Sei sulla pagina 1di 10

Neuron, Vol.

21, 467–476, September, 1998, Copyright 1998 by Cell Press

Neuroscience of Addiction Review

George F. Koob,* Pietro Paolo Sanna, framework for understanding the molecular basis of ad-
and Floyd E. Bloom diction (Koob and Le Moal, 1997; Koob et al., 1998).
Department of Neuropharmacology The underlying molecular and cellular changes that
The Scripps Research Institute occur with the transition from occasional drug use to
La Jolla, California 92037 pathological abuse and addiction are only partially un-
derstood as yet. To reach a more complete understand-
ing, these events will have to be integrated with the
Human addictions are chronically relapsing disorders animal models of the different elements of the addiction
characterized by compulsive drug taking, an inability process, including models of the transition from simple
to limit the intake of drugs, and the emergence of a drug taking to compulsive use at the molecular, cellular,
withdrawal syndrome during cessation of drug taking and behavioral levels. In this review, we will focus in
(dependence). The development of an addiction impacts particular on the factors that drive drug-seeking behav-
on several separate neurobiological processes, and ior at different stages of the addiction cycle (Koob and
these effects are both drug- and drug use–dependent. Le Moal, 1997), and we will place particular emphasis
In animal models of addiction, changes in specific neu- on trying to identify what is currently known and what
rotransmitter systems within a highly limited band of remains to be elucidated.
structures, including specific parts of the nucleus ac-
cumbens and amygdala, may underlie drug reward and Neurobiological Substrates for the Acute
the motivational effects associated with dependence. Reinforcing Effects of Drugs of Abuse
Changes in the signals mediated by several neurotrans- Animals and humans will readily self-administer the
mitters, including dopamine, opioid peptides, and corti- same classes of drugs, and such self-administration
cotropin-releasing factor (CRF), and in the regulation of defines these drugs as positive reinforcers (Headlee et
selected transcription factors within the neurons of this al., 1955). The powerful reinforcing properties of such
reward circuit, may underlie the vulnerability to relapse drugs are revealed by the efforts experimental animals
that characterizes addiction in humans. will perform to get them, such as pressing a lever multi-
ple times to receive an intravenous injection of the drug.
Animal Models of Addiction While early work focused on drug-taking behaviors in
Addiction, also known as substance dependence (Amer- dependent animals (largely primates), subsequent stud-
ican Psychiatric Association, 1994), is a chronically re- ies have replicated the same behaviors in nondepen-
lapsing disorder that is characterized by three major dent animals (largely rodents) (Schuster and Thompson,
elements: (1) compulsion to seek and take the drug, (2) 1969). Since rodents are more tractable experimentally,
loss of control in limiting intake, and (3) emergence of they have provided much recent insight into the perti-
a negative emotional state (e.g., dysphoria, anxiety, irri- nent circuits and transductive mechanisms for the acute
tability) when access to the drug is prevented (defined reinforcing effects of drugs of abuse (Koob and Bloom,
here as dependence) (Koob and Le Moal, 1997). Both 1988).
clinically and in experimental animals, the occasional Neuropharmacological studies have established an
use of an abusable drug is distinct from repeated drug important role for the dopaminergic system in the acute
use and the emergence of chronic drug addiction. The reinforcing effects of cocaine (see Table 1) (Woolverton
goal of current neuroscience research is to understand and Johnson, 1992). The midbrain dopamine system is
the cellular and molecular mechanisms that mediate the composed of two major projections: the nigrostriatal
transition between occasional, controlled drug use and system, which projects from the substantia nigra to the
the loss of behavioral control over drug seeking and corpus striatum, and the mesocorticolimbic dopamine
drug taking that defines chronic addiction (Koob and Le system, which projects from the ventral tegmental area
Moal, 1997). (VTA) to the nucleus accumbens, olfactory tubercle,
Much of the recent progress in understanding the frontal cortex, and amygdala. It is the mesocorticolimbic
mechanisms of addiction has derived from the develop- system that has been primarily implicated in the reinforc-
ment of animal models of addiction on specific drugs ing actions of drugs of abuse. Psychostimulants such
such as opiates, stimulants, and ethanol. These animal as cocaine and d-amphetamine elevate extracellular do-
models have localized the synaptic sites and transduc- pamine by inhibiting reuptake of dopamine by the dopa-
tive mechanisms in the nervous system on which drugs mine transporter and, in the case of d-amphetamine,
of abuse act initially and are beginning to be used to also by promoting reverse transport of dopamine. Dur-
explore how the nervous system adapts to drug use. ing intravenous cocaine self-administration, increased
While no animal model of addiction fully emulates the extracellular dopamine can be detected in the nucleus
human condition, animal models do permit investigation accumbens using in vivo microdialysis (Pettit and Jus-
of elements of the process of drug addiction, and the tice, 1989). In addition, selective destruction of meso-
data derived from these models provides an empirical corticolimbic dopamine neurons with the neurotoxin
6-hydroxydopamine (6-OHDA) eliminates cocaine self-
administration; similar decreases in cocaine self-admin-
* To whom correspondence should be addressed. istration occur when 6-OHDA lesions are restricted to
Neuron
468

Table 1. Neurobiological Substrates for the Acute Reinforcing


and stereotypy (Giros et al., 1996). However, such dopa-
Effects of Drugs of Abuse mine transporter–deficient mice still could be trained to
self-administer cocaine despite persistently high levels
Drug of Abuse Neurotransmitter Sites
of extracellular dopamine in dopaminergic terminal
Cocaine and Dopamine Nucleus accumbens fields, suggesting a more complex basis for the psycho-
amphetamines Serotonin Amygdala stimulant reinforcement (Rocha et al., 1998). Besides
Opiates Dopamine Ventral tegmental area
inhibiting the dopamine transporter, psychostimulants
Opioid peptides Nucleus accumbens
Nicotine Dopamine Ventral tegmental area also inhibit the reuptake of serotonin and noradrenaline,
Opioid peptides? Nucleus accumbens which may contribute to their reinforcing actions possi-
Amygdala? bly in part by modulating dopamine neurotransmission
THC Dopamine Ventral tegmental area (Parsons et al., 1995; Tanda et al., 1997b).
Opioid peptides? Much like psychostimulants, opiate drugs are readily
Ethanol Dopamine Ventral tegmental area
self-administered intravenously by animals, and the sys-
Opioid peptides Nucleus accumbens
Serotonin Amygdala temic and central administration of competitive opiate
GABA antagonists will decrease opiate reinforcement (re-
Glutamate viewed by Koob and Bloom, 1988; Di Chiara and North,
1992). The reinforcing actions of opiates appear to be
largely mediated by the m opioid receptor since selective
the dopamine fibers of the region of the nucleus accum- m antagonists decrease opioid reinforcement in a dose-
bens (Roberts et al., 1980). Multiple receptor subtypes dependent manner (Negus et al., 1993). In addition, mor-
exist for transducing the increase in extracellular dopa- phine reinforcement is abolished in mice with a targeted
mine induced by psychomotor stimulants into behav- disruption of the m opioid receptor gene (Matthes et al.,
ioral action. Antagonists for the dopamine D1, D2, and
1996).
D3 receptor subtypes all decrease the reinforcing prop-
The reinforcing properties of opiates utilize the same
erties of cocaine (Woolverton and Johnson, 1992; Caine
circuitry implicated in the actions of cocaine and am-
et al., 1995; Koob and Le Moal, 1997; Epping-Jordan et
phetamine stimulants but may involve additional sites
al., 1998a).
of interaction (Koob and Bloom, 1988) (Table 1). Block-
The neuronal interaction responsible for cocaine rein-
ade of opioid receptors either in the VTA or the nucleus
forcement and the motivation to seek the drug appears
accumbens will decrease heroin self-administration.
to reside within the nucleus accumbens (Chang et al.,
Furthermore, rats will lever press to administer opioid
1994; Carelli and Deadwyler, 1996; Peoples et al., 1997).
peptides in their nucleus accumbens or VTA, and opiate
Electrophysiological recordings in animals receiving in-
administration into these restricted brain regions will
travenous cocaine by self-administration have identified
reinforce drug-seeking behavior (reviewed by Di Chiara
several patterns of neuronal responses in the nucleus
and North, 1992; Shippenberg et al., 1992). Opiates, like
accumbens, all time-locked to the self-administered
drug infusion. One group of neurons fires just before other drugs of abuse, increase dopamine release in the
the lever press, and this anticipatory response may be nucleus accumbens (Di Chiara and Imperato, 1988; Pon-
an initiation or trigger mechanism. A second group of tieri et al., 1995) (see below). However, the reinforcing
neurons appears to change firing rate only after the effect of opiates in the nucleus accumbens persists
cocaine infusion, and these neurons may represent the when all dopamine projections there are destroyed, sug-
direct effects of reinforcement (Carelli and Deadwyler, gesting that their reinforcing actions may involve both
1996). Other neurons fire in proportion to the interinfu- a dopamine-dependent (VTA) and a dopamine-inde-
sion interval between consecutive self-administration pendent (nucleus accumbens) mechanism (Koob and
responses (Peoples and West, 1996). However, there Bloom, 1988).
appears to be a fourth type of neuronal firing pattern Sedative-hypnotics, including ethanol, are thought to
unique to cocaine self-administration; these “cocaine- produce their reinforcing actions through multiple neu-
specific cells” fire both before and after the cocaine- rotransmitter systems (Engel et al., 1992) (Table 1). One
reinforced response (Carelli and Deadwyler, 1996). Even of the major sites proposed for ethanol reinforcement
more intriguing is the observation that this subset of is modulation of GABA receptors (Liljequist and Engel,
neurons also fires to sensory stimuli (sounds or lights) 1982; Samson and Harris, 1992). GABA antagonists re-
that have been experimentally paired with cocaine deliv- verse many of the behavioral effects of ethanol (Liljequist
ery. Nucleus accumbens neurons may therefore mediate and Engel, 1982; Samson and Harris, 1992). Further-
conditioned drug responses (Carelli and Deadwyler, more, the benzodiazepine R0 15–4513 (termed an in-
1996). Similarly, conditioned sensory stimuli are strong verse agonist because it produces effects opposite to
elicitors of “craving” in cocaine-taking humans. those of typical benzodiazepines) will reverse some of
Recent studies using recombinant DNA techniques to the behavioral effects of ethanol, and dose-dependently
“knock out” specific genes involved in dopaminergic reduces oral ethanol self-administration in rats (Samson
neurotransmission may provide evidence of some re- and Harris, 1992). When potent GABA antagonists are
dundancy in the neurochemical basis of cocaine rein- microinjected into the brain, the most effective site to
forcement. In a mouse strain in which the gene for the reduce ethanol consumption is the central nucleus of
dopamine transporter was disrupted by homologous re- the amygdala (Hyttia and Koob, 1995).
combination, psychostimulants failed to alter baseline Ethanol reinforcement also appears to involve activa-
extracellular dopamine levels and failed to induce be- tion of brain dopamine systems. Acutely, ethanol con-
havioral effects such as enhanced locomotor activity sumption or systemic injection reduces the firing rate
Review
469

of pars reticulata GABA neurons, which are thought to Table 2. Drug Effects on Thresholds for Rewarding Brain
exert an inhibitory control over VTA dopaminergic neu- Stimulation
rons (Diana et al., 1993; Merue and Gessa, 1985), and
Withdrawal
increases concentrations of dopamine in the extracellu- Acute from Chronic
lar compartment in the nucleus accumbens. Dopamine Drug Class Administration Treatment
receptor antagonists injected into the nucleus accum-
Psychostimulants ↓ ↑
bens reduce lever pressing for ethanol in nondependent
(cocaine, amphetamines)
rats, and basal extracellular dopamine levels are in- Opiates ↓ ↑
creased in rats chronically consuming low doses of etha- (morphine, heroin)
nol (reviewed by Koob et al., 1994b). However, virtually Nicotine ↓ ↑
complete 6-hydroxydopamine denervation of the dopa- Sedative-Hypnotics ↓ ↑
mine inputs to the nucleus accumbens fails to alter vol- (ethanol)
THC ↓ ↑
untary responding for ethanol, suggesting that nucleus
accumbens dopamine transmission may not be critical
for reinforcing actions of ethanol (Koob et al., 1994b).
Moreover, manipulations that change brain serotonin dopamine system (Chen et al., 1991), and recent data
synaptic availability can decrease ethanol intake (Sellers suggest that THC can selectively increase the release
et al., 1992). For example, serotonin reuptake blockers of dopamine in the shell of the nucleus accumbens simi-
decrease ethanol intake but so do antagonists of the lar to other drugs of abuse (Tanda et al., 1997a).
serotonin-3 and serotonin-2c receptors, suggesting a
complex interaction of serotonin function and ethanol Negative Reinforcement Associated with Addiction
reinforcement (LeMarquand et al., 1994). Repeated drug use is thought to arise from the neuro-
Opiate antagonists also reduce ethanol self-adminis- chemical actions causing the positive reinforcing effects
tration in several animal models (Tabakoff and Hoffman, of a drug. However, the transition from occasional drug
1996). The central nucleus of the amygdala is particularly use to drug addiction has been thought to require an
sensitive to this opioid antagonism of oral ethanol self- additional source of reinforcement, the reduction of the
administration (Heyser, Roberts, and G. F. K., unpub- aversive (negative) emotional state arising from re-
lished data). Double-blind, placebo-controlled clinical peated use. Here, drug taking presumably removes the
trials showed that naltrexone can significantly reduce dysphoria, anxiety, irritability, and other unpleasant feel-
human ethanol consumption, frequency of relapse, and ings produced by drug abstinence. Other somatic physi-
“craving” for ethanol in detoxified alcoholics, suggesting cal signs, such as tremor, temperature changes, and
that the motivation to resume ethanol use may involve sweating, which also reflect the state of dependence,
opioid systems (Volpicelli et al., 1992). Finally, low doses presumably have little if any motivating properties on
of ethanol will sensitively inhibit the NMDA subtype of drug use (Koob, 1996; Koob and Le Moal, 1997). Indeed,
glutamate receptors; in drug discrimination studies, ani- one of the defining features of drug addiction is thought
mals substitute glutamate antagonists for ethanol (Ta- to be the establishment of such a negative emotional
bakoff and Hoffman, 1996). state (Russell, 1976). Consistent with this hypothesis,
Intravenous self-administration of nicotine is also all major drugs of abuse have been found to produce a
blocked by dopamine antagonists and dopamine-selec- negative emotional state in dependent humans during
tive lesions of the nucleus accumbens (Corrigall et al., acute abstinence. The combination of the positive rein-
1992; Dani and Heinemann, 1996). Nicotine withdrawal forcing effects of the drugs with reduction of the nega-
can also be produced by opioid antagonists (Dani and tive emotional states of drug abstinence provides a pow-
Heinemann, 1996). Nicotine thus may activate both the erful motivational force for the compulsive drug taking
dopamine system and some opioid peptide neurons in that characterizes addiction.
the same neural circuitry associated with other drugs One likely mechanism for this negative emotional state
of abuse (Corrigall et al., 1992) (see Table 1). Exactly may be a reduction in brain reward function. In studies
which type of nicotinic receptor subunit configuration employing intracranial self-stimulation behavior, to di-
mediates the reinforcing effects of nicotine is not clear, rectly study brain reward circuits, animals that have
but neurons in the VTA and nucleus accumbens express been made chronically dependent show increased re-
high levels of the a6, a2, and a3 subunits (Le Novere et ward thresholds (i.e., decreased reward) during with-
al., 1996). drawal (reviewed by Koob et al., 1993; Koob, 1996; Koob
Tetrahydrocannabinol (THC) shares effects in animal and Le Moal, 1997). These decreases in reward have
models of drug reinforcement similar to those of other been observed following the withdrawal from psycho-
drugs of abuse (Anthony et al., 1994). Upon acute admin- motor stimulants, opiates, ethanol, THC, and nicotine
istration, THC decreases reward thresholds in rats and are dose related to the amount of drug that had
(Gardner et al., 1988; Lepore et al., 1996), produces a been administered before withdrawal (Epping-Jordan et
place preference in rats (Lepore et al., 1995), and, as a al., 1998b; Gardner and Vorel, 1998) (Table 2).
synthetic THC analog, is intravenously self-administered
in mice (Fratta et al., 1997, Soc. Neurosci., abstract). Common Neuropharmacological Elements
THC binds to the cannabinoid-1 receptor, which is in Addiction
widely distributed throughout the brain but particularly The molecular and cellular basis for these changes in
in the extrapyramidal motor system of the rat (Herken- motivation to take drugs may reside in the neuroadapta-
ham et al., 1990). THC activates the mesocorticolimbic tions arising in the same neural elements that mediate
Neuron
470

Table 3. Neurotransmitters Implicated in the Motivational


Microinjections into the central nucleus of the amygdala
Effects of Withdrawal from Drugs of Abuse of lower doses of a CRF antagonist also reversed the
anxiogenic-like effects of ethanol withdrawal. Similar
↓ Dopamine
↓ Opioid peptides
doses of the CRF antagonist injected into the amygdala
↓ Serotonin reversed the aversive effects of opiate withdrawal
↓ GABA (Koob, 1996). In vivo microdialysis studies have shown
↑ Corticotropin-releasing factor an increase in extracellular CRF during ethanol, cocaine,
and THC withdrawal (Merlo-Pich et al., 1995; Rodriguez
de Fonseca et al., 1997; Richter and Weiss, personal
the acute reinforcing actions of these drugs. Earlier work communication). Thus, CRF activation may be a com-
suggested that these adaptations occur both within the mon element in the development of drug dependence
drug-sensitive reinforcement system and in additional and may contribute to motivational effects involving
systems outside the drug-sensitive reward system (Koob such subjective symptoms as increased stress and neg-
and Bloom, 1988). Several common elements have been ative affect (Koob, 1996).
identified that appear to change with chronic drug ad- The neurochemical changes highlighted above illus-
ministration and could underlie (or mediate) the compul- trate neuroadaptations common to all drugs of abuse
sive self-administration of all drugs (Koob, 1992; Koob, (Koob, 1996). However, even more intriguing is the pos-
1996; Koob and Le Moal, 1997) (Table 3). sibility that a neuroanatomical entity termed the ex-
Functional changes in the mesolimbic dopamine sys- tended amygdala (Heimer and Alheid, 1991) (Figure 1)
tem appear to be common to the chronic actions of all may represent a common anatomical substrate for acute
drugs of abuse (Nestler, 1996). Drug discontinuation is drug reward and the negative effects of compulsive drug
accompanied by biochemical and electrophysiological administration on reward function. The extended amyg-
evidence of decreases in dopamine function (Diana et dala is comprised of the medial subregion of the nucleus
al., 1993; Nestler, 1996; Weiss et al., 1996). During with- accumbens (termed the shell of the nucleus accum-
drawal from chronic ethanol administration, there is a bens), the bed nucleus of the stria terminalis, and the
decrease in extracellular dopamine in the nucleus ac- central nucleus of the amygdala (Heimer and Alheid,
cumbens, and, concomitantly, neurons of the mesolim- 1991). Heimer has noted that all of these regions share
bic dopamine system show dramatic reductions in certain cytoarchitectural and circuitry similarities (Heimer
spontaneous firing rates and patterns but not in the and Alheid, 1991). The extended amygdala receives nu-
number of spontaneously active neurons (Diana et al., merous afferents from limbic structures, such as the
1993; Weiss et al., 1996). These biochemical and electro- basolateral amygdala and hippocampus, and sends not
physiological changes can be reversed by ethanol ad- only efferents to the medial part of the ventral pallidum
ministration. Another neurotransmitter system impli- but also a large projection to the lateral hypothalamus,
cated in the acute reinforcing actions of drugs of abuse thus further defining the specific brain areas that inter-
whose function is altered during the development of face classical limbic (emotional) structures with the ex-
dependence is the opioid peptide system, as seen with trapyramidal motor system.
either chronic opiate usage (Self and Nestler, 1995; Nest- The structures comprising the extended amygdala
ler, 1996) or with other drugs of abuse (Di Chiara and may further define the neural substrates for the acute
North, 1992). Here, the functional changes are manifest reinforcing actions of drugs of abuse (Koob, 1992; Koob,
by a dramatic increased sensitivity to opioid receptor 1996). Acute administration of all the major drugs of
antagonists, probably mediated by alterations in opioid abuse produces increases in extracellular levels of do-
receptor signal transduction (see below) (Nestler, 1996; pamine in the shell of the nucleus accumbens (Pontieri
Widnell et al., 1996). et al., 1995). The ventromedial shell of the nucleus also
In addition, several neurotransmitter systems that are expresses high levels of the dopamine D3 receptor
not involved in the acute reinforcing effects of drugs of mRNA (Diaz et al., 1995), and the shell of the nucleus
abuse do appear to become involved following chronic accumbens is particularly sensitive to the cocaine an-
drug administration. These include several neuropep- tagonist activity of a dopamine D1 antagonist (Caine et
tides, notably dynorphin, neuropeptide FF (NPFF), and al., 1995). The central nucleus of the amygdala also has
CRF. Neurons containing the opioid peptide dynorphin a role in ethanol reinforcement. Microinjection of GABA
in the nucleus accumbens appear to be functionally antagonists or opioid peptide antagonists into the cen-
activated following chronic administration of cocaine tral nucleus can attenuate lever pressing for oral ethanol
(Hyman, 1996), while anti-opioid neuropeptides such as (Hyttia and Koob, 1995; Heyser, Roberts, and G. F. K.,
NPFF may be activated following chronic opiates (Malin unpublished data). Perhaps more intriguing is recent
et al., 1990; Lake et al., 1992). evidence that the extended amygdala may be an impor-
CRF, with its many actions on hormonal and behav- tant substrate for the changes in the reward system
ioral responses to stressors, may be a brain system en- associated with dependence. In animals dependent on
gaged during drug dependence by all drugs of abuse. ethanol, microinjections of previously ineffective doses
Activation of the pituitary adrenal axis long has been of a GABA agonist into the central nucleus of the amyg-
recognized as a characteristic of drug dependence and dala decreased ethanol self-administration (Roberts et
withdrawal in humans (Kreek, 1987). Rats treated re- al., 1996), suggesting that the GABAergic system has
peatedly with cocaine, nicotine, and ethanol show signif- been altered to become more responsive to agonists
icant anxiogenic-like responses following cessation of during the course of dependence. Thus, the same neuro-
chronic drug administration that are reversed with intra- chemical components in the extended amygdala in-
cerebroventricular administration of a CRF antagonist. volved in acute drug actions may become compromised
Review
471

Figure 1. Horizontal Section of a Rat Brain Depicting the Principal Structures of the Extended Amygdala
These structures include the central nucleus of the amygdala, the shell part of the nucleus accumbens, and the bed nucleus of the stria
terminalis. The drugs listed below each structure refer to potential sites of action of drug reinforcement during the addiction cycle, either
positive or negative. Redrawn with permission from Heimer and Alheid (1991).

during the development of dependence. One may spec- and Nestler, 1995; Hyman, 1996) (see Figure 3). An im-
ulate that additional neurochemical systems also may portant role for the D1 receptor–cAMP–CREB pathway
be engaged within the neurocircuitry of the extended in the neuroadaptation to chronic drug dependencies
amygdala (Koob and Bloom, 1988), in an attempt to is supported also by the recent evidence of effective
overcome the chronic presence of the perturbing drug anti-cocaine actions from dopamine D1 antagonists and
and to restore normal function despite the presence of effective anti-cocaine priming effects from D1 agonists
drug. The changes in CRF on the central nucleus of (i.e., ability of cocaine to reinstate responding in rats
the amygdala observed in dependent animals during that have stopped responding after cocaine has been
withdrawal support this hypothesis (Koob et al., 1994a, replaced by saline) (Self et al., 1996). Activation of Gi
1994b) (see Table 1, Table 2, and Figure 1). proteins, linked to D2 receptors, also appears to be
involved in the acute effects of cocaine since pertussis
toxin, which inactivates Gi proteins, produces a dopa-
Molecular, Cellular, and System Adaptations mine receptor antagonist-like effect on cocaine self-
Associated with Brain Motivational Systems administration (Self et al., 1994).
In addition to the changes in neurochemical systems In the VTA, repeated administration of cocaine pro-
that are common to the chronic administration of drugs duces transient decreases in Gi proteins that may lead
of abuse, there are molecular changes that may provide to D2 receptor subsensitivity (Nestler, 1994; Self and
not only the mechanism for the above neurochemical Nestler, 1995). More prolonged effects that persist up
changes but also the substrate for prolonged adaptation to one month in the nucleus accumbens include a super-
to chronic drug administration. Differences in adaptive sensitivity to D1-mediated responses (Henry and White,
responses at the molecular level may account for indi- 1991), increased levels of adenylyl cyclase and protein
vidual differences in vulnerability to dependence to dif- kinase A (PKA), decreased levels of Gi proteins (Self
ferent drugs. The challenge for future research is to link and Nestler, 1995; Nestler, 1996), and a decrease in the
these changes in plasticity to motivational actions of ability of cocaine to induce the immediate-early gene
dependence (Figure 2). c-fos, followed by the sustained expression of AP-1
Several molecular consequences of motivationally im- transcription factor complexes with altered composition
portant adaptations to chronic cocaine or amphetamine (see below).
administration have been identified. For example, acti- Chronic administration of cocaine also decreases the
vation of D1-like receptors stimulates a cascade of levels of neurofilament proteins in the VTA (Self and
events, including activation of Gs proteins and increased Nestler, 1995). Lower levels of neurofilament proteins
intracellular cyclic AMP formation that ultimately may are associated with decreased axonal transport, and
lead to phosphorylation of transcription factors such as this could decrease the amount of tyrosine hydroxylase
cyclic AMP response element–binding protein (CREB) transported from the VTA to the dopamine nerve termi-
and to the induction of immediate-early genes (see Self nals in the nucleus accumbens (Self and Nestler, 1995).
Neuron
472

Figure 2. Schematic Diagram Showing the


Relationship between Different Levels of
Analysis in the Study of Addiction and the
Role of Neuroadaptive Processes
Neuroadaptive mechanisms have been hy-
pothesized to contribute to compulsive be-
havior and addiction by acting at different
levels of the spiraling cycle of the develop-
ment of dependence (Koob and Le Moal,
1997). Both sensitization and counteradapta-
tion may contribute to changes in hedonic
responsiveness and set-points.

Such changes could be responsible for short-term re- Chronic ethanol exposure has been shown to compro-
ductions in extracellular levels of dopamine release dur- mise dopamine and GABAergic systems that are linked
ing drug withdrawal (Weiss et al., 1992), which then to the continued desire to consume ethanol (see above).
could trigger upregulation of the cyclic AMP system At the molecular level, chronic ethanol is associated
(Self and Nestler, 1995) (see above). with decreases in the ability of ethanol to potentiate
These adaptations within the mesolimbic dopamine GABA-stimulated Cl2 flux, decreased expression of the
system, and its receptor systems, not only could change a1 subunit of the GABA complex (Tabakoff and Hoffman,
the function of the dopamine system itself but also may 1996) and other subunits, and increased expression of
trigger a second major action, namely increased expres- the b subunit (Tabakoff and Hoffman, 1996). Chronic
sion of protachykinin and prodynorphin mRNAs. Dynor- ethanol also is associated with increases in specific
phin peptides in the nucleus accumbens, in turn, may subunits (NR1 and NR2A) of NMDA receptors (Tabakoff
decrease dopamine release via a presynaptic action on and Hoffman, 1996). The relationship of these molecular
k opioid receptors; k agonists are known to produce changes in ethanol-receptive elements to the motiva-
aversive effects in rodents and humans (Hyman, 1996). tional substrates outlined above may ultimately involve
Thus, chronic administration of psychomotor stimulants changes in the same transduction systems observed
would induce prodynorphin gene expression in the nu- for other drugs of abuse (Figures 2 and 3). A possible
cleus accumbens that opposes the effect of cocaine on anatomical substrate for the molecular action of GABA
reward. on motivational aspects of ethanol dependence is the
Chronic opiate administration is associated with lit- amygdala, a brain region considered important in medi-
tle evidence of changes in opioid peptide activity or ating emotional behavior in general. GABAergic neurons
changes in the number of any of the known opioid recep- in the central amygdala also have been proposed to
tors (but see Mansour et al., 1995; Zadina et al., 1997). mediate rapid changes in autonomic activity (Sun and
However, there is strong evidence that a dramatic en- Cassell, 1993), and acute ethanol induces expression of
hancement of sensitivity to the aversive effects of opioid the immediate-early gene c-fos, a marker of neuronal
antagonists can occur in brain areas implicated in the activation, in GABAergic neurons in the central amyg-
acute reinforcing effects of opiates (Stinus et al., 1990; dala (Morales, Criado, and P. P. S., unpublished data).
Self and Nestler, 1995; Nestler, 1996). The molecular Adaptation to chronic nicotine administration also
basis for this effect may be at the level of signal trans- may occur in the same neuronal circuits associated with
duction (see Figures 2 and 3). Acute administration of its initial molecular actions, namely nicotine on nicotinic
morphine decreases adenylate cyclase activity in the acetylcholine receptors located in the brain mesolimbic
nucleus accumbens, whereas chronic morphine treat- dopamine system. One recent hypothesis seeks to ex-
ment is associated with increases in second messenger plain nicotine tolerance and dependence on the basis
systems, including adenylate cyclase activity and PKA of a prolonged stability in the desensitized state of the
(Nestler, 1996). The hypothesis that these effects con- nicotinic acetylcholine receptor following the initial stim-
tribute to the motivational effects of opiates (e.g., toler- ulation (or “activation”) (Dani and Heinemann, 1996).
ance and dependence) is supported by studies showing Consistent with this desensitization hypothesis, long-
that direct administration into the nucleus accumbens term nicotine exposure causes an increase in the actual
of agents that inhibit Gai or activate protein kinase de- number of nicotinic acetylcholine receptors (Collins et
creases the reinforcing effects of opiates (Self et al., al., 1990).
1994). Chronic morphine administration also decreases Dani and Heinemann (Dani and Heinemann, 1996)
the level of the transcription factor CREB in the nucleus have proposed that nicotine stimulates the mesolimbic
accumbens. This provides a possible mechanism for dopamine system via activation of nicotinic acetylcho-
morphine to mediate the alterations in gene expression line receptors to produce the acute reinforcing effects
that may underlie the long-term changes in motivational of nicotine; with continued use, the inactivation of these
systems associated with opioid dependence (Widnell et receptors by desensitization would then lead to adaptive
al., 1996) (Figure 3). Genetic disruption of two of the tolerance. During abstinence, nicotine levels fall, and the
three types of CREB in mice dramatically reduced symp- increased nicotinic acetylcholine receptors, throughout
toms of morphine withdrawal and produced some re- the brain, begin to recover to a responsive state that
duction in tolerance to the analgesic effects of morphine may be dependent on the receptor subtype. Engag-
(Maldonado et al., 1996). ing nicotinic receptors in non-reward-related pathways
Review
473

Figure 3. Molecular Mechanisms of Neuro-


adaptation
Drugs of abuse, by acting on neurotransmit-
ter systems, affect the phenotypic and func-
tional properties of neurons through the gen-
eral mechanisms outlined in the diagram.
Shown are examples of ligand-gated ion
channels (1) such as the GABAA and the gluta-
mate NMDA receptor (NMR) and G protein–
coupled receptors (R) such as opioid, dopa-
mine (DA), or the cannabinoid CB1 receptors,
among others (2). The latter is activated by
endogenous cannabinoids such as anan-
damide. These receptors modulate the levels
of second messengers like cAMP and Ca21
(3), which in turn regulate the activity of pro-
tein kinase transducers (4). Such protein ki-
nases affect the functions of proteins located
in the cytoplasm, plasma membrane, and nu-
cleus (5–8). Among membrane proteins af-
fected are ligand-gated and voltage-gated
ion channels (6 and 7). Ethanol, for instance,
has been proposed to affect the GABAA re-
sponse via PKC phosphorylation and, at least
in Purkinje cells, via PKA phosphorylation
(Tabakoff and Hoffman, 1996). Gi and G o
proteins also can regulate potassium and cal-
cium channels directly through their bg sub-
units (9). Protein kinase transduction path-
ways also affect the activities of transcription
factors (8). Some of these factors, like CREB,
are regulated posttranslationally by phos-
phorylation; others, like Fos, are regulated
transcriptionally; still others, like Jun, are
regulated both posttranslationally and/or
transcriptionally. While membrane and cyto-
plasmic changes may be only local (e.g., den-
dritic domains or synaptic boutons), changes
in the activity of transcription factors may result in long-term functional changes. These may include changes in gene expression of proteins
involved in signal transduction (10) and/or neurotransmission (11–13), resulting in altered neuronal responses. For example, chronic exposure
to psychostimulants or opiates has been reported to increase levels of PKA (10) and adenylyl cyclase (11) in the nucleus accumbens and to
decrease levels of Gai (11) (Self and Nestler, 1995; Nestler, 1996). Moreover, chronic ethanol induces differential changes in subunit composition
in the GABAA and in the glutamate inotropic receptors (12) and increases expression of voltage-gated calcium channels (VGCC) (13) (Tabakoff
and Hoffman, 1996). Chronic exposure to drugs also alters the expression of transcription factors themselves (14). CREB expression, for
instance, is depressed in the nucleus accumbens and increased in the locus coeruleus by chronic morphine treatment (Nestler, 1996; Widnell
et al., 1996), while chronic cocaine and other chronic treatments induce a transition from Fos induction to the induction of the longer-lasting
Fos-related antigens (FRAs) (reviewed by Hyman, 1996). The receptor systems depicted in the figure may not coexist in the same cells. The
systematic elucidation of their fine distribution and colocalization as well as the definition of the role of pre- and postsynaptic mechanisms
are current and future challenges in the neuroscience of addiction.

could contribute to the aversive emotional states associ- (Aceto et al., 1995). In animals, a withdrawal syndrome
ated with nicotine withdrawal. According to this hypoth- is precipitated by administration of competitive THC
esis, smokers would be, in effect, ultimately medicating antagonists to animals treated chronically with canna-
themselves with nicotine to regulate the number of func- binoids (Aceto et al., 1995), and this precipitated canna-
tional nicotinic acetylcholine receptors (Dani and Heine- binoid withdrawal produced activation of limbic struc-
mann, 1996). Recent work shows that such changes tures as measured by c-fos activation and an increase
may be evident in the a subunit family, with the a4, in extracellular levels of CRF in the central nucleus of
a2, and a7 subunits showing inactivation with chronic the amygdala as seen by other major drugs of abuse
nicotine but the a3 subunit (which is similar in struc- (Rodriguez de Fonseca et al., 1997). Whether chronic
ture to the a6 subunit) showing resistance to desensiti- THC produces cellular and molecular changes in the
zation (Olale et al., 1997). Thus, one could speculate extended amygdala similar to other drugs of abuse will
that there are two powerful forces for development of be a challenge for future research.
nicotine addiction: desensitization of nicotinic receptors
in non-reward pathways (self-medication) and resis-
tance to desensitization in reward pathways (positive Craving and Relapse
reinforcement). Because drug addiction is a chronic relapsing disorder,
Marijuana is a drug of abuse and addiction in humans, the study of vulnerability to relapse is a particularly com-
and chronic high dose administration of THC or THC pelling challenge. However, the biological basis for the
analogs produces a dependence syndrome as mea- states of craving and protracted abstinence are difficult
sured by behavioral and subjective signs of withdrawal to define. Vulnerability to reinstatement of drug-taking
Neuron
474

behavior and ultimately a continuation of compulsive maintenance, withdrawal, and relapse. A particular chal-
drug use presumably reflects some underlying pro- lenge for future studies in the neuroscience of addiction
longed perturbation that emerges long after the with- will be to elucidate the neuroadaptive changes pro-
drawal reactions have subsided. A residual deficit state duced by chronic drug use in animal models of pro-
in the reward system, or sensitization of the reward tracted abstinence and relapse. Presumably, the an-
system to stimuli that predict drug effects, or both, could swers will be found not only in the same molecular and
be responsible for this vulnerability (Koob and Le Moal, cellular elements of the neurochemical systems and
1997) (Figure 2). neurocircuitry responsible for the positive and negative
Animal models of drug craving and relapse continue reinforcement associated with chronic drug use, but
to be developed and refined and are based largely on also in the multiple neuroadaptive mechanisms that es-
conditioned reinforcement where previously neutral tablish long-term memories of the drug rewards (Bailey
stimuli, such as drug injection paraphernalia or specific et al., 1996).
locations where drugs are obtained, have become
paired with the drug state or the drug withdrawal state Acknowledgments
(Koob, 1995). The neural substrates for such conditioned
positive reinforcement may involve elements of the ex- This is publication number 11130-NP from The Scripps Research
tended amygdala and its afferent circuits from the baso- Institute. Research was supported by National Institutes of Health
lateral amygdala (Everitt et al., 1991) and the mesolimbic grants DA04043, DA04398, and DA08467 from the National Institute
on Drug Abuse and AA06420 and AA08459 from the National Insti-
dopamine system. The neural substrates for any hypo-
tute on Alcohol Abuse and Alcoholism. The authors would like to
thetical conditioned negative reinforcement are largely thank Mike Arends for his valuable assistance with manuscript prep-
unknown. However, by definition, any conditioning that aration.
occurs during chronic drug use also involves the forma-
tion of subtle associations, but it is unknown whether References
the associations formed by the pairing of drugs with
previously neutral stimuli utilize the same circuits as Aceto, M.D., Scates, S.M., Lowe, J.A., and Martin, B.R. (1995). Can-
those for memories not related to drugs. nabinoid precipitated withdrawal by the selective cannabinoid re-
A possible molecular mechanism for such learned, ceptor antagonist, SR 141716A. Eur. J. Pharmacol. 282, R1–R2.
long-term neuroadaptation may involve alterations to American Psychiatric Association (1994). Diagnostic and Statistical
Manual of Mental Disorders, Fourth Edition (Washington, D.C.:
gene expression (Figure 3). For example, Nestler and
American Psychiatric Press).
coworkers have observed that acute cocaine induces
Anthony, J.C., Warner, L.A., and Kessler, R.C. (1994). Comparative
the transcription factor complex AP-1 in the striatum
epidemiology of dependence on tobacco, ethanol, controlled sub-
and nucleus accumbens. With continued exposure to stances, and inhalants: basic findings from the National Comorbidity
drugs, the composition of AP-1 gradually changes from Survey. Exp. Clin. Psychopharmacol. 2, 244–268.
one in which the immediate early-gene product, c-fos, Bailey, C.H., Bartsch, D., and Kandel, E.R. (1996). Toward a molecu-
is present, to one in which a longer-lived, Fos-related lar definition of long-term memory storage. Proc. Natl. Acad Sci.
antigen (termed “chronic FRA”) substitutes for c-fos USA 93, 13445–13452.
(Hope et al., 1994). Because of the longer half-life of Caine, S.B., Heinrichs, S.C., Coffin, V.L., and Koob, G.F. (1995).
chronic FRA, repeated exposure to cocaine leads to its Effects of the dopamine D1 antagonist SCH 23390 microinjected
progressive accumulation (Hope et al., 1994). Shifts of into the accumbens, amygdala or striatum on cocaine self-adminis-
AP-1 composition in relevant CNS regions may repre- tration in the rat. Brain Res. 692, 47–56.
sent a general neuroadaptive process that contributes Carelli, R.M., and Deadwyler, S.A. (1996). Dual factors controlling
to long-term functional changes (Rossetti and Carboni, activity of nucleus accumbens cell-firing during cocaine self-admin-
istration. Synapse 24, 308–311.
1995).
Chang, J.Y., Sawyer, S.F., Lee, R.-S., and Woodward, D.J. (1994).
Nicotine or cocaine self-administration also has been
Electrophysiological and pharmacological evidence for the role of
shown to elicit overlapping patterns of c-fos induction the nucleus accumbens in cocaine self-administration in freely mov-
followed by induction of FRA immunoreactivity in the ing rats. J. Neurosci. 14, 1224–1244.
nucleus accumbens and other regions, but not in the Chen, J.P., Paredes, W., Lowinson, J.H., and Gardner, E.L. (1991).
amygdala (Merlo-Pich et al., 1997). A recent study Strain-specific facilitation of dopamine efflux by delta 9-tetrahydro-
showed c-fos induction in the nucleus accumbens, cen- cannabinol in the nucleus accumbens of rat: an in vivo microdialysis
tral amygdala, and bed nucleus of the stria terminalis study. Neurosci. Lett. 129, 136–180.
following acute exposure to a synthetic cannabinoid Collins, A.C., Bhat, R.V., Pauly, J.R., and Marks, M.J. (1990). Modula-
agonist (Rodriguez de Fonseca et al., 1997). The differ- tion of nicotine receptors by chronic exposure to nicotinic agonists
ences in activation patterns of immediate-early genes and antagonists. In The Biology of Nicotine Dependence, G. Bock
and J. Marsh, eds. (New York: John Wiley), pp. 87–105.
and related proteins, such as the FRAs, by different
classes of drugs within neurons of the extended amyg- Corrigall, W.A., Franklin, K.B.J., Coen, K.M., and Clarke, P.B.S.
(1992). The mesolimbic dopaminergic system is implicated in the
dala may underlie their specific mechanisms of action.
reinforcing effects of nicotine. Psychopharmacology 107, 285–289.
Taken together, these observations support the po-
Dani, J.A., and Heinemann, S. (1996). Molecular and cellular aspects
tential contribution of alteration in gene expression to of nicotine abuse. Neuron 16, 905–908.
the long-term neuroadaptive changes associated with
Di Chiara, G., and Imperato, A. (1988). Drugs abused by humans
the motivational aspects of drug dependence. However, preferentially increase synaptic dopamine concentrations in the
of critical importance will be the correlation over time mesolimbic system of freely moving rats. Proc. Natl. Acad. Sci. USA
of the specific biochemical changes that account for 85, 5274–5278.
increased vulnerability for drug seeking in defined com- Di Chiara, G., and North, R.A. (1992). Neurobiology of opiate abuse.
ponents of all aspects of the addiction cycle: acquisition, Trends Pharmacol. Sci. 13, 185–193.
Review
475

Diana, M., Pistis, M., Carboni, S., Gessa, G.L., and Rossetti, Z.L. process and drug dependence: neurobiological mechanisms.
(1993). Profound decrement of mesolimbic dopaminergic neuronal Semin. Neurosci. 5, 351–358.
activity during ethanol withdrawal syndrome in rats: Electrophysio- Koob, G.F., Heinrichs, S.C., Menzaghi, F., Merlo-Pich, E., and Brit-
logical and biochemical evidence. Proc. Natl. Acad. Sci USA 90, ton, K.T. (1994a). Corticotropin releasing factor, stress and behavior.
7966–7969. Semin. Neurosci. 6, 221–229.
Diaz, J., Lévesque, D., Lammers, C.H., Griffon, N., Martres, M.-P., Koob, G.F., Rassnick, S., Heinrichs, S., and Weiss, F. (1994b). Alco-
Schwartz, J.-C., and Sokoloff, P. (1995). Phenotypical characteriza- hol, the reward system and dependence. In Toward a Molecular
tion of neurons expressing the dopamine D3 receptor in the rat Basis of Alcohol Use and Abuse (Series Title: Experientia Supple-
brain. Neuroscience 65, 731–745. mentum, Volume 71), B. Jansson, H. Jörnvall, U. Rydberg, L. Tere-
Engel, J.A., Enerback, C., Fahlke, C., Hulthe, P., Hard, E., Johannes- nius, and B.L. Vallee, eds. (Boston: Birkhauser-Verlag), pp. 103–114.
sen, K., Svensson, L., and Soderpalm, B. (1992). Serotonergic and Koob, G.F., Carrera, M.R.A., Gold, L.H., Heyser, C.J., Maldonado-
dopaminergic involvement in ethanol intake. In Novel Pharmacologi- Irizarry, C., Markou, A., Parsons, L.H., Roberts, A.J., Schulteis, G.,
cal Interventions for Alcoholism, C.A. Naranjo and E.M. Sellers, eds. Stinus, L., et al. (1998). Substance dependence as a compulsive
(New York: Springer), pp. 68–82. behavior. J. Psychopharmacol. 12, 39–48.
Epping-Jordan, M.P., Markou, A., and Koob, G.F. (1998a). The dopa- Kreek, M.J. (1987). Multiple drug abuse patterns and medical conse-
mine D1 receptor antagonist SCH 23390 injected into the dorsolat- quences. In Psychopharmacology: the Third Generation of Progress,
eral bed nucleus of the stria terminalis decreased cocaine reward H.Y. Meltzer, ed. (New York: Raven Press), pp. 1597–1604.
in the rat. Brain Res. 784, 105–115. Lake, J.R., Hebert, K.M., Payza, K., Deshotel, K.D., Hausam, D.D.,
Epping-Jordan, M.P., Watkins, S.S., Koob, G.F., and Markou, A. Witherspoon, W.E., Arcangeli, K.A., and Malin, D.H. (1992). Analog
(1998b). Dramatic decreases in brain reward function during nicotine of neuropeptide FF attenuates morphine tolerance. Neurosci. Lett.
withdrawal. Nature 392, 869–870. 146, 203–206.
Everitt, B.J., Morris, K.A., O’Brien, A., and Robbins, T.W. (1991). The LeMarquand, D., Pihl, R.O., and Benkelfat, C. (1994). Serotonin and
basolateral amygdala–ventral striatal system and conditioned place ethanol intake, abuse and dependence: findings of animal studies.
preference: further evidence of limbic–striatal interactions underly- Biol. Psychiatry 36, 395–421.
ing reward-related processes. Neuroscience 42, 1–18. Le Novere, N., Zoli, M., and Changeux, J.-P. (1996). Neuronal nico-
Gardner, E.L., and Vorel, S.R. (1998). Cannabinoid transmission and tinic receptor a6 subunit mRNA is selectively concentrated in cate-
reward-related events. Semin. Neurosci., in press. choliminergic nuclei of the rat brain. Eur. J. Neurosci. 8, 2428–2439.
Lepore, M., Vorel, S.R., Lowinson, J., and Gardner, E.L. (1995). Con-
Gardner, E.L., Paredes, W., Smith, D., Donner, A., Milling, C., Cohen,
ditioned place preference induced by delta 9-tetrahydrocannabinol:
D., and Morrison, D. (1988). Facilitation of brain stimulation reward
comparison with cocaine, morphine, and food reward. Life Sci. 56,
by delta-9-tetrahydrocannabinol. Psychopharmacology 96, 142–144.
2073–2080.
Giros, B., Jaber, M., Jones, S.R., Wightman, R.M., and Caron, M.G.
Lepore, M., Liu, X., Savage, V., Matalon, D., and Gardner, E.L. (1996).
(1996). Hyperlocomotion and indifference to cocaine and amphet-
Genetic differences in delta 9-tetrahydrocannabinol-induced facili-
amine in mice lacking the dopamine transporter. Nature 379,
tation of brain stimulation reward as measured by a rate-frequency
606–612.
curve-shift electrical brain stimulation paradigm in three different
Headlee, C.P., Coppock, H.W., and Nichols, J.R. (1955). Apparatus rat strains. Life Sci. 58, PL365–PL372.
and technique involved in a laboratory method of detecting the
Liljequist, S., and Engel, J. (1982). Effects of GABAergic agonists
addictiveness of drugs. J. Am. Pharm. Assoc. Sci. Edn. 44, 229–231.
and antagonists on various ethanol-induced behavioral changes.
Heimer, L., and Alheid, G. (1991). Piecing together the puzzle of Psychopharmacology 78, 71–75.
basal forebrain anatomy. In The Basal Forebrain: Anatomy to Func- Maldonado, R., Blendy, J.A., Tzavara, E., Gass, P., Roques, B.P.,
tion, T.C. Napier, P.W. Kalivas, and I. Hanin, eds. (New York: Plenum Hanoune, J., and Schutz, G. (1996). Reduction of morphine absti-
Press), pp. 1–42. nence in mice with a mutation in the gene encoding CREB. Science
Henry, D.J., and White, F.J. (1991). Repeated cocaine administration 273, 657–659.
causes persistent enhancement of D1 dopamine receptor sensitivity Malin, D.H., Lake, J.R., Hammond, M.V., Fowler, D.E., Rogillio, R.B.,
within the rat nucleus accumbens. J. Pharmacol. Exp. Ther. 258, Brown, S.L., Sims, J.L., Leecraft, B.M., and Yang, H.-Y.T. (1990).
882–890. FMRF-NH2-like mammalian octapeptide: possible role in opiate de-
Herkenham, M., Lynn, A.B., Little, M.D., Johnson, M.R., Melvin, L.S., pendence and abstinence. Peptides 11, 969–972.
de Costa, B.R., and Rice, K.C. (1990). Cannabinoid receptor localiza- Mansour, A., Watson, S.J., and Akil, H. (1995). Opioid receptors:
tion in brain. Proc. Natl. Acad. Sci. USA 87, 1932–1936. Past, present and future. Trends Neurosci. 18, 69–70.
Hope, B.T., Kelz, M.B., Duman, R.S., and Nestler, E.J. (1994). Chronic Matthes, H.W.D., Maldonado, R., Simonin, F., Valverde, O., Slowe,
electroconvulsive seizure (ECS) treatment results in expression of S., Kitchen, I., Befort, K., Dierich, A., Le Meur, M., Dolle, P., et
a long-lasting AP-1 complex in brain with altered composition and al. (1996). Loss of morphine-induced analgesia, reward effect and
characteristics. J. Neurosci. 14, 4318–4328. withdrawal symptoms in mice lacking the mu-opioid-receptor gene.
Hyman, S.E. (1996). Addiction to cocaine and amphetamine. Neuron Nature 383, 819–823.
16, 901–904. Merlo-Pich, E., Lorang, M., Yeganeh, M., Rodriguez de Fonseca, F.,
Hyttia, P., and Koob, G.F. (1995). GABAA receptor antagonism in the Raber, J., Koob, G.F., and Weiss, F. (1995). Increase of extracellular
extended amygdala decreases ethanol self-administration in rats. corticotropin-releasing factor-like immunoreactivity levels in the
Eur. J. Pharmacol. 283, 151–159. amygdala of awake rats during restraint stress and ethanol with-
Koob, G.F. (1992). Drugs of abuse: anatomy, pharmacology, and drawal as measured by microdialysis. J. Neurosci. 15, 5439–5447.
function of reward pathways. Trends Pharmacol. Sci. 13, 177–184. Merlo-Pich, E., Pagliusi, S.R., Tessari, M., Talabot-Ayer, D., Hooft
Koob, G.F. (1995). Animal models of drug addiction. In Psychophar- van Huijsduijnen, R., and Chiamulera, C. (1997). Common neural
macology, The Fourth Generation of Progress, F.E. Bloom and D.J. substrates for the addictive properties of nicotine and cocaine. Sci-
ence 275, 83–86.
Kupfer, eds. (New York: Raven Press), pp. 759–772.
Merue, G., and Gessa, G.L. (1985). Low doses of ethanol inhibit the
Koob, G.F. (1996). Drug addiction: the yin and yang of hedonic
firing of neurons in the substantia nigra, pars reticulata: a GABAergic
homeostasis. Neuron 16, 893–896.
effect? Brain Res. 360, 325–330.
Koob, G.F., and Bloom, F.E. (1988). Cellular and molecular mecha-
Negus, S.S., Henriksen, S.J., Mattox, A., Pasternak, G.W., Porto-
nisms of drug dependence. Science 242, 715–723.
ghese, P.S., Takemori, A.E., Weinger, M.B., and Koob, G.F. (1993).
Koob, G.F., and Le Moal, M. (1997). Drug abuse: hedonic homeo- Effect of antagonists selective for mu, delta and k opioid receptors
static dysregulation. Science 278, 52–58. on the reinforcing effects of heroin in rats. J. Pharmacol. Exp. Ther.
Koob, G.F., Markou, A., Weiss, F., and Schulteis, G. (1993). Opponent 265, 1245–1252.
Neuron
476

Nestler, E.J. (1994). Molecular neurobiology of drug addiction. Neu- Tabakoff, B., and Hoffman, P.L. (1996). Alcohol addiction: an enigma
ropsychopharmacology 11, 77–87. among us. Neuron 16, 909–912.
Nestler, E.J. (1996). Under siege: the brain on opiates. Neuron 16, Tanda, G., Pontieri, F.E., and Di Chiara, G. (1997a). Cannabinoid
897–900. and heroin activation of mesolimbic dopamine transmission by a
Olale, F., Gerzanich, V., Kuryatov, R., Wang, F., and Lindstrom, J. common mu1 opioid receptor mechanism. Science 276, 2048–2050.
(1997). Chronic nicotine exposure differentially affects the function Tanda, G., Pontieri, F.E., Frau, R., and Di Chiara, G. (1997b). Contri-
of human a3, a4, and a7 neuronal nicotinic receptor subtypes. J. bution of the noradrenaline carrier to the increase of extracellular
Pharmacol. Exp. Ther. 283, 675–683. dopamine in the rat prefrontal cortex by amphetamine and cocaine.
Parsons, L.H., Koob, G.F., and Weiss, F. (1995). Serotonin dysfunc- Eur. J. Neurosci. 9, 2077–2085.
tion in the nucleus accumbens of rats during withdrawal after unlim- Volpicelli, J.R., Alterman, A.I., Hayashida, M., and O’Brien, C.P.
ited access to intravenous cocaine. J. Pharmacol. Exp. Ther. 274, (1992). Naltrexone in the treatment of ethanol dependence. Arch.
1182–1191. Gen. Psychiatry 49, 876–880.
Peoples, L.L., and West, M.O. (1996). Phasic firing of single neurons Weiss, F., Markou, A., Lorang, M.T., and Koob, G.F. (1992). Basal
in the rat nucleus accumbens correlated with the timing of intrave- extracellular dopamine levels in the nucleus accumbens are de-
nous cocaine self-administration. J. Neurosci. 16, 3459–3473. creased during cocaine withdrawal after unlimited-access self-
Peoples, L.L., Uzwiak, A.J., Gee, F., and West, M.O. (1997). Operant administration. Brain Res. 593, 314–318.
behavior during sessions of intravenous cocaine infusion is neces-
Weiss, F., Parsons, L.H., Schulteis, G., Hyytia, P., Lorang, M.T.,
sary and sufficient for phasic firing of single nucleus accumbens
Bloom, F.E., and Koob, G.F. (1996). Ethanol self-administration re-
neurons. Brain Res. 757, 280–284.
stores withdrawal-associated deficiencies in accumbal dopamine
Pettit, H.-O., and Justice, J.B., Jr. (1989). Dopamine in the nucleus and 5-hydroxytryptamine release in dependent rats. J. Neurosci. 16,
accumbens during cocaine self-administration as studied by in vivo 3474–3485.
microdialysis. Pharmacol. Biochem. Behav. 34, 899–904.
Widnell, K., Self, D.W., Lane, S.B., Russell, D.S., Vaidya, V., Miseren-
Pontieri, F.E., Tanda, G., and Di Chiara, G. (1995). Intravenous co- dino, M.J.D., Rubin, C.S., Duman, R.S., and Nestler, E.J. (1996).
caine, morphine, and amphetamine preferentially increase extracel- Regulation of CREB expression: in vivo evidence for a functional
lular dopamine in the “shell” as compared with the “core” of the rat role in morphine action in the nucleus accumbens. J. Pharmacol.
nucleus accumbens. Proc. Natl. Acad. Sci. USA 92, 12304–12308. Exp. Ther. 276, 306–315.
Roberts, A.J., Cole, M., and Koob, G.F. (1996). Intra-amygdala mus- Woolverton, W.L., and Johnson, K.M. (1992). Neurobiology of co-
cimol decreases operant ethanol self-administration in dependent
caine abuse. Trends Pharmacol. Sci. 13, 193–200.
rats. Alcohol Clin. Exp. Res. 20, 1289–1298.
Zadina, J.E., Hackler, L., Ge, L.J., and Kastin, A.J. (1997). A potent
Roberts, D.C.S., Koob, G.F., Klonoff, P., and Fibiger, H.C. (1980).
and selective endogenous agonist for the m opiate receptor. Nature
Extinction and recovery of cocaine self-administration following
386, 499–502.
6-hydroxydopamine lesions of the nucleus accumbens. Pharmacol.
Biochem. Behav. 12, 781–787.
Note Added in Proof
Rocha, B.A., Fumagalli, F., Gainetdinov, R.R., Jones, S.R., Ator,
R., Giros, B., Miller, G.W., and Caron, M.G. (1998). Cocaine self- The data referred to as “Richter and Weiss, personal communica-
administration in dopamine-transporter knockout mice. Nature Neu- tion,” are now in press: Richter, R.M., and Weiss, F. (1998). In vivo
rosci. 1, 132–137. CRF release in rat amygdala is increased during cocaine withdrawal
Rodriguez de Fonseca, F., Carrera, M.R.A., Navarro, M., Koob, G.F., in self-administering rats. Synapse, in press.
and Weiss, F. (1997). Activation of corticotropin-releasing factor
in the limbic system during cannabinoid withdrawal. Science 276,
2050–2054.
Rossetti, Z.L., and Carboni, S. (1995). Ethanol withdrawal is associ-
ated with increased extracellular glutamate in the rat striatum. Eur.
J. Pharmacol. 283, 177–183.
Russell, M.A.H. (1976). What is dependence? In Drugs and Drug
Dependence, G. Edwards, M.A.H. Russell, D. Hawks, and M. Mac-
Cafferty, eds. (Lexington: MA, Lexington Books), pp. 182–187.
Samson, H.H., and Harris, R.A. (1992). Neurobiology of ethanol
abuse. Trends Pharmacol. Sci. 13, 206–211.
Schuster, C.R., and Thompson, T. (1969). Self-administration of and
behavioral dependence on drugs. Annu. Rev. Pharmacol. 9,
483–502.
Self, D.W., and Nestler, E.J. (1995). Molecular mechanisms of drug
reinforcement and addiction. Annu. Rev. Neurosci. 18, 463–495.
Self, D.W., Terwilliger, R.Z., Nestler, E.J., and Stein, L. (1994). Inacti-
vation of Gi and Go proteins in nucleus accumbens reduces both
cocaine and heroin reinforcement. J. Neurosci. 14, 6239–6247.
Self, D.W., Barnhart, W.J., Lehman, D.A., and Nestler, E.J. (1996).
Opposite modulation of cocaine-seeking behavior by D1- and D2 -
like dopamine receptor agonists. Science 271, 1586–1589.
Sellers, E.M., Higgins, G.A., and Sobell, M.B. (1992). 5-HT and etha-
nol abuse. Trends Pharmacol. Sci. 13, 69–75.
Shippenberg, T.S., Herz, A., Spanagel, R., Bals-Kubik, R., and Stein,
C. (1992). Conditioning of opioid reinforcement: neuroanatomical
and neurochemical substrates. Ann. NY Acad. Sci. 654, 347–356.
Stinus, L., Le Moal, M., and Koob, G.F. (1990). Nucleus accumbens
and amygdala are possible substrates for the aversive stimulus
effects of opiate withdrawal. Neuroscience 37, 767–773.
Sun, N., and Cassell, M.D. (1993). Intrinsic GABAergic neurons in
the rat central extended amygdala. J. Comp. Neurol. 330, 381–404.

Potrebbero piacerti anche