anxiety and mood disorders in epidemiological surveys, 1990-2014: A systematic review and meta-analysis. Drug and Alcohol Dependence, 154, 1–13. 10.1016/j.drugalcdep.2015.05.031 [DOI] [PubMed] [Google Scholar]
  135. Lambert JJ, Belelli D, Hill-Venning C, & Peters JA (1995). Neurosteroids and GABAA receptor function. Trends in Pharmacological Sciences, 16(9), 295–303. [DOI] [PubMed] [Google Scholar]
  136. Lambert JJ, Belelli D, Peden DR, Vardy AW, & Peters JA (2003). Neurosteroid modulation of GABAA receptors. Progress in Neurobiology, 71(1), 67–80. [DOI] [PubMed] [Google Scholar]
  137. Li X, & Slesinger PA (2022). GABAB Receptors and Drug Addiction: Psychostimulants and Other Drugs of Abuse. Current Topics in Behavioral Neurosciences, 52, 119–155. [DOI] [PubMed] [Google Scholar]
  138. Liang J, & Olsen RW (2014). Alcohol use disorders and current pharmacological therapies: The role of GABA(A) receptors. Acta Pharmacologica Sinica, 35(8), 981–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Liang J, Suryanarayanan A, Abriam A, Snyder B, Olsen RW, & Spigelman I (2007). Mechanisms of reversible GABAA receptor plasticity after ethanol intoxication. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 27(45), 12367–12377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Liang J, Zhang N, Cagetti E, Houser CR, Olsen RW, & Spigelman I (2006). Chronic intermittent ethanol-induced switch of ethanol actions from extrasynaptic to synaptic hippocampal GABAA receptors. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 26(6), 1749–1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Lingford-Hughes AR, Wilson SJ, Cunningham VJ, Feeney A, Stevenson B, Brooks DJ, & Nutt DJ (2005). GABA-benzodiazepine receptor function in alcohol dependence: A combined 11C-flumazenil PET and pharmacodynamic study. Psychopharmacology, 180(4), 595–606. [DOI] [PubMed] [Google Scholar]
  142. Lloyd-Evans E, & Waller-Evans H (2020). Biosynthesis and signalling functions of central and peripheral nervous system neurosteroids in health and disease. Essays in Biochemistry, 64(3), 591–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Locci A, & Pinna G (2017). Neurosteroid biosynthesis down-regulation and changes in GABAA receptor subunit composition: A biomarker axis in stress-induced cognitive and emotional impairment. British Journal of Pharmacology, 174(19), 3226–3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Lohoff FW, Clarke T-K, Kaminsky ZA, Walker RM, Bermingham ML, Jung J, Morris SW, Rosoff D, Campbell A, Barbu M, Charlet K, Adams M, Lee J, Howard DM, O’Connell EM, Whalley H, Porteous DJ, McIntosh AM, & Evans KL (2021). Epigenome-wide association study of alcohol consumption in N = 8161 individuals and relevance to alcohol use disorder pathophysiology: Identification of the cystine/glutamate transporter SLC7A11 as a top target. Molecular Psychiatry, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Lohoff FW, Roy A, Jung J, Longley M, Rosoff DB, Luo A, O’Connell E, Sorcher JL, Sun H, Schwandt M, Hodgkinson CA, Goldman D, Momenan R, McIntosh AM, Adams MJ, Walker RM, Evans KL, Porteous D, Smith AK, … Kaminsky ZA (2020). Epigenome-wide association study and multi-tissue replication of individuals with alcohol use disorder: Evidence for abnormal glucocorticoid signaling pathway gene regulation. Molecular Psychiatry, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Maguire J (2014). Stress-induced plasticity of GABAergic inhibition. Frontiers in Cellular Neuroscience, 8, 157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Makara GB, & Stark E (1974). Effects of gamma-aminobutyric acid (GABA) and GABA antagonist drugs on ACTH release. Neuroendocrinology, 16(3–4), 178–190. [DOI] [PubMed] [Google Scholar]
  148. Maldonado-Devincci AM, Cook JB, O’Buckley TK, Morrow DH, McKinley RE, Lopez MF, Becker HC, & Morrow AL (2014). Chronic intermittent ethanol exposure and withdrawal alters (3α,5α)-3-hydroxy-pregnan-20-one immunostaining in cortical and limbic brain regions of C57BL/6J mice. Alcoholism, Clinical and Experimental Research, 38(10), 2561–2571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Marcotte D (1998). Use of topiramate, a new anti-epileptic as a mood stabilizer. Journal of Affective Disorders, 50(2–3), 245–251. [DOI] [PubMed] [Google Scholar]
  150. Martinotti G, Di Nicola M, Reina D, Andreoli S, Focà F, Cunniff A, Tonioni F, Bria P, & Janiri L (2008). Alcohol Protracted Withdrawal Syndrome: The Role of Anhedonia. Substance Use & Misuse, 43(3–4), 271–284. [DOI] [PubMed] [Google Scholar]
  151. Mason BJ, Quello S, Goodell V, Shadan F, Kyle M, & Begovic A (2014). Gabapentin treatment for alcohol dependence: A randomized clinical trial. JAMA Internal Medicine, 174(1), 70–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. McEwen BS (1998). Stress, adaptation, and disease. Allostasis and allostatic load. Annals of the New York Academy of Sciences, 840, 33–44. [DOI] [PubMed] [Google Scholar]
  153. McEwen BS, & Stellar E (1993). Stress and the individual. Mechanisms leading to disease. Archives of Internal Medicine, 153(18), 2093–2101. [PubMed] [Google Scholar]
  154. McHugh RK, & Weiss RD (2019). Alcohol Use Disorder and Depressive Disorders. Alcohol Research: Current Reviews, 40(1), arcr.v40.1.01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Melcangi RC, Celotti F, Castano P, & Martini L (1993). Differential localization of the 5 alpha-reductase and the 3 alpha-hydroxysteroid dehydrogenase in neuronal and glial cultures. Endocrinology, 132(3), 1252–1259. [DOI] [PubMed] [Google Scholar]
  156. Mellon SH, & Deschepper CF (1993). Neurosteroid biosynthesis: Genes for adrenal steroidogenic enzymes are expressed in the brain. Brain Research, 629(2), 283–292. [DOI] [PubMed] [Google Scholar]
  157. Mellon SH, & Vaudry H (2001). Biosynthesis of neurosteroids and regulation of their synthesis. International Review of Neurobiology, 46, 33–78. [DOI] [PubMed] [Google Scholar]
  158. Meltzer-Brody S, Colquhoun H, Riesenberg R, Epperson CN, Deligiannidis KM, Rubinow DR, Li H, Sankoh AJ, Clemson C, Schacterle A, Jonas J, & Kanes S (2018). Brexanolone injection in post-partum depression: Two multicentre, double-blind, randomised, placebo-controlled, phase 3 trials. The Lancet, 392(10152), 1058–1070. [DOI] [PubMed] [Google Scholar]
  159. Mensah-Nyagan AG, Saredi S, Schaeffer V, Kibaly C, Meyer L, Melcangi RC, & Patte-Mensah C (2008). Assessment of neuroactive steroid formation in diabetic rat spinal cord using high-performance liquid chromatography and continuous flow scintillation detection. Neurochemistry International, 52(4), 554–559. [DOI] [PubMed] [Google Scholar]
  160. Miczek KA, Fish EW, & De Bold JF (2003). Neurosteroids, GABAA receptors, and escalated aggressive behavior. Hormones and Behavior, 44(3), 242–257. [DOI] [PubMed] [Google Scholar]
  161. Mihalek RM, Banerjee PK, Korpi ER, Quinlan JJ, Firestone LL, Mi ZP, Lagenaur C, Tretter V, Sieghart W, Anagnostaras SG, Sage JR, Fanselow MS, Guidotti A, Spigelman I, Li Z, DeLorey TM, Olsen RW, & Homanics GE (1999). Attenuated sensitivity to neuroactive steroids in gamma-aminobutyrate type A receptor delta subunit knockout mice. Proceedings of the National Academy of Sciences of the United States of America, 96(22), 12905–12910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Milivojevic V, Covault J, Angarita GA, Siedlarz K, & Sinha R (2019). Neuroactive steroid levels and cocaine use chronicity in men and women with cocaine use disorder receiving progesterone or placebo. The American Journal on Addictions, 28(1), 16–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Milivojevic V, Fox HC, Sofuoglu M, Covault J, & Sinha R (2016). Effects of progesterone stimulated allopregnanolone on craving and stress response in cocaine dependent men and women. Psychoneuroendocrinology, 65, 44–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Mitsuyama H, Little KY, Sieghart W, Devaud LL, & Morrow AL (1998). GABAA, Receptor α1, α4, and β3 Subunit mRNA and Protein Expression in the Frontal Cortex of Human Alcoholics. Alcoholism: Clinical and Experimental Research, 22(4), 815–822. [PubMed] [Google Scholar]
  165. Morley KC, Baillie A, Fraser I, Furneaux-Bate A, Dore G, Roberts M, Abdalla A, Phung N, & Haber PS (2018). Baclofen in the treatment of alcohol dependence with or without liver disease: Multisite, randomised, double-blind, placebo-controlled trial. The British Journal of Psychiatry: The Journal of Mental Science, 212(6), 362–369. [DOI] [PubMed] [Google Scholar]
  166. Morrow AL, Boero G, & Porcu P (2020). A Rationale for allopregnanolone treatment of alcohol use disorders: basic and clinical studies. Alcoholism: Clinical and Experimental Research, 44(2), 320–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Morrow AL, Porcu P, Boyd KN, & Grant KA (2006). Hypothalamic-pituitary-adrenal axis modulation of GABAergic neuroactive steroids influences ethanol sensitivity and drinking behavior. Dialogues in Clinical Neuroscience, 8(4), 463–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Midzak A, Zirkin B, Papadopoulos V (2015). Translocator protein: pharmacology and steroidogenesis. Biochem Soc Trans, 43(4), 572–578 [DOI] [PubMed] [Google Scholar]
  169. Moscato BS, Russell M, Zielezny M, Bromet E, Egri G, Mudar P, & Marshall JR (1997). Gender differences in the relation between depressive symptoms and alcohol problems: A longitudinal perspective. American Journal of Epidemiology, 146(11), 966–974. [DOI] [PubMed] [Google Scholar]
  170. Naudet F, & Braillon A (2018). Baclofen and alcohol in France. The Lancet. Psychiatry, 5(12), 961–962. [DOI] [PubMed] [Google Scholar]
  171. Nayeem N, Green TP, Martin IL, & Barnard EA (1994). Quaternary structure of the native GABAA receptor determined by electron microscopic image analysis. Journal of Neurochemistry, 62(2), 815–818. [DOI] [PubMed] [Google Scholar]
  172. Nickel C, Tritt K, Muehlbacher M, Pedrosa Gil F, Mitterlehner FO, Kaplan P, Lahmann C, Leiberich PK, Krawczyk J, Kettler C, Rother WK, Loew TH, & Nickel MK (2005). Topiramate treatment in bulimia nervosa patients: A randomized, double-blind, placebo-controlled trial. The International Journal of Eating Disorders, 38(4), 295–300. [DOI] [PubMed] [Google Scholar]
  173. Nutma E, Ceyzériat K, Amor S, Tsartsalis S, Millet P, Owen DR, Papadopoulos V, & Tournier BB (2021). Cellular sources of TSPO expression in healthy and diseased brain. European Journal of Nuclear Medicine and Molecular Imaging, 49(1), 146–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Nyberg S, Bäckström T, Zingmark E, Purdy RH, & Poromaa IS (2007). Allopregnanolone decrease with symptom improvement during placebo and gonadotropin-releasing hormone agonist treatment in women with severe premenstrual syndrome. Gynecological Endocrinology, 23(5), 257–266. [DOI] [PubMed] [Google Scholar]
  175. Olsen RW (2018). GABAA receptor: Positive and negative allosteric modulators. Neuropharmacology, 136(Pt A), 10–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Olsen RW, & Sieghart W (2008). International Union of Pharmacology. LXX. Subtypes of γ-Aminobutyric Acid A receptors: Classification on the basis of subunit composition, pharmacology, and function, update. Pharmacological Reviews, 60(3), 243–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Olsen RW, & Sieghart W (2009). GABAA receptors: Subtypes provide diversity of function and pharmacology. Neuropharmacology, 56(1), 141–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Olsen RW, & Spigelman I (2012). GABAA Receptor Plasticity in Alcohol Withdrawal. In Noebels JL, Avoli M, Rogawski MA, Olsen RW, & Delgado-Escueta AV (Eds.), Jasper’s Basic Mechanisms of the Epilepsies (4th ed.). National Center for Biotechnology Information; (US: ). [PubMed] [Google Scholar]
  179. O’Malley SS, & O’Connor PG (2011). Medications for unhealthy alcohol use: Across the spectrum. Alcohol Research & Health, 33(4), 300–312. [PMC free article] [PubMed] [Google Scholar]
  180. Overstreet DH, Knapp DJ, & Breese GR (2004). Modulation of multiple ethanol withdrawal-induced anxiety-like behavior by CRF and CRF1 receptors. Pharmacology Biochemistry and Behavior, 77(2), 405–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Owens MJ, Ritchie JC, & Nemeroff CB (1992). 5a-Pregnane-3a,21-diol-20-one (THDOC) attenuates mild stress-induced increases in plasma corticosterone via a non-glucocorticoid mechanism: Comparison with alprazolam. Brain Research, 573(2), 353–355. [DOI] [PubMed] [Google Scholar]
  182. Ozburn AR, Metten P, Potretzke S, Townsley KG, Blednov YA, & Crabbe JC (2020). Effects of Pharmacologically Targeting Neuroimmune Pathways on Alcohol Drinking in Mice Selectively Bred to Drink to Intoxication. Alcoholism, Clinical and Experimental Research, 44(2), 553–566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Pallarès M, Llidó A, Modòl L, Vallée M, & Darbra S (2015). Finasteride administration potentiates the disruption of prepulse inhibition induced by forced swim stress. Behavioural Brain Research, 289, 55–60. [DOI] [PubMed] [Google Scholar]
  184. Palmisano M, & Pandey SC (2017). Epigenetic mechanisms of alcoholism and stress-related disorders. Alcohol (Fayetteville, N.Y.), 60, 7–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Pandey SC, Kyzar EJ, & Zhang H (2017). Epigenetic basis of the dark side of alcohol addiction. Neuropharmacology, 122, 74–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Papadeas S, Grobin AC, & Morrow AL (2001). Chronic ethanol consumption differentially alters GABA(A) receptor alpha1 and alpha4 subunit peptide expression and GABA(A) receptor-mediated 36 Cl(−) uptake in mesocorticolimbic regions of rat brain. Alcoholism, Clinical and Experimental Research, 25(9), 1270–1275. [PubMed] [Google Scholar]
  187. Patchev VK, Hassan AHS, Holsboer F, & Almeida OFX(1996). The neurosteroid tetrahydroprogesterone attenuates the endocrine response to stress and exerts glucocorticoid-like effects on vasopressin gene transcription in the rat hypothalamus. Neuropsychopharmacology, 15(6), 533–540. [DOI] [PubMed] [Google Scholar]
  188. Patchev VK, Shoaib M, Holsboer F, & Almeida OFX (1994). The neurosteroid tetrahydroprogesterone counteracts corticotropin-releasing hormone-induced anxiety and alters the release and gene expression of corticotropin-releasing hormone in the rat hypothalamus. Neuroscience, 62(1), 265–271. [DOI] [PubMed] [Google Scholar]
  189. Patte-Mensah C, Kibaly C, Boudard D, Schaeffer V, Béglé A, Saredi S, Meyer L, & Mensah-Nyagan AG (2006). Neurogenic pain and steroid synthesis in the spinal cord. Journal of Molecular Neuroscience, 28(1), 17–31. [DOI] [PubMed] [Google Scholar]
  190. Paul SM, & Purdy RH. (1992). Neuroactive steroids. The FASEB Journal, 6(6), 2311–2322. [PubMed] [Google Scholar]
  191. Peltier MR, Verplaetse TL, Mineur YS, Gueorguieva R, Petrakis I, Cosgrove KP, Picciotto MR, & McKee SA (2021). Sex differences in progestogen- and androgen-derived neurosteroids in vulnerability to alcohol and stress-related disorders. Neuropharmacology, 187, 108499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Peltier MR, Verplaetse TL, Mineur YS, Petrakis IL, Cosgrove KP, Picciotto MR, & McKee SA (2019). Sex differences in stress-related alcohol use. Neurobiology of Stress, 10, 100149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Petrakis IL, & Simpson TL (2017). Posttraumatic Stress Disorder and Alcohol Use Disorder: A Critical Review of Pharmacologic Treatments. Alcoholism: Clinical and Experimental Research, 41(2), 226–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Pierucci-Lagha A, Covault J, Feinn R, Khisti RT, Morrow AL, Marx CE, Shampine LJ, & Kranzler HR (2006). Subjective effects and changes in steroid hormone concentrations in humans following acute consumption of alcohol. Psychopharmacology, 186(3), 451–461. [DOI] [PubMed] [Google Scholar]
  195. Piggott VM, Lloyd SC, Perrine SA, & Conti AC (2020). Chronic Intermittent Ethanol Exposure Increases Ethanol Consumption Following Traumatic Stress Exposure in Mice. Frontiers in Behavioral Neuroscience, 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Pollard MS, Tucker JS, & Green HD Jr. (2020). Changes in Adult Alcohol Use and Consequences During the COVID-19 Pandemic in the US. JAMA Network Open, 3(9), e2022942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Porcu P, Barron AM, Frye CA, Waif AA, Yang S-Y, He X-Y, Morrow AL, Panzica GC, & Melcangi RC (2016). Neurosteroidogenesis Today: Novel Targets for Neuroactive Steroid Synthesis and Action and Their Relevance for Translational Research. Journal of Neuroendocrinology, 28(2), 12351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Powell JG, Garland S, Preston K, & Piszczatoski C (2020). Brexanolone (Zulresso): Finally, an FDA-Approved Treatment for Postpartum Depression. Annals of Pharmacotherapy, 54(2), 157–163. [DOI] [PubMed] [Google Scholar]
  199. Purdy RH Morrow AL, Moore PH Jr, Paul SH (1991). Stress-induced levations of y-aminobutyric acid type A receptor-active steroids in the rat brain. Proc. Natl. Acad. Sci. USA, 88(10),4553–4557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Radley JJ, Arias CM, & Sawchenko PE (2006). Regional differentiation of the medial prefrontal cortex in regulating adaptive responses to acute emotional stress. Journal of Neuroscience, 26(50), 12967–12976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Rasmusson AM, King MW, Valovski I, Gregor K, Scioli-Salter E, Pineles SL, Hamouda M, Nillni YI, Anderson GM, & Pinna G (2019). Relationships between cerebrospinal fluid GABAergic neurosteroid levels and symptom severity in men with PTSD. Psychoneuroendocrinology, 102, 95–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Rasmusson AM, Pinna G, Paliwal P, Weisman D, Gottschalk C, Charney D, Krystal J, & Guidotti A (2006). Decreased cerebrospinal fluid allopregnanolone levels in women with posttraumatic stress disorder. Biological Psychiatry, 60(7), 704–713. [DOI] [PubMed] [Google Scholar]
  203. Rehm J, Anderson P, Barry J, Dimitrov P, Elekes Z, Feijão F, Frick U, Gual A, Gmel G, Kraus L, Marmet S, Raninen J, Rehm MX, Scafato E, Shield KD, Trapencieris M, & Gmel G (2015). Prevalence of and potential influencing factors for alcohol dependence in Europe. European Addiction Research, 21(1), 6–18. [DOI] [PubMed] [Google Scholar]
  204. Rehm J, & Shield KD (2019). Global burden of disease and the impact of mental and addictive disorders. Current Psychiatry Reports, 21(2), 10. [DOI] [PubMed] [Google Scholar]
  205. FDA approves drug for treatment of seizures associated with rare disease in patients two years of age and older. FDA (2022). https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-drug-treatment-seizures-associated-rare-disease-patients-two-years-age-and-older [Google Scholar]
  206. Reynaud M, Aubin H-J, Trinquet F, Zakine B, Dano C, Dematteis M, Trojak B, Paille F, & Detilleux M (2017). A randomized, placebo-controlled study of high-dose baclofen in alcohol-dependent patients-the ALPADIR study. Alcohol and Alcoholism (Oxford, Oxfordshire), 52(4), 439–446. [DOI] [PubMed] [Google Scholar]
  207. Richardson HN, Lee SY, O’Dell LE, Koob GF, & Rivier CL (2008). Alcohol self-administration acutely stimulates the hypothalamic-pituitary-adrenal axis, but alcohol dependence leads to a dampened neuroendocrine state. The European Journal of Neuroscience, 28(8), 1641–1653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Rivier C (2014). Role of hypothalamic corticotropin-releasing factor in mediating alcohol-induced activation of the rat hypothalamic–pituitary–adrenal axis. Frontiers in Neuroendocrinology, 35(2), 221–233. [DOI] [PubMed] [Google Scholar]
  209. 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. Journal of Neuroscience, 28(22), 5762–5771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Romeo E, Brancati A, De Lorenzo A, Fucci P, Furnari C, Pompili E, Sasso GF, Spalletta G, Troisi A, & Pasini A (1996). Marked decrease of plasma neuroactive steroids during alcohol withdrawal. Clinical Neuropharmacology, 19(4), 366–369. [DOI] [PubMed] [Google Scholar]
  211. Romeo E, Ströhle A, Spalletta G, di Michele F, Hermann B, Holsboer F, Pasini A, & Rupprecht R (1998). Effects of antidepressant treatment on neuroactive steroids in major depression. The American Journal of Psychiatry, 155(7), 910–913. [DOI] [PubMed] [Google Scholar]
  212. Rose AK, Shaw SG, Prendergast MA, & Little HJ (2010). The Importance of Glucocorticoids in AlcoholDependence and Neurotoxicity. Alcoholism: Clinical and Experimental Research, 34(12), 2011–2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Roy A, Mittal N, Zhang H, & Pandey SC (2002). Modulation of cellular expression of glucocorticoid receptor and glucocorticoid response element-DNA binding in rat brain during alcohol drinking and withdrawal. The Journal of Pharmacology and Experimental Therapeutics, 301(2), 774–784. [DOI] [PubMed] [Google Scholar]
  214. Rubinow DR, & Schmidt PJ (2018). Is there a role for reproductive steroids in the etiology and treatment of affective disorders? Dialogues in Clinical Neuroscience, 20(3), 187–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Rudolph U, & Knoflach F (2011). Beyond classical benzodiazepines: Novel therapeutic potential of GABAA receptor subtypes. Nature Reviews. Drug Discovery, 10(9), 685–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Rupprecht R, Rammes G, Eser D, Baghai TC, Schüle C, Nothdurfter C, Troxler T, Gentsch C, Kalkman HO, Chaperon F, Uzunov V, McAllister KH, Bertaina-Anglade V, La Rochelle CD, Tuerck D, Floesser A, Kiese B, Schumacher M, Landgraf R, … Kucher K (2009). Translocator protein (18 kD) as target for anxiolytics without benzodiazepine-like side effects. Science (New York, N.Y.), 325(5939), 490–93. [DOI] [PubMed] [Google Scholar]
  217. Rupprecht R, Ströhle A, Hermann B, di Michele F, Spalletta G, Pasini A, Holsboer F, & Romeo E (1998). Neuroactive steroid concentrations following metyrapone administration in depressed patients and healthy volunteers. Biological Psychiatry, 44(9), 912–914. [DOI] [PubMed] [Google Scholar]
  218. Russo SJ, & Nestler EJ (2013). The brain reward circuitry in mood disorders. Nature Reviews Neuroscience, 14(9), 609–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. SAMHSA, CBHSQ. (n.d.) Section 5 PE Tables – Results from the 2019 National Survey on Drug Use and Health: Detailed Tables. Retrieved February 1, 2022, from https://www.samhsa.gov/data/sites/default/files/reports/rpt29394/NSDUHDetailedTabs2019/NSDUHDetTabsSect5pe2019.htm
  220. Sakharkar AJ, Zhang H, Tang L, Shi G, & Pandey SC (2012). Histone deacetylases (HDAC)-induced histone modifications in the amygdala: A role in rapid tolerance to the anxiolytic effects of ethanol. Alcoholism: Clinical and Experimental Research, 36(1), 61–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. San José B, van de Mheen H, van Oers JA, Mackenbach JP, & Garretsen HF (2000). Adverse working conditions and alcohol use in men and women. Alcoholism, Clinical and Experimental Research, 24(8), 1207–1213. [DOI] [PubMed] [Google Scholar]
  222. Sanacora G, & Saricicek A (2007). GABAergic contributions to the pathophysiology of depression and the mechanism of antidepressant action. CNS & Neurological Disorders - Drug Targets, 6(2), 127–140. [DOI] [PubMed] [Google Scholar]
  223. Sanchez-Roige S, Palmer AA, Fontanillas P, Elson SL, 23andMe Research Team, the Substance Use Disorder Working Group of the Psychiatric Genomics Consortium, Adams MJ, Howard DM, Edenberg HJ, Davies G, Crist RC, Deary IJ, McIntosh AM, & Clarke T-K (2019). Genome-wide association study meta-analysis of the alcohol use disorders identification test (AUDIT) in two population-based cohorts. The American Journal of Psychiatry, 176 (2), 107–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Santos C, & Olmedo RE (2017). Sedative-hypnotic drug withdrawal syndrome: Recognition and treatment. Emergency Medicine Practice, 19(3), 1–20. [PubMed] [Google Scholar]
  225. Schmoutz CD, Guerin GF, & Goeders NE (2014). Role of GABA-active neurosteroids in the efficacy of metyrapone against cocaine addiction. Behavioural Brain Research, 271, 269–276. [DOI] [PubMed] [Google Scholar]
  226. Schuebel K, Gitik M, Domschke K, & Goldman D (2016). Making Sense of Epigenetics. The International Journal of Neuropsychopharmacology, 19(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Seal KH, Cohen G, Bertenthal D, Cohen BE, Maguen S, & Daley A (2011). Reducing barriers to mental health and social services for Iraq and Afghanistan veterans: Outcomes of an integrated primary care clinic. Journal of General Internal Medicine, 26(10), 1160–1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Sente A, Desai R, Naydenova K, Malinauskas T, Jounaidi Y, Miehling J, Zhou X, Masiulis S, Hardwick SW, Chirgadze DY, Miller KW, & Aricescu AR (2022). Differential assembly diversifies GABAA receptor structures and signalling. Nature, 604(7904), 190–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Serra M, Pisu MG, Dazzi L, Purdy RH, & Biggio G (2002). Prevention of the stress-induced increase in the concentration of neuroactive steroids in rat brain by long-term administration of mirtazapine but not of fluoxetine. Journal of Psychopharmacology, 16(2), 133–138. [DOI] [PubMed] [Google Scholar]
  230. Shen Q, Lal R, Luellen BA, Earnheart JC, Andrews AM, & Luscher B (2010). γ-Aminobutyric acid-type A receptor deficits cause hypothalamic-pituitary-adrenal axis hyperactivity and antidepressant drug sensitivity reminiscent of melancholic forms of depression. Biological Psychiatry, 68(6), 512–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Shinn AK, & Greenfield SF (2010). Topiramate in the treatment of substance-related disorders: A critical review of the literature. The Journal of Clinical Psychiatry, 71(5), 634–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Shu H-J, Eisenman LN, Jinadasa D, Covey DF, Zorumski CF, & Mennerick S (2004). Slow Actions of Neuroactive Steroids at GABAA Receptors. Journal of Neuroscience, 24(30), 6667–6675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Silberman Y, Ariwodola OJ, & Weiner JL (2012). B1-adrenoceptor activation is required for ethanol enhancement of lateral paracapsular GABAergic synapses in the rat basolateral amygdala. The Journal of Pharmacology and Experimental Therapeutics, 343(2), 451–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Silverman MN, & Sternberg EM (2012). Glucocorticoid regulation of inflammation and its functional correlates: From HPA axis to glucocorticoid receptor dysfunction. Annals of the New York Academy of Sciences, 1261(1), 55–63. h [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Simpson TL, Rise P, Browne KC, Lehavot K, & Kaysen D (2019). Clinical presentations, social functioning, and treatment receipt among individuals with comorbid life-time PTSD and alcohol use disorders versus drug use disorders: Findings from NESARC-III. Addiction, 114(6), 983–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Sinha R (2013). The clinical neurobiology of drug craving. Current Opinion in Neurobiology, 23(4), 649–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Smith RV, Havens JR, & Walsh SL (2016). Gabapentin misuse, abuse and diversion: A systematic review. Addiction (Abingdon, England), 111(7), 1160–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Soyka M, Preuss UW, Hesselbrock V, Zill P, Koller G, & Bondy B (2008). GABA-A2 receptor subunit gene (GABRA2) polymorphisms and risk for alcohol dependence. Journal of Psychiatric Research, 42(3), 184–191. [DOI] [PubMed] [Google Scholar]
  239. Spigelman I, Li Z, Liang J, Cagetti E, Samzadeh S, Mihalek RM, Homanics G , & Olsen RW (2003). Reduced Inhibition and Sensitivity to Neurosteroids in Hippocampus of Mice Lacking the GABAA Receptor δ Subunit. Journal of Neurophysiology, 90(2), 903–910. [DOI] [PubMed] [Google Scholar]
  240. Starkman BG, Sakharkar AJ, & Pandey SC (2012). Epigenetics-beyond the genome in alcoholism. Alcohol Research: Current Reviews, 34(3), 293–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Stell BM, Brickley SG, Tang CY, Farrant M, & Mody I (2003). Neuroactive steroids reduce neuronal excitability by selectively enhancing tonic inhibition mediated by δ subunit-containing GABAA receptors. Proceedings of the National Academy of Sciences, 100(24), 14439–14444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Stephens MAC, & Wand G (2012). Stress and the HPA axis: Role of glucocorticoids in alcohol dependence. Alcohol Research: Current Reviews, 34(4), 468–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Stetler C, & Miller GE (2011). Depression and hypothalamic-pituitary adrenal activation: A quantitative summary of four decades of research. Psychosomatic Medicine, 73(2), 114–126. [DOI] [PubMed] [Google Scholar]
  244. Ströhle A, Romeo E, Hermann B, Pasini A, Spalletta G, di Michele F, Holsboer F, & Rupprecht R (1999). Concentrations of 3α-reduced neuroactive steroids and their precursors in plasma of patients with major depression and after clinical recovery. Biological Psychiatry, 45(3), 274–277. [DOI] [PubMed] [Google Scholar]
  245. Sundström Poromaa I, & Gingnell M (2014). Menstrual cycle influence on cognitive function and emotion processing—From a reproductive perspective. Frontiers in Neuroscience, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Suzdak PD, Schwartz RD, Skolnick P, & Paul SM (1986). Ethanol stimulates gamma-aminobutyric acid receptor-mediated chloride transport in rat brain synaptoneurosomes. Proceedings of the National Academy of Sciences, 83(11), 4071–4075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Swendsen JD, Merikangas KR, Canino GJ, Kessler RC, Rubio-Stipec M, & Angst J (1998). The comorbidity of alcoholism with anxiety and depressive disorders in four geographic communities. Comprehensive Psychiatry, 39(4), 176–184. [DOI] [PubMed] [Google Scholar]
  248. Tasan RO, Nguyen NK, Weger S, Sartori SB, Singewald N, Heilbronn R, Herzog H, & Sperk G (2010). The central and basolateral amygdala are critical sites of neuropeptide Y/Y2 receptor-mediated regulation of anxiety and depression. Journal of Neuroscience, 30(18), 6282–6290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Tomaselli G, & Vallée M (2019). Stress and drug abuse-related disorders: The promising therapeutic value of neurosteroids focus on pregnenolone-progesterone-allopregnanolone pathway. Frontiers in Neuroendocrinology, 55, 100789. [DOI] [PubMed] [Google Scholar]
  250. Tonhajzerova I, & Mestanik M (2017). New Perspectives in the Model of Stress Response. Physiological Research, S173–S185. [DOI] [PubMed] [Google Scholar]
  251. Torres JM, & Ortega E (2003). Alcohol intoxication increases allopregnanolone levels in female adolescent humans. Neuropsychopharmacology, 28(6), 1207–1209. [DOI] [PubMed] [Google Scholar]
  252. Torres JM, & Ortega E (2004). Alcohol intoxication increases allopregnanolone levels in male adolescent humans. Psychopharmacology, 172(3), 352–355. [DOI] [PubMed] [Google Scholar]
  253. Uhart M, & Wand GS (2009). Stress, alcohol and drug interaction: An update of human research. Addiction Biology, 14(1), 43–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Ulrich-Lai YM, & Herman JP (2009). Neural regulation of endocrine and autonomic stress responses. Nature Reviews. Neuroscience, 10(6), 397–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Uzunova V, Sheline Y, Davis JM, Rasmusson A, Uzunov DP, Costa E, & Guidotti A (1998). Increase in the cerebrospinal fluid content of neurosteroids in patients with unipolar major depression who are receiving fluoxetine or fluvoxamine. Proceedings of the National Academy of Sciences, 95(6), 3239–3244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Valenzuela CF (1997). Alcohol and neurotransmitter interactions. Alcohol Health and Research World, 21(2), 144–148. [PMC free article] [PubMed] [Google Scholar]
  257. van den Brink W, Addolorato G, Aubin H-J, Benyamina A, Caputo F, Dematteis M, Gual A, Lesch O-M, Mann K, Maremmani I, Nutt D, Paille F, Perney P, Rehm J, Reynaud M, Simon N, Söderpalm B, Sommer WH, Walter H, & Spanagel R (2018). Efficacy and safety of sodium oxybate in alcohol-dependent patients with a very high drinking risk level. Addiction Biology, 23(4), 969–986. [DOI] [PubMed] [Google Scholar]
  258. VanDoren MJ, Matthews DB, Janis GC, Grobin AC, Devaud LL, & Morrow AL (2000). Neuroactive steroid 3alpha-hydroxy-5alpha-pregnan-20-one modulates electrophysiological and behavioral actions of ethanol. Journal of Neuroscience, 20(5), 1982–1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Vendruscolo LF, Barbier E, Schlosburg JE, Misra KK, Whitfield TW, Logrip ML, Rivier C, Repunte-Canonigo V, Zorrilla EP, Sanna PP, Heilig M, & Koob GF (2012). Corticosteroid-dependent plasticity mediates compulsive alcohol drinking in rats. Journal of Neuroscience, 32(22), 7563–7571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Vendruscolo LF, Estey D, Goodell V, Macshane LG, Logrip ML, Schlosburg JE, McGinn MA, Zamora-Martinez ER, Belanoff JK, Hunt HJ, Sanna PP, George O, Koob GF, Edwards S, & Mason BJ (2015). Glucocorticoid receptor antagonism decreases alcohol seeking in alcohol-dependent individuals. Journal of Clinical Investigation, 125 (8), 3193–3197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Verplaetse TL, Moore KE, Pittman BP, Roberts W, Oberleitner LM, Smith PH, Cosgrove KP, & McKee SA (2018). Intersection of stress and gender in association with transitions in past year DSM-5 substance use disorder diagnoses in the United States. Chronic Stress (Thousand Oaks, Calif.), 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Vieta E, Torrent C, Garcia-Ribas G, Gilabert A, Garcia-Parés G, Rodriguez A, Cadevall J, García-Castrillón J, Lusilla P, & Arrufat F (2002). Use of topiramate in treatment-resistant bipolar spectrum disorders. Journal of Clinical Psychopharmacology, 22(4), 431–435. [DOI] [PubMed] [Google Scholar]
  263. Volkow ND, Wang GJ, Hitzemann R, Fowler JS, Wolf AP, Pappas N, Biegon A, & Dewey SL (1993). Decreased cerebral response to inhibitory neurotransmission in alcoholics. The American Journal of Psychiatry, 150(2), 417–422. [DOI] [PubMed] [Google Scholar]
  264. Walker LC (2021). A balancing act: The role of pro- and anti-stress peptides within the central amygdala in anxiety and alcohol use disorders. Journal of Neurochemistry, 157(5), 1615–1643. [DOI] [PubMed] [Google Scholar]
  265. Wohlfarth KM, Bianchi MT, & Macdonald RL (2002). Enhanced neurosteroid potentiation of ternary GABA(A) receptors containing the delta subunit. The Journal of Neuroscience, 22(5), 1541–1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Wu K, Han W, Tian Q, Li Y, & Lu W (2021). Activity- and sleep-dependent regulation of tonic inhibition by Shisa7. Cell Reports, 34(12), 108899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Yazdi K, Fuchs-Leitner I, Rosenleitner J, & Gerstgrasser NW (2020). Impact of the COVID-19 pandemic on patients with alcohol use disorder and associated risk factors for relapse. Frontiers in Psychiatry, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Yoshizawa K, Okumura A, Nakashima K, Sato T, & Higashi T (2017). Role of allopregnanolone biosynthesis in acute stress-induced anxiety-like behaviors in mice. Synapse (New York, N.Y.), 71(8). [DOI] [PubMed] [Google Scholar]
  269. Young EA, Carlson NE, & Brown MB (2001). Twenty-four-hour ACTH and cortisol pulsatility in depressed women. Neuropsychopharmacology, 25(2), 267–276. [DOI] [PubMed] [Google Scholar]

RESOURCES