You are on page 1of 14

XVI-7

Functional Anatomy of Substance-Related Disorders


R.A. Wise and E.L. Gardner

For the most part, the functional anatomy relevant to substance cocaine (Jones, 1984). Addiction theory has, at least until recently,
abuse disorders remains to be identified. One problem is that been dominated by the assumption that the self-medication of
the field of substance-related disorders is not yet well defined. withdrawal distress differentiates between addiction and the so-
For the purposes of the present chapter, the substances under called ‘recreational’ use of drugs. The present chapter thus begins
discussion will be drugs of potential abuse. However, the category with a discussion of some of the issues and neuroanatomy of drug
of ‘food’ includes at least two classes of substances — sweets and dependence before turning to the topic of the neuroanatomy of drug
lipids — that can also be abused. The case of food underlines an reward.
important principle: substance abuse need not involve the kinds of
physiological dependence that have been assumed in the case of
drug addiction. Food is ‘abused’ only when it is taken above and
beyond any obvious physiological need. Current evidence suggests NEUROANATOMY OF DRUG DEPENDENCE
significant overlap in the brain circuitry relevant to foraging for Drug dependence develops with chronic drug use and is revealed in
food and foraging for drugs of abuse. For the most part, such ‘withdrawal symptoms’ that are seen upon interruption of drug use.
circuitry has been identified as critical for the substance of interest There is no common denominator of the range of drug dependence
to serve as a reward or ‘reinforcer’. The terms reward and reinforcer phenomena; the symptoms produced by withdrawal from a given
are descriptive, not explanatory; they merely identify substances or drug depend on the specific actions of that drug. Generally, the
events that are habit-forming. withdrawal symptoms associated with a given drug are opposite to
The concept of habit is central to the topic of substance abuse. the symptoms of intoxication with that drug. Thus withdrawal from
Again, the term is descriptive rather than explanatory. All substance stimulants is generally associated with depression and withdrawal
abuse reflects the habitual intake of a substance. Some habits are from depressants is generally associated with agitation. As a
benign, such as the habit of eating a small portion of pastry or ice general rule, dependence is seen as resulting from ‘compensatory’
cream following a meal. We invoke the notion of substance abuse
mechanisms: mechanisms that resist intoxication and tend, over
in relation to habits that are pursued compulsively despite negative
time, to re-establish normal equilibrium. These compensations also
consequences. The consequences most obviously associated with
result in tolerance (progressive desensitization) to the intoxicating
substance abuse are addiction and obesity. The transition from
effects of the drug and when the drug is withdrawn the unopposed
recreational drug use to addiction is gradual, just as is the transition
compensations produce the objective signs of the withdrawal
from overweight to obese, and we have no evidence to suggest that
syndrome. If a system has no tonic endogenous input, it is possible
the mechanisms of modest food or drug intake differ in anatomical
to observe tolerance of the response to an exogenous drug in
loci from those of excessive intake. Thus our current knowledge
the absence of any overt symptoms when the drug is withdrawn.
of the neuroanatomy of substance abuse is based largely on our
understanding of why individuals take food and drug substances in Usually, however, tolerance and dependence go together.
the first place. To the degree that drugs of different classes share common
In one sense, the task of identifying the neuroanatomy of drug actions, they tend to establish overlapping tolerance and dependence
abuse is an easier one than that of identifying the neuroanatomy syndromes and they are at least partially effective at alleviating each
of feeding: most drugs of abuse act at receptors that have been other’s withdrawal symptoms. The ability of one drug to cause tol-
identified and localized in the depths of the brain. Unlike foods, erance that extends to another drug is termed ‘cross-tolerance’; the
drug rewards do not rely on peripheral sensory pathways to ability of one drug to alleviate the withdrawal symptoms associated
reach the brain mechanisms serving motivational function. While with another drug is termed ‘cross-dependence’. Even drugs such
addictive drugs act on multiple receptor populations, and while as opiates, alcohol and barbiturates, which produce partial cross-
only a fraction of these populations serve motivational functions, tolerance and partial cross-dependence, have some actions — and
by injecting drugs directly into the brain and comparing the thus cause some adaptations — that are not shared (Kalant, 1977).
effectiveness at different injection sites, we are able to identify There is a good deal of cross-tolerance between depressant drugs
chemical trigger zones for the habit-forming actions of several drugs (opiates, alcohol, barbiturates and benzodiazepines) but little if any
of abuse. These trigger zones and the pathways that connect them between depressant drugs of abuse and stimulant drugs of abuse
can serve as a first step towards identifying the brain structures (Wise, 1987).
involved in substance-related disorders.
A classic issue in the field of drug abuse is the role of drug ‘Somatic’ Dependence Signs
dependence. Clear-cut dependence syndromes can develop with
chronic use of opiates, alcohol, barbiturates and benzodiazepines Pursuit of the anatomy of drug dependence mechanisms has
(Kalant, 1977), and more subtle dependence syndromes can develop been fuelled by classic addition theory, which held that the self-
with use of nicotine (Shiffman, 1979), cannabis (Jones, 1980) or medication of withdrawal distress was a central if not a defining

Biological Psychiatry : Edited by H. D’haenen, J.A. den Boer and P. Willner. ISBN 0-471-49198-5
 2002 John Wiley & Sons, Ltd.
510 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

property of addiction (Tatum et al., 1929; Himmelsbach, 1943; Lin- matter or into the fourth ventricle, which is fed by the aqueduct,
desmith, 1947; Goldstein and Goldstein, 1961; Collier, 1966; Jaffe systemic naloxone precipitates the somatic signs of teeth chattering
and Sharpless, 1968). Inasmuch as the withdrawal symptoms asso- and escape behaviours (Wei and Loh, 1976; Wei, 1981; Bozarth
ciated with such drugs as opiates and alcohol are unpleasant, and and Wise, 1984). One hypothesis holds that these symptoms are
inasmuch as addicts report that they are ‘sick’ during withdrawal responses to the fluctuations of body temperature that accompany
and that they take their drug to get ‘well’, this theory had obvious opiate withdrawal and result from opiate-induced disturbance of
face validity. From the perspective of classic addiction theory, the temperature-regulating mechanisms (Wei et al., 1974).
anatomy of drug dependence should be the anatomy of the brain cir- Of the sites that are readily reached by drug injections into the
cuits regulating thermoregulation, autonomic function and the other fourth ventricle, nucleus locus coeruleus and the periaqueductal
somatic signs of drug withdrawal — the signs that addicts report as grey have received most attention. The noradrenergic neurones of
aversive and liken to some of the symptoms of influenza. the locus coeruleus are inhibited by morphine; this inhibition under-
Our best knowledge of neuroanatomy of such dependence goes tolerance with repeated application (Aghajanian, 1978). In
phenomena involves opiate dependence. In part, this is because opiate-dependent animals, the opiate antagonist naloxone precip-
the opiate withdrawal syndrome has frequently served as the model itates accelerated firing of these neurones (Aghajanian, 1978); at
of other addictions. In part, it is because opiates can be injected least part of this hyperactivity appears to involve adaptations of
directly into the brain sites where they have various actions and the locus coeruleus itself (Ivanov and Aston-Jones, 2001). Part of
where they remain reasonably localized for many minutes. In part, this hyperactivity results from altered neuronal input to these cells
it is because opiate withdrawal symptoms can be precipitated by (Akaoka and Aston-Jones, 1991). Thus while the cells of the locus
administration of opiate antagonists, which can also be restricted coeruleus appear to be one anatomical substrate of dependence, the
to localized brain areas. Localization is relative, of course; drugs cells that provide input to the locus coeruleus are another. Presum-
injected into the brain can diffuse to adjacent structures, to the ably, the many cells that receive input from the locus coeruleus are
ventricular system and into the circulation, from which they can yet other such substrates.
reach distant targets of action. Adequate localization of the effects Indeed, most of the brain is involved directly or indirectly in
of direct drug injections into the brain requires the demonstration the opiate dependence syndrome. Signs of abnormal activity in
that higher doses are required to produce the same effects when opiate-withdrawn animals are seen in the abnormal expression of
the injections are in adjacent regions, into the ventricles or into early immediate genes in a variety of brain areas associated with
the circulation. This is a requirement that is frequently not met in autonomic function. Among these are the locus coeruleus and the
studies involving central drug injections. A5 noradrenergic cell group, the nucleus of the solitary tract, the
Localized brain injections of opiates and opiate antagonists have parabrachial nuclei, and the caudal and rostral ventrolateral medulla
been used to identify sites at which opiates act to cause the various (Stornetta et al., 1993). Moreover, it is not just autonomic control
symptoms of opioid intoxication and subsequent withdrawal, and systems that are altered by opiate dependence. Indeed, studies of
where opiate antagonists precipitate the various somatic symptoms early immediate gene expression confirm that opiate withdrawal is
of the opiate withdrawal syndrome in dependent laboratory animals. accompanied by abnormal neuronal activity in multiple structures
The somatic symptoms include diarrhoea, ear blanching, ptosis, and at all levels of the neuraxis. Among additional sites where
teeth chattering, ‘wet dog’ shakes, seminal emissions and in rats, immediate early genes are activated during opiate withdrawal are
attempts to escape from the test chamber. The human symptoms are the paraventricular nucleus of the hypothalamus, amygdala, ven-
similar: lacrimation, rhinorrhoea, yawning, sweating, restless sleep, tral tegmentum, nucleus accumbens, neostriatum, cerebral cortex,
dilated pupils, tremor, nausea and vomiting, diarrhoea, increased hippocampus, thalamus, cerebellum and various layers of the spinal
heart rate and blood pressure, chills, abdominal cramping, muscle cord (Hayward et al., 1990; Stornetta et al., 1993; Beckmann et al.,
spasms and sexual orgasm (both male and female). 1995; Rohde et al., 1996).
The brain mechanism of a given somatic withdrawal symptom The fact that opiate withdrawal effects are widespread in the cen-
is the brain mechanism involved in the normal function that is tral nervous system makes a general search for an anatomical basis
distorted by intoxication and subsequent withdrawal. The diarrhoea of opiate dependence a questionable enterprise. The problem is
during opiate withdrawal reflects an adaptation in the mechanism confounded when other drugs of abuse are considered, particularly
responsible for the constipation during opiate intoxication: the because there is no common denominator to stimulant and depres-
mechanism of µ-opioid inhibition of intestinal contractility. During sant somatic dependence signs (Wise, 1987). The affected circuitry
intoxication the intestine relaxes; during withdrawal it contracts is not only widespread; it also differs from drug to drug. Even
and expels the accumulated faeces. In the case of each withdrawal when we restrict consideration to the opiates, dependence appears
symptom, a mechanism specific to that symptom is involved. The to be more easily characterized in neurochemical than in anatomical
sweats and chills of opiate withdrawal reflect the actions of opiates terms. Opiate receptors are G-protein-coupled, and the exposure of
and opiate withdrawal on the mechanisms of regulation of body G-protein-coupled receptors to agonists usually results in desensiti-
temperature. Thus the anatomy of opiate withdrawal is, essentially, zation of the receptor (Ferguson et al., 1998; Pitcher et al., 1998).
the anatomy of opiate action. Not surprisingly, very many brain Other drugs of abuse also act on (e.g., cannabis) or via (e.g.,
structures are implicated in the full range of opiate and other drug cocaine, amphetamine and, more indirectly, nicotine) G-protein-
dependence symptoms. coupled receptors. Rebounding supersensitivity of such receptors
Among the first opiate withdrawal symptoms to be associated will, where the receptor usually receives endogenous input, result
with specific brain regions were the somatic signs of ptosis, teeth in withdrawal symptoms. This fact appears to confirm what has
chattering, wet dog shakes, and escape behaviour. These symptoms already been suggested: every site of opiate action can contribute
can be precipitated in opiate-dependent rats by injections of opiate to the complex phenomenon of opiate dependence. Localization of
antagonists into the ventricular system of the brain (Laschka et al., the mechanisms of opiate or other drug dependence thus requires
1976a). If the cerebral aqueduct is blocked so that antagonists a symptom-by-symptom characterization that may never be com-
injected into the lateral or the third ventrical do not have easy pleted. Interest in drug withdrawal symptoms has thus tended to
access to the fourth ventral, it is injections into the fourth ventricle focus on the specific symptoms most likely to contribute to the
(Laschka et al., 1976b) that are most effective. Injections into motivation for drug self-administration.
the nucleus locus coeruleus, at the lateral floor of the fourth Classic addiction theory — with its myriad withdrawal symp-
ventrical, are similarly effective (Maldonado et al., 1992). If the toms and syndromes — has always had as a primary weakness that
opiates themselves are infused locally into the periaqueductal grey it does not explain why non-dependent subjects initiate sufficient
FUNCTIONAL ANATOMY 511

drug intake to become dependent, nor does it explain why detox- A variety of neuroadaptations have been found within the
ified addicts are re-addicted so quickly. Broad dependence theory mesolimbic dopamine system and its primary terminal field, the
has thus been challenged by workers who hypothesize that it is the nucleus accumbens. These loci are central to current knowledge
generation of drug-induced positive reinforcement and euphoria, of brain reward circuitry, and neuroadaptations in these loci are
rather than the alleviation of withdrawal-induced dysphoria, that of potential relevance to drug dependence. The known neuroad-
motivates compulsive drug-seeking in addicts (McAuliffe and Gor- aptations range from morphological changes (Beitner-Johnson and
don, 1974; Bijerot, 1980; Wise and Bozarth, 1987; Gardner, 1992; Nestler, 1991; Beitner-Johnson et al., 1992; Sklair-Tavron et al.,
Robinson and Berridge, 1993; Di Chiara, 1998). This view gained 1996) to changes in neurotransmitter sensitivity (Bell et al., 2000;
support from the fact that stimulant drugs like cocaine, nicotine and Henry and White, 1991) and changes in intracellular signalling
cannabis can be taken compulsively despite withdrawal symptoms cascades and transcription factors (Cha et al., 1997; Chen et al.,
quite different from and much weaker than those associated with 1997; Fitzgerald et al., 1996; Hope et al., 1992; Kelz et al., 1999;
depressant drugs (Shiffman, 1979; Jones, 1980, 1984). Further sup- Nestler et al., 1990; Terwilliger et al., 1991). Some of these
port came from studies showing that lower animals would learn changes are associated with decreased sensitivity — tolerance — to
to take morphine, heroin, amphetamine, cocaine and related drugs the drug, while others are associated with increased sensitiv-
compulsively even if they were not first made dependent. Indeed, ity — sensitization. Mechanisms of tolerance are consistent with
animals will self-administer opiates in doses and limited access con- traditional dependence theory, but mechanisms of sensitization are
ditions that cause no overt somatic withdrawal symptoms (Deneau opposite to those associated with dependence theory, and the find-
et al., 1969; Woods and Schuster, 1971). Confirmation that opiates ing that addictive drugs can sensitize the nervous system to the
are strongly habit-forming independent of their ability to induce drug’s rewarding action raises another challenge to dependence the-
physical dependence came from the demonstration that rats self- ory and the question of whether its anatomy is part of the anatomy
administer morphine into the ventral tegmental area of the brain, of substance abuse.
a site at which infusions of doses that are effective elsewhere fail
to produce somatic signs of dependence, but not into the periaque- Sensitization in Reward Pathways
ductal grey, where infusions do cause somatic dependence signs
(Bozarth and Wise, 1984). With the waning influence of depen- Dependence theory is predicated upon the notion that increasing
dence theory, interest in the neuroanatomy of somatic dependence amounts of the drug are needed to ameliorate withdrawal symptoms.
signs has also waned, and dependence theory has come to focus on Dependence theory thus requires that the effects of repeated drug
intoxication-induced neuroadaptations within the specific circuitry experience decrease the sensitivity of the brain to the addictive
of drug reward. drug, and bias normal brain function in the opposite direction
from the acute drug state (Solomon and Corbit, 1973; Koob et al.,
Reward-Relevant Dependence Signs 1989). Some drug effects — in particular those associated with
somatic withdrawal symptoms — do appear to desensitize with
With the realization that opiate dependence was not a necessary chronic drug treatment. The reward-specific effects of amphetamine
condition for compulsive opiate self-administration, the assumption (Lett, 1989; Piazza et al., 1990; Lorrain et al., 2000), cocaine (Lett,
that some form of physical dependence must explain compulsive 1989; Horger et al., 1990; Shippenberg and Heidbreder, 1995) and
self-administration of addictive drugs became less attractive and morphine (Lett, 1989; Shippenberg et al., 1996), however, appear to
classic addiction theory became overshadowed by theories that undergo sensitization, at least with intermittent repeated treatment.
focused on the positive reinforcing properties of drugs for non- Thus while it is true that tolerance to the rewarding effects of
dependent subjects (Fibiger, 1978; Wise, 1978, 1988; Wise and cocaine can occur within a binge of cocaine self-administration
Bozarth, 1987; Di Chiara and Imperato, 1988; Gardner, 1992; (Emmett-Oglesby and Lane, 1992; Li et al., 1994), sensitization
Robinson and Berridge, 1993). The brain circuitry contributing to the rewarding effects of cocaine appear to occur between such
importantly to the positive reinforcing properties of drugs of abuse binges. While tolerance usually decreases within a few days of the
has been a target of intensive investigation and is discussed in some last drug administration, sensitization to intermittent psychomotor
detail in the next section. With the accumulating evidence as to the stimulants is very long lasting (Robinson and Becker, 1986).
neural substrates of drug reward has come a focusing of dependence The finding that between-session sensitization is much stronger
theory on those specific substrates. In the modern reformulation and longer-lasting than any obvious signs of tolerance to the
of dependence theory, neuroadaptations in the reward circuitry rewarding effects of these drugs has led to yet another positive
itself are posited to account for states of generalized depression reinforcement perspective on addiction (Robinson and Berridge,
and anhedonia and it is hypothesized that it is the self-medication 1993). This perspective has again raised the question of whether
of such states that adds the compulsive dimension to drug self- drug dependence phenomena play the significant role in addiction
administration (Solomon and Corbit, 1973; Leith and Barrett, 1976; that has been suggested by dependence theory. The sensitization-
Kokkinidis et al., 1980; Dackis and Gold, 1985; Kokkinidis and of-reward findings directly challenge all forms of dependence
McCarter, 1990; Frank et al., 1992; Koob and Bloom, 1988). or opponent process theory and raise, again, serious questions
The first suggestion that drugs of abuse might desensitize the about the relevance of the neuroanatomy of dependence to the
mechanisms of reward came from studies of the effects of drugs on neuroanatomy of substance abuse. While it is clear that there
rewarding electrical stimulation of the brain. While drugs of abuse are drug-induced neuroadaptations within the reward system itself,
usually potentiate the rewarding effects of lateral hypothalamic and the importance of drug-opposite or drug-like neuroadaptations for
related brain stimulation (Wise, 1996b), reward thresholds are ele- drug self-administration require direct experiments that have only
vated following withdrawal from chronic exposure to amphetamine recently begun (Carlezon et al., 1998; Kelz et al., 1999).
(Leith and Barrett, 1976; Kokkinidis et al., 1980; Wise and Munn,
1995), cocaine (Frank et al., 1988, 1992; Kokkinidis and McCarter,
1990; Markou and Koob, 1991), ethanol (Schulteis et al., 1995) and NEUROANATOMY OF DRUG REWARD
nicotine (Watkins et al., 2000). The period of elevation lasts, how-
ever, only a few days, far short of the period when peak craving While it does not make obvious sense to discuss a ‘trigger zone’
for opiates (Shalev et al., 2001) or cocaine (Grimm et al., 2001) for the rewarding effect of such natural incentives as food or
occurs. sexual contact, it is well established that food, like the psychomotor
512 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

stimulants and a number of other drugs of abuse, depends upon can reach a distant site of action in three ways. First, it may
dopamine neurotransmission for its rewarding effects (Wise et al., simply diffuse through the extracellular space to adjacent structures.
1978; Spyraki et al., 1982; Ettenberg and Camp, 1986). The Only if injections into surrounding structures are less effective
major dopamine systems originate in the midbrain and have long- and occur with greater latency can we conclude that a drug is
axon projections to the forebrain. These neurones originate as acting in a given target site. Second, drugs that readily cross
one embryonic group, but the somata spread laterally from the the blood– brain barrier can reach distal sites of action via the
midline and are somewhat arbitrarily subdivided into a nigrostriatal circulation. The obvious safeguard against this possibility is the
system, originating primarily in the substantia nigra and projecting demonstration that the same low doses are ineffective when given
primarily to the caudate nucleus, and a mesocorticolimbic system, intravenously. Finally drugs injected under hydraulic pressure can
originating primarily in the more medial ventral tegmental area follow the pressure gradient up the cannula shaft, and, if the cannula
and projecting primarily to the frontal and cingulate cortices, the penetrates a ventricle, to the ventricular pressure sink through
septum, hippocampus, amygdala and the nucleus accumbens. Of which it can reach distal circumventricular organs. This artefact
the various dopaminergic terminal fields, the nucleus accumbens led to misinterpretation of several studies involving the effects
is most clearly seen as serving motivational function (Mogenson of central hormone and neurotransmitter injections on drinking
et al., 1980). It appears to house trigger zones — regions where the behaviour (Johnson and Epstein, 1975). In the case of intracranial
drug binds and initiates its relevant pharmacological action — for self-administration of PCP, fluid egress up the cannula shaft is
the rewarding effects of at least three classes of addictive drug. the most likely of these possibilities. The lateral ventricle is just
above the nucleus accumbens, and a vertical penetration to the
shell of the nucleus accumbens usually involves penetration of the
Nucleus Accumbens
ventricle, establishing a pressure gradient from the cannula tip to
the ventricular system (where fluid flows naturally from the lateral
The nucleus accumbens appears to be the primary site of rewarding to the third and fourth ventricles and cerebrospinal aqueduct. The
action for amphetamine, one of two sites of rewarding action demonstration that PCP is self-administered into the shell of the
for cocaine and for phencyclidine, and one of two sites of nucleus accumbens (Carlezon and Wise, 1996) has two controls
putative rewarding action for addictive opiates. The mechanisms that increase confidence that the drug’s rewarding action is in the
of rewarding action of amphetamine and cocaine are secondary to nucleus accumbens. First, an angled cannula was used to avoid
their actions at the dopamine transporter, by which they elevate penetration of the ventricle. Second, rats did not self-administer
nucleus accumbens dopamine levels, whereas phencyclidine and PCP to a site in the core of the nucleus accumbens, 1 mm dorsal
morphine have rewarding actions due to their actions at receptors and slightly lateral — along the same cannula track — to the positive
for the neurotransmitter glutamate or for one or more of the reward sites in the shell of the accumbens. Such essential controls
endogenous opioid peptides, respectively. The rewarding action of (Routtenberg, 1972; Wise and Hoffman, 1992) are lacking in most
each appears to result in decreased activity in the medium-sized central injection studies, and this fact leaves interpretation of several
spiny neurones that comprise the output neurones of the nucleus central injection studies in doubt.
accumbens. While much less is known about the habit-forming
actions of cannabis, alcohol, barbiturates, benzodiazepines and
caffeine, there are reasons to suspect that the nucleus accumbens Amphetamine
also plays a part in the habit-forming effects of these potentially
addictive agents. Amphetamine binds to and reverses monoamine transporters, caus-
ing release of noradrenaline (norepinephrine), dopamine and sero-
tonin. The first evidence of a role for dopamine in drug reward came
Phencyclidine from studies in which intravenous amphetamine self-administration
was disrupted by treatment with dopamine-selective (but not
Phencyclidine (PCP) is a non-competitive antagonist at the NMDA- noradrenaline-selective) receptor antagonists (Yokel and Wise,
type excitatory amino acid receptor, where it blocks some of the 1975, 1976; Risner and Jones, 1976). Lesions of the nucleus accum-
excitatory effects of glutamate (Chaudieu et al., 1989; Ohmori bens, like treatment with dopamine receptor blockers, attenuate
et al., 1992). It is also, at higher concentrations, a dopamine intravenous amphetamine self-administration (Lyness et al., 1979).
uptake inhibitor (Gerhardt et al., 1987). It is self-administered Amphetamine is self-administered directly into the nucleus accum-
compulsively by a small but stable subset of humans (Crider, bens but not into the lateral ventricles (Hoebel et al., 1983); this
1986); it is also self-administered by laboratory animals (Balster self-administration of amphetamine is not evident if amphetamine is
et al., 1973), but not with the vigour or reliability that characterizes co-injected with dopamine antagonists (Phillips et al., 1994). Local
intravenous heroin or cocaine self-administration (Collins et al., injections of amphetamine into this region but not adjacent regions
1984). PCP has mixed rewarding and aversive actions when given also establish conditioned place preferences (Carr and White, 1983).
systemically (Barr et al., 1985; Crider, 1986; Iwamoto, 1986), but Thus the nucleus accumbens is a critical site for the habit-forming
has straightforward rewarding actions when given directly into the actions of amphetamine; amphetamine’s rewarding action depends
shell portion of the nucleus accumbens (Carlezon and Wise, 1996). on the activation of local dopamine receptors by the extracellular
PCP shares this rewarding action with NMDA antagonists that dopamine that accumulates (Zetterström et al., 1981) because of
do not affect dopamine uptake, and the rewarding action of these amphetamine’s action at the dopamine transporter.
agents in the nucleus accumbens is not attenuated by sulpiride, a D2 Once a period of amphetamine self-administration has been
dopamine antagonist that attenuates the rewarding effects of nucleus initiated, extracellular dopamine concentration appears to regulate
accumbens injections of the dopamine uptake inhibitor nomifensine the frequency of continued responding. Rats tend to respond to
(Carlezon and Wise, 1996). Thus PCP has dopamine-independent amphetamine until their nucleus accumbens dopamine levels are
rewarding actions at the NMDA receptor, where it binds at 10 times elevated three- to five-fold; they then wait until prior injections
lower concentration than is required for its binding at the dopamine are partially metabolized and their dopamine levels have fallen
transporter (Chaudieu et al., 1989; Ohmori et al., 1992). to approximately twice-normal levels before responding again
Before accepting the assumption that intracranial injections of (Ranaldi et al., 1999). Thus dopamine actions in the nucleus
a drug identify the systems where it acts, one should require accumbens are not only critical for whether or not amphetamine is
convincing evidence that the drug is not diffusing from the site rewarding; they also appear to determine how long a given injection
of injection to a distal site of action. A central drug injection will satiate an animal.
FUNCTIONAL ANATOMY 513

Cocaine aversive and act at the dopamine transporter to decrease dopamine


release (Thompson et al., 2000).
Cocaine, like amphetamine, is rewarding through its ability to ele-
vate nucleus accumbens dopamine levels (de Wit and Wise, 1977; Cannabinoids
Risner and Jones, 1980). Whereas amphetamine is a dopamine
releaser, cocaine is a dopamine uptake inhibitor (Heikkila et al., While there are no behavioural studies in which local injections
1975a, 1975b). While cocaine appears to be more effective as of cannabinoids were used to localize a rewarding site of action,
a reinforcer when injected into the frontal cortex (Goeders and there is evidence that cannabinoids can influence the mesolimbic
Smith, 1983), if animals are given sufficient exposure they will self- dopamine system. The major psychoactive constituent of marijuana
administer it directly into the nucleus accumbens (Carlezon et al., and hashish is 9 -tetrahydrocannabinol (THC). Partly for reasons of
1995), where dopamine-selective lesions attenuate the rewarding solubility, this substance appears to have its greatest abuse liability
effects of intravenous cocaine (Roberts et al., 1977, 1980). More- when smoked. Recently, however, intravenous self-administration
over, frontal cortex injections of cocaine appear to be reward- of THC has been demonstrated in squirrel monkeys (Tanda et al.,
ing because they trans-synaptically elevate nucleus accumbens 2000) and intravenous self-administration of a synthetic cannabi-
dopamine levels (Goeders and Smith, 1993). Nomifensine, a more noid has been shown in mice (Martellotta et al., 1998). Systemic
selective (Samanin et al., 1975) and efficacious (Nomikos et al., treatment with THC can also establish conditioned place prefer-
1990) dopamine uptake inhibitor, is also rewarding in the shell ences (Lepore et al., 1995; Valjent and Maldonado, 2000).
(but not the core) of the nucleus accumbens, and its effective- THC elevates nucleus accumbens dopamine levels when given
ness is blocked by co-administration of the D2 dopamine antagonist either systemically (Chen et al., 1990) or directly into the nucleus
sulpiride (Carlezon et al., 1995). This suggests that it is not the local accumbens (Chen et al., 1993), and it enhances potassium-induced
anaesthetic action of cocaine (Ritchie and Greene, 1985) in nucleus dopamine release in much the same way as do dopamine uptake
accumbens that accounts for its rewarding actions there. Self- inhibitors (Ng Cheong Ton et al., 1988). Such elevations appear to
administered doses of cocaine, like self-administered amphetamine, occur selectively in the shell of the nucleus accumbens (Tanda et al.,
elevate nucleus accumbens dopamine levels (Hurd et al., 1989; Pet- 1997a). When THC is given in combination with haloperidol, the
tit and Justice, 1989) and maintain them at levels two or more times dopamine release normally caused by haloperidol-induced blockade
above normal baseline levels (Wise et al., 1995b). of dopamine autoreceptors is enhanced, as it is by dopamine uptake
inhibitors (Gardner et al., 1990). Finally, THC’s effect on nucleus
accumbens levels of the dopamine metabolite 3-methoxytyramine
Opiates
resembles the effects of a dopamine uptake inhibitor (Heal et al.,
1990) like cocaine or nomifensine (Chen et al., 1994). It would
It is also thought that opiates trigger rewarding actions in the thus seem that at least some part of the habit-forming effects of
nucleus accumbens. Morphine (Olds, 1982) and the endogenous THC is triggered in the nucleus accumbens or somehow depends
opioid met-enkephalin (Goeders et al., 1984) are reported to be on nucleus accumbens dopaminergic function.
self-administered into this region, and morphine injections into
this region are reported to cause conditioned place preferences
(van der Kooy et al., 1982). Some concern must remain, however, Alcohol
as to whether opiate injections into this brain region have their
rewarding actions locally (Wise, 1989). In studies where nucleus Ethanol elevates nucleus accumbens dopamine levels (Imperato and
accumbens self-administration or conditioned place preference has Di Chiara, 1986; Gonzales and Weiss, 1998; Weiss et al., 1993),
been reported, anatomical controls that would rule out drug efflux and intra-accumbens microinjections of the dopamine D2 receptor
to the ventricular system or adjacent sites have not been reported. antagonist raclopride produce dose-orderly decreases in ethanol
This is troublesome because much lower doses of morphine are self- self-administration (Samson et al., 1993). Thus it seems likely that
administered or cause conditioned place preferences when injected alcohol has habit-forming actions mediated in part by the nucleus
into other brain sites (Bozarth and Wise, 1981, 1982; Bozarth, accumbens. In this case, however, there is no evidence to suggest
1987; Olmstead and Franklin, 1997). Moreover, some workers have that alcohol’s interface with drug reward circuitry is within the
reported nucleus accumbens opiates to be ineffective in each of nucleus accumbens.
these reward paradigms (Bozarth and Wise, 1982; Schildein et al.,
1998; Zangen et al., 2000). Caffeine
It is tempting, nonetheless, to suspect that opiates at high
enough concentrations can induce rewarding effects at nucleus The only links between caffeine and drug reward circuitry are
accumbens opiate receptors. There are several populations of opioid extremely speculative. There is no direct experimental evidence on
receptors in nucleus accumbens, and µ- and δ-opioids inhibit caffeine’s central sites of rewarding action. Indeed, although reports
nucleus accumbens medium spiny neurones (Hakan and Henriksen, do exist of successful caffeine self-administration in laboratory rats
1989; Jiang and North, 1992). There are µ-opioid receptors on (e.g., Collins et al., 1984), monkeys (e.g., Deneau et al., 1969)
the medium spiny output neurones of the nucleus accumbens and baboons (e.g., Griffiths et al., 1979), the bulk of the existing
(Wang et al., 1997). Activation of these receptors is thought to literature suggests that caffeine self-administration in laboratory
inhibit medium spiny neurone activity much as does dopamine and animals is inconsistent and sporadic at best (see reviews by Griffiths
systemic morphine (Hakan and Henriksen, 1989). In addition, there and Woodson, 1988; Griffiths and Mumford, 1995; Howell et al.,
are both µ- and δ-opioid receptors on the presynaptic terminals of 1997). More consistently, caffeine has been reported to augment
corticolimbic glutamate inputs to the nucleus accumbens. Actions the reinforcing properties (e.g., Schenk et al., 1994; Shoaib et al.,
at each of these receptor subtypes appear to inhibit release of the 1999), prolong extinction of self-administration (e.g., Kuzmin et al.,
excitatory amino acid glutamate onto medium spiny neurones (Jiang 1999) and trigger reinstatement of extinguished self-administration
and North, 1992), much as does blockade of NMDA receptors (Schenk and Partridge, 1999) of other addictive drugs.
by PCP. The two compounds that have been reported to be Caffeine is an antagonist at adenosine A1 and A2 receptors,
self-administered into the nucleus accumbens, morphine and met- and its behavioural effects appear to be mostly mediated by
enkephalin, each activate both µ- and δ-opioids. There are also such actions (Fredholm et al., 1999). As adenosine A1 (Goodman
κ-opioid receptors in the nucleus accumbens, but κ-opioids are and Snyder, 1982; Fastbom et al., 1987) and A2A (Dixon et al.,
514 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

1996; Rosin et al., 1998) receptors are densely distributed in the It is clear that self-administered intravenous doses of heroin
nucleus accumbens, as A2A receptors are extensively co-localized are sufficient to activate the mesolimbic dopamine system, as
with dopamine D2 receptors and preproenkephalin mRNA in self-administered intravenous heroin elevates nucleus accumbens
accumbens medium spiny neurones (Svenningsson et al., 1997, dopamine much as do self-administered amphetamine and cocaine
1998), and as A2A receptors are also co-localized (sparsely to be (Wise et al., 1995a). Thus — since it is unlikely that medium spiny
sure, but significantly above background levels) with dopamine neurones in the nucleus accumbens can discriminate the dopamine
D1 receptors in the accumbens (Svenningsson et al., 1997, 1998), overflow that results from heroin’s action at the dopamine cell
it may be surmised that adenosine receptor activation in the bodies from that resulting from amphetamine’s or cocaine’s actions
accumbens modulates meso-accumbens dopaminergic function. at the dopamine transporter — it seems clear that opiate actions
Indeed, considerable evidence exists that A1 or A2A receptor triggered in the ventral tegmental area must contribute significantly
activation inhibits dopaminergic function mediated through either to the rewarding effects of intravenous heroin. This suggestion
the D1 or D2 receptor, and that adenosine antagonism enhances is strengthened by the fact that the initiation of the next within-
meso-accumbens dopaminergic function (e.g., Turgeon et al., 1996; session lever-press for intravenous heroin, like those for intravenous
Le Moine et al., 1997; Moreau and Huber, 1999; Svenningsson amphetamine and cocaine, can be predicted by the fall of nucleus
et al., 1999; Poleszak and Malec, 2000). In a standard drug accumbens dopamine levels to a trigger point about twice the
discrimination assay in monkeys, adenosine antagonism generalizes normal dopamine concentration (Pocock et al., 1994).
dose-dependently and completely to eight different dopamine
receptor agonists that encompass a variety of mechanisms and sites Nicotine
of action, both pre- and postsynaptic (Holtzman, 1999). Further,
the discriminative stimulus properties of the non-selective A1 /A2 Nicotine also triggers its rewarding actions in the ventral tegmental
adenosine antagonist CGS-15943 are blocked dose-dependently and area. Here the critical receptors are on the dopaminergic neu-
completely by the dopamine D1 antagonist SCH-23390 and the rones themselves (Clarke and Pert, 1985). Intravenous nicotine
D2 antagonist eticlopride (Holtzman, 1999). Congruently, caffeine self-administration is blocked by dopamine antagonists and neu-
enhances dopamine turnover in the striatum (Waldeck, 1972, 1975; rotoxins as well as by chlorisondamine (Corrigall et al., 1992), a
Watanabe and Uramoto, 1986) and enhances striatal dopamine nicotinic channel blocker that is taken up and concentrated within
release in freely moving rats as measured by in vivo voltammetry the dopaminergic cell bodies of the ventral tegmental area and
(Morgan and Vestal, 1989). Recent evidence suggests that adenosine substantia nigra (El-Bizri et al., 1995). Nicotine increases dopamin-
and dopamine receptors form heterodimers in medium spiny ergic cell firing (Grenhoff et al., 1986; Mereu et al., 1987) and
neurones (Franco et al., 2000). All of these interesting findings elevates dopamine levels in the nucleus accumbens (Imperato et al.,
notwithstanding, it is only by tentative and arbitrary decision- 1986). Thus nicotine, like amphetamine, cocaine and morphine,
making that we include caffeine with habit-forming drugs having appears to be habit-forming because it elevates dopamine levels in
rewarding sites of action in the nucleus accumbens. There is no the nucleus accumbens.
evidence that caffeine has its rewarding action at trigger zones in Nicotine also elevates dopamine levels when infused directly into
the nucleus accumbens, and it is listed here simply because we the nucleus accumbens, where the cholinergic agonist carbachol is
could suggest no more relevant place for it. self-administered (Ikemoto et al., 1998a). However, as is the case
with ventral tegmental and nucleus accumbens administration of
Ventral Tegmental Area opioids, nucleus accumbens actions of nicotine appear to be weaker
than the ventral tegmental actions of nicotine (Benwell et al., 1993;
Opiates Nisell et al., 1994) and thus seem unlikely to account for the
rewarding effects of systemic nicotine (Corrigall et al., 1992).
A good deal of evidence suggests that the primary site of rewarding
action of opiates is in the ventral tegmental area, where µ-opioids
disinhibit the mesocorticolimbic dopamine system by inhibiting the Cannabinoids
GABAergic cells that normally hold their dopaminergic neighbours
under inhibitory control (Johnson and North, 1992). First, µ- and Like morphine, THC has actions in the ventral tegmental area
δ-opioids are self-administered into the ventral tegmental area (VTA) as well as in the nucleus accumbens. First, it stimulates
but not into an area just dorsal to it (Bozarth and Wise, 1981; dopaminergic cell firing and this effect is not blocked — while
Welzl et al., 1989; Devine and Wise, 1994; Zangen et al., 2000). at least part of the elevations seen in dialysis studies are (Chen
The µ-opioid DAMGO is 100 times more potent than the δ- et al., 1990; Tanda et al., 1997a) — by opiate antagonists (French,
opioid DPDPE. Ventral tegmental injections of these substances 1997; Melis et al., 2000). Second, local application of THC to
elevate nucleus accumbens dopamine levels (Leone et al., 1991; the ventral tegmental area elevates ventral tegmental but not
Devine et al., 1993), and DAMGO is here again 100 times more nucleus accumbens dopamine levels (Chen et al., 1993). This
potent than DPDPE. Opiate antagonists injected into the ventral effect is tentatively interpreted as a reflection of the inhibition
tegmental area decrease the rewarding effects of intravenous heroin, of dendritically released dopamine; it is not known if this local
although there is disagreement as to whether they do so more effect of THC in the VTA is naloxone-sensitive. Local injections
potently than antagonists injected into the nucleus accumbens (Britt of cannabinoids into the VTA have not been tested in behavioural
and Wise, 1983; Vaccarino et al., 1985). Opioid doses that are paradigms.
ineffective in the nucleus accumbens establish intracranial self-
administration and conditioned place preferences in the ventral Alcohol
tegmental area (Bozarth, 1987; Olmstead and Franklin, 1997;
Zangen et al., 2000). Indeed, in other behavioural tests opioids Ethanol increases the firing rate of dopamine neurones in the
have stronger behavioural effects when injected into the ventral rat VTA (Gessa et al., 1985; Brodie et al., 1990) and it is
tegmental area than when injected into the nucleus accumbens self-administered directly into the VTA (McBride et al., 1993;
(Broekkamp et al., 1976; Kalivas et al., 1983; Jenck et al., 1987; Gatto et al., 1994; Rodd-Henricks et al., 2000). This intracra-
West and Wise, 1988; Zangen et al., 2000) unless the nucleus nial self-administration of ethanol was seen in selectively bred
accumbens is pharmacologically or neurotoxically deprived of alcohol-preferring and ordinary Wistar rats, but not in selec-
dopamine (Stinus et al., 1985, 1986). tively bred alcohol-non-preferring rats. The animals performing
FUNCTIONAL ANATOMY 515

such intracranial ethanol self-administration show lever discrimina- Hauser, 1987; Beckstead, 1988). The evidence for this localization,
tion, extinction of self-administration when vehicle is substituted however, is based on the adjacent substantia nigra, where D1
for ethanol, and reinstatement of self-administration responding receptor-expressing GABAergic terminals synapse primarily on
on the active lever when ethanol is once again made available GABAergic output neurones of the zona reticulata. There is dense
(Rodd-Henricks et al., 2000). Sites dorsal to the VTA do not expression of D1 receptors in the zona reticulata of substantia
support ethanol self-administration (Gatto et al., 1994). In a map- nigra, and sparse expression in the zona compacta and VTA. This
ping study, posterior ventral tegmental sites were found to sup- density parallels the density of GABAergic cell bodies and of
port ethanol self-administration; anterior ventral tegmental sites GABAergic input in terminals that co-localize dynorphin, which
did not (Rodd-Henricks et al., 2000). As noted by McBride et al. identifies the D1 -expressing GABAergic medium spiny neurones
(1999), intracranial ethanol self-administration may involve brain of the striatum. This evidence suggests, very indirectly, that the D1
serotonin (5HT) mechanisms, especially 5HT3 receptors. Ethanol receptors in the VTA and zona compacta are likely to be localized
potentiates the depolarizing effects of 5HT at the 5HT3 receptor to GABAergic inputs to GABAergic rather than to dopaminergic
(Lovinger and White, 1991), and local 5HT3 agonist microinfusion targets. Ventral tegmental administration of D1 -type agonists causes
increases somatodendritic dopamine release in the VTA (Camp- increased glutamate as well as increased GABA efflux, however,
bell et al., 1996), suggesting that activation of 5HT3 receptors raising the possibility that D1 receptors may also be expressed on
in the VTA may activate meso-accumbens dopamine neurones. glutamatergic input to the VTA (Kalivas and Duffy, 1995).
Most provocatively, local 5HT3 antagonist microinfusion prevents The GABAergic anatomy of the VTA and substantia nigra
ethanol-induced somatodendritic dopamine release in the ventral is complex. The substantia nigra, and to a lesser extent the
tegmental area (Campbell et al., 1996), suggesting that ethanol’s VTA, contains GABAergic neurones with dominant projections
action on meso-accumbens dopamine neurones is mediated via VTA to the pedunculopontine tegmental nuclei (Gerfen et al., 1982),
5HT3 receptors. Ethanol’s actions on meso-accumbens dopamine and to the superior colliculus and medial thalamus (Williams and
neurones may also involve GABAA -(Harris et al., 1995) or NMDA- Faull, 1988). GABAergic cells of the VTA also project to the
mediated (Weight et al., 1991) mechanisms. nucleus accumbens (Van Bockstaele and Pickel, 1995). GABAergic
neurones also project to and inhibit (Johnson and North, 1992)
Barbiturates and Benzodiazepines their dopaminergic neighbours; it was originally suspected that this
inhibition was accomplished by GABAergic local circuit neurones
Barbiturates and benzodiazepines are self-administered and can be (Grace and Bunney, 1985), but recent evidence suggests that it
abused by humans. These drugs are also self-administered in ani- is collaterals from GABAergic projection neurones that account
mal models, more readily in primates than in rodents (Ator and for the tonic inhibition of nearby dopaminergic neurones (Tepper
Griffiths, 1987). Neither self-administration nor conditioned place et al., 1995).
preferences have been established with intracranial injections; thus Three lines of recent investigation suggest heterogeneity of
notions of the sites of rewarding action for these drugs are largely GABAergic participation in reward function. First, Ikemoto et al.
based on evidence that barbiturates and benzodiazepines act via an (1997b) have found that rats will self-inject the GABAA antagonist
interaction with GABAA receptors (e.g., Korpi et al., 1997; Chebib picrotoxin into the anterior but not the posterior VTA, and
and Johnston, 1999). Inasmuch as opiates are thought to have that this behaviour is disrupted by co-infusion of the GABAA
rewarding actions by decreasing GABAA inhibition of dopamin- agonist muscimol. However, when injected into the posterior
ergic neurones in the VTA, one possibility is that barbiturates and VTA, muscimol is rewarding (Ikemoto et al., 1998b). Zangen et al.
benzodiazepines have dopamine-dependent rewarding actions in the (2000) also found evidence of rostral–caudal differences in ventral
VTA. This fits with the finding that dopaminergic lesions of the tegmental reward; they report that endomorphin-1, the endogenous
nucleus accumbens produced by microinjections of the dopamin- ligand for the µ-opioid receptor (the rewarding effects of which
ergic cytotoxin 6-hydroxydopamine block diazepam-induced con- are thought to be mediated by µ-opioid receptors on GABAergic
ditioned place preference (Spyraki and Fibiger, 1988). However, neurones: Johnson and North, 1992), is vigorously self-administered
the GABAergic synaptology of the ventral region area is com- into the posterior VTA but much less vigorously self-administered
plex, as GABAergic neurones comprise not only afferents to but into the anterior VTA. Laviolette and van der Kooy (2001) have
also efferents from this region. Recent evidence suggests that some reported that the rewarding effects of ventral tegmental injections
GABAergic drug actions are reflected in dopaminergic output and of the GABAA agonist muscimol are dopamine-dependent whereas
others are reflected in non-dopaminergic output (Laviolette and van the rewarding effects of ventral tegmental injections of the GABAA
der Kooy, 2001). antagonist bicuculline are dopamine-independent.

Non-Dopamine Output from the VTA Medial Prefrontal Cortex

The view that rewarding brain stimulation, natural rewards like Cocaine
those of food and sexual contact, and several drug rewards owe
their ability to control behaviour to the fact that they elevate Cocaine is self-administered directly into the medial prefrontal
mesolimbic dopamine levels is widely but not uniformly accepted cortex (Goeders and Smith, 1983, 1986). It is difficult to imagine
(Wise, 1996b; but see Koob and Goeders, 1989; Bechara et al., how injections into this area could diffuse to the nucleus accumbens,
1998). Several recent findings suggest that the dopaminergic output the other area implicated in cocaine reward. The self-administration
of the VTA is not the only contribution of this region to reward of cocaine in this region is dopamine-dependent, as co-infusion of
function. the D2 dopamine antagonist sulpiride blocks the effect (Goeders
First, ventral tegmental microinjections of a D1 -type dopamine et al., 1986). It is not at all clear, however, that cocaine is effective
antagonist reduce the rewarding effects of intravenous cocaine in the frontal cortex — as it is in the nucleus accumbens — because
(Ranaldi and Wise, 2001). D1 -type dopamine receptors are not of its ability to block the dopamine transporter. The dopamine
expressed by the dopaminergic neurones of the VTA or adjacent transporter is sparsely distributed in the frontal cortex (Garris and
substantia nigra themselves; rather, the D1 receptor populations Wightman, 1994; Sesack et al., 1998), where it is the noradrenaline
in this brain region are thought to be localized primarily to transporter that seems to be most strongly implicated in dopamine
the terminals of GABAergic inputs to this region (Altar and clearance (Tanda et al., 1997b; Yamamoto and Novotney, 1998;
516 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

Morón et al., 2001). The noradrenaline transporter has greater NMDA receptor. It is not clear what binding site mediates the
affinity for dopamine than does the dopamine transporter, and in the rewarding effects of cocaine in the medial prefrontal cortex.
frontal cortex, unlike nucleus accumbens, it is abundant. Thus it is While the rewarding effects of cocaine in this region appear
probably cocaine’s ability to block the noradrenaline transporter in to be dopamine-dependent, since they are blocked by the D2
the frontal cortex that accounts for its self-administration into this dopamine antagonist sulpiride and are eliminated by dopamine-
area. Whatever the local site of action, self-administered cocaine in specific neurotoxic lesions (Goeders et al., 1986; Goeders and
the frontal cortex causes elevation of dopamine levels in the nucleus Smith, 1986), cocaine does not appear to have its rewarding effects
accumbens, and this presumably trans-synaptic effect is thought to as a result of actions at the dopamine transporter because this
be necessary for cocaine’s rewarding effects in the frontal cortex transporter is sparsely distributed in this region. Rather, cocaine’s
(Goeders and Smith, 1993). ability to elevate frontal cortex dopamine concentration appears
to depend on its blockade of the noradrenaline transporter, which
Phencyclidine apparently takes up dopamine into noradrenergic nerve terminals.
The findings reviewed above suggest a common reward circuitry
Phencyclidine is also self-administered into the medial prefrontal associated with a broad range of addictive drug classes. At the
cortex (Carlezon and Wise, 1996), where direct electrical stimula- core of the circuitry are the mesolimbic dopamine system and
tion is also rewarding (Routtenberg and Sloan, 1972; Corbett et al., the descending anatomical cascade of GABAergic neurones that
1982). In this case the mechanism of action is not well under- dominate the efferent pathway from nucleus accumbens. While this
stood, but it should be noted that the prefrontal cortex is intimately unifying theme is attractive, the evidence for actions of different
linked to the mesolimbic dopamine system. The ventral tegmen- drugs in this system is consistent but minimal in several cases. A
tal dopaminergic neurones project to the medial prefrontal cortex, good deal of additional evidence will be needed, either to tie each of
where they synapse on GABAergic interneurones and on the pyra- these drugs unequivocally to the suggested circuitry or to establish
midal cells that are the cortical output neurones (Goldman-Rakic separate, parallel, reward circuitry for the drug classes for which
et al., 1989; Sesack et al., 1995). The prefrontal cortex neurones, in current evidence remains limited. Moreover, we are hardly closer
turn, project to both the nucleus accumbens and the VTA (Christie now than we were 20 years ago (Nauta et al., 1978; Mogenson
et al., 1985; Sesack et al., 1989; Sesack and Pickel, 1992). The pri- et al., 1980) to understanding how the activity of these segments of
mary neurotransmitter of these neurones is glutamate (Druce et al., motivational circuitry interfaces with and controls the mechanisms
1982; Sandberg et al., 1985), and they co-localize the neuropeptide of behavioural action.
cholecystokinin (CCK) (Meyer et al., 1982; Hökfelt et al., 1988).
These neurones are of interest because the release of CCK in both
VTA and nucleus accumbens is more proportional to the strength
of rewarding electrical stimulation of the prefrontal cortex than is REFERENCES
the release of either dopamine or glutamate in either structure (You
et al., 1998). Aghajanian, G.K., 1978. Tolerance of locus coeruleus neurones to morphine
and suppression of withdrawal response by clonidine. Nature, 276,
186–188.
Conceptual Nervous System of Addiction Akaoka, H. and Aston-Jones, G., 1991. Opiate withdrawal-induced hyperac-
tivity of locus coeruleus neurons is substantially mediated by augmented
While only a portion of the anatomy of substance abuse is known, excitatory amino acid input. Journal of Neuroscience, 11, 3830–3839.
a preliminary sketch of several important links in the rewarding Altar, C.A. and Hauser, K., 1987. Topography of substantia nigra inner-
effects of drugs can be suggested. At its core is the mesolimbic vation by D1 receptor-containing striatal neurons. Brain Research, 410,
dopamine system with its cells of origin in the VTA and its most 1–11.
important terminals in the nucleus accumbens. The trigger zones for Ator, N.A. and Griffiths, R.R., 1987. Self-administration of barbiturates and
benzodiazepines: a review. Pharmacology, Biochemistry and Behavior ,
the habit-forming actions of nicotine and opiates are in the VTA; 27, 391–398.
the critical receptors are expressed by dopaminergic neurones and Balster, R.L., Johanson, C.E., Harris, R.T. and Schuster, C.R., 1973.
GABAergic neurones, respectively. The endogenous ligand for the Phencyclidine self-administration in the rhesus monkey. Pharmacology,
µ-opioid receptor in this region is endomorphin-1, which projects to Biochemistry and Behavior, 1, 167–172.
the VTA from the cells in either the hypothalamus or the nucleus of Barr, G.A., Paredes, W. and Bridger, W.H., 1985. Place conditioning with
the solitary tract (Martin-Schild et al., 1999); the endogenous ligand morphine and phencyclidine: dose dependent effects. Life Sciences , 36,
for the nicotinic receptor in this region is acetylcholine, which 363–368.
is released in the VTA by projections from the pedunculopontine Bechara, A., Nader, K. and van der Kooy, D., 1998. A two-separate-
tegmental nucleus (Oakman et al., 1995). motivational-systems hypothesis of opioid addiction. Pharmacology,
Trigger zones for the habit-forming actions of amphetamine, Biochemistry and Behavior, 59, 1–17.
Beckmann, A.M., Matsumoto, I. and Wilce, P.A., 1995. Immediate early
cocaine and phencyclidine are in the nucleus accumbens. The gene expression during morphine withdrawal. Neuropharmacology, 34,
binding sites for amphetamine and cocaine are the dopamine 1183–1189.
transporters on dopaminergic nerve terminals in this region; the Beckstead, R.M., 1988. Association of dopamine D1 and D2 receptors with
trigger zone for phencyclidine seems to be the NMDA receptor specific cellular elements in the basal ganglia of the cat: the uneven
on medium spiny neurones. The endogenous transmitter mediating topography of dopamine receptors in the striatum is determined by
the rewarding effects of amphetamine and cocaine is dopamine; it intrinsic striatal cells, not nigrostriatal axons. Neuroscience, 27, 851–863.
appears to require synergistic effects (see, e.g., Clark and White, Beitner-Johnson, D. and Nestler, E.J., 1991. Morphine and cocaine exert
1987) of both D1 -type and D 2 -type receptors for dopamine to be common chronic actions on tyrosine hydroxylase in dopaminergic brain
rewarding (Ikemoto et al., 1997a). The endogenous transmitter that, reward regions. Journal of Neurochemistry, 57, 344–347.
Beitner-Johnson, D., Guitart, X. and Nestler, E.J., 1992. Neurofilament
when blocked, accounts for the rewarding effects of phencyclidine
proteins and the mesolimbic dopamine system: common regulation by
would appear to be glutamate; glutamatergic afferents to the nucleus chronic morphine and chronic cocaine in the rat ventral tegmental area.
accumbens arise from a variety of cortical structures. Journal of Neuroscience, 12, 2165–2176.
Additional trigger zones for cocaine and phencyclidine effects Bell, K., Duffy, P. and Kalivas, P.W., 2000. Context-specific enhancement
are found in the medial prefrontal cortex; the receptor mediating of glutamate transmission by cocaine. Neuropsychopharmacology, 23,
the rewarding effects of phencyclidine again appears to be the 335–344.
FUNCTIONAL ANATOMY 517

Benwell, M.E.M., Balfour, D.J.K. and Lucchi, H.M., 1993. Influence of Christie, M.J., James, L.B. and Beart, P.M., 1985. An excitant amino-acid
tetrodotoxin and calcium on changes in extracellular dopamine levels projection from the medial prefrontal cortex to the anterior part of nucleus
evoked by systemic nicotine. Psychopharmacology , 112, 467–474. accumbens in the rat. Journal of Neurochemistry, 45, 477–482.
Bijerot, N., 1980. Addiction to pleasure: a biological and social- Clark, D. and White, F.J., 1987. D1 dopamine receptor — the search
psychological theory of addiction. In: Lettieri, D.J., Sayers, M. and for a function: a critical evaluation of the D1/D2 dopamine receptor
Pearson, H.W. (eds), Theories on Drug Abuse: Selected Contemporary classification and its function implications. Synapse, 1, 347–388.
Perspectives, pp. 246–255. National Institute on Drug Abuse, Clarke, P.B.S. and Pert, A., 1985. Autoradiographic evidence for nicotine
Rockville, MD. receptors on nigrostriatal and mesolimbic dopaminergic neurons. Brain
Bozarth, M.A., 1987. Neuroanatomical boundaries of the reward-relevant Research, 348, 355–358.
opiate-receptor field in the ventral tegmental area as mapped by the Collier, H.O.J., 1966. Tolerance, physical dependence and receptors: a
conditioned place preference method in rats. Brain Research, 414, 77–84. theory. Advances in Drug Research, 3, 171–188.
Bozarth, M.A. and Wise, R.A., 1981. Intracranial self-administration of Collins, R.J., Weeks, J.R., Cooper, M.M., Good, P.I. and Russell, R.R.,
morphine into the ventral tegmental area in rats. Life Sciences 28, 1984. Prediction of abuse liability of drugs using IV self-administration
551–555. by rats. Psychopharmacology , 82, 6–13.
Bozarth, M.A. and Wise, R.A., 1982. Localization of the reward-relevant Corbett, D., Laferriere, A. and Milner, P.M., 1982. Plasticity of the medial
opiate receptors. In: Harris, L.S. (ed.), Problems of Drug Dependence prefrontal cortex: facilitated acquisition of intracranial self-stimulation by
1981 , pp. 158–164. US Government Printing Office, Washington, DC. pretraining stimulation. Physiology and Behavior , 28, 532–534.
Corrigall, W.A., Franklin, K.B.J., Coen, K.M. and Clarke, P., 1992. The
Bozarth, M.A. and Wise, R.A., 1984. Anatomically distinct opiate receptor
mesolimbic dopaminergic system is implicated in the reinforcing effects
fields mediate reward and physical dependence. Science, 224, 516–517.
of nicotine. Psychopharmacology , 107, 285–289.
Britt, M.D. and Wise, R.A., 1983. Ventral tegmental site of opiate reward:
Crider, R., 1986. Phencyclidine: changing abuse patterns. NIDA Research
antagonism by a hydrophilic opiate receptor blocker. Brain Research, Monograph, 64, 163–173.
258, 105–108. Dackis, C.A. and Gold, M.S., 1985. New concepts in cocaine addiction:
Brodie, M.S., Shefner, S.A. and Dunwiddie, T.V., 1990. Ethanol increases the dopamine depletion hypothesis. Neuroscience and Biobehavioral
the firing rate of dopamine neurons of the rat ventral tegmental area Reviews, 9, 469–477.
in vitro. Brain Research, 508, 65–69. Deneau, G., Yanagita, T. and Seevers, M.H., 1969. Self-administration of
Broekkamp, C.L.E., Van den Bogaard, J.H., Heijnen, H.J., Rops, R.H., psychoactive substances by the monkey: a measure of psychological
Cools, A.R. and Van Rossum, J.M., 1976. Separation of inhibiting dependence. Psychopharmacologia , 16, 30–48.
and stimulating effects of morphine on self-stimulation behavior by Devine, D.P. and Wise, R.A., 1994. Self-administration of morphine,
intracerebral microinjections. European Journal of Pharmacology , 36, DAMGO, and DPDPE into the ventral tegmental area of rats. Journal of
443–446. Neuroscience, 14, 1978–1984.
Campbell, A.D., Kohl, R.R. and McBride, W.J., 1996. Serotonin-3 receptor Devine, D.P., Leone, P., Pocock, D. and Wise, R.A., 1993. Differential
and ethanol-stimulated somatodendritic dopamine release. Alcohol , 13, involvement of ventral tegmental mu, delta, and kappa opioid receptors in
569–574. modulation of basal mesolimbic dopamine release: in vivo microdialysis
Carlezon, W.A. Jr and Wise, R.A., 1996. Rewarding actions of phencycli- studies. Journal of Pharmacology and Experimental Therapeutics, 266,
dine and related drugs in nucleus accumbens shell and frontal cortex. 1236–1246.
Journal of Neuroscience, 16, 3112–3122. de Wit, H. and Wise, R.A., 1977. Blockade of cocaine reinforcement in
Carlezon, W.A. Jr, Devine, D.P. and Wise, R.A., 1995. Habit-forming rats with the dopamine receptor blocker pimozide but not with the
actions of nomifensine in nucleus accumbens. Psychopharmacology , 122, noradrenergic blockers phentolamine or phenoxybenzamine. Canadian
194–197. Journal of Psychology, 31, 195–203.
Carlezon, W.A., Thome, J., Olson, V.G. et al., 1998. Regulation of cocaine Di Chiara, G., 1998. A motivational learning hypothesis of the role of
reward by CREB. Science, 282, 2272–2275. mesolimbic dopamine in compulsive drug use. Journal of Psychophar-
Carr, G.D. and White, N.M., 1983. Conditioned place preference from intra- macology, 12, 54–67.
accumbens but not intra-caudate amphetamine injections. Life Science, Di Chiara, G. and Imperato, A., 1988. Drugs abused by humans prefer-
33, 2551–2557. entially increase synaptic dopamine concentrations in the mesolimbic
Cha, X.Y., Pierce, R.C., Kalivas, P.W. and Mackler, S.A., 1997. NAC-1, system of freely moving rats. Proceedings of the National Academy of
a rat brain mRNA, is increased in the nucleus accumbens three weeks Sciences USA, 85, 5274–5278.
after chronic cocaine self-administration. Journal of Neuroscience, 17, Dixon, A.K., Gubitz, A.K., Sirinathsinghji, D.J., Richardson, P.J. and
6864–6871. Freeman, T.C., 1996. Tissue distribution of adenosine receptor mRNAs
Chaudieu, I., Vignon, J., Chicheportiche, M., Kamenka, J.-M., Trouiller, in the rat. British Journal of Pharmacology, 118, 1461–1468.
G. and Chicheportiche, R., 1989. Role of the aromatic group in the Druce, D., Peterson, D., De Belleroche, J. and Bradford, H.F., 1982.
inhibition of phencyclidine binding and dopamine uptake by PCP analogs. Differential amino acid neurotransmitter release in rat neostriatum
Pharmacology, Biochemistry and Behavior , 32, 699–705. following lesioning of the cortico-striatal pathway. Brain Research, 247,
303–307.
Chebib, M. and Johnston, G.A., 1999. The ‘ABC’ of GABA receptors: a
El-Bizri, H., Rigdon, M.G. and Clarke, P.B.S., 1995. Intraneuronal accu-
brief review. Clinical and Experimental Pharmacology and Physiology,
mulation and persistence of radiolabel in rat brain following in vivo
26, 937–940.
administration of [3 H]-chlorisondamine. British Journal of Pharmacol-
Chen, J., Paredes, W., Li, J., Smith, D., Lowinson, J. and Gardner, E.L.,
ogy, 116, 2503–2509.
1990. 9 -Tetrahydrocannabinol produces naloxone-blockable enhance- Emmett-Oglesby, M.W. and Lane, J.D., 1992. Tolerance to the reinforcing
ment of presynaptic basal dopamine efflux in nucleus accumbens of effects of cocaine. Behavioural Pharmacology , 3, 193–200.
conscious, freely-moving rats as measured by intracerebral microdialysis. Ettenberg, A. and Camp, C.H., 1986. Haloperidol induces a partial
Psychopharmacology , 102, 156–162. reinforcement extinction effect in rats: implications for a dopamine
Chen, J., Marmur, R., Pulles, A., Paredes, W. and Gardner, E.L., 1993. involvement in food reward. Pharmacology, Biochemistry and Behavior ,
Ventral tegmental microinjection of 9 -tetrahydrocannabinol enhances 25, 813–821.
ventral tegmental somatodendritic dopamine levels but not forebrain Fastbom, J., Pazos, A., Probst, A. and Palacios, J.M., 1987. Adenosine
dopamine levels: evidence for local neural action by marijuana’s psy- A1 receptors in the human brain: a quantitative autoradiographic study.
choactive ingredient. Brain Research, 621, 65–70. Neuroscience, 22, 827–839.
Chen, J., Paredes, W. and Gardner, E.L., 1994. 9 -Tetrahydrocannabinol’s Ferguson, S.S., Zhang, J., Barak, L.S. and Caron, M.G., 1998. Molecular
enhancement of nucleus accumbens dopamine resembles that of reuptake mechanisms of G protein-coupled receptor desensitization and resensiti-
blockers rather than releasers: evidence from in vivo microdialysis zation. Life Sciences, 62, 1561–1565.
experiments with 3-methoxytyramine. NIDA Research Monograph, 141, Fibiger, H.C., 1978. Drugs and reinforcement mechanisms: a critical
312. review of the catecholamine theory. Annual Review of Pharmacology
Chen, J., Kelz, M.B., Hope, B.T., Nakabeppu, Y. and Nestler, E.J., 1997. and Toxicology, 18, 37–56.
Chronic Fos-related antigens: stable variants of deltaFosB induced in Fitzgerald, L.W., Ortiz, J., Hamedani, A.G. and Nestler, E.J., 1996. Drugs
brain by chronic treatments. Journal of Neuroscience, 17, 4933–4941. of abuse and stress increase the expression of GluR1 and NMDAR1
518 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

glutamate receptor subunits in the rat ventral tegmental area: common Grenhoff, J., Aston-Jones, G. and Svensson, T.H., 1986. Nicotinic effects
adaptations among cross-sensitizing agents. Journal of Neuroscience, 16, on the firing pattern of midbrain dopamine neurons. Acta Physiologica
274–282. Scandanavica, 128, 351–358.
Franco, R., Ferré, S., Agnati, L. et al., 2000. Evidence for adeno- Griffiths, R.R. and Mumford, G.K., 1995. Caffeine: a drug of abuse? In:
sine/dopamine receptor interactions: indications for heterodimerization. Bloom, F.E. and Kupfer, D.J. (eds), Psychopharmacology: The Fourth
Neuropsychopharmacology, 23 (Suppl. 4), S50–59. Generation of Progress, pp. 1699–1713. Raven Press, New York.
Frank, R.A., Martz, S. and Pommering, T., 1988. The effect of chronic Griffiths, R.R. and Woodson, P.P., 1988. Reinforcing properties of caffeine:
cocaine on self-stimulation train-duration thresholds. Pharmacology, studies in humans and laboratory animals. Pharmacology, Biochemistry
Biochemistry and Behavior , 29, 755–758. and Behavior, 29, 419–427.
Frank, R.A., Manderscheid, P.Z., Panicker, S., Williams, H.P. and Koko- Griffiths, R.R., Brady, J.V. and Bradford, L.D., 1979. Predicting the
ris, D., 1992. Cocaine euphoria, dysphoria, and tolerance assessed using abuse liability of drugs with animal self-administration procedures:
drug-induced changes in brain-stimulation reward. Pharmacology Bio- psychomotor stimulants and hallucinogens. In: Thompson, T. and Dews,
chemistry and Behavior, 42, 771–779. P.B. (eds), Advances in Behavioral Pharmacology, Vol. 2, pp. 163–208.
Fredholm, B.B., Battig, K., Holmen, J., Nehlig, A. and Zvartau, E.E., 1999. Academic Press, New York.
Actions of caffeine in the brain with special reference to factors that Grimm, J.W., Hope, B.T., Wise, R.A. and Shaham, Y., 2001. Incubation of
contribute to its widespread use. Pharmacological Reviews, 51, 83–133. cocaine craving after withdrawal. Nature, 412, 141–142.
French, E.D., 1997. 9 -Tetrahydrocannabinol excites rat VTA dopamine Hakan, R.L. and Henriksen, S.J., 1989. Opiate influences on nucleus
neurons through activation of cannabinoid CB1 but not opioid receptors. accumbens neuronal electrophysiology: dopamine and non-dopamine
Neuroscience Letters, 226, 159–162. mechanisms. Journal of Neuroscience, 9, 3538–3546.
Gardner, E.L., 1992. Brain reward mechanisms. In: Lowinson, J.H., Ruiz, P. Harris, R.A., Proctor, W.R., McQuilkin, S.J. et al., 1995. Ethanol increases
and Millman, R.B. (eds), Substance Abuse: a Comprehensive Textbook, GABAA responses in cells stably transfected with receptor subunits.
pp. 70–99. Williams & Wilkins, Baltimore MD. Alcoholism: Clinical and Experimental Research, 19, 226–232.
Gardner, E.L., Paredes, W. and Chen, J., 1990. Further evidence for 9 - Hayward, M.D., Duman, R.S. and Nestler, E.J., 1990. Induction of the c-
tetrahydrocannabinol as a dopamine reuptake blocker: brain microdialysis fos proto-oncogene during opiate withdrawal in the locus coeruleus and
studies. Society for Neuroscience Abstracts, 16, 1100. other regions of rat brain. Brain Research, 525, 256–266.
Garris, P.A. and Wightman, R.M., 1994. Different kinetics govern dopamin- Heal, D.J., Frankland, A.T.J. and Buckett, W.R., 1990. A new and highly
ergic transmission in the amygdala, prefrontal cortex, and striatum: an sensitive method for measuring 3-methoxytyramine using HPLC with
in vivo voltammetric study. Journal of Neuroscience, 14, 442–450. electrochemical detection: studies with drugs which alter dopamine
Gatto, G.J., McBride, W.J., Murphy, J.M., Lumeng, L. and Li, T.K., 1994. metabolism in the brain. Neuropharmacology, 29, 1141–1150.
Ethanol self-infusion into the ventral tegmental area by alcohol-preferring Heikkila, R.E., Orlansky, H., Mytilineou, C. and Cohen, G., 1975a.
rats. Alcohol , 11, 557–564. Amphetamine: evaluation of d- and l-isomers as releasing agents and
Gerfen, C.R., Staines, W.A., Arbuthnott, G.W. and Fibiger, H.C., 1982. uptake inhibitors for 3 H-dopamine and 3 H-norepinephrine in slices
Crossed connections of the substantia nigra in the rat. Journal of of rat neostriatum and cerebral cortex. Journal of Pharmacology and
Comparative Neurology, 20, 283–303. Experimental Therapeutics, 194, 47–56.
Heikkila, R.E., Orlansky, H. and Cohen, G., 1975b. Studies on the
Gerhardt, G.A., Pang, K. and Rose, G.M., 1987. In vivo electrochemical
distinction between uptake inhibition and release of (3 H)dopamine in
demonstration of the presynaptic actions of phencyclidine in rat caudate
rat brain tissue slices. Biochemical Pharmacology , 24, 847–852.
nucleus. Journal of Pharmacology and Experimental Therapeutics, 241,
Henry, D.J. and White, F.J., 1991. Repeated cocaine administration causes
714–721.
persistent enhancement of D1 dopamine receptor sensitivity within the
Gessa, G.L., Muntoni, F., Collu, M., Vargiu, L. and Mereu, G., 1985. Low
rat nucleus accumbens. Journal of Pharmacology and Experimental
doses of ethanol activate dopaminergic neurons in the ventral tegmental Therapeutics, 258, 882–890.
area. Brain Research, 348, 201–203. Himmelsbach, C.K., 1943. Morphine, with reference to physical depen-
Goeders, N.E. and Smith, J.E., 1983. Cortical dopaminergic involvement in dence. Federation Proceedings, 2, 201–203.
cocaine reinforcement. Science, 221, 773–775. Hoebel, B.G., Monaco, A.P., Hernandez, L., Aulisi, E.F., Stanley, B.G. and
Goeders, N.E. and Smith, J.E., 1986. Reinforcing properties of cocaine in Lenard, L., 1983. Self-injection of amphetamine directly into the brain.
the medial prefrontal cortex: primary action on presynaptic dopaminergic Psychopharmacology , 81, 158–163.
terminals. Pharmacology, Biochemistry and Behavior , 25, 191–199. Hökfelt, T., Herrera-Marschitz, M., Seroogy, K. et al., 1988. Immunohis-
Goeders, N.E. and Smith, J.E., 1993. Intracranial cocaine self-administration tochemical studies on cholecystokinin (CCK)-immunoreactive neurons
into the medial prefrontal cortex increases dopamine turnover in the in the rat using sequence specific antisera and with special reference to
nucleus accumbens. Journal of Pharmacology and Experimental Ther- the caudate nucleus and primary sensory neurons. Journal of Chemical
apeutics, 265, 592–600. Neuroanatomy, 1, 11–52.
Goeders, N.E., Lane, J.D. and Smith, J.E., 1984. Self-administration Holtzman, S.G., 1999. Discriminative effects of CGS 15943, a competitive
of methionine enkephalin into the nucleus accumbens. Pharmacology, adenosine receptor antagonist, have a dopamine component in moneys.
Biochemistry and Behavior , 20, 451–455. European Journal of Pharmacology , 376, 7–15.
Goeders, N.E., Dworkin, S.I. and Smith, J.E., 1986. Neuropharmacological Hope, B., Kosofsky, B., Hyman, S.E. and Nestler, E.J., 1992. Regulation
assessment of cocaine self-administration into the medial prefrontal of immediate early gene expression and AP-1 binding in the rat nucleus
cortex. Pharmacology Biochemistry and Behavior , 24, 1429–1440. accumbens by chronic cocaine. Proceedings of the National Academy of
Goldman-Rakic, P.S., Leranth, S.M., Williams, N., Mons, N. and Geffard, Sciences USA, 89, 5764–5768.
M., 1989. Dopamine synaptic complex with pyramidal neurons in primate Horger, B.A., Shelton, K. and Schenk, S., 1990. Preexposure sensitizes rats
cerebral cortex. Proceedings of the National Academy of Sciences USA, to the rewarding effects of cocaine. Pharmacology, Biochemistry and
86, 9015–9019. Behavior , 37, 707–711.
Goldstein, D.B. and Goldstein, A., 1961. Possible role of enzyme inhibition Howell, L.L., Coffin, V.L. and Spealman, R.D., 1997. Behavioral and
and repression in drug tolerance and addiction. Biochemical Pharmacol- physiological effects of xanthines in nonhuman primates. Psychophar-
ogy, 8, 48. macology, 129, 1–14.
Gonzales, R.A. and Weiss, F., 1998. Suppression of ethanol-reinforced Hurd, J.L., Weiss, F., Koob, G.F., Anden, N.-E. and Ungerstedt, U.,
behavior by naltrexone is associated with attenuation of the ethanol- 1989. Cocaine reinforcement and extracellular dopamine overflow in rat
induced increase in dialysate dopamine levels in the nucleus accumbens. nucleus accumbens: an in vivo microdialysis study. Brain Research, 498,
Journal of Neuroscience, 18, 10663–10671. 199–203.
Goodman, R.R. and Snyder, S.H., 1982. Autoradiographic localization of Ikemoto, S., Glazier, B.S., Murphy, J.M. and McBride, W.J., 1997a. Role
adenosine receptors in rat brain using [3 H]cyclohexyladenosine. Journal of dopamine D1 and D2 receptors in the nucleus accumbens in mediating
of Neuroscience, 2, 1230–1241. reward. Journal of Neuroscience, 17, 8580–8587.
Grace, A.A. and Bunney, B.S., 1985. Opposing effects of striatonigral Ikemoto, S., Murphy, J.M. and McBride, W.J., 1997b. Self-infusion of
feedback pathways on midbrain dopamine cell activity. Brain Research, GABAA antagonists directly into the ventral tegmental area and adjacent
333, 271–284. regions. Behavioral Neuroscience, 111, 369–380.
FUNCTIONAL ANATOMY 519

Ikemoto, S., Glazier, B.S., Murphy, J.M. and McBride, W.J., 1998a. Rats of cocaine-seeking behavior in mice. Journal of Pharmacology and
self-administer carbachol directly into the nucleus accumbens. Physiology Experimental Therapeutics , 290, 535–542.
and Behavior, 63, 811–814. Laschka, E., Herz, A. and Bläsig, J., 1976a. Sites of action of morphine
Ikemoto, S., Murphy, J.M. and McBride, W.J., 1998b. Regional differences involved in the development of physical dependence in rats I. Com-
within the rat ventral tegmental area for muscimol self-infusions. parison of precipitated morphine withdrawal after intraperitoneal and
Pharmacology, Biochemistry and Behavior , 61, 87–92. intraventricular injection of morphine antagonists. Psychopharmacologia,
Imperato, A. and Di Chiara, G., 1986. Preferential stimulation of dopamine 46, 133–139.
release in the nucleus accumbens of freely moving rats by ethanol. Laschka, E., Teschemacher, H., Mahraein, P. and Herz, A., 1976b.
Journal of Pharmacology and Experimental Therapeutics, 239, 219–228. Sites of action of morphine involved in the development of physical
Imperato, A., Mulas, A. and Di Chiara, G., 1986. Nicotine preferentially dependence of rats. II. Morphine withdrawal precipitated by application
stimulates dopamine release in the limbic system of freely moving rats. of morphine antagonists into restricted parts of the ventricular system
European Journal of Pharmacology , 132, 337–338. and by microinfusion into various brain areas. Psychopharmacologia,
Ivanov, A. and Aston-Jones, G., 2001. Local opiate withdrawal in locus 46, 141–147.
coeruleus neurons in vitro. Journal of Neurophysiology, 85, 2388–2397. Laviolette, S.R. and van der Kooy, D., 2001. GABAA receptors in the
Iwamoto, E.T., 1986. Place-aversion conditioned by phencyclidine in rats: ventral tegmental area control bidirectional reward signalling between
development of tolerance and pharmacological antagonism. Alcohol and dopaminergic and non-dopaminergic neural motivational systems. Euro-
Drug Research, 187, 555–556. pean Journal of Neuroscience, 13, 1009–1015.
Jaffe, J.H. and Sharpless, S.K., 1968. Pharmacological denervation super- Leith, N.J. and Barrett, R.J., 1976. Amphetamine and the reward system:
sensitivity in the central nervous system: a theory of physical dependence. evidence for tolerance and post-drug depression. Psychopharmacologia,
In: Wikler, A.H. (ed.), The Addictive States, pp. 226–246. Williams & 46, 19–25.
Wilkins, Baltimore. Le Moine, C., Svenningsson, P., Fredholm, B.B. and Bloch, B., 1997.
Jenck, F., Gratton, A. and Wise, R.A., 1987. Opioid receptor subtypes Dopamine-adenosine interactions in the striatum and the globus pallidus:
associated with ventral tegmental facilitation of lateral hypothalamic inhibition of striatopallidal neurons through either D2 or A2A receptors
brain stimulation reward. Brain Research, 423, 34–38. enhances D1 receptor-mediated effects on c-fos expression. Journal of
Jiang, Z.G. and North, R.A., 1992. Pre- and postsynaptic inhibition by Neuroscience, 17, 8038–8048.
opioids in rat striatum. Journal of Neuroscience, 12, 356–361. Leone, P., Pocock, D. and Wise, R.A., 1991. Morphine-dopamine interac-
Johnson, A.K. and Epstein, A.N., 1975. The cerebral ventricles as the tion: ventral tegmental morphine increases nucleus accumbens dopamine
avenue for the dipsogenic action of intracranial angiotensin. Brain release. Pharmacology Biochemistry and Behavior , 39, 469–472.
Research, 86, 399–418. Lepore, M., Vorel, S.R., Lowinson, J. and Gardner, E.L., 1995. Conditioned
Johnson, S.W. and North, R.A., 1992. Opioids excite dopamine neurons place preference induced by 9 -tetrahydrocannabinol: comparison with
by hyperpolarization of local interneurons. Journal of Neuroscience, 12, cocaine, morphine, and food reward. Life Sciences, 56, 2073–2080.
483–488. Lett, B.T., 1989. Repeated exposures intensify rather than diminish the
Jones, R.T., 1980. Human effects: an overview. In: Petersen, R.C. (ed.), rewarding effects of amphetamine, morphine, and cocaine. Psychophar-
Marijuana Research Findings: 1980 , pp. 54–80. US Government Print- macology, 98, 357–362.
ing Office, Washington, DC. Li, D.-H., Depoortere, R.Y. and Emmett-Oglesby, M.W., 1994. Tolerance
to the reinforcing effects of cocaine in a progressive ratio paradigm.
Jones, R.T., 1984. The pharmacology of cocaine. In: Grabowski, J. (ed.),
Psychopharmacology , 116, 326–332.
Cocaine: Pharmacology, Effects, and Treatment of Abuse, pp. 34–53. US
Lindesmith, A.R., 1947. Opiate Addiction. Principia Press, Bloomington,
Government Printing Office, Washington, DC.
IN.
Kalant, H., 1977. Comparative aspects of tolerance to, and dependence
Lorrain, D.S., Arnold, G.M. and Vezina, P., 2000. Previous exposure to
on, alcohol, barbiturates, and opiates. In: Gross, M.M. (ed.), Alcohol
amphetamine increases incentive to obtain the drug: long-lasting effects
Intoxication and Withdrawal , pp. 169–186. Plenum, New York.
revealed by the progressive ratio schedule. Behavioural Brain Research,
Kalivas, P.W. and Duffy, P., 1995. D1 receptors modulate glutamate 107, 9–19.
transmission in the ventral tegmental area. Journal of Neuroscience, 15, Lovinger, D.M. and White, G., 1991. Ethanol potentiation of 5-
5379–5388. hydroxytryptamine3 receptor-mediated ion current in neuroblastoma cells
Kalivas, P.W., Widerlov, E., Stanley, D., Breese, G. and Prange, A.J., 1983. and isolated adult mammalian neurons. Molecular Pharmacology, 40,
Enkephalin action on the mesolimbic system: a dopamine-dependent 263–270.
and a dopamine-independent increase in locomotor activity. Journal of Lyness, W.H., Friedle, N.M. and Moore, K.E., 1979. Destruction
Pharmacology and Experimental Therapeutics, 227, 229–237. of dopaminergic nerve terminals in nucleus accumbens: effect on
Kelz, M.B., Chen, J., Carlezon, W.A. et al., 1999. Expression of the d-amphetamine self-administration. Pharmacology, Biochemistry and
transcription factor DFosB in the brain controls sensitivity to cocaine. Behavior , 11, 553–556.
Nature, 401, 272–276. Maldonado, R., Stinus, L., Gold, L.H. and Koob, G.F., 1992. Role of
Kokkinidis, L. and McCarter, B.D., 1990. Postcocaine depression and different brain structures in the expression of the physical morphine
sensitization of brain-stimulation reward: analysis of reinforcement and withdrawal syndrome. Journal of Pharmacology and Experimental Ther-
performance effects. Pharmacology, Biochemistry and Behavior , 36, apeutics, 261, 669–677.
463–471. Markou, A. and Koob, G.F., 1991. Postcocaine anhedonia: an animal model
Kokkinidis, L., Zacharko, R.M. and Predy, P.A., 1980. Post-amphetamine of cocaine withdrawal. Neuropsychopharmacology, 4, 17–26.
depression of self-stimulation responding from the substantia nigra: Martellotta, M.C., Cossu, G., Frattore, L., Gessa, G.L. and Fratta, W., 1998.
reversal by tricyclic antidepressants. Pharmacology Biochemistry and Self-administration of the cannabinoid receptor agonist WIN 55, 212-2
Behavior , 13, 379–383. in drug-naive mice. Neuroscience, 85, 327–330.
Koob, G.F. and Bloom, F.E., 1988. Cellular and molecular mechanisms of Martin-Schild, S., Gerall, A.A., Kastin, A.J. and Zadina, J.E., 1999.
drug dependence. Science, 242, 715–723. Differential distribution of endomorphin 1- and endomorphin 2-like
Koob, G.F. and Goeders, N., 1989. Neuroanatomical substrates of drug immunoreactivities in the CNS of the rodent. Journal of Comparative
self-administration. In: Liebman, J.M. and Cooper, S.J. (eds), Neurophar- Neurology , 22, 450–471.
macological Basis of Reward, pp. 214–263. Oxford University Press, McAuliffe, W.E. and Gordon, R.A., 1974. A test of Lindesmith’s theory of
Oxford. addiction: the frequency of euphoria among long-term addicts. American
Koob, G.F., Stinus, L., Le Moal, M. and Bloom, F.E., 1989. Opponent Journal of Sociology, 79, 795–840.
process theory of motivation: neurobiological evidence from studies McBride, W.J., Murphy, J.M., Gatto, G.J. et al., 1993. CNS mechanisms of
of opiate dependence. Neuroscience and Biobehavioral Reviews, 13, alcohol self-administration. Alcohol and Alcoholism , 2(Suppl.), 463–467.
135–140. McBride, W.J., Murphy, J.M. and Ikemoto, S., 1999. Localization of
Korpi, E.R., Mattila, M.J., Wisden, W. and Luddens, H., 1997. GABA(A)- brain reinforcement mechanisms: intracranial self-administration and
receptor subtypes: clinical efficacy and selectivity of benzodiazepine site intracranial place-conditioning studies. Behavioural Brain Research , 101,
ligands. Annals of Medicine, 29, 275–282. 129–152.
Kuzmin, A., Johansson, B., Zvartau, E.E. and Fredholm, B.B., 1999. Melis, M., Gessa, G.L. and Diana, M., 2000. Different mechanisms for
Caffeine, acting on adenosine A1 receptors, prevents the extinction dopaminergic excitation induced by opiates and cannabinoids in the
520 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

rat midbrain. Progress in Neuro-Psychopharmacology and Biological and reinstatement of intravenous D-amphetamine self-administration.
Psychiatry, 24, 993–1006. Journal of Neuroscience, 19, 4102–4109.
Mereu, G., Yoon, K.-W.P., Boi, V., Gessa, G.L., Naes, L. and Westfall, T.C., Risner, M.E. and Jones, B.E., 1976. Role of noradrenergic and dopaminergic
1987. Preferential stimulation of ventral tegmental area dopaminergic processes in amphetamine self-administration. Pharmacology, Biochem-
neurons by nicotine. European Journal of Pharmacology, 141, 395–400. istry and Behavior, 5, 477–482.
Meyer, D.K., Beinfeld, M.C., Oertel, W.H. and Brownstein, M.J., 1982. Risner, M.E. and Jones, B.E., 1980. Intravenous self-administration of
Origin of the cholecystokinin containing fibers in the caudate–putamen. cocaine and norcocaine by dogs. Psychopharmacology , 71, 83–89.
Science, 215, 187–188. Ritchie, J.M. and Greene, N.M., 1985. Local anesthetics. In: Gilman, A.G.,
Mogenson, G.J., Jones, D.L. and Yim, C.Y., 1980. From motivation to Goodman, L.S., Rall, T.W. and Murad, F. (eds), The Pharmacological
action: functional interface between the limbic system and the motor Basis of Therapeutics , pp. 302–321. Macmillan, New York.
system. Progress in Neurobiology , 14, 69–97. Roberts, D.C.S., Corcoran, M.E. and Fibiger, H.C., 1977. On the role of
Moreau, J.L. and Huber, G., 1999. Central adenosine A 2A receptors: an ascending catecholaminergic systems in intravenous self-administration
overview. Brain Research Reviews, 31, 65–82. of cocaine. Pharmacology Biochemistry and Behavior , 6, 615–620.
Morgan, M.E. and Vestal, R.E., 1989. Methylxanthine effects on caudate Roberts, D.C.S., Koob, G.F., Klonoff, P. and Fibiger, H.C., 1980. Extinction
dopamine release as measured by in vivo electrochemistry. Life Sciences, and recovery of cocaine self-administration following 6-OHDA lesions
45, 2025–2039. of the nucleus accumbens. Pharmacology Biochemistry and Behavior ,
Mor ón, J.A., Brockington, A., Wise, R.A., Rocha, B.A. and Hope, B.T., 12, 781–787.
2001. Dopamine uptake through the norepinephrine transporter in brain Robinson, T.E. and Becker, J.B., 1986. Enduring changes in brain and
regions with low levels of the dopamine transporter: evidence from behavior produced by chronic amphetamine administration: a review and
knockout mouse lines. Journal of Neuroscience, 22, 389–395. evaluation of animal models of amphetamine psychosis. Brain Research
Nauta, W.J.H., Smith, G.P., Faull, R.L.M. and Domesick, V.B., 1978. Reviews, 11, 157–198.
Efferent connections and nigral afferents of the nucleus accumbens septi Robinson, T.E. and Berridge, K.C., 1993. The neural basis of drug craving:
in the rat. Neuroscience, 3, 385–401. an incentive-sensitization theory of addiction. Brain Research Reviews,
Nestler, E.J., Terwilliger, R.Z., Walker, J.R., Sevarino, K.A. and Duman, 18, 247–292.
R.S., 1990. Chronic cocaine treatment decreases levels of the G protein Rodd-Henricks, Z.A., McKenzie, D.L., Crile, R.S., Murphy, J.M. and
subunits Giα and Goα in discrete regions of rat brain. Journal of McBride, W.J., 2000. Regional heterogeneity for the intracranial self-
Neurochemistry , 55, 1079–1082. administration of ethanol within the ventral tegmental area of female
Ng Cheong Ton, J.M., Gerhardt, G.A., Friedemann, M. et al., 1988. Wistar rats. Psychopharmacology , 149, 217–224.
The effects of 9 -tetrahydrocannabinol on potassium-evoked release of Rohde, D.S., Detweiler, D.J. and Basbaum, A.I., 1996. Spinal cord mech-
dopamine in the rat caudate nucleus: an in vivo electrochemical and anisms of opioid tolerance and dependence: Fos-like immunoreactivity
in vivo microdialysis study. Brain Research, 451, 59–68. increases in subpopulations of spinal cord neurons during withdrawal.
Nisell, M., Nomikos, G.G. and Svensson, T.H., 1994. Infusion of nicotine Neuroscience, 72, 233–242.
in the ventral tegmental area or the nucleus accumbens of the rat Rosin, D.A., Robeva, A., Woodard, R.L., Guyenet, P.G. and Linden, J.,
differentially affects accumbal dopamine release. Pharmacology and 1998. Immunohistochemical localization of adenosine A2A receptors in
Toxicology, 75, 348–352. the rat central nervous system. Journal of Comparative Neurology, 401,
Nomikos, G.G., Damsma, G., Wenkstern, B.A. and Fibiger, H.C., 1990. 163–186.
In vivo characterization of locally applied dopamine uptake inhibitors by Routtenberg, A., 1972. Intracranial chemical injection and behavior: a
striatal microdialysis. Synapse, 6, 106–112. critical review. Behavioral Biology, 7, 601–641.
Oakman, S.A., Faris, P.L., Kerr, P.E., Cozzari, C. and Hartman, B.K., Routtenberg, A. and Sloan, M., 1972. Self-stimulation in the frontal cortex
1995. Distribution of pontomesencephalic cholinergic neurons projecting of Rattus norvegicus. Behavioral Biology, 7, 567–572.
to substantia nigra differs significantly from those projecting to ventral Samanin, R., Bernasconi, S. and Garattini, S., 1975. The effect of
tegmental area. Journal of Neuroscience, 15, 5859–5869. nomifensine on the depletion of brain serotonin and catecholamines
Ohmori, T., Koyama, T., Nakamura, F., Wang, P. and Yamashita, I., induced respectively by fenfluramine and 6-hydroxydopamine. European
1992. Effect of phencyclidine on spontaneous and N-methyl-D-aspartate Journal of Pharmacology, 34, 377–380.
(NMDA)-induced efflux of dopamine from superfused slices of rat Samson, H.H., Hodge, C.W., Tolliver, G.A. and Haraguchi, M., 1993. Effect
striatum. Neuropharmacology, 31, 461–467. of dopamine agonists and antagonists on ethanol-reinforced behavior:
Olds, M.E., 1982. Reinforcing effects of morphine in the nucleus accum- the involvement of the nucleus accumbens. Brain Research Bulletin, 30,
bens. Brain Research, 237, 429–440. 133–141.
Olmstead, M.C. and Franklin, K.B., 1997. The development of a con- Sandberg, M., Ward, H.K. and Bradford, H.F., 1985. Effect of cortico-
ditioned place preference to morphine: effects of microinjections into striate pathway lesion on the activities of enzymes involved in synthesis
various CNS sites. Behavioral Neuroscience, 111(6), 1324–1334. and metabolism of amino acid neurotransmitters in the striatum. Journal
Pettit, H.O. and Justice, J.B. Jr., 1989. Dopamine in the nucleus accumbens of Neurochemistry, 44, 42–47.
during cocaine self-administration as studied by in vivo microdialysis. Schenk, S. and Partridge, B., 1999. Cocaine-seeking produced by
Pharmacology, Biochemistry and Behavior , 34, 899–904. experimenter-administered drug injections: dose–effect relationships in
Phillips, G.D., Robbins, T.W. and Everitt, B.J., 1994. Bilateral intra- rats. Psychopharmacology , 147, 285–290.
accumbens self-administration of d-amphetamine: antagonism with Schenk, S., Valadez, A., Horger, B.A., Snow, S. and Wellman, P.J., 1994.
intra-accumbens SCH-23390 and sulpiride. Psychopharmacology , 114, Interactions between caffeine and cocaine in tests of self-administration.
477–485. Behavioural Pharmacology , 5, 153–158.
Piazza, P.V., Deminiere, J.M., Le Moal, M. and Simon, H., 1990. Schildein, S., Ågmo, A., Huston, J.P. and Schwarting, R.K.W., 1998.
Stress- and pharmacologically-induced behavioral sensitization increases Intraaccumbens injections of substance P, morphine and amphetamine:
vulnerability to acquisition of amphetamine self-administration. Brain effects on conditioned place preference and behavioral activity. Brain
Research, 514, 22–26. Research, 790, 185–194.
Pocock, D., Leeb, K. and Wise, R.A., 1994. Elevations and phasic Schulteis, G., Markou, A., Cole, M. and Koob, G.F., 1995. Decreased brain
fluctuations in nucleus accumbens dopamine (DA) during IV heroin self- reward produced by ethanol withdrawal. Proceedings of the National
administration. Society for Neuroscience Abstracts, 20, 1234. Academy of Sciences USA, 92, 5880–5884.
Poleszak, E. and Malec, D., 2000. Influence of adenosine receptor agonists Sesack, S.R. and Pickel, V.M., 1992. Prefrontal cortical efferents in the
and antagonists on amphetamine-influenced stereotypy in rats. Polish rat synapse on unlabeled neuronal targets of catecholamine terminals in
Journal of Pharmacology, 52, 423–429. the nucleus accumbens septi and on dopamine neurons in the ventral
Pitcher, J.A., Freedman, N.J. and Lefkowitz, R.J., 1998. G protein-coupled tegmental area. Journal of Comparative Neurology , 320, 145–160.
receptor kinases. Annual Review of Biochemistry , 67, 653–692. Sesack, S.R., Deutch, A.Y., Roth, R.H. and Bunney, B.S., 1989. Topo-
Ranaldi, R. and Wise, R.A., 2001. Blockade of D1 dopamine receptors in the graphical organization of the efferent projections of the medial prefrontal
ventral tegmental area decreases cocaine reward: possible role for den- cortex in the rat: an anterograde tract-tracing study with Phaseolus vul-
dritically released dopamine. Journal of Neuroscience, 21, 5841–5846. garis leucoagglutinin. Journal of Comparative Neurology, 290, 213–242.
Ranaldi, R., Pocock, D., Zereik, R. and Wise, R.A., 1999. Dopamine Sesack, S.R., Bressler, C.N. and Lewis, D.A., 1995. Ultrastructural asso-
fluctuations in the nucleus accumbens during maintenance, extinction, ciations between dopamine terminals and local circuit neurons in the
FUNCTIONAL ANATOMY 521

monkey prefrontal cortex: a study of calretinin-immunoreactive cells. Terwilliger, R.Z., Beitner-Johnson, D., Sevarino, K.A., Crain, S.M. and
Neuroscience Letters, 200, 9–12. Nestler, E.J., 1991. A general role for adaptations in G-proteins and the
Sesack, S.R., Hawrylak, V.A., Matus, C., Guido, M.A. and Levey, A.I., cyclic AMP system in mediating the chronic actions of morphine and
1998. Dopamine axon varicosities in the prelimbic division of the cocaine on neuronal function. Brain Research, 548, 100–110.
rat prefrontal cortex exhibit sparse immunoreactivity for the dopamine Thompson, A.C., Zapata, A., Justice, J.B. Jr., Vaughan, R.A., Sharpe, L.G.
transporter. Journal of Neuroscience, 18, 2697–2708. and Shippenberg, T.S., 2000. Kappa-opioid receptor activation modifies
Shalev, U., Morales, M., Hope, B., Yap, J. and Shaham, Y., 2001. Time- dopamine uptake in the nucleus accumbens and opposes the effects of
dependent changes in extinction behavior and stress-induced reinstate- cocaine. Journal of Neuroscience, 20, 9333–9340.
ment of drug seeking following withdrawal from heroin in rats. Psy- Turgeon, S.M., Pollack, A.E., Schusheim, L. and Fink, J.S., 1996. Effects of
chopharmacology, 156, 98–107. selective adenosine A1 and A2a agonists on amphetamine-induced loco-
Shiffman, S.M., 1979. The tobacco withdrawal syndrome. In: Krasnegor, motion and c-Fos in striatum and nucleus accumbens. Brain Research,
N.A. (ed.), Cigarette Smoking as a Dependence Process, pp. 158–184. 707, 75–80.
National Institute on Drug Abuse, Rockville, MD. Vaccarino, F.J., Bloom, F.E. and Koob, G.F., 1985. Blockade of nucleus
Shippenberg, T.S. and Heidbreder, C., 1995. Sensitization to the conditioned accumbens opiate receptors attenuates intravenous heroin reward in the
rewarding effects of cocaine: pharmacological and temporal character- rat. Psychopharmacology , 86, 37–42.
istics. Journal of Pharmacology and Experimental Therapeutics, 273, Valjent, E. and Maldonado, R., 2000. A behavioural model to reveal place
808–815. preference to 9-tetrahydrocannabinol in mice. Psychopharmacology ,
Shippenberg, T.S., Heidbreder, C. and Lefevour, A., 1996. Sensitization to 147, 436–438.
the conditioned rewarding effects of morphine: pharmacology and tem- Van Bockstaele, E.J. and Pickel, V.M., 1995. GABA-containing neurons in
poral characteristics. European Journal of Pharmacology , 299, 33–39. the ventral tegmental area project to the nucleus accumbens in rat brain.
Shoaib, M., Swanner, L.S., Yasar, S. and Goldberg, S.R., 1999. Chronic Brain Research, 682, 215–221.
caffeine exposure potentiates nicotine self-administration in rats. Psy- van der Kooy, D., Mucha, R.F., O’Shaughnessy, M. and Bucenieks, P.,
chopharmacology, 142, 327–333. 1982. Reinforcing effects of brain microinjections of morphine revealed
Sklair-Tavron, L., Shi, W.X., Lane, S.B., Harris, H.W., Bunney, B.S. by conditioned place preference. Brain Research, 243, 107–117.
and Nestler, E.J., 1996. Chronic morphine induces visible changes in Waldeck, B., 1972. Increased accumulation of (3 H)catecholamines formed
the morphology of mesolimbic dopamine neurons. Proceedings of the from ( 3 H)dopa after treatment with caffeine and aminophylline. Journal
National Academy of Sciences USA, 93, 11202–11207. of Pharmacy and Pharmacology, 24, 654–655.
Solomon, R.L. and Corbit, J.D., 1973. An opponent-process theory of Waldeck, B., 1975. Effect of caffeine on locomotor activity and central
motivation: II. Cigarette addiction. Journal of Abnormal Psychology, 81, catecholamine mechanisms: a study with special reference to drug
158–171. interaction. Acta Pharmacologica et Toxicologica, 36 (Suppl. 4), 1–23.
Spyraki, C. and Fibiger, H.C., 1988. A role of the mesolimbic dopamine Wang, H., Moriwaki, A., Wang, J.B., Uhl, G.R. and Pickel, V.M., 1997.
system in the reinforcing properties of diazepam. Psychopharmacology , Ultrastructural immunocytochemical localization of mu-opioid receptors
94, 133–137. in dendritic targets of dopaminergic terminals in the rat caudate–putamen
Spyraki, C., Fibiger, H.C. and Phillips, A.G., 1982. Attenuation by nucleus. Neuroscience, 81, 757–771.
haloperidol of place preference conditioning using food reinforcement. Watanabe, H. and Uramoto, H., 1986. Caffeine mimics dopamine receptor
Psychopharmacology , 77, 379–382. agonists without stimulation of dopamine receptors. Neuropharmacology,
Stinus, L., Winnock, M. and Kelley, A.E., 1985. Chronic neuroleptic 25, 577–581.
treatment and mesolimbic dopamine denervation induce behavioral Watkins, S.S., Stinus, L., Koob, G.F. and Markou, A., 2000. Reward
supersensitivity to opiates. Psychopharmacology , 85, 323–328. and somatic changes during precipitated nicotine withdrawal in rats:
Stinus, L., Nadaud, D., Jauregui, J. and Kelley, A.E., 1986. Chronic treat- centrally and peripherally mediated effects. Journal of Pharmacology and
ment with five different neuroleptics elicits behavioral supersensitivity to Experimental Therapeutics , 292, 1053–1064.
opiate infusion into the nucleus accumbens. Biological Psychiatry, 21, Wei, E.T., 1981. Enkephalin analogs and physical dependence. Journal of
34–48. Pharmacology and Experimental Therapeutics , 216, 12–18.
Stornetta, R.L., Norton, F.E. and Guyenet, P.G., 1993. Autonomic areas Wei, E.T. and Loh, H.H., 1976. Physical dependence on opiate-like peptides.
of rat brain exhibit increased Fos-like immunoreactivity during opiate Science, 193, 1262–1263.
withdrawal in rats. Brain Research, 624, 19–28. Wei, E.T., Tseng, L.F., Loh, H.H. and Way, E.L., 1974. Similarity of
Svenningsson, P., Le Moine, C., Kull, B., Sunahara, R., Bloch, B. and morphine abstinence signs to thermoregulatory behavior. Nature, 247,
Fredholm, B.B., 1997. Cellular expression of adenosine A2A receptor 398–399.
messenger RNA in the rat central nervous system with special reference Weight, F.F., Lovinger, D.M. and White, G., 1991. Alcohol inhibition of
to dopamine innervated areas. Neuroscience, 80, 1171–1185. NMDA channel function. Alcohol and Alcoholism , 1(Suppl.), 163–169.
Svenningsson, P., Le Moine, C., Aubert, I., Burbaud, P., Fredholm, B.B. and Weiss, F., Mitchiner, M., Bloom, F.E. and Koob, G.F., 1990. Free-
Bloch, B., 1998. Cellular expression of adenosine A2A receptor mRNA in choice responding for ethanol versus water in alcohol preferring (P)
the primate striatum. Journal of Comparative Neurology, 399, 229–240. and unselected Wistar rats is differentially modified by naloxone,
Svenningsson, P., Fourreau, L., Bloch, B., Fredholm, B.B., Gonon, F. bromocriptine, and methysergide. Psychopharmacology , 101, 178–186.
and Le Moine, C., 1999. Opposite tonic modulation of dopamine Weiss, F., Lorang, M.T., Bloom, F.E. and Koob, G.F., 1993. Oral alcohol
and adenosine on c-fos gene expression in striatopallidal neurons. self-administration stimulates dopamine release in the rat nucleus accum-
Neuroscience, 89, 827–837. bens: genetic and motivational determinants. Journal of Pharmacology
Tanda, G., Pontieri, F.E. and Di Chiara, G., 1997a. Cannabinoid and heroin and Experimental Therapeutics, 267, 250–258.
activation of mesolimbic dopamine transmission by a common µ1 opioid Welzl, H., Kuhn, G. and Huston, J.P., 1989. Self-administration of small
receptor mechanism. Science, 276, 2048–2050. amounts of morphine through glass micropipettes into the ventral
Tanda, G., Pontieri, F.E., Frau, R. and Di Chiara, G., 1997b. Contribution tegmental area of the rat. Neuropharmacology, 28, 1017–1023.
of blockade of the noradrenaline carrier to the increase of extracellular West, T.E.G. and Wise, R.A., 1988. Nucleus accumbens opioids facilitate
dopamine in the rat prefrontal cortex by amphetamine and cocaine. brain stimulation reward. Society for Neuroscience Abstracts, 14, 1102.
European Journal of Neuroscience, 9, 2077–2085. Williams, M.N. and Faull, R.L., 1988. The nigrotectal projection and
Tanda, G., Munzar, P. and Goldberg, S.R., 2000. Self-administration tectospinal neurons in the rat: a light and electron microscopic study
behavior is maintained by the psychoactive ingredient of marijuana in demonstrating a monosynaptic nigral input to identified tectospinal
squirrel monkeys. Nature Neuroscience, 3, 1073–1074. neurons. Neuroscience, 25, 533–562.
Tatum, A.L., Seevers, M.H. and Collins, K.H., 1929. Morphine addiction Wise, R.A., 1978. Catecholamine theories of reward: a critical review. Brain
and its physiological interpretation based on experimental evidence. Research, 152, 215–247.
Journal of Pharmacology and Experimental Therapeutics, 36, 447–475. Wise, R.A., 1987. The role of reward pathways in the development of drug
Tepper, J.M., Martin, L.P. and Anderson, D.R., 1995. GABAA receptor- dependence. Pharmacology and Therapeutics , 35, 227–263.
mediated inhibition of rat substantia nigra dopaminergic neurons Wise, R.A., 1988. The neurobiology of craving: implications for under-
by pars reticulata projection neurons. Journal of Neuroscience, 15, standing and treatment of addiction. Journal of Abnormal Psychology ,
3092–3103. 97, 118–132.
522 CLINICAL SYNDROMES: SUBSTANCE-RELATED DISORDERS

Wise, R.A., 1989. Opiate reward: sites and substrates. Neuroscience and Woods, J.H. and Schuster, C.R., 1971. Opiates as reinforcing stimuli.
Biobehavioral Reviews , 13, 129–133. In: Thompson, T. and Pickens, R. (eds), Stimulus Properties of Drugs,
Wise, R.A., 1996a. Neurobiology of addiction. Current Opinion in Neuro- pp. 163–175. Appleton, Century, Crofts, New York.
biology, 6, 243–251. Yamamoto, B.K. and Novotney, S., 1998. Regulation of extracellular
Wise, R.A., 1996b. Addictive drugs and brain stimulation reward. Annual dopamine by the norepinephrine transporter. Journal of Neurochemistry,
Review of Neuroscience, 19, 319–340. 71, 274–280.
Wise, R.A. and Bozarth, M.A., 1987. A psychomotor stimulant theory of Yokel, R.A. and Wise, R.A., 1975. Increased lever-pressing for amphetamine
addiction. Psychological Review, 94, 469–492. after pimozide in rats: implications for a dopamine theory of reward.
Wise, R.A. and Hoffman, D.C., 1992. Localization of drug reward mecha- Science, 187, 547–549.
nisms by intracranial injections. Synapse, 10, 247–263. Yokel, R.A. and Wise, R.A., 1976. Attenuation of intravenous amphetamine
Wise, R.A. and Munn, E., 1995. Withdrawal from chronic amphetamine reinforcement by central dopamine blockade in rats. Psychopharmacol-
elevates baseline intracranial self-stimulation thresholds. Psychopharma- ogy, 48, 311–318.
cology, 117, 130–136. You, Z.-B., Tzschentke, T.M., Brodin, E. and Wise, R.A., 1998. Electrical
Wise, R.A., Spindler, J., deWit, H. and Gerber, G.J., 1978. Neuroleptic- stimulation of the prefrontal cortex increases cholecystokinin, glutamate,
induced ‘anhedonia’ in rats: pimozide blocks the reward quality of food. and dopamine release in the nucleus accumbens: an in vivo microdialysis
Science, 201, 262–264. study in freely moving rats. Journal of Neuroscience, 18, 6492–6500.
Wise, R.A., Leone, P., Rivest, R. and Leeb, K., 1995a. Elevations of nucleus Zangen, A., Ikemoto, S. and Wise, R.A., 2000. Rewarding and locomotor
accumbens dopamine and DOPAC levels during intravenous heroin self- activity effects of endomorphin-1 in the rat brain. Society for Neuro-
administration. Synapse, 21, 140–148. science Abstracts, 26, 474.
Wise, R.A., Newton, P., Leeb, K., Burnette, B., Pocock, P. and Justice, J.B. Zetterström, T., Herrera-Marschitz, U. and Ungerstedt, U., 1981. Simulta-
Jr., 1995b. Fluctuations in nucleus accumbens dopamine concentration neous estimation of dopamine release and rotational behaviour induced
during intravenous cocaine self-administration in rats. Psychopharma- by d-amphetamine in 6-OH-DA denervated rats. Neuroscience Letters,
cology, 120, 10–20. Suppl. 7, S27.

You might also like