Skip to main content

REVIEW article

Front. Behav. Neurosci., 31 July 2013
Sec. Motivation and Reward
Volume 7 - 2013 | https://doi.org/10.3389/fnbeh.2013.00093

Involvement of the endogenous opioid system in the psychopharmacological actions of ethanol: the role of acetaldehyde

  • Area de Psicobiología, Universitat Jaume I, Castellón, Spain

Significant evidence implicates the endogenous opioid system (EOS) (opioid peptides and receptors) in the mechanisms underlying the psychopharmacological effects of ethanol. Ethanol modulates opioidergic signaling and function at different levels, including biosynthesis, release, and degradation of opioid peptides, as well as binding of endogenous ligands to opioid receptors. The role of β-endorphin and µ-opioid receptors (OR) have been suggested to be of particular importance in mediating some of the behavioral effects of ethanol, including psychomotor stimulation and sensitization, consumption and conditioned place preference (CPP). Ethanol increases the release of β-endorphin from the hypothalamic arcuate nucleus (NArc), which can modulate activity of other neurotransmitter systems such as mesolimbic dopamine (DA). The precise mechanism by which ethanol induces a release of β-endorphin, thereby inducing behavioral responses, remains to be elucidated. The present review summarizes accumulative data suggesting that the first metabolite of ethanol, the psychoactive compound acetaldehyde, could participate in such mechanism. Two lines of research involving acetaldehyde are reviewed: (1) implications of the formation of acetaldehyde in brain areas such as the NArc, with high expression of ethanol metabolizing enzymes and presence of cell bodies of endorphinic neurons and (2) the formation of condensation products between DA and acetaldehyde such as salsolinol, which exerts its actions via OR.

Ethanol and the Opioid System

Evidence indicates that ethanol modulates the activity of different components of the endogenous opioid system (EOS), with a large body of data supporting the implication of opioid ligands and receptors in the mediation of some of the psychopharmacological effects of ethanol.

The Endogenous Opioid System at a Glance

The opioid peptide precursors proopiomelanocortin (POMC), proenkephalin (PENK) or prodynorphin (PDYN) (Kieffer and Gavériaux-Ruff, 2002) are the source for the respective peptides β-endorphin, enkephalin, and dynorphin (Nylander and Roman, 2012). These endogenous ligands activate G-protein-coupled µ-, ∂-, and κ-opioid receptors (OR) (µ-OR, ∂-OR and κ-OR), which differ in their affinities and response profiles (Evans et al., 1992; Knapp et al., 1995; Kieffer and Evans, 2009). β-endorphin presents higher affinity for µ- than ∂-, and reduced affinity for κ-OR (Roth-Deri et al., 2008; Trigo et al., 2010). Enkephalin binding to ∂-OR is greater than that for µ-OR (Khachaturian et al., 1985; Raynor et al., 1994; Akil et al., 1998) and dynorphin shows specific affinity for κ-OR (Chavkin et al., 1982; Simon, 1991; Roth-Deri et al., 2008; Trigo et al., 2010). Ethanol can modulate opioidergic transmission at different levels, including synthesis, release, and degradation of opioid peptides, and binding of endogenous ligands to OR (for a review see, Méndez and Morales-Mulia, 2008). Since β-endorphin signaling has been specially implicated in the behavioral effects of ethanol, the present review will focus on the effects of ethanol on this component of the EOS. In this regard, although OR and ligands are widely distributed through the brain, there are important neuroanatomical determinants related to β-endorphin distribution that are worth highlighting. β-endorphin-synthesizing cell bodies are primarily located in the hypothalamic arcuate nucleus (NArc) (Chronwall, 1985). Important brain regions for drug-induced effects such as the nucleus accumbens (NAcb) are under tonic control of β-endorphinic innervations from the NArc (Chronwall, 1985; Khachaturian et al., 1985; Spanagel et al., 1992; Gianoulakis, 2001). These NArc β-endorphin projections exert this control through the direct activation of OR located at the NAcb and by an indirect pathway via OR in the ventral tegmental area (VTA), which in turn modulate NAcb activity via VTA-NAcb dopamine (DA) neurons (Mansour et al., 1988; Di Chiara and North, 1992; Spanagel et al., 1992).

Ethanol-Induced Modulation of β-Endorphinic Neurotransmission

Acute administration of ethanol induces the release of β-endorphin; an effect found in hypothalamic cell cultures and tissue preparations (Gianoulakis, 1990; Boyadjieva and Sarkar, 1994; de Waele et al., 1994; Reddy et al., 1995; De et al., 2002). Ethanol also produces in vivo increases in β-endorphin content at the level of the hypothalamus (Schulz et al., 1980; Patel and Pohorecky, 1989), NAcb (Anwer and Soliman, 1995; Olive et al., 2001; Marinelli et al., 2003a), midbrain including the VTA (Rasmussen et al., 1998; Jarjour et al., 2009) and the central amygdala (CeA) (Lam et al., 2008). Some studies, however, have found inconsistent results, probably related to procedural and methodological differences (Seizinger et al., 1983; Popp and Erickson, 1998; Rasmussen et al., 1998; Leriche and Méndez, 2010). Increased levels of enkephalin in the hypothalamus (Schulz et al., 1980; Seizinger et al., 1983; Milton et al., 1991) and NAcb (Marinelli et al., 2003b) have also been found after acute ethanol.

Long-term exposure to ethanol primarily induces a decrease in POMC expression (Boyadjieva and Sarkar, 1997; Rasmussen et al., 2002; Oswald and Wand, 2004) and in hypothalamic β-endorphin release and levels (Boyadjieva and Sarkar, 1994; Oswald and Wand, 2004). A limited number of studies reported an increase in biosynthesis of POMC and POMC mRNA expression (Seizinger et al., 1984; Gianoulakis et al., 1988) as well as an initial increase followed by a gradual return to normal levels (Wand, 1990). Also, some authors found an increase or no effect on β-endorphin release (Boyadjieva and Sarkar, 1994; Oswald and Wand, 2004). Discrepancies might be attributable to the method of ethanol administration, ethanol dose, time course of drug exposure, administration route and differences in the development of tolerance. Also, it has been observed that alcohol-induced changes depend on the brain region investigated as well as the species and strain of animals used (Gianoulakis, 2001; Méndez and Morales-Mulia, 2008).

Evidence of Behavioral Effects of Ethanol Mediated by the Endogenous Opioid System

Given that β-endorphin, and also enkephalin, activate μ-OR, extensive research has investigated the role of μ-OR in the behavioral effects of ethanol (Gianoulakis, 1993; Herz, 1997; Sanchis-Segura et al., 2000; Thorsell, 2013). Here we will focus on the involvement of these components of the EOS in several behavioral effects of ethanol, including psychomotor stimulation and sensitization, consumption, and associative learning (with a special focus on conditioned place preference (CPP)).

Psychomotor stimulation and sensitization

Increased psychomotor stimulation induced by ethanol in mice can be blocked with non-selective opioid receptor antagonists such as naloxone or naltrexone (Kiianmaa et al., 1983; Camarini et al., 2000; Sanchis-Segura et al., 2004; Pastor et al., 2005; Pastor and Aragon, 2006). Some pharmacological strategies have suggested the existence of three so-called subtypes of µ-OR; µ1, µ2, and, µ3 (Pasternak, 2001a,b; Cadet et al., 2003) and several studies have shown that μ- and specifically the µ1/2 - and µ3-OR subtypes, but not δ- or κ-OR, are involved in the motor stimulant effects of ethanol in adult mice (Pastor et al., 2005), and also in rats during early development (Arias et al., 2010; Pautassi et al., 2012). Other studies conducted in mice have suggested that this involvement of μ-OR in ethanol stimulation is debatable (Cunningham et al., 1998; Gevaerd et al., 1999; Holstein et al., 2005). Consistent with the EOS involvement, however, a lesion of the NArc produces a decrease in ethanol-induced stimulation in mice (Sanchis-Segura et al., 2000), and knockout mice deficient in β-endorphin showed attenuated ethanol-induced stimulation (Dempsey and Grisel, 2012). Also, in rats, naltrexone prevents activation produced by ethanol when locally administered in the NArc (Pastor and Aragon, 2008) and intra-VTA blockade of the μ-OR using either naltrexone or the irreversible and selective μ-OR antagonist β-funaltrexamine reduces ethanol-induced locomotor stimulation (Sánchez-Catalán et al., 2009). Additionally, chronic naltrexone, which upregulates μ-OR (Unterwald et al., 1998; Lesscher et al., 2003), enhances the stimulant effects of ethanol in mice (Sanchis-Segura et al., 2004).

A critical role of the EOS in the motor sensitizing effects of ethanol has also been proposed (Camarini et al., 2000; Miquel et al., 2003; Pastor and Aragon, 2006). Unspecific OR antagonism prevents development (Camarini et al., 2000) but not expression (Abrahao et al., 2008) of ethanol-induced locomotor sensitization. μ-OR are particularly involved in ethanol sensitization (Camarini et al., 2000), without a clear role of any of the µ-OR subtypes in mediating this process; µ1/2 -OR antagonism slowed down, but did not block development of sensitization (Pastor and Aragon, 2006). Facilitation of ethanol-induced sensitization found after a period of voluntary alcohol consumption in mice was also seen to be absent in μ-OR deficient CXBK mice (Tarragón et al., 2012). The involvement of μ-OR in ethanol sensitization might be related to ethanol-induced increases in β-endorphin release as a recent study demonstrated that β-endorphin-deficient mice do not show locomotor sensitization to ethanol (Dempsey and Grisel, 2012). Also, animals with selective lesions of the NArc show prevented sensitization to ethanol (Miquel et al., 2003; Pastor et al., 2011). Altogether these data suggest that opioids and specifically β-endorphins, via μ-OR, might be critical mediators of ethanol-induced neuroplasticity underlying psychomotor sensitization.

Ethanol consumption

Numerous studies conducted during the last few decades showed that systemic, as well as local administration of opioid antagonists decrease ethanol consumption under a variety of schedules in different animal species (for reviews see Herz, 1997; Gianoulakis, 2001; Oswald and Wand, 2004; Modesto-Lowe and Fritz, 2005). These conclusions have also been supported by the use of OR knockout mouse models (Roberts et al., 2000; Méndez and Morales-Mulia, 2008). This strong pre-clinical basis has lead to the use of opioid antagonists in alcoholism pharmacotherapy (O’Malley et al., 1992). In rodents, the use of non-selective, as well as selective μ-OR antagonists proved to be effective at reducing ethanol consumption (Méndez and Morales-Mulia, 2008). However, the effects of these manipulations have been seen to be, in some cases, non-specific; fat, saccharin, sucrose and water intake were also reduced by these manipulations (Krishnan-Sarin et al., 1995; Nielsen et al., 2008; Rao et al., 2008; Simms et al., 2008; Corwin and Wojnicki, 2009; Wong et al., 2009). These data are compatible with the interpretation that OR, and especially μ-OR might be a key mediator of the processing of positive reinforcement, both at emotional and motivational levels (Herz, 1997; Peciña and Berridge, 2005).

In general, data obtained with κ-OR or δ-OR manipulations are less conclusive. A recent review of the literature indicates that κ-OR stimulation generally antagonizes the reinforcing effects of alcohol whereas κ-OR blockade has no consistent effect (Wee and Koob, 2010). Dynorphin/κ-OR system appears to be involved in the negative reinforcing effects of ethanol by producing an aversive effect rather than by directly modulating the rewarding mechanism of ethanol (Wee and Koob, 2010; Walker et al., 2012). However, under an alcohol dependent-state, antagonism of κ-OR results effective in decreasing ethanol voluntary consumption (Wee and Koob, 2010; Walker et al., 2012). It has been reported that blockade of δ-OR either attenuates (Lê et al., 1993; Froehlich, 1995; Krishnan-Sarin et al., 1995; June et al., 1999; Hyytiä and Kiianmaa, 2001; Ciccocioppo et al., 2002), increases (Margolis et al., 2008) or has no effect on ethanol intake (Ingman et al., 2003). These discrepancies may be related to dynamic changes in δ-OR efficacy during ethanol exposure (Margolis et al., 2008). All these data support the participation of the POMC and PENK systems in maintaining alcohol consumption (Froehlich et al., 1991; Vengeliene et al., 2008).

Associative learning and conditioned place preference

It has been suggested that the EOS participates in the underlying mechanisms mediating conditioned effects induced by abused drugs, including ethanol. This implication is supported by two groups of experiments. On one hand, evidence indicates that OR antagonists attenuate cue-induced reinstatement of previously extinguished responding for ethanol self-administration (Lê et al., 1999; Ciccocioppo et al., 2002, 2003; Liu and Weiss, 2002; Burattini et al., 2006; Dayas et al., 2007; Marinelli et al., 2009), which suggests a role of EOS in cue-induced incentive motivational effects influencing ethanol-seeking behavior. This interpretation is consistent with clinical data showing that opioid antagonists increase abstinence duration periods in alcohol abusers (O’Malley et al., 1992), probably by reducing cue-induced seeking behavior. On the other hand, pretreatment with opioid receptor antagonism, while not influencing the acquisition of ethanol-induced CPP, reduces the expression and facilitates the extinction of this drug-free conditioned response (Bormann and Cunningham, 1997; Middaugh and Bandy, 2000; Kuzmin et al., 2003; Pastor et al., 2011). Mice lacking μ-OR also showed attenuated ethanol CPP (Hall et al., 2001). Further studies have suggested that expression of ethanol-induced CPP depends on OR located in the VTA, CeA, as well as anterior cingulated cortex (Bechtholt and Cunningham, 2005; Bie et al., 2009; Gremel et al., 2011). Additionally, a neurotoxic lesion of the β-endorphin neurons of the NArc, showed a facilitated extinction of ethanol-induced CPP (Pastor et al., 2011). β-endorphin and μ-OR appear to be therefore critically involved in the mechanisms underlying ethanol CPP. As Cunningham and collaborators have suggested, it is possible that altered opioid signaling might in turn alter conditioned motivation that normally maintains cue-induced seeking behavior during CPP testing (Cunningham et al., 1998). It is interesting to mention that pharmacological blockade of δ-OR with naltrindole in the CeA reduces expression of CPP induced by ethanol in rats (Bie et al., 2009). Activation of κ-OR has been shown to blunt acquisition of ethanol CPP (Logrip et al., 2009). Supporting these results, κ-OR knockout mice also showed enhanced ethanol CPP (Femenía and Manzanares, 2012).

Acetaldehyde: A Psychoactive Metabolite

The specific mechanism by which ethanol modulates the activity of the EOS remains to be understood. Evidence indicates that one possible mechanism might involve the role of acetaldehyde, the first metabolite of ethanol (Miquel et al., 2003; Sanchis-Segura et al., 2005b; Pastor and Aragon, 2008). Acetaldehyde is a psychoactive compound that produces behavioral and neurochemical effects suggested to mediate at least some of the effects of ethanol. Acetaldehyde is self-administered orally (Peana et al., 2010, 2012; Cacace et al., 2012) and directly into the brain (Brown et al., 1979; McBride et al., 2002; Rodd-Henricks et al., 2002; Peana et al., 2011). Its administration induces CPP (Smith et al., 1984; Quertemont and De Witte, 2001; Peana et al., 2009; Spina et al., 2010) as well as behavioral stimulation and sensitization when centrally administered (Arizzi et al., 2003; Correa et al., 2003a,b, 2009; Rodd et al., 2005; Arizzi-LaFrance et al., 2006; Sánchez-Catalán et al., 2009). The oxidation of ethanol to acetaldehyde in the brain is essentially mediated by the catalase-H2O2 system (Aragon et al., 1992a; Gill et al., 1992). Reduced brain catalase activity, which have been seen to decrease ethanol-derived central acetaldehyde formation in brain tissue preparations (Hamby-Mason et al., 1997) and in the brain of free-moving rats (Jamal et al., 2007), decreases ethanol consumption (Aragon and Amit, 1992; Koechling and Amit, 1994; Correa et al., 2004; Karahanian et al., 2011), ethanol-induced locomotor stimulation (Aragon et al., 1992b; Correa et al., 1999b, 2004; Sanchis-Segura et al., 1999a,b,c; Pastor et al., 2002; Pastor and Aragon, 2008), the anxiolityc effects of alcohol (Correa et al., 2008) and modulates ethanol-induced CPP (Font et al., 2008). Strategies aimed at increasing the production of brain acetaldehyde via an enhancement in activity of the enzymatic catalase system have also been used. These manipulations produced an increase in the motor stimulant properties of ethanol in mice (Correa et al., 1999a, 2000; Pastor et al., 2002). Other ethanol-induced effects such as taste aversion (Aragon et al., 1985) and social memory recognition have also been seen to be modulated by changes in brain catalase (Manrique et al., 2005).

Apart from brain catalase manipulation, the direct inactivation of acetaldehyde has also been shown to reduce ethanol effects, including drinking (Font et al., 2006a) and alcohol-induced relapse drinking (Orrico et al., 2013), CPP (Font et al., 2006b; Peana et al., 2008) and motor stimulation (Font et al., 2005; Martí-Prats et al., 2010; Pautassi et al., 2011).

Acetaldehyde-Induced Changes in the Opioidergic Neurotransmission

The NArc, the main site of β-endorphin synthesis in the brain, is one of areas with the highest levels of catalase expression (Moreno et al., 1995; Zimatkin and Lindros, 1996) and lower levels of the acetaldehyde-degrading enzyme aldehyde dehydrogenase (Zimatkin et al., 1992). Therefore, it has been thus suggested that catalase-dependent formation of acetaldehyde into the NArc might mediate ethanol-induced increases in the release of β-endorphin from the NArc in turn activating OR at the level of the VTA/NAcb to stimulate behavioral and neurophysiological actions (Sanchis-Segura et al., 2005a; Pastor and Aragon, 2008). Supporting this hypothesis, several authors (Reddy and Sarkar, 1993; Pastorcic et al., 1994; Reddy et al., 1995) have demonstrated that ethanol-induced increases in hypothalamic β-endorphin release are, indeed, mediated by acetaldehyde (Reddy and Sarkar, 1993; Pastorcic et al., 1994; Reddy et al., 1995). Hypothalamic cell cultures exposed to ethanol (12.5–100 µM) led to the formation of acetaldehyde (8–24 µM) and similar concentrations of acetaldehyde (12.5–50 µM) were able to stimulate β-endorphin release when tested in the absence of ethanol (Reddy and Sarkar, 1993; Pastorcic et al., 1994). Moreover, pre-treatment of hypothalamic cell cultures with catalase inhibitors caused dose-dependent decreases in ethanol-stimulated β-endorphin secretion (Reddy et al., 1995).

Another line of research linking the EOS and acetaldehyde is the investigation of the actions of salsolinol (for a review see Hipólito et al., 2012), the condensation product of DA and acetaldehyde. Salsolinol has been shown to alter enkephalin-receptor site binding (Lucchi et al., 1982) and other OR an effect that is blocked by naloxone (Fertel et al., 1980). Interestingly, intra-NAcb administration of salsolinol increases DA levels when microinjected in the core and decreases DA levels if the administration is in the NAcb shell (Hipólito et al., 2009) in a similar way to μ- and δ-OR agonists (Hipólito et al., 2008). It has been demonstrated that μ1-OR receptors exert a tonic modulatory control over activity of the DA system (Di Chiara and North, 1992; Devine et al., 1993). Thus, one possible mechanism by which salsolinol exerts its effects on the OR could be disinhibiting DA neurons in the VTA. Upholding this hypothesis, intra-posterior VTA administration of salsolinol induces a μ-OR dependent increase in DA levels in the NAcb shell (Hipólito et al., 2011). Accordingly, it has been recently shown that salsolinol excites DA neurons of the VTA, by activating µ-OR on local GABA interneurons (Xie et al., 2012).

Evidence of Behavioral Effects of Acetaldehyde Mediated by the Endogenous Opioid System

Whereas accumulating evidence indicates that the EOS participates in the behavioral effects of ethanol, only few studies have studied the involvement of this system in acetaldehyde effects. Self-administration of acetaldehyde appears to be mediated by the EOS; high doses of naloxone reduced intravenous acetaldehyde self-administration in rats, and naltrexone reduced the maintenance, the deprivation effect, and operant break points of acetaldehyde voluntary consumption (Myers et al., 1984; Peana et al., 2011). Treatment with naloxonazine, a specific µ1-OR antagonist reduces maintenance of acetaldehyde oral self-administration (Peana et al., 2011). Blockade of μ-OR using either naltrexone or the irreversible and selective μ-OR antagonist β-funaltrexamine suppress the locomotor stimulation effect of acetaldehyde when microinjected into the rat posterior VTA (Sánchez-Catalán et al., 2009). Additionally, Hipólito et al. (2010) have provided data supporting the hypothesis that acetaldehyde may mediate the actions of ethanol through a mechanism dependent on μ-OR activation. These authors showed that intra-posterior VTA injections of salsolinol induced locomotor stimulation and sensitization in rats; stimulation (but not sensitization) was prevented by μ-OR antagonism. Finally, Sanchis-Segura et al. (2005b) demonstrated that administration of a catalase inhibitor directly into the NArc is sufficient to prevent the effects of ethanol on rat locomotion. Conversely, locomotor stimulation induced by ethanol injected directly into the NArc, was prevented by catalase inhibition or naltrexone, indicating a link between the behavioral effects of a reduction in acetaldehyde formation and the antagonism of μ-OR (Pastor and Aragon, 2008). The NArc, therefore, may represent a critical site to link two independent but related hypotheses: (1) the hypothesis proposing that acetaldehyde may mediate some of the psychopharmacological actions attributed to ethanol (Aragon et al., 1992a; Smith et al., 1997; Quertemont et al., 2005; Correa et al., 2012) and (2) the hypothesis that suggests that the β-endorphin/µ-OR system participate in the reinforcing and psychomotor effects of ethanol (Stinus et al., 1980; Herz, 1997; Gianoulakis, 2001; Sanchis-Segura et al., 2005b; Pastor and Aragon, 2008). Early findings also suggested a role of the opioidergic system in mediating CPP induced by salsolinol in rats (Matsuzawa et al., 2000). Antagonism of μ-OR attenuated CPP induced by salsolinol when achieved under fear stress (Matsuzawa et al., 2000). Moreover, intra-posterior VTA administration of salsolinol, that produced CPP in rats, also produced an increase in DA in the NAcb that was suppressed by β-funaltrexamine administration (Hipólito et al., 2011).

Summary and Perspectives

In the present review we have summarized consistent results indicating that the EOS, and particularly β-endorphin and μ-OR, are critically involved in the psychopharmacological effects of ethanol. Additionally, we have reviewed a large body of data that indicates that the first metabolite of ethanol, acetaldehyde, might be responsible for the activation of β-endorphin release and μ-OR signaling after ethanol administration. There are two main lines of research suggesting a link between acetaldehyde and the EOS: (1) formation of acetaldehyde in brain areas such as the NArc, with high expression of ethanol metabolizing enzymes and presence of cell bodies of endorphinic neurons and (2) the formation of condensation products between DA and acetaldehyde such as salsolinol, which exerts its actions via μ-OR. To a certain degree both lines of research show important incompatibility. The fact that the lesions of the NArc are sufficient to block ethanol-induced behaviors challenge the putative role of salsolinol formed in other non-hypothalamic areas. Future studies will need to explore how to reconcile those two sets of data, and to clarify what is sufficient and/or necessary for acetaldehyde to induce behavioral responses mediated by the EOS. Finally, it is interesting to mention that most of the data suggesting a role of the EOS in acetaldehyde-induced behavioral effects have been linked to acetaldehyde-induced changes in the opioid system that are suggested to impact behavior via modulation of the DA system (Peana et al., 2011). Ethanol as well as acetaldehyde activate firing of dopaminergic neurons in the VTA (Foddai et al., 2004; Diana et al., 2008) and stimulate DA transmission in the NAcb (Melis et al., 2007; Enrico et al., 2009; Sirca et al., 2011), effects that are prevented by D-penicillamine, a sequestering agent of acetaldehyde (Enrico et al., 2009). A recent study demonstrates that in rats, ethanol and acetaldehyde induce via DA D1 receptors, ERK phosphorylation in the NAcb and extended amygdala (Vinci et al., 2010). This effect is blocked by D-penicillamine and by naltrexone, suggesting that the opiodergic modulation of the reinforcing properties of acetaldehyde could be mediated by the dopaminergic system (Vinci et al., 2010; Peana et al., 2011). There are other effects such as ethanol-induced CPP, ethanol drinking in some non-operant conditions and even ethanol-induced sensitization that appear to have a less straightforward involvement of DA signaling (Risinger et al., 1992; Broadbent et al., 1995; Spina et al., 2010; Young et al., 2013). Future research will need to investigate DA-dependent and independent mechanisms by which acetaldehyde might induce behavioral responses via its modulation of the EOS.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

This work was supported by grants from Fundación Bancaixa (P1-1A2011-05), Spain.

References

Abrahao, K. P., Quadros, I. M., and Souza-Formigoni, M. L. (2008). Morphine attenuates the expression of sensitization to ethanol, but opioid antagonists do not. Neuroscience 156, 857–864. doi: 10.1016/j.neuroscience.2008.08.012

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Akil, H., Owens, C., Gutstein, H., Taylor, L., Curran, E., and Watson, S. (1998). Endogenous opioids: overview and current issues. Drug Alcohol Depend. 51, 127–140. doi: 10.1016/s0376-8716(98)00071-4

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Anwer, J., and Soliman, M. R. (1995). Ethanol-induced alterations in beta-endorphin levels in specific rat brain regions: modulation by adenosine agonist and antagonist. Pharmacology 51, 364–369. doi: 10.1159/000139348

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Aragon, C. M., and Amit, Z. (1992). The effect of 3-amino-1,2,4-triazole on voluntary ethanol consumption: evidence for brain catalase involvement in the mechanism of action. Neuropharmacology 31, 709–712. doi: 10.1016/0028-3908(92)90150-n

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Aragon, C. M., Pesold, C. N., and Amit, Z. (1992b). Ethanol-induced motor activity in normal and acatalasemic mice. Alcohol 9, 207–211. doi: 10.1016/0741-8329(92)90055-f

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Aragon, C. M., Rogan, F., and Amit, Z. (1992a). Ethanol metabolism in rat brain homogenates by a catalase-H2O2 system. Biochem. Pharmacol. 44, 93–98. doi: 10.1016/0006-2952(92)90042-h

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Aragon, C. M., Spivak, K., and Amit, Z. (1985). Blockade of ethanol induced conditioned taste aversion by 3-amino-1,2,4-triazole: evidence for catalase mediated synthesis of acetaldehyde in rat brain. Life Sci. 37, 2077–2084. doi: 10.1016/0024-3205(85)90579-x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Arias, C., Molina, J. C., and Spear, N. E. (2010). Differential role of mu, delta and kappa opioid receptors in ethanol-mediated locomotor activation and ethanol intake in preweanling rats. Physiol. Behav. 99, 348–454. doi: 10.1016/j.physbeh.2009.11.012

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Arizzi, M. N., Correa, M., Betz, A. J., Wisniecki, A., and Salamone, J. D. (2003). Behavioral effects of intraventricular injections of low doses of ethanol, acetaldehyde, and acetate in rats: studies with low and high rate operant schedules. Behav. Brain Res. 147, 203–210. doi: 10.1016/s0166-4328(03)00158-x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Arizzi-LaFrance, M. N., Correa, M., Aragon, C. M., and Salamone, J. D. (2006). Motor stimulant effects of ethanol injected into the substantia nigra pars reticulata: importance of catalase-mediated metabolism and the role of acetaldehyde. Neuropsychopharmacology 31, 997–1008. doi: 10.1038/sj.npp.1300849

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bechtholt, A. J., and Cunningham, C. L. (2005). Ethanol-induced conditioned place preference is expressed through a ventral tagmental area dependent mechanism. Behav. Neurosci. 119, 213–223. doi: 10.1037/0735-7044.119.1.213

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bie, B., Zhu, W., and Pan, Z. Z. (2009). Ethanol-induced delta-opioid receptor modulation of glutamate synaptic transmission and conditioned place preference in central amygdala. Neuroscience 160, 348–358. doi: 10.1016/j.neuroscience.2009.02.049

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Bormann, N. M., and Cunningham, C. L. (1997). The effects of naloxone on expression and acquisition of ethanol place conditioning in rats. Pharmacol. Biochem. Behav. 58, 975–982.

Pubmed Abstract | Pubmed Full Text

Boyadjieva, N. I., and Sarkar, D. K. (1994). Effects of chronic alcohol on immunoreactive beta-endorphin secretion from hypothalamic neurons in primary cultures: evidence for alcohol tolerance, withdrawal, and sensitization responses. Alcohol. Clin. Exp. Res. 18, 1497–1501. doi: 10.1006/mcne.1994.1071

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Boyadjieva, N. I., and Sarkar, D. K. (1997). Effects of ethanol on basal and prostaglandin E1-induced increases in beta-endorphin release and intracellular cAMP levels in hypothalamic cells. Alcohol. Clin. Exp. Res. 21, 1005–1009. doi: 10.1111/j.1530-0277.1997.tb04245.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Broadbent, J., Grahame, N. J., and Cunningham, C. L. (1995). Haloperidol prevents ethanol-stimulated locomotor activity but fails to block sensitization. Psychopharmacology 120, 475–482. doi: 10.1007/bf02245821

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Brown, Z. W., Amit, Z., and Rockman, G. E. (1979). Intraventricular self-administration of acetaldehyde, but not ethanol, in naive laboratory rats. Psychopharmacology 64, 271–276. doi: 10.1007/bf00427509

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Burattini, C., Gill, T. M., Aicardi, G., and Janak, P. H. (2006). The ethanol self-administration context as a reinstatement cue: acute effects of naltrexone. Neuroscience 139, 877–887. doi: 10.1016/j.neuroscience.2006.01.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cacace, S., Plescia, F., Barberi, I., and Cannizzaro, C. (2012). Acetaldehyde oral self-administration: evidence from the operant-conflict paradigm. Alcohol. Clin. Exp. Res. 36, 1278–1287. doi: 10.1111/j.1530-0277.2011.01725.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cadet, P., Mantione, K. J., and Stefano, G. B. (2003). Molecular identification and functional expression of mu 3, a novel alternatively spliced variant of the human mu opiate receptor gene. J. Immunol. 170, 5118–5123.

Pubmed Abstract | Pubmed Full Text

Camarini, R., Nogueira Pires, M. L., and Calil, H. M. (2000). Involvement of the opioid system in the development and expression of sensitization to the locomotor-activating effect of ethanol. Int. J. Neuropsychopharmacol. 3, 303–309. doi: 10.1017/s146114570000211x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chavkin, C., James, I. F., and Goldstein, A. (1982). Dynorphin is a specific endogenous ligand of the kappa opioid receptor. Science 215, 413–415. doi: 10.1126/science.6120570

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Chronwall, B. M. (1985). Anatomy and physiology of the neuroendocrine arcuate nucleus. Peptides 6 (Suppl. 2), 1–11. doi: 10.1016/0196-9781(85)90128-7

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ciccocioppo, R., Martin-Fardon, R., and Weiss, F. (2002). Effect of selective blockade of mu(1) or delta opioid receptors on reinstatement of alcohol-seeking behavior by drug-associated stimuli in rats. Neuropsychopharmacology 27, 391–399. doi: 10.1016/s0893-133x(02)00302-0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ciccocioppo, R., Lin, D., Martin-Fardon, R., and Weiss, F. (2003). Reinstatement of ethanol-seeking behavior by drug cues following single versus multiple ethanol intoxication in the rat: effects of naltrexone. Psychopharmacology 168, 208–215. doi: 10.1007/s00213-002-1380-z

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Arizzi, M. N., Betz, A., Mingote, S., and Salamone, J. D. (2003a). Open field locomotor effects in rats after intraventricular injections of ethanol and the ethanol metabolites acetaldehyde and acetate. Brain Res. Bull. 62, 197–202. doi: 10.1016/j.brainresbull.2003.09.013

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Arizzi, M. N., Betz, A., Mingote, S., and Salamone, J. D. (2003b). Locomotor stimulant effects of intraventricular injections of low doses of ethanol in rats: acute and repeated administration. Psychopharmacology 170, 368–375. doi: 10.1007/s00213-003-1557-0

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Arizzi-LaFrance, M. N., and Salamone, J. D. (2009). Infusions of acetaldehyde into the arcuate nucleus of the hypothalamus induce motor activity in rats. Life Sci. 84, 321–327. doi: 10.1016/j.lfs.2008.12.013

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Manrique, H. M., Font, L., Escrig, M. A., and Aragon, C. M. (2008). Reduction in the anxiolytic effects of ethanol by centrally formed acetaldehyde: the role of catalase inhibitors and acetaldehyde-sequestering agents. Psychopharmacology 200, 455–464. doi: 10.1007/s00213-008-1219-3

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Miquel, M., and Aragon, C. M. (2000). Lead acetate potentiates brain catalase activity and enhances ethanol-induced locomotion in mice. Pharmacol. Biochem. Behav. 66, 137–142. doi: 10.1016/s0091-3057(00)00204-5

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Miquel, M., Sanchis-Segura, C., and Aragon, C. M. (1999a). Acute lead acetate administration potentiates ethanol-induced locomotor activity in mice: the role of brain catalase. Alcohol. Clin. Exp. Res. 23, 799–805.

Pubmed Abstract | Pubmed Full Text

Correa, M., Miquel, M., Sanchis-Segura, C., and Aragon, C. M. (1999b). Effects of chronic lead administration on ethanol-induced locomotor and brain catalase activity. Alcohol 19, 43–49. doi: 10.1016/s0741-8329(99)00023-3

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Salamone, J. D., Segovia, K. N., Pardo, M., Longoni, R., Spina, L., et al. (2012). Piecing together the puzzle of acetaldehyde as a neuroactive agent. Neurosci. Biobehav. Rev. 36, 404–430. doi: 10.1016/j.neubiorev.2011.07.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Correa, M., Sanchis-Segura, C., Pastor, R., and Aragon, C. M. (2004). Ethanol intake and motor sensitization: the role of brain catalase activity in mice with different genotypes. Physiol. Behav. 82, 231–40. doi: 10.1016/j.physbeh.2004.03.033

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Corwin, R. L., and Wojnicki, F. H. (2009). Baclofen, raclopride, and naltrexone differentially affect intake of fat and sucrose under limited access conditions. Behav. Pharmacol. 20, 537–548. doi: 10.1097/fbp.0b013e3283313168

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Cunningham, C. L., Henderson, C. M., and Bormann, N. M. (1998). Extinction of ethanol-induced conditioned place preference and conditioned place aversion: effects of naloxone. Psychopharmacology 139, 62–70. doi: 10.1007/s002130050690

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Dayas, C. V., Liu, X., Simms, J. A., and Weiss, F. (2007). Distinct patterns of neural activation associated with ethanol seeking: effects of naltrexone. Biol. Psychiatry 61, 979–989. doi: 10.1016/j.biopsych.2006.07.034

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

De, A., Boyadjieva, N., and Sarkar, D. K. (2002). Role of protein kinase C in control of ethanol-modulated beta-endorphin release from hypothalamic neurons in primary cultures. J. Pharmacol. Exp. Ther. 301, 119–128. doi: 10.1124/jpet.301.1.119

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

de Waele, J. P., Kiianmaa, K., and Gianoulakis, C. (1994). Spontaneous and ethanol-stimulated in vitro release of beta-endorphin by the hypothalamus of AA and ANA rats. Alcohol. Clin. Exp. Res. 18, 1468–1473. doi: 10.1111/j.1530-0277.1994.tb01452.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Dempsey, S., and Grisel, J. E. (2012). Locomotor sensitization to EtOH: contribution of β-Endorphin. Front. Mol. Neurosci. 5:87. doi: 10.3389/fnmol.2012.00087

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Devine, D. P., Leone, P., and Wise, R. A. (1993). Mesolimbic dopamine neurotransmission is increased by administration of mu-opioid receptor antagonists. Eur. J. Pharmacol. 243, 55–64. doi: 10.1016/0014-2999(93)90167-g

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Di Chiara, G., and North, R. A. (1992). Neurobiology of opiate abuse. Trends Pharmacol. Sci. 13, 185–193. doi: 10.1016/0165-6147(92)90062-b

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Diana, M., Peana, A. T., Sirca, D., Lintas, A., Melis, M., and Enrico, P. (2008). Crucial role of acetaldehyde in alcohol activation of the mesolimbic dopamine system. Ann. NY Acad. Sci. 1139, 307–317. doi: 10.1196/annals.1432.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Enrico, P., Sirca, D., Mereu, M., Peana, A. T., Lintas, A., Golosio, A., et al. (2009). Acetaldehyde sequestering prevents ethanol-induced stimulation of mesolimbic dopamine transmission. Drug Alcohol Depend. 100, 265–271. doi: 10.1016/j.drugalcdep.2008.10.010

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Evans, C. J., Keith, D. E. Jr., Morrison, H., Magendzo, K., and Edwards, R. H. (1992). Cloning of a delta opioid receptor by functional expression. Science 258, 1952–1955. doi: 10.1126/science.1335167

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Femenía, T., and Manzanares, J. (2012). Increased ethanol intake in prodynorphin knockout mice is associated to changes in opioid receptor function and dopamine transmission. Addict. Biol. 17, 322–337. doi: 10.1111/j.1369-1600.2011.00378.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Fertel, R. H., Greenwald, J. E., Schwarz, R., Wong, L., and Bianchine, J. (1980). Opiate receptor binding and analgesic effects of the tetrahydroisoquinolines salsolinol and tetrahydropapaveroline. Res. Commun. Chem. Pathol. Pharmacol. 27, 3–16.

Pubmed Abstract | Pubmed Full Text

Foddai, M., Dosia, G., Spiga, S., and Diana, M. (2004). Acetaldehyde increases dopaminergic neuronal activity in the VTA. Neuropsychopharmacology 29, 530–536. doi: 10.1038/sj.npp.1300326

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Font, L., Aragon, C. M., and Miquel, M. (2006a). Voluntary ethanol consumption decreases after the inactivation of central acetaldehyde by D-penicillamine. Behav. Brain Res. 171, 78–86. doi: 10.1016/j.bbr.2006.03.020

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Font, L., Miquel, M., and Aragon, C. (2005). Prevention of ethanol induced behavioral stimulation by D-penicillamine: a sequestration agent for acetaldehyde. Alcohol. Clin. Exp. Res. 29, 1156–1164. doi: 10.1097/01.alc.0000171945.30494.af

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Font, L., Miquel, M., and Aragon, C. (2006b). Ethanol-induced conditioned place preference, but not aversion, is blocked by treatment with D-penicillamine, an inactivation agent for acetaldehyde. Psychopharmacology 184, 56–64. doi: 10.1007/s00213-005-0224-z

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Font, L., Miquel, M., and Aragon, C. M. (2008). Involvement of brain catalase activity in the acquisition of ethanol-induced conditioned place preference. Physiol. Behav. 93, 733–741. doi: 10.1016/j.physbeh.2007.11.026

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Froehlich, J. C. (1995). Genetic factors in alcohol self-administration. J. Clin. Psychiatry 56(Suppl. 7), 15–23.

Pubmed Abstract | Pubmed Full Text

Froehlich, J. C., Zweifel, M., Harts, J., Lumeng, L., and Li, T. K. (1991). Importance of delta opioid receptors in maintaining high alcohol drinking. Psychopharmacology 103, 467–472. doi: 10.1007/bf02244246

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gevaerd, M. S., Sultowski, E. T., and Takahashi, R. N. (1999). Combined effects of diethylpropion and alcohol on locomotor activity of mice: participation of the dopaminergic and opioid systems. Braz. J. Med. Biol. Res. 32, 1545–1550. doi: 10.1590/S0100-879X1999001200015

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gianoulakis, C. (1990). Characterization of the effects of acute ethanol administration on the release of beta-endorphin peptides by the rat hypothalamus. Eur. J. Pharmacol. 180, 21–29. doi: 10.1016/0014-2999(90)90588-w

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gianoulakis, C. (1993). Endogenous opioids and excessive alcohol consumption. J. Psychiatry Neurosci. 18, 148–156.

Pubmed Abstract | Pubmed Full Text

Gianoulakis, C. (2001). Influence of the endogenous opioid system on high alcohol consumption and genetic predisposition to alcoholism. J. Psychiatry Neurosci. 26, 304–318.

Pubmed Abstract | Pubmed Full Text

Gianoulakis, C., Hutchison, W. D., and Kalant, H. (1988). Effects of ethanol treatment and withdrawal on biosynthesis and processing of proopiomelanocortin by the rat neurointermediate lobe. Endocrinology 122, 817–825. doi: 10.1210/endo-122-3-817

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gill, K., Menez, J. F., Lucas, D., and Deitrich, R. A. (1992). Enzymatic production of acetaldehyde from ethanol in rat brain tissue. Alcohol. Clin. Exp. Res. 16, 910–915. doi: 10.1111/j.1530-0277.1992.tb01892.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Gremel, C. M., Young, E. A., and Cunningham, C. L. (2011). Blockade of opioid receptors in anterior cingulate cortex disrupts ethanol-seeking behavior in mice. Behav. Brain Res. 219, 358–362. doi: 10.1016/j.bbr.2010.12.033

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hall, F. S., Sora, I., and Uhl, G. R. (2001). Ethanol consumption and reward are decreased in µ-opiate receptor knockout mice. Psychopharmacology 154, 43–49. doi: 10.1007/s002130000622

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hamby-Mason, R., Chen, J. J., Schenker, S., Perez, A., and Henderson, G. I. (1997). Catalase mediates acetaldehyde formation from ethanol in fetal and neonatal rat brain. Alcohol. Clin. Exp. Res. 21, 1063–72. doi: 10.1111/j.1530-0277.1997.tb04255.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Herz, A. (1997). Endogenous opioid systems and alcohol addiction. Psychopharmacology 129, 99–111. doi: 10.1007/s002130050169

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hipólito, L., Martí-Prats, L., Sánchez-Catalán, M. J., Polache, A., and Granero, L. (2011). Induction of conditioned place preference and dopamine release by salsolinol in posterior VTA of rats: involvement of μ-opioid receptors. Neurochem. Int. 59, 559–562. doi: 10.1016/j.neuint.2011.04.014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hipólito, L., Sánchez-Catalán, M. J., Granero, L., and Polache, A. (2009). Local salsolinol modulates dopamine extracellular levels from rat nucleus accumbens: shell/core differences. Neurochem. Int. 55, 187–192. doi: 10.1016/j.neuint.2009.02.014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hipólito, L., Sánchez-Catalán, M. J., Martí-Prats, L., Granero, L., and Polache, A. (2012). Revisiting the controversial role of salsolinol in the neurobiological effects of ethanol: old and new vistas. Neurosci. Biobehav. Rev. 36, 362–378. doi: 10.1016/j.neubiorev.2011.07.007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hipólito, L., Sánchez-Catalán, M. J., Zanolini, I., Polache, A., and Granero, L. (2008). Shell/core differences in mu- and delta-opioid receptor modulation of dopamine efflux in nucleus accumbens. Neuropharmacology 55, 183–189. doi: 10.1016/j.neuropharm.2008.05.012

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hipólito, L., Sánchez-Catalán, M. J., Zornoza, T., Polache, A., and Granero, L. (2010). Locomotor stimulant effects of acute and repeated intrategmental injections of salsolinol in rats : role of μ-opioid receptors. Psychopharmacology 209, 1–11. doi: 10.1007/s00213-009-1751-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Holstein, S. E., Pastor, R., Meyer, P. J., and Phillips, T. J. (2005). Naloxone does not attenuate the locomotor effects of ethanol in FAST, SLOW, or two heterogeneous stocks of mice. Psychopharmacology 182, 277–289. doi: 10.1007/s00213-005-0066-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Hyytiä, P., and Kiianmaa, K. (2001). Suppression of ethanol responding by centrally administered CTOP and naltrindole in AA and Wistar rats. Alcohol. Clin. Exp. Res. 25, 25–33. doi: 10.1111/j.1530-0277.2001.tb02123.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Ingman, K., Salvadori, S., Lazarus, L., Korpi, E. R., and Honkanen, A. (2003). Selective delta-opioid receptor antagonist N,N(CH3)2-Dmt-Tic-OH does not reduce ethanol intake in alcohol-preferring AA rats. Addict. Biol. 8, 173–179. doi: 10.1080/1355621031000117400

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jamal, M., Ameno, K., Uekita, I., Kumihashi, M., Wang, W., and Ijiri, I. (2007). Catalase mediates acetaldehyde formation in the striatum of free-moving rats. Neurotoxicology 28, 1245–1248. doi: 10.1016/j.neuro.2007.05.002

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Jarjour, S., Bai, L., and Gianoulakis, C. (2009). Effect of acute ethanol administration on the release of opioid peptides from the midbrain including the ventral tegmental area. Alcohol. Clin. Exp. Res. 33, 1033–1043. doi: 10.1111/j.1530-0277.2009.00924.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

June, H. L., Grey, C., Warren-Reese, C., Durr, L. F., Ricks-Cord, A., Johnson, A., et al. (1999). The opioid receptor antagonist nalmefene reduces responding maintained by ethanol presentation: preclinical studies in ethanol-preferring and outbred wistar rats. Alcohol. Clin. Exp. Res. 22, 2174–2185. doi: 10.1111/j.1530-0277.1998.tb05931.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Karahanian, E., Quintanilla, M. E., Tampier, L., Rivera-Meza, M., Bustamante, D., Gonzalez-Lira, V., et al. (2011). Ethanol as a prodrug: brain metabolism of ethanol mediates its reinforcing effects. Alcohol. Clin. Exp. Res. 35, 606–612. doi: 10.1111/j.1530-0277.2011.01439.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Khachaturian, H., Lewis, M. E., Alessi, N. E., and Watson, S. J. (1985). Time of origin of opioid peptide-containing neurons in the rat hypothalamus. J. Comp. Neurol. 236, 538–546. doi: 10.1002/cne.902360409

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kieffer, B. L., and Evans, C. J. (2009). Opioid receptors: from binding sites to visible molecules in vivo. Neuropharmacology 56(Suppl. 1), 205–212. doi: 10.1016/j.neuropharm.2008.07.033

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kieffer, B. L., and Gavériaux-Ruff, C. (2002). Exploring the opioid system by gene knockout. Prog. Neurobiol. 66, 285–306. doi: 10.1016/s0301-0082(02)00008-4

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kiianmaa, K., Hoffman, P., and Tabakoff, B. (1983). Antagonism of the behavioral effects of ethanol by naltrexone in BALB/c, C57BL/6, and DBA/2 mice. Psychopharmacology 79, 291–294. doi: 10.1007/bf00433403

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Knapp, R. J., Malatynska, E., Collins, N., Fang, L., Wang, J. Y., Hruby, V. J., et al. (1995). Molecular biology and pharmacology of cloned opioid receptors. FASEB J. 9, 516–525.

Pubmed Abstract | Pubmed Full Text

Koechling, U. M., and Amit, Z. (1994). Effects of 3-amino-1,2,4-triazole on brain catalase in the mediation of ethanol consumption in mice. Alcohol 11, 235–239. doi: 10.1016/0741-8329(94)90036-1

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Krishnan-Sarin, S., Portoghese, P. S., Li, T. K., and Froehlich, J. C. (1995). The delta 2-opioid receptor antagonist naltriben selectively attenuates alcohol intake in rats bred for alcohol preference. Pharmacol. Biochem. Behav. 52, 153–159. doi: 10.1016/0091-3057(95)00080-g

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Kuzmin, A., Sandin, J., Terenius, L., and Ogren, S. O. (2003). Acquisition, expression, and reinstatement of ethanol-induced conditioned place preference in mice: effects of opioid receptor-like 1 receptor agonists and naloxone. J. Pharmacol. Exp. Ther. 304, 310–318. doi: 10.1124/jpet.102.041350

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lam, M. P., Marinelli, P. W., Bai, L., and Gianoulakis, C. (2008). Effects of acute ethanol on opioid peptide release in the central amygdala: an in vivo microdialysis study. Psychopharmacology 201, 261–271. doi: 10.1007/s00213-008-1267-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lê, A. D., Poulos, C. X., Harding, S., Watchus, J., Juzytsch, W., and Shaham, Y. (1999). Effects of naltrexone and fluoxetine on alcohol self-administration and reinstatement of alcohol seeking induced by priming injections of alcohol and exposure to stress. Neuropsychopharmacology 21, 435–444. doi: 10.1016/s0893-133x(99)00024-x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lê, A. D., Poulos, C. X., Quan, B., and Chow, S. (1993). The effects of selective blockade of delta and mu opiate receptors on ethanol consumption by C57BL/6 mice in a restricted access paradigm. Brain Res. 630, 330–332. doi: 10.1016/0006-8993(93)90672-a

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Leriche, M., and Méndez, M. (2010). Ethanol exposure selectively alters beta-endorphin content but not [3H]-DAMGO binding in discrete regions of the rat brain. Neuropeptides 44, 9–16. doi: 10.1016/j.npep.2009.11.009

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lesscher, H. M., Bailey, A., Burbach, J. P., Van Ree, J. M., Kitchen, I., and Gerrits, M. A. (2003). Receptor-selective changes in mu-, delta- and kappa-opioid receptors after chronic naltrexone treatment in mice. Eur. J. Neurosci. 17, 1006–1012.

Pubmed Abstract | Pubmed Full Text

Liu, X., and Weiss, F. (2002). Additive effect of stress and drug cues on reinstatement of ethanol seeking: exacerbation by history of dependence and role of concurrent activation of corticotropin-releasing factor and opioid mechanisms. J. Neurosci. 22, 7856–7861.

Pubmed Abstract | Pubmed Full Text

Logrip, M. L., Janak, P. H., and Ron, D. (2009). Blockade of ethanol reward by the kappa opioid receptor. Alcohol 43, 359–365. doi: 10.1016/j.alcohol.2009.05.001

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Lucchi, L., Bosio, A., Spano, P. F., and Trabucchi, M. (1982). Action of ethanol and salsolinol on opiate receptor function. Brain Res. 232, 506–510. doi: 10.1016/0006-8993(82)90297-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Manrique, H. M., Miquel, M., and Aragon, C. M. (2005). Brain catalase mediates potentiation of social recognition memory produced by ethanol in mice. Drug Alcohol Depend. 79, 343–350. doi: 10.1016/j.drugalcdep.2005.02.007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Mansour, A., Khachaturian, H., Lewis, M. E., Akil, H., and Watson, S. J. (1988). Anatomy of CNS opioid receptors. Trends Neurosci. 11, 308–314. doi: 10.1016/0166-2236(88)90093-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Margolis, E. B., Fields, H. L., Hjelmstad, G. O., and Mitchell, J. M. (2008). Delta-opioid receptor expression in the ventral tegmental area protects against elevated alcohol consumption. J. Neurosci. 28, 12672–12681. doi: 10.1523/jneurosci.4569-08.2008

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Marinelli, P. W., Funk, D., Harding, S., Li, Z., Juzytsch, W., and Lê, A. D. (2009). Roles of opioid receptor subtypes in mediating alcohol seeking induced by discrete cues and context. Eur. J. Neurosci. 30, 671–678. doi: 10.1111/j.1460-9568.2009.06851.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Marinelli, P. W., Quirion, R., and Gianoulakis, C. (2003a). Estradiol valerate and alcohol intake: a comparison between Wistar and Lewis rats and the putative role of endorphins. Behav. Brain Res. 139, 59–67. doi: 10.1016/s0166-4328(02)00057-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Marinelli, P. W., Quirion, R., and Gianoulakis, C. (2003b). A microdialysis profile of beta-endorphin and catecholamines in the rat nucleus accumbens following alcohol administration. Psychopharmacology 169, 60–67. doi: 10.1007/s00213-003-1490-2

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Martí-Prats, L., Sánchez-Catalán, M. J., Hipólito, L., Orrico, A., Zornoza, T., Polache, A., et al. (2010). Systemic administration of D-penicillamine prevents the locomotor activation after intra-VTA ethanol administration in rats. Neurosci. Lett. 483, 143–147. doi: 10.1016/j.neulet.2010.07.081

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Matsuzawa, S., Suzuki, T., and Misawa, M. (2000). Involvement of mu-opioid receptor in the salsolinol-associated place preference in rats exposed to conditioned fear stress. Alcohol. Clin. Exp. Res. 24, 366–372. doi: 10.1111/j.1530-0277.2000.tb04624.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

McBride, W. J., Li, T. K., Deitrich, R. A., Zimatkin, S., Smith, B. R., and Rodd-Henricks, Z. A. (2002). Involvement of acetaldehyde in alcohol addiction. Alcohol. Clin. Exp. Res. 26, 114–119.

Pubmed Abstract | Pubmed Full Text

Melis, M., Enrico, P., Peana, A. T., and Diana, M. (2007). Acetaldehyde mediates alcohol activation of the mesolimbic dopamine system. Eur. J. Neurosci. 26, 2824–33. doi: 10.1111/j.1460-9568.2007.05887.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Méndez, M., and Morales-Mulia, M. (2008). Role of mu and delta opioid receptors in alcohol drinking behaviour. Curr. Drug Abuse Rev. 1, 239–252. doi: 10.2174/1874473710801020239

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Middaugh, L. D., and Bandy, A. L. (2000). Naltrexone effects on ethanol consumption and response to ethanol conditioned cues in C57BL/6 mice. Psychopharmacology 151, 321–327. doi: 10.1007/s002130000479

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Milton, G. W., Verhaert, P. D., and Downer, R. G. (1991). Immunofluorescent localization of dopamine-like and leucine-enkephalin-like neurons in the supraoesophageal ganglia of the american cockroach, periplaneta americana. Tissue Cell 23, 331–340. doi: 10.1016/0040-8166(91)90051-t

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Miquel, M., Font, L., Sanchis-Segura, C., and Aragon, C. M. (2003). Neonatal administration of monosodium glutamate prevents the development of ethanol-but not psychostimulant-induced sensitization: a putative role of the arcuate nucleus. Eur. J. Neurosci. 17, 2163–2170. doi: 10.1046/j.1460-9568.2003.02646.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Modesto-Lowe, V., and Fritz, E. M. (2005). The opioidergic-alcohol link: implications for treatment. CNS Drugs 19, 693–707. doi: 10.2165/00023210-200519080-00005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Moreno, S., Mugnaini, E., and Cerù, M. P. (1995). Immunocytochemical localization of catalase in the central nervous system of the rat. J. Histochem. Cytochem. 43, 1253–1267. doi: 10.1177/43.12.8537642

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Myers, W., Ng, K., and Singer, G. (1984). Effects of naloxone and buprenorphine on intravenous acetaldehyde self-injection in rats. Physiol. Behav. 33, 449–455. doi: 10.1016/0031-9384(84)90168-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nielsen, C. K., Simms, J. A., Pierson, H. B., Li, R., Saini, S. K., Ananthan, S., et al. (2008). A novel delta opioid receptor antagonist, SoRI-9409, produces a selective and long-lasting decrease in ethanol consumption in heavy-drinking rats. Biol. Psychiatry 64, 974–981. doi: 10.1016/j.biopsych.2008.07.018

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Nylander, I., and Roman, E. (2012). Neuropeptides as mediators of the early-life impact on the brain; implications for alcohol use disorders. Front. Mol. Neurosci. 5:77. doi: 10.3389/fnmol.2012.00077

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Olive, M. F., Koenig, H. N., Nannini, M. A., and Hodge, C. W. (2001). Stimulation of endorphin neurotransmission in the nucleus accumbens by ethanol, cocaine, and amphetamine. J. Neurosci. 21, RC184.

Pubmed Abstract | Pubmed Full Text

O’Malley, S. S., Jaffe, A. J., Chang, G., Schottenfeld, R. S., Meyer, R. E., and Rounsaville, B. (1992). Naltrexone and coping skills therapy for alcohol dependence. A controlled study. Arch. Gen. Psychiatry 49, 881–887. doi: 10.1001/archpsyc.1992.01820110045007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Orrico, A., Hipólito, L., Sánchez-Catalán, M. J., Martí-Prats, L., Zornoza, T., Granero, L., et al. (2013). Efficacy of D-penicillamine, a sequestering acetaldehyde agent, in the prevention of alcohol relapse-like drinking in rats. Psychopharmacology doi: 10.1007/s00213-013-3065-1. [Epub ahead of print]

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Oswald, L. M., and Wand, G. S. (2004). Opioids and alcoholism. Physiol. Behav. 81, 339–358. doi: 10.1016/j.physbeh.2004.02.008

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pasternak, G. W. (2001a). The pharmacology of mu analgesics: from patients to genes. Neuroscientist 7, 220–231. doi: 10.1177/107385840100700307

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pasternak, G. W. (2001b). Insights into mu opioid pharmacology the role of mu opioid receptor subtypes. Life Sci. 68, 2213–229. doi: 10.1016/S0024-3205(01)01008-6

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pastor, R., and Aragon, C. M. (2006). The role of opioid receptor subtypes in the development of behavioral sensitization to ethanol. Neuropsychopharmacology 31, 1489–1499. doi: 10.1038/sj.npp.1300928

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pastor, R., and Aragon, C. M. (2008). Ethanol injected into the hypothalamic arcuate nucleus induces behavioral stimulation in rats: an effect prevented by catalase inhibition and naltrexone. Behav. Pharmacol. 19, 698–705. doi: 10.1097/fbp.0b013e328315ecd7

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pastor, R., Font, L., Miquel, M., Phillips, T. J., and Aragon, C. M. (2011). Involvement of the beta-endorphin neurons of the hypothalamic arcuate nucleus in ethanol-induced place preference conditioning in mice. Alcohol. Clin. Exp. Res. 35, 2019–2029. doi: 10.1111/j.1530-0277.2011.01553.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pastor, R., Sanchis-Segura, C., and Aragon, C. M. (2002). Ethanol-stimulated behaviour in mice is modulated by brain catalase activity and H2O2 rate of production. Psychopharmacology 165, 51–59. doi: 10.1007/s00213-002-1241-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pastor, R., Sanchis-Segura, C., and Aragon, C. (2005). Effect of selective antagonism of mu(1)-, mu(1/2)-, mu(3)-, and delta-opioid receptors on the locomotor-stimulating actions of ethanol. Drug Alcohol Depend. 78, 289–295. doi: 10.1016/j.drugalcdep.2004.11.007

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pastorcic, M., Boyadjieva, N., and Sarkar, D. K. (1994). Comparison of the effects of alcohol and acetaldehyde on proopiomelanocortin mRNA levels and beta-endorphin secretion from hypothalamic neurons in primary cultures. Mol. Cell. Neurosci. 5, 580–586. doi: 10.1006/mcne.1994.1071

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Patel, V. A., and Pohorecky, L. A. (1989). Acute and chronic ethanol treatment on beta-endorphin and catecholamine levels. Alcohol 6, 59–63. doi: 10.1016/0741-8329(89)90074-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pautassi, R. M., Nizhnikov, M. E., Acevedo, M. B., and Spear, N. E. (2012). Early role of the κ opioid receptor in ethanol-induced reinforcement. Physiol. Behav. 105, 1231–1241. doi: 10.1016/j.physbeh.2012.01.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Pautassi, R. M., Nizhnikov, M. E., Fabio, M. C., and Spear, N. E. (2011). An acetaldehyde-sequestering agent inhibits appetitive reinforcement and behavioral stimulation induced by ethanol in preweanling rats. Pharmacol. Biochem. Behav. 97, 462–469. doi: 10.1016/j.pbb.2010.10.005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Peana, A. T., Assaretti, A. R., Muggironi, G., Enrico, P., and Diana, M. (2009). Reduction of ethanol-derived acetaldehyde induced motivational properties by L-cysteine. Alcohol. Clin. Exp. Res. 33, 43–48. doi: 10.1111/j.1530-0277.2008.00809.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Peana, A. T., Enrico, P., Assaretti, A. R., Pulighe, E., Muggironi, G., Nieddu, M., et al. (2008). Key role of ethanol-derived acetaldehyde in the motivational properties induced by intragastric ethanol: a conditioned place preference study in the rat. Alcohol. Clin. Exp. Res. 32, 249–258. doi: 10.1111/j.1530-0277.2007.00574.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Peana, A. T., Muggironi, G., and Diana, M. (2010). Acetaldehyde-reinforcing effects; a study on oral self-administration behavior. Front. Psychiatry 1:23. doi: 10.3389/fpsyt.2010.00023

Pubmed Abstract | Pubmed Full Text

Peana, A. T., Muriggironi, G., Fois, G. R., Zinellu, M., Sirca, D., and Diana, M. (2012). Effect of (L)-cysteine on acetaldehyde self-administration. Alcohol 46, 489–497. doi: 10.1016/j.alcohol.2011.10.004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Peana, A. T., Muggironi, G., Fois, G. R., Zinellu, M., Vinci, S., and Acquas, E. (2011). Effect of opioid receptor blockade on acetaldehyde self-administration and ERK phosphorylation in the rat nucleus accumbens. Alcohol 45, 773–783. doi: 10.1016/j.alcohol.2011.06.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Peciña, S., and Berridge, K. C. (2005). Hedonic hot spot in nucleus accumbens shell: where do mu-opioids cause increased hedonic impact of sweetness?. J. Neurosci. 25, 11777–11786. doi: 10.1523/jneurosci.2329-05.2005

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Popp, R. L., and Erickson, C. K. (1998). The effect of an acute ethanol exposure on the rat brain POMC opiopeptide system. Alcohol 16, 139–148. doi: 10.1016/s0741-8329(98)00003-2

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Quertemont, E., and De Witte, P. (2001). Conditioned stimulus preference after acetaldehyde but not ethanol injections. Pharmacol. Biochem. Behav. 68, 449–454. doi: 10.1016/s0091-3057(00)00486-x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Quertemont, E., Tambour, S., and Tirelli, E. (2005). The role of acetaldehyde in the neurobehavioral effects of ethanol: a comprehensive review of animal studies. Prog. Neurobiol. 75, 247–274. doi: 10.1016/j.pneurobio.2005.03.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Rao, R. E., Wojnicki, F. H., Coupland, J., Ghosh, S., and Corwin, R. L. (2008). Baclofen, raclopride, and naltrexone differentially reduce solid fat emulsion intake under limited access conditions. Pharmacol. Biochem. Behav. 89, 581–590. doi: 10.1016/j.pbb.2008.02.013

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Rasmussen, D. D., Boldt, B. M., Wilkinson, C. W., and Mitton, D. R. (2002). Chronic daily ethanol and withdrawal: 3. Forebrain pro-opiomelanocortin gene expression and implications for dependence, relapse, and deprivation effect. Alcohol. Clin. Exp. Res. 26, 535–546. doi: 10.1111/j.1530-0277.2002.tb02572.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Rasmussen, D. D., Bryant, C. A., Boldt, B. M., Colasurdo, E. A., Levin, N., and Wilkinson, C. W. (1998). Acute alcohol effects on opiomelanocortinergic regulation. Alcohol. Clin. Exp. Res. 22, 789–801. doi: 10.1111/j.1530-0277.1998.tb03870.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Raynor, K., Kong, H., Chen, Y., Yasuda, K., Yu, L., Bell, G. I., et al. (1994). Pharmacological characterization of the cloned kappa-, delta-, and mu-opioid receptors. Mol. Pharmacol. 45, 330–334.

Pubmed Abstract | Pubmed Full Text

Reddy, B. V., and Sarkar, D. K. (1993). Effect of alcohol, acetaldehyde, and salsolinol on beta-endorphin secretion from the hypothalamic neurons in primary cultures. Alcohol. Clin. Exp. Res. 17, 1261–1267. doi: 10.1111/j.1530-0277.1993.tb05239.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Reddy, B. V., Boyadjieva, N., and Sarkar, D. K. (1995). Effect of ethanol, propanol, butanol, and catalase enzyme blockers on beta-endorphin secretion from primary cultures of hypothalamic neurons: evidence for a mediatory role of acetaldehyde in ethanol stimulation of beta-endorphin release. Alcohol. Clin. Exp. Res. 19, 339–344. doi: 10.1111/j.1530-0277.1995.tb01512.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Risinger, F. O., Dickinson, S. D., and Cunningham, C. L. (1992) Haloperidol reduces ethanol-induced motor activity stimulation but not conditioned place preference. Psychopharmacology 107, 453–456. doi: 10.1007/bf02245175

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Roberts, A. J., McDonald, J. S., Heyser, C. J., Kieffer, B. L., Matthes, H. W., Koob, G. F., et al. (2000). Mu-Opioid receptor knockout mice do not self-administer alcohol. J. Pharmacol. Exp. Ther. 293, 1002–1008.

Pubmed Abstract | Pubmed Full Text

Rodd, Z., Bell, R. L., Zhang, Y., Murphy, J. M., Goldstein, A., Zaffaroni, A., et al. (2005). Regional heterogeneity for the intracranial self-administration of ethanol and acetaldehyde within the ventral tegmental area of alcohol-preferring (P) rats: involvement of dopamine and serotonin. Neuropsychopharmacology 30, 330–338. doi: 10.1038/sj.npp.1300561

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Rodd-Henricks, Z. A., Melendez, R. I, Zaffaroni, A., Goldstein, A., McBride, W. J., and Lu, T. K. (2002). The reinforcing effects of acetaldehyde in the posterior ventral tegmental area of alcohol-preferring rats. Pharmacol. Biochem. Behav. 72, 55–64. doi: 10.1016/s0091-3057(01)00733-x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Roth-Deri, I., Green-Sadan, T., and Yadid, G. (2008). Beta-endorphin and drug-induced reward and reinforcement. Prog. Neurobiol. 86, 1–21. doi: 10.1016/j.pneurobio.2008.06.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sánchez-Catalán, M. J., Hipólito, L., Zornoza, T., Polache, A., and Granero, L. (2009). Motor stimulant effects of ethanol and acetaldehyde injected into the posterior ventral tegmental area of rats: role of opioid receptors. Psychopharmacology 204, 641–653. doi: 10.1007/s00213-009-1495-6

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Correa, M., and Aragon, C. M. (2000). Lession on the hypothalamic arcuate nucleus by estradiol valerate results in a blockade of ethanol-induced locomotion. Behav. Brain Res. 114, 57–63. doi: 10.1016/s0166-4328(00)00183-2

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Correa, M., Miquel, M., and Aragon, C. M. (2005b). Catalase inhibition in the arcuate nucleus blocks ethanol effects on the locomotor activity of rats. Neurosci. Lett. 376, 66–70. doi: 10.1016/j.neulet.2004.11.025

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Grisel, J. E., Olive, M. F., Ghozland, S., Koob, G. F., Roberts, A. J., et al. (2005a). Role of the endogenous opioid system on the neuropsychopharmacological effects of ethanol: new insights about an old question. Alcohol. Clin. Exp. Res. 29, 1522–1527. doi: 10.1097/01.alc.0000174913.60384.e8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Miquel, M., Correa, M., and Aragon, C. M. (1999a). Daily injections of cyanamide enhance both ethanol-induced locomotion and brain catalase activity. Behav. Pharmacol. 10, 459–465. doi: 10.1097/00008877-199909000-00004

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Miquel, M., Correa, M., and Aragon, C. M. (1999b). The catalase inhibitor sodium azide reduces ethanol-induced locomotor activity. Alcohol 19, 37–42. doi: 10.1016/s0741-8329(99)00016-6

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Miquel, M., Correa, M., and Aragon, C. M. (1999c). Cyanamide reduces brain catalase and ethanol-induced locomotor activity: is there a functional link? Psychopharmacology 144, 83–89. doi: 10.1007/s002130050980

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sanchis-Segura, C., Pastor, R., and Aragon, C. M. (2004). Opposite effects of acute versus chronic naltrexone administration on ethanol-induced locomotion. Behav. Brain Res. 153, 61–67. doi: 10.1016/j.bbr.2003.11.003

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Schulz, R., Wüster, M., Duka, T., and Herz, A. (1980). Acute and chronic ethanol treatment changes endorphin levels in brain and pituitary. Psychopharmacology 68, 221–227. doi: 10.1007/bf00428107

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Seizinger, B. R., Bovermann, K., Maysinger, D., Höllt, V., and Herz, A. (1983). Differential effects of acute and chronic ethanol treatment on particular opioid peptide systems in discrete regions of rat brain and pituitary. Pharmacol. Biochem. Behav. 18(Suppl. 1), 361–369. doi: 10.1016/0091-3057(83)90200-9

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Seizinger, B. R., Höllt, V., and Herz, A. (1984). Effects of chronic ethanol treatment on the in vitro biosynthesis of pro-opiomelanocortin and its posttranslational processing to beta-endorphin in the intermediate lobe of the rat pituitary. J. Neurochem. 43, 607–613. doi: 10.1111/j.1471-4159.1984.tb12778.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Simms, J. A., Steensland, P., Medina, B., Abernathy, K. E., Chandler, L. J., Wise, R., et al. (2008). Intermittent access to 20% ethanol induces high ethanol consumption in Long-Evans and Wistar rats. Alcohol. Clin. Exp. Res. 32, 1816–1823. doi: 10.1111/j.1530-0277.2008.00753.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Simon, E. J. (1991). Opioid receptors and endogenous opioid peptides. Med. Res. Rev. 11, 357–374. doi: 10.1002/med.2610110402

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Sirca, D., Enrico, P., Mereu, M., Peana, A. T., and Diana, M. (2011). L-cysteine prevents ethanol-induced stimulation of mesolimbic dopamine transmission. Alcohol. Clin. Exp. Res. 35, 862–869. doi: 10.1111/j.1530-0277.2010.01416.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Smith, B. R., Amit, Z., and Splawinsky, J. (1984). Conditioned place preference induced by intraventricular infusions of acetaldehyde. Alcohol 1, 193–195. doi: 10.1016/0741-8329(84)90097-1

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Smith, B. R., Aragon, C. M. G., and Amit, Z. (1997). Catalase and the production of central acetaldehyde: a possible mediator of the psychopharmacological effects of ethanol. Addict. Biol. 2, 277–289. doi: 10.1080/13556219772570

CrossRef Full Text

Spanagel, R., Herz, A., and Shippenberg, T. S. (1992). Opposing tonically active endogenous opioid systems modulate the mesolimbic dopaminergic pathway. Proc. Natl. Acad. Sci. U S A 89, 2046–2050. doi: 10.1073/pnas.89.6.2046

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Spina, L., Longoni, R., Vinci, S., Ibba, F., Peana, A. T., Muggironi, G., et al. (2010). Role of dopamine D1 receptors and extracellular signal regulated kinase in the motivational properties of acetaldehyde as assessed by place preference conditioning. Alcohol. Clin. Exp. Res. 34, 607–616. doi: 10.1111/j.1530-0277.2009.01129.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Stinus, L., Koob, G. F., Ling, N., Bloom, F. E., and Le Moal, M. (1980). Locomotor activation induced by infusion of endorphins into the ventral tegmental area: evidence for opiate-dopamine interactions. Proc. Natl. Acad. Sci. U S A 77, 2323–2327. doi: 10.1073/pnas.77.4.2323

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Tarragón, E., Baliño, P., Aragon, C. M., and Pastor, R. (2012). Ethanol drinking-in-the-dark facilitates behavioral sensitization to ethanol in C57BL/6J, BALB/cByJ, but not in mu-opioid receptor deficient CXBK mice. Pharmacol. Biochem. Behav. 101, 14–23. doi: 10.1016/j.pbb.2011.11.014

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Thorsell, A. (2013). The μ-opioid receptor and treatment response to naltrexone. Alcohol Alcohol. 48, 402–408. doi: 10.1093/alcalc/agt030

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Trigo, J. M., Martin-García, E., Berrendero, F., Robledo, P., and Maldonado, R. (2010). The endogenous opioid system: a common substrate in drug addiction. Drug Alcohol Depend. 108, 183–194. doi: 10.1016/j.drugalcdep.2009.10.011

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Unterwald, E. M., Anton, B., To, T., Lam, H., and Evans, C. J. (1998). Quantitative immunolocalization of mu opioid receptors: regulation by naltrexone. Neuroscience 85, 897–905. doi: 10.1016/s0306-4522(97)00659-3

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Vengeliene, V., Bilbao, A., Molander, A., and Spanagel, R. (2008). Neuropharmacology of alcohol addiction. Br. J. Pharmacol. 154, 299–315. doi: 10.1038/bjp.2008.30

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Vinci, S., Ibba, F., Longoni, R., Spina, L., Spiga, S., and Acquas, E. (2010). Acetaldehyde elicits ERK phosphorylation in the rat nucleus accumbens and extended amygdala. Synapse 64, 916–927. doi: 10.1002/syn.20811

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Walker, B. M., Valdez, G. R., McLaughlin, J. P., and Bakalkin, G. (2012). Targeting dynorphin/kappa opioid receptor systems to treat alcohol abuse and dependence. Alcohol 46, 359–370. doi: 10.1016/j.alcohol.2011.10.006

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wand, G. S. (1990). Differential regulation of anterior pituitary corticotrope function is observed in vivo but not in vitro in two lines of ethanol-sensitive mice. Alcohol. Clin. Exp. Res. 14, 100–106. doi: 10.1111/j.1530-0277.1990.tb00454.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wee, S., and Koob, G. F. (2010). The role of the dynorphin-kappa opioid system in the reinforcing effects of drugs of abuse. Psychopharmacology 210, 121–135. doi: 10.1007/s00213-010-1825-8

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Wong, K. J., Wojnicki, F. H., and Corwin, R. L. (2009). Baclofen, raclopride, and naltrexone differentially affect intake of fat/sucrose mixtures under limited access conditions. Pharmacol. Biochem. Behav. 92, 528–536. doi: 10.1016/j.pbb.2009.02.002

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Xie, G., Hipólito, L., Zuo, W., Polache, A., Granero, L., Krnjevic, K., et al. (2012). Salsolinol stimulates dopamine neurons in slices of posterior ventral tegmental area indirectly by activating-opioid receptors. J. Pharmacol. Exp. Ther. 341, 43–50. doi: 10.1124/jpet.111.186833

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Young, E. A., Dreumont, S. E., and Cunningham, C. L. (2013). Role of nucleus accumbens dopamine receptor subtypes in the learning and expression of alcohol-seeking behavior. Neurobiol. Learn. Mem. doi: 10.1016/j.nlm.2013.05.004. [Epub ahead of print]

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zimatkin, S. M., and Lindros, K. O. (1996). Distribution of catalase in rat brain: aminergic neurons as possible targets for ethanol effects. Alcohol Alcohol. 31, 167–174. doi: 10.1093/oxfordjournals.alcalc.a008128

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Zimatkin, S. M., Rout, U. K., Koivusalo, M., Bühler, R., and Lindros, K. O. (1992). Regional distribution of low-Km mitochondrial aldehyde dehydrogenase in the rat central nervous system. Alcohol. Clin. Exp. Res. 16, 1162–1167. doi: 10.1111/j.1530-0277.1992.tb00713.x

Pubmed Abstract | Pubmed Full Text | CrossRef Full Text

Keywords: ethanol, acetaldehyde, endogenous opioid system, salsolinol, behavior, animal

Citation: Font L, Luján MÁ and Pastor R (2013) Involvement of the endogenous opioid system in the psychopharmacological actions of ethanol: the role of acetaldehyde. Front. Behav. Neurosci. 7:93. doi:10.3389/fnbeh.2013.00093

Received: 03 May 2013; Accepted: 10 July 2013;
Published online: 31 July 2013.

Edited by:

Merce Correa, University Jaume I, Spain

Reviewed by:

Elio Acquas, University of Cagliari, Italy
María J. Sánchez-Catalán, Centre National de Recherche Scientifique (CNRS), Institute of Cellular and Integrative Neurosciences (INCI) UPR3212, France

Copyright © 2013 Font, Luján and Pastor. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Laura Font, Area de Psicobiología, Universitat Jaume I, Avda. Sos Baynat s/n, 12071, Castellón, Spain e-mail: laura.font@uji.es

Download