+ All Categories
Home > Documents > Neurotransmitter and neuromodulatory mechanisms involved in alcohol abuse and alcoholism

Neurotransmitter and neuromodulatory mechanisms involved in alcohol abuse and alcoholism

Date post: 27-Feb-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
32
~ Pergamon 0197-0186(94)00139-1 Neurochem. Int. Vol. 26, No. 4, pp. 305 336, 1995 Copyright © 1995 Elsevier Science Ltd Printed in Great Britain. All rights reserved 0197q)186/95 $29.00+0.00 INVITED REVIEW NEUROTRANSMITTER AND NEUROMODULATORY MECHANISMS INVOLVED IN ALCOHOL ABUSE AND ALCOHOLISM IGAL NEVO* and MICHEL HAMON INSERM U.288, Neurobiologie Cellulaire et Fonctionnelle, Facult6 de M6decine Piti6-Salp~tri~re, 91, boulevard de l'H6pital, 75634 Paris Cedex 13, France CRITIQUES by: R. D. MYERS, B. A. MCMILLEN and A. ADELL; and B. E. LEONARD Abstract--Acute or chronic consumption of alcohol interferes differentially with transmission processes in the CNS, affecting many--if not all--of the known neurotransmitter systems. Conversely, selective phar- macological manipulations of some of these neurotransmitter systems have been shown to reduce ethanol intake and preference as well as the severity of the ethanol withdrawal syndrome in animal models, certain compounds having even been employed successfully in the clinic. This review examines the studies which have attempted to elucidate the roles of these neurotransmitter systems in the mechanisms involved in the various aspects of alcohol abuse and alcoholism, with an emphasis on recent developments. The brain's major amino acid transmitter systems--inhibitory 7-aminobutyric acid (GABA) and excitatory glu- tamate-have been widely studied over the past decade, with the general consensus that acute ethanol facilitates GABAergic transmission (by enhancing chloride conductance through the GABAA receptor) and inhibits glutamatergic function (by decreasing cationic conductance through the NMDA receptor). Conversely, the development of tolerance associated with chronic ethanol consumption leads to a reduced GABAergic and increased glutamatergic function. Interactions between ethanol and the monoaminergic transmitter systems are complex. Dopaminergic and noradrenergic mechanisms, along with the endogenous opioid systems of the brain, seem to be implicated in the rewarding effects of ethanol via activation of positive reinforcement pathways, while the serotonergic system mediates negative reinforcement. A number of ligands of the dopaminergic, serotonergic and opioidergic receptors involved in ethanol consumption- related behaviors have been recognized for their effects in reducing ethanol preference and/or alleviating symptoms of the ethanol withdrawal syndrome in various animal models. Several of these substances are being used with success clinically. Studies of the central cholinergic system in alcoholics have provided clues to the mechanisms underlying the deleterious effects of ethanol on learning and memory, and evidence of a reduced central cholinergic activity has been reported in alcohol-dependent patients. Interestingly, acetylcholine-rich grafts and cholinomimetic drugs have been found to ameliorate ethanol-induced behavioral deficits in alcoholized rats. More generally, basic studies on alcohol's effects on central neuro- transmission certainly hold the key to the development of new strategies for the treatment of alcoholism. Ethanol is the second most commonly abused psycho- tropic drug after caffeine, and in most of the world today, alcoholism has emerged as a major social and health problem. The neurobiological consequences of ethanol abuse have been shown to be quite serious; prolonged exposure of rats to ethanol results in den- dritic hypertrophy and neuronal loss in various brain regions (Riley and Walker, 1978 ; Pierce et al., 1993), while chronic ethanol abuse in humans is associated with diffuse cortical and cerebellar atrophy, degener- ation of cerebellar Purkinje cells, and reduction in the size and number of neurons in the frontal cortex * Author to whom all correspondence should be addressed. 305
Transcript

~ Pergamon 0197-0186(94)00139-1

Neurochem. Int. Vol. 26, No. 4, pp. 305 336, 1995 Copyright © 1995 Elsevier Science Ltd

Printed in Great Britain. All rights reserved 0197q)186/95 $29.00+0.00

INVITED REVIEW

N E U R O T R A N S M I T T E R A N D N E U R O M O D U L A T O R Y M E C H A N I S M S INVOLVED IN ALCOHOL ABUSE A N D

ALCOHOLISM

IGAL NEVO* and MICHEL HAMON INSERM U.288, Neurobiologie Cellulaire et Fonctionnelle, Facult6 de M6decine Piti6-Salp~tri~re,

91, boulevard de l'H6pital, 75634 Paris Cedex 13, France

CRITIQUES

by: R. D. MYERS, B. A. MCMILLEN and A. ADELL; and B. E. LEONARD

Abstract--Acute or chronic consumption of alcohol interferes differentially with transmission processes in the CNS, affecting many-- if not all--of the known neurotransmitter systems. Conversely, selective phar- macological manipulations of some of these neurotransmitter systems have been shown to reduce ethanol intake and preference as well as the severity of the ethanol withdrawal syndrome in animal models, certain compounds having even been employed successfully in the clinic. This review examines the studies which have attempted to elucidate the roles of these neurotransmitter systems in the mechanisms involved in the various aspects of alcohol abuse and alcoholism, with an emphasis on recent developments. The brain's major amino acid transmitter systems--inhibitory 7-aminobutyric acid (GABA) and excitatory glu- t amate -have been widely studied over the past decade, with the general consensus that acute ethanol facilitates GABAergic transmission (by enhancing chloride conductance through the GABAA receptor) and inhibits glutamatergic function (by decreasing cationic conductance through the NMDA receptor). Conversely, the development of tolerance associated with chronic ethanol consumption leads to a reduced GABAergic and increased glutamatergic function. Interactions between ethanol and the monoaminergic transmitter systems are complex. Dopaminergic and noradrenergic mechanisms, along with the endogenous opioid systems of the brain, seem to be implicated in the rewarding effects of ethanol via activation of positive reinforcement pathways, while the serotonergic system mediates negative reinforcement. A number of ligands of the dopaminergic, serotonergic and opioidergic receptors involved in ethanol consumption- related behaviors have been recognized for their effects in reducing ethanol preference and/or alleviating symptoms of the ethanol withdrawal syndrome in various animal models. Several of these substances are being used with success clinically. Studies of the central cholinergic system in alcoholics have provided clues to the mechanisms underlying the deleterious effects of ethanol on learning and memory, and evidence of a reduced central cholinergic activity has been reported in alcohol-dependent patients. Interestingly, acetylcholine-rich grafts and cholinomimetic drugs have been found to ameliorate ethanol-induced behavioral deficits in alcoholized rats. More generally, basic studies on alcohol's effects on central neuro- transmission certainly hold the key to the development of new strategies for the treatment of alcoholism.

E thano l is the second mos t commonly abused psycho- tropic drug after caffeine, and in mos t of the world today, a lcohol ism has emerged as a major social and heal th problem. The neurobiological consequences of e thanol abuse have been shown to be quite serious;

pro longed exposure of rats to e thanol results in den- dritic hyper t rophy and neurona l loss in various bra in regions (Riley and Walker, 1978 ; Pierce et al., 1993), while chronic e thanol abuse in h u m a n s is associated with diffuse cortical and cerebellar a t rophy, degener- a t ion of cerebellar Purkinje cells, and reduct ion in the size and n u m b e r of neurons in the f ronta l cortex * Author to whom all correspondence should be addressed.

305

3()6 Invited Reviev,

(Charness ez al., 1989). It therefore follows that a great deal of effort has been devoted to the elucidation of the neurobiological mechanisms underlying the urge to consume alcohol.

There is convincing evidence that genetic factors can contribute to the predisposition to alcoholism, and that there are subtypes of alcoholics who are affected to different degrees by environmental and genetic factors (Cloninger, 1987). In this respect, the development of animal models in which rodents will voluntarily consume ethanol solutions over water in a free-choice situation has been of great use in the search for the neurobiological substrate(s) of ethanol preference and reinforcement. Such models can be divided into two principal categories: (1) animals induced to prefer ethanol over water in a free-choice situation subsequent to behavioral conditioning or pharmacological manipulations, and (2) genetically selected strains of animals bred for differences in etha- nol preference, initial sensitivity, or severity of with- drawal. An early study by Fuller (1964), which was confirmed in later investigations (Belknap et al.,

1993), of a number of inbred mouse strains found that C57BL/6J mice voluntarily consume more than l0 g/kg of ethanol per day, while mice of the DBA/2J strain strongly avoid ethanol, typically consuming 0.1-2 g/kg per day. Several rat strains exhibiting a preference for ethanol have been developed (see Table 1 ), including the UChB (high alcohol-consuming) and UChA (low alcohol-consuming) lines bred at the Uni-

versity of Chile (Mardones, 1960), the AA (alcohol- preferring) and ANA (alcohol-avoiding) lines bred in the laboratories of Alko Ltd, the Finnish State Alco- hol Company (Eriksson, 1968L the P (alcohol-pre- ferring) and NP (alcohol non-preferring) lines of the University of Indiana (Lumeng et al., 1977), the HAD (high alcohol-drinking) and LAD (low alcohol-drink- ing) lines, developed by the same group from a foun- dation stock of the genetically heterogeneous N/Nih rat (Lumeng et al., 1986), and the sP (Sardinian alco- hol-preferring) and sNP (Sardinian alcohol non-pre- ferring) lines developed at the University of Cagliari (Fadda et al., 1989). Alcohol-preferring rats from these strains will voluntarily consume sufficient quan- tities of 10-30% ethanol solutions (i.e. at the same ethanol concentrations as those found in many alcoholic beverages) to attain pharmacologically rel- evant blood alcohol concentrations. In addition, they win work to obtain ethanol, and will develop tolerance and physical dependence (Hunt, 1993). Rodent lines have also been selectively bred for an initial sensitivity to acute ethanol (Table 1 ). These include the AT (alco- hol tolerant) and ANT (alcohol non-tolerant) lines of rats outbred for differential ethanol-induced motor impairment on the tilting plane (Eriksson and Rusi, 1981 ), the HAS (high alcohol-sensitive) and LAS (low alcohol-sensitive) rat lines selected for ethanol- induced ataxia from a genetically heterogeneous N/Nih breeding stock (Palmer et al., 1987), the SS (short-sleep, ethanol-insensitive) and LS (long-sleep,

Table I. Rodent lines bred for differences in ethanol preference, initial sensitivity to acute ethanol, or severity of ethanol withdrawal syndrome (see text for details)

(a) I, ines hred /br d([l~'rence.s in ethum*l pr{j~,rence

Alcohol-preferring Alcohol non-preferring Species References

UChB UChA rat Mardones (1960) C57BL,'6J DBA/2J mouse Fuller (I 964)

AA ANA rat Eriksson (1968) P NP rat Lumeng et al. (19771

HAD LAD rat Lumeng et al. (1986) sP sNP rat Fadda et al. (1989)

(h) Lines bred/or d([lerences in initial sensitivity to acute ethanol

Low sensitivity High sensitivity

SS LS mouse McClearn and Kakihana (1973) AT ANT rat Eriksson and Rusi (1981)

LAS HAS rat Palmer et al. (1987) HOT COLD mouse Phillips et al. (1990)

(c) Lines bred./or d![]~,rences in severity o/ethanol withdrawal syndrome

Withdrawal-seizure Withdrawal-seizure resistant prone

WSR WSP mouse Crabbe et al. (1985)

Invited Review 307

ethanol-sensitive) lines of mice selected from a gen- etically heterogeneous stock (HS/Ibg) for differential sensitivity to the acute hypnotic effect of ethanol as determined by sleep time, i.e. the duration of loss of the righting reflex (McClearn and Kakihana, 1973), and the COLD (ethanol-sensitive) and HOT (ethanol- resistant) mouse lines genetically selected for their hypothermic response to acute ethanol (Phillips et al., 1990). Finally, two lines of mice have been genetically selected for resistance (withdrawal-seizure resistant, WSR) and susceptibility (withdrawal-seizure prone, WSP) to handling-induced convulsions during etha- nol withdrawal by Crabbe et al. (1985).

Prior to the early 1970s, the generally accepted view was that ethanol exerted its central effects via a non- specific fluidization of neuronal membranes (e.g. Seeman, 1972), since it was considered doubtful, given the simple chemical structure of ethanol, that a specific receptor for this molecule could exist. As the role of neurotransmitters and their receptors in the CNS began to be better understood, it became apparent that ethanol was in fact able to alter central neuro- transmission. It is now clear that there is no single neurotransmitter system which can be selected as being responsible for mediating all of the central effects of ethanol, and that ethanol in fact has many targets in the CNS. This review will provide an over- view of the numerous studies which have attempted to elucidate the role of the brain's neurotransmitter systems in mediating the reinforcing effects of ethanol and the neurobiological consequences of alcohol abuse.

AMINO ACID NEUROTRANSMITFERS

Since the early 1980s, many studies have con- centrated on defining a role for the brain's major inhibitory and excitatory amino acid neuro- transmitters, y-aminobutyric acid (GABA) and glu- tamate, respectively, in the central actions of ethanol.

Inhibitory Amino Acids

G amma-aminobutyric acid ( G A B A ) A great deal of effort has been devoted to examining

the relationship between ethanol's CNS actions and the GABAA receptor, a GABA-gated chloride channel which contains multiple binding sites. In addition to a site which binds the agonists GABA and muscimol and the antagonist bicuculline, there is also a binding site for benzodiazepine (BZD) agonists, antagonists and inverse agonists, a site which binds the chloride

channel antagonists picrotoxin and t-butyl- bicyclophosphorothionate (TBPS), and a recognition site for depressant/anticonvulsant barbiturates. Moreover, this receptor is composed of distinct pro- tein subunits, each of which has 1-6 variants (~j~,/~-3, YJ 3, 6 and p~ 2) which can be arranged in different combinations, producing a number of possible isoforms.

Behavioral studies. Behavioral studies have demon- strated the ability of GABA mimetic drugs to potent- iate ethanol's sedative and incoordinating effects in rodents, whereas GABA antagonists and inverse agonists have been shown to attenuate these effects (Allan and Harris, 1987a).

In addition, the BZD partial inverse agonist Ro 15- 4513 was shown to block the high-dose depressant effects of ethanol on spontaneous locomotor activity, without affecting its low-dose stimulatory effects (Becker, 1988), a finding which further supports a role for the GABAA receptor complex in mediating the central depressant effects of ethanol. Moreover, rod- ent strains selected for differences in ethanol sensitivity have been found to display differential sensitivities to BZD (Marley et al., 1988), and, reciprocally, mice selected for differential diazepam sensitivity display differences in ethanol effects (Gallaher and Gionet, 1988). As expected from the existence of a BZD- ethanol link, it has been shown that chronic ethanol administration confers cross-tolerance to BZD with respect to several different behavioral parameters (L6 et al., 1986).

Most data linking the GABAergic system to ethanol actions are concerned with the mediation of the direct CNS effects of ethanol. There are also several lines of evidence, however, which support a role for this neurotransmitter system in ethanol-induced positive reinforcement, by way of an enhancement of GABA- ergic function. Notably, an early study demonstrated the maintenance of alcohol preference during alcohol withdrawal with the BZD agonist diazepam (Deutsch and Walton, 1977). In more recent studies, the partial inverse agonist at the BZD receptor, Ro 15-4513, was found to reduce ethanol intake by several lines of rats, including an alcohol-preferring strain, without modifying other consummatory behaviors (e.g. McBride et al., 1988). The same effect was observed with the BZD inverse agonist Ro 19-4603 in another alcohol-preferring rat strain (Balakleevsky et al., 1990).

In addition to GABAA receptors, the guanine nucleotide-binding protein-coupled GABAB receptor might also participate in the GABAergic mediation of at least some of the central effects of ethanol. In

308 Invited Review

particular, Allan and Harris (1989) showed that the GABAB antagonist, phaclofen, blocked ethanol- induced hypothermia, motor incoordination, narcosis and locomotor stimulation in mice, while exhibiting no effects when administered alone. More recently, Humeniuk et al. (1993), using a number of GABA, agonists and antagonists, concluded that the GABAu receptor may indeed be involved in the locomotor stimulation induced by low doses of ethanol, and that it is an agonist rather than antagonist activity at this receptor which would lead to an attenuation of the effects of ethanol.

Biochemical and electrophysioloqical studies. Studies conducted in the 1970s showed that barbiturates and BZD produce many of their CNS effects by enhancing GABAergic inhibition by way of allosteric modu- lation of the GABAA receptor (see Harris et al., 1992). As initial studies demonstrated a certain similarity in the actions of BZD and ethanol, this led to the suggestion that these two classes of drugs may share a common mechanism of action (Frye eta[ . , 1980).

Nevertheless, studies performed in rodents sub- sequent to acute or chronic exposure to ethanol (pro- ducing brain ethanol concentrations corresponding to moderate-to-severe intoxication, i.e. up to 100 raM) have generally found little or no change in the binding of antagonists or agonists to the GABA site on GABAA receptors (Unwin and Taberner, 1980; Buck and Harris, 1990a,b). However, certain studies did note an increase in the density of low-affinity GABAA receptors in mouse and rat brain upon acute ethanol intoxication and a decrease when the rodents were subjected to chronic ethanol intoxication (e.g. Ticku and Burch, 1980). In a study comparing the AT and ANT rat lines, Korpi et al. (1992a) noted a lower binding of the GABA,~ agonist [~H]muscimol to GABAA receptors in the ANT rats.

Whereas tolerance to the effects of chronic ethanol administration on chloride movements develops, the sensitivity of the chloride channel to GABA is not reduced (Allan and Harris, 1987b). This would seem to suggest that a part of the GABAA receptor complex other than that which binds GABA may be affected ; indeed, Mhatre et al. (1988) noted an increase in the binding of the BZD partial inverse agonist [~H]Ro 15- 4513 in the cerebral cortex and cerebellum, but not in the hippocampus or striatum, of rats chronically intoxicated by intragastric intubation, although an- other group had previously t\mnd no change m BZD inverse agonist binding in any of the brain areas exam- ined after chronic ethanol intubation of rats (Tam- borska and Marangos, 1986). Subsequently, however. Mhatre and Ticku (1989) showed that the binding

o1" BZD inverse agonists following chronic ethanol exposure of cultured spinal cord neurons was increased for 24 h after removal of ethanol from the culture medium, with a reversal of the effect within 48 h, a time course corresponding to the appearance and disappearance of anxiety and seizures subsequent to ethanol withdrawal in vivo. Moreover, Korpi et al.

(1992a) found an enhanced affinity for [3H]Ro 15- 4513 in the alcohol-sensitive ANT rats as compared to their AT counterparts. In contrast, binding of BZD agonists and antagonists in rodents subjected to chronic ethanol intoxication has generally been found to be unchanged (e.g. Buck and Harris, 1990b). One group, however, noted a reduction in the density of [3H]flunitrazepam binding in the frontal cortex (Freund and Ballinger, 1988) and hippocampus (Freund and Ballinger, 1989) of alcoholics, with no changes in other areas examined. Conversely, an enhancement by ethanol of the binding of [3H]flu- mazenil (Ro 15-1788) in several mouse brain regions has been reported by Mosaddeghi et al. (1992).

In an in vitro study of the GABAA receptor com- plex's picrotoxin/convulsant site, high concentrations (EC~0 ~ 500 raM) of ethanol (lethal in vi~;o) were found to inhibit [35S]TBPS binding (Thyagarajan and Ticku, 1985),

In vitro studies of 3~'C1 uptake by isolated brain membrane vesicles and cultured mammalian brain cells have demonstrated a potentiation of GABA-acti- vated chloride flux by moderate concentrations ( < 100 raM) of ethanol (Suzdak et al., 1986a ; Aguayo, 1990), an effect which could be blocked by the GABA antag- onists picrotoxin and bicuculline as well as by the BZD partial inverse agonist Ro 15-4513 (Suzdak el al., 1986b). According to Hunt (1993), this effect could provide a molecular basis to account at least in part for the central depressant action of ethanol. Moreover, it would appear that GABA-mediated chloride movements are differentially affected by etha- nol in genetically selected rodent strains, as ethanol has a greater efficacy in promoting GABA-mediated chloride influx in mice selectively bred for high ethanol sensitivity than in their low ethanol-sensitive counter- parts (Allan et al., 1988); in high and low alcohol- sensitive rats, it is also the ethanol-insensitive line which displays resistance to ethanol enhancement of GABA-activated chloride influx (Harris and Allan, 1989).

Several groups have also examined the effects of ethanol on mRNA levels of the different GABAA receptor subunits. Montpied et al. (1991) noted that chronic ethanol treatment produces a decrease in the levels of ~t and ~_,, but not :<, subunit mRNA in the

Invited Review 309

rat cerebral cortex, an effect also found with acute ethanol administration (Morrow et al., 1989). In a study using alcoholized WSP and WSR mice, Buck et al. (1991) demonstrated that chronic ethanol treat- ment increases the content of y3 mRNA in both lines, and decreases the content of ~ mRNA in WSP but not WSR mice, and that of ~6 in WSR but not WSP mice. Conversely, Hirouchi et al. (1993) found that ~ subunit mRNA was significantly increased in mice exposed to continuous ethanol inhalation for more than 5 days (resulting in dependence upon ethanol), but returned to its normal level 8 h after termination of the ethanol inhalation. Studies on the encoded pro- teins themselves indicated a reduction in the levels of the :q, ~2 and ~3 subunits in the cerebral cortex and of the ~1 subunit in the cerebellum of chronically ethanol- intoxicated rats (Mhatre et al., 1993).

It should be noted that whereas a majority of studies have come out in favor of a modulation of GABA receptor sensitivity by ethanol, others do not agree with this view (Mancillas et al., 1986; White et al., 1990). Differences in the methods used for the prep- aration of the tissues, in the housing conditions, and in the method of sacrifice of the animals might account for these discrepancies. For instance, membrane ves- icles prepared from the cerebral cortex of individually housed rats have been shown to exhibit lower values of GABA-stimulated chloride influx than their pair- housed counterparts (Thielen et al., 1993). Regional differences in the subunit composition of GABAA receptors could also contribute to the variable results from one group to another, if sensitivity to ethanol depends on a specific ~f17 combination (Criswell et al., 1993). In fact, Wafford et al. (1991) had previously shown that the 72L variant (the ~2 subunit has two slice variants which vary by eight amino acids, the "long" (72L) and the "short" (72S) forms) was necess- ary for a potentiation of GABA A receptor function by ethanol and BZD. According to Criswell et al. (1993), ethanol would enhance GABA response in areas where type-l-BZD binding, of composition ~Jfl~Y2, is especially abundant : medial septum, inferior colliculus, substantia nigra reticulata, ventral pal- lidum and diagonal band of Broca, but not in areas where it is poorly represented : lateral septum, ventral tegmental area and hippocampus. Interestingly, most electrophysiological investigations concluding on a lack of effect of ethanol on GABAergic responses came from studies in the hippocampus (e.g. Mancillas et al., 1986).

To date, the situation regarding the effects of etha- nol on central GABAergic neurotransmission is there- fore far from being completely elucidated. It would

appear that acute ethanol, at high doses, generally facilitates GABAergic neurotransmission, whereas chronic ethanol would exert the opposite effect (see Fig. 1). Whether such long-term changes in GABA- ergic systems participate in the tolerance/dependence phenomena associated with chronic ethanol exposure is still a matter for debate.

Glycine (at strychnine-sensitive receptors)

Gonzalez (1993) recently showed that there was no difference between chronically alcoholized or ethanol- withdrawn rats and ethanol-naive controls in the sen- sitivity of these animals to seizures induced by the glycine antagonist strychnine, in spite of the increased sensitivity of similarly treated animals to seizures induced by the GABAA antagonist picrotoxin. He concluded that ethanol exposure and withdrawal may not significantly alter the function of glycinergic neurotransmission and that the ethanol withdrawal syndrome is likely to be indicative of alterations in specific neuronal mechanisms, rather than of a gen- eralized state of CNS hyperexcitability. However, Criswell et al. (1993) claim that there exist ethanol- sensitive and -insensitive responses in the brain for glycine (as for GABA and glutamate--see respective sections) ; there is some evidence that ethanol affects responses to glycine on a regionally specific basis, with an enhancement of responses in the spinal cord (Celantano et al., 1988), but no effect in the cortex of the cat (Nestoros, 1980) or in the inferior colliculus of the rat (Simson et al., 1991). Moreover, binding studies in the hippocampus of ethanol-fed mice found no change in the number ofglycine binding sites (Snell et al., 1991).

The sum of these results would seem to suggest that glycinergic synapses per se are not a major target for ethanol under acute or chronic conditions.

Exci tatory Amino Acids

Glutamate Evidence has accumulated over the past decade

that, in contrast to its potentiation of GABA inhibi- tory function, acute ethanol can antagonize the effects of glutamate, the other major brain amino acid neuro- transmitter, which is involved in the mediation of excitatory synaptic transmission. For instance, sys- temic injection of ethanol (2 g/kg) was shown to inhibit the excitation of noradrenergic locus coeruleus neurons as produced by microiontophoretic appli- cation of glutamate, N M D A or quisqualate, while the response of these neurons to similarly applied

31(I Invited Review

Ethanol

q picro

BA ;cimol

cuculline

agonists I . . . . . . ~ antagonists

inverse agonists

Fig. 1. Summary of the effects of acute and chronic exposure to ethanol (in unselected animal strains) on the interaction of various ligands with their respective binding sites on the GABAA receptor. • positive

effect, @ negative effect.

acetylcholine was unaffected by the ethanol treatment (Engberg and Hajos, 1992).

Glutamate receptors in the mammalian CNS are divided into three principal subtypes, based on ligand specificity : ionotropic e-amino-3-hydroxy-5-methyl- 4-isoxazole propionic acid (AMPA) 'kainate, metabo- tropic quisqualate, and ionotropic N-methyl-D-aspar- tate (NMDA) receptors. The NMDA receptor, which is coupled to a voltage-sensitive ton channel per- meable to calcium and monovalent cations such as sodium and potassium, has been implicated in the mechanisms involved in a number of physiological and pathological processes, ranging from the synaptic plasticity underlying learning and memory (Cotman et al., 1989) to the development of epileptiform

seizures (Dingledine et al., 1986) and neurotoxicity (Lovinger, 1993a).

Ethanol and N M D A receptors. Applied acutely at concentrations known to correspond to blood alcohol levels associated with moderate alcohol intake in humans (Wallgren and Barry, 1970), ethanol has been shown by electrophysiological techniques to potently and selectively inhibit the action of agonists at the NMDA receptor (Hoffman et al., 1989; Lovinger et

al.. 1989). In addition, in vivo microdialysis studies demonstrated a decrease in the extracellular con- centrations of glutamate in the striatum after adminis- tration of 2 g/kg ethanol to awake rats, as well as an inhibition by ethanol of the striatal release of glutamate induced by local application of NMDA

Invited Review 311

(Carboni et al., 1993). Using electrophysiological techniques, and employing selective antagonists of the non-NMDA glutamate receptor subtypes in order to isolate NMDA-mediated EPSPs, Lovinger et al. (1990) showed that ethanol-induced inhibition at the NMDA receptor concerns the agonist action of both the endogenous ligand, glutamate, and exogenous NMDA. These authors also presented evidence indi- cating that the potency of different alcohols for inhibiting the NMDA-activated current is linearly related to their potency for producing intoxication, suggesting that ethanol-induced inhibition of NMDA-mediated responses may contribute to etha- nol intoxication. In a recent study, Simson et al. (1993) found that ethanol, applied locally via electro- osmosis, potently inhibited NMDA-evoked neuronal activity in the inferior colliculus and hippocampus, but not in the lateral septum. They suggest that there may exist two types of N M D A receptor--one sen- sitive to ethanol and the other insensitive.

Through a process referred to as excitotoxicity, excessive neuronal stimulation by glutamate leads to degeneration of neurons and subsequently to neuronal death. While the activation of non-NMDA receptors (especially of the kainate subtype) is known to be able to contribute to neurotoxicity, it is the large increase in intracellular calcium provoked by NMDA receptor stimulation which is though to be a key event in ini- tiating the neurotoxic processes leading to delayed neuronal death (Chandler et al., 1993). Chronic etha- nol consumption has also been reported to cause neurotoxicity, with dendritic hypertrophy and neu- ronal loss in the hippocampus (dentate gyrus) and cerebellum of rats (Riley and Walker, 1978 ; Pierce et al., 1993), and with diffuse cortical and cerebellar atrophy, degeneration of cerebellar Purkinje cells, and selective reduction in the size and number of neurons in the frontal cortex in humans (Charness et al., 1989). However, as the mechanism for this neuronal cell loss is unknown, it has been postulated that a potentiation by chronically administered ethanol of endogenous glutamatergic function (Iorio et al., 1992) may account in part for alcohol's neurotoxicity. Indeed, whereas acute ethanol can block NMDA toxicity in neurons cultured from rat cerebral cortex (Lustig et al., 1992), recent studies have shown that chronic exposure of rat cerebrocortical cultures to ethanol is able to enhance the neurotoxic potency of NMDA (Chandler et al., 1993 ;Ahern et al., 1994).

Glycine modulatory site (strychnine-insensitive). Although it is generally accepted that ethanol can inhibit NMDA receptor function, there is little evi- dence as to the mechanism by which this inhibition is

produced. Woodward and Gonzales (1990) suggested that ethanol may be acting at a strychnine-insensitive glycine binding site, where an interaction between N M D A or glutamate and glycine may be necessary for NMDA receptor-mediated effects (Forsythe et al., 1988), to produce its inhibition of NMDA-mediated processes ; this suggestion was based on the ability of exogenously added glycine to completely reverse the inhibitory effects of ethanol on NMDA-stimulated endogenous dopamine release from rat striatal slices (Woodward and Gonzales, 1990). Other studies added their support to this hypothesis by demon- strating the ability of glycine or glycine agonists to reverse the inhibitory effects of ethanol on NMDA- stimulated increases in intracellular calcium in rat neurons (Rabe and Tabakoff, 1990). According to Peoples and Weight (1992), glycine and ethanol (50 mM) interact in a non-competitive manner in inhibit- ing NMDA-activated currents. This view is supported by a recent study in which Woodward (1994), using a selective competitive glycine antagonist, showed that ethanol did not significantly alter the glycine antag- onist's inhibitory potency, which would be expected if the two compounds were competing for the glycine site.

Ethanol and n o n - N M D A receptors. As agonists of the AMPA/kainate receptor subtype--also coupled to an ion channel--produce neurotoxic effects as well, it was of interest to examine whether ethanol exerts the same actions on this subtype as on the N M D A receptors. In an electrophysiological study of the effect of ethanol on kainate, AMPA and N M D A receptor- operated channels, Dildy-Mayfield and Harris (1992), using Xenopus oocytes expressing mRNA from rat hippocampus and cerebellum, demonstrated a similar degree of inhibition by ethanol of NMDA and non- NMDA ion channel-coupled receptors; in certain regions, the ethanol sensitivity of some non-NMDA receptors was even found to surpass that of the N M D A subtypes. Other studies, however, failed to find a difference in the electrophysiological respon- siveness to AMPA and kainate of neurons in hippo- campal slices from rats exposed to ethanol in utero as compared with controls (Martin et al., 1992). Fur- thermore, in a study of recombinant AMPA-type glu- tamate receptors expressed in mammalian cells, Lovinger (1993b) showed that inhibition of kainate- activated current could be observed, but only in the presence of intoxicating ethanol concentrations ; how- ever, the potency of ethanol for inhibiting current in cortical neurons was lower than that for inhibiting the function of the recombinant receptors. Nevertheless, as the concentrations of ethanol which affect recom-

312 Invited Review

binant AMPA receptors are within the range of con- centrations found during moderate to severe intoxication, the author suggests that if receptors with such sensitivity are present in vivo, they may con- tribute to intoxication ; this speculation is supported by a report suggesting that #7 vivo ethanol adminis- tration inhibits neuronal firing stimulated by the AMPA receptor agonist quisqualate in the locus coeruleus (Engberg and Hajos, 1992). In a study of metabotropic glutamate receptor function in offspring of prenatally ethanol-exposed rats, Queen et al. (1993) found a reduction in metabotropic glutamate recep- tor-activated phosphoinositide hydrolysis when a 5% (but not 3.35%) ethanol solution was given to the pregnant dams. As metabotropic receptor activation has been implicated in the facilitation of NMDA receptor-dependent long-term potentiation (LTP), the authors argue that the effect of higher blood ethanol concentrations, which will also affect the metabo- tropic receptor subtype, may lead to an even greater impact of prenatal ethanol exposure on LTP than occurs when NMDA receptor function alone is affec- ted by maternal consumption of more moderate quan- tities of ethanol.

Receptor hhldin9 studies. Studies on tile effects of ethanol on the receptor binding sites for excitatory amino acids have produced contradictory results (a general summary of the actions of ethanol on the NMDA receptor is provided in Fig. 2). Whilst an early study had shown that acute and chronic adminis- tration of small to moderate doses of ethanol increased [~H]glutamate binding to rat brain synaptic membranes (Michaelis et a/., 1978). Savage et al.

(1992) reported a reduction in the density of NMDA- sensitive [~H]glutamate binding sites in the hippo- campal formation of 45-day old rat offspring who had been exposed to alcohol throughout the gestational period, although they did not observe this reduction in other brain regions containing relatively high den- sities of NMDA receptors. In an #l vitro binding assay using rat cortical membranes, De Montis et ul. (1991) showed that low concentrations (0.1 10 I*M) of etha- nol increased the affinity of [3H]MK-801 binding, without modifying the B~,~. while higher con- centrations (1 100 raM) failed to modify [3H]MK-801 binding, and even higher concentrations (200 mM) actually caused a negative modulation of the binding of the radioligand. On the other hand, chronic exposure of rodents to ethanol leads to an increase in the number of [3H]MK-801 binding sites in certain brain areas (Snell et al., 1993), reflecting an up-regu- lation of NMDA receptors ; this more closely reflects what one would expect to find upon chronic ethanol

intoxication, given the inhibition of NMDA receptor- mediated activity produced by acute ethanol. More- over, a recent study by Trevisan et al. (1994) demon- strated the ability of chronic ethanol exposure to dramatically up-regulate NMDA receptor subunit 1 protein ~an essential component of the NMDA receptor--in the hippocampus but not in the nucleus accumbens, cerebral cortex, or striatum of the rat. In contrast, the ethanol treatment did not modify the levels of receptor subunits which make up the AMPA- type receptor, nor did chronic treatment with other psychotropic drugs mimic the effects of ethanol on the levels of NMDA receptor subunit 1 (Trevisan et al.,

1994). Behavioral studies. In behavioral studies, support

can be found for the idea that ethanol may function as a selective NMDA receptor antagonist at low con- centrations. Indeed, in pigeons and mice (Grant et al.,

1991) and in rats (Grant and Colombo, 1992) trained to discriminate ethanol from vehicle, the non- competitive NMDA antagonists dizocilpine (MK- 801) and phencyclidine (PCP) substituted fully for ethanol, and drug discrimination procedures in rats have shown that ethanol is able to act as an antagonist of NMDA discrimination and to produce dis- criminative stimulus effects similar to those of com- petitive and noncompetitive NMDA antagonists (Balster et al., 1992). In addition, ethanol was found to be able to attenuate NMDA-mediated thermal hyperalgesia produced both after acute intrathecal NMDA administration and in a model of neuropathic pain (Meller et al., 1993), Reciprocally, NMDA recep- tor antagonists are able to modulate alcohol pref- erence (Lamblin et al., 1993) and to prevent the effects associated with the ethanol withdrawal syndrome (Liljequist, 1991). In line with this last finding, a greater number ofhippocampal MK-801 binding sites has been found in WSP mice than in the WSR line (Valverius et al., 1990).

Several laboratories have reported evidence tbr the involvement of the NMDA receptor in forms of syn- aptic plasticity such as LTP (Collingridge et al., 1983 ; Harris et al., 1984), a base for memory formation (Teyler and DiScenna, 1984). Thus, the inhibition by ethanol of the initiation of LTP in the hippocampus (Morrisett and Swartzwelder, 1993) is consistent with the negative influence of ethanol on NMDA-mediated synaptic transmission (see above). The possible link between LTP and memory formation might explain why ethanol, after acute (Mello, 1972), chronic (Walker and Hunter, 1978) or prenatal (Streissguth et al., 1990) exposure, exerts deleterious effects on learning and retention.

Invited Review 313

Acute Ethanol

glycine D-serine

o 6 6 5,7-dichlorokynurenate )-AP5

2PP

. . . . . . ~ agonists antagonists I

Fig. 2. Effects of acute and chronic exposure to ethanol (in unselected animal strains) on the interaction of various ligands with their respective binding sites on the glutamatergic NMDA receptor. *Low con- centrations only. HA966 = (RS)-3-amino-l-hydroxypyrrolidin-2-one; MK-801 = dizocilpine; PCP = phencyclidine; D-AP5 =D(-)-2-amino-5-phosphonopentanoic acid; CPP= 3-((RS)-2-carboxy-piper-

azin-4-yl)-propyl-1-phosphonic acid. ~ positive effect, ~ negative effect.

DOPAMINE The existence of a dopaminergic substrate for the

facilitation of brain reward mechanisms became apparent from early studies showing that antagonism of the dopaminergic system could attenuate brain stimulation reward and block the reinforcing effects of cocaine and amphetamine (Kelly et al., 1975). Moreover, the inhibition of dopamine (DA) uptake by both of these drugs and the release of DA by amphetamine, having as an effect the potentiation of the actions of DA postsynatpically and, behaviorally, the facilitation of intracranial self-stimulation (Stein, 1964), strengthen the case for an implication of the

dopaminergic system in the abuse potential of a drug and brain reward (Wise and Bozarth, 1987). A brain microdialysis study examining the differential involve- ment of the anatomically and functionally distinct subdivisions of the dopaminergic system has further shown that drugs abused by humans stimulate DA release from the major terminal area of the mesolimbic DA system (the nucleus accumbens) to a far greater degree than from the dorsal caudate nucleus, the major terminal area of the nigrostriatal DA pathway, while drugs with aversive properties reduced DA release in these two areas (Di Chiara and Imperato, 1988).

Numerous studies provide evidence that alcohol-

314 Invited Review

preferring rats consume ethanol for its central rein- forcing actions. Indeed, it has been shown that P rats will voluntarily self-administer ethanol intra- gastrically (Waller et al., 1984) or intracerebrally (Gatto et al.. 1990). Interestingly, lower con- centrations of DA and its metabolites have been found in the nucleus accumbens of P (Murphy et al., 1987) and HAD (Gongwer et al., 1989) rats than in their NP and LAD counterparts. It therefore follows that much effort has been devoted to determining whether DA also underlies ethanol preference.

In 1974, Seeman and Lee showed that high brain ethanol concentrations could release DA in the stri- atum, although low to moderate concentrations were found to have no effect. Later studies, however, using more moderate ( 1 3 g/kg) doses of ethanol, were able to demonstrate an increase in DA turnover and release (Imperato and Di Chiara, 1986) in various brain areas during ethanol intoxication, and conversely, a dra- matic decrease in DA release during ethanol with- drawal in the nucleus accumbens (Diana et al., 1993) and striatum (Rossetti et al., 1992a), which could be reversed by a challenge ethanol dose. Electro- physiological studies demonstrated the ability of low doses of acute ethanol to increase the firing rate of DA neurons in the substantia nigra (Mereu et al.,

1984) and ventral tegmental area (VTA) in rivo (Gessa et al., 1985) and in vitro (Brodie e t a [ . . 1990). Conversely, the number of spontaneously active DA neurons in the VTA (Shen and Chiodo. 1993) and their firing rates (Diana et al., 1993) are reduced dur- ing acute ethanol withdrawal, both of which changes can be reversed by ethanol administration (Diana el a/.. 1993). Diana et al. (1993) suggested that the depressed activity of the mesolimbic DA system dur- ing the ethanol withdrawal syndrome may be relevant to the dysphoric state associated with ethanol with- drawal in humans.

Receptor binding studies have been conducted m unselected animal lines exposed to ethanol and tbl- lowing ethanol withdrawal, as well as in selected alco- hol-preferring and non-preferring rat lines. In an initial study using the AA and ANA rat lines, Korpi et al. (1987) found a modest reduction in D~ receptor number in the caudate nucleus of AA rats, but con- cluded that the difference observed between the two lines was too small to be considered of importance for genetically determined differences in alcohol pref- erence. A later study by Stefanini el a/. (1992) in sP rats confirmed the modest decrease observed by Korpi et al. (1987) in the caudate, but also noted decreases in limbic areas, notably in the nucleus accumbens and olfactory tubercle ; they found these differences to be

due to a decrease in B ...... with no change in Kd. In the light of these results, they claimed that a link probably exists between ethanol preference and the reduction in D: receptor number in these limbic areas, as D: receptors in the accumbens and olfactory tubercle are the first target of DA released by mesolimbic DA nerve terminals. Recently, McBride et al. (1993a) pro- vided further confirmation for a decrease in the den- sity of D 2 receptors in P rats ; their autoradiographical study showed a reduction in D: receptor labeling in the caudate-putamen, nucleus accumbens and VTA of P rats. and binding assays using membrane prep- arations indicated that this difference in the caudate- putameu was due to a decrease in B ....... with no change in the affinity of the receptors. A study by De Montis et al. (1993) revealed that sP rats also present modi- lications in D~ receptor binding ; while no change was detected in the caudate-putamen, the limbic areas of the sP rats presented a lower B ...... of D~ sites, with no change in Kd. Moreover, the sP rats exhibited a less pronounced responsiveness than their sNP counter- parts to DA-stimulated adenylate cyclase activity.

Support for a reduced D2 receptor function in alcoholism was provided by a clinical study of the growth hormone response to the DA agonist apo- morphine a well-accepted measure of postsynaptic D~ receptor function (Nair et al.. 1982). While early studies had demonstrated an increase in postsynaptic [), receptor function during the early alcohol with- drawal period in alcoholic patients (Anunziato et al.,

1983), Balldin et al. (1993) showed that long-term abstinent alcoholic subjects had a significantly lower maximal growth hormone response to apomorphine than control subjects.

Studies using unselected rat lines have produced less easily interpretable results than those obtained with alcohol-preferring strains. Certain studies found binding (B ...... but not Kd) to Dt (Hruska, 1988: Log- rano et al., 1993) and D~ (Hruska, 1988) receptors to be increased following chronic ethanol treatment, but Lucchi et al. (1988) found a decrease in D~ binding and Fuchs et al. (1987) reported that ethanol can reverse the up-regulation of D~ receptors due to chronic haloperidol treatment. Hamdi and Prasad (1992) suggested that in rats, chronic ethanol intake may cause a time-dependent bidirectional change in striatal D~ receptors, as they found an initial decrease in B ..... (with no change in Ka) of these receptors during the first week of consumption of an ethanol liquid diet, followed by a reversal of this effect and an increased B .... during the second week with this regimen. The latter change was still present during the third week. although to a lesser extent, and by the

Invited Review 315

fourth week, there was once again a significantly lower density of D~ receptors ; Bmax values stabilized to con- trol levels from the sixth week on, in spite of con- tinuous ethanol liquid diet.

An autoradiographical study by Rommelspacher et

al. (1992) concluded that there was a decrease in the number (but not the affinity) of D2 receptor binding sites in the caudate nucleus, nucleus accumbens and olfactory tubercle of unselected rats 24 h after alcohol withdrawal. However, May (1992) failed to confirm this observation and found no alteration in the Bmax or Kd values of striatal D~ or D2 receptors in ethanol- withdrawn rats. Nevertheless, this author noted that the high affinity (G-protein-coupled) state of the Dt receptor had a 5-fold increase in affinity for DA in ethanol-withdrawn rats as compared to control ani- mals (May, 1992).

A great deal of attention has also been devoted to behavioral studies examining the effects of various manipulations of dopaminergic neurotransmission on ethanol intake (see Table 2). An early study by Myers and Melchior (1975) suggested a potential role for DA in the mediation of ethanol drinking behavior by showing that intracerebroventricular injection of the DA (and noradrenergic) neurotoxin 6-hydroxy- dopamine (6-OHDA) resulted in a decreased pref- erence for ethanol in the rat, a result which was replicated by several other groups (e.g. Brown and Amit, 1977). Kiianmaa et al. (1975), however, found that 6-OHDA lesions of the mesencephalic tegmental area enhanced ethanol intake. Consistent with the data reported by Ld et al. (1981 a), it is possible that the mechanism underlying this enhancement of ethanol intake is by an impairment of the mechanism leading to ethanol tolerance. A more recent study also man- aged to induce significant increases in ethanol pref- erence by specific 6-OHDA lesions of the nucleus

accumbens and olfactory tubercle--two areas of the limbic system implicated in DA-mediated reward pathways--but not of adjacent brain areas in the rat (Quarfordt et al., 1991). The authors proposed that the resulting degeneration of DA-containing fibers and terminals causes a reversal of the rat's recognized aversion to ethanol by blunting the drug's aversive qualities. In contrast, Rassnick et al. (1993a) found no alteration in ethanol self-administration in trained rats by 6-OHDA lesions of the nucleus accumbens and olfactory tubercle, concluding that while the mesolimbic DA system may contribute to the rein- forcing actions of ethanol, it is not critical for main- taining ethanol reinforcement. These apparently contradictory results may derive from the different animal strains (Sprague Dawley in the study by Quar- fordt et al. (1991) and Wistar in that of Rassnick et

al. (1993a)) and experimental paradigms (free-choice between water and different concentrations (3-30%) of ethanol in the study by Quarfordt et al. (1991), and self-administration of water or a 10% ethanol solution by lever-pressing in that of Rassnick et al. (1993a)) employed in the two studies.

Despite early reports that treatments with the DA receptor antagonists haloperidol (Davis et al., 1978) or pimozide (Brown et al., 1982) were ineffective in altering ethanol consummatory behavior, thereby suggesting that the DA system was apparently not involved in the reinforcing effects of ethanol (see review by Deitrich, 1976), later findings have been more favorable toward a role for DA in the mech- anism underlying ethanol preference. Notably, in a double-blind clinical trial with 50 chronic alcoholics, the rated craving for alcohol was reduced from strong to very mild by administration of the DA agonist bromocriptine (Borg, 1983). In addition, a positive correlation seems to exist between DA activity and

Table 2. Effects of pharmacological manipulations modifying the CNS activity of monoaminergic systems on alcohol consumption in unselected rat lines (or clinical trials ; see text for details)

Monoamine CNS Alcohol System Activity Drinking References

7 7 Hodge et al. (1992) /~ ~ Borg (1983) ; Rassnick et al. (1993) ; Lan~a (1994)

DA "~ ,7 Kiianmaa et al, (1975) ; L~ et al. (1981a) ; Quarfordt et al. (1991) x, x, Myers and Melchior (1975) ; Samson et al. (1993) ; Hodge et al. (1993) N Davis et al. (1978) ; Brown et al. (1982) ; Rassnick et al. (1993a)

NE 7 ~ Docherty et al. (1993) "~ "~ Brown and Amit (1977) ; Davis et al. (1978) ; Amit and Brown (1982)

7 ~ Myers et al. (1972) ; Krasner et al. (1975) ; Naranjo et al. (1986) ; Bruno (1989) ; Gill and 5-HT Amit (1989) ; Lu et al. (1993)

"x, Myers and Veale (1968) ; Schreiber et al. (1993)

7 increase ; "~ decrease ; no change.

316 Invited Review

ethanol-reinforced responding, as microinjections of the non-specific DA agonist d-amphetamine and the D2/D3 agonist quinpirole into the nucleus accumbens increase ethanol-reinforced lever pressing (Hodge et

al., 1992), while responding is decreased when the D, antagonist raclopride is injected into this area (Sam- son et al., 1993) or upon i.p. injection of the mixed D~/D 2 agonist SDZ-205,152 (Rassnick e ta / . , 1993b). Consistent with the idea that reductions in responding lbllowing antagonist injection into the nucleus accum- bens may be due to decreased DA activity, the injec- tion of quinpirole into the VTA, which decreases cell firing rates, presumably by activating somatodendritic DA autoreceptors (Chiodo, 1988), also resulted in a decrease in ethanol-reinforced responding (Hodge et

al_ 1993). In a study using P rats, Weiss et al. (1990) showed

that i.p. injection of these rats with bromocriptine produced a shift in preference from ethanol toward water by inhibiting responding for ethanol while enhancing water consumption. This finding was con- firmed by Dyr et al. (1993) in HAD rats, with the D2/D3 agonist quinpirole causing a decrease in alcohol drinking with no concomitant decrease in the intake of food or a palatable saccharin solution in these animals. However, the D: antagonist, spiperone, had a differential effect on alcohol intake according to the dose administered, with no effect at the lowest dose, an increase at the intermediate dose, and a decrease at the highest dose administered. An increase in ethanol consumption with intermediate doses of another Dr antagonist, sulpiride, microinjected directly into the nucleus accumbens, was also reported in P rats (Levy et al., 1991). The interpretation proposed for this phenomenon was that an intermediate dose of these antagonists may cause a partial blockade of D 2 recep- tors, the effect of which would logically be a reduction in the reinforcing efficacy of ethanol. This would lead the P rats to increase their already high ethanol intake in an attempt to overcome the reduced reinforcing efficacy of ethanol. On the other hand, the complete blockade of D: receptors produced by higher doses would completely eliminate the reinforcing effects of ethanol, thus producing a decrease in alcohol con- sumption. A decrease in alcohol intake has also been shown to occur in rats systemically treated with high doses of the D2 antagonists pimozide and haloperidol (Pfeffer and Samson, 1988).

Very recently, Langa (1994) found a 40 50% reduction in voluntary ethanol intake by rats which had received intrastriatal transplantation of fetal dopaminergic grafts from the ventral mesencephalon, but not in those which were sham-operated or in which

the transplanted grafts were devoid of DA cells. The author concluded that a reduction in chronic alcohol intake may result from an increase in tonus of the dopaminergic mesolimbic system.

The past few years have seen the development of a controversy over the possible genetic implication of the D2 receptor gene in alcoholism. A first report by Blum et al. (1990) found a correlation between alcoholism and the A1 allele of the Taq 1 restriction fragment length polymorphism (RFLP) in the Y region of the D2 receptor gene. This conclusion was subsequently confirmed in a number of studies (for review, see Noble, 1993). In contrast, a number of other studies disagreed with this association (Bolos et

al., 1990; Gelernter et al., 1993). In support of the original findings of Blum et al. (1990), a comp- lementary receptor binding study by the same group (Noble et al., 1991)--using the same sample of brain tissues from deceased alcoholics and nonalcoholics as had been employed for the first study--found that individuals with the AI allele had less D~ receptor binding sites (lower Bm~) in the caudate nuclei than those without the allele ; in fact, a progressive decrease in D_~ receptor binding site density was observed with A2/A2, A1/A2 and A1/A1 genotypes, respectively. Moreover, in a more recent study of a new RFLP located closer to the regulatory regions of the D2 receptor gene, Blum et al. (1993) found an increased prevalence of the 5' Taq l BI allele in alcoholics as compared to nonalcoholics or less severe alcoholics, with no difference between less severe alcoholics and nonalcoholics ; the authors concluded that both the 5' and 3' regions of the D2 receptor gene are associated with severe alcoholism, suggesting that this gene may have an etiological role in some severe alcoholics. Detractors of this theory, however, claim that the reported positive associations between alcoholism and the A I polymorphism result from an inappropriate selection of controls, as alcoholics were not excluded in some instances, and in other studies, the sample of alcoholics did not distinguish between different degrees of severity of the disease (Noble and Blum, 1991). Moreover, Barr and Kidd (1993) showed that ethnic makeup can generate significant population differences in allele frequencies of the D2 receptor gene, varying from prevalences of as low as 0.09 for Yemenite Jews to as high as 0.75 for the Muskoke Amerindians. Thus, due to the significant het- erogeneity among both the alcoholic and control groups used in the studies to date, Gelernter et al. (1993) undertook a reanalysis of the data presented in the literature as a whole, in which the samples with the highest and lowest A1 allele frequencies were

Invited Review 317

removed. The conclusion of this analysis was that the difference between alcoholics and controls was not statistically significant.

In a recent development, a common study was undertaken by many of the authors who had fueled the debate over the putative existence of an association between alcoholism and the Taq I A1 RFLP (Gejman et al., 1994). In this examination of the abnormalities of the D2 receptor gene coding sequences possibly associated with schizophrenia or alcoholism, it was revealed that, although there exist 3 infrequent DNA variants that predict altered amino acid sequence of the receptor, none of these is associated with either alcoholism or schizophrenia, and there are thus no structural coding abnormalities in the D2 receptor gene in alcoholism or schizophrenia. It should be noted that the results of this study do not exclude the existence of a genetic defect in the O2 receptor gene, which could be located in a part of the genomic region not examined, such as exon 1 (a noncoding exon), introns, promoter (located at least 250 kb from the site of the Taq I A RFLP), or other regulatory sequences. However, as explained by Gejman et al. (1994), a valid, etiologically significant disease associated with an RFLP that is not in the coding or regulatory region of a known gene implies that there exists a mutation in that gene within a small distance of the RFLP; thus, the observed absence of a structural mutation in the gene renders interpretations of the association reported by many authors rather difficult.

Another receptor thought to play an important role in the reward process of addiction behavior is the D3 receptor, which is specifically located in the limbic areas of the brain, notably in the nucleus accumbens. However, a recent investigation on three independent brain tissue samples from the groups of Blum, Noble and Geijer (in which a positive association between the D2 receptor gene and alcohol dependence had previously been reported) indicates that there was no difference in the frequencies of the alleles of the Bal I RFLP of the D3 receptor gene between alcoholic and control subjects (P. Gorwood, personal com- munication). This suggests that there is no association between a given polymorphism of the D3 receptor gene and alcoholism.

NOREPINEPHRINE

The involvement of norepinephrine (NE) in alcoholism has also been widely examined. A number of early studies implicated noradrenergic systems in the mediation of ethanol self-administration, as neurochemical lesions of noradrenergic neurons

(Brown and Amit, 1977) or inhibition of NE synthesis (Davis et aL, 1978) were found to result in a sup- pression of voluntary ethanol consumption (see Table 2). Despite the suggestion that DA may be the sub- strate of all reward (Wise, 1980), but congruent with a number of early studies which had observed increases in NE turnover following the administration of ethanol (e.g Corrodi et aL, 1966), Amit and Brown (1982) therefore suggested that ethanol reinforcement may be an exception, in that the reward mechanism through which it acts appears to be subserved by nor- adrenergic rather than dopaminergic mechanisms.

However, the NE and DA systems should not necessarily be completely dissociated from one another, with respect to their involvement in the mediation of ethanol reinforcement, as evidence for the participation of dopaminergic mechanisms in this phenomenon has repeatedly been reported in the literature (see above). In this context, Wise and Bozarth (1985) proposed that while DA is likely to be the principal transmitter responsible for the central mediation of ethanol reward, ethanol reinforcement may occur via inhibition of noradrenergic activity in the locus coeruleus, the effect of which would be a release of DA neurons from tonic noradrenergic inhi- bition.

The effects of chronic or acute ethanol treatment on the noradrenergic system have also been examined. Early studies showed that single-dose ethanol administration stimulates the synthesis and turnover of cerebral NE without modifying its concentrations (e.g. Carlsson and Lindqvist, 1973). However, rat pups exposed to ethanol pre- or postnatally were found to have lower whole brain and hypothalamic levels of NE (Detering et al., 1980). A more recent study using the technique of in vivo brain microdialysis found that long-term alcohol treatment of non-selec- ted rats resulted in an increased release of the cat- echolamine in the hippocampus (Huttunen, 1991), and a chronoamperometric study in the cerebellar cor- tex of unselected rats found that ethanol, applied locally, inhibited NE reuptake (Lin et al., 1993). Upon acute ethanol administration to unselected rats, Ros- setti et al. (1992b) found a biphasic effect on NE release in the frontal cortex, as a low dose (0.2 g/kg) raised NE outflow, while a higher dose (2 g/kg) inhibited the release of NE ; the authors suggest that the decrease in cortical NE output may reflect the sedative-hypnotic properties of ethanol at high doses, whereas the increased NE release may represent a biochemical correlative of the arousal and increased alertness elicited by low doses of ethanol.

Studies with genetically selected lines of rodents

318 Invited Review

have shown differential cerebral noradrenergic activi- ties. Hetlevuo et al. (1990) demonstrated that the basal utilization rate of NE was similar between the AT and ANT rat lines, although the increased formation of the NE metabolite MHPG by the AT rats suggested that these rats had a higher noradrenergic activity in the limbic forebrain, hypothalamus and cerebellum. In addition, higher NE levels in the midbrain of LS mice than in their SS counterparts were reported by Sbafik et al. (1991).

In man, cerebrospinal fluid (CSF) levels of the NE metabolite MOPEG (which are considered to reflect central noradrenergic function) are increased fol- lowing absorption of ethanol (Borg el al., 1983). A genetic component in alcoholism-related nor- adrenergic transmission has also been raised, as alco- hol-dependent subjects have higher MOPEG levels than control subjects, while non-intoxicated subjects who have a family history of alcoholism have lower inherent MOPEG levels (Borg et al. 1983). A later study by Liljeberg et al. (1987) showed that CSF MOPEG levels are negatively correlated with the degree of alcohol craving, as well as with signs of euphoria and various intellectual capacities.

Studies of the effects of ethanol on central adre- nergic receptors have produced somewhat equivocal findings. In a study of 17-day old offspring exposed to ethanol in u tero , Wigal et al. (1990) noted a reduction in the number (Bm~,x) of /~-adrenoceptor binding sites in the hippocampus with no change in Kd, while Hadjivanova et al. (1991) found an increase in the affinity (decrease in Kd) of these receptors, with no change in their B ...... in the hippocampus of 8-week old rats perinatally exposed to ethanol. On the other hand, Rommelspacher et al. (1989) found that ethanol prevented a decrease in the number of//-adrenoceptor binding sites in the cerebral cortex induced by chronic treatment of rats with the tricyclic antidepressant desi- pramine. However, Wong et al. (1988) failed to observe any significant difference in binding to ~-, ~-2- and fi-adrenergic receptors in cerebral cortical and hippocampal membranes between P and NP rats, and Pandey et al. (1992) did not find any change in ~- adrenoceptor binding in the cerebral cortex of un- selected rats after short- and long-term ethanol administration. In a study of postmortem brains of ethanol-intoxicated nonalcoholic subjects and matched controls, Meana et al. (1992) showed that acute ethanol intoxication did not produce any changes in ~2-adrenoceptor binding. In contrast, Hu et al.

(1993) observed an increase in the binding of the ~2- adrenoceptor antagonist [3H]rauwolscine to the neu- ronal cell line NG108-15 after long-term (2 days)

treatment with 100 mM ethanol, as well as a large increase in the cell levels of ~2H- and ~2c-adrenoceptor mRNAs, leading the authors to suggest that clinically attainable concentrations of ethanol can regulate ~- adrenoceptor gene expression within the time frame of a single drinking session.

A limited number of investigations have also employed specific ligands of the adrenergic receptors in order to study the involvement of the noradrenergic system in the central effects of alcohol. In these studies, ~_~-adrenoceptor antagonists of different chemical classes were shown to be able to reverse the ataxic (Durcan et al., 1991, 1992) and hypothermic (Durcan et al., 1991) effects of ethanol in mice. In another study, Docherty et al. (1993) induced a reduction in the rats' intake of ethanol in a limited access procedure by administration of the fl-adrenergic agonist iso- proterenol via metered aerosol inhalation just before ethanol availability.

SEROTONIN

Since Myers and Veale first implicated an alteration of the cerebral metabolism of serotonin (5-hydroxy- tryptamine: 5-HT) in alcohol-related consummatory behavior in 1968, the indoleamine has increasingly become the focus of attention of investigators striving to determine the nature of the biochemical basis of the preference for ethanol. Myers and Veale (1968) noted a reduction in the rats' preference for a solution of ethanol in a wide range of concentrations upon oral administration of p-chlorophenylalanine (p-CPA), a tryptophan hydroxylase inhibitor which lowers 5-HT content in the brain without significantly altering levels of other neurotransmitters. Such a reduction in etha- nol preference by depleters of cerebral 5-HT has since been confirmed by numerous other authors, both in normal outbred and alcohol-preferring rat strains (Frey et al., 1970 ; Schreiber et al., 1993), as well as in clinical studies in humans (Naranjo e t al. , 1986). In 1975, Frankel et al. provided the first evidence that serotonergic neurotransmission was involved in the development of tolerance to the effects of ethanol, by demonstrating an impairment of the acquisition of tolerance by p - C P A administration. The same group (L~ et al., 1981b) subsequently showed that a similar effect could be obtained by degeneration of brain 5- HT neurons with the neurotoxin 5,7-dihydroxy- tryptamine (5,7-DHT) or by electrolytic lesions of the median raphe nucleus (an area with a high con- centration of serotonergic cell bodies), and that con- versely, the augmentation of brain serotonergic neurotransmission by administration of tryptophan

Invited Review 319

had the result of facilitating the development of tol- erance to the effects of ethanol (L~ et al., 1979).

Today, there is considerable evidence from both animal and human studies that 5-HT plays a role in the modulation of alcohol intake and/or dependence, and in the development of tolerance to ethanol. Para- doxically (in view of the findings of decreased ethanol consumption by depleters of cerebral 5-HT), the gen- eral consensus is that there exists an inverse relation- ship between the functioning of the cerebral serotonergic system and ethanol drinking preference (see Table 2). Pharmacological manipulations that reduce cerebral 5-HT concentrations increase ethanol intake in animals; conversely, procedures which increase 5-HT release or turnover produce a reduction in ethanol intake. Thus, substances which increase serotonergic neurotransmission, such as the 5-HT pre- cursors 5-hydroxytryptophan (5-HTP) and tryp- tophan, 5-HT uptake inhibitors, 5-HT releasers, and specific 5-HT receptor agonists, have been found to reduce ethanol consumption in both humans and lab- oratory animals. Moreover, studies using genetically bred lines of alcohol-preferring, alcohol-naive rats (P/NP, HAD/LAD) have demonstrated lower basal concentrations of 5-HT and its main metabolite, 5- hydroxyindoleacetic acid (5-HIAA) (Murphy et al., 1987; Gongwer et al., 1989), as well as a decreased turnover rate (Morinan, 1987) and a reduction in the number of 5-HT fibers (Zhou et al. 1991), in various brain areas of these rats as compared to their gen- etically bred, non alcohol-preferring counterparts. However, other authors using different lines of alco- hol-preferring and non-preferring rats (AA/ANA) have failed to find such differences (Ahtee and Eriks- son, 1972; Korpi et al., 1988).

The lesion of serotonergic cell bodies results in an increase in ethanol consumption in alcohol non-pre- ferring rats, while no such effect is observed in their alcohol-preferring counterparts (Ferreira and Soares- da-Silva, 1991), presumably due to their initial low basal cerebral 5-HT contents. The results of these animal studies are complemented by clinical findings, which tend to support the idea o fa hypofunctioning of the 5-HT system in alcoholic patients; indeed, drugs which activate serotonergic transmission appear to be of therapeutic value in reducing ethanol consumption (Sellers et al., 1992). In addition, a decrease in CSF tryptophan levels was observed following acute etha- nol consumption by healthy volunteers, this tending to indicate an increase in 5-HT synthesis during ethanol intoxication, while abstinent alcoholic patients pre- sented elevated basal CSF tryptophan levels and low CSF levels of 5-HIAA, which would further support

the view that alcoholics have a lower inherent 5-HT turnover (Beck et al., 1984). The idea that alcoholic subjects have lower baseline 5-HIAA levels in the CSF was first proposed in 1979 by Ballenger and his co- workers. These levels are transiently elevated during acute ethanol consumption, but during non-con- suming periods, there occurs a further depletion, which merely serves to aggravate the cycle. This and other evidence along the same line has led to the formulation of a hypothesis whereby subjects pre- disposed to the consumption of alcohol would have lower, genetically determined 5-HT brain levels, and that drinking reflects an attempt to correct the initial deficit in 5-HT neurotransmission. The genetic com- ponent of the predisposition to alcoholism is sup- ported by the finding of low central serotonergic transmission in non-drinking adult children of alcoholics (Hunt, 1990), and by an increased platelet 5-HT uptake in alcoholics and their descendants (Ernouf et al., 1993). In addition, a significant associ- ation between a particular genotype (LL) of the enzyme that catalyzes the rate-limiting step in the biosynthesis of 5-HT, tryptophan hydroxylase, and low CSF 5-HIAA concentrations has recently been reported in impulsive alcoholic violent offenders (Nielsen et al., 1994).

Receptor studies

Binding studies in animal models of alcoholism and using postmortem human brain samples from alcoholics unfortunately do not provide a very clear picture of the possible effects of alcohol on 5-HT receptors (for a general overview, see Fig. 3). One reason for inconsistencies in the results is likely differ- ences in the animal models and mode of ethanol administration, which often vary significantly from one group to another. Wong et al. (1988) found an increased [3H]5-HT binding to 5-HT~ receptors in cortical and hippocampal membrane preparations of P rats, as compared to their NP counterparts; this same group later examined binding of [3H]8-OH- DPAT to 5-HTIA receptors in cortical and hippo- campal membrane preparations of P rats, where they found a higher density of binding sites (Bmax) and higher affinity (lower Kd) in both areas (Wong et al., 1990). In a subsequent study, a higher Bmax was found, with no significant change in Kd, for binding of [3H]8- OH-DPAT to 5-HT~A receptors in cortical membrane preparations of P rats (Wong et al., 1993). An increase in 5-HT~A receptor binding in the medial nucleus accumbens and medial prefrontal cortex has also been reported by McBride et al. (1990), who used a quan- titative autoradiographie technique for measurement

320 Invited Review'

~5-HT Neuron~

Firing

Somato-Dendrific Autoreceptors:

©

- - Pre-synaptic - ~ Receptors:

/ / - - ~ 5-~IT - / I Ethan°ll

V -- ~'O V ~ N~[, Post-syna tic I ( ~ Recep~

Fig. 3. EffEcts of acute and chronic exposure to ethanol (in unselected animal strains) on the variouf 5-HT receptor subtypes. • positive effect. O negative effect.

Invited Review 321

of these receptors in P rats. The authors' interpret- ation of these results is that an up-regulation of 5-HT1A receptors may have developed in the P rats as a compensatory mechanism to adapt to a lower 5- HT innervation (Zhou et al., 1991) and/or function. However, Korpi et al. (1992b) found no change in [3H]5-HT binding to 5-HTj receptors in various brain regions of AA as compared to ANA rats, and another group (Ulrichsen, 1991), in a study of 5-HTIA recep- tors during severe chronic ethanol intoxication and withdrawal in the rat, observed a decrease in the Bma x of [3H] 8-OH-DPAT binding in hippocampal (but not cortical) membrane preparations, with no change in Kd, in both the intoxication and withdrawal phases. Pandey et al. (1992) also noted no change in [3H]8- OH-DPAT binding in cortical or hippocampal mem- branes of rats subjected to short-term or long-term ethanol treatment or upon withdrawal. However, in the human brain postmortem, Dillon et al. (1991) found a decreased binding of [3H]8-OH-DPAT to 5- HTIA receptors in several cortical gyri of alcoholics.

Studies of the 5-HT2A receptor subtype produced results showing either decreases in various brain areas of alcohol-preferring rats as compared to non-pre- ferring rats (McBride et al., 1990, 1993b), or no change (Wong et al., 1988; Korpi et al., 1992b) in the density of corresponding binding sites. In man, Simonsson and Ailing (1988) demonstrated a reduced function of the 5-HT2A receptor on platelets obtained from alcoholics, shortly after a period of abuse, by examining 5-HT-stimulated phosphoinositide hydro- lysis; a subsequent study (Simonsson et al., 1992) detected no difference between post-withdrawal alcoholics and controls, showing that this reduction in 5-HT2 receptor function is a state-dependent rather than a trait-dependent marker. A decrease in 5-HT2A receptor number and 5-HT-stimulated [3H]inositol-1- phosphate formation has been found in rat cerebral cortex after long-term ethanol exposure and with- drawal by Pandey et al. (1992), who attributed these changes to a possible reduction in coupling of 5-HT2A receptors to G-protein or phospholipase C.

Very little data is available concerning the binding of specific ligands to the other subtypes of 5-HT recep- tors. One study found a lower density of 5-HTIB recep- tors, as measured by [125I](-)iodocyanopindolol binding, in the medial nucleus accumbens of P rats as compared to NP rats (McBride et al., 1990), while another study found no difference in [3H]LY 278584 binding to 5-HT3 receptors in various brain regions of AA and ANA rats (Korpi et al., 1992b). Nevertheless, a direct effect of alcohol on the 5-HT3 receptor has been reported by Lovinger and White (1991), who

found that 5-HT3 receptor-mediated ion current in neuroblastoma cells and isolated adult mammalian neurons is potentiated by ethanol.

Pharmacological manipulations affecting ethanol con- sumption

5 - H T precursor loading. In 1972, Myers et al. demonstrated the inhibitory effect of i.p. and i.c.v. 5- HTP, the precursor of 5-HT, on ethanol consumption in the rat. This effect was also observed by Alvarado et al. (1990) in UChB rats, which exhibited a decrease in specific appetite for ethanol (i.e. no change in food or total fluid intake) upon i.p. 5-HTP administration. The same, specific effect was obtained upon adminis- tration of an L-5-HTP derivative, triptosine, to out- bred rats with an experimentally-induced preference for ethanol (Ernouf et al., 1992). However, it cannot be excluded that these decreases in consumption may be provoked by a taste aversion to ethanol induced by such compounds (Zabik and Roache, 1983).

Serotonin uptake modulators. Specific 5-HT uptake blockers have long been known to have a suppressant effect on ethanol consumption in animal models, including alcohol-preferring rat strains, and have been shown to be of therapeutic value in reducing ethanol intake in problem drinkers (Naranjo et al., 1986). In fact, most work to date at the preclinical and clinical levels, with respect to treatment of problem drinking via modulation of the serotonergic system, has been done with 5-HT uptake blockers such as fluvoxamine, fluoxetine, citalopram and zimelidine (Gill and Amit, 1989). Overall, clinical studies with these and other uptake inhibitors have yielded positive results in the short-term management of problem drinkers (Nar- anjo et al., 1986, 1992), although none have dem- onstrated clinical efficacy in the long-term treatment of the heavy drinker (Thomas, 1991).

However, analogous to the effects described above upon administration of the 5-HT synthesis inhibitor pCPA, there is a general consensus that the effects of 5-HT uptake inhibitors in decreasing ethanol con- sumption are secondary to their actions on con- summatory behaviors in general, and on caloric regulation (Myers and Quarfordt, 1991). Indeed, most animal studies carried out to date with 5-HT uptake inhibitors have found a decrease in food and/or total fluid consumption concomitant to the reduction in ethanol drinking, and clinical studies often report a weight loss in subjects treated with these compounds. Nevertheless, in a study examining the effects of fluoxetine on the intragastric self-administration of ethanol in the P line of rats, Murphy et al. (1988) found that 10 mg/kg of this drug actually increased

322 Invited Review

sell-administration of water and drinking of a flavored solution associated with the ingestion of ethanol, with no significant reduction in food intake, while reducing self-administration of a 20% ethanol solution. In addition, using an ethanol-reinforced responding paradigm, Haraguchi et al. (1990) observed no decrease in body weight in nonfood- or fluid-deprived rats upon fluoxetine administration, at doses pro- ducing a decrease in the oral self-administration of 10% ethanol, and Lu et al. (1993) found that fluox- etine reduced both ethanol and water consumption in rats, although the suppression of ethanol intake was significantly greater than that of water intake. Also in man, Naranjo et al. (1992) noted no change in sub- jects' body weights or self-reported consumption of non-alcoholic drinks upon treatment with citalopram. Thus, according to some authors, the ethanol intake- reducing effect of the 5-HT uptake inhibitors would primarily be due to their actions on satiety mech- anisms in general, while others contend that these drugs exert a greater effect on ethanol drinking than on other ingestive behaviors. This latter view is sup- ported by studies examining the consumption of pal- atable substances, which have shown that the effects of 5-HT uptake blockers are greater upon substances with more pronounced reinforcing efficacies (Leander, 1987).

Studies have also been carried out with a 5-HT uptake enhancer which, one might imagine, should have the effect of increasing ethanol intake. Para- doxically, Daoust et al. (1992) found that the 5-HT uptake enhancer tianeptine also decreased ethanol intake in rats in which a preference for ethanol had been induced by alcoholization. Contrary to what was obtained for most of the 5-HT uptake blockers, however, tianeptine had no inhibitory effect on food intake. The common effect of tianeptine and the uptake blockers may derive from the fact that both classes of drugs enhance 5-HT metabolism/function through an increased availability oftryptophan for 5- HT synthesis, due to their common inhibitory influ- ence on liver tryptophan pyrrolase activity.

5 - H T releasers. As with 5-HT uptake inhibitors, 5- HT releasers, such as fenfluramine or dexfen- fluramine, have generally been found to decrease etha- nol consumption in animal models of alcoholism (Krasner et al., 1975 ; Lu et al., 1993), consistent with the idea that increasing 5-HT concentrations in the synaptic cleft attenuates ethanol intake. In common with the uptake blockers, 5-HT releasers may act via a potent anorectic effect, yet the results obtained by Lu et al. (1993) showed that the ethanol intake-reducing effect of fenfluramine is signiiicantly greater than

that on the reduction of water or sucrose solution intake.

Spec!/ ic 5 - H T receptor [igands. Studies using alco- hol-preferring rodent strains (Kostowski and Dyr, 1992), monkeys (Collins and Myers, 1987), and clini- cal studies in humans (Bruno, 1989) have nearly all reported a decrease in alcohol consumption with the 5-HTIA agonists buspirone, ipsapirone, 8-OH-DPAT, or gepirone; furthermore, in most cases, the effect observed was specific for ethanol consumption, as food and/or total fluid intake was either not affected or increased. Moreover, Schreiber et al. (1993) demonstrated that the ethanol preference-reducing effect of ipasapirone (injected s.c.) could be com- pletely blocked by the (non-selective) 5-HTIA antag- onist spiperone (s.c.). The same authors also reported a reduction in the ethanol consumption of alcohol- preferring rats by microinjection of 8-OH-DPAT into the dorsal raphe nucleus (a brain area rich in somato- dendritic 5-HTiA autoreceptors), with no significant effect observed upon injection into the nucleus accum- bens: based on these data, Schreiber et al. (1993) suggested that 5-HT~A receptor agonists reduced etha- nol preference via stimulation of presynaptic auto- receptors on raphe cell bodies. Interestingly, Blomqvist et al. (1994) recently demonstrated the ability of the 5-HT~A receptor agonists 8-OH-DPAT, ipsapirone and buspirone to antagonize ethanol- stimulated locomotor activity, an effect which has been proposed to be homologous to ethanol's rein- forcing properties (Wise and Bozarth, 1987).

In a number of studies, administration of 5-HT3 receptor antagonists (e.g. ondansetron, ICS 205-930, MDL 72222, zacopride) has also been shown: (i) to attenuate volitional ethanol consumption in various animal models (Knapp and Pohorecky, 1992), in alco- hol-preferring rats (Fadda et al., 1991 ; Meert, 1993) and in human alcohol abusers (Toneatto et al., 1991) ; (ii) to attenuate ethanol-induced hyperlocomotion (Rajachandran et al., 1993); (iii) to block the dis- criminative stimulus effects of ethanol (Grant and Barrett, 1991); (iv) to prevent or antagonize the behavioral consequences of ethanol withdrawal in rats (Kostowski et al., 1993); (v) to antagonize ethanol- stimulated DA release in the nucleus accumbens (Car- boni et al., 1989; see also below); and (vi) to reduce the desire to drink and prevent withdrawal in human subjects (Fozard, 1992). Taken together, these data suggest that 5-HT3 receptor antagonists may bring about their inhibitory effect on voluntary ethanol con- sumption by reducing both ethanol's rewarding and intoxicating properties. Nevertheless, once again, the degree of specificity of the actions of these drugs on

Invited Review 323

ethanol consumption is questionable, with many inconsistencies from one study to the other with regard to the effects of 5-HT3 antagonists on general consummatory behavior (Sellers et al., 1992).

The 5-HTIDt~ receptor (5-HT m in rodents ; see Hum- phrey et aL, 1993) is probably another subtype to be implicated in the phenomenon of alcoholism. Indeed, following the observation by George et al. (1990) of an ethanol-like behavioral response in abstinent alcoholics upon administration of the non-selective 5- HT agonist mCPP (m-chlorophenylpiperazine), which acts at 5-HTID#, 5-HT2A and 5-HT2c receptors, Grant and Colombo (1993) noted the ability of the non-selective 5-HT agonist TFMPP (m-trifluoro- methyl-phenylpiperazine), which has a similar affinity for the 5-HTID~, 5-HT~D~, 5-HT~A, and 5-HT2c recep- tors, to substitute for the discriminative stimulus effects of ethanol; as investigations of the dis- criminative stimulus effects of TFMPP have min- imized a role for 5-HT~A and 5-HT2c receptors, a 5- HTIDa mechanism has been suggested for the mediation of this effect of ethanol. Nevertheless, fur- ther investigations with more selective ligands of 5- HTID~.qDt3 receptors are needed to actually assess the involvement of these receptors in alcohol abuse and alcoholism.

The 5-HT2 receptor family has also, of late, become the focus of attention. Indeed, decreases in alcohol consumption have been observed with 5-HT2A/2C agonists such as DOI and MK212, although with a concomitant decrease in food intake (Sellers et al., 1992). According to certain studies, the 5-HT2A/2c antagonist ritanserin attenuates alcohol consumption in rats with a developed preference for ethanol without affecting other consummatory behaviors (Panocka and Massi, 1992; Meert, 1993) and in chronic alcoholics (Monti and Alterwain, 1991). However, these findings were not confirmed by Myers and Lank- ford (1993), who claimed that the former results were most likely due to the fact that a low concentration (3 %) ethanol solution, rather than the rats' maximally preferred concentration, was used in previous studies (Panocka and Massi, 1992 ; Meert, 1993). Long-term treatment with the 5-HT2 antagonist amperozide (which also possesses some DA-releasing properties), on the other hand, was found to irreversibly suppress ethanol drinking, without producing any apparent side effects, in rats in which alcohol preference was pharmacologically induced with the aldehyde dehydrogenase inhibitor cyanamide (Myers et al., 1993a). Similar results were obtained in P rats, while no effect was observed on their NP counterparts, pre-

sumably due to their initial low level of alcohol con- sumption (Myers et al., 1993b). A recent study tested a promising new drug, FG 5893, a "novel second- generation amperozide-like drug", which combines 5- HT1A agonist and 5-HT2 antagonist properties (Singh et al., 1993). Administration of this compound resulted in a significant, long-lasting reduction in intake of a maximally preferred concentration of etha- nol in rats with cyanamide-induced alcohol pref- erence, while producing no change in body weight, food intake or total fluid consumption. These results thus corroborate previous evidence implicating 5- HT~A and 5-HT2 receptor subtypes in the addictive property of alcohol. Given these promising initial results, further animal (and possibly clinical) studies with drugs sharing a similar mechanism of action are warranted.

Serotonin-dopamine interactions

The involvement of the mesolimbic DA system in mechanisms of positive reinforcement is well docu- mented (see above). In vivo microdialysis studies in the nucleus accumbens have shown that ethanol administration (by i.p. injection or by local appli- cation through the microdialysis probe) will pref- erentially stimulate DA (Imperato and Di Chiara, 1986; Yoshimoto et aL, 1991) but also 5-HT (Yoshi- moto et al., 1991) release in this area and 5-HT release in the frontal cortex of sP rats (Portas et al., 1994). Electrophysiological data agree with the latter obser- vations, since ethanol concentrations corresponding to those attained during moderate alcohol intoxi- cation in vivo enhance the firing rate of dorsal raphe nucleus neurons (Chu and Keenan, 1987). Several observations support the existence of functional inter- actions between 5-HT and DA neurotransmission, notably within the context of the central reinforcing effect of ethanol. In fact, Herv6 et al. (1987) have shown that 5-HT neurons in anterior raphe nuclei extend processes which make synaptic contacts on DA cells located in the VTA and which project into the nucleus accumbens. Furthermore, microinjections of 5-HT into the VTA (Guan and McBride, 1989) or of excitatory agents directly into the dorsal raphe nucleus (Yoshimoto and McBride, 1992) are capable of enhancing the release of DA in the nucleus accum- bens. One line of evidence indicates that the mech- anism of this excitatory action of 5-HT may be via the 5-HT3 receptor, whose stimulation by agonists increases DA release in the nucleus accumbens (Jiang et al., 1990). Indeed, systemic administration (Car- boni et al., 1989) or local application through a micro- dialysis probe (Yoshimoto et al., 1991) of 5-HT3

324 Invited Review

antagonists has been shown to attenuate the stimu- latory effects of ethanol (i.p. or local application) on DA release in the nucleus accumbens. This may be the mechanism by which drugs which antagonize 5-HT~ receptor function act to produce their observed behavioral effects on ethanol preference (see above).

MONOAMINE OXIDASE

In the early attempts to elucidate the neurobiological bases of alcohol intoxication, particular attention was paid to the main enzymatic pathway responsible for the degradation of the catecholamines and serotonin, that catalyzed by monoamine oxidase (MAO, EC 1.4.3.4). The brain is known to contain both of the isozyme forms of the enzyme, MAO-A and MAO-B, whereas platelets contain only the -B'" form. While MAO-A is mainly localized in catecholamine-containing neurons in the brain, the serotonergic neurons of the raphe complex and glial cells contain predominantly the "B'" form ot" the enzyme (Abell et al., 1988).

Observations as to the inhibitory effects of ethanol in vitro Oll the activity of this enzyme were made as early as the 1960's (Maynard and Schenker, 1962). Sub- sequently, Feldstein et al. (1964) demonstrated a decreased urinary recovery of 5-HIAA (the main metab- olite of 5-HT, obtained by degradation by MAO) upon ethanol consumption. However, Palaic et al. (1971) found no change in brain MAO activity after either acute or chronic ethanol treatment.

Following the introduction of the estimation of plate- let MAO activity in the mid 1960s (Paasonen and Sol- atunturi, 1965), a large number of studies on the connections between platelet MAO activity and alcohol- ism began to appear in the early 1970s. Because of the accessibility of platelets, the study of MAO activity in humans was made possible, with the hope that platelet MAO activity might reflect MAO-B expression in the CNS (Youdim, 1988). The general consensus is that subjects exhibiting alcohol abuse present lower platelet MAO activity than controls (e.g. Oreland et al., 1981). Schuckit et al. (1982) demonstrated that even non- alcoholic relatives of alcoholics had a reduced platelet MAO activity as compared to controls with no alcoholic relatives.

An in t:itro study of MAO activity in human tissues by Tabakoff et al. (1985) provided evidence that ethanol inhibits MAO-B in platelets, and that it was this form of the enzyme which is also preferentially inhibited by ethanol in the brain. Conversely, the effect of ethanol on the "A" isoform of the enzyme may be different, as Kono et al. (1993) showed that the incubation of cub

tured human placental cells (which contain only MAO- A) with pharmacologically relevant concentrations of ethanol (3(7>70 raM) significantly increased MAO activity.

ACETYLCHOLINE

Chronic ethanol intake is known to provoke a loss of memory and cognitive impairment in man (Lish- man, 1990) and to disrupt memory processes in vari- ous animal models (Arendt et al. , 1989; Hodges et

al., 1991 ; Beracochea e t al. , 1992). By virtue of the implication of the cholinergic system in cognition and memory (Drachman, 1977), numerous studies have tbcused on the involvement of acetylcholine (ACh) in ethanol-induced impairment of cognitive functions. It thus appeared that chronic alcoholic subjects with KorsakoWs syndrome, a disease characterized by an irreversible loss of memory, confabulation and dis- orientation (Lishman, 1990), exhibited a loss of chol- inergic neurons in the nucleus basalis of Meynert (Arendt et al., 1983). Moreover, the activity of choline acetyltransferase (CHAT), the enzyme for ACh syn- thesis, was found to be reduced in several brain areas of chronic alcoholics (Antuono et al. , 1980).

The loss of cholinergic neurons observed in chronic alcoholic subjects was replicated by Arendt et al.

(1988) using an animal model after prolonged (12 weeks) ethanol intake. These authors also reported decreases in ACh content and in the activities of both ChAT and acetylcholinesterase (ACHE), the enzyme which degrades ACh, in various brain areas of the rats which had been exposed to ethanol for 12 weeks. These results were later confirmed by another study, in which 28 weeks of ethanol intake provoked profound decreases in ChAT and AChE activities and ACh levels in various brain areas of alcoholized rats, as well as decreases in ACh synthesis and release (evoked #7 v i tro by K + depolarization), and in high affinity choline uptake (Arendt et al., 1989). Chronic ethanol treatment was also found to decrease the cholinergic facilitation of population spikes in the CAI region of the hippocampus without affecting cholinergic inhi- bition of field EPSPs, indicating that alcohol appears to selectively disrupt a subset of cholinergic effector systems within hippocampal neurons (Rothberg et al.,

1993). In further support of the idea that chronic ethanol

provokes a depression of brain cholinergic activity, an interesting series of experiments demonstrated that grafting of ACh-rich fetal basal forebrain transplants into the cortex and hippocampus of alcoholized rats improved cholinergic and behavioral deficits and

Invited Review 325

memory functions impaired by treatment with ethanol (Arendt et al. , 1989 ; Hodges e t al. 1991). Moreover, cholinomimetic drugs have been shown to ameliorate behavioral performances impaired by chronic ethanol. In particular, physostigmine completely reversed alco- hol-induced deficits in T-maze spontaneous alter- nation rates (Beracochea et al. , 1992). Furthermore, performance of alcohol-treated rats in the radial maze was improved by the cholinergic agonists arecoline and nicotine (and disrupted by the antagonists scop- olamine and mecamylamine ; Hodges et al. , 1991).

Chronic ethanol, both in man and in animal models, has also been shown to produce changes in cholinergic receptor binding sites. Hellstr/Sm-Lindahl et al. (1993) noted marked reductions in the total number of mus- carinic receptors, as well as m~ and m2 subtypes, in the thalamus of a group of old (>59 yr), but not young (19-57 yr), alcoholics (in post-mortem brain samples), whereas the age-related decrease in cortical muscarinic receptor binding sites did not differ between controls and chronic alcoholics. The same study detected no difference in cortical and thalamic nicotinic receptor binding sites between alcoholics and controls. Other authors also reported losses of mus- carinic receptors in various brain areas (Antuono e t al. , 1980), including the frontal cortex, hippocampus and putamen of human alcohol abusers (Freund and Ballinger, 1991). Conversely, Hu e t al. (1993) found that long-term (2 days) treatment of NGI08-15 cells with ethanol increased the binding of the muscarinic acetylcholine receptor antagonist [3H]quinuclidinyl benzilate (which labels total muscarinic binding sites) and Pick et al. (1993) noted that exposure of mouse pups to ethanol increased the density of muscarinic receptors in the hippocampus. In contrast, Rothberg e t al. (1993) failed to find a difference between control and chronically alcoholized rats in muscarinic recep- tor subtype (m~-ms) densities in the hippocampus. Wahlstr6m and Nordberg (1991) also observed no differences in muscarinic or nicotinic receptor binding in various brain areas between groups of differentially alcoholized rats and controls.

ENDOGENOUS OPIOIDS

There is considerable evidence that the reinforcing effects of ethanol are also mediated, at least in part, by the brain's endogenous opioid systems, and that these systems may contribute to the adaptive neuronal responses provoked by ethanol intoxication. It is known, for example, that a number of the behavioral and pharmacological effects of ethanol, such as hypo- thermia, euphoria, analgesia and motor activation, as

well as the development of tolerance and dependence, are similar to those produced by opiates (Kalant, 1977). Moreover, the opiate antagonist naloxone has been shown to be able to inhibit the development of dependence to ethanol in mice (Blum e t al. , 1977), while cross-tolerance can develop between ethanol and the opiate agonist morphine (Khanna et al. , 1979). Interestingly, Blum e t al. (1983) found that there is an inverse correlation between ethanol con- sumption and brain levels of the endogenous opioid peptide Met-enkephalin, and a recent study by Gian- oulakis e t al. (1992) has revealed differences between AA and ANA rats in the hypothalamic mRNA con- tents of proopiomelanocortin, the precursor of the endogenous opioid//-endorphin, and in the content of/%endorphin-like immunoreactivity in distinct brain areas.

The traditional view of the involvement of endogen- ous opioids in the voluntary consumption of alcohol is referred to as the opioid deficiency hypothesis (Reid e t al. , 1991), which states that, analogous to what is believed to be the case for serotonergic neuro- transmission (see section on Serotonin), alcoholics suffer from a deficiency in central opioidergic neural activity, which is compensated for by the consumption of alcohol. The mechanism for this compensation may be via an alteration of the binding properties of the opiate receptors or of the release and synthesis of the endogenous opioids themselves. Alternatively, by- products of ethanol metabolism, the tetrahydro- isoquinolines and fl-carbolines (condensation pro- ducts of acetaldehyde with catecholamines and indoleamines, respectively), may mediate ethanol reinforcement through interactions with central endogenous opiate systems (Davis and Walsh, 1970).

Two opioid receptors have been more particularly implicated in alcohol effects, the p and 6 subtypes. An early study by Altshuler et al. (1980) revealed that the blockade of these receptors by naltrexone reduced rhesus monkeys' intravenous self-administration of ethanol, suggesting that opiate receptor blockade had diminished the reinforcing properties of ethanol. Simi- larly, naloxone (Weiss e t al. , 1990; Hyytig and Sin- clair, 1993), naltrexone (Altshuler e t al. , 1980) and the opioid antagonist nalmefene (Hubbell et al. , 1991) reduce rats' preference for ethanol. Furthermore, Froehlich e t al. (1991 ) demonstrated the efficacy of the selective 6-opioid antagonist ICI 174,864 in reducing ethanol consumption in rats, as well as the ability of thiorphan, an enkephalinase inhibitor which has the effect of potentiating activation of the 6-receptor, to increase ethanol intake. However, Hyytig (1993)

326 Invited Review

found that the #-opioid antagonist CTOP suppressed alcohol drinking in AA rats, while the ,5-opioid antag- onist ICI 174,864 was without effect. Conversely, low doses of morphine (1 2 mg/kg) increased ethanol preference in rats (Hubbell et al., 1993).

Certain functional consequences of ethanol intake may also be mediated by the opioidergic system, as naloxone has proven effective in attenuating the etha- nol withdrawal syndrome (Blum et al., 1977), and ICI 174,864, microinjected into discrete brain regions, has been shown to block ethanol-induced hypothermia and sedation (Widdowson, 1987).

Based on these observations in animal studies, nal- trexone has been applied with success in the treatment of alcohol dependence in man (Volpicelli et al., 1992).

It is well accepted that the stimulation of DA release in the nucleus accumbens is a prerequisite for the expression of the rewarding properties of systemically injected morphine (Di Chiara and lmperato, 1988). Such a release is also observed upon microinjection of the endogenous opioid peptide Met-enkephalin into the VTA (Di Chiara and lmperato, 1988). Moreover, chronic morphine treatment has been shown to pro- duce a significant decrease in DA transporter density in the anterior basal forebrain, an area which includes the nucleus accumbens, without producing any effect on binding to the DA transporter in striatal mem- branes or on binding to the 5-HT transporter in these areas (Simantov, 1993). These results support the data obtained in other studies, which had suggested that opiate reward is dependent upon a dopaminergic sub- strate (Bozarth and Wise, 1981). In a recent study employing the technique of in vivo microdialysis, Benjamin el al. (1993) found the administration of naltrexone to reverse the ethanol-induced DA release in the nucleus accumbens. Similarly, Acquas et al. (1993) observed an inhibition of ethanol-stimulated (but not cocaine-stimulated) DA release in the nucleus accumbens using naltrindole, a specific antagonist of the (5-receptor subtype. These observations support the findings of a previous in vitro study by Widdowson and Holman (1992), which showed that both naloxone and ICI 174,864 inhibited the increase in DA release induced by the addition of 75 mM ethanol to the incubation medium of rat striatal slices.

Clearly, all these results correspond with the idea that ethanol affects DA neurotransmission through the activation of an opioid intermediary link ending with the stimulation of ~ and/or ll receptors.

One of the mechanisms which can mediate the potentiation by ethanol of the opioidergic modulation of neural reward mechanisms is via adaptive changes in the density or affinity ofopioid receptors. A number

of studies have demonstrated the ability of acute etha- nol exposure to selectively inhibit opioid binding to ~%opioid receptors by promoting ligand dissociation (e.g. Charness et al., 1983), whereas long-term ethanol exposure up-regulates 6-opioid receptors in NG 108- 15 cells (Charness et al., 1983) and in rodent brain (Gianoulakis, 1983), and increases 6-opioid receptor gene expression (Charness et al., 1993) in NG 108-15 cells.

OTHER NEUROPEPTIDES

In addition to the endogenous opioids, a number of other neuropeptides have been examined for their involvement in the mechanisms underlying alcoholism and/or the CNS actions of ethanol. Notably, the brain-gut neuropeptides cholecystokinin (CCK) and bombesin have been shown to inhibit intake of alcohol solutions and feeding behaviors in a variety of species (e.g. Kulkosky and Glazner, 1988), including rats selectively bred for ethanol sensitivity (Kulkosky et

al., 1993). The reduction in free choice of ethanol solutions observed in non-deprived animals (Toth et

al.. 1990) suggests that these neuropeptides may func- tion endogenously in the control of the motivation to consume alcohol, as signals of satiation with ethanol (Kulkosky et al., 1989). In addition, there is evidence for the involvement of neurotensin in the effects of ethanol. In particular, a down-regulation of the recep- tors of this peptide was observed upon chronic administration of ethanol to mice (Campbell and Erwin, 1993). Furthermore, genetic-based differences in the brain levels of this peptide and its receptors have been described in mice bred for differential sen- sitivity to ethanol (Erwin et al., 1993). A role for the tachykinins in alcohol preference has also been proposed, as selective agonists at the NK3 tachykinin receptors have been shown to selectively inhibit alco- hol intake in sP rats (Ciccocioppo et al., 1994).

CONCLUSION

As this review has shown, the CNS effects of ethanol are multifaceted, with diverse actions on several neurotransmitter systems.

When a low dose of alcohol is ingested, the dopa- mineric, serotonergic and noradrenergic systems are stimulated (Engel and Liljequist, 1983). While acti- vation of the dopaminergic system will initiate positive reinforcement mechanisms in the brain's limbic structures, enhancement of noradrenergic neuro- transmission, releasing NE particularly in the hypo- thalamus and midbrain, might be associated with

Invited Review 327

ethanol-induced euphoria and a general excitation of central functions. However, during acute intoxication after intake of a high dose of alcohol, a feeling of psychic and motor inhibition will prevail, char- acterized by a generally sedated and dysphoric state accompanied by anxiolysis and muscle relaxation; this aspect is presumably mediated by reduction of dopaminergic and noradrenergic transmission on the one hand, and by ethanol-induced enhancement of the binding of GABA to the GABAA receptor complex on the other hand, resulting in a potentiation of GABA's general central depressant actions.

Upon prolonged alcohol consumption, tolerance and physical dependence will develop, rendering neu- ronal function "normal" in the presence of ethanol and "abnormal" in its absence (Charness et al., 1989). However, potentiation by chronic ethanol of glu- tamatergic function will enhance the endogenous amino acid's neurotoxic effects, possibly initiating a process leading to delayed neuronal death. Loss of cholinergic neurons, concomitant with a global etha- nol-induced inhibition of cholinergic function, may contribute to the well-documented memory-dis- rupting effects of chronic alcohol consumption.

Upon withdrawal of ethanol following a period of chronic consumption, decreased release or hypo- sensitivity of the catecholamine receptors may account for irritability, while post-withdrawal modi- fications in GABAergic function may contribute to a lowering of the threshold for the appearance of withdrawal seizures. These symptoms can be readily reversed by intake of a moderate dose of alcohol, thus improving, for a short period of time, CNS functions.

It is likely that the reinforcing properties of ethanol are mediated mainly by the dopaminergic system. In turn, the ethanol-induced increase in DA release in the nucleus accumbens, which presumably represents the neurobiological substrate for reward, may be modulated by serotonergic and opioidergic neuro- transmission. Indeed, the release of DA is facilitated by the activation of 5-HT3 receptors, as blockade of these receptors prevents the DA release observed upon administration of ethanol. Similarly, blockade of/~- and/or ~i-opioid receptors also inhibits the ethanol- stimulated release of DA, suggesting a facilitatory role for the brain's endogenous opioids in the development of ethanol preference.

REFERENCES

Abell C. W., Denney R. M. and Westlund K. N. (1988) Localization and function of monoamine oxidases A and B. In : Progress in Catecholamine Research, Part A : Basic

Aspects and Peripheral Mechanisms (Dahlstrrm A., Belmaker R. H. and Sandler M., eds), pp. 103 108. Alan R. Liss, New York.

Acquas E., Meloni M. and Di Chiara G. (1993) Blockade of 6-opioid receptors in the nucleus accumbens prevents ethanol-induced stimulation of dopamine release. Eur. J. Pharmac. 230, 239-241.

Aguayo L. G. (1990) Ethanol potentiates the GABAA-acti- vated CI- current in mouse hippocampal and cortical neu- rons. Eur. J. Pharmac. 187, 127-130.

Ahern K. V., Lustig H. S. and Greenberg D. A. (1994) Enhancement of NMDA toxicity and calcium responses by chronic exposure of cultured cortical neurons to etha- nol. Neurosci. Lett. 165, 211-214.

Ahtee L. and Eriksson K. (1972) 5-hydroxytryptamine and 5-hydroxyindoleacetic acid content in brain of rat strains selected for their alcohol intake. Physiol. Behav. 8, 123- 126.

Allan A. M. and Harris R. A. (1987a) Involvement of neu- ronal chloride channels in ethanol intoxication, tolerance and dependence. In: Recent Developments in Alcoholism (Galanter M., ed.), pp. 313-325, Vol. 5. Plenum Press, New York.

Allan A. M. and Harris R. A. (1987b) Acute and chronic ethanol treatments alter GABA receptor-operated chlo- ride channels. Pharmac. Biochem. Behav. 27, 665~70.

Allan A. M. and Harris R. A. (1989) A new alcohol antag- onist: phaclofen. Life Sci. 45, 1771-1779.

Allan A. M., Spuhler K. P. and Harris R. A. (1988) Gamma aminobutyric acid-activated chloride channels: relation- ships in ethanol sensitivity. J. Pharmac. exp. Ther. 244, 866-870.

Altshuler H. L., Phillips P. E. and Feinhandler D. A. (1980) Alteration of ethanol self-administration by naltrexone. Life Sci. 26, 679-688.

Alvarado R., Contreras S., Segovia-Riquelrne N. and Mar- dones J. (1990) Effects of serotonin uptake blockers and of 5-hydroxytryptophan on the voluntary consumption of ethanol, water and solid food by UChA and UChB rats. Alcohol7, 315 319.

Amit Z. and Brown Z. W. (1982) Actions of drugs of abuse on brain reward systems: a reconsideration with specific attention to alcohol. Pharmac. Biochem. Behav. 17, 233- 238.

Antuono P., Sorbi S., Bracco L., Fusco T. and Amaducci L. (1980) A discrete sampling technique in senile dementia of the Alzheimer type and alcoholic dementia : study of the cholinergic system. In : Agin 9 of the Brain and Dementia (Amaducci L., Davison A. N. and Antuono P., eds), pp. 151-158, Vol. 13. Raven Press, New York.

Anunziato L., Amoroso S., Di Renzo G., Argenzio F., Auri- lio C., Grella A. and Quattrone A. (1983) Increased GH responsiveness to dopamine receptor stimulation in alco- hol addicts during the late withdrawal syndrome. Life Sci. 33, 2651~655.

Arendt T., Bigl V., Arendt A. and Tennstedt A. (1983) Loss of neurons in the nucleus hasalis of Meynert in Alzheimer's disease, paralysis agitans and Korsakoff's disease. Acta Neuropath. 6, 101-108.

Arendt T., Hennig D., Gray J. A. and Marchbanks R. (1988) Loss of neurons in the rat basal forebrain cholinergic projection system after prolonged intake of ethanol. Brain Res. Bull. 21, 563-570.

Arendt T., Allen Y., Marchbanks R. M., Schugens M. M., Sinden J., Lantos P. L. and Gray J. A. (1989) Cholinergic

328 Invited Review

system and memory in the rat : effects of chronic ethanol, embryonic basal forebrain transplants and excitotoxic lesions of the cholinergic basal forebrain projection system. Neuroscienee 33, 435~462.

Balakleevsky A., Colombo G., Fadda F. and Gessa G. k. (1990) Ro 19-4603, a benzodiazepine receptor inverse agonist, attenuates voluntary ethanol consumption in rats selectively bred for high ethanol preference. Alcohol Alco- hol. 25, 449M52.

Balldin J., Berggren U., Lindstedt G. and Sundkler A. (1993) Further neuroendocrine evidence for reduced D: dopa- mine receptor function in alcoholism. Druq Alcohol Depend. 32, 159 162.

Ballenger J., Goodwin F. and Major L. (1979) Alcohol and central serotonin metabolism in man. Arch. Gen. Psyehiat. 36, 224-227.

Balster R. L., Grech D. M. and Bobelis D. J. (1992) Drug discrimination analysis of ethanol as an N-methyl-l~-aspar- tate receptor antagonist. Eur. J. Pharmac. 222, 39M2.

Barr C. L. and Kidd K. K. (1993) Population frequencies of the A1 allele at the dopamine D~ receptor locus. Biol. Psychiat. 34, 204-209.

Beck O., Borg S., Edman G., Fyro B., Oxenstierna G. and Sedvalt G. (1984) 5-hydroxytryptophol in human cer- ebrospinal fluid: conjugation, concentration gradient, relationship to 5-hydroxyindoleacetic acid, and influence of hereditary factors..L Neurochem. 43, 58 61.

Becker H. C. (1988) Effects of the imidazobenzodiazepine Ro 15-4513 on the stimulant and depressant actions of ethanol on spontaneous locomotor activity. L(/i, Sci. 43, 643 650.

Belknap J. K., Crabbe J. C. and Young E. R. (1993l Vol- untary consumption of ethanol in 15 inbred mouse strains. Psychopharmacology 112, 503 510.

Benjamin D., Grant E. R. and Pohorecky k. A. (1993) Nal- trexone reverses ethanol-induced dopamine release in the nucleus accumbens in awake, freely moving rats. Bra#7 Res. 621,137 140.

Beracochea D., Micheau J. and Jafl;ard R. (1992) Memory deficits following chronic alcohol consumption in mice: relationships with hippocampal and cortical cholinergic activities. Pharmac. Bioehem. Behav, 42, 749 753.

Blomqvist O., S6derpalm B. and Engel J. A. (1994) 5-HT~.~ receptor agonists reduce ethanol-induced locomotor activity in mice. Ah.ohol 11, 157 161.

Blum K., Futterman S., Wallace J. E. and Schwertner H. A. (1977) Naloxone-induced inhibition of ethanol depen- dence in mice. Nature 265, 49 51.

Blum K., Elston S. F. A., DeLallo L., Briggs A. H. and Wallace J. E. (1983) Ethanol acceptance as a function of genotype amounts of brain [Met]enkephalin. Proc. nam. Acad. Sci. U.S.A. 80, 6510 6512.

Blum K., Noble E. P., Sheridan P. J., Montgomery A., Rit- chie T., Jagadeeswaran P., Nogami H., Briggs A. H and Cohn J. B. (1990) Allelic association of human dopamine D~ receptor gene in alcoholism. J A M A 263, 2055 2060.

Blum K., Noble E. P., Sheridan P. J., Montgomery A., Rit- chie T., Ozkaragoz T., Fitch R. J., Wood R., Finley O. and Sadlack F. (1993) Genetic predisposition in alcoholism: association of the D2 dopamine receptor Taq 1 BI RFLP with severe alcoholics. Ah'ohol 10, 59--67.

Bolos A. M., Dean M., Lucas-Derse S.. Ramsburg M.. Brown G. L. and Goldman D. (1990) Population and pedigree studies reveal a lack of association between the

dopamine D: receptor gene and alcoholism. J A M A 264, 3156-3160.

Borg V. (1983) Bromocriptine in the prevention of alcohol abuse. Acta Psvchiat. Stand. 68, 100-110.

Borg S., Kvande H. and Sedvall G. (1983) Monoamine metabolites in lumbar cerebro-spinal fluid and urine in alcoholic patients before and after treatment with disul- firam. Ak~ohol Ah'ohol. 18, 61-65.

Bozarth M. A. and Wise R. A. (1981) Heroin reward is dependent upon a dopaminergic substrate. Li]b Sci. 28, 551 555.

Brodie M. S., Shether S. A. and Dunwiddie T. V. (1990) Ethanol increases the firing rate of dopamine neurons in the rat ventral tegmental area in vitro. Brain Res. 508, 65 69.

Brown Z. W. and Amit Z. (1977) The effects ot selective catecholamine depletions by 6-hydroxydopamine on etha- 11ol preference in rats. Neurosci. Lett. 5, 333-336.

Brown Z. W., Gill K., Abitbol M. and Amit Z. (1982) Lack of effect of dopamine receptor blockade on voluntary ethanol consumption in rats. Behav. neural Biol. 36, 291 294.

Bruno F. (1989) Buspirone in the treatment of alcoholic patients 22, (Suppl. 1) Psychopatholo,qy 49 59.

Buck K. J. and Harris R. A. (1990a) Benzodiazepine agonist and inverse agonist actions on GABAA receptor-operated chloride channels. I. Acute effects of ethanol. J. Pharmac. exp. Ther. 253, 70(~712.

Buck K. J. and Harris R. A. (1990b) Benzodiazepine agonist and inverse agonist actions on GABAA receptor-operated channels, ll. Chronic effects of ethanol. J. Pharmac. exp. Ther. 253, 713 719.

Buck K. J., Hahner L., Sikela J. and Harris R. A. (1991) Chronic ethanol treatment alters brain levels of ),-amino- butyric acidA receptor subunit mRNAs: relationship to genetic differences in ethanol withdrawal seizure severity. J. Neuroehem. 57, 1452 1455.

Campbell A. D. and Erwin V. G. (19931 Chronic ethanol administration downregulates neurotensin receptors in long- and short-sleep mice. Pharmae. Biochem. Behav. 45, 95 106.

Carboni E., Acquas E., Frau R. and Di Chiara G. (1989) Differential inhibitory effect of a 5-HT3 antagonist on drug-induced stimulation of dopamine release. Eur. J. Pharmac. 164, 515--519.

Carboni S., lsola R., Gessa G. L. and Rossetti Z. L. (1993) Ethanol prevents the glutamate release induced by N- methyl-D-aspartate in the rat striatum. Neurosci. Lett. 152, 133 136.

Carlsson A. and Lindqvist M. (1973) Effect of ethanol on the hydroxylation of tyrosine and tryptophan in rat brain in vit,o. J. Pharm. Pharmac. 25, 437M40.

Celantano J. J., Gibbs T. T. and Farb D. H. (1988) Ethanol potentiates GABA- and glycine-induced chloride currents in chick spinal cord neurons. Brain Res. 455, 377 380.

Chandler L. J., Newsom H., Sumners C. and Crews F. (1993) Chronic ethanol exposure potentiates NMDA excito- toxcity in cerebral cortical neurons. J. Neurochem. 60, 1578 1581.

Charness M. E., Gordon A. S. and Diamond 1. (1983) Etha- nol modulation of opiate receptors in cultured neural cells. Science 222, 1246 1248.

(2harness M. E., Simon R. P. and Greenberg D. A. (1989) Ethanol and the nervous system. N. Enql. J. Med. 321, 442 454.

Charness M. E., Hu G., Edwards R. H. and Querimit L. A.

Invited Review 329

(1993) Ethanol increases 6-opioid receptor gene expression in neuronal cell lines. Mol. Pharmac. 44, 1119-1127.

Chiodo L. A. (1988) Dopamine-containing neurons in the mammalian central nervous system: electrophysiology and pharmacology. Neurosci. Biobehav. Rev. 12, 49-91.

Chu N.-S. and Keenan L. (1987) Responses of midbrain dorsal raphe neurons to ethanol studied in brainstem slices. Alcohol 4, 373-374.

Ciccocioppo R., Panocka I., Pompei P., De Caro G. and Massi M. (1994) Selective agonists at NK3 tachykinin receptors inhibit alcohol intake in Sardinian alcohol-pre- ferring rats. Brain Res. Bull. 33, 71-77.

Cloninger C. R. (1987) Neurogenetic adaptive mechanisms in alcoholism. Science 236, 410-416.

Collingridge G. L., Kehl S. J. and McLennan H. (1983) Excitatory amino acids in synaptic transmission in the Schaffer collateral-commissural pathway of the rat hippo- campus. J. Physiol. (Lond.) 334, 33-46.

Collins D. M. and Myers R. (1987) Buspirone attenuates volitional alcohol intake in the chronically drinking monkey. Alcohol 4, 49-56.

Corrodi H., Fuxe K. and H6kfelt T. (1966) The effect of ethanol on the activity of central catecholamine neurones in rat brain. J. Pharm. Pharmac. 18, 821-823.

Cotman C. W., Bridges R. J., Taube J. S., Clark A. S., Geddes J. W. and Monaghan D. T. (1989) The role of the NMDA receptor in central nervous system plasticity and pathology. J. N I H Res. 1, 65-74.

Crabbe J. C., Kosobud A., Young E. R., Tam B. R. and McSwigan J. D. (1985) Bidirectional selection for sus- ceptibility to ethanol withdrawal seizures in Mus musculus. Behav. Genet. 15, 521-536.

Criswell H. E., Simson P. E., Duncan G. E., McCown T. J., Herbert J. S., Morrow A. L. and Breese G. R. (1993) Molecular basis for regionally specific action of ethanol on y-aminobutryric acidA receptors: generalization to other ligand-gated ion channels. J. Pharmac. exp. Ther. 267, 552-557.

Daoust M., Compagnon P., Legrand E. and MocaEr E. (1992) Tianeptine, a specific serotonin uptake enhancer, decreases ethanol intake in rats. Alcohol Alcohol. 27, 15- 17.

Davis V. E. and Walsh M. J. (1970) Alcohol, amines and alkaloids: a possible basis for alcohol addiction. Science 167, 1005-1007.

Davis W. M., Smith S. G. and Werner T. E. (1978) Nor- adrenergic role in self-administration of ethanol. Pharmac. Biochem. Behav. 9, 369-374.

Deitrich R. A. (1976) Biochemical aspects of alcoholism. Psychoneuroendocrinology 1, 325 346.

De Montis M. G., Devoto P., Giorgi G., Tagliamonte A. and Gessa G. L. (1991) Ethanol, at micromolar con- centrations, increases the affinity of [3H]MK-801 binding in rat brain. Eur. J. Pharmac. 199, 139-140.

De Montis M. G., Gambarana C., Gessa G. L., Meloni D., Tagliamonte A. and Stefanini E. (1993) Reduced [3H]SCH 23390 binding and DA-sensitive adenylyl cyclase in the limbic system of ethanol-preferring rats. Alcohol Alcohol. 28, 397-400.

Detering N., Collins R. M. Jr., Hawkins R. L., Ozand P. T. and Karahasan A. (1980) Comparative effects of ethanol and malnutrition on the development of catecholamine neurons: changes in norepinephrine turnover. J. Neuro- chem. 34, 1788-1791.

Deutsch J. A. and Walton N. Y. (1977) Diazepam main-

tenance of alcohol preference during alcohol withdrawal. Science 198, 307-309.

Diana M., Pistis M., Carboni S., Gessa G. L. and Rossetti Z. L. (1993) Profound decrement of mesolimbic dopa- minergic neuronal activity during ethanol withdrawal syn- drome in rats: electrophysiological and biochemical evidence. Proc. natn. Acad. Sci. U.S.A. 90, 7966-7969.

Di Chiara G. and Imperato A. (1988) Drugs abused by humans preferentially increase synaptic dopamine con- centrations in the mesolimbic system of freely moving rats. Proc. natn. Acad. Sci. U.S.A. 85, 5274~5278.

Dildy-Mayfield J. E. and Harris R. A. (1992) Comparison of ethanol sensitivity of rat brain kainate, OL-Ct-amino- 3-hydroxy-5-methyl-4-isoxalone proprionic acid and N- methyl-D-aspartate receptors expressed in Xenopus oocy- tes. J. Pharmac. exp. Ther. 262, 487-494.

Dillon K. A., Gross-lsseroff R., Israeli M. and Biegon A. (1991) Autoradiographic analysis of serotonin 5-HT~A receptor binding in the human brain postmortem : effects of age and alcohol. Brain Res. 554, 56454.

Dingledine R., Hynes M. A. and King G. L. (1986) Involve- ment of N-methyl-D-aspartate receptors in epileptiform bursting in the rat hippocampal slice. J. Physiol. (Lond.) 380, 175-189.

Docherty J. M., Harding S. and Grupp L. A. (1993) Reduction of ethanol intake by aerosol inhalation of a fl- adrenergic agonist : new route new treatment approach? Alcohol. : Clin. exp. Res. 17, 1051-1054.

Drachman D. A. (1977) Cognitive function in man. Does the cholinergic system have a special role? Neurology 27, 738-790.

Durcan M. J., Lister R. G. and Linnoila M. (1991) Evidence for central u2-adrenoceptors, not imidazoline binding sites, mediating the ethanol-attenuating properties of ct2-adreno- ceptor antagonists. J. Pharmac. exp. Ther. 258, 576-582.

Durcan M. J., Lister R. G. and Linnoila M. (1992) The effects of two different inhibitors of PNMT and their inter- actions with ethanol. NeuroReport 3, 349-352.

Dyr W., McBride W. J., Lumeng L., Li T.-K. and Murphy J. M. (1993) Effects of D~ and D 2 dopamine receptor agents on ethanol consumption in the high-alcohol-drink- ing (HAD) line of rats. Alcohol 10, 207-212.

Engberg B. and Hajos M. (1992) Ethanol attenuates the response of locus coeruleus neurons to excitatory amino acid agonists in vivo. Naunyn-Schmiedeberg's Arch. Phar- mac. 345, 222-226.

Engel J. A. and Liljequist S. (1983) The involvement of different central neurotransmitters in mediating the stimu- latory and sedative effects of ethanol. In : Stress and Alco- hol Use (Pohorecky L. A. and Brick J., eds), pp. 153-169. Elsevier Science Publishing, New York.

Eriksson K. (1968) Genetic selection for voluntary alcohol consumption in the albino rat. Science 159, 739-741.

Eriksson K. and Rusi M. (1981) Finnish selection studies on alcohol-related behaviors: general outline. In: Develop- ment o f Animal Models as Pharmacogenetic Tools (McClearn G. E., Deitrich R. A. and Erwin G., eds), Proc. Workshop, Boulder, Colorado, 1978, NIAAA Research Monograph No. 6, pp. 87-117. U.S. Government Printing Office, Washington.

Ernouf D., Daoust M., Poulain D. and Narcisse G. (1992) Triptosine, an L-5-hydroxytryptophan derivative, reduces alcohol consumption in alcohol-preferring rats. Alcohol Alcohol. 27, 273-276.

Ernouf D., Compagnon P., Lothion P., Narcisse G., Brnard

330 Invited Review

J. Y. and Daoust M. (19933 Platelets [~H]5-HT uptake in descendants from alcoholic patients : a potential risk factor for alcohol dependence? Lile Sci. 52, 989 995.

Erwin V. G., Radcliffe R., Hinkle B. and Jones B. C. ( 19933 Genetic-based differences in neurotensin levels and recep- tors in brains ofLS X SS mice. Peptides 14, 821 828.

Fadda F., Mosca E., Colombo G. and Gessa G. L. (1989) Effect of spontaneous ingestion of ethanol on brain dopa- mine metabolism. Life Sci. 44, 281 287.

Fadda F., Garau G., Marchei F., Colombo G. and Gessa G. L. (1991) MDL 72222, a selective 5-HT, receptor antag- onist, suppresses voluntary ethanol consumption in alco- hol-preferring rats. Alcohol Ah'ohol. 26, 107 I10.

Feldstein A., Hoagland H., Wong K. and Freeman H. (1964) Biogenic amines, biogenic aldehydes, and alcohol. Quart. J. Studies Ah'oho125, 218-225.

Ferriera L. and Soares-da-Silva P. (19913 5-Hydroxy- tryptamine and alcoholism. Human P~3'chopharmac. 6, $21-24.

Forsythe I. D., Westbrook G. L. and Mayer M. L. (19883 Modulation of excitatory synaptic transmission by glycine and zinc in cultures of mouse hippocampal neurons. J. Neurosci. 8, 3733 3741.

Fozard J. R. (1992) Pharmacological relevance of 5-HT, receptors. Int. Acad. Biomed. Drug. Res. 1, 45 55.

Frankel D., Khanna J. M., LeBlanc A. E. and Kalant H. (1975) Effect of p-chlorophenylalanine on the acquisition of tolerance to ethanol and pentobarbital. Psycho- pharmacology 44, 246-252.

Freund G. and Ballinger W. E. (1988) Decrease of benzo- diazepine receptors in frontal cortex of alcoholics. Alcohol 5, 275 282.

Freund G. and Ballinger W. E. (19893 Loss of muscarinic and benzodiazepine neuroreceptors from hippocampus of alcohol abusers. Ah'ohol Alcohol. 6, 23 31.

Freund G. and Ballinger W. E. (1991) Loss o1 synaptic receptors can precede morphologic changes induced by alcoholism. Alcohol Ah.ohol. Suppl. 1, 385 391.

Frey H. H., Magnussen M. P. and Nielsen C. K. (1970) The effect of p-chloroamphetamine on the consumption of ethanol by rats. Arch. Int. Pharmacod.vn. 183, 165 172.

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.

Frye G. D., Vogel R. A., Mailman R. B., Ondrusek M. G., Wilson J. H., Mueller R. A. and Breese G. R. (1980) A comparison of behavioral and neurochemical effects of ethanol and chlordiazepoxide. In : Ah'ohol and Aldehyde Metabolizin9 ,~vstems (Thurman R. D., ed.), pp. 727 737, Vol. 4. Plenum Press, New York.

Fuchs V., Coper H. and Rommelspacher H. (1987) The effect of ethanol and haloperidol on dopamine receptor (D2) density. Neuropharmacology 26, 1231-1233.

Fuller J. L. (1964) Measurement of alcohol preference in genetic experiments. J. comp. Physiol. Psychol. 57, 85 88.

Gallaher E. J. and Gionet S. E. (1988) Initial sensitivity and tolerance to ethanol in mice genetically selected for diazepam sensitivity. Ah.ohol. : Clin. exp. Res. 12, 77 80.

Gatto G. J., Murphy J. M., McBride W. J., Lumeng L. and Li T.-K. (1990) I ntracranial self-administration of ethanol into the ventral tegmental area of alcohol-preferring (P) rats. Ah'ohol. : Clin. exp. Res. 14, 291.

Gejman P. V., Ram A., Gelernter J.. Friedman E., Cao Q, Pickar D., Blum K., Noble E. P., Kranzler H. R., O'Malley S., Hamer D. H., Whitsitt F., Rao P., DeLisi L. E., Virk-

kunen M., Linnoila M., Goldman D. and Gershon E. S. (1994) No structural mutation in the dopamine D2 recep- tor gene in alcoholism or schizophrenia: analysis using denaturing gradient gel electrophoresis. JAMA 271,204- 208.

Gelernter J., Goldman D. and Risch N. (1993) The AI allele at the D2 dopamine receptor gene and alcoholism. JAMA 269, 1673 1677.

George D. T., Benkelfat C., Murphy D. L., Schmitz J. and Linnoila M. (1990) Ethanol-like behavioral response in abstinent alcoholics infused with the 5-HT agonist mCPP. Biol. Ps~Thiat. 27, 176A.

Gessa G. L., Muntoni F., Collu M., Vargiu L. and Mereu G. (1985) Low doses of ethanol activate dopaminergic neurons in the ventral tegmental area. Brain Res. 348, 201 203.

Gianoulakis C. (1983) Long-term ethanol alters the binding of [~H]-opiates to brain membranes. Life Sci. 33, 725-733.

Gianoulakis C., De Waele J. P. and Kiianmaa K. (1992) Differences in the brain and pituitary fl-endorphin system between the alcohol-preferring AA and alcohol-avoiding ANA rats. Alcohol. : Clin. exp. Res. 16, 453--459.

Gill K. and Amit Z. (1989) Serotonin uptake blockers and voluntary alcohol consumption. A review of recent studies. In: Recent Developments in Alcoholism (Galanter M., ed.), pp. 225 248, Vol. 4. Plenum Press, New York.

Gongwer M. A., Murphy J. M., McBride W. J., Lumeng L. and Li T.-K. (1989) Regional brain contents of serotonin, dopamine and their metabolites in the selectively bred high- and low-alcohol drinking lines of rats. Ah'ohol 6, 317 320.

Gonzatez L. P. (1993) Sensitivity to strychnine seizures is unaltered during ethanol withdrawal. Alcohol. : Clin. exp. Res. 17, 1029 1034.

Grant K. A. and Barrett J. E. (1991) Blockade of the dis- criminative stimulus effects of ethanol with 5-HT3 receptor antagonists. Pschopharmacoloyy 104, 451~456.

Grant K. A. and Colombo G. (1992) Discriminative stimulus effects of ethanol: effect of training dose on the sub- stitution of N-methyl-o-aspartate antagonists. J. Pharmac. exp. Ther. 264, 1241-1247.

Grant K. A. and Colombo G. (19933 Substitution of the 5-HT~ agonist trifluoromethylphenylpiperazine (TFMPP) for the discriminative stimulus effects of ethanol: effect of training dose. Psychopharmacology 113, 26-30.

Grant K. A., Knisely J. S., Tabakoff B., Barrett J. E. and Balster R. L. (19913 Ethanol-like discriminative stimulus effects of noncompetitive N-methyl-D-aspartate antag- onists. Behav. Pharmac. 2, 87-90.

Guan X.-M. and McBride W. J. (1989) Serotonin micro- infusion into the ventral tegmental area increases accum- bens dopamine release. Brain Res. Bull. 23, 541-547.

Hadjivanova K., Petkov V. D., Alova L., Petkov V. V. and Vuglenova Y. (1991) Changes in brain muscarinic and beta-adrenoreceptors of the offspring of ethanol-con- suming mothers (experiments on rats). Acta Physiol. Phar- mac. Bulg. 17, 91-97.

Hamdi A. and Prasad C. (1992) Bidirectional changes in striatal D~-dopamine receptor density during chronic etha- nol intake. Life Sci. 52, 251-257.

Haraguchi M., Samson H. H. and Tolliver G. A. (1990) Reduction in oral ethanol self-administration in the rat by the 5-HT uptake blocker fluoxetine. Pharmac. Biochem. Behat,. 35, 25~262.

Harris E. W., Ganong A. H. and Cotman C. W. (1984)

Invited Review 331

Long-term potentiation in the hippocampus involves acti- vation of N-methyl-D-aspartate receptors. Brain Res. 323, 132-137.

Harris R. A. and Allan A. M. (1989) Alcohol intoxication : ion channels and genetics. F A SE B J. 3, 1689-1695.

Harris R. A., Brodie M. S. and Dunwiddie T. V. (1992) Possible substrates of ethanol reinforcement : GABA and dopamine. Ann. N Y Acad. Sci. 654, 61-69.

Hellevuo K., Kiianmaa K. and Kim C. (1990) Effect of ethanol on brain catecholamines in rat lines developed for differential ethanol-induced motor impairment. Alcohol 7, 159-163.

Hellstr0m-Lindahl E., Winblad B. and Nordberg A. (1993) Muscarinic and nicotinic receptor changes in the cortex and thalamus of brains of chronic alcoholics. Brain Res. 620, 42-48.

Herv6 D., Pickel V. M., Joh T. H. and Beaudet A. (1987) Serotonin axon terminals in the ventral tegmental area of the rat : fine structure and synaptic input to dopaminergic neurons. Brain Res. 435, 71-83.

Hirouchi M., Hashimoto T. and Kuriyama K. (1993) Alter- ation of GABAA receptor ~-subunit mRNA in mouse brain following continuous ethanol inhalation. Eur. J. Pharmac. 247, 127-130.

Hodge C. W., Samson H. H. and Haraguchi M. (1992) Microinjections of dopamine agonists in n. accumbens increase ethanol reinforced responding. Pharmac. Biochem. Behav. 43, 249-254.

Hodge C. W., Haraguchi M., Erickson H. and Samson H. H. (1993) Ventral tegmental microinjections of quinpirole decrease ethanol and sucrose-reinforced responding. Alco- hol. : Clin. exp. Res. 17, 370-375.

Hodges H., Allen Y., Sinden J., Mitchell S. N., Arendt T., Lantos P. L. and Gray J. A. (1991) The effects of chol- inergic drugs and cholinergic-rich foetal neural transplants on alcohol-induced deficits in radial maze performance in rats. Behav. Brain Res. 43, 7 28.

Hoffman P. L., Rabe C. S., Moses F. and Tabakoff B. (1989) N-methyl-D-aspartate receptors and ethanol : inhibition of calcium flux and cyclic GMP production. J. Neurochem. 52, 1937-1940.

Hruska R. E. (1988) Effect of ethanol administration on striatal D~ and D2 dopamine receptors. J. Neurochem. 50, 1929-1933.

Hu G., Querimit L. A., Downing L. A. and Charness M. E. (1993) Ethanol differentially increases 72-adrenergic and muscarinic acetylcholine receptor gene expression in NG108-15 cells. J. biol. Chem. 268, 23441-23447.

Hubbell C. L., Marglin S. H., Spitalnic S. J., Abelson M. L., Wild K. D. and Reid L. D. (1991) Opioidergic, sero- tonergic, and dopaminergic manipulations and rats' intake of a sweetened alcoholic beverage. Alcohol 8, 355-367.

Hubbell C. L., Mankes R. F. and Reid L. D. (1993) A small dose of morphine leads rats to drink more alcohol and achieve higher blood alcohol concentrations. Alcohol. : Clin. exp. Res. 17, 104(~1043.

Humeniuk R. E., White J. M. and Ong J. (1993) The role of GABAa receptors in mediating the stimulatory effects of ethanol in mice. Psychopharmacology 111, 219-224.

Humphrey P. P. A., Hartig P. and Hoyer D. (1993) A pro- posed new nomenclature for 5-HT receptors. Trends Phar- mac. Sci. 14, 233-236.

Hunt W. A. (1990) Brain mechanisms that underlie the rein- forcing effect of ethanol. In : Why People Drink (Cox W. M., ed.), pp. 51 83, Vol. 4. Gardner Press Inc., New York.

Hunt W. A. (1993) Neuroscience research: how has it con- tributed to our understanding of alcohol abuse and alcoholism? A review. Alcohol. : Clin. exp. Res. 17, 1055 1065.

Huttunen P. (1991) Microdialysis of extracellular nor- adrenaline in the hippocampus of the rat after long-term alcohol intake. Brain Res. 560, 225 228.

Hyyti~ P. (1993) Involvement of #-opioid receptors in alco- hol drinking by alcohol-preferring AA rats. Pharmac. Biochem. Behav. 45, 697-701.

Hyyti~i P. and Sinclair J. D. (1993) Responding for oral ethanol after naloxone treatment by alcohol-preferring AA rats. Alcohol. : Clin. exp. Res. 17, 631~536.

Imperato A., Di Chiara G. (1986) Preferential stimulation of dopamine release in the nucleus accumbens of freely moving rats by ethanol. J. Pharmac. exp. Ther. 239, 219- 228.

Iorio K. R., Reinlib L., Tabakoff B. and Hoffman P. L. (1992) Chronic exposure of cerebellar granule cells to etha- nol results in increased N-methyl-D-aspartate receptor function. Molec. Pharmac. 41, 1142-1148.

Jiang L. H., Ashby C. R. Jr, Kasser R. J. and Wang R. Y. (1990) The effect of intraventricular administration of the 5-HT 3 receptor agonist 2-methylserotonin on the release of dopamine in the nucleus accumbens : an in vivo chrono- coulometric study. Brain Res. 513, 156--160.

Kalant H. (1977) Comparative aspects of tolerance to, and dependence on, alcohol, barbiturates and opiates. In: Alco- hol Intoxication and Withdrawal (Gross M. M., ed.), pp. 169-185, Vol. 11 lB. Plenum Press, New York.

Kelly P. H., Seviour P. W. and Iversen S. D. (1975) Amphet- amine and apomorphine responses in the rat following 6- OHDA lesions of the nucleus accumbens septi and corpus striatum. Brain Res. 94, 507-522.

Khanna J. M., L~ A. D., Kalant H. and LeBlanc A. E. (1979) Cross-tolerance between ethanol and morphine with respect to their hypothermic effects. Eur. J. Pharmac. 59, 145-149.

Kiianmaa K., Fuxe K., Jonsson G. and Ahtee L. (1975) Evidence for involvement of central NA neurones in alco- hol intake. Increased alcohol consumption after degener- ation of the NA pathway to the cortex cerebri. Neurosci. Lett. 1, 41~45.

Knapp D. J. and Pohorecky L. A. (1992) Zacopride, a 5- HT 3 receptor antagonist, reduces voluntary ethanol con- sumption in rats. Pharmac. Biochem. Behav. 41, 847-850.

Kono H., Link Y. C., Zuspan F. P., Lee A. C. and Yajima A. (1993) Effect of ethanol and progesterone on monoamine oxidase activity in cultured cells of human term placenta. Am. J. Obstet. Gynec. 161, 136-140.

Korpi E. R., Sinclair J. D. and Malminen O. (1987) Dopa- mine D2 receptor binding in striatal membranes of rat lines selected for differences in alcohol-related behaviors. Pharmac. Toxic. 61, 94-97.

Korpi E. R., Sinclair J. D., Kaheinen P., Vitamaa T., Hel- levuo K. and Kiianmaa K. (1988) Brain regional and adrenal monoamine concentrations and behavioral responses to stress in alcohol-preferring AA and alcohol- avoiding ANA rats. Alcohol 5, 417-425.

Korpi E. R., Uusi-Oukari M., Castren E., Suzdak P. D., Sepp~il~i T., Sarviharju M. and Tuominen K. (1992a) Cer- ebellar GABAA receptors in two rat lines selected for high and low sensitivity to moderate alcohol doses: phar- macological and genetic studies. Alcohol 9, 225-231.

Korpi E. R., Paivarinta P., Abi-Dargham A., Honkanen A.,

332 Invited Review

Laruelle M., Tuominen K. and Hilakivi L. A. (1992b) Binding of serotonergic ligands to brain membranes of alcohol-preferring AA and alcohol avoiding ANA rats. Alcohol9, 369 374.

Kostowski W. and Dyr W. (1992) Effects of 5-HT~A receptor agonists on ethanol preference in the rat. Alcohol 9, 283 286.

Kostowski W., Dyr W. and Krzascik P. (1993) The abilities of 5-HT3 receptor antagonist ICS 205-930 to inhibit alco- hol preference and withdrawal seizures in rats, Alcohol 10, 369 373.

Krasner N., Moore M. R., Dow J., Thompson G. G. and Goldberg A. (1975) Fenfluramine and 780SE in exper- imental alcoholism in rats. Postgrad. Med. J. 51, 76 79.

Kulkosky P. J. and Glazner G. W. (1988) Dose-additive inhibition of intake of ethanol by cholecystokinin and bombesin. Ak'ohol. : Clin. evp. Res. 12, 277 281.

Kulkosky P. J., Sanchez M. R., Foderaro M. A. and Chiu N. (1989) Cholecystokinin and satiation with alcohol. Alcohol 6, 395402.

Kulkosky P. J., Clayborne Y. J. and Sandoval S. L. (1993) Cholecystokinin and bombesin inhibit ethanol and food intake in rats selectively bred for ethanol sensitivity. Alco- hol. : Clin. exp. Res. 17, 545 551.

Lamblin F., Deuceuninck D. and De Witte P. (1993) Modu- lation of alcohol preference by NMDA antagonists in male rats. Alcohol Ah'ohol. 28, 639-647.

Lanqa A. J. (1994) Reduction of voluntary alcohol intake in the rat by modulation of the dopaminergic mesolimbic system : transplantation of ventral mesencephalic cell sus- pensions. Neuroscience 58, 359 369.

L6 A. D., Khanna J. M., Kalant H. and LeBlanc A. E. (1979) Effect of tryptophan on the acquisition of tolerance to ethanol-induced motor impairment and hypothermia. Psychopharmacolo#y 61, 125 129.

L6 A. D., Khanna J. M., Kalant H. and LeBlanc A. E. ( 1981 a) Effect of modification of brain serotonin (5-HT). norepinephrine (NE) and dopamine (DA) on ethanol tol- erance. Psychopharmacology 75, 231 235.

L6 A. D., Khanna J. M., Kalant H. and LeBlanc A. E (1981b) The effect of lesions in the dorsal, median and magnus raphe nuclei on the development of tolerance to ethanol. J. Pharmac. exp. Ther. 218, 525 529.

L6 A. D., Khanna J. M., Kalant H. and Grossi E. (1986) Tolerance to and cross-tolerance among ethanol, pento- barbital and chlordiazepoxide. Pharmac. Biochem. Behav. 24, 93-98.

Leander J. D. (1987) Fluoxetine suppresses palatability- induced ingestion. Psychopharmacology 91,285 287.

Levy A. D., Murphy J. M., McBride W. J., Lumeng L. and Li T.-K. ( 1991 ) Microinjection of sulpiride into the nucleus accumbens increases ethanol drinking in alcohol-pre- ferring (P) rats. Alcohol Ah'ohol. Sttl~pl. 1,417~420.

Liljeberg P., Mossberg D. and Borg S. (1987) Clinical con- ditions and central noradrenergic activity in long term abstinent alcoholic patients. Alcohol Ah'ohol. Suppl. 1, 615~18.

Liljequist S. (1991) The competitive NMDA receptor-antag- onist, CGP 39551, inhibits ethanol withdrawal seizures. Eur. J. Pharmac. 192, 197 198.

Link A. M.-Y., Bickford P. C., Palmer M. R. and Gerhardt G. A. (1993) Ethanol inhibits the uptake of exogenous norepinephrine from the extracellular space of the rat cer- ebellum. Neurosci. Lett. 164, 71 75.

Lishman W. A. (1990) Alcohol and the brain. Br. J. Psychiat. 156, 635- 644.

Lograno D. E., Matteo F., Trabucchi M., Govoni S., Cag- iano R., Lacomba C. and Cuomo V. (1993) Effects of chronic ethanol intake at a low dose on the rat brain dopaminergic system. Alcohol 10, 45~49.

Lovinger D. M. (1993a) Excitotoxicity and alcohol-related brain damage. Alcohol. : Clin. exp. Res. 17, 19-27.

Lovinger D. M. (1993b) High ethanol sensitivity of recom- binant AMPA-type glutamate receptors expressed in mammalian cells. Neurosci. Lett. 159, 83-87.

Lovinger D. M. and White G. (1991) Ethanol potentiation of 5-hydroxytryptamine3 receptor-mediated ion current in neuroblastoma cells and isolated adult mammalian neurons. Mol. Pharmac. 40, 263 270.

Lovinger D. M., White G. and Weight F. F. (1989) Ethanol inhibits NMDA-activated ion current in hippocampal neurons. Science 243, 1721 1724.

Lovinger D. M., White G. and Weight F. F. (1990) NMDA receptor-mediated synaptic excitation selectively inhibited by ethanol in hippocampal slice from adult rat. J. Neurosci. 10, 1372-1379.

Lu M. R., Wagner G. C. and Fisher H. (1993) Ethanol consumption following acute fenfluramine, fluoxetine, and dietary tryptophan. Pharmae. Biochem. Behav. 44, 931 937.

Lucchi L., Moresco R. M., Govoni S. and Trabucchi M. (1988) Effect of chronic ethanol treatment on dopamine receptor subtypes in rat striatum. Brain Res. 449, 347-351.

Lumeng L., Hawkins T. D. and Li T.-K. (1977) New strains of rats with alcohol preference and nonpreference. In: Alcohol and Aldehyde Metabolizing Systems (Thurman R. G., Williamson J. R., Drott H. R. and Chance B., eds), pp. 537 544, Vol. 3. Academic Press, New York.

Lumeng L., Doolittle D. P. and Li T.-K. (1986) New dupli- cate lines of rats that differ in voluntary alcohol consump- tion. Alcohol Alcohol. 21, A125.

Lustig H. S., Chan J. and Greenberg D. A. (1992) Ethanol inhibits excitotoxicity in cerebral cortical cultures. Neuro- sci. Lett. 135, 259-261.

Mancillas J. R., Siggins G. R. and Bloom F. E. (1986) Sys- temic ethanol: selective enhancement of responses to acetylcholine and somatostatin in hippocampus. Science 231, 161 163.

Mardones J. (1960) Experimentally induced changes in the free selection of ethanol. Int. Ret,. Neurobiol. 2, 41-76.

Marley R. J., Freund R. K. and Wehner J. M. (1988) Differ- ential response to flurazepam in long-sleep and short-sleep mice. Pharmac. Biochem. Behav. 31,453~458.

Martin D., Savage D. D. and Swartzwelder H. S. (1992) Effects of prenatal ethanol exposure on hippocampal ion- otropic-quisqualate and kainate receptors. Alcohol. : Clin. exp. Res. 16, 81(~821.

May T. (1992) Striatal dopamine Drlike receptors have higher affinity for dopamine in ethanol-treated rats. Eur. J. Pharmac. 215, 313 316.

Maynard L. S. and Schenker V. (1962) Monoamine oxidase inhibition by ethanol in vitro. Nature 196, 575-576.

McBride W. J., Murphy J. M., Lumeng L. and Li T.-K. (1988) Effects of Ro 15-4513, fluoxetine and desipramine on the intake of ethanol, water and food by the alcohol- preferring (P) and -nonpreferring (NP) lines of rats. Phar- mat'. Biochem. Behat~. 30, 1045-1050.

McBride W. J., Murphy J. M., Lumeng L. and Li T.-K. {1990) Serotonin, dopamine and GABA involvement in

Invited Review 333

alcohol drinking of selectively bred rats. Alcohol 7, 199- 205.

McBride W. J., Chernet E., Dyr W., Lumeng L. and Li T.- K. (1993a) Densities of dopamine D2 receptors are reduced in CNS regions of alcohol-preferring P rats. Alcohol 10, 387-390.

McBride W. J., Chernet E., Rabold J. A., Lumeng L. and Li T.-K. (1993b) Serotonin-2 receptors in the CNS of alco- hol-preferring and -nonpreferring rats. Pharmac. Biochem. Behav. 46, 631-636.

McClearn G. E. and Kakihana R. (1973) Selective breeding for ethanol sensitivity in mice. Behav. Genet. 3, 409-410.

Meana J. J., Gabilondo A. M., Barturen F. and Garcia- Sevilla J. A. (1992) Acute ethanol intoxication may not alter ct2-adrenoceptors in the human brain. Psycho- pharmacology 107, 132-134.

Meert T. F. (1993) Effects of various serotonergic agents on alcohol intake and alcohol preference in Wistar rats selected at two different levels of alcohol preference. Alco- hol Alcohol. 28, 157-170.

Meller S. T., Dykstra C., Pechman P. S., Maves T. J. and Gebhart G. F. (1993) Ethanol dose-dependently atten- uates NMDA-mediated thermal hyperalgesia in the rat. Neurosci. Lett. 154, 137-140.

Mello N. (1972) Behavioral studies of alcoholism. In: The Biology of Alcoholism (Kissin B. and Begleiter H., eds), pp. 219-291, Vol. 2. Plenum Press, New York.

Mereu G., Fadda F. and Gessa G. L. (1984) Ethanol stimu- lates the firing rate of nigral dopaminergic neurons in unanesthetized rats. Brain Res. 292, 63-69.

Mhatre M. and Ticku M. J. (1989) Chronic ethanol treat- ment selectively increases the binding of inverse agonists for benzodiazepine binding sites in cultured spinal cord neurons. J. Pharmac. exp. Ther. 251, 164-168.

Mhatre M., Mehta A. K. and Ticku M. K. (1988) Chronic ethanol administration increases the binding of the benzo- diazepine inverse agonist and alcohol antagonist [3H]Ro 15-4513 in rat brain. Eur. J. Pharmac. 153, 141-145.

Mhatre M. C., Pena G., Sieghart W. and Ticku M. K. (1993) Antibodies specific for GABAa receptor ct subunits reveal that chronic alcohol treatment down-regulates ct-subunit expression in rat brain regions. J. Neurochem. 61, 1620- 1625.

Michaelis E. K., Mulvaney M. J. and Freed W. J. (1978) Effects of acute and chronic ethanol intake on syn- aptosomal glutamate binding activity. Biochem. Pharmac. 27, 1685-1691.

Monti J. M. and Alterwain P. (1991) Ritanserin decreases alcohol intake in chronic alcoholics (letter). Lancet 337, 60.

Montpied P., Morrow A. L., Karanian J. W., Ginns E. I., Martin B. M. and Paul S. M. (1991) Prolonged ethanol inhalation decreases 7-aminobutyric acidA receptor ct sub- unit mRNAs in the rat cerebral cortex. Mol. Pharmac. 39, 157-163.

Morinan A. (1987) Reduction in striatal 5-hydroxy- tryptamine turnover following chronic administration of ethanol to rats. Alcohol Alcohol. 22, 53-60.

Morrisett R. A. and Swartzwelder H. S. (1993) Attenuation of hippocampal long-term potentiation by ethanol: a patch-clamp analysis of glutamatergic and GABAergic mechanisms. J. Neurosci. 13, 2264-2272.

Morrow A. L., Montpied P., Ginns E. I., Martin B. M. and Paul S. M. (1989) Acute ethanol administration decreases

the level of GABA receptor alpha-subunit mRNAs in rat cerebral cortex. Soc. Neurosci. Abstr. 15, 488.

Mosaddeghi M., Burke T. F. and Moerschbaecher J. M. (1992) Effects of acute administration of alcohol on in vivo binding of [3H]Ro 15-1788 to mouse brain. J. Pharmac. exp. Ther. 263, 639-645.

Murphy J. M., McBride W. J., Lumeng L. and Li T.-K. (1987) Contents of monoamines in forebrain regions of alcohol-preferring (P) and non-preferring (NP) lines of rats. Pharmac. Biochem. Behav. 26, 389-392.

Murphy J. M., Waller M. B., Gatto G. J., McBride W. J., Lumeng L. and Li T.-K. (1988) Effects of fiuoxetine on the intra-gastric self-administration of ethanol in the alcohol- preferring P line of rats. Alcohol 5, 283~86.

Myers R. D. and Lankford M. F. (1993) Failure of the 5- HT2 receptor antagonist, ritanserin, to alter preference for alcohol in drinking rats. Pharmac. Biochem. Behav. 45, 233-237.

Myers R. D. and Melchior C. L. (1975) Alcohol drinking in the rat after destruction of serotonergic and catechol- aminergic neurons in the brain, Chem. Path. Pharmac. 10, 363-378.

Myers R. D. and Quarfordt S. (1991) Alcohol drinking atten- uated by sertraline in rats with 6-OHDA or 5,7-DHT lesions of N. accumbens: a caloric response? Pharmac. Biochem. Behav. 40, 923-928.

Myers R. D. and Veale W. L. (1968) Alcohol preference in the rat : reduction following depletion of brain serotonin. Science 16tl, 1469-1471.

Myers R. D., Evans J. E. and Yaksh T. L. (1972) Ethanol preference in the rat : interactions between brain serotonin and ethanol, acetaldeydehyde, paraldehyde, 5-HTP and 5- HTOL. Neuropharmacology 11, 539-549.

Myers R. D., Lankford M. and Bj6rk A. (1993a) Irreversible suppression of alcohol drinking in cyanamide-treated rats after sustained delivery of the 5-HT2 antagonist ampero- zide. Alcohol 10, 117-125.

Myers R. D., Lankford M. F. and Bj6rk A. (1993b) 5- HT2 receptor blockade by amperozide suppresses ethanol drinking in genetically preferring rats. Pharmac. Biochem. Behav. 45, 741 747.

Nair N. P. V., Lal S., Iskandar H. I., Etienne P., Wood P. N. and Guyda H. (1982) Effect of sulpiride, an atypical neuroleptic, on apomorphine-induced growth hormone secretion. Brain Res. Bull. 8, 587-591.

Naranjo C. A., Sellers E. M. and Larwin M. O. (1986) Modulation of ethanol intake by serotonin uptake inhibi- tors. J. clin. Psychiat. 47 (Suppl.), 16-22.

Naranjo C. A., Poulos C. X., Bremner K. E. and Lanct6t K. L. (1992) Citalopram decreases desirability, liking, and consumption of alcohol in alcohol-dependent drinkers. Clin. Pharmac. Ther. 51, 729-739.

Nestoros J. (1980) Ethanol specifically potentiates GABA- mediated neurotransmission in feline cerebral cortex. Sci- ence 2119, 708-710.

Nielsen D. A., Goldman D., Virkkunen M., Tokola R., Rawlings R. and Linnoila M. (1994) Suicidality and 5-hydroxyindoleacetic acid concentration associated with a tryptophan hydroxylase polymorphism. Arch. Gen. Psychiat. 51, 34-38.

Noble E. P. (1993) The D 2 dopamine receptor gene : a review of association studies in alcoholism. Behav. Genet. 23, 119- 129.

Noble E. P. and Blum K. (1991) The dopamine D2 receptor gene and alcoholism. JAMA 265, 2667.

334 Invited Review

Noble E. P., Blum K., Ritchie T., Montgomery A. and Sheri- dan P. J. (1991) Allelic association of the D_, dopamine receptor gene with receptor binding characteristics in alcoholism. Arch. Gem Psychiat. 48, 648-656.

Oreland L., Wiberg/~. and Fowler C. J. (19813 Monoamine oxidase activity in platelets as related to monoamine oxi- dase activity and monoaminergic function in the brain, In : Recent Advances in Neurop~vchopharmacoloqy. Advances in the Biosciences (Angrist et al., eds), pp. 195 201, Vol. 3. Pergamon Press, Oxford.

Paasonen M. K. and Solatunturi E. (19653 Monoamine oxi- dase in mammalian blood platelets. Ann. Med. Exp. Biol. Fenn. 43, 98 100.

Palaic D., Desaty J., Albert J. M. and Panisset J. C. (19713 Effect of ethanol on metabolism and subcellular dis- tribution of serotonin in rat brain. Brain Res. 25, 381 386.

Palmer M. R., Wang Y., Fossom L. H. and Spuhler K. P. (1987) Genetic correlation of ethanol-induced ataxia and cerebellar Purkinje neuron depression among inbred strains and selected lines of rats. Ah'ohol. : Clin. exp. Res. I1,494-501.

Pandey S. C., Piano M. R., Schwertz D. W., Davis J. M. and Pandey G. N. (1992) Effect of ethanol administration and withdrawal on serotonin receptor subtypes and receptor- mediated phosphoinositide hydrolysis in rat brain. Ah'ohol. : Clin. exp. Res. 16, 1110 1116.

Panocka 1. and Massi M. (1992) Long-lasting suppression of alcohol preference in rats lbllowing serotonin receptor blockade by ritanserin. Brain Res. Bull. 28, 493 496.

Peoples R. W. and Weight F. F. (19923 Ethanol inhibition of N-methyl-o-aspartate-activated ion current in rat hippocampal neurons is not competitive with glycine. Brain Res. 571,342-344.

Pfeffer A. O. and Samson H. H. (1988) Hatoperidol and apomorphine effects on ethanol reinforcement in free feed- ing rats. Pharmac. Biochem. Behav. 29, 343-- 350.

Phillips T. J., Terdal E. S. and Crabbe J. C. (19903 Response to selection for sensitivity to ethanol hypothermia : genetic analyses. Behav. Genet. 20, 473 480.

Pick C. G., Cooperman M., Trombka D., Rogel-Fuchs Y. and Yanai J. (19933 Hippocampal cholinergic alterations and related behavioral deficits after early exposure to etha- nol. Int. J. dev. Neurosci. 11,379 385.

Pierce D. R., Serbus D. C. and Light K. E. (1993) Intragastric intubation of alcohol during postnatal development of rats results in selective cell loss in the cerebellum. Alcohol. : Clin. exp. Res. 17, 1275-1280.

Portas C. M.. Devoto P. and Gessa G. L. (1994) Effect of ethanol on extracellular 5-hydroxytryptamine output in rat frontal cortex. Eur. J. Pharmac. .-Era,. Toxic. Pharmac. Sect. 270, 123-125.

Quarfordt S. D., Kalmus G. W. and Myers R. D. (19913 Ethanol drinking following 6-OHDA lesions of nucleus accumbens and tuberculum olfactorium of the rat. Ak.oho/ 8, 211 217.

Queen S. A., Sanchez C. F., Lopez S. R., Paxton L. L. and Savage D. D. (1993) Dose- and age-dependent effects of prenatal ethanol exposure on hippocampal metabotropic- glutamate receptor-stimulated phosphoinositide hydroly- sis. Alcohol. : Clin. exp. Res. 17, 887-893.

Rabe C. S. and Tabakoff B. (1990) Glycine site-directed agonists reverse the actions of ethanol at the N-methyl-D- aspartate receptor. Mol. Pharmac. 38, 753 757.

Rajachandran L., Spear N. E. and Spear L. P. (19933 Effects of the combined administration of the 5-HT~ antagonist

MDL 72222 and ethanol on conditioning in the peri- adolescent and adult rat. Pharmac. Biochem. Behav. 46, 535 542.

Rassnick S., Stinus L. and Koob G. F. (1993a) The effects of 6-hydroxydopamine lesions of the nucleus accumbens and the mesolimbic dopamine system on oral self-adminis- tration of ethanol in the rat. Brain Res. 623, 16-24.

Rassnick S., Pulvirenti L. and Koob G. F. (1993b) SDZ- 205,152, a novel dopamine receptor agonist, reduces oral ethanol self-administration in rats. Ah'ohol 10, 127-132.

Reid L, D., Delconte J. D., Nichols M. L., Bilsky E. J. and Hubbell C. L. (1991) Tests of opioid deficiency hypotheses of alcoholism. Alcohol 8, 247 257.

Riley J. N. and Walker D. W. (1978) Morphological alter- ations in hippocampus after long-term alcohol con- sumption in mice. Science 201,646-648.

Rommelspacher H., Wolffgramm J. and Widjaja S. (1989) Effects of desipramine on rat behavior are prevented by concomitant treatment with ethanol. Pharmac. Biochem. Behav. 32, 533 542.

Rommelspacher H., Raeder C., Kaulen P. and Brfining G. (1992) Adaptive changes of dopamine-D2 receptors in rat brain following ethanol withdrawal : a quantitative auto- radiographic investigation. Alcohol 9, 355 362.

Rossetti Z. L., Hmaidan Y. and Gessa G. L. (1992a) Marked inhibition of mesolimbic dopamine release: a common feature of ethanol, morphine, cocaine and amphetamine abstinence in rats. Eur. J. Pharmac. 221,227 234.

Rossetti Z. L., Longu G., Mercuro G., Hmaidan Y. and Gessa G. L. (1992b) Biphasic effect of ethanol on nor- adrenaline release in the frontal cortex of awake rats. Ah'ohol Alcohol. 27, 477480.

Rothberg B. S., Yasuda R. P., Satkus S. A., Wolfe B. B. and Hunter B. E. (1993) Effects of chronic ethanol on cholinergic actions in rat hippocampus: electro- physiological studies and quantification of m~-m5 mus- carinic receptor subtypes. Brain Res. 631, 227-234.

Samson H. H., Hodge C. W., Tolliver G. A. and Haraguchi M. (1993) Effect of dopamine agonists and antagonists on ethanol-reinforced behavior: the involvement of the nucleus accumbens. Brain Res. Bull. 30, 133 141.

Savage D. D., Queen S. A., Sanchez C. F., Paxton L. L., Mahoney J. C., Goodlett C. R. and West J. R. (1992) Prenatal ethanol exposure during the last third of gestation in rat reduces hippocampal NMDA agonist binding site density in 45-day-old offspring. Alcohol 9, 3741.

Schreiber R., Opitz K., Glaser T. and DeVry J. (1993) lpsa- pirone and 8-OH-DPAT reduce ethanol preference in rats : involvement of presynaptic 5-HT~A receptors. Psycho- pharmacology 112, 100-110.

Schuckit M. A., Shaskan E., Duby J., Vega R. and Moss M. (19823 Platelet monoamine oxidase activity in relatives of alcoholics. Arch. Gen. Psychiat. 39, 137--140.

Seeman P. (1972) The membrane actions of anesthetics and tranquilizers. Pharmac. Rev. 24, 583-655.

Seeman P. and Lee T. (1974) The dopamine-releasing actions of neuroleptics and ethanol. J. Pharmac. exp. Ther. 190, 131 135.

Sellers E. M., Higgins G. A. and Sobell M. B. (19923 5-HT and alcohol abuse. Trends Pharmac. Sci. 13, 69 75.

Shafik E. N., Aiken S. P. and McArdle J. J. (1991) Regional catecholamine levels in brains of normal and ethanol-tol- erant long-sleep and short-sleep mice. Brain Res. 563, 44- 48.

Shen R.-Y. and Chiodo L. A. (19933 Acute withdrawal after

Invited Review 335

repeated ethanol treatment reduces the number of spon- taneously active dopaminergic neurons in the ventral teg- mental area. Brain Res. 622, 289-293.

Simantov R. (1993) Chronic morphine alters dopamine transporter density in the rat brain: possible role in the mechanism of drug addiction. Neurosci. Lett. 163, 121 124.

Simonsson P. and Ailing C. (1988) The 5-hydroxytryptamine stimulated formation of inositol monophosphate is inhibited in platelets from alcoholics. Life Sci. 42, 385- 391.

Simonsson P., Berglund M., Oreland L., Moberg A.-L. and Ailing C. (1992) Serotonin-stimulated phosphoinositide hydrolysis in platelets from post-withdrawal alcoholics. Alcohol Alcohol. 27, 607~512.

Simson P. E., Criswell H. E. and Breese G. R. (1991) Ethanol potentiates GABA-mediated inhibition in the inferior col- liculus : evidence for local ethanol/GABA interactions. J. Pharmac. exp. Ther. 259, 1288-1293.

Simson P. E., Criswell H. E. and Breese G. R. (1993) Inhi- bition of NMDA-evoked electrophysiological activity by ethanol in selected brain regions: evidence for ethanol- sensitive NMDA-evoked responses. Brain Res. 607, 9-16.

Singh G. K., Kalamus G. W., Bjrrk A. K. and Myers R. D. (1993) Alcohol drinking in rats is attenuated by the mixed 5-HTt agonist/5-HT2 antagonist FG 5893. Alcohol 10, 243-248.

Snell L. D., Tabakoff B. and Hoffman P. L. (1991) The density of NMDA but not glycine binding sites is increased in ethanol-dependent mice. Alcohol. : Clin. exp. Res. 15, 333.

Snell L. D., Tabakoff B. and Hoffman P. L. (1993) Radio- ligand binding to the N-methyl-D-aspartate receptor/ ionophore complex : alterations by ethanol in vitro and by chronic in vivo ethanol ingestion. Brain Res. 602, 91-98.

Stefanini E., Frau M., Garau M. G., Garau B., Fadda F. and Gessa G. L. (1992) Alcohol-preferring rats have fewer dopamine D2 receptors in the limbic system. Alcohol Alco- hol. 27, 127-130.

Stein L. (1964) Self-stimulation of the brain and the central stimulant action of amphetamine. Fedn. Proc. 23, 836- 850.

Streissguth A., Barr H. and Sampson P. (1990) Moderate prenatal alcohol exposure : effects on child I. Q. and learn- ing problems at age 7 1/2 years. Alcohol. : Clin. exp. Res. 14, 662-669.

Suzdak P. D., Schwartz R. D., Skolnick P. and Paul S. M. (1986a) Ethanol stimulates gamma-aminobutyric acid receptor-mediated chloride transport in rat brain syn- aptoneurosomes. Proc. hath. Acad. Sci. U.S.A. 83, 4071 4075.

Suzdak P. D., Glowa J. R., Crawley J. N., Schwartz R. D., Skolnick P. and Paul S. M. (1986b) A selective imid- azobenzodiazepine antagonist of ethanol in the rat. Sci- ence 234, 1243-1247.

Tabakoff B., Lee J. M., De Leon-Jones F. and Hoffman P. L. (1985) Ethanol inhibits the activity of the B form of monoamine oxidase in human platelet and brain tissue. Psychopharmacology 87, 152 156.

Tamborska E. and Marangos P. J. (1986) Brain benzo- diazepine binding sites in ethanol dependent and with- drawal states. Life Sci. 38, 465-472.

Teyler T. and DiScenna P. (1984) Long-term potentiation as a candidate mnemonic device. Brain Res. Rev. 7, 1528.

Thielen R. J., McBride W. J., Lumeng L. and Li T.-K. (1993)

Housing conditions alter GABAA receptor of alcohol-pre- ferring and -nonpreferring rats. Pharmac, Biochem. Behat,. 46, 723-727.

Thomas R. (1991) Fluvoxamine and alcoholism. In: Flu- voxamine : New Perspectives in Clinical Practice (Cobb J., ed.), pp. 85-92. Clinical Neuroscience Publishers, Oxford.

Thyagarajan R. and Ticku M. K. (1985) The effect of in vitro and in vivo ethanol administration on [~sS]t-butyl- bicyclophosphorothionate binding in C57 mice. Brain Res. Bull. 15, 343-345.

Ticku M. K. and Burch T. (1980) Alterations in gamma- aminobutyric acid receptor sensitivity following acute and chronic ethanol treatments. J. Neurochem. 34, 417-423.

Toneatto T., Romach M. K., Sobell L. C., Somer G. R. and Sellers E. M. (1991) Ondansetron, a 5-HT3 antagonist, reduces alcohol consumption in alcohol abusers. Alcohol. : Clin. exp. Res. 15, 382.

Toth P., Shaw C., Perlanski E. and Grupp L. A. (1990) Cholecystokinin octapeptide reduces ethanol intake in food- and water-sated rats. Pharmac. Biochem. Behav. 35, 493-495.

Trevisan L., Fitzgerald L. W., Brose N., Gasic G. P., Heine- mann S. F., Duman R. S. and Nestler E. J. (1994) Chronic ingestion of ethanol up-regulates NMDARI receptor subunit immunoreactivity in rat hippocampus. J. Neur- ochem. 62, 1635-t638.

Ulrichsen J. (1991) Alterations in serotonin receptor sub- types in ethanol-dependent rats. Alcohol Alcohol. 26, 567- 573.

Unwin J. W. and Taberner P. V. (1980) Sex and strain differences in GABA receptor binding after chronic etha- nol drinking in mice. Neuropharrnacology 19, 1257-1259.

Valverius P., Crabbe J. C., Hoffman P. L. and Tabakoff B. (1990) NMDA receptors in mice bred to be prone or resistant to ethanol withdrawal seizures. Eur. J. Pharmac. 184, 185-189.

Volpicelli J. R., Alterman A. I., Hayashida M. and O'Brien C. P. (1992) Naltrexone in the treatment of alcohol depen- dence. Arch. Gen. Psychiat. 49, 876-880.

Wafford K. A., Burnett D. M., Leidenheimer N. J., Burt D. R., Wang J. B., Kofuji P., Dunwiddie T. V., Harris R. A. and Sikela J. M. (1991) Ethanol sensitivity of the GABAA receptor expressed in Xenopus oocytes requires 8 amino acids contained in the y2L subunit. Neuron 7, 27-33.

Wahlstrrm G. and Nordberg A. (1991) Changes in drinking behaviour and cholinergic binding sites induced by inter- mittent long-term ethanol treatment in the male rat. Alco- hol Alcohol. 26, 575 584.

Walker D. W. and Hunter B. E. (1978) Short-term memory impairment following chronic alcohol consumption in rats. Neuropsychologia 16, 545-553.

Waller M. B., McBride W. J., Gatto G. J., Lumeng L. and Li T.-K. (1984) lntragastric self-infusion of ethanol by ethanol-preferring and -nonpreferring lines of rats. Science 225, 78-80.

Wallgren H. and Barry H. III (eds) (1970) Actions o f Alcohol, Vol. 1. Elsevier Science Publishing, New York.

Weiss F., Mitchiner M., Bloom F. E. and Koob G. F. (1990) Free-choice responding for ethanol versus water in alcohol preferring (P) and unselected Wistar rats is differentially modified by naloxone, bromocriptine, and methysergide. Psychopharmacology 101, 178-186.

White G., Lovinger D. M. and Weight F. F. (1990) Ethanol inhibits NMDA-activated current but does not alter

336 Invited Review

GABA-activated current in an isolated adult mammalian neuron. Brain Res. 507, 332-336.

Widdowson P. S. (1987) The effect of neurotensin, TRH, and the ,5-opioid receptor antagonist ICI 174864 on alcohol- induced narcosis in rats. Brain Res. 424, 281 289.

Widdowson P. S. and Holman R. B. (1992) Ethanol-induced increase in endogenous dopamine release may involve endogenous opiates. J. Neurochem. 59, 157 163.

Wigal S. B., Amsel A. and Wilcox R. E. (1990) Fetal ethanol exposure diminishes hippocampal /~-adrenergic receptor density while sparing muscarinic receptors during devel- opment. Det'. Brain Res. 55, 161 169.

Wise R. A. (1980) Actions of drugs of abuse on brain reward systems. Pharmac. Biochem. Behat,. 13 (Suppl.), 213-223.

Wise R. A. and Bozarth M. A. (1985) Actions of abused drugs on reward systems in the brain, in : Neurotoxicologv (Blum K. and Manzo L., eds), pp. 111 133. Marcel Dekker, New York.

Wise R. A. and Bozarth M. A. (1987) A psychomotor stimu- lant theory of addiction. P,~:vchol. Rel~. 94, 469492.

Wong D. T.. Lumeng L.. Threlkeld P. G.. Reid L. R. and Li T.-K. (1988) Serotonergic and adrenergic receptors in alcohol-preferring and non-preferring rats. J. Neural Transm. 71,207-218.

Wong D. T., Threlkeld P. G., Lumeng L. and Li T.-K. (1990) Higher density of serotonin-lA receptors in the hippocampus and cerebral cortex of alcohol-preferring P rats. L!fe Sci. 46, 231 235.

Wong D. T., Reid L. R., Li T.-K. and Lumeng L. (1993) Greater abundance of serotonin~n receptor in some brain areas of alcohol-preferring (P) rats compared to non- preferring (NP) rats. Pharmac. Biochem. Behar'. 46, 173 177.

Woodward J. J. (1994) A comparison of the effects of ethanol and the competitive glycine agonist 7-chlorokynurenic acid on N-methyl-o-aspartic acid-induced neurotrans- mitter release from rat hippocampal slices. J. Neurochem. 62, 987-991.

Woodward J. J. and Gonzales R. A. (1990) Ethanol inhi- bition of N-methyl-D-aspartate-stimulated endogenous dopamine release from rat striatal slices: reversal by glycine. J. Neurochem. 54, 712 715.

Yoshimoto K. and McBride W. J. (1992) Regulation of nucleus accumbens dopamine release by the dorsal raphe nucleus in the rat. Neurochem. Res. 17, 401-407.

Yoshimoto K., McBride W. J., Lumeng L. and Li T.-K. (1991) Alcohol stimulates the release of dopamine and serotonin in the nucleus accumbens. Alcohol 9, 17--22.

Youdim M. B. H. (1988) Platelet monoamine oxidase B : use and misuse. Experientia 44, 137-141.

Zabik J. E. and Roache J. D. (1983) 5-hydroxytryptophan- induced conditioned taste aversion to ethanol in the rat. Pharmac. Biochem. Behat,. 18, 785-790.

Zhou F. C., Bledsoe S., Lumeng L. and Li T.-K. (1991) Immunostained serotonergic fibers are decreased in selec- ted brain regions of alcohol-preferring rats. Alcohol8, 425 431.


Recommended